Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Biochem J ; 477(23): 4581-4597, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33155636

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disease associated with the accumulation of amyloid-beta oligomers (AßO). Recent studies have demonstrated that mitochondria-specific autophagy (mitophagy) contributes to mitochondrial quality control by selectively eliminating the dysfunctional mitochondria. Mitochondria motility, which is regulated by Miro1, is also associated with neuronal cell functions. However, the role played by Miro1 in the mitophagy mechanism, especially relative to AßO and neurodegenerative disorders, remains unknown. In this study, AßO induced mitochondrial dysfunction, enhanced Parkin-mediated mitophagy, and reduced mitochondrial quantities in hippocampal neuronal cells (HT-22 cells). We demonstrated that AßO-induced mitochondrial fragmentation could be rescued to the elongated mitochondrial form and that mitophagy could be mitigated by the stable overexpression of Miro1 or by pretreatment with N-acetylcysteine (NAC)-a reactive oxygen species (ROS) scavenger-as assessed by immunocytochemistry. Moreover, using time-lapse imaging, under live cell-conditions, we verified that mitochondrial motility was rescued by the Miro1 overexpression. Finally, in hippocampus from amyloid precursor protein (APP)/presenilin 1 (PS1)/Tau triple-transgenic mice, we noted that the co-localization between mitochondria and LC3B puncta was increased. Taken together, these results indicated that up-regulated ROS, induced by AßO, increased the degree of mitophagy and decreased the Miro1 expression levels. In contrast, the Miro1 overexpression ameliorated AßO-mediated mitophagy and increased the mitochondrial motility. In AD model mice, AßO induced mitophagy in the hippocampus. Thus, our results would improve our understanding of the role of mitophagy in AD toward facilitating the development of novel therapeutic agents for the treatment of AßO-mediated diseases.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Mitofagia , Neurônios/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Animais , Linhagem Celular , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Ubiquitina-Proteína Ligases/genética , Proteínas rho de Ligação ao GTP/genética
2.
Int J Mol Sci ; 21(22)2020 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-33233448

RESUMO

In recent decades, many studies on the treatment and prevention of pancreatic cancer have been conducted. However, pancreatic cancer remains incurable, with a high mortality rate. Although mouse models have been widely used for preclinical pancreatic cancer research, these models have many differences from humans. Therefore, large animals may be more useful for the investigation of pancreatic cancer. Pigs have recently emerged as a new model of pancreatic cancer due to their similarities to humans, but no pig pancreatic cancer cell lines have been established for use in drug screening or analysis of tumor biology. Here, we established and characterized an immortalized miniature pig pancreatic cell line derived from primary pancreatic cells and pancreatic cancer-like cells expressing K-rasG12D regulated by the human PTF1A promoter. Using this immortalized cell line, we analyzed the gene expression and phenotypes associated with cancer cell characteristics. Notably, we found that acinar-to-ductal transition was caused by K-rasG12D in the cell line constructed from acinar cells. This may constitute a good research model for the analysis of acinar-to-ductal metaplasia in human pancreatic cancer.


Assuntos
Pâncreas/metabolismo , Neoplasias Pancreáticas/genética , Lesões Pré-Cancerosas/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Animais , Linhagem Celular , Transformação Celular Neoplásica/genética , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/genética , Pâncreas/patologia , Ductos Pancreáticos/metabolismo , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Transdução de Sinais/genética , Suínos , Porco Miniatura
3.
Cell Biol Toxicol ; 35(6): 573-588, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31147869

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder caused by amyloid beta oligomers (AßO), which induce cell death by triggering oxidative stress and endoplasmic reticulum (ER) stress. Oxidative stress is regulated by antioxidant enzymes, including peroxiredoxins. Peroxiredoxins (Prx) are classified into six subtypes, based on their localization and cysteine residues, and protect cells by scavenging hydrogen peroxide (H2O2). Peroxiredoxin 4 (Prx4) is unique in being localized to the ER; however, whether Prx4 protects neuronal cells from AßO-induced toxicity remains unclear, although Prx4 expression is upregulated in AßO-induced oxidative stress and ER stress. In this study, we established HT-22 cells in which Prx4 was either overexpressed or silenced to investigate its role in AßO-induced toxicity. AßO-stimulation of HT-22 cells with overexpressed Prx4 caused decreases in both AßO-induced ROS and ER stress (followed by ER expansion). In contrast, AßO stimulation caused increases in both ROS and ER stress that were notably higher in HT-22 cells with silenced Prx4 expression than in HT-22 cells. Consequently, Prx4 overexpression decreased apoptotic cell death and ameliorated the AßO-induced increase in intracellular Ca2+. Therefore, we conclude that Prx4 has a protective effect against AßO-mediated oxidative stress, ER stress, and neuronal cell death. Furthermore, these results suggest that Prx4 may be a target for preventing AßO toxicity in AD. Graphical abstract .


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peroxirredoxinas/metabolismo , Peptídeos beta-Amiloides/fisiologia , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Peroxirredoxinas/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Reprod Biol Endocrinol ; 16(1): 79, 2018 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-30111318

RESUMO

BACKGROUND: Mounting evidence shows that ROS regulation by various antioxidants is essential for the expression of enzymes involved in steroidogenesis and maintenance of progesterone production by the corpus luteum (CL). However, the underlying mechanisms of peroxiredoxin 1 (PRDX1), an antioxidant enzyme, in luteal function for progesterone production in mice have not been reported. The aim of this study was to evaluate the functional link between PRDX1 and progesterone production in the CL of Prdx1 knockout (K/O) mice in the functional stage of CL. METHODS: The expression pattern of the unfolded protein response (UPR) signaling pathways, endoplasmic reticulum (ER) stress-induced apoptosis related genes and peroxiredoxins 1 (PRDX1) were investigated by western blotting analysis in CL tissue of 10 weeks mice during functional stage of CL. The protein levels of these genes after ER-stress inducer tunicamycin (Tm), ER-stress inhibitor tauroursodeoxycholic acid (TUDCA) and ROS scavenger, N-acetylcysteine (NAC) stimulation by intraperitoneal (i.p) injection were also investigated in CL tissue of wild type (WT) mice. Finally, we examined progesterone production and UPR signaling related gene expression in CL tissue of Prdx1 K/O mice. RESULTS: We demonstrated that PRDX1 deficiency in the functional stage activates the UPR signaling pathways in response to ER stress-induced apoptosis. Interestingly, CL number, serum progesterone levels, and steroidogenic enzyme expression in Prdx1 K/O mice decreased significantly, compared to those in wild type mice. Levels of UPR signaling pathway markers (GRP78/BIP, P50ATF6, and phosphorylated (p)-eIF2) and ER-stress associated apoptotic factors (CHOP, p-JNK, and cleaved caspase-3) were dramatically increased in the CL tissue of Prdx1 K/O mice. In addition, administration of the NAC, reduced progesterone production and activated ER-stress-induced UPR signaling in the CL tissue obtained from the ovary of Prdx1 K/O mice. Taken together, these results indicated that reduction in serum progesterone levels and activation of ER-stress-induced UPR signaling are restored by NAC injection in the CL of Prdx1 K/O mice. CONCLUSION: These observations provide the first evidence regarding the basic mechanisms connecting PRDX1 and progesterone production in the functional stage of CL.


Assuntos
Corpo Lúteo/metabolismo , Peroxirredoxinas/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas , Acetilcisteína/farmacologia , Animais , Apoptose/genética , Colagogos e Coleréticos/farmacologia , Corpo Lúteo/efeitos dos fármacos , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Feminino , Sequestradores de Radicais Livres/farmacologia , Expressão Gênica/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peroxirredoxinas/genética , Progesterona/sangue , Ácido Tauroquenodesoxicólico/farmacologia
5.
Neurochem Int ; 155: 105312, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35231558

RESUMO

Recently, Parkin has been reported to induce endoplasmic reticulum (ER) stress. In addition, amyloid beta oligomers (AßO), hallmarks of Alzheimer's disease (AD), also increase ER stress in neurons. Because a mutation in the Parkin gene is a well-known predominant cause of familial Parkinson's disease (PD), Parkin has been well studied in PD but has not been well researched in AD. In this study, we investigated the role of AßO-mediated Parkin associated with ER stress in AD. For AD-based research, we used AßO treatments in mouse hippocampus-derived HT-22 cells. We stably expressed Parkin in HT-22 cells to confirm the hypothesis and used siParkin for downregulation of Parkin expression. Moreover, using hippocampi from amyloid precursor protein/presenilin 1/Tau triple transgenic mice (3xTg-AD mice), which are used for AD models, we confirmed the relationship between ER stress and Parkin in vivo. We observed that ATF4 upregulated AßO-increases in Parkin. Parkin overexpression aggravated ER stress in AßO-treated HT-22 cells and the hippocampi of 3xTg-AD mice. Parkin downregulation led to no significant change when compared to AßO-treated cells. Moreover, Parkin-mediated ER stress was not related to oxidative stress. Our study indicates that AßO-induced ATF4 upregulated Parkin levels and that Parkin increases ER stress as a positive feedback loop. Through this study, our findings provide a foundation for future studies on the specific mechanisms related to the role of Parkin in AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Morte Celular , Estresse do Retículo Endoplasmático , Retroalimentação , Camundongos , Camundongos Transgênicos , Ubiquitina-Proteína Ligases/genética
6.
Inflammation ; 44(2): 714-724, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33150538

RESUMO

Excessive microglial cell activation in the brain can lead to the production of various neurotoxic factors (e.g., pro-inflammatory cytokines, nitric oxide) which can, in turn, initiate neurodegenerative processes. Recent research has been reported that mitochondrial dynamics regulate the inflammatory response of lipopolysaccharide (LPS). Isoliquiritigenin (ISL) is a compound found in Glycyrrhizae radix with anti-inflammatory and antioxidant properties. In this study, we investigated the function of ISL on the LPS-induced pro-inflammatory response in BV-2 microglial cells. We showed that ISL reduced the LPS-induced increase in pro-inflammatory mediators (e.g., nitric oxide and pro-inflammatory cytokines) via the inhibition of ERK/p38/NF-κB activation and the generation of reactive oxygen species (ROS). Furthermore, ISL inhibited the excessive mitochondrial fission induced by LPS, regulating mitochondrial ROS generation and pro-inflammatory response by suppressing the calcium/calcineurin pathway to dephosphorylate Drp1 at the serine 637 residue. Interestingly, the ISL pretreatment reduced the number of apoptotic cells and levels of cleaved caspase3/PARP, compared to LPS-treated cells. Our findings suggested that ISL ameliorated the pro-inflammatory response of microglia by inhibiting dephosphorylation of Drp1 (Ser637)-dependent mitochondrial fission. This study provides the first evidence for the effects of ISL against LPS-induced inflammatory response related and its link to mitochondrial fission and the calcium/calcineurin pathway. Consequently, we also identified the protective effects of ISL against LPS-induced microglial apoptosis, highlighting the pharmacological role of ISL in microglial inflammation-mediated neurodegeneration.


Assuntos
Anti-Inflamatórios/farmacologia , Chalconas/farmacologia , Inflamação/tratamento farmacológico , Microglia/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Animais , Anti-Inflamatórios/uso terapêutico , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Calcineurina/metabolismo , Cálcio/metabolismo , Linhagem Celular , Chalconas/uso terapêutico , Citocinas/metabolismo , Citometria de Fluxo , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Lipopolissacarídeos , Camundongos , Microglia/fisiologia , Dinâmica Mitocondrial/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Cells ; 10(8)2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34440805

RESUMO

Vascularization of tissues, organoids and organ-on-chip models has been attempted using endothelial cells. However, the cultured endothelial cells lack the capacity to interact with other somatic cell types, which is distinct from developing vascular cells in vivo. Recently, it was demonstrated that blood vessel organoids (BVOs) recreate the structure and functions of developing human blood vessels. However, the tissue-specific adaptability of BVOs had not been assessed in somatic tissues. Herein, we investigated whether BVOs infiltrate human cerebral organoids and form a blood-brain barrier. As a result, vascular cells arising from BVOs penetrated the cerebral organoids and developed a vessel-like architecture composed of CD31+ endothelial tubes coated with SMA+ or PDGFR+ mural cells. Molecular markers of the blood-brain barrier were detected in the vascularized cerebral organoids. We revealed that BVOs can form neural-specific blood-vessel networks that can be maintained for over 50 days.


Assuntos
Vasos Sanguíneos/fisiologia , Encéfalo/irrigação sanguínea , Neovascularização Fisiológica/fisiologia , Organoides/irrigação sanguínea , Vasos Sanguíneos/citologia , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Encéfalo/citologia , Técnicas de Cocultura , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio/citologia , Endotélio/metabolismo , Humanos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Organoides/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo
8.
J Biomech Eng ; 132(2): 025001, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20370251

RESUMO

Orthopaedic research on in vitro forces applied to bones, tendons, and ligaments during joint loading has been difficult to perform because of limitations with existing robotic simulators in applying full-physiological loading to the joint under investigation in real time. The objectives of the current work are as follows: (1) describe the design of a musculoskeletal simulator developed to support in vitro testing of cadaveric joint systems, (2) provide component and system-level validation results, and (3) demonstrate the simulator's usefulness for specific applications of the foot-ankle complex and knee. The musculoskeletal simulator allows researchers to simulate a variety of loading conditions on cadaver joints via motorized actuators that simulate muscle forces while simultaneously contacting the joint with an external load applied by a specialized robot. Multiple foot and knee studies have been completed at the Cleveland Clinic to demonstrate the simulator's capabilities. Using a variety of general-use components, experiments can be designed to test other musculoskeletal joints as well (e.g., hip, shoulder, facet joints of the spine). The accuracy of the tendon actuators to generate a target force profile during simulated walking was found to be highly variable and dependent on stance position. Repeatability (the ability of the system to generate the same tendon forces when the same experimental conditions are repeated) results showed that repeat forces were within the measurement accuracy of the system. It was determined that synchronization system accuracy was 6.7+/-2.0 ms and was based on timing measurements from the robot and tendon actuators. The positioning error of the robot ranged from 10 microm to 359 microm, depending on measurement condition (e.g., loaded or unloaded, quasistatic or dynamic motion, centralized movements or extremes of travel, maximum value, or root-mean-square, and x-, y- or z-axis motion). Algorithms and methods for controlling specimen interactions with the robot (with and without muscle forces) to duplicate physiological loading of the joints through iterative pseudo-fuzzy logic and real-time hybrid control are described. Results from the tests of the musculoskeletal simulator have demonstrated that the speed and accuracy of the components, the synchronization timing, the force and position control methods, and the system software can adequately replicate the biomechanics of human motion required to conduct meaningful cadaveric joint investigations.


Assuntos
Pé/fisiologia , Articulação do Joelho/fisiologia , Movimento/fisiologia , Postura/fisiologia , Tendões/fisiologia , Algoritmos , Fenômenos Biomecânicos , Cadáver , Humanos , Movimento (Física) , Software
9.
Cell Death Dis ; 11(3): 204, 2020 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-32205843

RESUMO

Iron is an essential element for cellular functions, including those of neuronal cells. However, an imbalance of iron homeostasis, such as iron overload, has been observed in several neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Iron overload causes neuronal toxicity through mitochondrial fission, dysregulation of Ca2+, ER-stress, and ROS production. Nevertheless, the precise mechanisms between iron-induced oxidative stress and iron toxicity related to mitochondria and endoplasmic reticulum (ER) in vivo are not fully understood. Here, we demonstrate the role of peroxiredoxin 5 (Prx5) in iron overload-induced neurotoxicity using Prx5-deficient mice. Iron concentrations and ROS levels in mice fed a high iron diet were significantly higher in Prx5-/- mice than wildtype (WT) mice. Prx5 deficiency also exacerbated ER-stress and ER-mediated mitochondrial fission via Ca2+/calcineurin-mediated dephosphorylation of Drp1 at Serine 637. Moreover, immunoreactive levels of cleaved caspase3 in the CA3 region of the hippocampus were higher in iron-loaded Prx5-/- mice than WT mice. Furthermore, treatment with N-acetyl-cysteine, a reactive oxygen species (ROS) scavenger, attenuated iron overload-induced hippocampal damage by inhibiting ROS production, ER-stress, and mitochondrial fission in iron-loaded Prx5-/- mice. Therefore, we suggest that iron overload-induced oxidative stress and ER-mediated mitochondrial fission may be essential for understanding iron-mediated neuronal cell death in the hippocampus and that Prx5 may be useful as a novel therapeutic target in the treatment of iron overload-mediated diseases and neurodegenerative diseases.


Assuntos
Hipocampo/metabolismo , Sobrecarga de Ferro/metabolismo , Dinâmica Mitocondrial/fisiologia , Neurônios/metabolismo , Peroxirredoxinas/deficiência , Animais , Morte Celular/fisiologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Estresse do Retículo Endoplasmático , Feminino , Hipocampo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Peroxirredoxinas/genética , Gravidez , Transdução de Sinais
10.
Foot Ankle Int ; 30(8): 767-72, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19735634

RESUMO

BACKGROUND: One of the more serious diabetic complications is Charcot neuroarthropathy (CN), a disease that results in arch collapse and permanent foot deformity. However, very little is known about the etiology of CN. From a mechanical standpoint, it is likely that there is a ;;vicious circle'' in terms of (i) arch collapse causing increased midfoot joint pressures, and (ii) increased joint contact pressures exacerbating the collapse of midfoot bones. This study focused on assessment of peak joint pressure difference between diabetic and non-diabetic cadaver feet during simulated walking. We hypothesized that joint pressures are higher for diabetics than normal population. MATERIALS AND METHODS: Sixteen cadaver foot specimens (eight control and eight diabetic specimens) were used in this study. Human gait at 25% of typical walking speed (averaged stance duration of 3.2s) was simulated by a custom-designed Universal Musculoskeletal Simulator. Four medial midfoot joint pressures (the first metatarsocuneiform, the medial naviculocuneiform, the middle naviculocuneiform, and the first intercuneiform) were measured dynamically during full stance. RESULTS: The pressures in each of the four measured midfoot joints were significantly greater in the diabetic feet (p = 0.015, p = 0.025, p < 0.001, and p = 0.545, respectively). CONCLUSION: Across all four tested joints, the diabetic cadaver specimens had, on average, 46% higher peak pressures than the control cadaver feet during the simulated stance phase. CLINICAL RELEVANCE: This finding suggests that diabetic patients could be predisposed to arch collapse even before there are visible signs of bone or joint abnormalities.


Assuntos
Artropatia Neurogênica/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Robótica , Articulações Tarsianas/fisiopatologia , Caminhada/fisiologia , Suporte de Carga/fisiologia , Idoso , Idoso de 80 Anos ou mais , Articulação do Tornozelo/fisiopatologia , Artropatia Neurogênica/etiologia , Artropatia Neurogênica/patologia , Cadáver , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Neurológicos , Amplitude de Movimento Articular
11.
Neurotoxicology ; 74: 242-251, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31408635

RESUMO

Diethylhexyl phthalate (DEHP) is used in many plastic products, such as perfumes, lunch boxes, bags, and building materials. As DEHP is not covalently bound to the plastic, humans can be easily exposed to it. DEHP induces neurobehavioral changes and neuronal cell death; however, the exact mechanism behind this is still unclear. We hypothesized that the neurotoxic mechanism is related to DEHP-induced oxidative stress leading to apoptosis through mitochondrial fission. We demonstrated that DEHP-induced oxidative stress triggers neuronal cell death via mitochondrial fission in mouse hippocampal HT-22 cells. Furthermore, we identified that peroxiredoxin 5 (Prx5), an antioxidant enzyme induced by DEHP, prevents DEHP-induced mitochondrial fission by inhibiting the production of reactive oxygen species. We conclude that Prx5 may be a promising therapeutic target for mitigating DEHP-induced neuronal cell death.


Assuntos
Morte Celular/genética , Dietilexilftalato/toxicidade , Hipocampo/patologia , Mitocôndrias/genética , Neurônios/efeitos dos fármacos , Peroxirredoxinas/farmacologia , Animais , Apoptose/genética , Linhagem Celular , Técnicas de Silenciamento de Genes , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Camundongos , Mitocôndrias/ultraestrutura , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Peroxirredoxinas/genética , Espécies Reativas de Oxigênio
12.
Neurotoxicology ; 68: 133-141, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30048666

RESUMO

Numerous studies suggest that glutamate toxicity is a major contributor to neuronal dysfunction and death in several neurodegenerative diseases. In our previous study, isoliquiritigenin (ISL) isolated from Glycyrrhiza uralensis showed neuroprotective effects against neuronal cell death mediated by intracellular reactive oxygen species (ROS) generation and loss of mitochondrial membrane potential. However, the mechanisms by which ISL protects against glutamate-induced oxidative stress are unknown. In the present study, we focused on the cellular and molecular mechanisms underlying the inhibition of ROS production and induction of mitochondrial dysfunction by ISL in glutamate-stimulated HT22 mouse hippocampal neuron cells. The results revealed that ISL inhibited glutamate-induced mitochondrial ROS production and decline of glutathione levels and ATP generation in HT22 cells. Interestingly, we discovered that ISL prevents glutamate-induced mitochondrial fission by inhibiting the dephosphorylation of Drp1 at the serine 637 residue, which is a regulatory factor of mitochondrial dynamics, and both a S637D mutation of Drp1, which resulted in a phosphorylation-mimetic form of Drp1 at Ser637, and mitochondria-targeted antioxidant Mito-TEMPO inhibited glutamate-induced mitochondrial fission. Furthermore, ISL also prevented the increase of intracellular calcium accompanied by activation of calcineurin, which is a key regulator of dephosphorylation of Drp1 (Ser637), in glutamate-treated HT22 cells. Taken together, our results demonstrated that ISL protects against glutamate-induced mitochondrial fission by inhibiting the increase of mitochondrial ROS and intracellular calcium, which are accompanied by dephosphorylation of Drp1 (Ser637), and consequently attenuates glutamate-induced neuronal cell death. Therefore, these findings suggest that ISL exhibits the potential for protection against glutamate toxicity. These results may contribute to the development of new drugs and novel strategies for the treatment of neurodegenerative disorders related to glutamate toxicity.


Assuntos
Calcineurina/metabolismo , Chalconas/administração & dosagem , Dinaminas/metabolismo , Ácido Glutâmico/toxicidade , Hipocampo/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Animais , Linhagem Celular , Hipocampo/metabolismo , Humanos , Camundongos , Neurônios/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
13.
Free Radic Biol Med ; 123: 96-106, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29778464

RESUMO

Reactive oxygen species (ROS) produced in biological reactions have been shown to contribute to ovarian aging. Peroxiredoxin 2 (Prx2) is an antioxidant enzyme that protects cells by scavenging ROS; however, its effect on age-related, oxidative stress-associated ovarian failure has not been reported. Here, we investigated its role in age-related ovarian dysfunction and 4-vinylcyclohexene diepoxide (VCD)-induced premature ovarian failure using Prx2-deficient mice. Compared to those in wildtype (WT) mice, serum levels of anti-Müllerian hormone, 17ß-estradiol, and progesterone and numbers of follicles and corpora lutea were significantly lower in 18-month-old Prx2-/- mice. Moreover, levels of Bax, cytochrome c, cleaved caspase-3, and phosphorylated JNK proteins were higher and numbers of apoptotic (terminal deoxynucleotidyl transferase dUTP nick end labeling-positive) cells were considerably greater in 18-month-old Prx2-/- ovaries than WT ovaries. Furthermore, the effects of the ovarian toxicant VCD in significantly enhancing ROS levels and apoptosis through activation of JNK-mediated apoptotic signaling were more pronounced in Prx2-/- than WT mouse embryonic fibroblasts. Expression of the steroidogenic proteins StAR, CYP11A1, and 3ß-HSD and serum levels of 17ß-estradiol and progesterone were also reduced to a greater extent in Prx2-/- mice than WT mice after VCD injection. This reduced steroidogenesis was rescued by addition of the Prx mimic ebselen or JNK inhibitor SP600125. This constitutes the first report that Prx2 deficiency leads to acceleration of age-related or VCD-induced ovarian failure by activation of the ROS-induced JNK pathway. These findings suggest that Prx2 plays an important role in preventing accelerated ovarian failure by inhibiting ROS-induced JNK activation.


Assuntos
Envelhecimento , Sistema de Sinalização das MAP Quinases , Doenças Ovarianas/patologia , Folículo Ovariano/patologia , Estresse Oxidativo , Peroxirredoxinas/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose , Carcinógenos/toxicidade , Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/metabolismo , Corpo Lúteo/patologia , Cicloexenos/toxicidade , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças Ovarianas/induzido quimicamente , Doenças Ovarianas/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Transdução de Sinais , Compostos de Vinila/toxicidade
14.
Int J Biochem Cell Biol ; 102: 10-19, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29906559

RESUMO

Iron is an essential element for neuronal as well as cellular functions. However, Iron overload has been known to cause neuronal toxicity through mitochondrial fission, dysregulation of Ca2+, endoplasmic reticulum (ER) stress, and reactive oxygen species (ROS) production. Nevertheless, the precise mechanisms of iron-induced oxidative stress and mitochondria- and ER-related iron toxicity in neuronal cells are not fully understood. In this study, we demonstrated that iron overload induces ROS production earlier in the ER than in the mitochondria, and peroxiredoxin 5 (Prx5), which is a kind of antioxidant induced by iron overload, prevents iron overload-induced mitochondrial fragmentation mediated by contact with ER and translocation of Drp1, by inhibiting ROS production and calcium/calcineurin pathway in HT-22 mouse hippocampal neuronal cells. Moreover, Prx5 also prevented iron overload-induced ER-stress and cleavage of caspase-3, which consequently attenuated neuronal cell death. Therefore, we suggested that iron overload induces oxidative stress in the ER earlier than in the mitochondria, thereby increasing ER stress and calcium levels, and consequently causing mitochondrial fragmentation and neuronal cell death. So we thought that this study is essential for understanding iron toxicity in neurons, and Prx5 may serve as a new therapeutic target to prevent iron overload-induced diseases and neurodegenerative disorders.


Assuntos
Morte Celular , Estresse do Retículo Endoplasmático , Hipocampo/patologia , Sobrecarga de Ferro/patologia , Mitocôndrias/patologia , Neurônios/patologia , Peroxirredoxinas/metabolismo , Animais , Calcineurina/metabolismo , Cálcio/metabolismo , Linhagem Celular , Sobrecarga de Ferro/metabolismo , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
15.
Org Lett ; 9(1): 141-4, 2007 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-17192105

RESUMO

[reaction: see text] The Prins cyclization strategy was successfully applied in the total synthesis of (-)-blepharocalyxin D, a cytotoxic dimeric diarylheptanoid isolated from Alpinia blepharocalyx.


Assuntos
Piranos/síntese química , Cristalografia por Raios X , Modelos Moleculares , Estrutura Molecular , Piranos/química
16.
Biomol Ther (Seoul) ; 25(3): 308-314, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28173642

RESUMO

Urotensin II (UII) is a mitogenic and hypertrophic agent that can induce the proliferation of vascular cells. UII inhibition has been considered as beneficial strategy for atherosclerosis and restenosis. However, currently there is no therapeutics clinically available for atherosclerosis or restenosis. In this study, we evaluated the effects of a newly synthesized UII receptor (UT) antagonist, KR- 36996, on the proliferation of SMCs in vitro and neointima formation in vivo in comparison with GSK-1440115, a known potent UT antagonist. In primary human aortic SMCs (HASMCs), UII (50 nM) induced proliferation was significantly inhibited by KR-36996 at 1, 10, and 100 nM which showed greater potency (IC50: 3.5 nM) than GSK-1440115 (IC50: 82.3 nM). UII-induced proliferation of HASMC cells was inhibited by U0126, an ERK1/2 inhibitor, but not by SP600125 (inhibitor of JNK) or SB202190 (inhibitor of p38 MAPK). UII increased the phosphorylation level of ERK1/2. Such increase was significantly inhibited by KR-36996. UII-induced proliferation was also inhibited by trolox, a scavenger for reactive oxygen species (ROS). UII-induced ROS generation was also decreased by KR-36996 treatment. In a carotid artery ligation mouse model, intimal thickening was dramatically suppressed by oral treatment with KR-36996 (30 mg/kg) which showed better efficacy than GSK-1440115. These results suggest that KR-36996 is a better candidate than GSK-1440115 in preventing vascular proliferation in the pathogenesis of atherosclerosis and restenosis.

17.
Antioxid Redox Signal ; 27(11): 715-726, 2017 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-28358580

RESUMO

AIMS: Aberrant Cdk5 (cyclin-dependent kinase 5) and oxidative stress are crucial components of diverse neurodegenerative disorders, including Alzheimer's disease (AD). We previously reported that a change in peroxiredoxin (Prx) expression is associated with protection from neuronal death. The aim of the current study was to analyze the role of Prx in regulating Cdk5 activation in AD. RESULTS: We found that of the six Prx subtypes, Prx5 was increased the most in cellular (N2a-APPswe cells) model of AD. Prx5 in the brain of APP (amyloid precursor protein) transgenic mouse (Tg2576) was more increased than a nontransgenic mouse. We evaluated Prx5 function by using overexpression (Prx5-WT), a mutation in the catalytic residue (Prx5-C48S), and knockdown. Increased neuronal death and Cdk5 activation by amyloid beta oligomer (AßO) were rescued by Prx5-WT expression, but not by Prx5-C48S or Prx5 knockdown. Prx5 plays a role in Cdk5 regulation by inhibiting the conversion of p35 to p25, which is increased by AßO accumulation. Prx5 is also upregulated in both the cytosol and mitochondria and it protects cells from AßO-mediated oxidative stress by eliminating intracellular and mitochondrial reactive oxygen species. Moreover, Prx5 regulates Ca2+ and Ca2+-mediated calpain activation, which are key regulators of p35 cleavage to p25. Innovation and Conclusion: Our study represents the first demonstration that Prx5 induction is a key factor in the suppression of Cdk5-related neuronal death in AD and we show that it functions via regulation of Ca2+-mediated calpain activation. Antioxid. Redox Signal. 27, 715-726.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Quinase 5 Dependente de Ciclina/metabolismo , Peroxirredoxinas/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Cálcio/metabolismo , Calpaína/metabolismo , Linhagem Celular , Quinase 5 Dependente de Ciclina/genética , Citosol/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Humanos , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mutação , Regulação para Cima
18.
Toxicology ; 365: 17-24, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27481217

RESUMO

Iron is necessary for neuronal functions; however, excessive iron accumulation caused by impairment of iron balance could damage neurons. Neuronal iron accumulation has been observed in several neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. Nevertheless, the precise mechanisms underlying iron toxicity in neuron cells are not fully understood. In this study, we investigated the mechanism underlying iron overload-induced mitochondrial fragmentation in HT-22 hippocampal neuron cells that were incubated with ferric ammonium citrate (FAC). Mitochondrial fragmentation via dephosphorylation of Drp1 (Ser637) and increased apoptotic neuronal death were observed in FAC-stimulated HT-22 cells. Furthermore, the levels of intracellular calcium (Ca(2+)) were increased by iron overload. Notably, chelation of intracellular Ca(2+) rescued mitochondrial fragmentation and neuronal cell death. In addition, iron overload activated calcineurin through the Ca(2+)/calmodulin and Ca(2+)/calpain pathways. Pretreatment with the calmodulin inhibitor W13 and the calpain inhibitor ALLN attenuated iron overload-induced mitochondrial fragmentation and neuronal cell death. Therefore, these findings suggest that Ca(2+)-mediated calcineurin signals are a key player in iron-induced neurotoxicity by regulating mitochondrial dynamics. We believe that our results may contribute to the development of novel therapies for iron toxicity related neurodegenerative disorders.


Assuntos
Sinalização do Cálcio , Sobrecarga de Ferro/patologia , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Calcineurina/metabolismo , Cálcio/metabolismo , Calmodulina/antagonistas & inibidores , Calmodulina/metabolismo , Calpaína/antagonistas & inibidores , Calpaína/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Dinaminas/genética , Dinaminas/metabolismo , Compostos Férricos/química , Hipocampo/citologia , Ferro/toxicidade , Leupeptinas/farmacologia , Camundongos , Mitocôndrias/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Compostos de Amônio Quaternário/química , Sulfonamidas/farmacologia
19.
Biomol Ther (Seoul) ; 24(5): 523-8, 2016 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27582556

RESUMO

Urotensin II (UII) is a potent vasoactive peptide and mitogenic agent to induce proliferation of various cells including vascular smooth muscle cells (VSMCs). In this study, we examined the effects of a novel UII receptor (UT) antagonist, KR-36676, on vasoconstriction of aorta and proliferation of aortic SMCs. In rat aorta, UII-induced vasoconstriction was significantly inhibited by KR-36676 in a concentration-dependent manner. In primary human aortic SMCs (hAoSMCs), UII-induced cell proliferation was significantly inhibited by KR-36676 in a concentration-dependent manner. In addition, KR-36676 decreased UII-induced phosphorylation of ERK, and UII-induced cell proliferation was also significantly inhibited by a known ERK inhibitor U0126. In mouse carotid ligation model, intimal thickening of carotid artery was dramatically suppressed by oral treatment with KR-36676 (30 mg/ kg/day) for 4 weeks compared to vehicle-treated group. From these results, it is indicated that KR-36676 suppress UII-induced proliferation of VSMCs at least partially through inhibition of ERK activation, and that it also attenuates UII-induced vasoconstriction and vascular neointima formation. Our study suggest that KR-36676 may be an attractive candidate for the pharmacological management of vascular dysfunction.

20.
Free Radic Biol Med ; 99: 392-404, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27585948

RESUMO

Microglial activation is a hallmark of neurodegenerative diseases. ROS activates microglia by regulating transcription factors to express pro-inflammatory genes and is associated with disruption of Ca2+ homeostasis through thiol redox modulation. Recently, we reported that Prx5 can regulate activation of microglia cells by governing ROS. In addition, LPS leads to excessive mitochondrial fission, and regulation of mitochondrial dynamics involved in a pro-inflammatory response is important for the maintenance of microglial activation. However, the precise relationship among these signals and the role of Prx5 in mitochondrial dynamics and microglial activation is still unknown. In this study, we demonstrated that Ca2+/calcineurin-dependent de-phosphorylation of Drp1 induces mitochondrial fission and regulates mitochondrial ROS production, which influences the expression of pro-inflammatory mediators in LPS-induced microglia cells. Moreover, it is likely that cytosolic and Nox-derived ROS were upstream of mitochondrial fission and mitochondrial ROS generation in activated microglia cells. Prx5 regulates LPS-induced mitochondrial fission through modulation of Ca2+/calcineurin-dependent Drp1 de-phosphorylation by eliminating Nox-derived and cytosolic ROS. Therefore, we suggest that mitochondrial dynamics may be essential for understanding pro-inflammatory responses and that Prx5 may be used as a new therapeutic target to prevent neuroinflammation and neurodegenerative diseases.


Assuntos
Calcineurina/metabolismo , Cálcio/metabolismo , Dinaminas/metabolismo , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , Dinâmica Mitocondrial/efeitos dos fármacos , Peroxirredoxinas/metabolismo , Animais , Calcineurina/genética , Linhagem Celular Transformada , Dinaminas/genética , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/genética , NADPH Oxidase 1/genética , NADPH Oxidase 1/metabolismo , NADPH Oxidase 2/genética , NADPH Oxidase 2/metabolismo , Oxirredução , Peroxirredoxinas/genética , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA