Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Toxicol In Vitro ; 89: 105564, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36736710

RESUMO

Epidemiological studies indicate that human and animal exposure to environmental mercury (Hg) disrupts normal immune system function, but the molecular mechanism responsible for this is still unresolved. We have previously utilized phospho-proteomic mass spectrometry to demonstrate that in the absence of B Cell Receptor (BCR) stimulation, exposure of B cells to Hg induces significant changes to a great many elements of the BCR signaling pathway in a concentration dependent manner. In this report, we have extended those initial findings by utilizing mass spectrometry to evaluate in detail the effect of low-level Hg exposure on BCR induced phospho-proteomic changes. Specifically, murine WEHI-231 B lymphoma cells were exposed to environmentally relevant levels of Hg with or without concomitant BCR stimulation. The cellular phospho-proteomes were then profiled by LC-MS/MS. We found that for low-level exposures, Hg interference with signal transduction across the BCR pathway was predominantly associated with modification of phosphorylation of 12 phosphosites located on seven different proteins. Nine sites were serine, two sites tyrosine and one site threonine. Most of these sites are novel, in the sense that only the two tyrosine and one of the serine sites have previously been reported to be associated with BCR signaling.


Assuntos
Mercúrio , Animais , Camundongos , Humanos , Fosfosserina/metabolismo , Fosfosserina/farmacologia , Mercúrio/toxicidade , Cromatografia Líquida , Proteômica , Linhagem Celular , Espectrometria de Massas em Tandem , Transdução de Sinais , Receptores de Antígenos de Linfócitos B/metabolismo , Proteínas/metabolismo , Fosforilação , Tirosina/metabolismo
2.
Cancer Commun (Lond) ; 42(3): 205-222, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35191227

RESUMO

BACKGROUND: Understanding how the tumor microenvironment is shaped by various factors is important for the development of new therapeutic strategies. Tumor cells often undergo spontaneous apoptotic cell death in tumor microenvironment, these apoptotic cells are histologically co-localized with immunosuppressive macrophages. However, the mechanism by which tumor cell apoptosis modulates macrophage polarization is not fully understood. In this study, we aimed to explore the tumor promoting effects of apoptotic tumor cells and the signal pathways involved. METHODS: Apoptotic cells and macrophages in tumors were detected by immunohistochemical staining. Morphological analysis was performed with Giemsa staining. Lipids generated from apoptotic cells were detected by liquid chromatography-mass spectrometry. Phosphatidylserine-containing liposomes were prepared to mimic apoptotic cells. The expression of protein was determined by real-time PCR, immunohistochemistry enzyme-linked immunosorbent assay and Western blotting. Mouse malignant ascites and subcutaneous tumor models were designed for in vivo analysis. Transgenic mice with specific genes knocked out and inhibitors specific to certain proteins were used for the mechanistic studies. RESULTS: The location and the number of apoptotic cells were correlated with that of macrophages in several types of carcinomas. Phosphatidylserine, a lipid molecule generated in apoptotic cells, induced polarization and accumulation of M2-like macrophages in vivo and in vitro. Moreover, sustained administration of phosphoserine promoted tumor growth in the malignant ascites and subcutaneous tumor models. Further analyses suggested that phosphoserine induced a M2-like phenotype in macrophages, which was related to the activation of phosphoserine receptors including T-cell immunoglobin mucin 4 (TIM4) and the FAK-SRC-STAT3 signaling pathway as well as elevated the expression of the histone demethylase Jumonji domain-containing protein 3 (JMJD3). Administration of specific inhibitors of these pathways could reduce tumor progression. CONCLUSIONS: This study suggest that apoptotic cell-generated phosphoserine might be a notable signal for immunosuppressive macrophages in tumors, and the related pathways might be potential therapeutic targets for cancer therapy.


Assuntos
Neoplasias , Fosfatidilserinas , Animais , Apoptose , Ascite/metabolismo , Histona Desmetilases com o Domínio Jumonji , Macrófagos/metabolismo , Camundongos , Neoplasias/metabolismo , Fosfatidilserinas/metabolismo , Fosfatidilserinas/farmacologia , Fosfosserina/metabolismo , Fosfosserina/farmacologia , Fator de Transcrição STAT3/metabolismo , Microambiente Tumoral
3.
PLoS One ; 13(9): e0203211, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30192865

RESUMO

BACKGROUND: Sphingosine kinase phosphorylates sphingosine to generate sphingosine 1 phosphate (S1P) following stimulation of the five plasma membrane G-protein-coupled receptors. The objective of this study is to clarify the role of S1P and its receptors (S1PRs), especially S1PR3 in airway epithelial cells. METHODS: The effects of S1P on asthma-related genes expression were examined with the human bronchial epithelial cells BEAS-2B and Calu-3 using a transcriptome analysis and siRNA of S1PRs. To clarify the role of CCL20 in the airway inflammation, BALB/c mice were immunized with ovalbumin (OVA) and subsequently challenged with an OVA-containing aerosol to induce asthma with or without intraperitoneal administration of anti-CCL20. Finally, the anti-inflammatory effect of VPC 23019, S1PR1/3 antagonist, in the OVA-induced asthma was examined. RESULTS: S1P induced the expression of some asthma-related genes, such as ADRB2, PTGER4, and CCL20, in the bronchial epithelial cells. The knock-down of SIPR3 suppressed the expression of S1P-inducing CCL20. Anti-CCL20 antibody significantly attenuated the eosinophil numbers in the bronchoalveolar lavage fluid (P<0.01). Upon OVA challenge, VPC23019 exhibited substantially attenuated eosinophilic inflammation. CONCLUSIONS: S1P/S1PR3 pathways have a role in release of proinflammatory cytokines from bronchial epithelial cells. Our results suggest that S1P/S1PR3 may be a possible candidate for the treatment of bronchial asthma.


Assuntos
Brônquios/imunologia , Brônquios/metabolismo , Quimiocina CCL20/metabolismo , Lisofosfolipídeos/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Anti-Inflamatórios/farmacologia , Asma/tratamento farmacológico , Asma/genética , Asma/metabolismo , Brônquios/patologia , Linhagem Celular , Modelos Animais de Doenças , Eosinofilia/tratamento farmacológico , Eosinofilia/patologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Receptores Adrenérgicos beta 2/genética , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Receptores de Prostaglandina E Subtipo EP4/genética , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato
4.
J Neurochem ; 140(5): 776-786, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28054340

RESUMO

Fingolimod, a sphingosine-1-phosphate receptor (S1PR) agonist, is clinically available to treat multiple sclerosis and is showing promise in treating stroke. We investigated if fingolimod provides long-term protection from experimental neonatal germinal matrix hemorrhage (GMH), aiming to support a potential mechanism of acute fingolimod-induced protection. GMH was induced in P7 rats by infusion of collagenase (0.3 U) into the right ganglionic eminence. Animals killed at 4 weeks post-GMH received low- or high-dose fingolimod (0.25 or 1.0 mg/kg) or vehicle, and underwent neurocognitive testing before histopathological evaluation. Subsequently, a cohort of animals killed at 72 h post-GMH received 1.0 mg/kg fingolimod; the specific S1PR1 agonist, SEW2871; or fingolimod co-administered with the S1PR1/3/4 inhibitor, VPC23019, or the Rac1 inhibitor, EHT1864. All drugs were injected intraperitoneally 1, 24, and 48 h post-surgery. At 72 h post-GMH, brain water content, extravasated Evans blue dye, and hemoglobin were measured as well as the expression levels of phospho-Akt, Akt, GTP-Rac1, Total-Rac1, ZO1, occludin, and claudin-3 determined. Fingolimod significantly improved long-term neurocognitive performance and ameliorated brain tissue loss. At 72 h post-GMH, fingolimod reduced brain water content and Evans blue dye extravasation as well as reversed GMH-induced loss of tight junctional proteins. S1PR1 agonism showed similar protection, whereas S1PR or Rac1 inhibition abolished the protective effect of fingolimod. Fingolimod treatment improved functional and morphological outcomes after GMH, in part, by tempering acute post-hemorrhagic blood-brain barrier disruption via the activation of the S1PR1/Akt/Rac1 pathway.


Assuntos
Cloridrato de Fingolimode/farmacologia , Hemorragias Intracranianas/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Água Corporal/metabolismo , Encéfalo/patologia , Química Encefálica/efeitos dos fármacos , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Cognição/efeitos dos fármacos , Feminino , Hemorragias Intracranianas/metabolismo , Hemorragias Intracranianas/psicologia , Contagem de Leucócitos , Masculino , Oxidiazóis/farmacologia , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Gravidez , Pironas/farmacologia , Quinolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Tiofenos/farmacologia , Proteínas de Junções Íntimas/metabolismo , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores
5.
Biochem Biophys Res Commun ; 476(4): 627-634, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27282481

RESUMO

Endothelial nitric oxide synthase (eNOS) plays a crucial role in vascular homeostasis. Lysophospholipid interaction with sphingosine 1-phosphat (S1P) receptors results in eNOS activation in different cells. In endothelial cells, eNOS activation via S1P1 or S1P3 was shown controversially. The aim of this study is to investigate the meaning of both S1P receptors for eNOS activation in human endothelial cells. Therefore, several S1P1 and S1P3 agonists in combination with antagonists and specific RNAi approach were used. eNOS activation was measured in human umbilical vein endothelial cells (HUVEC) via DAF2-DA-based fluorescence microscopy. For investigation of the signaling pathway, agonists/antagonist studies, RNAi approach, Luminex™ multiplex, and Western Blot were used. In HUVEC, both the S1P1 agonist AUY954 as well as the S1P1,3 agonist FTY720P induced eNOS activation in a time- and dose-dependent manner. Other S1P1 agonists activated eNOS to a lesser extent. The AUY954-induced eNOS activation was blocked by the S1P1 antagonist W146, the combination of W146 and the S1P3 antagonist CAY10444 and the S1P1,3 antagonist VPC23019, but not by CAY10444 indicating the meaning of S1P1 for the AUY954-induced eNOS activation. The FTY720P-induced eNOS activation was blocked only by the combination of W146 and CAY10444 and the combined S1P1,3 antagonist VPC23019, but not by W146 or CAY10444 indicating the importance of both S1P1 and S1P3 for FTY720-induced eNOS activation. These results were confirmed using specific siRNA against S1P1 and S1P3. The S1P1,3 activation results in Akt phosphorylation and subsequent activation of eNOS via phosphorylation at serine(1177) and dephosphorylation at threonine(495). Beside former investigations with rather unspecific S1P receptor activation these data show potent selective S1P1 activation by using AUY954 and with selective S1P receptor inhibition evidence was provided that both S1P1 and S1P3 lead to downstream activation of eNOS in HUVEC in the same experimental setting. Inhibition or knockdown of one of these receptor subtypes did not abolish the eNOS activation and subsequent NO production.


Assuntos
Células Endoteliais/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Anilidas/farmacologia , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo III/química , Organofosfatos/farmacologia , Organofosfonatos/farmacologia , Fosforilação , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato , Tiazolidinas/farmacologia , Tiofenos/farmacologia , beta-Alanina/análogos & derivados , beta-Alanina/farmacologia
6.
J Pharm Sci ; 103(11): 3457-3463, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25266204

RESUMO

The clinical use of therapeutic proteins can be complicated by the development of anti-product antibodies. We have previously observed that O-phospho-l-serine (OPLS) reduced antibody response to FVIII in Hemophilia-A (HA) mice. However, the mechanism underlying this observation is not clear. We hypothesize that OPLS reduces immunogenicity by inducing tolerogenic properties in dendritic cells (DCs). We tested this hypothesis using in vivo, in vitro, and ex vivo methods. Naive HA mice that were pre-exposed to FVIII in the presence of OPLS showed substantially lower antibody response following rechallenge with OPLS free FVIII as compared with dexamethasone-pretreated mice. Exposure of OPLS to bone-marrow-derived dendritic cells (BMDCs) in culturing conditions resulted in an increase in the regulatory cytokine TGF-ß and a decrease in proinflammatory cytokines TNF-α and IL12p70. This was accompanied by a significant reduction in upregulation of costimulatory marker CD40, as measured by flow cytometry. Furthermore, ex vivo matured BMDCs in the presence of FVIII and OPLS failed to elicit a robust immune response in HA mice compared with FVIII-treated BMDCs. Our data suggest that OPLS modulates the immune response by altering the function and maturation of DCs, resulting in the induction of tolerogenic properties. © 2014 Wiley Periodicals, Inc. and the American Pharmacists Association J Pharm Sci 103:3457-3463, 2014.


Assuntos
Adjuvantes Imunológicos/farmacologia , Coagulantes/farmacologia , Células Dendríticas/efeitos dos fármacos , Fator VIII/farmacologia , Hemofilia A/tratamento farmacológico , Tolerância Imunológica/efeitos dos fármacos , Fosfosserina/farmacologia , Animais , Anticorpos/sangue , Antígenos CD40/metabolismo , Células Cultivadas , Coagulantes/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Fator VIII/imunologia , Hemofilia A/sangue , Hemofilia A/genética , Hemofilia A/imunologia , Interleucina-12/metabolismo , Camundongos Transgênicos , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Tissue Eng Part A ; 20(3-4): 474-85, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24229073

RESUMO

Calcium phosphates (CaP) are considered as biomaterials of choice for the treatment of critical-sized bone defects. Novel injectable CaP materials integrating poly(epsilon-lysine) generation 3 dendrons tethered with phosphoserine were obtained by sol-gel synthesis. This type of dendron was integrated to mimic the biochemical structure of noncollagenous proteins present in the forming osteoids during bone repair. Sol-gel synthesis was coupled with a dialysis process able to equilibrate the materials at a physiological pH value. Fourier transform infrared spectroscopy (FTIR) showed the successful retention of the dendrons after gel dialysis, whereas X-ray diffraction analysis demonstrated both the pH-tuned formation of a hydroxyapatite crystalline phase within the gel and the complete removal of ammonium nitrate deriving from the sol-gel reaction solvent. Scanning electron microscopy images confirmed the presence of crystalline domains in gels synthesized at pH 9.0. Injectability tests showed that the optimized formulations fulfilled the rheological properties required to minimally invasive surgical procedures. Cytotoxicity tests on osteoblast-like MG-63 cells as well as morphology and viability studies showed that the dendrons induced a significantly higher level of cell proliferation at early incubation time. Differentiation of the cell was also clearly enhanced at longer incubation time as demonstrated by both alkaline phosphatase activity and expression of typical markers. Altogether, the data from this work indicate the clinical potential of the osteoid-mimicking CaP cements in minimally invasive bone surgery.


Assuntos
Fosfatos de Cálcio/farmacologia , Diferenciação Celular/efeitos dos fármacos , Dendrímeros/farmacologia , Géis/farmacologia , Células-Tronco Mesenquimais/citologia , Fosfosserina/farmacologia , Polilisina/farmacologia , Fosfatase Alcalina/metabolismo , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Géis/síntese química , Géis/química , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Injeções , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/enzimologia , Oxazinas/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X , Xantenos/metabolismo
8.
Cell Biol Int ; 38(3): 309-17, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24155130

RESUMO

Phosphoserine has potential effectiveness as a simple substrate in preparing bone replacement materials, which could enhance bone forming ability. However, there is a need to investigate the independent effect of phosphoserine on osteogenic differentiation of human adipose stem cells (hADSCs). hADSCs were cultured in an osteogenic medium with phosphoserine. Cell proliferation was analysed by CCK8 and osteogenic differentiation was measured by alkaline phosphatase (ALP) activity, von Kossa staining and real time-polymerase chain reaction (RT-PCR). No stimulatory effect of phosphoserine on cell proliferation was noted at Days 1, 4 and 7. Deposition of calcium increased after the addition of phosphoserine. mRNA expression of type I collagen (COL-I), alkaline phosphatase (ALP), osteocalcin (OCN), Osterix, bone morphogenetic protein-2 (BMP-2) and RUNX2 increased markedly with phosphoserine treatment. The BMP-2 antagonist, noggin, and its receptor kinase inhibitors, dorsomorphin and LDN-193189, attenuated phosphoserine-promoted ALP activity. BMP-responsive and Runx2-responsive reporters were activated by phosphoserine treatment. Thus phosphoserine can promote osteogenic differentiation of hADSCs, probably by activating BMP and Runx2 pathways, which could be a promising approach for enhancing osteogenic capacity of cell-based construction in bone tissue engineering.


Assuntos
Adipócitos/efeitos dos fármacos , Proteína Morfogenética Óssea 2/metabolismo , Osteogênese/efeitos dos fármacos , Fosfosserina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Adipócitos/citologia , Adulto , Fosfatase Alcalina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Feminino , Humanos , Masculino , Osteocalcina/metabolismo , Células Estromais/citologia
9.
Neuropharmacology ; 62(2): 815-24, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21933676

RESUMO

Although the neuromodulator adenosine plays an important role in many central nervous system physiological and pathological processes, the properties and mechanisms of extracellular adenosine production are still unclear. In previous work, we determined that two forms of adenosine release can be evoked in the molecular layer of the cerebellum: one independent of ionotropic glutamate receptor activation (evoked by a train of stimuli) and one mainly dependent on the activation of ionotropic glutamate receptors (evoked by a single stimulus in 4-aminopyridine). Here we have investigated how these different forms of adenosine release are modulated by metabotropic receptors (A(1), GABA(B) and mGlu4). Although both types of adenosine release are inhibited by the activation of metabotropic receptors, single stimulus-evoked release was much more potently inhibited suggesting differential coupling between receptors and adenosine release mechanisms. Metabotropic receptor antagonists revealed that endogenous A(1) receptor activation plays the major role in controlling adenosine release and determine the relationship between stimulus strength and adenosine release. The major mechanism of modulation is through control of ionotropic glutamate receptor activation with block of metabotropic receptors inducing glutamate receptor-dependent adenosine release. In contrast to metabotropic receptor agonists, which inhibit adenylyl cyclase, activation of adenylyl cyclase (with forskolin) increased both glutamate receptor-dependent and independent adenosine release. This is the first time that the control of adenosine release by endogenous modulators has been studied and like classical neurotransmitters, adenosine release is controlled by an interplay of presynaptic modulators. This article is part of a Special Issue entitled 'Post-Traumatic Stress Disorder'.


Assuntos
Adenosina/metabolismo , Cerebelo/metabolismo , Neurônios/metabolismo , Receptor A1 de Adenosina/metabolismo , Receptores de GABA-B/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Antagonistas do Receptor A1 de Adenosina/farmacologia , Animais , Cerebelo/efeitos dos fármacos , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Antagonistas de Receptores de GABA-B/farmacologia , Masculino , Neurônios/efeitos dos fármacos , Fosfosserina/farmacologia , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Tionucleotídeos/farmacologia
10.
Chem Biol ; 16(9): 928-36, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19778720

RESUMO

This overview focuses on the (alpha,alpha-difluoromethylene)phosphonate mimic of phosphoserine (pCF(2)Ser) and its application to the study of kinase-mediated signal transduction-pathways of great interest to drug development. The most versatile modes of access to these chemical biological tools are discussed, organized by method of PCF(2)-C bond formation. The pCF(2)-Ser mimic may be site-specifically incorporated into peptides (SPPS) and proteins (expressed protein ligation). This isopolar, dianionic pSer mimic results in a "constitutive phosphorylation" phenotype and is seen to support native protein-protein interactions that depend on serine phosphorylation. Signal transduction pathways studied with this chemical biological approach include the regulation of p53 tumor suppressor protein activity and of melatonin production. Given these successes, the future is bright for the use of such "teflon phospho-amino acid mimics" to map kinase-based signaling pathways.


Assuntos
Ácido Clodrônico/análogos & derivados , Fosfosserina/química , Transdução de Sinais , Ácido Clodrônico/síntese química , Ácido Clodrônico/química , Ácido Clodrônico/farmacologia , Sistema de Sinalização das MAP Quinases , Melatonina/metabolismo , Fosforilação , Fosfosserina/síntese química , Fosfosserina/farmacologia , Domínios e Motivos de Interação entre Proteínas , Proteína Supressora de Tumor p53/metabolismo
11.
Aging Cell ; 7(1): 47-57, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18005252

RESUMO

This study aims to investigate the roles of the protein kinase A (PKA)- and caspase-dependent pathways in amyloid beta-peptide 31-35 (Abeta[31-35])-induced apoptosis, and the mechanisms of neuroprotection by group III metabotropic glutamate receptor (mGluR) activation against apoptosis induced by Abeta[31-35] in cortical neurons. We demonstrated that Abeta[31-35] induces neuronal apoptosis as well as a significant increase in caspase-3, -8 and -9. Activation of group III mGluRs by l-serine-O-phosphate and (R,S)-4-phosphonophenylglycine (two group III mGluR agonists), which attenuate the effects of Abeta[31-35], provides neuroprotection to the cortical neurons subjected to Abeta[31-35]. We also showed that Rp-cAMP, an inhibitor of cAMP-dependent PKA, has the ability to protect neurons from Abeta[31-35]-induced apoptosis and to reverse almost completely the effects of Abeta[31-35] on the activities of caspase-3. Further, we found that Sp-cAMP, an activator of cAMP-dependent PKA, can significantly abolish the l-serine-O-phosphate- and (R,S)-4-phosphonophenylglycine-induced neuroprotection against apoptosis, and decrease caspase-3, -8 and -9 in the Abeta[31-35]-treated neurons. Our findings suggest that neuronal apoptosis induced by Abeta[31-35] is mediated by the PKA-dependent pathway as well as the caspase-dependent intrinsic and extrinsic apoptotic pathways. Activation of group III mGluRs protects neurons from Abeta[31-35]-induced apoptosis by blocking the caspase-dependent pathways. Inhibition of the PKA-dependent pathway might also protect neurons from Abeta[31-35]-induced apoptosis by blocking the caspase-dependent pathways. Taken together, our observations suggest that Abeta[31-35] might have the ability to activate PKA, which in turn activates the caspase-dependent intrinsic and extrinsic apoptotic pathways, inducing apoptosis in the cortical neurons.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Córtex Pré-Frontal/metabolismo , Animais , Caspase 3/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Glicina/análogos & derivados , Glicina/farmacologia , Marcação In Situ das Extremidades Cortadas , Neurônios/citologia , Neurônios/metabolismo , Fosfosserina/farmacologia , Córtex Pré-Frontal/citologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Transdução de Sinais
12.
Chem Biol ; 14(9): 1019-30, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17884634

RESUMO

High-throughput identification of small molecules that selectively modulate molecular, cellular, or systems-level properties of the mammalian brain is a significant challenge. Here we report the chemical genetic identification of the orphan ligand phosphoserine (P-Ser) as an enhancer of neurogenesis. P-Ser inhibits neural stem cell/progenitor proliferation and self-renewal, enhances neurogenic fate commitment, and improves neuronal survival. We further demonstrate that the effects of P-Ser are mediated by the group III metabotropic glutamate receptor 4 (mGluR4). siRNA-mediated knockdown of mGluR4 abolished the effects of P-Ser and increased neurosphere proliferation, at least in part through upregulation of mTOR pathway activity. We also found that P-Ser increases neurogenesis in human embryonic stem cell-derived neural progenitors. This work highlights the tremendous potential of developing effective small-molecule drugs for use in regenerative medicine or transplantation therapy.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Neurônios/citologia , Fosfosserina/farmacologia , Receptores de Glutamato Metabotrópico/fisiologia , Células-Tronco/citologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Ligantes , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Fosfosserina/metabolismo , Proteínas Quinases/metabolismo , Células-Tronco/efeitos dos fármacos , Serina-Treonina Quinases TOR
13.
J Neurosci Res ; 84(2): 268-77, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16752416

RESUMO

Altered glial function that leads to oxidative stress and excitotoxicity may contribute to the initiation or progression of neuronal death in neurodegenerative diseases. We report the pivotal role of astroglial Group II and III metabotropic glutamate receptors (mGluR) against neurotoxicity. Activation of Group II or III mGluR on astrocytes with selective agonists DCG-IV or L-AP4 respectively inhibited astroglial lipopolysaccharide (LPS)-conditioned medium induced apoptosis of primary cultured mesencephalic neurons. Specific Group II or III mGluR antagonists APICA or MSOP completely abolished the neuroprotective effects of DCG-IV and L-AP4. Morphologic analysis showed that DCG-IV or L-AP4 could also attenuate the astroglial neurotoxicity to dopaminergic neurons. Measurement of extracellular glutamate concentration and [(3)H]-glutamate uptake showed that the restoration of glutamate uptake capability in LPS-treated astrocytes might be involved in the neuroprotective effects of activating astroglial Group II or III mGluR. Furthermore, we found that the repression of astroglial uptake function could be revived by GSH, and both Group II and III mGluR agonists could recover the endogenous reduced glutathione (GSH) level in LPS-treated astrocytes. These results suggested that the possible mechanisms of neuroprotection by either Type II or Type III mGluR activation may involve restoration of endogenous GSH, in turn affording recovery of astroglial capability to take up glutamate.


Assuntos
Astrócitos/efeitos dos fármacos , Encéfalo/metabolismo , Ácido Glutâmico/metabolismo , Lipopolissacarídeos/toxicidade , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Apoptose/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/efeitos dos fármacos , Células Cultivadas , Ciclopropanos/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Glutationa/metabolismo , Glicina/análogos & derivados , Glicina/farmacologia , Imuno-Histoquímica , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Fosfosserina/farmacologia , Propionatos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/efeitos dos fármacos
14.
J Agric Food Chem ; 54(3): 773-8, 2006 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-16448181

RESUMO

The protective effects of hen egg yolk phosvitin phosphopeptides (PPPs) against hydrogen peroxide (H2O2)-induced oxidative stress were evaluated in an in vitro assay using human intestinal epithelial cells. Caco-2 cells were stimulated with 1 mM H2O2 for 6 h, and the secretion of IL-8, a proinflammatory mediator, was determined by ELISA as a biomarker of oxidative stress. The inhibition of H2O2-induced IL-8 secretion from Caco-2 cells was observed by pretreatment for 2 h with PPPs, but not with phosvitin. PPPs also suppressed the formation of malondialdehyde in H2O2-treated Caco-2 cells. Furthermore, intracellular glutathione levels and glutathione reductase activity were elevated by the addition of PPPs. The protective effects of PPPs against H2O2-induced oxidative stress were almost the same as that of glutathione, and PPPs with a high content of phosphorus exhibited higher protective activity than PPPs without phosphorus; however, phosphoserine itself did not show any significant antioxidative stress activity. These findings suggest that oligophosphopeptides from hen egg yolk phosvitin possess novel antioxidative activity against oxidative stress in intestinal epithelial cells and that phosphorus and peptide structure seem to have a key role in the activity.


Assuntos
Gema de Ovo/química , Estresse Oxidativo/efeitos dos fármacos , Fosfopeptídeos/farmacologia , Fosvitina/química , Animais , Células CACO-2 , Galinhas , Feminino , Glutationa/análise , Glutationa Redutase/análise , Humanos , Peróxido de Hidrogênio/farmacologia , Interleucina-8/metabolismo , Malondialdeído/metabolismo , Fosfosserina/farmacologia , Fosvitina/farmacologia
15.
J Immunol ; 171(9): 4672-9, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14568942

RESUMO

Apoptotic cells are phagocytosed as soon as they appear in vivo. In this study, we first determined precisely at what stage apoptotic cells are phagocytosed by macrophages, and then examined the subsequent cytokine production. Phagocytosis was confirmed by flow cytometry and confocal laser microscopy, whereas the subsequent response was examined by ELISA and RT-PCR for quantitative and semiquantitative measurement of the protein and mRNA levels of cytokines, respectively. Even the cell populations containing very early apoptotic cells, such as IL-2-dependent CTLL-2 cells cultured in the absence of IL-2 for 4 h and a murine leukemic cell line, P388 cells, treated with etoposide for 5 h, were phagocytosed by macrophages. Although the cell populations containing the very early apoptotic cells used in this study were FITC-Annexin V-negative and did not show a decrease in cell size as compared with untreated cells, they showed a very small increase in phosphatidylserine on the cell surface, as detected with Cy3-Annexin V, and a decrease in mitochondrial membrane potential, indicating that the cell populations had already started the apoptotic process. Phagocytosis of such populations containing very early apoptotic cells was inhibited by phospho-L-serine much more significantly than Arg-Gly-Asp-Ser. In addition, macrophages hardly produced either proinflammatory or anti-inflammatory cytokines after phagocytosis, thus being an almost null response. These results are contrary to the generally accepted concept that the phagocytosis of apoptotic cells leads to the production of anti-inflammatory cytokines, suggesting instead that cells starting to undergo apoptosis are quickly phagocytosed by macrophages without any inflammation in vivo.


Assuntos
Apoptose/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Citocinas/biossíntese , Citocinas/genética , Humanos , Interleucina-8/antagonistas & inibidores , Interleucina-8/biossíntese , Células de Kupffer/citologia , Células de Kupffer/imunologia , Leucemia P388 , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos Alveolares/citologia , Macrófagos Alveolares/imunologia , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/imunologia , Camundongos , Monócitos/citologia , Monócitos/imunologia , Oligopeptídeos/farmacologia , Fagocitose/efeitos dos fármacos , Fosfosserina/farmacologia , RNA Mensageiro/biossíntese , Fatores de Tempo
16.
Pharmacol Toxicol ; 93(2): 71-6, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12899668

RESUMO

The effect of N-palmitoyl-L-serine phosphoric acid (L-NASPA), which has been used as an inhibitor of lysophosphatidic acid receptors, on intracellular Ca2+ concentration ([Ca2+]i) in human osteosarcoma MG63 cells was measured by using fura-2. L-NASPA (0.1-10 microM) caused a rapid and transient plateau [Ca2+]i rise in a concentration-dependent manner (EC50=0.5 microM). The L-NASPA-induced [Ca2+]i rise was partly reduced by removal of extracellular Ca2+ but was not altered by L-type voltage-gated Ca2+ channel blockers. In Ca2+-free medium, thapsigargin, an inhibitor of the endoplasmic reticulum Ca2+-ATPase, induced a [Ca2+]i rise, after which the increasing effect of L-NASPA on [Ca2+]i was completely inhibited; also, pretreatment with L-NASPA partly reduced thapsigargin-induced [Ca2+]i rise. U73122, an inhibitor of phospholipase C, abolished histamine (but not L-NASPA)-induced [Ca2+]i rise. Overnight incubation with 1 microM L-NASPA did not affect cell proliferation, but 10-20 microM L-NASPA exerted 4% and 15% inhibition, respectively. Collectively, L-NASPA rapidly increased [Ca2+]i in MG63 cells by evoking both extracellular Ca2+ influx and intracellular Ca2+ release, and is cytotoxic at higher concentrations.


Assuntos
Cálcio/química , Citosol/efeitos dos fármacos , Citosol/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Palmitatos/farmacologia , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Animais , Cálcio/fisiologia , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Bovinos , Relação Dose-Resposta a Droga , Corantes Fluorescentes , Fura-2 , Humanos , Fosfolipases/metabolismo , Células Tumorais Cultivadas
17.
Neuropsychopharmacology ; 27(4): 565-75, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12377393

RESUMO

Amphetamine is an indirect dopamine receptor agonist and increases glutamate release in the striatum. Activation of group I metabotropic glutamate receptors (mGluRs) upregulates cAMP response element-binding protein (CREB) and Elk-1 phosphorylation via extracellular signal-regulated kinase 1 and 2 (ERK1/2) in the striatum in vivo. In the present study the role of mGluRs in the regulation of ERK1/2 pathways leading to CREB and Elk-1 phosphorylation by amphetamine was investigated using immunohistochemistry and Western blot in the rat dorsal striatum. Acute administration of amphetamine (5 mg/kg, i.p.) caused increases in phosphorylated (p)CREB, pElk-1, and pERK1/2 immunoreactivity. Intrastriatal blockade of group I mGluRs with N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC; 25 nmol) significantly attenuated amphetamine-induced pCREB, pElk-1, pERK1/2, and Fos immunoreactivity in both medial and lateral areas of the striatum. Systemic injection of an mGluR5 antagonist, 2-methyl-6-(phenylethynyl)pyridine hydrochloride (MPEP; 10 mg/kg, i.p.), also blocked the amphetamine induction of these phosphoproteins. In contrast, intrastriatal blockade of group II/III mGluRs with (RS)-alpha-methylserine-o-phosphate monophenyl ester (MSOPPE; 25 nmol) did not affect amphetamine-induced increases in all the four markers. Similarly, intrastriatal dantrolene (2 or 20 nmol) that blocks intracellular Ca(2+) release from ryanodine-sensitive stores did not affect amphetamine effects. Injection of PHCCC, MPEP, MSOPPE, or dantrolene alone did not alter basal levels of the three phosphoproteins and Fos. These data suggest that acute amphetamine is able to facilitate the phosphorylation of CREB, Elk-1, and ERK1/2 signaling proteins and Fos gene expression via a group I mGluR-dependent mechanism in the dorsal striatum.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Anfetamina/farmacologia , Proteínas de Ligação a DNA , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Neostriado/efeitos dos fármacos , Fosfosserina/análogos & derivados , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Fatores de Transcrição/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Transtornos Relacionados ao Uso de Anfetaminas/fisiopatologia , Animais , Benzopiranos/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dantroleno/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Imuno-Histoquímica , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Relaxantes Musculares Centrais/farmacologia , Neostriado/metabolismo , Fosforilação/efeitos dos fármacos , Fosfosserina/farmacologia , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-fos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Piridinas/farmacologia , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima/fisiologia , Proteínas Elk-1 do Domínio ets , Proteínas Quinases p38 Ativadas por Mitógeno
18.
Lipids ; 37(1): 53-9, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11876263

RESUMO

Cholesteryl-3beta-phosphoserine (CPHS) is a synthetic steroid affecting intracellular cholesterol transport. To compare CPHS with the well-known inhibitors progesterone and U18666A, we examined cholesterol transport in three human cell lines: the monocytic U-937, the endothelial ECV-304, and the lymphoid Jurkat. Under low density lipoprotein (LDL) loading, CPHS inhibited cholesterol esterification in U-937 and ECV-304 cells but not in Jurkat cells. In contrast, CPHS inhibited the mobilization of plasma membrane cholesterol induced by 25-hydroxycholesterol, brefeldin A, or sphingomyelinase in all cell lines. In cells pulse-labeled with [3 H]cholesterol, CPHS decreased incorporation of cholesterol and inhibited its esterification. In prelabeled cells, CPHS promoted cholesterol efflux and enhanced the cyclodextrin-mediated removal of plasma membrane cholesterol. CPHS did not affect endogenous cholesterol synthesis nor acylcoenzyme A:cholesterol acyltransferase activity. These data suggest that, unlike progesterone and U18666A, CPHS inhibits intracellular cholesterol transport by specifically affecting the movements of cholesterol in the plasma membrane. Owing to this restricted site of action, CPHS may help to clarify the role of the plasma membrane in cholesterol trafficking. For example, the lack of an effect of CPHS on the esterification of LDL-derived cholesterol in Jurkat cells suggests that most of the LDL-derived cholesterol in these cells is directly delivered to the endoplasmic reticulum without cycling through the plasma membrane.


Assuntos
Colesterol/análogos & derivados , Colesterol/metabolismo , Colesterol/farmacologia , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Androstenos/farmacologia , Transporte Biológico Ativo/efeitos dos fármacos , Linhagem Celular , Membrana Celular/metabolismo , Ésteres do Colesterol/metabolismo , LDL-Colesterol/metabolismo , Ciclodextrinas/farmacologia , Retículo Endoplasmático/metabolismo , Esterificação , Humanos , Líquido Intracelular/metabolismo , Células Jurkat , Progesterona/farmacologia , Células U937
19.
Brain Res Mol Brain Res ; 94(1-2): 75-84, 2001 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-11597767

RESUMO

Cyclic AMP response element-binding protein (CREB) is a major transcriptional activator at the calcium and cAMP response-element (CaCRE). Phosphorylated (p)CREB facilitates gene expression in striatal neurons. Elk-1 is another transcriptional regulator at the serum response element in the upstream promoter region of the CaCRE. Elk-1 is phosphorylated by extracellular signal-regulated kinases (ERK) and may also contribute to the regulation of gene expression. To evaluate putative roles of group I metabotropic glutamate receptors (mGluRs) in CREB, Elk-1, and ERK phosphorylation, the group I selective agonist, 3,5-dihydroxyphenylglycine (DHPG), was infused into the dorsal striatum at doses of 125, 250, or 500 nmol in freely moving rats. Semi-quantitative immunohistochemistry demonstrated that DHPG significantly increased levels of pCREB, pElk-1, and pERK immunoreactivity of ipsilateral dorsal striatum in a dose dependent manner. The increased immunoreactivity by 500 nmol DHPG was significantly blocked by intrastriatal infusion of the group I selective antagonist, n-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC, 25 nmol), but not by the group II/III antagonist, (RS)-alpha-methylserine-o-phosphate monophenyl ester (MSOPPE, 25 nmol). These data suggest that group I mGluR activation is positively linked to signaling cascades resulting in CREB, Elk-1, and ERK phosphorylation in the striatum in vivo.


Assuntos
Corpo Estriado/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA , Metoxi-Hidroxifenilglicol/análogos & derivados , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Fatores de Transcrição , Animais , Comportamento Animal/efeitos dos fármacos , Benzopiranos/farmacologia , Relação Dose-Resposta a Droga , Masculino , Metoxi-Hidroxifenilglicol/farmacologia , Fosforilação , Fosfosserina/análogos & derivados , Fosfosserina/farmacologia , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Proteínas Elk-1 do Domínio ets
20.
FEBS Lett ; 494(1-2): 121-4, 2001 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-11297747

RESUMO

Mildly oxidized low-density lipoprotein activates platelets through lysophosphatidic acid (LPA). Hence, the platelet-activating properties attributed to native low-density lipoprotein (nLDL) might be caused by LPA contamination. We show that nLDL enhances thrombin receptor-activating peptide (TRAP)-induced fibrinogen binding to alpha(IIb)beta(3). The LPA receptor blocker N-palmitoyl-L-serine-phosphoric acid did not affect nLDL-enhanced fibrinogen binding induced by TRAP, but reduced TRAP-induced binding. cAMP and inhibitors of protein kinase C and Ca(2+) rises completely blocked ligand binding by TRAP and nLDL/TRAP. Inhibitors of p38(MAPK) and ADP secretion interfered only partially. Blockade of Rho-kinase increased ligand binding 2-3-fold. We conclude that nLDL enhances TRAP-induced fibrinogen binding independent of LPA.


Assuntos
Fibrinogênio/metabolismo , Lipoproteínas LDL/metabolismo , Lisofosfolipídeos/metabolismo , Ativação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G , Difosfato de Adenosina/metabolismo , Cálcio/metabolismo , AMP Cíclico/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipoproteínas LDL/farmacologia , Palmitatos/metabolismo , Palmitatos/farmacologia , Fragmentos de Peptídeos/farmacologia , Fosfosserina/análogos & derivados , Fosfosserina/metabolismo , Fosfosserina/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Ácidos Lisofosfatídicos , Transdução de Sinais , Quinases Associadas a rho
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA