Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Aging Cell ; : e14184, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38687090

RESUMEN

Cellular senescence contributes to inflammatory kidney disease via the secretion of inflammatory and profibrotic factors. Protease-activating receptor 2 (PAR2) is a key regulator of inflammation in kidney diseases. However, the relationship between PAR2 and cellular senescence in kidney disease has not yet been described. In this study, we found that PAR2-mediated metabolic changes in renal tubular epithelial cells induced cellular senescence and increased inflammatory responses. Using an aging and renal injury model, PAR2 expression was shown to be associated with cellular senescence. Under in vitro conditions in NRK52E cells, PAR2 activation induces tubular epithelial cell senescence and senescent cells showed defective fatty acid oxidation (FAO). Cpt1α inhibition showed similar senescent phenotype in the cells, implicating the important role of defective FAO in senescence. Finally, we subjected mice lacking PAR2 to aging and renal injury. PAR2-deficient kidneys are protected from adenine- and cisplatin-induced renal fibrosis and injury, respectively, by reducing senescence and inflammation. Moreover, kidneys lacking PAR2 exhibited reduced numbers of senescent cells and inflammation during aging. These findings offer fresh insights into the mechanisms underlying renal senescence and indicate that targeting PAR2 or FAO may be a promising therapeutic approach for managing kidney injury.

2.
Antioxidants (Basel) ; 12(11)2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-38001800

RESUMEN

Chronic kidney disease (CKD) is a kidney structure and function abnormality. CKD development and progression are strongly influenced by oxidative stress and inflammatory responses, which can lead to tubulointerstitial fibrosis. Unfortunately, there are no effective or specific treatments for CKD. We investigated the potential of the thiobarbiturate-derived compound MHY1025 to alleviate CKD by reducing oxidative stress and inflammatory responses. In vitro experiments using NRK52E renal tubular epithelial cells revealed that MHY1025 significantly reduced LPS-induced oxidative stress and inhibited the activation of the NF-κB pathway, which is involved in inflammatory responses. Furthermore, treatment with MHY1025 significantly suppressed the expression of fibrosis-related genes and proteins induced by TGFß in NRK49F fibroblasts. Furthermore, we analyzed the MHY1025 effects in vivo. To induce kidney fibrosis, mice were administered 250 mg/kg folic acid (FA) and orally treated with MHY1025 (0.5 mg/kg/day) for one week. MHY1025 effectively decreased the FA-induced inflammatory response in the kidneys. The group treated with MHY1025 exhibited a significant reduction in cytokine and chemokine expression and decreased immune cell marker expression. Decreased inflammatory response was associated with decreased tubulointerstitial fibrosis. Overall, MHY1025 alleviated renal fibrosis by directly modulating renal epithelial inflammation and fibroblast activation, suggesting that MHY1025 has the potential to be a therapeutic agent for CKD.

3.
Cell Commun Signal ; 21(1): 215, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596656

RESUMEN

BACKGROUND: Toll-like receptor 7 (TLR7) is an endosomal TLR activated by single-stranded RNA, including endogenous microRNAs. Although TLR7 is known to promote inflammatory responses in pathophysiological conditions, its role in renal fibrosis has not been investigated. Here, we aim to investigate the inflammatory roles of TLR7 in kidney inflammation and fibrosis. METHODS: TLR7 knockout mice (Tlr7 -/-) subjected to AD-induced kidney injury were utilized to examine the role of TLR7 in kidney fibrosis. To elucidate the role of TLR7 in renal epithelial cells, NRK52E rat renal tubule epithelial cells were employed. RESULTS: Under fibrotic conditions induced by an adenine diet (AD), TLR7 was significantly increased in damaged tubule epithelial cells, where macrophages were highly infiltrated. TLR7 deficiency protected against AD-induced tubular damage, inflammation, and renal fibrosis. Under in vitro conditions, TLR7 activation increased NF-κB activity and induced chemokine expression, whereas TLR7 inhibition effectively blocked NF-κB activation. Furthermore, among the known TLR7 endogenous ligands, miR-21 was significantly upregulated in the tubular epithelial regions. In NRK52E cells, miR-21 treatment induced pro-inflammatory responses, which could be blocked by a TLR7 inhibitor. When the TLR7 inhibitor, M5049, was administered to the AD-induced renal fibrosis model, TLR7 inhibition significantly attenuated AD-induced renal inflammation and fibrosis. CONCLUSIONS: Overall, activation of TLR7 by endogenous miR-21 in renal epithelial cells contributes to inflammatory responses in a renal fibrosis model, suggesting a possible therapeutic target for the treatment of renal fibrosis. Video Abstract.


Asunto(s)
Enfermedades Renales , MicroARNs , Receptor Toll-Like 7 , Animales , Ratones , Ratas , Adenina , Células Epiteliales , Inflamación , MicroARNs/genética , FN-kappa B , Transducción de Señal , Enfermedades Renales/genética , Enfermedades Renales/patología , Fibrosis
4.
Int J Mol Sci ; 24(5)2023 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-36902313

RESUMEN

The peroxisome proliferator-activated receptor (PPAR) nuclear receptor has been an interesting target for the treatment of chronic diseases. Although the efficacy of PPAR pan agonists in several metabolic diseases has been well studied, the effect of PPAR pan agonists on kidney fibrosis development has not been demonstrated. To evaluate the effect of the PPAR pan agonist MHY2013, a folic acid (FA)-induced in vivo kidney fibrosis model was used. MHY2013 treatment significantly controlled decline in kidney function, tubule dilation, and FA-induced kidney damage. The extent of fibrosis determined using biochemical and histological methods showed that MHY2013 effectively blocked the development of fibrosis. Pro-inflammatory responses, including cytokine and chemokine expression, inflammatory cell infiltration, and NF-κB activation, were all reduced with MHY2013 treatment. To demonstrate the anti-fibrotic and anti-inflammatory mechanisms of MHY2013, in vitro studies were conducted using NRK49F kidney fibroblasts and NRK52E kidney epithelial cells. In the NRK49F kidney fibroblasts, MHY2013 treatment significantly reduced TGF-ß-induced fibroblast activation. The gene and protein expressions of collagen I and α-smooth muscle actin were significantly reduced with MHY2013 treatment. Using PPAR transfection, we found that PPARγ played a major role in blocking fibroblast activation. In addition, MHY2013 significantly reduced LPS-induced NF-κB activation and chemokine expression mainly through PPARß activation. Taken together, our results suggest that administration of the PPAR pan agonist effectively prevented renal fibrosis in both in vitro and in vivo models of kidney fibrosis, implicating the therapeutic potential of PPAR agonists against chronic kidney diseases.


Asunto(s)
Enfermedades Renales , FN-kappa B , Ratones , Animales , FN-kappa B/metabolismo , Enfermedades Renales/metabolismo , Inflamación/metabolismo , Modelos Animales de Enfermedad , PPAR gamma/metabolismo , Quimiocinas/metabolismo , Fibrosis , Fibroblastos/metabolismo
5.
J Gerontol A Biol Sci Med Sci ; 78(1): 60-68, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35867996

RESUMEN

The age-associated functional decline of the kidney is accompanied by structural changes including glomerular sclerosis and interstitial fibrosis. Aging kidneys also exhibit increased vulnerability in stressful environmental conditions. In this study, we assessed the differences in responses between young and aged animals to folic acid (FA)-induced renal fibrosis. To monitor the effects of aging on FA-induced kidney fibrosis, we administered FA (250 mg/kg) to young (6-month old) and aged (20-month old) rats. The development of severe fibrosis was only detected in aged rat kidneys, which was accompanied by increased kidney injury and inflammation. Furthermore, we found that FA-treated aged rats had significantly lower farnesoid X receptor (FXR) expression in the tubular epithelial cells than the rats not treated with FA. Interestingly, the extent of inflammation was severe in the kidneys of aged rat, where the FXR expression was low. To explore the role of FXR in kidney inflammation, in vitro studies were performed using NRK52E kidney tubule epithelial cells. NF-κB activation by lipopolysaccharide treatment induces chemokine production in NRK52E cells. The activation of FXR by obeticholic acid significantly reduced the transcriptional activity of NF-κB and chemokine production. In contrast, FXR knockdown increased LPS-induced chemokine production in NRK52E cells. Finally, FXR-knockout mice that were administered FA showed increased inflammation and severe fibrosis. In summary, we demonstrated that diminished FXR expression in the epithelial cells of the renal tubules exacerbated the fibrotic response in aged rat kidneys by upregulating pro-inflammatory NF-κB activation.


Asunto(s)
Enfermedades Renales , FN-kappa B , Ratones , Ratas , Animales , FN-kappa B/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/farmacología , Riñón/patología , Fibrosis , Inflamación/metabolismo , Enfermedades Renales/patología , Quimiocinas/metabolismo
6.
Biochim Biophys Acta Mol Basis Dis ; 1868(10): 166474, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35772632

RESUMEN

A high-fat diet (HFD) is a major risk factor for chronic kidney disease. Although HFD promotes renal injury, characterized by increased inflammation and oxidative stress leading to fibrosis, the underlying mechanism remains elusive. Here, we investigated the role and mechanism of protease-activating receptor 2 (PAR2) activation during HFD-induced renal injury in C57/BL6 mice. HFD for 16 weeks resulted in kidney injury, manifested by increased blood levels of blood urea nitrogen, increased levels of oxidative stress with inflammation, and structural changes in the kidney tubules. HFD-fed kidneys showed elevated PAR2 expression level in the tubular epithelial region. To elucidate the role of PAR2, PAR2 knockout mice and their littermates were administered HFD. PAR2 deficient kidneys showed reduced extent of renal injury. PAR2 deficient kidneys showed significantly decreased levels of inflammatory gene expression and macrophage infiltration, followed by reduced accumulation of extracellular matrix proteins. Using NRK52E kidney epithelial cells, we further elucidated the mechanism and role of PAR2 activation during renal injury. Palmitate treatment increased PAR2 expression level in NRK52E cells and scavenging of oxidative stress blocked PAR2 expression. Under palmitate-treated conditions, PAR2 agonist-induced NF-κB activation level was higher with increased chemokine expression level in the cells. These changes were attenuated by the depletion of oxidative stress. Taken together, our results suggest that HFD-induced PAR2 activation is associated with increased levels of renal oxidative stress, inflammatory response, and fibrosis.


Asunto(s)
Dieta Alta en Grasa , Riñón , Receptor PAR-2 , Animales , Dieta Alta en Grasa/efectos adversos , Fibrosis , Inflamación/metabolismo , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Palmitatos , Receptor PAR-2/genética
7.
Arch Pharm Res ; 45(3): 159-173, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35334088

RESUMEN

Renal fibrosis is defined by excessive extracellular matrix (ECM) accumulation and is associated with a decreased kidney function. Increased inflammation and infiltration of inflammatory cells are the key features of renal fibrosis development; however, the mechanism of how inflammation starts is still un-known. Here, we show that the activation of epithelial Protease-activating receptor 2 (PAR2) signaling plays an important role in the initiation of inflammation via increased chemokine expression and inflammatory cell induction. In the adenine diet-induced renal fibrosis mouse model, PAR2 expression was significantly increased in the renal tubule region. Kidneys from PAR2-knockout mice were protected from adenine diet-induced renal fibrosis, kidney dysfunction, and inflammation. Using NRK52E kidney epithelial cells, we further elucidated the mechanisms underlying these processes. Activation of PAR2 signaling pathway by PAR2 agonist specifically increased the levels of chemokines, including MCP1 and MCP3, via the MAPK-NF-κB signaling pathway. Inhibition of the MAPK signaling pathway attenuated PAR2 agonist-induced NF-κB activation, chemokine expression, and macrophage cell induction. Furthermore, PAR2 activation directly increased mesenchymal cell markers in epithelial cells. Taken together, we found that increased PAR2 expression and the PAR2/MAPK signaling pathway promote renal fibrosis by increasing the inflammatory responses and promoting EMT process.


Asunto(s)
Enfermedades Renales , Péptido Hidrolasas , Receptor PAR-2 , Animales , Fibrosis , Riñón/patología , Enfermedades Renales/metabolismo , Ratones , Péptido Hidrolasas/metabolismo , Receptor PAR-2/metabolismo , Transducción de Señal
8.
Aging Dis ; 12(7): 1713-1728, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34631216

RESUMEN

FoxOs and their post-translational modification by phosphorylation, acetylation, and methylation can affect epigenetic modifications and promote the expression of downstream target genes. Therefore, they ultimately affect cellular and biological functions during aging or occurrence of age-related diseases including cancer, diabetes, and kidney diseases. As known for its key role in aging, FoxOs play various biological roles in the aging process by regulating reactive oxygen species, lipid accumulation, and inflammation. FoxOs regulated by PI3K/Akt pathway modulate the expression of various target genes encoding MnSOD, catalases, PPARγ, and IL-1ß during aging, which are associated with age-related diseases. This review highlights the age-dependent differential regulatory mechanism of Akt/FoxOs axis in metabolic and non-metabolic organs. We demonstrated that age-dependent suppression of Akt increases the activity of FoxOs (Akt/FoxOs axis upregulation) in metabolic organs such as liver and muscle. This Akt/FoxOs axis could be modulated and reversed by antiaging paradigm calorie restriction (CR). In contrast, hyperinsulinemia-mediated PI3K/Akt activation inhibited FoxOs activity (Akt/FoxOs axis downregulation) leading to decrease of antioxidant genes expression in non-metabolic organs such as kidneys and lungs during aging. These phenomena are reversed by CR. The results of studies on the process of aging and CR indicate that the Akt/FoxOs axis plays a critical role in regulating metabolic homeostasis, redox stress, and inflammation in various organs during aging process. The benefical actions of CR on the Akt/FoxOs axis in metabolic and non-metabolic organs provide further insights into the molecular mechanisms of organ-differential roles of Akt/FoxOs axis during aging.

9.
Toxicology ; 463: 152973, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34619300

RESUMEN

Chronic kidney disease (CKD) is characterized by persistent abnormalities in kidney function, accompanied by structural changes. Interstitial fibrosis, characterized by the accumulation of extracellular matrix (ECM) proteins, is frequently detected during CKD development. Given the multiple underlying causes of CKD, numerous animal models have been developed to advance our understanding of human nephropathy. Herein, we compared two reliable toxin-induced mouse kidney fibrosis models in terms of fibrosis and inflammation. Administration of folic acid (250 mg/kg, intraperitoneal injection) or an adenine diet (0.25 % for three weeks) afforded similar effects on kidney function, as detected by increased serum nitrogen levels. In addition, the kidneys exhibited a similar extent of tubule dilation and kidney damage. The degree of fibrosis was compared using various biological methods. Although both models developed a significant fibrotic phenotype, the adenine diet-fed model showed a marginally higher increase in fibrosis than the folic acid model, as reflected by increased kidney ECM gene and protein levels. We further compared inflammatory responses in the kidneys. Interestingly, pro-inflammatory responses, including cytokine expression and immune cell infiltration, were significantly increased in adenine diet-fed kidneys. Furthermore, collagen expression was identified in the macrophage-infiltrated region, implying the importance of inflammation in fibrogenesis. Collectively, we observed that the adenine diet-fed kidney fibrosis model presented a higher inflammatory response with increased fibrosis when compared with the folic acid-induced kidney fibrosis model, indicating the importance of the inflammatory response in fibrosis development.


Asunto(s)
Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/patología , Insuficiencia Renal Crónica/fisiopatología , Adenina/toxicidad , Animales , Fibrosis , Ácido Fólico/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL
10.
Oxid Med Cell Longev ; 2021: 6683033, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34211632

RESUMEN

Protease-activated receptor 2 (PAR2) is a member of G-protein-coupled receptors and affects ligand-modulated calcium signaling. Although PAR2 signaling promotes obesity and adipose tissue inflammation in high fat- (HF-) fed conditions, its role in adipocyte differentiation under nonobesogenic conditions needs to be elucidated. Here, we used several tissues and primary-cultured adipocytes of mice lacking PAR2 to study its role in the development of adipose tissues. C57BL/6J mice with PAR2 deficiency exhibited a mild lipodystrophy-like phenotype in a chow diet-fed condition. When adipocyte differentiation was examined using primary-cultured preadipocytes, PAR2 deficiency led to a notable decrease in adipocyte differentiation and related protein expression, and PAR2 agonist treatment elevated adipocyte differentiation. Regarding the mechanism, PAR2-deficient preadipocytes exhibited impaired mitochondrial energy consumption. Further studies indicated that calcium-related signaling pathways for mitochondrial biogenesis are disrupted in the adipose tissues of PAR2-deficient mice and PAR2-deficient preadipocytes. Also, a PAR2 antagonist elevated mitochondrial reactive oxygen species and reduced the MitoTracker fluorescent signal in preadipocytes. Our studies revealed that PAR2 is important for the development of adipose tissue under basal conditions through the regulation of mitochondrial biogenesis and adipocyte differentiation.


Asunto(s)
Adipocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor PAR-2/deficiencia , Animales , Diferenciación Celular , Masculino , Ratones , Mitocondrias
11.
J Nutr Biochem ; 95: 108769, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34000413

RESUMEN

Protease-activated receptor 2 (PAR2) is a member of G protein-coupled receptors. There are two types of PAR2 signaling pathways: Canonical G-protein signaling and ß-arrestin signaling. Although PAR2 signaling has been reported to aggravate hepatic steatosis, the exact mechanism is still unclear, and the role of PAR2 in autophagy remains unknown. In this study, we investigated the regulatory role of PAR2 in autophagy during high-fat diet (HFD)-induced hepatic steatosis in mice. Increased protein levels of PAR2 and ß-arrestin-2 and their interactions were detected after four months of HFD. To further investigate the role of PAR2, male and female wild-type (WT) and PAR2-knockout (PAR2 KO) mice were fed HFD. PAR2 deficiency protected HFD-induced hepatic steatosis in male mice, but not in female mice. Interestingly, PAR2-deficient liver showed increased AMP-activated protein kinase (AMPK) activation with decreased interaction between Ca2+/calmodulin-dependent protein kinase kinase ß (CAMKKß) and ß-arrestin-2. In addition, PAR2 deficiency up-regulated autophagy in the liver. To elucidate whether PAR2 plays a role in the regulation of autophagy and lipid accumulation in vitro, PAR2 was overexpressed in HepG2 cells. Overexpression of PAR2 decreased AMPK activation with increased interaction of CAMKKß with ß-arrestin-2 and significantly inhibited autophagic responses in HepG2 cells. Inhibition of autophagy by PAR2 overexpression further exacerbated palmitate-induced lipid accumulation in HepG2 cells. Collectively, these findings suggest that the increase in the PAR2-ß-arrestin-2-CAMKKß complex by HFD inhibits AMPK-mediated autophagy, leading to the alleviation of hepatic steatosis.


Asunto(s)
Adenilato Quinasa/metabolismo , Autofagia/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Enfermedad del Hígado Graso no Alcohólico/inducido químicamente , Receptor PAR-2/metabolismo , Adenilato Quinasa/genética , Animales , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Receptor PAR-2/genética , Regulación hacia Arriba , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
12.
Endocrinol Metab (Seoul) ; 36(1): 171-184, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33677938

RESUMEN

BACKGROUND: Protease-activated protein-2 (PAR2) has been reported to regulate hepatic insulin resistance condition in type 2 diabetes mice. However, the mechanism of lipid metabolism through PAR2 in obesity mice have not yet been examined. In liver, Forkhead box O1 (FoxO1) activity induces peroxisome proliferator-activated receptor γ (PPARγ), leading to accumulation of lipids and hyperlipidemia. Hyperlipidemia significantly influence hepatic steatoses, but the mechanisms underlying PAR2 signaling are complex and have not yet been elucidated. METHODS: To examine the modulatory action of FoxO1 and its altered interaction with PPARγ, we utilized db/db mice and PAR2-knockout (KO) mice administered with high-fat diet (HFD). RESULTS: Here, we demonstrated that PAR2 was overexpressed and regulated downstream gene expressions in db/db but not in db+ mice. The interaction between PAR2/ß-arrestin and Akt was also greater in db/db mice. The Akt inhibition increased FoxO1 activity and subsequently PPARγ gene in the livers that led to hepatic lipid accumulation. Our data showed that FoxO1 was negatively controlled by Akt signaling and consequently, the activity of a major lipogenesis-associated transcription factors such as PPARγ increased, leading to hepatic lipid accumulation through the PAR2 pathway under hyperglycemic conditions in mice. Furthermore, the association between PPARγ and FoxO1 was increased in hepatic steatosis condition in db/db mice. However, HFD-fed PAR2-KO mice showed suppressed FoxO1-induced hepatic lipid accumulation compared with HFD-fed control groups. CONCLUSION: Collectively, our results provide evidence that the interaction of FoxO1 with PPARγ promotes hepatic steatosis in mice. This might be due to defects in PAR2/ß-arrestin-mediated Akt signaling in diabetic and HFD-fed mice.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hígado Graso , Animales , Lípidos , Lipogénesis , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Exp Gerontol ; 142: 111108, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33130113

RESUMEN

Age- or high fat diet (HFD)-associated renal structural changes are commonly associated with a decline in renal function. Although HFD causes injurious effects in various organs during aging, its effects on age-associated renal fibrosis have not yet been investigated. In this study, we show that a short-term HFD significantly induces renal fibrosis by causing loss of mitochondrial integrity in aged Sprague-Dawley (SD) rats. To evaluate the effects of short-term HFD intake on age-associated renal fibrosis, we administered HFD in young and aged SD rats for 15 days. Our results showed that a short-term HFD significantly increased the renal fibrosis and inflammation in aged rats. Moreover, mitochondrial integrity and the expression of fatty acid oxidation-related proteins decreased in the kidneys of the HFD-fed aged rats. Further, NRK52E renal tubular epithelial cells subjected to lipid stress by treatment with oleic acid showed a reduced amount of mitochondrial OXPHOS-related proteins. Our results suggest that short-term HFD affects mitochondrial integrity and exacerbates inflammation leading to renal fibrosis, especially in aged rats. We conclude that the mitochondrial integrity in kidney tissues is important in HFD-induced renal fibrosis development during aging.


Asunto(s)
Dieta Alta en Grasa , Enfermedades Renales , Animales , Dieta Alta en Grasa/efectos adversos , Fibrosis , Riñón/patología , Enfermedades Renales/etiología , Enfermedades Renales/patología , Ratas , Ratas Sprague-Dawley
14.
Appl Immunohistochem Mol Morphol ; 28(8): 579-592, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32902936

RESUMEN

Chronic inflammation is a major risk factor underlying aging and age-associated diseases. It impairs normal lipid accumulation, adipose tissue function, and mitochondrial function, which eventually lead to insulin resistance. Peroxisome proliferator-activated receptors (PPARs) critically regulate gluconeogenesis, lipid metabolism, and the lipid absorption and breakdown process, and PPAR activity decreases in the liver during aging. In the present study, we investigated the ability of 2-(4-(5,6-methylenedioxybenzo[d]thiazol-2-yl)-2-methylphenoxy)-2-methylpropanoic acid (MHY2013), synthesized PPARα/PPARß/PPARγ pan agonist, to suppress the inflammatory response and attenuate insulin resistance in aged rat liver. Six- and 20-month-old rats were divided into 4 groups: young and old rats fed ad libitum; and old rats fed ad libitum supplemented with MHY2013 (1 mg and 5 mg/kg/d for 4 wk). We found that MHY2013 supplementation efficiently downregulated the activity of nuclear factor-κB through JNK/ERK/p38 mitogen-activated protein kinase signaling in the liver of aged rats. In addition, MHY2013 treatment increased hepatic insulin signaling, and the downstream signaling activity of FOXO1, which is negatively regulated by Akt. Downregulation of Akt increases expression of FOXO1, which acts as a transcription factor and increases transcription of interleukin-1ß, leading to hepatic inflammation. The major finding of this study is that MHY2013 acts as a therapeutic agent against age-related inflammation associated with insulin resistance by activating PPARα, PPARß, and PPARγ. Thus, the study provides evidence for the anti-inflammatory properties of MHY2013, and the role it plays in the regulation of age-related alterations in signal transduction pathways.


Asunto(s)
Resistencia a la Insulina , Interleucina-1beta/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Activados del Proliferador del Peroxisoma/agonistas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Envejecimiento/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Quinasas Janus/metabolismo , Hígado/metabolismo , Hígado/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , PPAR alfa/agonistas , PPAR gamma/agonistas , PPAR-beta/agonistas , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
Exp Gerontol ; 134: 110891, 2020 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-32114077

RESUMEN

Chronic inflammation is a complex and unresolved inflammatory response with low-grade multivariable patterns that aggravate systemic pathophysiological conditions and the aging process. To redefine and delineate these age-related complex inflammatory phenomena at the molecular, cellular, and systemic levels, the concept of "Senoinflammation" was recently formulated. In this review, we describe the accumulated data on both the multiphase systemic inflammatory process and the cellular proinflammatory signaling pathway. We also describe the proinflammatory mechanisms underlying the metabolic molecular pathways in aging. Additionally, we review age-related lipid accumulation, the role of the inflammatory senescence-associated secretory phenotype (SASP), the involvement of cytokine/chemokine secretion, endoplasmic reticulum (ER) stress, insulin resistance, and autophagy. The last section of the review highlights the modulation of the senoinflammatory process by the anti-aging and anti-inflammatory action of calorie restriction (CR). Evidence from aging and CR research strongly suggests that SASP from senescent cells may be the major source of secreted cytokines and chemokines during aging. A better understanding of the mechanisms underpinning the senoinflammatory response and the mitigating role of CR will provide insights into the molecular mechanisms of chronic inflammation and aging for potential interventions.

16.
Brain Res ; 1730: 146642, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31930999

RESUMEN

Neuroinflammation is often associated with astrocyte and microglial activations particularly in Parkinson's disease (PD) and other brain damage such as Alzheimer's disease. Therefore, the modulation of glial activation offers a possible target for treating PD-associated pathologies. Here, we evaluated the neuroprotective effects of usnic acid, a naturally occurring dibenzofuran derivative found in several lichen species in an acute mouse model of PD. Male mice were administered with vehicle or usnic acid (5 or 25 mg/kg) for 10 consecutive days, and then on day 11, MPTP (20 mg/kg, i.p.) was administered four times (with 2hrs intervals between injections) to induce PD pathologies. It was found that MPTP-induced motor dysfunction and neuronal loss were ameliorated in the usnic acid-treated mice versus vehicle-treated controls. Further study revealed that usnic acid effectively inhibited MPP+-induced glial activation in primary astrocytes by blocking NF-κB activation. Taken together, these findings suggest that usnic acid could be considered potentially useful therapeutic candidates for PD and other neurodegenerative diseases associated with neuroinflammation.


Asunto(s)
Antiinflamatorios/administración & dosificación , Benzofuranos/administración & dosificación , Encéfalo/efectos de los fármacos , Encefalitis/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Trastornos Parkinsonianos/complicaciones , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Encefalitis/etiología , Masculino , Ratones Endogámicos C57BL , Neuroglía/efectos de los fármacos , Neuroglía/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Trastornos Parkinsonianos/inducido químicamente , Prueba de Desempeño de Rotación con Aceleración Constante
17.
J Gerontol A Biol Sci Med Sci ; 75(3): 452-458, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31112599

RESUMEN

Age-associated renal fibrosis is commonly observed, with a decline in renal function during aging. Although peroxisome proliferator-activated receptors α/ß (PPARα/ß) activation has been shown to exert beneficial effects on age-associated renal changes, its effects on age-associated renal fibrosis have not been investigated yet. Here, we show that the PPARα/ß activator, MHY2013, can significantly alter lipid metabolism in renal tubule epithelial cells and attenuate renal fibrosis in aged Sprague Dawley (SD) rats. We found that MHY2013 significantly increased nuclear translocation and activity of PPARα/ß in NRK52E renal epithelial cells. Moreover, the enhanced PPARα/ß activity increased the expression of fatty acid oxidation-associated PPARα/ß target genes. In addition, transforming growth factor-ß (TGF-ß)- and oleic acid-induced lipid accumulation and fibrosis-associated gene expression were decreased in NRK52E cells by MHY2013 pretreatment. To evaluate the effects of MHY2013 on age-associated renal fibrosis, aged SD rates were orally administered MHY2013 (1 and 5 mg/kg) daily for 1 month. MHY2013 efficiently increased PPARα/ß activation and reduced renal lipid accumulation in aged SD rat kidneys. Furthermore, renal fibrosis was significantly decreased by MHY2013, indicating the importance of renal lipid metabolism in age-associated renal fibrosis. Taken together, our results suggest that activation of PPARα/ß signaling during aging prevents age-associated renal fibrosis.


Asunto(s)
Enfermedades Renales/patología , Enfermedades Renales/prevención & control , Riñón/patología , PPAR alfa/agonistas , PPAR-beta/agonistas , Factores de Edad , Animales , Fibrosis/prevención & control , Masculino , PPAR alfa/efectos de los fármacos , PPAR-beta/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
18.
Aging (Albany NY) ; 11(12): 4125-4144, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31246177

RESUMEN

Decreased forkhead box O1 (FoxO1) activity induces hyperlipidemia and increased PPARγ, leading to hyperlipidemia in association with endoplasmic reticulum (ER) stress. In the liver, aging and comorbidities such as hyperlipidemia and diabetes significantly influence a wide variety of steatosis, but the underlying mechanisms are complex and remain elusive.To establish the modulatory role of FoxO1 and the functional consequences of its altered interaction with PPARγ in the present study, we utilized a cell culture system, aged rats and diabetic db/db mice.We found that, under ER stress, FoxO1 induces PPARγ-mediated lipid accumulation in aged rat livers. Our data showed that the FoxO1-induced hepatic lipid accumulation was negatively regulated by Akt signaling. PPARγ, a key lipogenesis transcription factor, was increased in aged liver, resulting in lipid accumulation via hepatic ER stress under hyperglycemic conditions. We further demonstrated that loss of FoxO1 causes a decline in PPARγ expression and reduces lipid accumulation. In addition, the interaction between FoxO1 and PPARγ was shown to induce hepatic steatosis in aging and db/db mice.We provide evidence that, in aged rats, FoxO1 interaction with PPARγ promotes hepatic steatosis, due to hyperglycemia-induced ER stress, which causes an impairment in Akt signaling, such in aging-related diabetes.


Asunto(s)
Envejecimiento , Estrés del Retículo Endoplásmico , Hígado Graso , Proteínas del Tejido Nervioso/metabolismo , PPAR gamma/metabolismo , Animales , Línea Celular Tumoral , Glucosa/metabolismo , Metabolismo de los Lípidos , Masculino , Proteínas del Tejido Nervioso/genética , PPAR gamma/genética , Ratas , Ratas Sprague-Dawley
19.
Food Chem Toxicol ; 129: 239-248, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31029726

RESUMEN

Dibutyl phthalate (DBP) is commonly used plasticizer and a known endocrine disruptor that can cause birth defects and developmental disorders. Although several studies have reported that DBP has neurotoxic effects on neurite outgrowth and on learning and memory, its neurotoxic effects on neural progenitor cells (NPCs) have not been investigated. The present study was undertaken to determine the effects of DBP on NPCs and hippocampal neurogenesis. At high concentration DBP (500 µM) retarded NPC proliferation without affecting cell viability by arresting the cell cycle at G1 but did not cause cell death. DNA damage was observed by examining p53 expression and by γH2AX staining. DBP-treated cells had elevated ROS levels and exhibited mitochondrial dysfunction, which can cause DNA damage. Adult hippocampal neurogenesis was investigated using BrdU immunohistochemistry in young C57BL/6 mice intraperitoneally administrated with vehicle or DBP (10 or 50 mg/kg) for 2 weeks. DBP administered mice were found to have significantly fewer newly generated neurons in hippocampi, and the Morris water maze test revealed that DBP (50 mg/kg) impaired spatial learning and memory. Taken together, these findings suggest that DBP has harmful effects on NPCs and hippocampal neurogenesis and that DBP exposure could lead to learning and memory dysfunctions.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Dibutil Ftalato/toxicidad , Hipocampo/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Plastificantes/toxicidad , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Hipocampo/citología , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Células-Madre Neurales/citología , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
20.
Aging (Albany NY) ; 11(4): 1283-1304, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30811347

RESUMEN

ß-Hydroxybutyrate (HB) is a ketone body used as an energy source that has shown anti-inflammatory effects similar to calorie restriction (CR); Here, PGC-1α, an abundantly expressed co-factor in the kidney, was reported to interact with both FoxO1 and NF-κB although the definitive interactive mechanism has not yet been reported. In this study, we investigated whether renal aging-related inflammation is modulated by HB. We compared aged rats administered with HB to calorie restricted rats and examined the modulation of FoxO1 and the NF-κB pathway through interactions with PGC-1α. We found that in aged rats treated with HB, pro-inflammatory signaling changes were reversed and showed effects comparable to CR. As FoxO1 and its target genes catalase/MnSOD were upregulated by HB treatment and PGC-1α selectively interacted with FoxO1, not with NF-κB, and ameliorated the renal inflammatory response. These findings were further confirmed using FoxO1 overexpression and siRNA transfection in vitro. Our findings suggest that HB suppressed aging-related inflammation as a CR mimetic by enabling the co-activation and selective interaction between FoxO1 and PGC-1α. This study demonstrates the potential therapeutic role of HB as a CR mimetic, which ameliorates inflammation by a novel mechanism where FoxO1 outcompetes NF-κB by interacting with PGC-1α in aging kidneys.


Asunto(s)
Ácido 3-Hidroxibutírico/farmacología , Envejecimiento , Restricción Calórica , Inflamación/tratamiento farmacológico , Proteínas del Tejido Nervioso/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Animales , Antiinflamatorios/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Proteínas del Tejido Nervioso/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Ratas , Ratas Sprague-Dawley , Organismos Libres de Patógenos Específicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...