Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
PLoS Pathog ; 17(1): e1009241, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33481946

RESUMO

The gastrointestinal epithelium guides the immune system to differentiate between commensal and pathogenic microbiota, which relies on intimate links with the type I IFN signal pathway. Epithelial cells along the epithelium provide the front line of host defense against pathogen infection in the gastrointestinal tract. Increasing evidence supports the regulatory potential of long noncoding RNAs (lncRNAs) in immune defense but their role in regulating intestinal epithelial antimicrobial responses is still unclear. Cryptosporidium, a protozoan parasite that infects intestinal epithelial cells, is an important opportunistic pathogen in AIDS patients and a common cause of diarrhea in young children in developing countries. Recent advances in Cryptosporidium research have revealed a strong type I IFN response in infected intestinal epithelial cells. We previously identified a panel of host cell lncRNAs that are upregulated in murine intestinal epithelial cells following microbial challenge. One of these lncRNAs, NR_033736, is upregulated in intestinal epithelial cells following Cryptosporidium infection and displays a significant suppressive effect on type I IFN-controlled gene transcription in infected host cells. NR_033736 can be assembled into the ISGF3 complex and suppresses type I IFN-mediated gene transcription. Interestingly, upregulation of NR_033736 itself is triggered by the type I IFN signaling. Moreover, NR_033736 modulates epithelial anti-Cryptosporidium defense. Our data suggest that upregulation of NR_033736 provides negative feedback regulation of type I IFN signaling through suppression of type I IFN-controlled gene transcription, and consequently, contributing to fine-tuning of epithelial innate defense against microbial infection.


Assuntos
Criptosporidiose/imunologia , Cryptosporidium/imunologia , Interferon Tipo I/metabolismo , RNA Longo não Codificante/genética , Transdução de Sinais , Animais , Animais Recém-Nascidos , Criptosporidiose/parasitologia , Diarreia/imunologia , Diarreia/parasitologia , Células Epiteliais/parasitologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/parasitologia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/parasitologia , Intestinos/parasitologia , Camundongos , Transcrição Gênica , Regulação para Cima
2.
J Neuroinflammation ; 18(1): 16, 2021 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-33407594

RESUMO

BACKGROUND: Microglia are resident immunocompetent and phagocytic cells in the CNS. Pro-inflammatory microglia, stimulated by microbial signals such as bacterial lipopolysaccharide (LPS), viral RNAs, or inflammatory cytokines, are neurotoxic and associated with pathogenesis of several neurodegenerative diseases. Long non-coding RNAs (lncRNA) are emerging as important tissue-specific regulatory molecules directing cell differentiation and functional states and may help direct proinflammatory responses of microglia. Characterization of lncRNAs upregulated in proinflammatory microglia, such as NR_126553 or 2500002B13Rik, now termed Nostrill (iNOS Transcriptional Regulatory Intergenic LncRNA Locus) increases our understanding of molecular mechanisms in CNS innate immunity. METHODS: Microglial gene expression array analyses and qRT-PCR were used to identify a novel long intergenic non-coding RNA, Nostrill, upregulated in LPS-stimulated microglial cell lines, LPS-stimulated primary microglia, and LPS-injected mouse cortical tissue. Silencing and overexpression studies, RNA immunoprecipitation, chromatin immunoprecipitation, chromatin isolation by RNA purification assays, and qRT-PCR were used to study the function of this long non-coding RNA in microglia. In vitro assays were used to examine the effects of silencing the novel long non-coding RNA in LPS-stimulated microglia on neurotoxicity. RESULTS: We report here characterization of intergenic lncRNA, NR_126553, or 2500002B13Rik now termed Nostrill (iNOS Transcriptional Regulatory Intergenic LncRNA Locus). Nostrill is induced by LPS stimulation in BV2 cells, primary murine microglia, and in cortical tissue of LPS-injected mice. Induction of Nostrill is NF-κB dependent and silencing of Nostrill decreased inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production in BV2 and primary microglial cells. Overexpression of Nostrill increased iNOS expression and NO production. RNA immunoprecipitation assays demonstrated that Nostrill is physically associated with NF-κB subunit p65 following LPS stimulation. Silencing of Nostrill significantly reduced NF-κB p65 and RNA polymerase II recruitment to the iNOS promoter and decreased H3K4me3 activating histone modifications at iNOS gene loci. In vitro studies demonstrated that silencing of Nostrill in microglia reduced LPS-stimulated microglial neurotoxicity. CONCLUSIONS: Our data indicate a new regulatory role of the NF-κB-induced Nostrill and suggest that Nostrill acts as a co-activator of transcription of iNOS resulting in the production of nitric oxide by microglia through modulation of epigenetic chromatin remodeling. Nostrill may be a target for reducing the neurotoxicity associated with iNOS-mediated inflammatory processes in microglia during neurodegeneration.


Assuntos
Microglia/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , RNA Longo não Codificante/biossíntese , Transcrição Gênica/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Feminino , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Síndromes Neurotóxicas/genética , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Óxido Nítrico Sintase Tipo II/genética , RNA Longo não Codificante/genética , Transcrição Gênica/efeitos dos fármacos
3.
Mol Biol Rep ; 48(1): 563-584, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33387198

RESUMO

Emerging evidence suggests that microglia can support neurogenesis. Little is known about the mechanisms by which microglia regulate the cortical environment and stimulate cortical neurogenesis. We used an in vitro co-culture model system to investigate the hypothesis that microglia respond to soluble signals from cortical cells, particularly following mechanical injury, to alter the cortical environment and promote cortical cell proliferation, differentiation, and survival. Analyses of cortical cell proliferation, cell death, neurogenic protein expression, and intracellular signaling were performed on uninjured and injured cortical cells in co-culture with microglial cell lines. Microglia soluble cues enhanced cortical cell viability and proliferation cortical cells. Co-culture of injured cortical cells with microglia significantly reduced cell death of cortical cells. Microglial co-culture significantly increased Nestin + and α-internexin + cortical cells. Multiplex ELISA and RT-PCR showed decreased pro-inflammatory cytokine production by microglia co-cultured with injured cortical cells. Inhibition of AKT phosphorylation in cortical cells blocked microglial-enhanced cortical cell viability and expression of neurogenic markers in vitro. This in vitro model system allows for assessment of the effect of microglial-derived soluble signals on cortical cell viability, proliferation, and stages of differentiation during homeostasis or following mechanical injury. These data suggest that microglia cells can downregulate inflammatory cytokine production following activation by mechanical injury to enhance proliferation of new cells capable of neurogenesis via activation of AKT intracellular signaling. Increasing our understanding of the mechanisms that drive microglial-enhanced cortical neurogenesis during homeostasis and following injury in vitro will provide useful information for future primary cell and in vivo studies.


Assuntos
Córtex Cerebral/crescimento & desenvolvimento , Microglia/citologia , Neurogênese/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Sobrevivência Celular/genética , Células Cultivadas , Córtex Cerebral/metabolismo , Técnicas de Cocultura , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Filamentos Intermediários/genética , Microglia/metabolismo , Nestina/genética , Neurônios/metabolismo , Cultura Primária de Células , Ratos , Transdução de Sinais/genética
4.
J Immunol ; 201(12): 3630-3640, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30446564

RESUMO

Cryptosporidium is an important opportunistic intestinal pathogen for immunocompromised individuals and a common cause of diarrhea in young children in developing countries. Gastrointestinal epithelial cells play a central role in activating and orchestrating host immune responses against Cryptosporidium infection, but underlying molecular mechanisms are not fully understood. We report in this paper that C. parvum infection causes significant alterations in long noncoding RNA (lncRNA) expression profiles in murine intestinal epithelial cells. Transcription of a panel of lncRNA genes, including NR_045064, in infected cells is controlled by the NF-κB signaling. Functionally, inhibition of NR_045064 induction increases parasite burden in intestinal epithelial cells. Induction of NR_045064 enhances the transcription of selected defense genes in host cells following C. parvum infection. Epigenetic histone modifications are involved in NR_045064-mediated transcription of associated defense genes in infected host cells. Moreover, the p300/MLL-associated chromatin remodeling is involved in NR_045064-mediated transcription of associated defense genes in intestinal epithelial cells following C. parvum infection. Expression of NR_045064 and associated genes is also identified in intestinal epithelium in C57BL/6J mice following phosphorothioate oligodeoxynucleotide or LPS stimulation. Our data demonstrate that lncRNAs, such as NR_045064, play a role in regulating epithelial defense against microbial infection.


Assuntos
Criptosporidiose/genética , Cryptosporidium parvum/fisiologia , Mucosa Intestinal/fisiologia , RNA Longo não Codificante/genética , Animais , Anti-Infecciosos , Linhagem Celular , Criptosporidiose/imunologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Imunidade/genética , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo
5.
Infect Immun ; 87(4)2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30642905

RESUMO

Cryptosporidium, a protozoan parasite that infects the gastrointestinal epithelium and other mucosal surfaces in humans and animals, is an important opportunistic pathogen in AIDS patients and one of the most common enteric pathogens affecting young children in developing regions. This parasite is referred to as a "minimally invasive" mucosal pathogen, and epithelial cells play a central role in activating and orchestrating host immune responses. We previously demonstrated that Cryptosporidium parvum infection stimulates host epithelial cells to release exosomes, and these released exosomes shuttle several antimicrobial peptides to carry out anti-C. parvum activity. In this study, we detected the upregulation of inflammatory genes in the liver and spleen following C. parvum intestinal infection in neonatal mice. Interestingly, exosomes released from intestinal epithelial cells following C. parvum infection could activate the nuclear factor kappa B signaling pathway and trigger inflammatory gene transcription in isolated primary splenocytes. Several epithelial cell-derived proteins and a subset of parasite RNAs were detected in the exosomes released from C. parvum-infected intestinal epithelial cells. Shuttling of these effector molecules, including the high mobility group box 1 protein, was involved in the induction of inflammatory responses in splenocytes induced by the exosomes released from infected cells. Our data indicate that exosomes released from intestinal epithelial cells upon C. parvum infection can activate immune cells by shuttling various effector molecules, a process that may be relevant to host systemic responses to Cryptosporidium infection.


Assuntos
Criptosporidiose/imunologia , Criptosporidiose/parasitologia , Cryptosporidium parvum/fisiologia , Células Epiteliais/imunologia , Exossomos/imunologia , Intestinos/imunologia , Baço/citologia , Animais , Criptosporidiose/genética , Células Epiteliais/parasitologia , Exossomos/genética , Feminino , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/imunologia , Humanos , Intestinos/parasitologia , Fígado/imunologia , Fígado/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/imunologia , Baço/imunologia , Baço/parasitologia
6.
J Immunol ; 199(10): 3571-3582, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28993514

RESUMO

Long noncoding RNAs, a newly identified class of noncoding RNAs, are important regulators of gene expression in innate immunity. We report in this study that the transcription of FIRRE, a conserved long noncoding RNA between humans and mice, is controlled by NF-κB signaling in macrophages and intestinal epithelial cells. Functionally, FIRRE appears to positively regulate the expression of several inflammatory genes in macrophages or intestinal epithelial cells in response to LPS stimulation via posttranscriptional mechanisms. Specifically, FIRRE physically interacts with heterogeneous nuclear ribonucleoproteins U, regulating the stability of mRNAs of selected inflammatory genes through targeting the AU-rich elements of their mRNAs in cells following LPS stimulation. Therefore, our data indicate a new regulatory role for NF-κB-responsive FIRRE in the posttranscriptional regulation of inflammatory genes in the innate immune system.


Assuntos
Sequência Conservada/genética , Inflamação/genética , Mucosa Intestinal/imunologia , Macrófagos/imunologia , NF-kappa B/metabolismo , RNA Longo não Codificante/genética , Elementos de Resposta/genética , Animais , Regulação da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Imunidade Inata , Camundongos , RNA Longo não Codificante/metabolismo , RNA Interferente Pequeno/genética , Células U937
7.
Eur Arch Otorhinolaryngol ; 276(8): 2283-2287, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31177327

RESUMO

PURPOSE: To assess autonomic nerve function in patients with laryngopharyngeal reflux disease (LPRD) and determine the correlation between LPRD and autonomic nerve dysfunction. METHODS: Patients with suspected LPRD who visited our outpatient department were assessed using the reflux symptom index (RSI) and reflux finding score (RFS) scales. Eighty-one suspected LPRD patients with RSI > 13 and RFS > 7 were examined using 5-min short-range heart rate variability, and all were given proton pump inhibitor diagnostic treatment. RESULTS: The root mean square of successive R-R intervals, high-frequency (HF) power, standardized HF, and HF % were significantly lower in the case group than in the control group (p < 0.05); however, the low frequency (LF)/HF ratio was significantly higher in the case group (p < 0.05). There were no significant differences in the standard deviation of the average normal-to-normal interval, total power, LF power, and LF % between the two groups (p > 0.05). RSI, RFS, and disease duration were negatively correlated with HF power (r = - 0.89, -0.77, and -0.315, respectively; p < 0.05). The LF/HF ratio and disease duration were positively correlated (r = 0.315, p < 0.05). CONCLUSIONS: Autonomic nerve dysfunction was observed in our patients with LPRD. LPRD severity was significantly correlated with autonomic nerve dysfunction and negatively correlated with vagal nerve function.


Assuntos
Doenças do Sistema Nervoso Autônomo/diagnóstico , Doenças do Sistema Nervoso Autônomo/etiologia , Vias Autônomas/fisiopatologia , Refluxo Laringofaríngeo/complicações , Adulto , Idoso , Feminino , Frequência Cardíaca , Humanos , Refluxo Laringofaríngeo/diagnóstico , Refluxo Laringofaríngeo/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Inibidores da Bomba de Prótons/uso terapêutico
8.
J Infect Dis ; 218(8): 1336-1347, 2018 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-30052999

RESUMO

Intestinal infection by Cryptosporidium is known to cause epithelial cell migration disorder but the underlying mechanisms are unclear. Previous studies demonstrated that a panel of parasite RNA transcripts of low protein-coding potential are delivered into infected epithelial cells. Using multiple models of intestinal cryptosporidiosis, we report here that C. parvum infection induces expression and release of the dickkopf protein 1 (Dkk1) from intestinal epithelial cells. Delivery of parasite Cdg7_FLc_1030 RNA to intestinal epithelial cells triggers transactivation of host Dkk1 gene during C. parvum infection. Release of Dkk1 is involved in C. parvum-induced inhibition of cell migration of epithelial cells, including noninfected bystander cells. Moreover, Dkk1-mediated suppression of host cell migration during C. parvum infection involves inhibition of Cdc42/Par6 signaling. Our data support the hypothesis that attenuation of intestinal epithelial cell migration during Cryptosporidium infection involves parasite Cdg7_FLc_1030 RNA-mediated induction and release of Dkk1 from infected cells.


Assuntos
Cryptosporidium parvum/metabolismo , Células Epiteliais/parasitologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Intestinal/citologia , RNA de Protozoário/farmacologia , Animais , Linhagem Celular , Cryptosporidium parvum/genética , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Ativação Transcricional
9.
J Biol Chem ; 292(30): 12375-12382, 2017 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-28615453

RESUMO

Long non-coding RNAs (lncRNAs) have emerged as potential key regulators of the inflammatory response, particularly by modulating the transcriptional control of inflammatory genes. lncRNAs may act as an enhancer or suppressor to inflammatory transcription, function as scaffold molecules through interactions with RNA-binding proteins in chromatin remodeling complexes, and modulate dynamic and epigenetic control of inflammatory transcription in a gene-specific and time-dependent fashion. Here, we will review recent literature regarding the role of lncRNAs in transcriptional control of inflammatory responses. Better understanding of lncRNA regulation of inflammation will provide novel targets for the development of new therapeutic strategies.


Assuntos
Inflamação/genética , RNA Longo não Codificante/genética , Transcrição Gênica , Animais , Humanos , Inflamação/metabolismo , RNA Longo não Codificante/metabolismo
10.
Cytokine ; 110: 29-38, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29698843

RESUMO

Creatinine is the breakdown product of creatine, a key participant in the generation of ATP and is traditionally considered to be a biologically inert waste product. Based on our earlier work, we analyzed the effects of creatinine hydrochloride on the expression of tumor necrosis factor-alpha (TNF-α), a pro-inflammatory cytokine, in a human T cell line, as well as human and mouse macrophage cell lines. Exposing cells to creatinine hydrochloride significantly reduced TNF-α mRNA and protein levels compared to control-treated cultures in all cell lines tested. Lipopolysaccharide (LPS), a potent inducer of inflammation, was employed with in mouse macrophage cell lines to induce high levels of TNF-α in order to determine whether creatinine hydrochloride could reduce preexisting inflammation. Cells treated with LPS and creatinine hydrochloride had significantly reduced TNF-α levels compared to cells treated with LPS alone. As the NF-κB signaling pathway represents a major mechanism of TNF-α generation, nuclear extracts were examined for NF-κB pathway activation. Cells exposed to CRN had significantly lower levels of NF-κB in the nucleus compared to control-treated cells. Together, these results support the hypothesis that CRN can alter anti-inflammatory responses by interfering with the activation of the NF-κB pathway.


Assuntos
Creatinina/metabolismo , Regulação para Baixo/fisiologia , Macrófagos/metabolismo , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Células Jurkat , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , NF-kappa B/metabolismo , Células RAW 264.7 , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Linfócitos T/efeitos dos fármacos , Células THP-1/efeitos dos fármacos , Células THP-1/metabolismo
11.
FASEB J ; 31(3): 1215-1225, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27979905

RESUMO

Long intergenic noncoding RNAs (lincRNAs) are long noncoding transcripts (>200 nt) from the intergenic regions of annotated protein-coding genes. We report here that the lincRNA gene lincRNA-Tnfaip3, located at mouse chromosome 10 proximal to the tumor necrosis factor α-induced protein 3 (Tnfaip3) gene, is an early-primary response gene controlled by nuclear factor-κB (NF-κB) signaling in murine macrophages. Functionally, lincRNA- Tnfaip3 appears to mediate both the activation and repression of distinct classes of inflammatory genes in macrophages. Specifically, induction of lincRNA-Tnfaip3 is required for the transactivation of NF-κB-regulated inflammatory genes in response to bacterial LPSs stimulation. LincRNA-Tnfaip3 physically interacts with the high-mobility group box 1 (Hmgb1), assembling a NF-κB/Hmgb1/lincRNA-Tnfaip3 complex in macrophages after LPS stimulation. This resultant NF-κB/Hmgb1/lincRNA-Tnfaip3 complex can modulate Hmgb1-associated histone modifications and, ultimately, transactivation of inflammatory genes in mouse macrophages in response to microbial challenge. Therefore, our data indicate a new regulatory role of NF-κB-induced lincRNA-Tnfaip3 to act as a coactivator of NF-κB for the transcription of inflammatory genes in innate immune cells through modulation of epigenetic chromatin remodeling.-Ma, S., Ming, Z., Gong, A.-Y., Wang, Y., Chen, X., Hu, G., Zhou, R., Shibata, A., Swanson, P. C., Chen, X.-M. A long noncoding RNA, LincRNA-Tnfaip3, acts as a coregulator of NF-κB to modulate inflammatory gene transcription in mouse macrophages.


Assuntos
Ativação de Macrófagos/genética , Macrófagos/imunologia , NF-kappa B/genética , RNA Longo não Codificante/genética , Animais , Linhagem Celular , Montagem e Desmontagem da Cromatina , Proteína HMGB1/metabolismo , Histonas/metabolismo , Camundongos , NF-kappa B/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética
12.
Cell Microbiol ; 19(11)2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28655069

RESUMO

Cryptosporidial infection causes dysregulated transcription of host genes key to intestinal epithelial homeostasis, but the underlying mechanisms remain obscure. Previous studies demonstrate that several Cryptosporidium parvum (C. parvum) RNA transcripts are selectively delivered into epithelial cells during host cell invasion and may modulate gene transcription in infected cells. We report here that C. parvum infection suppresses the transcription of LRP5, SLC7A8, and IL33 genes in infected intestinal epithelium. Trans-suppression of these genes in infected host cells is associated with promoter enrichment of suppressive epigenetic markers (i.e., H3K9me3). Cdg7_FLc_0990, a C. parvum RNA that has previously demonstrated to be delivered into the nuclei of infected epithelial cells, is recruited to the promoter regions of LRP5, SLC7A8, and IL33 genes. Cdg7_FLc_0990 appears to be recruited to their promoter regions together with G9a, a histone methyltransferase for H3K9 methylation. The PR domain zinc finger protein 1, a G9a-interacting protein, is required for the assembly of Cdg7_FLc_0990 to the G9a complex and gene-specific enrichment of H3K9 methylation. Our data demonstrate that cryptosporidial infection induces epigenetic histone methylations in infected cells through nuclear transfer of parasite Cdg7_Flc_0990 RNA transcript, resulting in transcriptional suppression of the LRP5, SLC7A8, and IL33 genes.


Assuntos
Sistema y+ de Transporte de Aminoácidos/biossíntese , Cryptosporidium parvum/genética , Cadeias Leves da Proteína-1 Reguladora de Fusão/biossíntese , Interleucina-33/biossíntese , Mucosa Intestinal/parasitologia , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/biossíntese , Transcrição Gênica/genética , Sistema y+ de Transporte de Aminoácidos/genética , Animais , Linhagem Celular , Criptosporidiose/parasitologia , Criptosporidiose/patologia , Cryptosporidium parvum/patogenicidade , Epigênese Genética , Células Epiteliais/parasitologia , Cadeias Leves da Proteína-1 Reguladora de Fusão/genética , Proteínas de Choque Térmico HSP72/genética , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Interleucina-33/genética , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Metilação , Camundongos , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Regiões Promotoras Genéticas/genética , Interferência de RNA , RNA de Protozoário/genética , RNA Interferente Pequeno/genética
13.
J Immunol ; 196(6): 2799-2808, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26880762

RESUMO

Long intergenic noncoding RNAs (lincRNAs) are long noncoding transcripts (>200 nt) from the intergenic regions of annotated protein-coding genes. One of the most highly induced lincRNAs in macrophages upon TLR ligation is lincRNA-Cox2, which was recently shown to mediate the activation and repression of distinct classes of immune genes in innate immune cells. We report that lincRNA-Cox2, located at chromosome 1 proximal to the PG-endoperoxide synthase 2 (Ptgs2/Cox2) gene, is an early-primary inflammatory gene controlled by NF-κB signaling in murine macrophages. Functionally, lincRNA-Cox2 is required for the transcription of NF-κB-regulated late-primary inflammatory response genes stimulated by bacterial LPS. Specifically, lincRNA-Cox2 is assembled into the switch/sucrose nonfermentable (SWI/SNF) complex in cells after LPS stimulation. This resulting lincRNA-Cox2/SWI/SNF complex can modulate the assembly of NF-κB subunits to the SWI/SNF complex, and ultimately, SWI/SNF-associated chromatin remodeling and transactivation of the late-primary inflammatory-response genes in macrophages in response to microbial challenge. Therefore, our data indicate a new regulatory role for NF-κB-induced lincRNA-Cox2 as a coactivator of NF-κB for the transcription of late-primary response genes in innate immune cells through modulation of epigenetic chromatin remodeling.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Inflamação/imunologia , Macrófagos Peritoneais/fisiologia , Microglia/fisiologia , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Montagem e Desmontagem da Cromatina , Cromossomos Humanos Par 1/genética , Ciclo-Oxigenase 2/genética , Humanos , Imunidade Inata/genética , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Longo não Codificante/genética , RNA Interferente Pequeno/genética , Ativação Transcricional/genética
14.
Parasitol Res ; 117(3): 831-840, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29374323

RESUMO

To counteract host immunity, Cryptosporidium parvum has evolved multiple strategies to suppress host antimicrobial defense. One such strategy is to reduce the production of the antimicrobial peptide beta-defensin 1 (DEFB1) by host epithelial cells but the underlying mechanisms remain unclear. Recent studies demonstrate that a panel of parasite RNA transcripts of low protein-coding potential are delivered into infected host cells and may modulate host gene transcription. Using in vitro models of intestinal cryptosporidiosis, in this study, we analyzed the expression profile of host beta-defensin genes in host cells following infection. We found that C. parvum infection caused a significant downregulation of the DEFB1 gene. Interestingly, downregulation of DEFB1 gene was associated with host delivery of Cdg7_FLc_1000 RNA transcript, a C. parvum RNA that has previously demonstrated to be delivered into the nuclei of infected host cells. Knockdown of Cdg7_FLc_1000 in host cells could attenuate the trans-suppression of host DEFB1 gene and decreased the parasite burden. Therefore, our data suggest that trans-suppression of DEFB1 gene in intestinal epithelial cells following C. parvum infection involves host delivery of parasite Cdg7_FLc_1000 RNA, a process that may be relevant to the epithelial defense evasion by C. parvum at the early stage of infection.


Assuntos
Criptosporidiose/genética , Cryptosporidium parvum , Mucosa Intestinal/parasitologia , RNA de Protozoário/fisiologia , beta-Defensinas/genética , Animais , Linhagem Celular , Criptosporidiose/imunologia , Criptosporidiose/parasitologia , Cryptosporidium parvum/genética , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Camundongos
15.
J Infect Dis ; 217(1): 122-133, 2017 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-28961856

RESUMO

Intestinal infection by Cryptosporidium parvum causes inhibition of epithelial turnover, but underlying mechanisms are unclear. Previous studies demonstrate that a panel of parasite RNA transcripts of low protein-coding potential are delivered into infected epithelial cells. Using in vitro and in vivo models of intestinal cryptosporidiosis, we report here that host delivery of parasite Cdg7_FLc_1000 RNA results in inhibition of epithelial cell migration through suppression of the gene encoding sphingomyelinase 3 (SMPD3). Delivery of Cdg7_FLc_1000 into infected cells promotes the histone methyltransferase G9a-mediated H3K9 methylation in the SMPD3 locus. The DNA-binding transcriptional repressor, PR domain zinc finger protein 1, is required for the assembly of Cdg7_FLc_1000 into the G9a complex and associated with the enrichment of H3K9 methylation at the gene locus. Pathologically, nuclear transfer of Cryptosporidium parvum Cdg7_FLc_1000 RNA is involved in the attenuation of intestinal epithelial cell migration via trans-suppression of host cell SMPD3.


Assuntos
Movimento Celular , Criptosporidiose/patologia , Cryptosporidium parvum/patogenicidade , Regulação para Baixo , Células Epiteliais/fisiologia , RNA de Protozoário/metabolismo , Esfingomielina Fosfodiesterase/biossíntese , Animais , Linhagem Celular , Modelos Animais de Doenças , Epigênese Genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Humanos , Enteropatias/patologia , Metilação , Camundongos , Processamento de Proteína Pós-Traducional
16.
J Infect Dis ; 215(4): 636-643, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28007919

RESUMO

Cryptosporidium parvum is an important opportunistic parasite pathogen for immunocompromised individuals and a common cause of diarrhea in young children. Previous studies have identified a panel of RNA transcripts of very low protein-coding potential in C. parvum. Using an in vitro model of human intestinal cryptosporidiosis, we report here that some of these C. parvum RNA transcripts were selectively delivered into the nuclei of host epithelial cells during C. parvum infection. Nuclear delivery of several such parasitic RNAs, including Cdg7_FLc_0990, involved heat-shock protein 70-mediated nuclear importing mechanism. Overexpression of Cdg7_FLc_0990 in intestinal epithelial cells resulted in significant changes in expression levels of specific genes, with significant overlapping with alterations in gene expression profile detected in host cells after C. parvum infection. Our data demonstrate that C. parvum transcripts of low protein-coding potential are selectively delivered into epithelial cells during infection and may modulate gene transcription in infected host cells.


Assuntos
Criptosporidiose/genética , Células Epiteliais/parasitologia , Interações Hospedeiro-Patógeno/genética , RNA de Protozoário/genética , Transcrição Gênica , Linhagem Celular , Cryptosporidium parvum/patogenicidade , Células Epiteliais/metabolismo , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Intestinos/citologia , Intestinos/parasitologia , Transcriptoma
17.
FASEB J ; 30(3): 1187-97, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26578685

RESUMO

Long intergenic noncoding RNAs (lincRNAs) can regulate the transcription of inflammatory genes and thus may represent a new group of inflammatory mediators with a potential pathogenic role in inflammatory diseases. Here, our genome-wide transcriptomic data show that TNF-α stimulation caused up-regulation of 171 lincRNAs and down-regulation of 196 lincRNAs in murine intestinal epithelial cells in culture. One of the up-regulated lincRNAs, lincRNA-Cox2, is an early-responsive lincRNA induced by TNF-α through activation of the NF-ĸB signaling pathway. Knockdown of lincRNA-Cox2 resulted in reprogramming of the gene expression profile in intestinal epithelial cells in response to TNF-α stimulation. Specifically, lincRNA-Cox2 silencing significantly (P < 0.05) enhanced the transcription of Il12b, a secondary late-responsive gene induced by TNF-α. Mechanistically, lincRNA-Cox2 promoted the recruitment of the Mi-2/nucleosome remodeling and deacetylase (Mi-2/NuRD) repressor complex to the Il12b promoter region. Recruitment of the Mi-2/NuRD complex was associated with decreased H3K27 acetylation and increased H3K27 dimethylation at the Il12b promoter region, which might contribute to Il12b trans-suppression by lincRNA-Cox2. Thus, our data demonstrate a novel mechanism of epigenetic modulation by lincRNA-Cox2 on Il12b transcription, supporting an important role for lincRNAs in the regulation of intestinal epithelial inflammatory responses.


Assuntos
Interleucina-12/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , RNA Longo não Codificante/genética , Fator de Necrose Tumoral alfa/metabolismo , Acetilação , Animais , Linhagem Celular , Ciclo-Oxigenase 2/metabolismo , Regulação para Baixo , Epigenômica/métodos , Células Epiteliais/metabolismo , Histonas/genética , Interleucina-12/metabolismo , Mucosa Intestinal/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Nucleossomos/genética , Regiões Promotoras Genéticas , RNA Longo não Codificante/metabolismo , Transcrição Gênica , Ativação Transcricional , Fator de Necrose Tumoral alfa/genética , Regulação para Cima
18.
PLoS Pathog ; 9(4): e1003261, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23592986

RESUMO

Exosomes are membranous nanovesicles released by most cell types from multi-vesicular endosomes. They are speculated to transfer molecules to neighboring or distant cells and modulate many physiological and pathological procedures. Exosomes released from the gastrointestinal epithelium to the basolateral side have been implicated in antigen presentation. Here, we report that luminal release of exosomes from the biliary and intestinal epithelium is increased following infection by the protozoan parasite Cryptosporidium parvum. Release of exosomes involves activation of TLR4/IKK2 signaling through promoting the SNAP23-associated vesicular exocytotic process. Downregulation of let-7 family miRNAs by activation of TLR4 signaling increases SNAP23 expression, coordinating exosome release in response to C. parvum infection. Intriguingly, exosomes carry antimicrobial peptides of epithelial cell origin, including cathelicidin-37 and beta-defensin 2. Activation of TLR4 signaling enhances exosomal shuttle of epithelial antimicrobial peptides. Exposure of C. parvum sporozoites to released exosomes decreases their viability and infectivity both in vitro and ex vivo. Direct binding to the C. parvum sporozoite surface is required for the anti-C. parvum activity of released exosomes. Biliary epithelial cells also increase exosomal release and display exosome-associated anti-C. parvum activity following LPS stimulation. Our data indicate that TLR4 signaling regulates luminal exosome release and shuttling of antimicrobial peptides from the gastrointestinal epithelium, revealing a new arm of mucosal immunity relevant to antimicrobial defense.


Assuntos
Criptosporidiose/imunologia , Cryptosporidium parvum/imunologia , Exossomos/metabolismo , Mucosa Intestinal/imunologia , Receptor 4 Toll-Like/metabolismo , Apresentação de Antígeno , Catelicidinas/metabolismo , Linhagem Celular , Ativação Enzimática , Células Epiteliais/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , MicroRNAs/biossíntese , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Interferência de RNA , Transdução de Sinais/imunologia , Esporozoítos/imunologia , Esporozoítos/metabolismo , beta-Defensinas/metabolismo
19.
PLoS Pathog ; 8(5): e1002702, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615562

RESUMO

Cryptosporidium is a protozoan parasite that infects the gastrointestinal epithelium and causes a diarrheal disease. Toll-like receptor (TLR)- and NF-κB-mediated immune responses from epithelial cells, such as production of antimicrobial peptides and generation of reactive nitrogen species, are important components of the host's defense against cryptosporidial infection. Here we report data demonstrating a role for miR-27b in the regulation of TLR4/NF-κB-mediated epithelial anti-Cryptosporidium parvum responses. We found that C. parvum infection induced nitric oxide (NO) production in host epithelial cells in a TLR4/NF-κB-dependent manner, with the involvement of the stabilization of inducible NO synthase (iNOS) mRNA. C. parvum infection of epithelial cells activated NF-κB signaling to increase transcription of the miR-27b gene. Meanwhile, downregulation of KH-type splicing regulatory protein (KSRP) was detected in epithelial cells following C. parvum infection. Importantly, miR-27b targeted the 3'-untranslated region of KSRP, resulting in translational suppression. C. parvum infection decreased KSRP expression through upregulating miR-27b. Functional manipulation of KSRP or miR-27b caused reciprocal alterations in iNOS mRNA stability in infected cells. Forced expression of KSRP and inhibition of miR-27b resulted in an increased burden of C. parvum infection. Downregulation of KSRP through upregulating miR-27b was also detected in epithelial cells following LPS stimulation. These data suggest that miR-27b targets KSRP and modulates iNOS mRNA stability following C. parvum infection, a process that may be relevant to the regulation of epithelial anti-microbial defense in general.


Assuntos
Criptosporidiose/imunologia , Cryptosporidium parvum/imunologia , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptor 4 Toll-Like/imunologia , Transativadores/metabolismo , Animais , Linhagem Celular , Cryptosporidium parvum/genética , Células Epiteliais/metabolismo , Trato Gastrointestinal/parasitologia , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Splicing de RNA , RNA Mensageiro , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Transcrição Gênica
20.
J Immunol ; 188(3): 1266-74, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22219331

RESUMO

Aberrant cellular responses to proinflammatory cytokines, such as TNF-α, are pathogenic features in most chronic inflammatory diseases. A variety of extracellular and intracellular feedback pathways has evolved to prevent an inappropriate cellular reaction to these proinflammatory cytokines. In this study, we report that TNF-α treatment of human and mouse cholangiocytes and hepatocytes downregulated expression of p300/CBP-associated factor (PCAF), a coactivator and an acetyltransferase that promotes histone acetylation and gene transcription. Of these upregulated microRNAs in TNF-α-treated cells, miR-181a/b (miR-181a and miR-181b) suppressed translation of PCAF mRNA. Functional manipulation of miR-181a/b caused reciprocal alterations in PCAF protein expression in cultured cholangiocytes and hepatocytes. Inhibition of miR-181a/b function with anti-miRs blocked TNF-α-induced suppression of PCAF expression. Promoter recruitment of PCAF was shown to be associated with TNF-α-induced transcription of inflammatory genes. Intriguingly, pretreatment of cells with TNF-α inhibited transcription of inflammatory genes in response to subsequent TNF-α stimulation. Overexpression of PCAF or inhibition of miR-181a/b function with anti-miRs attenuated the inhibitory effects of TNF-α pretreatment on epithelial inflammatory response to subsequent TNF-α stimulation. Downregulation of PCAF and the inhibitory effects of TNF-α pretreatment on liver epithelial inflammatory response were further confirmed in a mouse model of TNF-α i.p. injection. These data suggest that PCAF is a target for miR-181a/b, and downregulation of PCAF by TNF-α provides negative feedback regulation to inflammatory reactions in liver epithelial cells, a process that may be relevant to the epigenetic fine-tuning of epithelial inflammatory processes in general.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Retroalimentação Fisiológica , Hepatócitos/metabolismo , Inflamação , MicroRNAs/genética , Fator de Necrose Tumoral alfa/farmacologia , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Animais , Humanos , Inflamação/genética , Camundongos , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa