Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38325866

RESUMEN

Regardless of the underlying etiology, renal fibrosis is the final histological outcome of progressive kidney disease. Unilateral ureteral obstruction (UUO) is an ideal and reproducible experimental rodent model of renal fibrosis, which is characterized by tubulointerstitial inflammatory responses, accumulation of extracellular matrix, tubular dilatation and atrophy, and fibrosis. The magnitude of UUO-induced renal fibrosis is experimentally manipulated by the species chosen, animal age, and the severity and duration of the obstruction, while relief of the obstruction allows the animal to recover from fibrosis. The pathogenesis of renal fibrosis is complex and multifactorial and is orchestrated by activation of renin-angiotensin system (RAS), oxidative stress, inflammatory response, transforming growth factor beta 1-Smad pathway, activated myofibroblasts, cell death (apoptosis, autophagy, ferroptosis, and necroptosis), destruction of intracellular organelles, and signaling pathway. The current therapeutic approaches have limited efficacy. Inhibition of RAS and use of antioxidants and antidiabetic drugs, such as inhibitors of sodium-glucose cotransporter 2 and dipeptidyl peptidase-4, have recently gained attention as therapeutic strategies to prevent renal scarring. This literature review highlights the state of the art regarding the molecular mechanisms relevant to the management of renal fibrosis caused by UUO.

2.
PLoS One ; 18(6): e0286903, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37310976

RESUMEN

The angiotensin receptor neprilysin inhibitor LCZ696 affords superior cardioprotection and renoprotection compared with renin-angiotensin blockade monotherapy, but the underlying mechanisms remain elusive. Herein, we evaluated whether LCZ696 attenuates renal fibrosis by inhibiting ASK1/JNK/p38 mitogen-activated protein kinase (MAPK)-mediated apoptosis in a rat model of unilateral ureteral obstruction (UUO) and in vitro. Rats with UUO were treated daily for 7 days with LCZ696, valsartan, or the selective ATP competitive inhibitor of apoptosis signal-regulating kinase 1 (ASK1), GS-444217. The effects of LCZ696 on renal injury were examined by assessing the histopathology, oxidative stress, intracellular organelles, apoptotic cell death, and MAPK pathways. H2O2-exposed human kidney 2 (HK-2) cells were also examined. LCZ696 and valsartan treatment significantly attenuated renal fibrosis caused by UUO, and this was paralleled by downregulation of proinflammatory cytokines and decreased inflammatory cell influx. Intriguingly, LCZ696 had stronger effects on renal fibrosis and inflammation than valsartan. UUO-induced oxidative stress triggered mitochondrial destruction and endoplasmic reticulum stress, which resulted in apoptotic cell death; these effects were reversed by LCZ696. Both GS-444217 and LCZ696 hampered the expression of death-associated ASK1/JNK/p38 MAPKs. In H2O2-treated HK-2 cells, LCZ696 and GS-444217 increased cell viability but decreased the production of intracellular reactive oxygen species and MitoSOX and apoptotic cell death. Both agents also deactivated H2O2-stimulated activation of ASK1/JNK/p38 MAPKs. These findings suggest that LCZ696 protects against UUO-induced renal fibrosis by inhibiting ASK1/JNK/p38 MAPK-mediated apoptosis.


Asunto(s)
Enfermedades Renales , Proteína Quinasa 14 Activada por Mitógenos , Obstrucción Ureteral , Humanos , Animales , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos , Neprilisina , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/tratamiento farmacológico , Receptores de Angiotensina , Peróxido de Hidrógeno , MAP Quinasa Quinasa Quinasa 5 , Valsartán/farmacología , Antihipertensivos , Antivirales , Apoptosis
3.
PLoS One ; 17(10): e0274116, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36223414

RESUMEN

Renal fibrosis represents the final common outcome of chronic kidney disease of virtually any etiology. However, the mechanism underlying the evolution of renal fibrosis remains to be addressed. This study sought to clarify whether RIP1-RIP3-mediated necroptosis is involved in renal fibrosis via Wnt3α/ß-catenin/GSK-3ß signaling in vitro and in a rat model of unilateral ureteral obstruction (UUO). Rats with UUO were administered RIP inhibitors (necrostatin-1 or GSK872) or ß-catenin/TCF inhibitor ICG-001 daily for 7 consecutive days. UUO caused significant renal tubular necrosis and overexpression of RIP1-RIP3-MLKL axis proteins, and was accompanied by activation of the NLRP3 inflammasome and renal fibrosis. Oxidative stress caused by UUO was closely associated with endoplasmic reticulum stress and mitochondrial dysfunction, which resulted in apoptotic cell death via Wnt3α/ß-catenin/GSK-3ß signaling. All of these effects were abolished by an RIP inhibitor (necrostatin-1 or GSK872) or ICG-001. In H2O2-treated HK-2 cells, both RIP inhibitor and ICG-001 decreased intracellular reactive oxygen species production and apoptotic cells, but increased cell viability. Activated Wnt3α/ß-catenin/GSK-3ß signaling was decreased by either RIP inhibitor or ICG-001. Our findings suggest that RIP1-RIP3-mediated necroptosis contributes to the development of renal fibrosis via Wnt3α/ß-catenin/GSK-3ß signaling in UUO and may be a therapeutic target for protection against renal scarring of other origins.


Asunto(s)
Enfermedades Renales , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Obstrucción Ureteral , Animales , Fibrosis , Glucógeno Sintasa Quinasa 3 beta , Peróxido de Hidrógeno , Inflamasomas , Enfermedades Renales/complicaciones , Proteína con Dominio Pirina 3 de la Familia NLR , Necroptosis , Ratas , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Obstrucción Ureteral/complicaciones , beta Catenina/metabolismo
4.
5.
Int Immunopharmacol ; 108: 108868, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35636077

RESUMEN

OBJECTIVE: Coenzyme Q10 (CoQ10) protects against various types of injury, but its role in preventing renal scarring in chronic kidney disease remains an open question. Herein, we evaluated whether CoQ10 attenuates renal fibrosis by interfering with necroinflammation in a rat model of unilateral ureteral obstruction (UUO) and in vitro. METHODS: Rats with UUO were treated daily with CoQ10 or an RIP inhibitor (necrostatin-1 or GSK872) for 7 days. The influence of CoQ10 on renal injury caused by UUO was evaluated by histopathology and analysis of gene expression, oxidative stress, intracellular organelles, apoptosis, and Wnt3α/ß-catenin/GSK-3ß signaling·H2O2-exposed human kidney (HK-2) cells were also examined after treatment with CoQ10 or an RIP inhibitor. RESULTS: UUO induced marked renal tubular necrosis, upregulation of RIP1-RIP3-MLKL axis proteins, activation of the NLRP3 inflammasome, and evolution of renal fibrosis. UUO-induced oxidative stress evoked excessive endoplasmic reticulum stress and mitochondrial dysfunction, which triggered apoptotic cell death through Wnt3α/ß-catenin/GSK-3ß signaling. All of these effects were mitigated by CoQ10 or an RIP inhibitor. In H2O2-treated HK-2 cells, CoQ10 or an RIP inhibitor suppressed the expression of RIP1-RIP3-MLKL proteins and pyroptosis-related cytokines, and hindered the production of intracellular reactive oxygen species as shown by MitoSOX Red staining and apoptotic cell death but increased cell viability. The CoQ10 or Wnt/ß-catenin inhibitor ICG-001 deactivated H2O2-stimulated activation of Wnt3α/ß-catenin/GSK-3ß signaling. CONCLUSION: These findings suggest that CoQ10 attenuates renal fibrosis by inhibiting RIP1-RIP3-MLKL-mediated necroinflammation via Wnt3α/ß-catenin/GSK-3ß signaling in UUO.


Asunto(s)
Enfermedades Renales , Obstrucción Ureteral , Animales , Fibrosis , Glucógeno Sintasa Quinasa 3 beta , Peróxido de Hidrógeno/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/patología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología , Proteínas Quinasas/metabolismo , Ratas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ubiquinona/análogos & derivados , Obstrucción Ureteral/tratamiento farmacológico , beta Catenina
6.
Korean J Intern Med ; 36(6): 1437-1449, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34666433

RESUMEN

BACKGROUND/AIMS: Cigarette smoking is an important modifiable risk factor in kidney disease progression. However, the underlying mechanisms for this are lacking. This study aimed to assess whether nicotine (NIC), a major toxic component of cigarette smoking, would exacerbates tacrolimus (TAC)-induced renal injury. METHODS: Sprague-Dawley rats were treated daily with NIC, TAC, or both drugs for 4 weeks. The influence of NIC on TAC-caused renal injury was examined via renal function, histopathology, oxidative stress, mitochondria, endoplasmic reticulum (ER) stress, and programmed cell death (apoptosis and autophagy). RESULTS: Both NIC and TAC significantly impaired renal function and histopathology, while combined NIC and TAC treatment aggravated these parameters beyond the effects of either alone. Increased oxidative stress, ER stress, mitochondrial dysfunction, proinf lammatory and profibrotic cytokine expressions, and programmed cell death from either NIC or TAC were also aggravated by the two combined. CONCLUSION: Our observations suggest that NIC exacerbates chronic TAC nephrotoxicity, implying that smoking cessation may be beneficial for transplant smokers taking TAC.


Asunto(s)
Nicotina , Tacrolimus , Animales , Apoptosis , Riñón/fisiología , Nicotina/toxicidad , Ratas , Ratas Sprague-Dawley , Tacrolimus/toxicidad
7.
Acta Pharmacol Sin ; 42(1): 77-87, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32555441

RESUMEN

Reducing immunosuppressant-related complications using conventional drugs is an efficient therapeutic strategy. L-carnitine (LC) has been shown to protect against various types of renal injury. In this study, we investigated the renoprotective effects of LC in a rat model of chronic tacrolimus (TAC) nephropathy. SD rats were injected with TAC (1.5 mg · kg-1 · d-1, sc) for 4 weeks. Renoprotective effects of LC were assessed in terms of renal function, histopathology, oxidative stress, expression of inflammatory and fibrotic cytokines, programmed cell death (pyroptosis, apoptosis, and autophagy), mitochondrial function, and PI3K/AKT/PTEN signaling. Chronic TAC nephropathy was characterized by severe renal dysfunction and typical histological features of chronic nephropathy. At a molecular level, TAC markedly increased the expression of inflammatory and fibrotic cytokines in the kidney, induced oxidative stress, and led to mitochondrial dysfunction and programmed cell death through activation of PI3K/AKT and inhibition of PTEN. Coadministration of LC (200 mg · kg-1 · d-1, ip) caused a prominent improvement in renal function and ameliorated histological changes of kidneys in TAC-treated rats. Furthermore, LC exerted anti-inflammatory and antioxidant effects, prevented mitochondrial dysfunction, and modulated the expression of a series of apoptosis- and autophagy-controlling genes to promote cell survival. Human kidney proximal tubular epithelial cells (HK-2 cells) were treated with TAC (50 µg/mL) in vitro, which induced production of intracellular reactive oxygen species and expression of an array of genes controlling programmed cell death (pyroptosis, apoptosis, and autophagy) through interfering with PI3K/AKT/PTEN signaling. The harmful responses of HK-2 cells to TAC were significantly attenuated by cotreatment with LC and the PI3K inhibitor LY294002 (25 µM). In conclusion, LC treatment protects against chronic TAC nephropathy through interfering the PI3K/AKT/PTEN signaling.


Asunto(s)
Antiinflamatorios/uso terapéutico , Apoptosis/efectos de los fármacos , Carnitina/uso terapéutico , Enfermedades Renales/prevención & control , Sustancias Protectoras/uso terapéutico , Transducción de Señal/efectos de los fármacos , Animales , Antiinflamatorios/química , Autofagia/efectos de los fármacos , Carnitina/química , Línea Celular , Cromonas/farmacología , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Masculino , Mitocondrias/efectos de los fármacos , Morfolinas/farmacología , Estrés Oxidativo/efectos de los fármacos , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Sustancias Protectoras/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piroptosis/efectos de los fármacos , Ratas Sprague-Dawley , Estereoisomerismo , Tacrolimus
8.
Korean J Intern Med ; 36(Suppl 1): S180-S195, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32942841

RESUMEN

BACKGROUND/AIMS: Accumulating evidence indicates that L-carnitine (LC) protects against multiorgan damage through its antioxidant properties and preservation of the mitochondria. Little information is available about the effects of LC on renal fibrosis. This study examined whether LC treatment would provide renoprotection in a rat model of unilateral ureteral obstruction (UUO) and in vitro. METHODS: Sprague-Dawley rats that underwent UUO were treated daily with LC for 7 or 14 days. The influence of LC on renal injury caused by UUO was evaluated by histopathology, and analysis of gene expression, oxidative stress, mitochondrial function, programmed cell death, and phosphatidylinositol 3-kinase (PI3K)/ AKT/forkhead box protein O 1a (FoxO1a) signaling. In addition, H2O2-exposed human kidney cells (HK-2) were treated with LC. RESULTS: LC treatment inhibited expression of proinflammatory and profibrotic cytokines, and was followed by a significant attenuation of tubulointerstitial inflammation and fibrosis. The increased oxidative stress caused by UUO was associated with mitochondrial dysfunction and excessive apoptosis and autophagy via PI3K/AKT/FoxO1a-dependent signaling, and this was abrogated by administration of LC. In H2O2-exposed HK-2 cells, LC decreased intracellular production of reactive oxygen species, and suppressed expression of profibrotic cytokines and reduced the number of apoptotic cells. CONCLUSION: LC protects against the progression of tubulointerstitial fibrosis in an obstructed kidney.


Asunto(s)
Obstrucción Ureteral , Animales , Carnitina , Fibrosis , Peróxido de Hidrógeno , Riñón/patología , Fosfatidilinositol 3-Quinasas , Ratas , Ratas Sprague-Dawley , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/patología
9.
Front Pharmacol ; 12: 798381, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069210

RESUMEN

Dapagliflozin, a sodium-glucose cotransporter-2 inhibitor, offers renoprotection in diabetes. However, potential for use in nondiabetic kidney disease remains unknown. Herein, we assessed whether dapagliflozin alleviates renal fibrosis by interfering with necroinflammation in a rat model of unilateral ureteral obstruction (UUO) and in vitro. After induction of UUO, rats were administered dapagliflozin daily for seven consecutive days. UUO induced significant renal tubular necrosis and overexpression of RIP1-RIP3-MLKL axis proteins; these coincided with NLRP3 inflammasome activation, and subsequent development of renal fibrosis. Oxidative stress caused by UUO is tightly associated with endoplasmic reticulum stress and mitochondrial dysfunction, leading to apoptotic cell death through Wnt3α/ß-catenin/GSK-3ß signaling; all of which were abolished by both dapagliflozin and specific RIP inhibitors (necrostatin-1 and GSK872). In H2O2-treated HK-2 cells, dapagliflozin and RIP inhibitors suppressed overexpression of RIP1-RIP3-MLKL proteins and pyroptosis-related cytokines, decreased intracellular reactive oxygen species production and apoptotic cell death, whereas cell viability was improved. Moreover, activated Wnt3α/ß-catenin/GSK-3ß signaling was inhibited by dapagliflozin and Wnt/ß-catenin inhibitor ICG-001. Our findings suggest that dapagliflozin ameliorates renal fibrosis by inhibiting RIP1-RIP3-MLKL-mediated necroinflammation via Wnt3α/ß-catenin/GSK-3ß signaling in UUO.

10.
Acta Pharmacol Sin ; 41(12): 1597-1608, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32300244

RESUMEN

Tissue kallikrein has protective function against various types of injury. In this study, we investigated whether exogenous pancreatic kininogenase (PK) conferred renoprotection in a rat model of unilateral ureteral obstruction (UUO) and H2O2-treated HK-2 cells in vitro. SD rats were subjected to UUO surgery, then PK (7.2 U/g per day, ip) was administered for 7 or 14 days. After the treatment, rats were euthanized; the obstructed kidneys were harvested for further examination. We found that PK administration significantly attenuated interstitial inflammation and fibrosis, and downregulated the expression of proinflammatory (MCP-1, TLR-2, and OPN) and profibrotic (TGF-ß1 and CTGF) cytokines in obstructed kidney. UUO-induced oxidative stress, closely associated with excessive apoptotic cell death and autophagy via PI3K/AKT/FoxO1a signaling, which were abolished by PK administration. We further showed that PK administration increased the expression of bradykinin receptors 1 and 2 (B1R and B2R) mRNA and the production of NO and cAMP in kidney tissues. Coadministration with either B1R antagonist (des-Arg9-[Leu8]-bradykinin) or B2R antagonist (icatibant) abrogated the renoprotective effects of PK, and reduced the levels of NO and cAMP in obstructed kidney. In H2O2-treated HK-2 cells, addition of PK (6 pg/mL) significantly decreased ROS production, regulated the expression of oxidant and antioxidant enzymes, suppressed the expression of TGF-ß1 and MCP-1, and inhibited cell apoptosis. Our data demonstrate that PK treatment protects against the progression of renal fibrosis in obstructed kidneys.


Asunto(s)
Fibrosis/prevención & control , Calicreínas/uso terapéutico , Riñón/metabolismo , Páncreas/enzimología , Sustancias Protectoras/uso terapéutico , Obstrucción Ureteral/complicaciones , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Fibrosis/etiología , Fibrosis/patología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/patología , Sistema Calicreína-Quinina/efectos de los fármacos , Riñón/patología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Obstrucción Ureteral/patología
11.
Korean J Intern Med ; 34(5): 1078-1090, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29432674

RESUMEN

BACKGROUND/AIMS: Evidence suggests that Shen-Kang (SK), a traditional Chinese herbal medicine, protects against various types of renal injury. In this study, we evaluated whether SK treatment confers renoprotection in a rat model of chronic tacrolimus (TAC) nephropathy. METHODS: Rats were treated daily with TAC (1.5mg/kg, subcutaneously) and SK (450 mg/kg, intravenously) for 4 weeks. The effects of SK on TAC-induced renal injury were assessed by measuring renal function, urine albumin excretion, histopathology, inflammatory cell infiltration, expression of profibrotic (transforming growth factor ß1 [TGF-ß1] and TGF-ß inducible gene-h3 [ßig-h3]) and proinflammatory cytokines, oxidative stress, and apoptotic cell death. RESULTS: Administration of SK preserved glomerular integrity (fractional mesangial area and Wilms tumor 1-positive glomeruli), attenuated tubulointerstitial fibrosis, and reduced the number of ectodermal dysplasia 1-positive cells, and this was paralleled by improved urine albumin excretion and renal dysfunction. At the molecular level, SK treatment suppressed expression of TGF-ß1/Smad2/3, ßig-h3, and proinflammatory cytokines. Oxidative stress and apoptotic cell death were significantly decreased with SK treatment, and apoptosis-related genes were regulated toward cell survival (active caspase-3 and the B-cell lymphoma-2/Bcl2-associated X [Bcl-2/Bax] ratio). CONCLUSION: SK protects against TAC-induced renal injury.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Enfermedades Renales/prevención & control , Riñón/efectos de los fármacos , Sustancias Protectoras/farmacología , Tacrolimus , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Citocinas/metabolismo , Citoprotección , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
12.
BMC Nephrol ; 19(1): 63, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540150

RESUMEN

BACKGROUND: Accumulating evidence suggests that a decrease in brain-derived neurotrophic factor (BDNF) level induces a variety of psychiatric and neurological disorders. However, the expression and role of BDNF in the kidney have not been explored. The present study examined the expression of BDNF and tropomyosin-related kinase (Trk) receptors in an experimental model of chronic cyclosporine A (CsA) nephropathy. METHODS: Sprague-Dawley rats on a salt-deplete diet were treated daily for four weeks with vehicle or CsA. Urine profiles, apoptotic cell death, oxidative stress (8-hydroxy-2'-deoxyguanosine, 8-OHdG), and expression of BDNF and Trk receptors (TrkB and TrkC) were compared between groups. The impact of vasopressin infusion on the urine-concentrating ability was examined by measuring the expression of aquaporin-2 (AQP-2) and BDNF and urine profiles in normal and CsA-treated rats. RESULTS: Compared with the vehicle-treated rats, rats given CsA had enhanced urine volume and declined urine osmolality. Immunohistochemistry and immunoblotting showed that BDNF and Trk receptors were constitutively expressed in kidneys from vehicle-treated rats. This was confirmed by double immunofluorescent staining for Na-K-ATPase-α1, AQP-1, and AQP-2. By contrast, the expression of these factors decreased in kidneys from CsA-treated rats (BDNF: 51.1 ± 19.5% vs. 102.0 ± 30.3%, p < 0.01). Downregulation of BDNF was accompanied by impairment of urine osmolality, and this was reversed by exogenous infusion of vasopressin. Notably, the number of TUNEL-positive cells correlated negatively with BDNF expression and positively with urinary 8-OHdG excretion. CONCLUSIONS: BDNF is expressed in the collecting duct of the kidney and may be associated with urine-concentrating ability in an experimental model of chronic CsA-induced nephropathy. Our study provides a new avenue for further investigation of chronic CsA nephropathy.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Ciclosporina/toxicidad , Inmunosupresores/toxicidad , Riñón/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Expresión Génica , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
13.
PLoS One ; 9(6): e100798, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24959755

RESUMEN

BACKGROUND: Tacrolimus (TAC)-induced pancreatic islet injury is one of the important causes of new-onset diabetes in transplant recipients. This study was performed to evaluate whether a dipeptidyl peptidase IV (DPP IV) inhibitor is effective in improving TAC-induced diabetes mellitus by reducing pancreatic islet injury. METHODS: Rats were treated with TAC (1.5 mg/kg, subcutaneously) and the DPP IV inhibitor MK-0626 (10 or 20 mg/kg, oral gavage) for 4 weeks. The effect of MK-0626 on TAC-induced diabetes was evaluated by assessing pancreatic islet function, histopathology. TAC-induced incretin dysfunction was also examined based on active glucagon-like peptide-1 (GLP-1) levels in the serum after glucose loading. The protective effect of MK-0626 was evaluated by measuring markers of oxidative stress, oxidative resistance, and apoptosis. To determine whether enhanced GLP-1 signaling is associated with these protective effects, we measured the expression of the GLP-1 receptor (GLP-1R) and the effect of the GLP-1 analog exendin-4 on cell viability and oxidative stress in isolated islets. RESULTS: MK-0626 treatment attenuated TAC-induced pancreatic islet dysfunction and islet morphology. TAC treatment led to a defect in active GLP-1 secretion; however, MK-0626 reversed these effects. TAC treatment increased the level of 8-hydroxy-2'-deoxyguanosine (8-OHdG), the number of apoptotic death, and the level of active caspase-3, and decreased the level of manganese superoxide dismutase and heme oxygenase-1; MK-0626 treatment reversed these changes. MK-0626 treatment restored the expression of GLP-1R, and direct administration of exendin-4 to isolated islets reduced TAC-induced cell death and 8-OHdG expression. CONCLUSIONS: The DPP IV inhibitor MK-0626 was an effective antidiabetic agent that exerted antioxidative and antiapoptotic effects via enhanced GLP-1 signaling in TAC-induced diabetics.


Asunto(s)
Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Islotes Pancreáticos/efectos de los fármacos , Sustancias Protectoras/farmacología , Tacrolimus/efectos adversos , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Daño del ADN/efectos de los fármacos , Desoxiguanosina/análogos & derivados , Desoxiguanosina/sangre , Desoxiguanosina/orina , Diabetes Mellitus Experimental , Islotes Pancreáticos/patología , Masculino , Ratas , Ratas Sprague-Dawley
14.
Nephron Exp Nephrol ; 126(3): 148-56, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24855017

RESUMEN

BACKGROUND: Leflunomide (LEF) and benazepril have renoprotective effects on diabetic nephropathy (DN) through their anti-inflammatory and anti-fibrotic activities. This study investigated whether combined treatment using LEF and benazepril affords superior protection compared with the respective monotherapies. METHODS: Diabetes was induced with streptozotocin (STZ, 65 mg/kg) by intraperitoneal injection in male Wistar rats. Two weeks after STZ injection, diabetic rats were treated daily for 12 weeks with LEF (10 mg/kg), benazepril (10 mg/kg), or a combination of both. Basic parameters (body weight, fasting blood glucose level, and 24 h urinary protein excretion), histopathology, inflammatory [inflammatory cell infiltration (ED-1), monocyte chemoattractant protein-1 (MCP-1), and Toll-like receptor-2 (TLR-2)] and glomerulosclerotic factors [transforming growth factor-ß1 (TGF-ß1) and connective tissue growth factor (CTGF)], and oxidative stress (8-hydroxy-2'-deoxyguanosine, 8-OHdG) were studied. RESULTS: Benazepril or LEF treatment significantly prevented body weight loss and 24 h urinary protein excretion induced by diabetes; combined treatment with LEF and benazepril further improved these parameters compared with giving each drug alone (all p < 0.01). Increased expression of inflammatory (MCP-1 and TLR-2) and glomerulosclerotic (TGF-ß1 and CTGF) factors in diabetic rat kidney was reduced by treatment with either LEF or benazepril and was further reduced by the combined administration of the two drugs (p < 0.01). These effects were accompanied by suppression of urinary 8-OHdG excretion. There was no significant between-group difference in blood glucose level. CONCLUSIONS: LEF treatment lessens DN, and combined treatment with LEF and benazepril provides synergistic effects in preventing DN.


Asunto(s)
Benzazepinas/administración & dosificación , Diabetes Mellitus Experimental/tratamiento farmacológico , Nefropatías Diabéticas/tratamiento farmacológico , Isoxazoles/administración & dosificación , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antifibrinolíticos/administración & dosificación , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/patología , Sinergismo Farmacológico , Leflunamida , Masculino , Ratas , Ratas Wistar , Resultado del Tratamiento
15.
Transplantation ; 98(1): 22-8, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24825522

RESUMEN

BACKGROUND: Drug-drug interaction between everolimus (EVR) and tacrolimus (TAC) is still undetermined. We evaluated whether EVR enhances TAC-induced organ injury through drug-drug interaction. METHODS: Tacrolimus (6 mg/kg) was given to rats with or without EVR (1 or 2 mg/kg) orally for 4 weeks. The influences of EVR on TAC-induced organ injury were evaluated in terms of nephrotoxicity and pancreatic islet dysfunction. Drug-drug interaction was evaluated by measuring the level of each drug in the blood and target tissue, and the correlation between the two drugs was observed in the blood and target tissue. The concentration of 8-hydroxy-2'-deoxyguanosine in blood or urine was measured as a marker of oxidative stress, and correlation between drug levels and oxidative stress was also evaluated. RESULTS: Tacrolimus treatment alone did not cause overt renal or pancreatic islet injury, but the addition of EVR significantly enhanced the TAC-induced organ injury, as demonstrated by aggravated nephrotoxicity and pancreatic islet dysfunction. The combination of EVR and TAC significantly increased each drug level in the target tissues as well as in blood, and there was good correlation between the two drugs in blood and target organs. The serum and urinary levels of 8-hydroxy-2'-deoxyguanosine were significantly increased in the TAC+EVR group compared with the TAC- or EVR-alone group and were well correlated with drug levels in blood and tissues. CONCLUSIONS: Everolimus enhances TAC-induced target organ injury by increasing oxidative stress via pharmacological interaction in blood and target tissue. This finding provides a better understanding of the effects of EVR when used in combination with TAC.


Asunto(s)
Inmunosupresores/toxicidad , Islotes Pancreáticos/efectos de los fármacos , Riñón/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Sirolimus/análogos & derivados , Tacrolimus/toxicidad , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Biomarcadores/sangre , Biomarcadores/orina , Desoxiguanosina/análogos & derivados , Desoxiguanosina/sangre , Desoxiguanosina/orina , Interacciones Farmacológicas , Quimioterapia Combinada , Everolimus , Inmunosupresores/sangre , Inmunosupresores/farmacocinética , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Riñón/metabolismo , Riñón/patología , Masculino , Ratas , Ratas Sprague-Dawley , Sirolimus/sangre , Sirolimus/farmacocinética , Sirolimus/toxicidad , Tacrolimus/sangre , Tacrolimus/farmacocinética
16.
Nephrology (Carlton) ; 19(8): 490-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24796922

RESUMEN

AIMS: Chronic cyclosporine (CsA) treatment induces autophagic cell death characterized by excessive autophagosome formation and decreased autophagic clearance. In this study, we evaluated the influence of ginseng treatment on autophagy in chronic CsA nephropathy. METHODS: Mice were treated with CsA (30 mg/kg) with or without Korean red ginseng (KRG) extract (0.2, 0.4 g/kg) for 4 weeks. The effect of KRG on CsA-induced autophagosome formation was measured using phospholipid-conjugated form of LC3-II, beclin-1, and autophagic vacuoles were visualized with electron microscopy. Autophagic clearance was evaluated by accumulation of p62/sequestosome 1 (p62) and ubiquitin, then double immunolabeling for p62 and either LC3-II or ubiquitin. To demonstrate the association between the effects of KRG treatment on autophagy and apoptosis, double immunolabelling for LC3-II and active caspase-3 was performed. Multiple autophagy pathways were also examined. RESULTS: KRG co-treatment significantly decreased the expression of LC3-II, beclin-1, and the number of autophagic vacuoles compared with the CsA group, and these changes were accompanied by improvements in renal dysfunction and fibrosis. CsA-induced accumulation of p62 and ubiquitin was also decreased by KRG treatment, and these proteins were colocalized with LC3-II and with each other. KRG treatment simultaneously reduced the expression of both active caspase-3 and LC3-II in the injured area. KRG treatment during chronic CsA nephropathy induced the expression of AKT/mTOR, which is a pathway that inhibits autophagy, and reduced AMPK expression. CONCLUSION: Ginseng treatment attenuated CsA-induced excessive autophagosome formation and autophagic aggregates. These findings suggest that ginseng has a renoprotective effect against CsA-induced autophagic cell death.


Asunto(s)
Autofagia/efectos de los fármacos , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/prevención & control , Panax , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Animales , Enfermedad Crónica , Ciclosporina/efectos adversos , Enfermedades Renales/inducido químicamente , Masculino , Ratones , Ratones Endogámicos ICR
17.
PLoS One ; 8(8): e72685, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24009697

RESUMEN

BACKGROUND: This study was performed to investigate whether ginseng has a protective effect in an experimental mouse model of cyclosporine-induced pancreatic injury. METHODS: Mice were treated with cyclosporine (30 mg/kg/day, subcutaneously) and Korean red ginseng extract (0.2 or 0.4 g/kg/day, oral gavage) for 4 weeks while on a 0.01% salt diet. The effect of ginseng on cyclosporine-induced pancreatic islet dysfunction was investigated by an intraperitoneal glucose tolerance test and measurements of serum insulin level, ß cell area, macrophage infiltration, and apoptosis. Using an in vitro model, we further examined the effect of ginseng on a cyclosporine-treated insulin-secreting cell line. Oxidative stress was measured by the concentration of 8-hydroxy-2'-deoxyguanosine in serum, tissue sections, and culture media. RESULTS: Four weeks of cyclosporine treatment increased blood glucose levels and decreased insulin levels, but cotreatment with ginseng ameliorated the cyclosporine-induced glucose intolerance and hyperglycemia. Pancreatic ß cell area was also greater with ginseng cotreatment compared with cyclosporine monotherapy. The production of proinflammatory molecules, such as induced nitric oxide synthase and cytokines, and the level of apoptotic cell death also decreased in pancreatic ß cell with ginseng treatment. Consistent with the in vivo results, the in vitro study showed that the addition of ginseng protected against cyclosporine-induced cytotoxicity, inflammation, and apoptotic cell death. These in vivo and in vitro changes were accompanied by decreases in the levels of 8-hydroxy-2'-deoxyguanosine in pancreatic ß cell in tissue section, serum, and culture media during cotreatment of ginseng with cyclosporine. CONCLUSIONS: The results of our in vivo and in vitro studies demonstrate that ginseng has a protective effect against cyclosporine-induced pancreatic ß cell injury via reducing oxidative stress.


Asunto(s)
Panax/química , Enfermedades Pancreáticas/tratamiento farmacológico , Enfermedades Pancreáticas/metabolismo , Extractos Vegetales/administración & dosificación , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Ciclosporina/efectos adversos , Modelos Animales de Enfermedad , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/patología , Pruebas de Función Renal , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Nitritos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Páncreas/efectos de los fármacos , Páncreas/patología , Páncreas/fisiopatología , Enfermedades Pancreáticas/inducido químicamente
18.
Transplantation ; 96(2): 146-53, 2013 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-23765110

RESUMEN

BACKGROUND: Cyclosporine A (CsA)-associated oxidative stress has been proposed as an important mechanism of renal injury. This study was designed to examine whether N-acetylcysteine (NAC), a well-known antioxidant, affects Klotho, antiaging gene, expression and its signaling pathway in an experimental model of chronic CsA nephropathy. METHODS: Mice maintained on a low-sodium diet were given vehicle (olive oil, 1 mL/kg/day), CsA (30 mg/kg/day), NAC (150 mg/kg/day), or a combination of CsA and NAC for 4 weeks. The effect of NAC on CsA-induced renal injury was evaluated with basic parameters, histopathology, and markers of oxidative stress [8-hydroxy-2'-deoxyguanosine (8-OHdG) excretion and manganese superoxide dismutase (MnSOD) expression]. The influence of NAC on Klotho and its signal pathway (p-AKT and p-FoxO1) in CsA-treated mouse kidney was evaluated with immunohistochemistry and/or immunoblot. RESULTS: Concomitant administration of CsA and NAC significantly improved renal function and attenuated tubulointerstitial fibrosis, and these changes were accompanied by decreased urinary 8-OHdG level and increased MnSOD expression. NAC treatment preserved Klotho gene expression compared with CsA treatment alone (P < 0.05), and this correlated with urinary 8-OHdG excretion (r = -0.934) and MnSOD expression (r = 0.873, P < 0.001 for both). Concomitant treatment of CsA and NAC translocated FoxO1 from the cytoplasm to the nucleus, implicating dephosphorylation of FoxO1 by NAC in p-AKT/p-FoxO1 pathway. CONCLUSION: NAC treatment preserves Klotho expression and modifies p-AKT/p-FoxO1 pathway in chronic CsA nephropathy.


Asunto(s)
Acetilcisteína/farmacología , Ciclosporina/efectos adversos , Glucuronidasa/genética , Glucuronidasa/metabolismo , Inmunosupresores/efectos adversos , Enfermedades Renales/prevención & control , 8-Hidroxi-2'-Desoxicoguanosina , Acetilcisteína/administración & dosificación , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Enfermedad Crónica , Desoxiguanosina/análogos & derivados , Desoxiguanosina/orina , Modelos Animales de Enfermedad , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Expresión Génica/efectos de los fármacos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Proteínas Klotho , Masculino , Ratones , Ratones Endogámicos ICR , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/metabolismo
19.
PLoS One ; 8(3): e59693, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555748

RESUMEN

The aim of this study was to investigate whether hATMSCs protect against cyclosporine (CsA)-induced renal injury. CsA (7.5 mg/kg) and hATMSCs (3×10(6)/5 mL) were administered alone and together to rats for 4 weeks. The effect of hATMSCs on CsA-induced renal injury was evaluated by assessing renal function, interstitial fibrosis, infiltration of inflammatory cells, and apoptotic cell death. Four weeks of CsA-treatment produced typical chronic CsA-nephropathy. Combined treatment with CsA and hATMSCs did not prevent these effects and showed a trend toward further renal deterioration. To evaluate why hATMSCs aggravated CsA-induced renal injury, we measured oxidative stress, a major mechanism of CsA-induced renal injury. Both urine and serum 8-hydroxydeoxyguanosine(8-OHdG) levels were higher in the CsA+hATMSCs group than in the CsA group (P<0.05). An in vitro study showed similar results. Although the rate of apoptosis did not differ significantly between HK-2 cells cultured in hATMSCs-conditioned medium and those cultured in DMEM, addition of CsA resulted in greater apoptosis in HK-2 cells cultured in hATMSCs-conditioned medium. Addition of CsA increased oxidative stress in the hATMSCs-conditioned medium. The results of our study suggest that treatment with hATMSCs may aggravate CsA-induced renal injury because hATMSCs cause oxidative stress in the presence of CsA.


Asunto(s)
Tejido Adiposo/citología , Ciclosporina/efectos adversos , Riñón/efectos de los fármacos , Riñón/metabolismo , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/citología , Estrés Oxidativo/efectos de los fármacos , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Medios de Cultivo Condicionados , Desoxiguanosina/análogos & derivados , Desoxiguanosina/sangre , Desoxiguanosina/orina , Fibrosis , Humanos , Riñón/patología , Riñón/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
20.
Am J Nephrol ; 37(5): 421-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23594788

RESUMEN

BACKGROUND: This study was performed to investigate whether ginseng extract has a protective effect in an experimental mouse model of chronic cyclosporine (CsA) nephropathy. METHODS: Mice were treated with CsA (30 mg/kg/day, subcutaneously) with or without Korean red ginseng extract (KRG) (0.2, 0.4 g/kg/day, orally) on a 0.01% salt diet for 4 weeks. The effect of KRG on CsA-induced renal injury was evaluated by assessing renal function and pathology, mediators of inflammation, tubulointerstitial fibrosis and apoptotic cell death. Using an in vitro model, we also examined the effect of KRG on CsA-treated proximal tubular cells (HK-2). Oxidative stress was measured by assessing 8-hydroxy-2'-deoxyguanosine (8-OHdG) levels in 24-hour urine, tissue sections, and culture media. RESULTS: Four weeks of CsA treatment caused renal dysfunction, typical pathologic lesions and apoptotic cell death. KRG treatment reduced serum creatinine and blood urea nitrogen and histopathology and increased creatinine clearance. Proinflammatory and profibrotic molecules such as induced nitric oxide synthase, cytokines, transforming growth factor (TGF)-ß1 and TGF-ß1-inducible gene h3 and apoptotic cell death, also decreased with KRG treatment. Consistent with these results, in vitro studies showed that addition of KRG protected against CsA-induced morphological changes, cytotoxicity, inflammation, and apoptotic cell death as demonstrated by annexin V binding. These changes were accompanied by decrease in the level of 8-OHdG in urine and culture supernatant after KRG treatment. CONCLUSION: The results of our in vivo and in vitro studies demonstrate that KRG has a protective effect in CsA-induced renal injury via reducing oxidative stress.


Asunto(s)
Ciclosporina/efectos adversos , Inmunosupresores/efectos adversos , Enfermedades Renales/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Panax , Extractos Vegetales/uso terapéutico , Animales , Biomarcadores/metabolismo , Línea Celular , Interacciones de Hierba-Droga , Humanos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Masculino , Ratones , Fitoterapia , Extractos Vegetales/farmacología , Distribución Aleatoria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...