Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropharmacology ; 238: 109652, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37422180

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disease. However, no curative or modifying therapy is known. Inosine is a purine nucleoside that increases brain-derived neurotrophic factor (BDNF) expression in the brain through adenosine receptors. Herein, we investigated the neuroprotective effects of inosine and elucidated the mechanisms underlying its pharmacological action. Inosine rescued SH-SY5Y neuroblastoma cells from MPP+ injury in a dose-dependent manner. Inosine protection correlated with BDNF expression and the activation of its downstream signaling cascade, as the TrkB receptor inhibitor, K252a and siRNA against the BDNF gene remarkably reduced the protective effects of inosine. Blocking the A1 or A2A adenosine receptors diminished BDNF induction and the rescuing effect of inosine, indicating a critical role of adenosine A1 and A2A receptors in inosine-related BDNF elevation. We assessed whether the compound could protect dopaminergic neurons from MPTP-induced neuronal injury. Beam-walking and challenge beam tests revealed that inosine pretreatment for 3 weeks reduced the MPTP-induced motor function impairment. Inosine ameliorated dopaminergic neuronal loss and MPTP-mediated astrocytic and microglial activation in the substantia nigra and striatum. Inosine ameliorated the depletion of striatal dopamine and its metabolite following MPTP injection. BDNF upregulation and the activation of its downstream signaling pathway seemingly correlate with the neuroprotective effects of inosine. To our knowledge, this is the first study to demonstrate the neuroprotective effects of inosine against MPTP neurotoxicity via BDNF upregulation. These findings highlight the therapeutic potential of inosine in dopaminergic neurodegeneration in PD brains.


Assuntos
Neuroblastoma , Doenças Neurodegenerativas , Fármacos Neuroprotetores , Doença de Parkinson , Humanos , Camundongos , Animais , Dopamina/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Regulação para Cima , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/tratamento farmacológico , Neurônios Dopaminérgicos , Substância Negra , Inosina/farmacologia , Inosina/metabolismo , Inosina/uso terapêutico , Camundongos Endogâmicos C57BL , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/metabolismo
2.
Mol Ther ; 31(3): 890-908, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36566348

RESUMO

Mesenchymal stem cells (MSCs) are ubiquitous multipotent cells that exhibit significant therapeutic potentials in a variety of disorders. Nevertheless, their clinical efficacy is limited owing to poor survival, low rate of engraftment, and impaired potency upon transplantation. Spheroidal three-dimensional (3D) culture of MSCs (MSC3D) has been proven to better preserve their in vivo functional properties. However, the molecular mechanisms underlying the improvement in MSC function by spheroid formation are not clearly understood. NLRP3 inflammasomes, a key component of the innate immune system, have recently been shown to play a role in cell fate decision of MSCs. The present study examined the role of NLRP3 inflammasomes in the survival and potency of MSC spheroids. We found that MSC3D led to decreased activation of NLRP3 inflammasomes through alleviation of ER stress in an autophagy-dependent manner. Importantly, downregulation of NLRP3 inflammasomes signaling critically contributes to the enhanced survival rate in MSC3D through modulation of pyroptosis and apoptosis. The critical role of NLRP3 inflammasome suppression in the enhanced therapeutic efficacy of MSC spheroids was further confirmed in an in vivo mouse model of DSS-induced colitis. These findings suggest that 3D culture confers survival and functional advantages to MSCs by suppressing NLRP3 inflammasome activation.


Assuntos
Colite , Inflamassomos , Células-Tronco Mesenquimais , Animais , Camundongos , Colite/induzido quimicamente , Colite/genética , Colite/imunologia , Inflamassomos/genética , Inflamassomos/imunologia , Células-Tronco Mesenquimais/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Transdução de Sinais , Técnicas de Cultura de Células em Três Dimensões
3.
J Nutr Biochem ; 112: 109212, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36370926

RESUMO

Dietary restriction through low-calorie intake or intermittent fasting benefits many organs, including the brain. This study investigated the neuroprotective effects of fasting in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. We found that fasting every other day rather than weekly increased the levels of brain-derived neurotrophic factor and glial-derived neurotrophic factor in the nigrostriatal pathway. Therefore, we maintained the animals on alternate-day fasting for 2 weeks and injected MPTP (30 mg/kg/day, intraperitoneally [i.p.]) for five days. We observed that alternate-day fasting attenuated MPTP-induced dopaminergic neuronal loss and astroglial activation in the substantia nigra and the striatum. Moreover, neurochemical analysis using high-performance liquid chromatography showed that alternate-day fasting reduced MPTP-induced depletion of striatal dopamine. Consistent with these results, behavioral tests showed that fasting suppressed the motor impairment caused by MPTP. Furthermore, fasting increased the phosphorylation of phosphatidylinositol-3-kinase and protein kinase B, which are downstream signaling molecules of neurotrophic factors. Fasting also increased the phosphorylation of extracellular signal-regulated protein kinase and cAMP response element-binding protein, further supporting the involvement of neurotrophic factors in the observed neuroprotective effects. Hence, our results demonstrated the dopaminergic neuroprotection of intermittent fasting in an MPTP mouse model of Parkinson's disease, supporting the idea that fasting could be an instrumental tool for preventing neurodegeneration in the brain.


Assuntos
Fármacos Neuroprotetores , Doença de Parkinson , Animais , Camundongos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Jejum Intermitente , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/metabolismo , Doença de Parkinson/metabolismo , Substância Negra
4.
Front Pharmacol ; 13: 997701, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36225577

RESUMO

MicroRNA (miRNA)-mediated striatal gene regulation may play an important role in methamphetamine (METH) addiction. This study aimed to identify changes in novel miRNAs and their target genes during METH self-administration and investigate their roles in METH-induced locomotion. RNA sequencing analysis revealed that mir-183-5p was upregulated in the striatum of METH self-administered rats, and target gene prediction revealed that the glucocorticoid receptor (GR) gene, Nr3c1, was a potential target gene for mir-183-5p. We confirmed that single and repeated METH administrations increased METH-induced locomotion and plasma corticosterone levels in rats. Additionally, increased miR-185-5p expression and decreased GR gene expression were observed only in the repeated-METH-injection group but not in the single-injection group. We then investigated the effects of miR-183-5p on METH-induced locomotion using a miR-183-5p mimic and inhibitor. Injection of a mir-183-5p mimic in the striatum of rats attenuated METH-induced locomotion, whereas injection of a miR-183-5p inhibitor enhanced the locomotor activity in METH-administered rats. Furthermore, the miR-183-5p mimic reduced the phosphorylation of tyrosine hydroxylase (TH) whereas the inhibitor increased it. Taken together, these results indicate that repeated METH injections increase striatal miR-183-5p expression and regulate METH-induced locomotion by regulating GR expression in rats, thereby suggesting a potential role of miR-183-5p as a novel regulator of METH-induced locomotion.

5.
Sci Adv ; 8(34): eabn8614, 2022 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-36001671

RESUMO

Immunomodulation is an essential consideration for cell replacement procedures. Unfortunately, lifelong exposure to nonspecific systemic immunosuppression results in immunodeficiency and has toxic effects on nonimmune cells. Here, we engineered hybrid spheroids of mesenchymal stem cells (MSCs) with rapamycin-releasing poly(lactic-co-glycolic acid) microparticles (RAP-MPs) to prevent immune rejection of islet xenografts in diabetic C57BL/6 mice. Hybrid spheroids were rapidly formed by incubating cell-particle mixture in methylcellulose solution while maintaining high cell viability. RAP-MPs were uniformly distributed in hybrid spheroids and sustainably released RAP for ~3 weeks. Locoregional transplantation of hybrid spheroids containing low doses of RAP-MPs (200- to 4000-ng RAP per recipient) significantly prolonged islet survival times and promoted the generation of regional regulatory T cells. Enhanced programmed death-ligand 1 expression by MSCs was found to be responsible for the immunomodulatory performance of hybrid spheroids. Our results suggest that these hybrid spheroids offer a promising platform for the efficient use of MSCs in the transplantation field.


Assuntos
Células-Tronco Mesenquimais , Esferoides Celulares , Animais , Humanos , Imunomodulação , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transplante Heterólogo
6.
Mol Neurobiol ; 59(9): 5874-5890, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35804280

RESUMO

Boswellia serrata gum is a natural product that showed beneficial effects on neurodegenerative diseases in recent studies. In this study, we investigated the effects of Boswellia serrata resin on rotenone-induced dopaminergic neurotoxicity. Firstly, we attempted to see if the resin can induce AMP-activated protein kinase (AMPK) signaling pathway which has been known to have broad neuroprotective effects. Boswellia increased AMPK phosphorylation and reduced phosphorylation of mammalian target of rapamycin (p-mTOR) and α-synuclein (p-α-synuclein) in the striatum while increased the expression level of Beclin1, a marker for autophagy and brain-derived neurotrophic factor. Next, we examined the neuroprotective effects of the Boswellia extract in the rotenone-injected mice. The results showed that Boswellia evidently attenuated the loss of the nigrostriatal dopaminergic neurons and microglial activation caused by rotenone. Moreover, Boswellia ameliorated rotenone-induced decrease in the striatal dopamine and impairment in motor function. Accumulation of α-synuclein meditated by rotenone was significantly ameliorated by Boswellia. Also, we showed that ß-boswellic acid, the active constituents of Boswellia serrata gum, induced AMPK phosphorylation and attenuated α-synuclein phosphorylation in SHSY5 cells. These results suggest that Boswellia protected the dopaminergic neurons from rotenone neurotoxicity via activation of the AMPK pathway which might be associated with attenuation of α-synuclein aggregation and neuroinflammation. Further investigations are warranted to identify specific molecules in Boswellia which are responsible for the neuroprotection.


Assuntos
Boswellia , Fármacos Neuroprotetores , Síndromes Neurotóxicas , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Boswellia/metabolismo , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Mamíferos/metabolismo , Metanol/metabolismo , Metanol/farmacologia , Camundongos , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Síndromes Neurotóxicas/metabolismo , Extratos Vegetais/metabolismo , Extratos Vegetais/farmacologia , Rotenona/farmacologia , alfa-Sinucleína/metabolismo
7.
J Control Release ; 346: 1-19, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35398173

RESUMO

Parkinson's disease (PD) is a debilitating neurodegenerative condition characterized by the loss of dopaminergic neurons within the substantia nigra. The specific molecular mechanisms through which PD-associated neuronal loss occurs remain unclear, and there is no available effective treatment against PD-related neurodegeneration. Resveratrol (RSV) has exhibited promising neuroprotective effects via antioxidant and anti-inflammatory activity. However, its poor bioavailability in the brain represents a challenge for its application in PD treatment. In this study, we synthesized RSV-loaded PLGA nanoparticles (RSV-PLGA-NPs) conjugated with lactoferrin (Lf) to enhance RSV diffusion into the brain and assessed whether this formulation improved the neuroprotective effects of RSV in experimental PD models. The Lf-conjugated RSV-PLGA-NPs (Lf-RSV-PLGA-NPs) exhibited enhanced internalization into SH-SY5Y and human brain microvascular endothelial cells as compared to RSV-PLGA-NPs and free RSV. Further, Lf-RSV-PLGA-NPs were more effective than RSV-PLGA-NPs and free RSV in attenuating the MPP+-induced generation of reactive oxygen species, reduction of mitochondrial membrane potential, and cell death. Importantly, Lf conjugation specifically increased the accumulation of RSV-PLGA-NPs in the brain as determined via bioluminescent imaging analyses. Our formulation substantially enhanced the neuroprotective effects of RSV in the MPTP-induced PD model. Hence, Lf-RSV-PLGA-NPs represent a promising tool for improving RSV bioavailability and neuroprotection within the brain.


Assuntos
Nanopartículas , Neuroblastoma , Fármacos Neuroprotetores , Doença de Parkinson , Barreira Hematoencefálica , Células Endoteliais , Humanos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Resveratrol
8.
Antioxid Redox Signal ; 37(13-15): 1030-1050, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35286219

RESUMO

Aims: Raloxifene, a selective estrogen receptor (ER) modulator, has been reported to exert the tumor-suppressive effects in both ER-positive and ER-negative cancer cells; however, the mechanisms underlying its ER-independent anti-cancer effects are poorly understood. The NLRP3 inflammasome, a critical component of the innate immune system, has recently received growing attention owing to its multifaceted roles in various aspects of cancer development. The present study aimed at examining the involvement of NLRP3 inflammasomes in the anti-breast cancer effects of raloxifene and its underlying mechanisms. Results: Raloxifene significantly inhibited the activation of NLRP3 inflammasomes in various breast cancer cell lines. Importantly, forced expression of a gain-of-function variant of NLRP3 rescued breast cancer cells from growth arrest by raloxifene, suggesting that the suppression of NLRP3 inflammasomes activation mediates the raloxifene-induced inhibition of breast cancer growth. Mechanistically, raloxifene suppressed NLRP3 inflammasomes activation by lowering the cellular levels of reactive oxygen species (ROS) through the modulation of redox signaling mediated via aryl hydrocarbon receptor (AhR)-nuclear factor erythroid 2-related factor 2 (Nrf2)-heme oxygenase-1 (HO-1) axis or the impaired generation of mitochondrial ROS in a mitophagy-dependent manner. Further, the blockage of AhR signaling or inhibition of mitophagy abolished the tumor-suppressive effect of raloxifene in a human breast tumor xenograft model. Innovation: We elucidate a novel molecular mechanism underlying the breast tumor suppressing effect of raloxifene. Conclusion: The results observed in this study suggest that the modulation of NLRP3 inflammasomes activation is a critical event in the inhibition of breast tumor growth by raloxifene. Antioxid. Redox Signal. 37, 1030-1050.


Assuntos
Neoplasias da Mama , Inflamassomos , Humanos , Feminino , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Mitofagia , Receptores de Hidrocarboneto Arílico , Espécies Reativas de Oxigênio/metabolismo , Cloridrato de Raloxifeno/farmacologia , Oxirredução , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo
9.
Biochem Pharmacol ; 192: 114719, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34352280

RESUMO

The neuropathological hallmark of Parkinson's disease (PD) is the preferential loss of dopaminergic neurons in the substantia nigra and presence of Lewy bodies in the dying neurons. Though specific molecular mechanisms for the neurodegeneration remains to be clarified, mitochondrial dysfunction and increased oxidative stress are major players associated with PD pathogenesis and these pathogenic mechanisms can be reproduced in cells and animals by application of various neurotoxins such as MPP+. In this study, we attempted to determine the neuroprotective effects of methylene blue (MB) against 1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity, and to elucidate its action mechanism. We observed that MB attenuated MPP+-induced apoptotic cell death in SH-SY5Y cells and the mescencephalic dopaminergic neurons. In addition, MB protected the cells against MPP+-induced oxidative stress and mitochondrial dysfunction as evidenced by restoration of mitochondrial complex I activity and ATP levels, and attenuation of oxidative stress. Moreover, we demonstrated that MB induced antioxidant molecules, and activated Nrf2 pathway through AKT activation. These results indicate that MB protects the neurons from MPP+-induced toxicity through activation of antioxidant system, thereby reducing the oxidative stress and mitochondrial impairment, implying the potential use of MB in the treatment of neurodegenerative diseases such as PD.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Azul de Metileno/farmacologia , Mitocôndrias/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Neuroproteção/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Herbicidas/toxicidade , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/agonistas , Neuroproteção/fisiologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/fisiologia
10.
Neurochem Int ; 148: 105120, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34197898

RESUMO

Oxidative stress and mitochondrial dysfunction are now widely accepted as the major factors involved in the pathogenesis of Parkinson's disease (PD). Rotenone, a commonly used environmental toxin also reproduces these principle pathological features of PD. Hence, it is used frequently to induce experimental PD in cells and animals. In this study, we evaluated the neuroprotective effects of metformin against rotenone-induced toxicity in SH-SY5Y cells. Metformin treatment clearly rescued these cells from rotenone-mediated cell death via the reduction of the cytosolic and mitochondrial levels of reactive oxygen species and restoration of mitochondrial function. Furthermore, metformin upregulated PGC-1α, the master regulator of mitochondrial biogenesis and key antioxidant molecules, including glutathione and superoxide dismutase. We demonstrated that the drug exerted its cytoprotective effects by activating nuclear factor erythroid 2-related factor 2 (Nrf2)/heme-oxygenase (HO)-1 pathway, which in turn, is dependent on AKT activation by metformin. Thus, our results implicate that metformin provides neuroprotection against rotenone by inhibiting oxidative stress in the cells by inducing antioxidant system via upregulation of transcription mediated by Nrf2, thereby restoring the rotenone-induced mitochondrial dysfunction and energy deficit in the cells.


Assuntos
Hipoglicemiantes/farmacologia , Metformina/farmacologia , Doenças Mitocondriais/prevenção & controle , Fator 2 Relacionado a NF-E2/genética , Proteína Oncogênica v-akt/genética , Estresse Oxidativo/efeitos dos fármacos , Rotenona/antagonistas & inibidores , Rotenona/toxicidade , Transdução de Sinais/efeitos dos fármacos , Desacopladores/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Espécies Reativas de Oxigênio/metabolismo
11.
Arch Pharm Res ; 44(7): 689-701, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34302237

RESUMO

Human immunodeficiency virus 1 (HIV-1) infection can cause several HIV-associated neurocognitive disorders a variety of neurological impairments characterized by the loss of cortical and subcortical neurons and decreased cognitive and motor function. HIV-1 gp120, the major envelope glycoprotein on viral particles, acts as a binding protein for viral entry and is known to be an agent of neuronal cell death. To determine the mechanism of HIV-1 gp120-induced memory dysfunction, we performed mouse intracerebroventricular (i.c.v.) infusion with HIV-1 gp120 protein (300 ng per mouse) and investigated memory impairment and amyloidogenesis. Infusion of the HIV-1 gp120 protein induced memory dysfunction, which was evaluated using passive avoidance and water maze tests. Infusion of HIV-1 gp120 induced neuroinflammation, such as the release of iNOS and COX-2 and the activation of astrocytes and microglia and increased the mRNA and protein levels of IL-6, ICAM-1, M-CSF, TIM, and IL-2. In particular, we found that the infusion of HIV-1 gp120 induced the accumulation of amyloid plaques and signs of elevated amyloidogenesis, such as increased expression of amyloid precursor protein and BACE1 and increased ß-secretase activity. Therefore, these studies suggest that HIV-1 gp120 may induce memory impairment through Aß accumulation and neuroinflammation.


Assuntos
Encéfalo/patologia , Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/complicações , Transtornos da Memória/virologia , Doenças Neuroinflamatórias/virologia , Proteínas Amiloidogênicas/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/virologia , Proteína gp120 do Envelope de HIV/administração & dosagem , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/metabolismo , HIV-1/patogenicidade , Humanos , Infusões Intraventriculares , Masculino , Transtornos da Memória/imunologia , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos ICR , Doenças Neuroinflamatórias/imunologia , Doenças Neuroinflamatórias/patologia
12.
Bioorg Chem ; 106: 104493, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33268010

RESUMO

Suntamide A (1), a new cyclic peptide, was isolated from Cicadidae Periostracum. The gross structure of 1 was elucidated by detailed analysis of HRMS and 1D/2D NMR spectra, and the absolute configuration was established by C3 Marfey's method. We extended our study to examine biological activity of 1, and found that 1 protected SH-SY5Y cells against rotenone-induced neurotoxicity. This effect of 1 seemed to be attributed to antioxidant induction and protection of mitochondria from rotenone-caused injury. Along with augmentation of the antioxidant system by 1, there was an evident activation of Nrf2, a transcription factor involved in the activation of the antioxidant system. These results indicate that 1 rescued the cells from rotenone-mediated neurotoxicity by enhancing antioxidant capacity via induction of Nrf2, suggesting that the compound could be used as a therapeutic intervention in neurodegenerative diseases such as Parkinson's disease.


Assuntos
Antioxidantes/farmacologia , Hemípteros/química , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/farmacologia , Peptídeos Cíclicos/farmacologia , Animais , Antioxidantes/química , Antioxidantes/isolamento & purificação , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estrutura Molecular , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/isolamento & purificação , Peptídeos Cíclicos/química , Peptídeos Cíclicos/isolamento & purificação , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Rotenona/antagonistas & inibidores , Rotenona/farmacologia , Relação Estrutura-Atividade , Células Tumorais Cultivadas
14.
Biochem Pharmacol ; 180: 114193, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32800853

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disease, which is characterized by the progressive loss of dopaminergic neurons in the substantia nigra, leading to a decrease in striatal dopamine. There is no antiparkinsonian therapy that offers a true disease-modifying treatment till date and there is an urgent need for a safe and effective neuroprotective or neurorestorative therapy. Our previous study demonstrated that metformin upregulated dopamine in the mouse brain and provided significant neuroprotection in animal model of PD. Therefore, we designed this study to investigate the molecular mechanism underlying such pharmacological effect of metformin. Herein, we found that metformin enhanced the phosphorylation of tyrosine hydroxylase (TH) which was accompanied by increase in brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and activation of their downstream signaling pathways in the mouse brain and SH-SY5Y cells. We further investigated the role of the neurotrophic factors in the activation of TH and observed that both BDNF and GDNF-induction were essential for metformin-induced TH activation. We found that the AMPK/aPKCζ/CREB pathway was essential for metformin-induced GDNF upregulation and TH activation. Thus, this study reveals the TH-activating property of metformin in the brain via induction of neurotrophic factors along with the signaling mechanism. These results potentiate the candidacy of metformin not only as a neuroprotective agent, but also as restorative therapy for the treatment of PD.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dopamina/biossíntese , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Metformina/farmacologia , Proteína Quinase C/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Hipoglicemiantes/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
15.
Biochim Biophys Acta Gen Subj ; 1864(8): 129631, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32418902

RESUMO

BACKGROUND: AMP-activated protein kinase (AMPK) exerts its anti-inflammatory effects by suppressing redox-sensitive nuclear factor kappa B (NF-κB) and pro-inflammatory cytokines including TNF-α. However, it is unclear whether AMPK regulates anti-inflammatory cytokine expressions in the presence of oxidative stress-induced inflammation. We sought to elucidate the mechanisms whereby AMPK regulates inflammatory cytokine expressions under NADPH oxidase (NOX)-induced oxidative stress. METHODS: HT-29 human colonic epithelial cells transfected with AMPKα shRNA and mouse models with AMPKα knocked out in epithelial cells (AMPKαfl/fl-Vil-Cre) or macrophages (AMPKαfl/fl-Lyz2-Cre) were used to examine the effects of AMPK and NOX on signaling pathways and cytokine expressions. RESULTS: In HT-29 cells, 5-hydroxytryptamine (5-HT)-induced NOX activity was enhanced by AMPKα silencing, and resulted in inflammatory cell death. AMPKα deletion specific for colon epithelial cells (AMPKαfl/fl-Vil-Cre) or macrophages (AMPKαfl/fl-Lyz2-Cre) intensified 5-HT- or dextran sulfate sodium (DSS)-induced upregulations of NOX2, TNF-α, and IL-6, but completely abolished basal and 5-HT- or DSS-induced upregulation of IL-10 in colon epithelium. Furthermore, 5-HT- and DSS-induced changes were accompanied by marked upregulations of increased inflammatory signaling pathways linked to NF-κB, AP-1, and STAT3 transcription factors, and to GATA, a cell fate-directing signaling. In addition, AMPKα deletion significantly fortified 5-HT- or DSS-induced downregulations of cytoprotective signaling pathways (Nrf2, HIF-1α, and KLF4). CONCLUSION: Basal AMPKα maintains an anti-inflammatory state by inhibiting NOX, balancing pro-/anti-inflammatory signaling pathways, and directing IL-10 production. When these regulatory roles of AMPK are diminished by oxidative stress, colon epithelium undergoes inflammation despite IL-10 production.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Inflamação/metabolismo , Interleucina-10/biossíntese , Proteínas Quinases Ativadas por AMP/genética , Inativação Gênica , Células HT29 , Humanos , Fator 4 Semelhante a Kruppel , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , Transdução de Sinais
16.
Bioorg Med Chem Lett ; 30(11): 127145, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32249119

RESUMO

Two new chlorinated secondary metabolites, saccharochlorines A and B (1 and 2), were isolated from the saline cultivation of a marine-derived bacterium Saccharomonospora sp. (KCTC-19160). The chemical structures of the saccharochlorines were elucidated by 2D NMR and MS spectroscopic data. Saccharochlorines A and B (1 and 2) exhibit weak inhibition of ß-secretase (BACE1) in biochemical inhibitory assay, but they induced the release of Aß (1-40) and Aß (1-42) in H4-APP neuroglial cells. This discrepancy might be derived from the differences between the cellular and sub-cellular environments or the epigenetic stimulation of BACE1 expression.


Assuntos
Acrilatos/química , Actinobacteria/química , Acrilatos/isolamento & purificação , Acrilatos/metabolismo , Acrilatos/farmacologia , Actinobacteria/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Conformação Molecular , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Fragmentos de Peptídeos/metabolismo
17.
Stem Cell Res Ther ; 11(1): 63, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-32127052

RESUMO

BACKGROUND: Multiple system atrophy (MSA) is a sporadic neurodegenerative disorder of unknown etiology, but is closely associated with damage to dopaminergic neurons. MSA progression is rapid. Hence, long-term drug treatments do not have any therapeutic benefits. We assessed the inhibitory effect of mesenchymal stem cells (MSCs) on double-toxin-induced dopaminergic neurodegenerative MSA. RESULTS: Behavioral disorder was significantly improved and neurodegeneration was prevented following MSC transplantation. Proteomics revealed lower expression of polyamine modulating factor-binding protein 1 (PMFBP1) and higher expression of 3-hydroxymethyl-3-methylglutaryl-CoA lyase (HMGCL), but these changes were reversed after MSC transplantation. In the in vitro study, the 6-OHDA-induced effects were reversed following co-culture with MSC. However, PMFBP1 knockdown inhibited the recovery effect due to the MSCs. Furthermore, HMGCL expression was decreased following co-culture with MSCs, but treatment with recombinant HMGCL protein inhibited the recovery effects due to MSCs. CONCLUSIONS: These data indicate that MSCs protected against neuronal loss in MSA by reducing polyamine- and cholesterol-induced neural damage.


Assuntos
Células da Medula Óssea/metabolismo , Colesterol/efeitos adversos , Células-Tronco Mesenquimais/metabolismo , Atrofia de Múltiplos Sistemas/prevenção & controle , Atrofia de Múltiplos Sistemas/terapia , Poliaminas/efeitos adversos , Animais , Humanos , Masculino , Atrofia de Múltiplos Sistemas/patologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar
18.
Cancers (Basel) ; 12(3)2020 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-32155890

RESUMO

Adiponectin, an adipokine predominantly derived from adipose tissue, exhibits potent antitumor properties in breast cancer cells. However, its mechanisms of action remain elusive. Inflammasomes-intracellular multimeric protein complexes-modulate cancer cell growth in a complicated manner, as well as playing a role in the innate immune system. Herein, we examined the potential role of inflammasomes in the antitumor activity of adiponectin and found that globular adiponectin (gAcrp) significantly suppressed inflammasomes activation in breast cancer cells both in vitro and in vivo conditions, as determined by decreased expression of inflammasomes components, including NOD-like receptor pyrin domain-containing protein 3 (NLRP3) and the apoptosis-associated speck-like protein containing a CARD (ASC), and inhibition of interleukin-1ß and caspase-1 activation. Treatment with pharmacological inhibitors of inflammasomes caused decrease in cell viability, apoptosis induction, and G0/G1 cell cycle arrest, suggesting that inflammasomes activation is implicated in the growth of breast cancer cells. In addition, treatment with gAcrp generated essentially similar results to those of inflammasomes inhibitors, further indicating that suppression of breast cancer cell growth by gAcrp is mediated via modulation of inflammasomes. Mechanistically, gAcrp suppressed inflammasomes activation through sestrin2 (SESN2) induction, liver kinase B1 (LKB-1)-dependent AMP-activated protein kinase (AMPK) phosphorylation, and alleviation of endoplasmic reticulum (ER) stress. Taken together, these results demonstrate that gAcrp inhibits growth of breast cancer cells by suppressing inflammasomes activation, at least in part, via SESN2 induction and AMPK activation-dependent mechanisms.

19.
Exp Neurol ; 323: 113082, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31669069

RESUMO

Chitinase 3-like 1 (Chi3L1) plays a major role in the pathogenesis of inflammatory diseases. We investigated the effect of Chi3L1 knockout on stroke development. Ischemia/reperfusion was induced by middle cerebral artery occlusion (MCAO) in Chi3L1 knockout and wildtype mice. Significantly increased infarct volume and decreased neurological deficit scores at 24 h after ischemia/reperfusion were found in Chi3L1 knockout mice compared to wildtype mice. Moreover, ischemic neuronal cell death was increased in Chi3L1 knockout mice through increased oxidative stress and release of IL-6 and IL-1ß but IL-10 and IL-4 were reduced. Furthermore, expression of inflammation-related proteins (iNOS, COX-2, Iba-1, and GFAP) was significantly increased in Chi3L1 knockout mice compared to wildtype. In microglia isolated from MCAO-injured Chi3L1 knockout mice, expression of M1 markers (iNOS, CD86, IL-1ß, and IL-6) was increased and M2 markers (Arg1, Mrc1, IL-10, and IL-4Ra) was decreased. In BV-2 cells, knockdown of Chi3L1 increased TNF-α- and INF-γ-induced expression of iNOS, COX-2, and Iba-1, but decreased the expression of Arg1, MRC1, and IL-4 receptor-alpha (IL-4Rα). Expression of IL-4Rα, an important factor of M2 polarization, and its downstream signals p-JAK1, p-JAK3, and p-STAT6, was much reduced in the knockout mice. Additionally, in BV-2 cells, knockdown of Chi3L1 by siRNA Chi3L1 decreased rhTNF-α- and INF-γ-induced expression of IL-4Rα, p-JAK1, p-JAK3, and p-STAT6. Furthermore, treatment with AS1517499 abolished Chi3L1 knockdown-induced reduced IL-4Rα and Arg1 but not CD86 expression. Our results indicate that deletion of Chi3L1 accelerates stroke development through enhancement of neuroinflammation by markedly decreasing STAT6-dependent M2 macrophage polarization.


Assuntos
Proteína 1 Semelhante à Quitinase-3/deficiência , Inflamação/metabolismo , Microglia/metabolismo , Fator de Transcrição STAT6/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Camundongos , Camundongos Knockout , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/patologia
20.
Transl Neurodegener ; 8: 26, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31592103

RESUMO

BACKGROUND: Neuroinflammation and accumulation of ß-amyloid (Aß) play a significant role in the onset and progression of Alzheimer's disease (AD). Our previous study demonstrated that signal transducer and activator of transcription-3 (STAT3) plays a major role in neuroinflammation and amyloidogenesis. METHODS: In the present study, we investigated the inhibitory effect of bee venom phospholipase A2 (bvPLA2) on memory deficiency in Tg2576 mice, which demonstrate genetic characteristics of AD and the mechanism of its action at the cellular and animal level. For in vivo study, we examined the effect of bvPLA2 on improving memory by conducting several behavioral tests with the administration of bvPLA2 (1 mg/kg) to Tg2576 mice. For in vitro study, we examined the effect of bvPLA2 on amyloidogenesis and neuroinflammation by treating bvPLA2 on LPS-activated BV2 cells. RESULTS: We found that bvPLA2 alleviated memory impairment in Tg2576 mice, as demonstrated in the behavioral tests assessing memory. In the bvPLA2-treated group, Aß, amyloid precursor protein (APP), and ß-secretase 1 (BACE1) levels and ß-secretase activity were significantly decreased. Expression of pro-inflammatory cytokines and inflammation-related proteins decreased in the brain of bvPLA2-treated group, whereas anti-inflammatory cytokines increased. In addition, bvPLA2 reduced STAT3 phosphorylation in the brains of the bvPLA2-treated group. At the cellular level, bvPLA2 inhibits production of nitric oxide, pro-inflammatory cytokines, and inflammation-related proteins including p-STAT3. Additionally, bvPLA2 inhibits the production of Aß in cultured BV-2 cells. Results from the docking experiment, pull-down assay, and the luciferase assay show that bvPLA2 directly binds STAT3 and, thus, regulates gene expression levels. Moreover, when the STAT3 inhibitor and bvPLA2 were administered together, the anti-amyloidogenic and anti-inflammatory effects were further enhanced than when they were administered alone. CONCLUSION: These results suggest that bvPLA2 could restore memory by inhibiting the accumulation of Aß and inflammatory responses via blockage of STAT3 activity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...