Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Genet ; 13: 1058817, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36685876

RESUMO

Black and Hispanic American patients frequently develop earlier onset of multiple sclerosis (MS) and a more severe disease course that can be resistant to disease modifying treatments. The objectives were to identify differential methylation of genomic DNA (gDNA) associated with disease susceptibility and treatment responses in a cohort of MS patients from underrepresented minority populations. Patients with MS and controls with non-inflammatory neurologic conditions were consented and enrolled under an IRB-approved protocol. Approximately 64% of donors identified as Black or African American and 30% as White, Hispanic-Latino. Infinium MethylationEPIC bead arrays were utilized to measure epigenome-wide gDNA methylation of whole blood. Data were analyzed in the presence and absence of adjustments for unknown covariates in the dataset, some of which corresponded to disease modifying treatments. Global patterns of differential methylation associated with MS were strongest for those probes that showed relative demethylation of loci with lower M values. Pathway analysis revealed unexpected associations with shigellosis and amoebiasis. Enrichment analysis revealed an over-representation of probes in enhancer regions and an under-representation in promoters. In the presence of adjustments for covariates that included disease modifying treatments, analysis revealed 10 differentially methylated regions (DMR's) with an FDR <1E-77. Five of these genes (ARID5B, BAZ2B, RABGAP1, SFRP2, WBP1L) are associated with cancer risk and cellular differentiation and have not been previously identified in MS studies. Hierarchical cluster and multi-dimensional scaling analysis of differential DNA methylation at 147 loci within those DMR's was sufficient to differentiate MS donors from controls. In the absence of corrections for disease modifying treatments, differential methylation in patients treated with dimethyl fumarate was associated with immune regulatory pathways that regulate cytokine and chemokine signaling, axon guidance, and adherens junctions. These results demonstrate possible associations of gastrointestinal pathogens and regulation of cellular differentiation with MS susceptibility in our patient cohort. This work further suggests that analyses can be performed in the presence and absence of corrections for immune therapies. Because of their high representation in our patient cohort, these results may be of specific relevance in the regulation of disease susceptibility and treatment responses in Black and Hispanic Americans.

2.
Immunobiology ; 225(2): 151883, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31818507

RESUMO

Prior work demonstrated that epithelial V-like antigen (EVA), a cell surface adhesion molecule, is expressed in B lymphocytes and is necessary for the efficacy of anti-alpha4 integrin treatment of experimental autoimmune encephalomyelitis (EAE), the mouse model of human multiple sclerosis. EVA deficiency is associated with a severe clinical phenotype of EAE in the presence or absence of treatment. Histological analysis revealed enhanced B cell-mediated autoimmunity and deposition of antibody and complement within the brain and spinal cord. Here our goal was to determine the molecular mechanism of EVA regulation of B lymphocyte function. Analysis of bone marrow from MOG-immunized mice revealed increased expansion of CD11c+ B cells in EVA-deficient mice as compared to wild type controls. In vitro studies of mouse bone marrow B lymphocytes revealed enhanced proliferation of the CD11c+ population in response to the Tlr7/8 agonist R848. An increase in R848-induced proliferation of CD11c+ B cells was also seen in vitro in Daudi cells, a human B cell line, following knockdown of the mpzl2 gene that encodes EVA. These mechanisms were characterized further by global expression analysis of bone marrow from immunized EVA-deficient and wild type control mice. These data revealed increased expression of B cell associated genes and decreased expression of the anti-viral oligoadenylate synthase genes, Oas1 and Oas2, in the knockout condition. In Daudi cells, R848 treatment induced an increase in Oas2 expression in control cells that was not observed in EVA-deficient cells. EVA deficiency also was associated with increased transcription of an Epstein-Barr virus gene during lytic replication. These results suggest EVA expression and signaling prevent expansion of CD11c+ B lymphocytes, a cellular phenotype associated with autoimmunity, increase expression of anti-viral oligoadenylate synthase genes, and reduce replication of a DNA virus.


Assuntos
Antivirais/imunologia , Linfócitos B/imunologia , Antígeno CD11c/imunologia , Animais , Autoimunidade/imunologia , Linfócitos B/virologia , Medula Óssea/imunologia , Medula Óssea/virologia , Encéfalo/imunologia , Encéfalo/virologia , Moléculas de Adesão Celular/imunologia , Proliferação de Células/fisiologia , Células Cultivadas , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/virologia , Herpesvirus Humano 4/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Transdução de Sinais/imunologia , Medula Espinal/imunologia , Medula Espinal/virologia
3.
Immunobiology ; 224(1): 80-93, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30391100

RESUMO

Prior work demonstrated that a splice variant of SCN5A, a voltage-gated sodium channel gene, acts as a cytoplasmic sensor for viral dsRNA in human macrophages. Expression of this channel also polarizes macrophages to an anti-inflammatory phenotype in vitro and in vivo. Here we utilized global expression analysis of splice variants to identify novel channel-dependent signaling mechanisms. Pharmacological activation of voltage-gated sodium channels in human macrophages, but not treatment with cytoplasmic poly I:C, was associated with splicing of a retained intron in transcripts of PPP1R10, a regulator of phosphatase activity and DNA repair. Microarray analysis also demonstrated expression of a novel sodium channel splice variant, human macrophage SCN10A, that contains a similar exon deletion as SCN5A. SCN10A localizes to cytoplasmic and nuclear vesicles in human macrophages. Simultaneous expression of human macrophage SCN5A and SCN10A was required to decrease expression of the retained intron and increase protein expression of PPP1R10. Channel activation also increased protein expression of the splicing factor EFTUD2, and knockdown of EFTUD2 prevented channel dependent splicing of the retained PPP1R10 intron. Knockdown of the SCN5A and SCN10A variants in human macrophages reduced the severity of dsDNA breaks induced by treatment with bleomycin and type 1 interferon. These results suggested that human macrophage SCN5A and SCN10A variants mediate an innate immune signaling pathway that limits DNA damage through increased expression of PPP1R10. The functional significance of this pathway is that it may prevent cytotoxicity during inflammatory responses.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Inflamação/metabolismo , Macrófagos/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Fatores de Alongamento de Peptídeos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Células Cultivadas , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Humanos , Imunidade Inata , Inflamação/genética , Análise em Microsséries , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Fatores de Alongamento de Peptídeos/genética , Processamento Pós-Transcricional do RNA , Proteínas de Ligação a RNA/genética , Ribonucleoproteína Nuclear Pequena U5/genética , Transdução de Sinais , Regulação para Cima
4.
J Biol Chem ; 289(51): 35326-40, 2014 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-25368329

RESUMO

Pattern recognition receptors contain a binding domain for pathogen-associated molecular patterns coupled to a signaling domain that regulates transcription of host immune response genes. Here, a novel mechanism that links pathogen recognition to channel activation and downstream signaling is proposed. We demonstrate that an intracellular sodium channel variant, human macrophage SCN5A, initiates signaling and transcription through a calcium-dependent isoform of adenylate cyclase, ADCY8, and the transcription factor, ATF2. Pharmacological stimulation with a channel agonist or treatment with cytoplasmic poly(I:C), a mimic of viral dsRNA, activates this pathway to regulate expression of SP100-related genes and interferon ß. Electrophysiological analysis reveals that the SCN5A variant mediates nonselective outward currents and a small, but detectable, inward current. Intracellular poly(I:C) markedly augments an inward voltage-sensitive sodium current and inhibits the outward nonselective current. These results suggest human macrophage SCN5A initiates signaling in an innate immune pathway relevant to antiviral host defense. It is postulated that SCN5A is a novel pathogen sensor and that this pathway represents a channel activation-dependent mechanism of transcriptional regulation.


Assuntos
Imunidade Inata/imunologia , Macrófagos/imunologia , Canal de Sódio Disparado por Voltagem NAV1.5/imunologia , Transdução de Sinais/imunologia , Fator 2 Ativador da Transcrição/genética , Fator 2 Ativador da Transcrição/imunologia , Fator 2 Ativador da Transcrição/metabolismo , Adenilil Ciclases/genética , Adenilil Ciclases/imunologia , Adenilil Ciclases/metabolismo , Animais , Antígenos Nucleares/genética , Antígenos Nucleares/imunologia , Antígenos Nucleares/metabolismo , Antivirais/farmacologia , Autoantígenos/genética , Autoantígenos/imunologia , Autoantígenos/metabolismo , Western Blotting , Células Cultivadas , AMP Cíclico/imunologia , AMP Cíclico/metabolismo , Perfilação da Expressão Gênica , Células HEK293 , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/genética , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Poli I-C/farmacologia , Ligação Proteica/imunologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
5.
J Neuropathol Exp Neurol ; 72(6): 489-504, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23656992

RESUMO

Multiple sclerosis (MS) is the most common nontraumatic cause of neurologic disability in young adults. Despite treatment, progressive tissue injury leads to accumulation of disability in many patients. Here, our goal was to develop an immune-mediated strategy to promote tissue repair and clinical recovery in an MS animal model. We previously demonstrated that a variant of the voltage-gated sodium channel NaV1.5 is expressed intracellularly in human macrophages, and that it regulates cellular signaling. This channel is not expressed in mouse macrophages, which has limited the study of its functions. To overcome this obstacle, we developed a novel transgenic mouse model (C57BL6), in which the human macrophage NaV1.5 splice variant is expressed in vivo in mouse macrophages. These mice were protected from experimental autoimmune encephalomyelitis, the mouse model of MS. During active inflammatory disease, NaV1.5-positive macrophages were found in spinal cord lesions where they formed phagocytic cell clusters; they expressed markers of alternative activation during recovery. NaV1.5-positive macrophages that were adoptively transferred into wild-type recipients with established experimental autoimmune encephalomyelitis homed to lesions and promoted recovery. These results suggest that NaV1.5-positive macrophages enhance recovery from CNS inflammatory disease and could potentially be developed as a cell-based therapy for the treatment of MS.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/prevenção & controle , Macrófagos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Recuperação de Função Fisiológica/genética , Animais , Linhagem Celular , Encefalomielite Autoimune Experimental/patologia , Humanos , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.5/biossíntese , Distribuição Aleatória , Recuperação de Função Fisiológica/fisiologia
6.
FEMS Immunol Med Microbiol ; 63(3): 319-27, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22092558

RESUMO

Phagocytosis and intracellular processing of mycobacteria by macrophages are complex cellular processes that require spatial and temporal coordination of particle uptake, organelle movement, activation of signaling pathways, and channel-mediated ionic flux. Recent work demonstrated that human macrophage NaV1.5, an intracellular voltage-gated sodium channel expressed on late endosomes, enhances endosomal acidification and phagocytosis. Here, using bacillus Camille-Guerin (BCG) as a model of mycobacterial infection, we examined how this channel regulates phagocytosis and phagosome maturation in human macrophages. Knockdown of NaV1.5 reduced high capacity uptake of labeled BCG. BCG-containing, NaV1.5-expressing cells demonstrated localization of NaV1.5 and Rab-7 positive endosomes and mitochondria to periphagosome regions that was not observed in NaV1.5-deficient cells. Knockdown of the channel reduced the initial calcium response following bacterial challenge and prevented the generation of prolonged and localized calcium oscillations during phagosome maturation. Inhibition of the mitochondrial Na(+) /Ca(2+) exchanger also prevented prolonged calcium oscillations during phagosome maturation. These results suggest that NaV1.5 and mitochondrial-dependent calcium signaling regulate mycobacteria phagocytosis and phagosome maturation in human macrophages through spatial-temporal coordination of calcium signaling within a unique subcellular region.


Assuntos
Sinalização do Cálcio , Endossomos/microbiologia , Macrófagos/microbiologia , Mycobacterium bovis/imunologia , Fagocitose , Canais de Sódio/metabolismo , Células Cultivadas , Endossomos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Canais de Sódio/genética
7.
Neurosci Lett ; 495(2): 115-20, 2011 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-21440040

RESUMO

Prior work demonstrated that immune surveillance of the brain occurs primarily through the blood-cerebrospinal (CSF) fluid barrier rather than the blood-brain barrier endothelium. Recently, we identified epithelial V-like antigen (EVA), an immunoglobulin-like adhesion molecule, as a regulator of blood-CSF barrier integrity in a mouse model. Here we characterized EVA expression and function in human choroid plexus epithelial cells and analyzed its role in CD4 T lymphocyte adhesion. In human choroid plexus epithelial cells and a subset of CD4 T lymphocytes, EVA is expressed at high levels. Epithelial adhesion of T lymphocytes is inhibited by a blocking monoclonal antibody that recognizes EVA. T cell adhesion elicits calcium flux in choroid plexus epithelial cells that also can be blocked by an EVA-specific antibody. EVA-positive cell-cell contacts between epithelial and T cells are associated with increased complexity of cytoskeletal epithelial morphology. These results demonstrate that EVA is expressed in human choroid plexus epithelial cells and CD4 T lymphocytes and regulates CD4+ T lymphocyte adhesion to human choroid plexus epithelial cells in vitro. These data suggest a novel mechanism to regulate CNS immune surveillance.


Assuntos
Moléculas de Adesão Celular/biossíntese , Plexo Corióideo/metabolismo , Células Epiteliais/fisiologia , Adulto , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Linfócitos T CD4-Positivos/fisiologia , Sinalização do Cálcio , Adesão Celular , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/imunologia , Células Cultivadas , Humanos
8.
J Biol Chem ; 284(12): 8114-26, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19136557

RESUMO

Voltage-gated sodium channels initiate electrical signaling in excitable cells such as muscle and neurons. They also are expressed in non-excitable cells such as macrophages and neoplastic cells. Previously, in macrophages, we demonstrated expression of SCN8A, the gene that encodes the channel NaV1.6, and intracellular localization of NaV1.6 to regions near F-actin bundles, particularly at areas of cell attachment. Here we show that a splice variant of NaV1.6 regulates cellular invasion through its effects on podosome and invadopodia formation in macrophages and melanoma cells. cDNA sequence analysis of SCN8A from THP-1 cells, a human monocyte-macrophage cell line, confirmed the expression of a full-length splice variant that lacks exon 18. Immunoelectron microscopy demonstrated NaV1.6-positive staining within the electron dense podosome rosette structure. Pharmacologic antagonism with tetrodotoxin (TTX) in differentiated THP-1 cells or absence of functional NaV1.6 through a naturally occurring mutation (med) in mouse peritoneal macrophages inhibited podosome formation. Agonist-mediated activation of the channel with veratridine caused release of sodium from cationic vesicular compartments, uptake by mitochondria, and mitochondrial calcium release through the Na/Ca exchanger. Invasion by differentiated THP-1 and HTB-66 cells, an invasive melanoma cell line, through extracellular matrix was inhibited by TTX. THP-1 invasion also was inhibited by small hairpin RNA knockdown of SCN8A. These results demonstrate that a variant of NaV1.6 participates in the control of podosome and invadopodia formation and suggest that intracellular sodium release mediated by NaV1.6 may regulate cellular invasion of macrophages and melanoma cells.


Assuntos
Processamento Alternativo , Estruturas da Membrana Celular/metabolismo , Macrófagos Peritoneais/metabolismo , Melanoma/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Sódio/metabolismo , Animais , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Éxons , Humanos , Macrófagos Peritoneais/patologia , Melanoma/patologia , Camundongos , Camundongos Mutantes , Mitocôndrias/metabolismo , Mutação , Canal de Sódio Disparado por Voltagem NAV1.6 , Invasividade Neoplásica , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Veratridina/farmacologia
9.
J Immunol ; 178(12): 7822-32, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17548620

RESUMO

Voltage-gated sodium channels expressed on the plasma membrane activate rapidly in response to changes in membrane potential in cells with excitable membranes such as muscle and neurons. Macrophages also require rapid signaling mechanisms as the first line of defense against invasion by microorganisms. In this study, our goal was to examine the role of intracellular voltage-gated sodium channels in macrophage function. We demonstrate that the cardiac voltage-gated sodium channel, NaV1.5, is expressed on the late endosome, but not the plasma membrane, in a human monocytic cell line, THP-1, and primary human monocyte-derived macrophages. Although the neuronal channel, NaV1.6, is also expressed intracellularly, it has a distinct subcellular localization. In primed cells, NaV1.5 regulates phagocytosis and endosomal pH during LPS-mediated endosomal acidification. Activation of the endosomal channel causes sodium efflux and decreased intraendosomal pH. These results demonstrate a functionally relevant intracellular voltage-gated sodium channel and reveal a novel mechanism to regulate macrophage endosomal acidification.


Assuntos
Endossomos/metabolismo , Macrófagos/imunologia , Fagocitose , Canais de Sódio/fisiologia , Ácidos/metabolismo , Membrana Celular/química , Células Cultivadas , Endossomos/química , Endossomos/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Lipopolissacarídeos/farmacologia , Macrófagos/ultraestrutura , Canal de Sódio Disparado por Voltagem NAV1.5 , Canal de Sódio Disparado por Voltagem NAV1.6 , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/metabolismo , Fagossomos/química , Fagossomos/metabolismo , Canais de Sódio/análise , Canais de Sódio/genética , Canais de Sódio/metabolismo , Tetrodotoxina/farmacologia , Veratridina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA