Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 18(1): 15-25, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27841869

RESUMO

The lymph node periphery is an important site for many immunological functions, from pathogen containment to the differentiation of helper T cells, yet the cues that position cells in this region are largely undefined. Here, through the use of a reporter for the signaling lipid S1P (sphingosine 1-phosphate), we found that cells sensed higher concentrations of S1P in the medullary cords than in the T cell zone and that the S1P transporter SPNS2 on lymphatic endothelial cells generated this gradient. Natural killer (NK) cells are located at the periphery of the lymph node, predominantly in the medulla, and we found that expression of SPNS2, expression of the S1P receptor S1PR5 on NK cells, and expression of the chemokine receptor CXCR4 were all required for NK cell localization during homeostasis and rapid production of interferon-γ by NK cells after challenge. Our findings elucidate the spatial cues for NK cell organization and reveal a previously unknown role for S1P in positioning cells within the medulla.


Assuntos
Proteínas de Transporte de Ânions/metabolismo , Células Endoteliais/imunologia , Células Matadoras Naturais/imunologia , Linfonodos/imunologia , Lisofosfolipídeos/metabolismo , Receptores CXCR4/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Proteínas de Transporte de Ânions/genética , Diferenciação Celular , Movimento Celular , Células Cultivadas , Quimiotaxia , Homeostase , Interferon gama/metabolismo , Ativação Linfocitária/genética , Lisofosfolipídeos/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CXCR4/genética , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais , Esfingosina/química , Esfingosina/metabolismo , Linfócitos T Auxiliares-Indutores/fisiologia
2.
Nature ; 566(7743): E6, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30670873

RESUMO

In this Article, the top label in Fig. 5d should read 'DISH 3/16' instead of 'DISH 3/17'. This error has been corrected online.

3.
Nature ; 563(7733): 639-645, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30464338

RESUMO

The diversity and complexity of the human brain are widely assumed to be encoded within a constant genome. Somatic gene recombination, which changes germline DNA sequences to increase molecular diversity, could theoretically alter this code but has not been documented in the brain, to our knowledge. Here we describe recombination of the Alzheimer's disease-related gene APP, which encodes amyloid precursor protein, in human neurons, occurring mosaically as thousands of variant 'genomic cDNAs' (gencDNAs). gencDNAs lacked introns and ranged from full-length cDNA copies of expressed, brain-specific RNA splice variants to myriad smaller forms that contained intra-exonic junctions, insertions, deletions, and/or single nucleotide variations. DNA in situ hybridization identified gencDNAs within single neurons that were distinct from wild-type loci and absent from non-neuronal cells. Mechanistic studies supported neuronal 'retro-insertion' of RNA to produce gencDNAs; this process involved transcription, DNA breaks, reverse transcriptase activity, and age. Neurons from individuals with sporadic Alzheimer's disease showed increased gencDNA diversity, including eleven mutations known to be associated with familial Alzheimer's disease that were absent from healthy neurons. Neuronal gene recombination may allow 'recording' of neural activity for selective 'playback' of preferred gene variants whose expression bypasses splicing; this has implications for cellular diversity, learning and memory, plasticity, and diseases of the human brain.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Variação Genética/genética , Neurônios/citologia , Neurônios/patologia , Recombinação Genética , Processamento Alternativo/genética , Animais , DNA Complementar/análise , DNA Complementar/genética , DNA Polimerase Dirigida por DNA/metabolismo , Éxons/genética , Feminino , Humanos , Íntrons/genética , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Especificidade de Órgãos , Mutação Puntual/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Análise de Sequência de DNA , Deleção de Sequência/genética
4.
J Cardiothorac Vasc Anesth ; 38(6): 1347-1352, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38521629

RESUMO

OBJECTIVE: This study aimed to delineate the recovery patterns of regional oxygen saturation (SrO2) in pediatric cardiac surgery patients subjected to remote ischemic preconditioning (RIPC), utilizing near-infrared spectroscopy (NIRS) for quantification. It also sought to establish the correlation between these perfusion patterns and postoperative clinical outcomes. DESIGN: A prospective longitudinal observational study. SETTING: The study was conducted at Fundación Valle Del Lili, a high-complexity service provider institution in Fundación Valle Del Lili. PARTICIPANTS: Pediatric patients (younger than 18 years of age) scheduled for elective cardiac surgery requiring cardiopulmonary bypass between August 2022 and July 2023. INTERVENTIONS: RIPC was performed after anesthetic induction, involving cycles of ischemia and reperfusion on a lower limb. Monitoring included SrO2 using NIRS. MEASUREMENTS AND MAIN RESULTS: The study identified 4 distinct patterns of SrO2 during RIPC. Findings demonstrated a significant association between the negative SrO2 pattern and increased postoperative adverse events, including extended hospital stays and higher mortality, while a positive pattern was associated with better outcomes. CONCLUSIONS: Specific patterns of SrO2 response to RIPC may serve as important indicators for risk stratification in congenital heart surgery. This study illustrated the potential of NIRS in detecting hypoxic states and predicting postoperative outcomes, emphasizing the need for standardized clinical interpretation of RIPC patterns.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Saturação de Oxigênio , Espectroscopia de Luz Próxima ao Infravermelho , Humanos , Estudos Prospectivos , Masculino , Feminino , Procedimentos Cirúrgicos Cardíacos/métodos , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Lactente , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Saturação de Oxigênio/fisiologia , Pré-Escolar , Criança , Precondicionamento Isquêmico/métodos , Estudos Longitudinais , Adolescente , Resultado do Tratamento , Cardiopatias Congênitas/cirurgia
5.
Pediatr Cardiol ; 45(4): 780-786, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38421480

RESUMO

Congenital heart diseases impact millions annually, with pediatric care lacking suitable risk assessment tools. This research seeks to illuminate the association between the global longitudinal strain (GLS) and the subsequent impact on postoperative outcomes, contributing to a deeper understanding of its predictive value in the pediatric population affected by congenital heart diseases. An observational, analytic, longitudinal, and prospective study was conducted from May 2022 to May 2023, including all patients under 18 undergoing heart surgery with cardiopulmonary bypass (CBP). Patients not classifiable within the Risk Adjustment for Congenital Heart Surgery were excluded. Using transesophageal echocardiography, GLS was measured pre- and post-CPB. Receiver operating characteristic curve analysis determined GLS cut-off points for 30-day mortality risk, using Youden's method for optimal sensitivity and specificity. Bivariate and multivariate analysis identified the relationships between clinical variables. Eighty-nine patients undergoing congenital heart surgery were included. Fifteen deaths occurred. The area under the curve (AUC) for each GLS classification (pre, post, index) demonstrated effective discriminatory capacity (> 0.70) in predicting 30-day mortality. Pre-CBP GLS showed the strongest predictive power (AUC 0.833, IQR: 0.731 - 0.936) with a cut-off point of 12. Values lower than the cut-off point of pre-CPB GLS correlated with increased vasoactive-inotropic Scores and longer mechanical ventilation. GLS measurement is a reproducible method for assessing ventricular function in pediatric heart surgery, showing potential as a prognostic tool. This study marks the initial effort to establish cut-off points for preoperative GLS, postoperative GLS, and the strain index.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Cardiopatias Congênitas , Criança , Humanos , Deformação Longitudinal Global , Cardiopatias Congênitas/diagnóstico por imagem , Cardiopatias Congênitas/cirurgia , Hospitais , Valor Preditivo dos Testes , Estudos Prospectivos , Volume Sistólico , Função Ventricular Esquerda
6.
Nat Immunol ; 12(10): 992-1001, 2011 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-21857655

RESUMO

It is established that the transcription factor E2A and its antagonist Id3 modulate the checkpoints consisting of the precursor to the T cell antigen receptor (pre-TCR) and the TCR. Here we demonstrate that Id3 expression was higher beyond the pre-TCR checkpoint, remained high in naive T cells and showed a bimodal pattern in the effector-memory population. We show how E2A promoted T lineage specification and how pre-TCR-mediated signaling affected E2A genome-wide occupancy. Thymi in Id3-deficient mice had aberrant development of effector-memory cells, higher expression of the chemokine receptor CXCR5 and the transcriptional repressor Bcl-6 and, unexpectedly, T cell-B cell conjugates and B cell follicles. Collectively, our data show how E2A acted globally to orchestrate development into the T lineage and that Id3 antagonized E2A activity beyond the pre-TCR checkpoint to enforce the naive fate of T cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas Inibidoras de Diferenciação/fisiologia , Linfócitos T/imunologia , Animais , Memória Imunológica , Imunofenotipagem , Antígenos Comuns de Leucócito/análise , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores CXCR5/análise , Baço/imunologia , Timo/imunologia
7.
Nat Immunol ; 12(12): 1221-9, 2011 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-22057289

RESUMO

During infection, naive CD8(+) T cells differentiate into effector cells, which are armed to eliminate pathogens, and memory cells, which are poised to protect against reinfection. The transcriptional program that regulates terminal differentiation into short-lived effector-memory versus long-lived memory cells is not clearly defined. Through the use of mice expressing reporters for the DNA-binding inhibitors Id2 and Id3, we identified Id3(hi) precursors of long-lived memory cells before the peak of T cell population expansion or upregulation of cell-surface receptors that indicate memory potential. Deficiency in Id2 or Id3 resulted in loss of distinct CD8(+) effector and memory populations, which demonstrated unique roles for these inhibitors of E-protein transcription factors. Furthermore, cytokines altered the expression of Id2 and Id3 differently, which provides insight into how external cues influence gene expression.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Regulação da Expressão Gênica , Memória Imunológica/imunologia , Proteína 2 Inibidora de Diferenciação/metabolismo , Proteínas Inibidoras de Diferenciação/metabolismo , Subpopulações de Linfócitos T/imunologia , Transcrição Gênica , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Citocinas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Infecções/genética , Infecções/imunologia , Infecções/microbiologia , Proteína 2 Inibidora de Diferenciação/genética , Proteínas Inibidoras de Diferenciação/genética , Lectinas Tipo C , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Receptores Imunológicos/metabolismo , Subpopulações de Linfócitos T/citologia , Transcrição Gênica/efeitos dos fármacos
8.
FASEB J ; 36(2): e22132, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34986275

RESUMO

Ponesimod is a sphingosine 1-phosphate (S1P) receptor (S1PR) modulator that was recently approved for treating relapsing forms of multiple sclerosis (MS). Three other FDA-approved S1PR modulators for MS-fingolimod, siponimod, and ozanimod-share peripheral immunological effects via common S1P1 interactions, yet ponesimod may access distinct central nervous system (CNS) mechanisms through its selectivity for the S1P1 receptor. Here, ponesimod was examined for S1PR internalization and binding, human astrocyte signaling and single-cell RNA-seq (scRNA-seq) gene expression, and in vivo using murine cuprizone-mediated demyelination. Studies confirmed ponesimod's selectivity for S1P1 without comparable engagement to the other S1PR subtypes (S1P2,3,4,5 ). Ponesimod showed pharmacological properties of acute agonism followed by chronic functional antagonism of S1P1 . A major locus of S1P1 expression in the CNS is on astrocytes, and scRNA-seq of primary human astrocytes exposed to ponesimod identified a gene ontology relationship of reduced neuroinflammation and reduction in known astrocyte disease-related genes including those of immediate early astrocytes that have been strongly associated with disease progression in MS animal models. Remarkably, ponesimod prevented cuprizone-induced demyelination selectively in the cingulum, but not in the corpus callosum. These data support the CNS activities of ponesimod through S1P1 , including protective, and likely selective, effects against demyelination in a major connection pathway of the brain, the limbic fibers of the cingulum, lesions of which have been associated with several neurologic impairments including MS fatigue.


Assuntos
Astrócitos/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Substâncias Protetoras/farmacologia , Receptores de Esfingosina-1-Fosfato/metabolismo , Tiazóis/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Sistema Nervoso Central/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos
9.
FASEB J ; 34(7): 8833-8842, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32929779

RESUMO

LPA1 is one of six known receptors (LPA1-6) for lysophosphatidic acid (LPA). Constitutive Lpar1 null mutant mice have been instrumental in identifying roles for LPA-LPA1 signaling in neurobiological processes, brain development, and behavior, as well as modeling human neurological diseases like neuropathic pain. Constitutive Lpar1 null mutant mice are protected from partial sciatic nerve ligation (PSNL)-induced neuropathic pain, however, the cell types that are functionally responsible for mediating this protective effect are unknown. Here, we report the generation of an Lpar1flox/flox conditional null mutant mouse that allows for cre-mediated conditional deletion, combined with a PSNL pain model. Lpar1flox/flox mice were crossed with cre transgenic lines driven by neural gene promoters for nestin (all neural cells), synapsin (neurons), or P0 (Schwann cells). CD11b-cre transgenic mice were also used to delete Lpar1 in microglia. PSNL-initiated pain responses were reduced following cre-mediated Lpar1 deletion with all three neural promoters as well as the CD11b promoter, supporting involvement of Schwann cells, central and/or peripheral neurons, and microglia in mediating pain. Interestingly, rescue responses were nonidentical, implicating distinct roles for Lpar1-expressing cell types. Our results with a new Lpar1 conditional mouse mutant expand an understanding of LPA1 signaling in the PSNL model of neuropathic pain.


Assuntos
Microglia/patologia , Neuralgia/patologia , Neurônios/patologia , Receptores de Ácidos Lisofosfatídicos/fisiologia , Células de Schwann/patologia , Nervo Isquiático/cirurgia , Animais , Feminino , Marcação de Genes , Ligadura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Neuralgia/etiologia , Neuralgia/metabolismo , Neurônios/metabolismo , Células de Schwann/metabolismo , Transdução de Sinais
11.
Proc Natl Acad Sci U S A ; 115(42): 10804-10809, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30262650

RESUMO

Somatic copy number variations (CNVs) exist in the brain, but their genesis, prevalence, forms, and biological impact remain unclear, even within experimentally tractable animal models. We combined a transposase-based amplification (TbA) methodology for single-cell whole-genome sequencing with a bioinformatic approach for filtering unreliable CNVs (FUnC), developed from machine learning trained on lymphocyte V(D)J recombination. TbA-FUnC offered superior genomic coverage and removed >90% of false-positive CNV calls, allowing extensive examination of submegabase CNVs from over 500 cells throughout the neurogenic period of cerebral cortical development in Mus musculus Thousands of previously undocumented CNVs were identified. Half were less than 1 Mb in size, with deletions 4× more common than amplification events, and were randomly distributed throughout the genome. However, CNV prevalence during embryonic cortical development was nonrandom, peaking at midneurogenesis with levels triple those found at younger ages before falling to intermediate quantities. These data identify pervasive small and large CNVs as early contributors to neural genomic mosaicism, producing genomically diverse cellular building blocks that form the highly organized, mature brain.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Variações do Número de Cópias de DNA , Regulação da Expressão Gênica no Desenvolvimento , Neurogênese/genética , Análise de Célula Única/métodos , Sequenciamento Completo do Genoma/métodos , Animais , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Genoma , Genômica , Camundongos , Camundongos Endogâmicos C57BL
12.
J Lipid Res ; 60(1): 212-217, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30463988

RESUMO

Lysophosphatidic acid (LPA) activates cognate G protein-coupled receptors (GPCRs) to initiate biological signaling cascades. Lysophospholipid (LP) receptor binding properties remain incompletely assessed because of difficulties with ligand lipophilicity and lipid "stickiness." These inherent attributes produce high levels of nonspecific binding within cell-membrane preparations used to assess GPCRs, as has been shown in classical binding assays using radiolabeled ligands, making accurate measurements of lipid binding kinetics difficult to achieve. Backscattering interferometry (BSI) is an optical technology that measures molecular binding interactions by reporting changes in the refractive index of a solution after binding events. Here, we report the use of BSI to assess LPA1 for its ability to bind to naturally occurring lipids and a synthetic LPA1 antagonist (ONO-9780307), under both primary- and competition-binding conditions. Assessment of 12 different lipids demonstrated that the known LP ligand, 1-oleoyl-LPA, as well as an endocannabinoid metabolite, anandamide phosphate, are specific ligands for LPA1, whereas other LPs tested were not. Newly determined dissociation constants (Kd values) for orthosteric lipid ligands approximated 10-9 M, substantially lower (i.e., with higher affinity) than measured Kd values in classical binding or cell-based assays. These results demonstrate that BSI may have particular utility in assessing binding interactions between lipid receptors and their lipid ligands and could provide new screening approaches for lipid receptor identification and drug discovery.


Assuntos
Interferometria/métodos , Luz , Lisofosfolipídeos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Ligação Competitiva , Linhagem Celular , Ligantes , Ligação Proteica , Espalhamento de Radiação , Especificidade por Substrato
13.
J Biol Chem ; 288(41): 29882-9, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-24003216

RESUMO

The fetal development of the mammalian eyelid involves the expansion of the epithelium over the developing cornea, fusion into a continuous sheet covering the eye, and a splitting event several weeks later that results in the formation of the upper and lower eyelids. Recent studies have revealed a significant number of molecular signaling components that are essential mediators of eyelid development. Receptor-mediated sphingosine 1-phosphate (S1P) signaling is known to influence diverse biological processes, but its involvement in eyelid development has not been reported. Here, we show that two S1P receptors, S1P2 and S1P3, are collectively essential mediators of eyelid closure during murine development. Homozygous deletion of the gene encoding either receptor has no apparent effect on eyelid development, but double-null embryos are born with an "eyes open at birth" defect due to a delay in epithelial sheet extension. Both receptors are expressed in the advancing epithelial sheet during the critical period of extension. Fibroblasts derived from double-null embryos have a deficient response to epidermal growth factor, suggesting that S1P2 and S1P3 modulate this essential signaling pathway during eyelid closure.


Assuntos
Desenvolvimento Embrionário/genética , Pálpebras/metabolismo , Receptores de Lisoesfingolipídeo/genética , Animais , Western Blotting , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Anormalidades do Olho/embriologia , Anormalidades do Olho/genética , Pálpebras/embriologia , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Lisofosfolipídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Gravidez , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato
14.
Proc Natl Acad Sci U S A ; 108(2): 751-6, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21177428

RESUMO

Sphingosine 1-phosphate (S1P), a lysophospholipid, has gained relevance to multiple sclerosis through the discovery of FTY720 (fingolimod), recently approved as an oral treatment for relapsing forms of multiple sclerosis. Its mechanism of action is thought to be immunological through an active phosphorylated metabolite, FTY720-P, that resembles S1P and alters lymphocyte trafficking through receptor subtype S1P(1). However, previously reported expression and in vitro studies of S1P receptors suggested that direct CNS effects of FTY720 might theoretically occur through receptor modulation on neurons and glia. To identify CNS cells functionally contributing to FTY720 activity, genetic approaches were combined with cellular and molecular analyses. These studies relied on the functional assessment, based on clinical score, of conditional null mouse mutants lacking S1P(1) in CNS cell lineages and challenged by experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. All conditional null mutants displayed WT lymphocyte trafficking that responded normally to FTY720. In marked contrast, EAE was attenuated and FTY720 efficacy was lost in CNS mutants lacking S1P(1) on GFAP-expressing astrocytes but not on neurons. In situ hybridization studies confirmed that astrocyte loss of S1P(1) was the key alteration in functionally affected mutants. Reductions in EAE clinical scores were paralleled by reductions in demyelination, axonal loss, and astrogliosis. Receptor rescue and pharmacological experiments supported the loss of S1P(1) on astrocytes through functional antagonism by FTY720-P as a primary FTY720 mechanism. These data identify nonimmunological CNS mechanisms of FTY720 efficacy and implicate S1P signaling pathways within the CNS as targets for multiple sclerosis therapies.


Assuntos
Astrócitos/citologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Regulação da Expressão Gênica , Esclerose Múltipla/tratamento farmacológico , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Cloridrato de Fingolimode , Imunossupressores/farmacologia , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Esfingosina/farmacologia
15.
J Biol Chem ; 287(21): 17608-17617, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22461625

RESUMO

Lysophosphatidic acid (LPA) is a bioactive lipid that serves as an extracellular signaling molecule acting through cognate G protein-coupled receptors designated LPA(1-6) that mediate a wide range of both normal and pathological effects. Previously, LPA(1), a G(αi)-coupled receptor (which also couples to other G(α) proteins) to reduce cAMP, was shown to be essential for the initiation of neuropathic pain in the partial sciatic nerve ligation (PSNL) mouse model. Subsequent gene expression studies identified LPA(5), a G(α12/13)- and G(q)-coupled receptor that increases cAMP, in a subset of dorsal root ganglion neurons and also within neurons of the spinal cord dorsal horn in a pattern complementing, yet distinct from LPA(1), suggesting its possible involvement in neuropathic pain. We therefore generated an Lpar5 null mutant by targeted deletion followed by PSNL challenge. Homozygous null mutants did not show obvious base-line phenotypic defects. However, following PSNL, LPA(5)-deficient mice were protected from developing neuropathic pain. They also showed reduced phosphorylated cAMP response element-binding protein expression within neurons of the dorsal horn despite continued up-regulation of the characteristic pain-related markers Caα(2)δ(1) and glial fibrillary acidic protein, results that were distinct from those previously observed for LPA(1) deletion. These data expand the influences of LPA signaling in neuropathic pain through a second LPA receptor subtype, LPA(5), involving a mechanistically distinct downstream signaling pathway compared with LPA(1).


Assuntos
Gânglios Espinais/metabolismo , Lisofosfolipídeos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais , Animais , AMP Cíclico/genética , AMP Cíclico/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Gânglios Espinais/patologia , Deleção de Genes , Lisofosfolipídeos/genética , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Neuralgia/genética , Receptores de Ácidos Lisofosfatídicos/genética , Elementos de Resposta/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Biochim Biophys Acta ; 1823(2): 245-54, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22019450

RESUMO

Sphingosine-1-phosphate-induced α1B-adrenergic receptor desensitization and phosphorylation were studied in rat-1 fibroblasts stably expressing enhanced green fluorescent protein-tagged adrenoceptors. Sphingosine-1-phosphate induced adrenoceptor desensitization and phosphorylation through a signaling cascade that involved phosphoinositide 3-kinase and protein kinase C activities. The autocrine/paracrine role of sphingosine-1-phosphate was also studied. It was observed that activation of receptor tyrosine kinases, such as insulin growth factor-1 (IGF-I) and epidermal growth factor (EGF) receptors increased sphingosine kinase activity. Such activation and consequent production of sphingosine-1-phosphate appear to be functionally relevant in IGF-I- and EGF-induced α1B-adrenoceptor phosphorylation and desensitization as evidenced by the following facts: a) expression of a catalytically inactive (dominant-negative) mutant of sphingosine kinase 1 or b) S1P1 receptor knockdown markedly reduced this growth factor action. This action of sphingosine-1-phosphate involves EGF receptor transactivation. In addition, taking advantage of the presence of the eGFP tag in the receptor construction, we showed that S1P was capable of inducing α1B-adrenergic receptor internalization and that its autocrine/paracrine generation was relevant for internalization induced by IGF-I. Four distinct hormone receptors and two autocrine/paracrine mediators participate in IGF-I receptor-α1B-adrenergic receptor crosstalk.


Assuntos
Comunicação Autócrina/fisiologia , Lisofosfolipídeos/metabolismo , Comunicação Parácrina/fisiologia , Receptores Adrenérgicos alfa 1/metabolismo , Esfingosina/análogos & derivados , Animais , Receptores ErbB/antagonistas & inibidores , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Ratos , Receptor IGF Tipo 1/metabolismo , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/metabolismo
17.
Glia ; 61(12): 2009-22, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24115248

RESUMO

Schwann cell (SC) migration is an important step preceding myelination and remyelination in the peripheral nervous system, and can be promoted by peptide factors like neuregulins. Here we present evidence that a lipid factor, lysophosphatidic acid (LPA), influences both SC migration and peripheral myelination through its cognate G protein-coupled receptor (GPCR) known as LPA1 . Ultrastructural analyses of peripheral nerves in mouse null-mutants for LPA1 showed delayed SC-to-axon segregation, polyaxonal myelination by single SCs, and thinner myelin sheaths. In primary cultures, LPA promoted SC migration through LPA1 , while analysis of conditioned media from purified dorsal root ganglia neurons using HPLC/MS supported the production of LPA by these neurons. The heterotrimeric G-alpha protein, Gαi , and the small GTPase, Rac1, were identified as important downstream signaling components of LPA1 . These results identify receptor mediated LPA signaling between neurons and SCs that promote SC migration and contribute to the normal development of peripheral nerves through effects on SC-axon segregation and myelination.


Assuntos
Axônios/metabolismo , Movimento Celular/fisiologia , Lisofosfolipídeos/farmacologia , Bainha de Mielina/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Células de Schwann/metabolismo , Animais , Axônios/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Camundongos , Bainha de Mielina/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Células de Schwann/citologia , Células de Schwann/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
18.
Cell Rep ; 42(12): 113545, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38064339

RESUMO

Vitamin B12 (B12) deficiency causes neurological manifestations resembling multiple sclerosis (MS); however, a molecular explanation for the similarity is unknown. FTY720 (fingolimod) is a sphingosine 1-phosphate (S1P) receptor modulator and sphingosine analog approved for MS therapy that can functionally antagonize S1P1. Here, we report that FTY720 suppresses neuroinflammation by functionally and physically regulating the B12 pathways. Genetic and pharmacological S1P1 inhibition upregulates a transcobalamin 2 (TCN2)-B12 receptor, CD320, in immediate-early astrocytes (ieAstrocytes; a c-Fos-activated astrocyte subset that tracks with experimental autoimmune encephalomyelitis [EAE] severity). CD320 is also reduced in MS plaques. Deficiency of CD320 or dietary B12 restriction worsens EAE and eliminates FTY720's efficacy while concomitantly downregulating type I interferon signaling. TCN2 functions as a chaperone for FTY720 and sphingosine, whose complex induces astrocytic CD320 internalization, suggesting a delivery mechanism of FTY720/sphingosine via the TCN2-CD320 pathway. Taken together, the B12-TCN2-CD320 pathway is essential for the mechanism of action of FTY720.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Animais , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Cloridrato de Fingolimode/metabolismo , Astrócitos/metabolismo , Esfingosina/metabolismo , Vitamina B 12/farmacologia , Vitamina B 12/uso terapêutico , Vitamina B 12/metabolismo , Transcobalaminas/metabolismo , Transcobalaminas/uso terapêutico , Propilenoglicóis/metabolismo , Propilenoglicóis/farmacologia , Propilenoglicóis/uso terapêutico , Vitaminas , Imunossupressores/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo
19.
ACS Omega ; 7(5): 4185-4193, 2022 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-35155912

RESUMO

The modification of the rutile TiO2(110) surface with dopamine represents the best example of the functionalization of TiO2-based nanoparticles with catecholamines, which is of great interest for sunlight harvesting and drug delivery. However, there is little information on the dopamine-TiO2(110) adsorption complex in terms of thermodynamic properties and structural parameters such as bond coordination and orientation of the terminal ethyl-amino group. Here, we report a density functional theory (DFT) investigation of dopamine adsorption on the TiO2(110) surface using the optB86b-vdW functional with a Hubbard-type correction to the Ti 3d orbitals, where U eff = 3 eV. Guided by available X-ray photoelectron spectroscopy (XPS) and near-edge X-ray absorption fine structure (NEXAFS) data, our simulations identify enolate species with bidentate coordination at a submonolayer coverage, which are bonded to two neighboring 5-fold-coordinated Ti atoms at the TiO2(110) surface through both deprotonated oxygen atoms of the dopamine, i.e., in a bridging fashion. The process is highly exothermic, involving an adsorption energy of -2.90 eV. Calculated structural parameters suggest that the molecule sits approximately upright on the surface with the amino group interacting with the π-like orbitals of the aromatic ring, leading to a gauche-like configuration. The resulting NH···π hydrogen bond in this configuration can be broken by overcoming an energy barrier of 0.22 eV; in this way, the amino group rotation leads to an anti-like conformation, making this terminal group able to bind to other biomolecules. This mechanism is endothermic by 0.07 eV. Comparison of existing spectroscopic data with DFT modeling shows that our computational setup can reproduce most experimentally determined parameters such as tilt angles from NEXAFS and chemical shifts in XPS, which allows us to identify the preferred mode of adsorption of dopamine on the TiO2(110) surface.

20.
Front Cell Neurosci ; 16: 918041, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35783097

RESUMO

Multiple sclerosis (MS) is an immune-mediated demyelinating disease that alters central nervous system (CNS) functions. Relapsing-remitting MS (RRMS) is the most common form, which can transform into secondary-progressive MS (SPMS) that is associated with progressive neurodegeneration. Single-nucleus RNA sequencing (snRNA-seq) of MS lesions identified disease-related transcriptomic alterations; however, their relationship to non-lesioned MS brain regions has not been reported and which could identify prodromal or other disease susceptibility signatures. Here, snRNA-seq was used to generate high-quality RRMS vs. SPMS datasets of 33,197 nuclei from 8 normal-appearing MS brains, which revealed divergent cell type-specific changes. Notably, SPMS brains downregulated astrocytic sphingosine kinases (SPHK1/2) - the enzymes required to phosphorylate and activate the MS drug, fingolimod. This reduction was modeled with astrocyte-specific Sphk1/2 null mice in which fingolimod lost activity, supporting functionality of observed transcriptomic changes. These data provide an initial resource for studies of single cells from non-lesioned RRMS and SPMS brains.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA