Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Int J Mol Sci ; 25(6)2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38542262

RESUMEN

Graves' ophthalmopathy (GO), or thyroid eye disease (TED), is the most frequent extrathyroidal manifestation of Graves' disease (GD). Inflammation and subsequent aberrant tissue remodeling with fibrosis are important pathogenesis. There are many proposed mechanisms and molecular pathways contributing to tissue remodeling and fibrosis in GO, including adipogenesis, fibroblast proliferation and myofibroblasts differentiation, oxidative stress, endoplasmic reticulum (ER) stress, hyaluronan (HA) and glycosaminoglycans (GAGs) accumulation in the extracellular matrix (ECM) and new concepts of epigenetics modification, such as histone modification, DNA methylation, non-coding RNAs, and gut microbiome. This review summarizes the current understanding of ECM proteins and associated tissue remodeling in the pathogenesis and potential mediators for the treatment of GO.


Asunto(s)
Enfermedad de Graves , Oftalmopatía de Graves , Humanos , Oftalmopatía de Graves/genética , Oftalmopatía de Graves/metabolismo , Órbita/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Enfermedad de Graves/metabolismo , Fibrosis
2.
Biomedicines ; 9(12)2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34944687

RESUMEN

Graves' ophthalmopathy (GO) is the most common extrathyroidal manifestation of Graves' disease. It is characterized initially by an inflammatory process, followed by tissue remodeling and fibrosis, leading to proptosis, exposure keratopathy, ocular motility limitation, and compressive optic neuropathy. The pathogenic mechanism is complex and multifactorial. Accumulating evidence suggests the involvement of oxidative stress in the pathogenesis of GO. Cigarette smoking, a major risk factor for GO, has been shown to induce reactive oxygen species (ROS) generation and oxidative damage in GO orbital fibroblasts. In addition, an elevation in ROS and antioxidant enzymes is observed in tears, blood, and urine, as well as orbital fibroadipose tissues and fibroblasts from GO patients. In vitro and in vivo studies have examined the efficacy of various antioxidant supplements for GO. These findings suggest a therapeutic role of antioxidants in GO patients. This review summarizes the current understanding of oxidative stress in the pathogenesis and potential antioxidants for the treatment of GO.

3.
Biomolecules ; 11(10)2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34680057

RESUMEN

Pirfenidone is a pyridinone derivative that has been shown to inhibit fibrosis in animal models and in patients with idiopathic pulmonary fibrosis. Its effect on orbital fibroblasts remains poorly understood. We investigated the in vitro effect of pirfenidone in transforming growth factor-ß1 (TGF-ß1)-induced myofibroblast transdifferentiation and extracellular matrix (ECM) homeostasis in primary cultured orbital fibroblasts from patients with Graves' ophthalmopathy (GO). The expression of fibrotic proteins, including α-smooth muscle actin (α-SMA), connective tissue growth factor (CTGF), fibronectin, and collagen type I, was determined by Western blots. The activities of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) responsible for the ECM homeostasis were examined. After pretreating the GO orbital fibroblasts with pirfenidone (250, 500, and 750 µg/mL, respectively) for one hour followed by TGF-ß1 for another 24 h, the expression of α-SMA, CTGF, fibronectin, and collagen type I decreased in a dose-dependent manner. Pretreating the GO orbital fibroblasts with pirfenidone not only abolished TGF-ß1-induced TIMP-1 expression but recovered the MMP-2/-9 activities. Notably, pirfenidone inhibited TGF-ß1-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK), the critical mediators in the TGF-ß1 pathways. These findings suggest that pirfenidone modulates TGF-ß1-mediated myofibroblast differentiation and ECM homeostasis by attenuating downstream signaling of TGF-ß1.


Asunto(s)
Oftalmopatía de Graves/genética , Miofibroblastos/efectos de los fármacos , Piridonas/farmacología , Factor de Crecimiento Transformador beta1/farmacología , Actinas/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Colágeno Tipo I/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Matriz Extracelular/genética , Fibroblastos/efectos de los fármacos , Fibronectinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Oftalmopatía de Graves/patología , Homeostasis/genética , Humanos , Metaloproteinasas de la Matriz/genética , Miofibroblastos/citología , Cultivo Primario de Células , Inhibidores Tisulares de Metaloproteinasas/genética , Factor de Crecimiento Transformador beta1/genética
4.
Int J Mol Sci ; 22(6)2021 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-33799469

RESUMEN

Transforming growth factor-ß1 (TGF-ß1)-induced myofibroblast transdifferentiation from orbital fibroblasts is known to dominate tissue remodeling and fibrosis in Graves' ophthalmopathy (GO). However, the signaling pathways through which TGF-ß1 activates Graves' orbital fibroblasts remain unclear. This study investigated the role of the mitogen-activated protein kinase (MAPK) pathway in TGF-ß1-induced myofibroblast transdifferentiation in human Graves' orbital fibroblasts. The MAPK pathway was assessed by measuring the phosphorylation of p38, c-Jun N-terminal kinase (JNK), and extracellular-signal-regulated kinase (ERK) by Western blots. The expression of connective tissue growth factor (CTGF), α-smooth muscle actin (α-SMA), and fibronectin representing fibrogenesis was estimated. The activities of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) responsible for extracellular matrix (ECM) metabolism were analyzed. Specific pharmacologic kinase inhibitors were used to confirm the involvement of the MAPK pathway. After treatment with TGF-ß1, the phosphorylation levels of p38 and JNK, but not ERK, were increased. CTGF, α-SMA, and fibronectin, as well as TIMP-1 and TIMP-3, were upregulated, whereas the activities of MMP-2/-9 were inhibited. The effects of TGF-ß1 on the expression of these factors were eliminated by p38 and JNK inhibitors. The results suggested that TGF-ß1 could induce myofibroblast transdifferentiation in human Graves' orbital fibroblasts through the p38 and JNK pathways.


Asunto(s)
Transdiferenciación Celular/genética , MAP Quinasa Quinasa 4/genética , Factor de Crecimiento Transformador beta1/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Actinas/genética , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibronectinas/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Miofibroblastos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología
5.
Sci Rep ; 8(1): 7276, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29739987

RESUMEN

Connective tissue growth factor (CTGF) associated with transforming growth factor-ß (TGF-ß) play a pivotal role in the pathophysiology of many fibrotic disorders. However, it is not clear whether this interaction also takes place in GO. In this study, we investigated the role of CTGF in TGF-ß-induced extracellular matrix production and myofibroblast transdifferentiation in Graves' orbital fibroblasts. By Western blot analysis, we demonstrated that TGF-ß1 induced the expression of CTGF, fibronectin, and alpha-smooth muscle actin (α-SMA) in Graves' orbital fibroblasts. In addition, the protein levels of fibronectin and α-SMA in Graves' orbital fibroblasts were also increased after treatment with a recombinant human protein CTGF (rhCTGF). Moreover, we transfected the orbital fibroblasts with a small hairpin RNA of CTGF gene (shCTGF) to knockdown the expression levels of CTGF, which showed that knockdown of CTGF significantly diminished TGF-ß1-induced expression of CTGF, fibronectin and α-SMA proteins in Graves' orbital fibroblasts. Furthermore, the addition of rhCTGF to the shCTGF-transfected orbital fibroblasts could restore TGF-ß1-induced expression of fibronectin and α-SMA proteins. Our findings demonstrate that CTGF is an essential downstream mediator for TGF-ß1-induced extracellular matrix production and myofibroblast transdifferentiation in Graves' orbital fibroblasts and thus may provide with a potential therapeutic target for treatment of GO.


Asunto(s)
Transdiferenciación Celular/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Oftalmopatía de Graves/genética , Factor de Crecimiento Transformador beta1/genética , Actinas/genética , Adulto , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica/genética , Oftalmopatía de Graves/fisiopatología , Humanos , Masculino , Miofibroblastos/metabolismo , Órbita/patología
6.
J Formos Med Assoc ; 117(3): 212-219, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28438434

RESUMEN

BACKGROUND/PURPOSE: Deregulation of metabolic pathways is one of the hallmarks of cancer progression. Connective tissue growth factor (CTGF/CCN2) acts as a tumor suppressor in oral squamous cell carcinoma (OSCC). However, the role of CTGF in modulating cancer metabolism is still unclear. METHODS: OSCC cells stably overexpressing CTGF (SAS/CTGF) and shRNA against CTGF (TW2.6/shCTGF) were established. Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were examined by the Seahorse XF24 analyzer. The expression of CTGF and mitochondrial biogenesis related genes was measured by real-time polymerase chain reaction or Western blot analysis. RESULTS: CTGF decreased OCR, ECAR, adenosine triphosphate (ATP) generation, mitochondrial DNA (mtDNA), and mitochondrial transcription factor A (mtTFA) protein expression in OSCC cells. Overexpression of mtTFA restored CTGF-decreased OCR, ECAR, mtDNA copy number, migration and invasion of SAS/CTGF cells. Immunoprecipitation assay showed a higher level of ubiquitinated mtTFA protein after CTGF treatment. MG132, an inhibitor of proteasomal degradation, reversed the effect of CTGF on mtTFA protein expression in SAS cells. CONCLUSION: CTGF can decrease glycolysis, mitochondrial oxidative phosphorylation, ATP generation, and mtDNA copy number by increasing mtTFA protein degradation through ubiquitin proteasome pathway and in turn reduces migration and invasion of OSCC cells. Therefore, CTGF may be developed as a potential additive therapeutic drug for oral cancer in the near future.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/fisiología , Proteínas de Unión al ADN/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Neoplasias de la Boca/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina/fisiología , Línea Celular Tumoral , Humanos
7.
Br J Ophthalmol ; 101(5): 676-680, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27543288

RESUMEN

AIMS: To examine the expression of connective tissue growth factor (CTGF) in human cultured orbital fibroblasts from patients with Graves' ophthalmopathy (GO) and investigate whether a correlation exists between the presence of CTGF protein and clinical parameters of the disease. METHODS: The protein expression levels of CTGF were analysed by western blots in cultured orbital fibroblasts from 10 patients with GO and 7 age-matched normal controls. Associations between the protein expression of CTGF and the clinical factors of GO, including clinical demographics, thyroid function, clinical activity score (CAS) and ophthalmopathy index (OI), was evaluated. RESULTS: The mean protein expression levels of CTGF in the GO orbital fibroblasts were significantly higher than those of normal controls (p<0.001). Based on further analysis, the protein expression levels of CTGF in the GO orbital fibroblasts had significant correlation with gender (p=0.029), serum levels of thyrotropin receptor antibodies (p=0.029), CAS (p=0.048) and OI (p=0.043). Especially, there was a significant correlation between protein expression levels of CTGF and lid oedema (p=0.037), proptosis (p=0.045) and corneal involvement (p=0.001). CONCLUSIONS: Our findings revealed that the protein expression levels of CTGF in the GO orbital fibroblasts were significantly highly expressed than those of normal controls, and the elevated CTGF was associated with clinical characteristics and evolution, indicating CTGF may play a role in the pathogenesis and pathophysiology of GO.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Oftalmopatía de Graves/metabolismo , Adulto , Autoanticuerpos/sangre , Blefaroptosis/fisiopatología , Western Blotting , Estudios de Casos y Controles , Células Cultivadas , Exoftalmia/fisiopatología , Femenino , Fibroblastos/metabolismo , Oftalmopatía de Graves/inmunología , Humanos , Masculino , Persona de Mediana Edad , Receptores de Tirotropina/inmunología
8.
Oxid Med Cell Longev ; 2016: 4676289, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27340508

RESUMEN

Cigarette smoking is the most important risk factor for the development or deterioration of Graves' ophthalmopathy. Smoke-induced increased generation of reactive oxygen species may be involved. However, it remains to be clarified how orbital fibroblasts are affected by cigarette smoking. Our study demonstrated that Graves' orbital fibroblasts have exaggerated response to cigarette smoke extract challenge along with increased oxidative stress, fibrosis-related genes expression, especially connective tissue growth factor, and intracellular levels of transforming growth factor-ß1 and interleukin-1ß. The findings obtained in this study provide some clues for the impact of cigarette smoking on Graves' ophthalmopathy and offer a theoretical basis for the potential and rational use of antioxidants in treating Graves' ophthalmopathy.


Asunto(s)
Fibroblastos/efectos de los fármacos , Oftalmopatía de Graves/metabolismo , Órbita/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Humo/efectos adversos , Fumar/efectos adversos , Adulto , Antioxidantes/farmacología , Estudios de Casos y Controles , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , Oftalmopatía de Graves/genética , Oftalmopatía de Graves/patología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Órbita/metabolismo , Órbita/patología , Estrés Oxidativo/genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
9.
PLoS One ; 10(11): e0143514, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26599235

RESUMEN

Graves' ophthalmopathy (GO) is a disfiguring and sometimes blinding disease, which is characterized by inflammation and swelling of orbital tissues, with fibrosis and adipogenesis being predominant features. The aim of this study is to investigate whether the expression levels of fibrosis-related genes, especially that of connective tissue growth factor (CTGF), are altered in orbital fibroblasts of patients with GO. The role of oxidative stress in the regulation of CTGF expression in GO orbital fibroblasts is also examined. By a SYBR Green-based real time quantitative PCR (RT-QPCR), we demonstrated that the mRNA expression levels of fibronectin, apolipoprotein J, and CTGF in cultured orbital fibroblasts from patients with GO were significantly higher than those of age-matched normal controls (p = 0.007, 0.037, and 0.002, respectively). In addition, the protein expression levels of fibronectin, apolipoprotein J, and CTGF analyzed by Western blot were also significantly higher in GO orbital fibroblasts (p = 0.046, 0.032, and 0.008, respectively) as compared with the control. Furthermore, after treatment of orbital fibroblasts with a sub-lethal dose of hydrogen peroxide (200 µM H2O2), we found that the H2O2-induced increase of CTGF expression was more pronounced in the GO orbital fibroblasts as compared with those in normal controls (20% vs. 7%, p = 0.007). Importantly, pre-incubation with antioxidants including N-acetylcysteine (NAC) and vitamin C, respectively, resulted in significant attenuation of the induction of CTGF in GO orbital fibroblasts in response to H2O2 (p = 0.004 and 0.015, respectively). Taken together, we suggest that oxidative stress plays a role in the alteration of the expression of CTGF in GO orbital fibroblasts that may contribute to the pathogenesis and progression of GO. Antioxidants may be used in combination with the therapeutic agents for effective treatment of GO.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/genética , Fibroblastos/metabolismo , Expresión Génica , Oftalmopatía de Graves/genética , Adulto , Antioxidantes/metabolismo , Antioxidantes/farmacología , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibrosis/genética , Regulación de la Expresión Génica , Oftalmopatía de Graves/patología , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba , Adulto Joven
10.
Toxins (Basel) ; 7(1): 81-96, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25584429

RESUMEN

Malic acid (MA) has been commonly used in cosmetic products, but the safety reports in skin are sparse. To investigate the biological effects of MA in human skin keratinocytes, we investigated the potential cytotoxicity and apoptotic effects of MA in human keratinocyte cell lines (HaCaT). The data showed that MA induced apoptosis based on the observations of DAPI staining, DNA fragmentation, and sub-G1 phase in HaCaT cells and normal human epidermal keratinocytes (NHEKs). Flow cytometric assays also showed that MA increased the production of mitochondrial superoxide (mito-SOX) but decreased the mitochondrial membrane potential. Analysis of bioenergetics function with the XF 24 analyzer Seahorse extracellular flux analyzer demonstrated that oxygen consumption rate (OCR) was significantly decreased whereas extracellular acidification rate (ECAR) was increased in MA-treated keratinocytes. The occurrence of apoptosis was proved by the increased expressions of FasL, Fas, Bax, Bid, caspases-3, -8, -9, cytochrome c, and the declined expressions of Bcl-2, PARP. MA also induced endoplasmic reticulum stress associated protein expression such as GRP78, GADD153, and ATF6α. We demonstrated that MA had anti-proliferative effect in HaCaT cell through the inhibition of cell cycle progression at G0/G1, and the induction of programmed cell death through endoplasmic reticulum stress- and mitochondria-dependent pathways.


Asunto(s)
Queratinocitos/efectos de los fármacos , Malatos/toxicidad , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Daño del ADN , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Epidérmicas , Humanos , Queratinocitos/metabolismo , Mitocondrias/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
11.
Curr Pharm Des ; 20(35): 5510-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24606797

RESUMEN

Mitochondria are the organelles producing most of the energy and play important roles in a variety of biochemical functions in human cells. Mitochondrial defects can cause ATP deficiency and overproduction of reactive oxygen species, which are the major hallmarks of mitochondrial diseases. Abundant evidence has suggested that mitochondrial dysfunction-elicited oxidative stress can play an important role in the pathogenesis and progression of mitochondrial diseases. Mitochondria can respond to energy deficiency by the retrograde signaling to trigger a number of molecular events to help the human cells to cope with physiological or environmental changes. In this article, we first describe oxidative stress-induced cellular responses including metabolic adaptation, compensatory increase of mitochondrial biogenesis, upregulation of antioxidant enzymes, and alteration of protein acetylation in human cells with mitochondrial dysfunction. In this regard, we review recent findings to elucidate the mechanisms by which human cells motivate their mitochondria and the antioxidant defense system to respond to energy deficiency and oxidative stress, which contribute to the adaptive metabolic reprogramming in mitochondrial diseases. In addition, we emphasize the critical role of the activation of AMPK, Sirt1 and Sirt3 in the metabolic adaptation of human cells harboring mitochondrial DNA mutations. Recent studies have revealed that AMPK and sirtuins-mediated signaling pathways are involved in metabolic reprogramming, which is effected by upregulation of antioxidant defense system and mitochondrial protein acetylation, in human cells with mitochondrial dysfunction. Finally, we discuss several potential modulators of bioenergetic function such as coenzyme Q10, mitochondria-targeting antioxidants, resveratrol, and L-carnitine based on recent findings from studies on human cells and animal models of mitochondrial diseases. Elucidation of the signaling pathway of this adaptive response to oxidative stress triggered by mitochondrial dysfunction may enable us to gain a deeper insight into the communication between mitochondria and the nucleus and guide us to develop novel therapeutic agents for effective treatment of mitochondrial diseases.


Asunto(s)
Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Estrés Oxidativo/fisiología , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Humanos , Mitocondrias/efectos de los fármacos , Enfermedades Mitocondriales/fisiopatología , Enfermedades Mitocondriales/terapia , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
12.
Biochim Biophys Acta ; 1840(4): 1331-44, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24513455

RESUMEN

BACKGROUND: Mitochondrial DNA (mtDNA) mutations are an important cause of mitochondrial diseases, for which there is no effective treatment due to complex pathophysiology. It has been suggested that mitochondrial dysfunction-elicited reactive oxygen species (ROS) plays a vital role in the pathogenesis of mitochondrial diseases, and the expression levels of several clusters of genes are altered in response to the elevated oxidative stress. Recently, we reported that glycolysis in affected cells with mitochondrial dysfunction is upregulated by AMP-activated protein kinase (AMPK), and such an adaptive response of metabolic reprogramming plays an important role in the pathophysiology of mitochondrial diseases. SCOPE OF REVIEW: We summarize recent findings regarding the role of AMPK-mediated signaling pathways that are involved in: (1) metabolic reprogramming, (2) alteration of cellular redox status and antioxidant enzyme expression, (3) mitochondrial biogenesis, and (4) autophagy, a master regulator of mitochondrial quality control in skin fibroblasts from patients with mitochondrial diseases. MAJOR CONCLUSION: Induction of adaptive responses via AMPK-PFK2, AMPK-FOXO3a, AMPK-PGC-1α, and AMPK-mTOR signaling pathways, respectively is modulated for the survival of human cells under oxidative stress induced by mitochondrial dysfunction. We suggest that AMPK may be a potential target for the development of therapeutic agents for the treatment of mitochondrial diseases. GENERAL SIGNIFICANCE: Elucidation of the adaptive mechanism involved in AMPK activation cascades would lead us to gain a deeper insight into the crosstalk between mitochondria and the nucleus in affected tissue cells from patients with mitochondrial diseases. This article is part of a Special Issue entitled Frontiers of Mitochondrial Research.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Adaptación Biológica/fisiología , Fenómenos Fisiológicos Celulares , Enfermedades Mitocondriales/etiología , Estrés Oxidativo/fisiología , Autofagia/fisiología , Humanos , Mitocondrias/fisiología , Enfermedades Mitocondriales/metabolismo
13.
Biochem J ; 459(1): 149-60, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24476000

RESUMEN

MERRF (myoclonus epilepsy associated with ragged-red fibres) is a maternally inherited mitochondrial encephalomyopathy with various syndromes involving both muscular and nervous systems. The most common mutation in MERRF syndrome, the A8344G mutation in mtDNA, has been associated with severe defects in the respiratory function of mitochondria. In the present study, we show that there is a significant decrease in CA8 (carbonic anhydrase-related protein VIII) in cybrids harbouring the MERRF A8344G mutation. CA8 deficiency and mutations were found to be associated with a distinctive lifelong gait disorder in wdl (Waddles) mice and novel syndromes characterized by cerebellar ataxia and mental retardation in humans. The results of the present study showed that overexpression of CA8 in MERRF cybrids significantly decreased cell death induced by STS (staurosporine) treatment, suggesting a protective function of CA8 in cells harbouring the A8344G mutation of mtDNA. Interestingly, an increase in the formation of LC3-II (microtubule-associated protein 1 light chain 3-II) was found in the cybrids with down-regulated CA8 expression, suggesting that reduced expression of CA8 leads to autophagy activation. Furthermore, cybrids exhibiting down-regulated CA8 showed increased cytosolic Ca2+ signals and reduced levels of phospho-Akt compared with those in the cybrids with overexpressed CA8, indicating that phospho-Akt is involved in the protection of cells by CA8. Our findings suggest that CA8 is involved in the autophagic pathway and may have a protective role in cultured cells from patients with MERRF. Targeting CA8 and the downstream autophagic pathway might help develop therapeutic agents for treatment of MERRF syndrome in the future.


Asunto(s)
Biomarcadores de Tumor/genética , ADN Mitocondrial/genética , Síndrome MERRF/genética , Mutación/fisiología , Biomarcadores de Tumor/biosíntesis , Muerte Celular/genética , Línea Celular , ADN Mitocondrial/biosíntesis , Humanos , Síndrome MERRF/metabolismo
14.
Exp Biol Med (Maywood) ; 238(5): 450-60, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23856898

RESUMEN

Under normal physiological conditions, reactive oxygen species (ROS) serve as 'redox messengers' in the regulation of intracellular signalling, whereas excess ROS may induce irreversible damage to cellular components and lead to cell death by promoting the intrinsic apoptotic pathway through mitochondria. In the aging process, accumulation of mitochondria DNA mutations, impairment of oxidative phosphorylation as well as an imbalance in the expression of antioxidant enzymes result in further overproduction of ROS. This mitochondrial dysfunction-elicited ROS production axis forms a vicious cycle, which is the basis of mitochondrial free radical theory of aging. In addition, several lines of evidence have emerged recently to demonstrate that ROS play crucial roles in the regulation of cellular metabolism, antioxidant defence and posttranslational modification of proteins. We first discuss the oxidative stress responses, including metabolites redistribution and alteration of the acetylation status of proteins, in human cells with mitochondrial dysfunction and in aging. On the other hand, autophagy and mitophagy eliminate defective mitochondria and serve as a scavenger and apoptosis defender of cells in response to oxidative stress during aging. These scenarios mediate the restoration or adaptation of cells to respond to aging and age-related disorders for survival. In the natural course of aging, the homeostasis in the network of oxidative stress responses is disturbed by a progressive increase in the intracellular level of the ROS generated by defective mitochondria. Caloric restriction, which is generally thought to promote longevity, has been reported to enhance the efficiency of this network and provide multiple benefits to tissue cells. In this review, we emphasize the positive and integrative roles of mild oxidative stress elicited by mitochondria in the regulation of adaptation, anti-aging and scavenging pathway beyond their roles in the vicious cycle of mitochondrial dysfunction in the aging process.


Asunto(s)
ADN Mitocondrial/metabolismo , Longevidad , Mitocondrias/metabolismo , Mutación , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/genética , Autofagia/genética , ADN Mitocondrial/genética , Humanos , Mitocondrias/genética , Mitocondrias/patología , Mitofagia/genética , Procesamiento Proteico-Postraduccional
15.
Mol Vis ; 19: 927-34, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23687429

RESUMEN

PURPOSE: To investigate the biphasic effects of hydrogen peroxide (H2O2) on the orbital fibroblasts of patients with Graves' ophthalmopathy (GO) and the relation to antioxidants and proinflammatory cytokines. METHODS: Proliferation of cultured orbital fibroblasts from patients with GO and normal controls was evaluated in response to various concentrations of H2O2. The effect of low concentrations of H2O2 (6.25 µM) on the cellular proliferation and induction of intracellular proinflammatory cytokines, and reactive oxygen species of orbital fibroblasts were assessed. Protective effects of N-acetylcysteine and vitamin C on GO fibroblasts in response to 6.25 µM H2O2 stimulation were also investigated. RESULTS: When the GO fibroblasts were exposed to H2O2 at a concentration of 50 µM or above, significant cytotoxicity was observed. In contrast, lower concentrations of H2O2 (3.125-25 µM) increased the survival of GO fibroblasts with the peak cellular proliferation at 6.25 µM H2O2. However, this biphasic effect of H2O2 on the viability of orbital fibroblasts was not found in normal controls. In addition, 6.25 µM H2O2 led to significant elevation of the levels of transforming growth factor, beta 1, interleukin-1ß, and superoxide anion in GO fibroblasts, but no significant change in the normal controls. Pretreatment with N-acetylcysteine or vitamin C reversed the enhanced proliferation capacity and the induction of transforming growth factor, beta 1, interleukin-1ß and superoxide anion of GO fibroblasts in response to 6.25 µM H2O2. CONCLUSIONS: These findings revealed the biphasic effect of H2O2 on cellular proliferation of GO orbital fibroblasts. Importantly, a low level of H2O2 can stimulate proliferation of GO orbital fibroblasts and induce the production of proinflammatory cytokines, which can be inhibited by pretreatment with antioxidants. This provides a theoretical basis for the rational use of antioxidant in treating GO at an early stage.


Asunto(s)
Antioxidantes/uso terapéutico , Fibroblastos/patología , Oftalmopatía de Graves/tratamiento farmacológico , Oftalmopatía de Graves/patología , Órbita/patología , Estrés Oxidativo , Sustancias Protectoras/uso terapéutico , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Adulto , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Ácido Ascórbico/uso terapéutico , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Interleucina-1beta/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Factor de Crecimiento Transformador beta/metabolismo
16.
Chem Res Toxicol ; 26(5): 662-73, 2013 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-23617821

RESUMEN

Quantum dots (QDs) are one of most utilized nanomaterials in nanocrystalline semiconductors. QDs emit near-infrared fluorescence and can be applied as probes for detecting vasculature and imaging in biological systems. Since QDs have potential in clinical application, the toxicity of QDs needs to be carefully evaluated. In our present study, we elucidate the cytotoxic mechanisms of QDs using a mouse renal adenocarcinoma (RAG) cell line. QDs in RAG cells increased intracellular reactive oxygen species (ROS) levels and induced autophagy at 6 h, leading to subsequent apoptosis at 24 h. QDs entered the cells and were located within the endoplasmic reticulum (ER), endosome, and lysosome at 6 h and endosome, lysosome, and mitochondria at 24 h. However, QDs only affected mitochondrial function and did not induce ER stress. N-Acetylcysteine, an antioxidant agent, reduced intracellular ROS levels and decreased QD-induced autophagy but enhanced QD-induced cell death. Moreover, 3-methylamphetamine (an autophagy inhibitor) also reduced the cell viability in QD-treated cells. These findings suggest that ROS plays an essential role in the regulation of QD-induced autophagy, which subsequently enhances cell survival. Taken together, these results suggest that oxidative stress-induced autophagy is a defense/survival mechanism against the cytotoxicity of QD.


Asunto(s)
Antineoplásicos/toxicidad , Autofagia/efectos de los fármacos , Cadmio/toxicidad , Nanopartículas del Metal/toxicidad , Estrés Oxidativo/efectos de los fármacos , Puntos Cuánticos , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
17.
Int J Mol Sci ; 14(3): 6414-35, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-23519108

RESUMEN

The human skin is an integral system that acts as a physical and immunological barrier to outside pathogens, toxicants, and harmful irradiations. Environmental ultraviolet rays (UV) from the sun might potentially play a more active role in regulating several important biological responses in the context of global warming. UV rays first encounter the uppermost epidermal keratinocytes causing apoptosis. The molecular mechanisms of UV-induced apoptosis of keratinocytes include direct DNA damage (intrinsic), clustering of death receptors on the cell surface (extrinsic), and generation of ROS. When apoptotic keratinocytes are processed by adjacent immature Langerhans cells (LCs), the inappropriately activated Langerhans cells could result in immunosuppression. Furthermore, UV can deplete LCs in the epidermis and impair their migratory capacity, leading to their accumulation in the dermis. Intriguingly, receptor activator of NF-κB (RANK) activation of LCs by UV can induce the pro-survival and anti-apoptotic signals due to the upregulation of Bcl-xL, leading to the generation of regulatory T cells. Meanwhile, a physiological dosage of UV can also enhance melanocyte survival and melanogenesis. Analogous to its effect in keratinocytes, a therapeutic dosage of UV can induce cell cycle arrest, activate antioxidant and DNA repair enzymes, and induce apoptosis through translocation of the Bcl-2 family proteins in melanocytes to ensure genomic integrity and survival of melanocytes. Furthermore, UV can elicit the synthesis of vitamin D, an important molecule in calcium homeostasis of various types of skin cells contributing to DNA repair and immunomodulation. Taken together, the above-mentioned effects of UV on apoptosis and its related biological effects such as proliferation inhibition, melanin synthesis, and immunomodulations on skin residential cells have provided an integrated biochemical and molecular biological basis for phototherapy that has been widely used in the treatment of many dermatological diseases.

18.
J Invest Dermatol ; 133(7): 1890-900, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23370535

RESUMEN

Arsenic causes several human cancers. Arsenic-induced Bowen's disease (As-BD), the most common arsenical cancer, is characterized by increased proliferation, dysplasia, and individual cell apoptosis, all of which involve mitochondria. We reported that arsenic causes aberrant keratinocyte proliferation through mtTFA-mediated mitochondrial biogenesis in As-BD. Increasing mitochondrial biogenesis causes cells to undergo oxidative stress. However, how arsenic induces oxidative stress and causes mtDNA damage in arsenical cancers remains largely unknown. Using tissues from As-BD patients and arsenic-treated keratinocytes, we determined the oxidative stress, antioxidant enzymes, DNA-repair enzymes, and 8-hydroxy-2'-deoxyguanosine (8-OHdG) level in mtDNA by immunofluorescence, real-time PCR, and western blot. The results showed that oxidative stress was enhanced in both As-BD and arsenic-treated keratinocytes. Antioxidant enzymes including manganese-superoxide anion and copper/zinc-superoxide anion and DNA-repair enzymes were upregulated concomitantly in tissues and cells. In arsenic-treated keratinocytes, increased mitochondrial oxidative stress and the 8-OHdG level in mtDNA were attenuated by pretreatment with ascorbic acid, a potent antioxidant. Further, we found several somatic mutations in the ND4, ND5, and ND6 genes of mtDNA in lesional but not in perilesional skin from As-BD patients. Taken together, the results suggest that oxidative damage and mutations to mtDNA might be involved in the arsenical skin cancers in the context of mitochondrial biogenesis.


Asunto(s)
Arsénico/efectos adversos , Enfermedad de Bowen/inducido químicamente , Enfermedad de Bowen/fisiopatología , Daño del ADN/fisiología , ADN Mitocondrial/fisiología , Estrés Oxidativo/fisiología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/fisiopatología , 8-Hidroxi-2'-Desoxicoguanosina , Anciano , Anciano de 80 o más Años , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Arsénico/farmacología , Enfermedad de Bowen/genética , Estudios de Casos y Controles , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Enzimas Reparadoras del ADN/metabolismo , ADN Mitocondrial/genética , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/patología , Persona de Mediana Edad , Mutación/genética , Neoplasias Cutáneas/genética , Superóxidos/metabolismo
19.
Kaohsiung J Med Sci ; 28(7 Suppl): S53-62, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22871604

RESUMEN

The safety of quantum dots (QDs) 705 was evaluated in this study. Mice were treated with QD705 (intravenous) at a single dose of (40 pmol) for 4, 12, 16, and 24 weeks. Effects of QD705 on kidneys were examined. While there was a lack of histopathology, reduction in renal functions was detected at 16 weeks. Electron microscopic examination revealed alterations in proximal convoluted tubule (PCT) cell mitochondria at even much earlier time, including disorientation and reduction of mitochondrial number (early change), mitochondrial swelling, and later compensatory mitochondrial hypertrophy (enlargement mitochondria: giant mitochondria with hyperplastic inner cristae) as well as mitochondrial hyperplasia (increase in mitochondrial biogenesis and numbers) were observed. Such changes probably represent compensatory attempts of the mitochondria for functional loss or reduction of mitochondria in QD705 treated animals. Moreover, degeneration of mitochondria (myelin-figure and cytoplasmic membranous body formation) and degradation of cytoplasmic materials (isolated cytoplasmic pockets of degenerated materials and focal cytoplasmic degradation) also occurred in later time points (16-24 weeks). Such mitochondrial changes were not identical with those induced by pure cadmium. Taken together, we suggest that mitochondria appeared to be the target of QD705 toxicity and specific mitochondrial markers may be useful parameters for toxicity assessments of QDs or other metal-based nanomaterials.


Asunto(s)
Colorantes Fluorescentes/toxicidad , Mitocondrias/efectos de los fármacos , Puntos Cuánticos , Animales , Nitrógeno de la Urea Sanguínea , Cadmio/farmacología , Cadmio/toxicidad , Creatinina/sangre , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Colorantes Fluorescentes/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Masculino , Ratones , Ratones Endogámicos ICR , Microscopía Electrónica de Transmisión , Mitocondrias/metabolismo , Mitocondrias/patología , Tamaño Mitocondrial/efectos de los fármacos , Selenio/farmacología , Selenio/toxicidad , Telurio/farmacología , Telurio/toxicidad
20.
Photomed Laser Surg ; 30(10): 579-86, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22891782

RESUMEN

OBJECTIVE: This study investigated the clinical effects of intravascular laser irradiation of blood (ILIB) therapy on oxidative stress and mitochondrial dysfunction in subjects with chronic spinal cord injury (SCI) resulting from trauma. BACKGROUND DATA: Little is known about how ILIB may generate antioxidant defenses in humans, and there is still a lack of randomized, sham-control studies to indicate its influence on different metabolic pathways. METHODS: Twenty-four chronic SCI subjects (assigned to a sham and a study group), and 12 normal subjects were recruited. The study group underwent 1 h daily of ILIB for 15 days over 3 weeks. The sham group underwent ILIB with no laser power. RESULTS: Baseline measurements established higher oxidative stress and mitochondrial dysfunction in the SCI subjects than in the normal subjects. At day 15 of therapy, the study group revealed a significantly higher mitochondrial DNA (mtDNA) copy number, white blood cell adenosine triphosphate (WBC ATP) synthesis, and total antioxidant capacity (TAC) with significantly reduced malondialdehyde (MDA), than did the sham group. The study group intragroup comparison revealed significantly increased mtDNA copy numbers, WBC ATP synthesis, and TAC, with significantly reduced MDA, compared with its baseline measurements. The sham group intragroup comparisons demonstrated no statistical differences. Low-density lipoprotein (LDL) in the study group was significantly reduced at days 10 and 15, with significantly higher high-density lipoprotein (HDL) at day 45. CONCLUSIONS: Our study results contribute to the knowledge about the effectiveness of ILIB in alleviating oxidative stress and mitochondrial dysfunction in chronic SCI patients.


Asunto(s)
Sangre/efectos de la radiación , Procedimientos Endovasculares/métodos , Terapia por Luz de Baja Intensidad/métodos , Enfermedades Mitocondriales/radioterapia , Estrés Oxidativo/efectos de la radiación , Traumatismos de la Médula Espinal/radioterapia , Adulto , Anciano , Análisis de Varianza , Biomarcadores/sangre , Distribución de Chi-Cuadrado , Enfermedad Crónica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Mitocondriales/sangre , Enfermedades Mitocondriales/etiología , Estudios Prospectivos , Valores de Referencia , Medición de Riesgo , Método Simple Ciego , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/diagnóstico , Traumatismos de la Médula Espinal/metabolismo , Estadísticas no Paramétricas , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...