Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1397117, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39040107

RESUMEN

Intestinal epithelial cells possess the requisite molecular machinery to initiate cell-intrinsic defensive responses against intracellular pathogens, including intracellular parasites. Interferons(IFNs) have been identified as cornerstones of epithelial cell-intrinsic defense against such pathogens in the gastrointestinal tract. Long non-coding RNAs (lncRNAs) are RNA transcripts (>200 nt) not translated into protein and represent a critical regulatory component of mucosal defense. We report here that lncRNA Nostrill facilitates IFN-γ-stimulated intestinal epithelial cell-intrinsic defense against infection by Cryptosporidium, an important opportunistic pathogen in AIDS patients and a common cause of diarrhea in young children. Nostrill promotes transcription of a panel of genes controlled by IFN-γ through facilitating Stat1 chromatin recruitment and thus, enhances expression of several genes associated with cell-intrinsic defense in intestinal epithelial cells in response to IFN-γ stimulation, including Igtp, iNos, and Gadd45g. Induction of Nostrill enhances IFN-γ-stimulated intestinal epithelial defense against Cryptosporidium infection, which is associated with an enhanced autophagy in intestinal epithelial cells. Our findings reveal that Nostrill enhances the transcription of a set of genes regulated by IFN-γ in intestinal epithelial cells. Moreover, induction of Nostrill facilitates the IFN-γ-mediated epithelial cell-intrinsic defense against cryptosporidial infections.


Asunto(s)
Criptosporidiosis , Interferón gamma , Mucosa Intestinal , ARN Largo no Codificante , Interferón gamma/metabolismo , ARN Largo no Codificante/genética , Criptosporidiosis/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Mucosa Intestinal/metabolismo , Animales , Humanos , Transcripción Genética , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/parasitología , Ratones , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT1/genética , Cryptosporidium/genética , Cryptosporidium/inmunología , Regulación de la Expresión Génica , Autofagia/inmunología
2.
Front Immunol ; 14: 1205468, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37346046

RESUMEN

Cryptosporidium is a zoonotic apicomplexan parasite that infects the gastrointestinal epithelium and other mucosal surfaces in humans. It is an important opportunistic pathogen in AIDS patients and a leading cause of infectious diarrhea and diarrheal-related death in children worldwide. The intestinal epithelial cells provide the first line of defense against Cryptosporidium infection and play a central role in activating and regulating the host's antiparasitic response. Increasing evidence suggests that long noncoding RNAs (lncRNAs) participate in host-pathogen interactions and play a regulatory role in the pathogenesis of diseases but the underlying molecular mechanisms are not fully understood. We previously identified a panel of host lncRNAs that are upregulated in murine intestinal epithelial cells following Cryptosporidium infection, including U90926. We demonstrate here that U90926 is acting in a pro-parasitic manner in regulating intestinal epithelial cell-autonomous antiparasitic defense. Inhibition of U90926 resulted in a decreased infection burden of the parasite while overexpression of U90926 showed an increase in infection burden in cultured murine intestinal epithelial cells. Induction of U90926 suppressed transcription of epithelial defense genes involved in controlling Cryptosporidium infection through epigenetic mechanisms. Specifically, transcription of Aebp1, which encodes the Aebp1 protein, a potent modulator of inflammation and NF-κB signaling, was suppressed by U90926. Gain- or loss-of-function of Aebp1 in the host's epithelial cells caused reciprocal alterations in the infection burden of the parasite. Interestingly, Cryptosporidium carries the Cryptosporidium virus 1 (CSpV1), a double-stranded (ds) RNA virus coding two dsRNA fragments, CSpV1-dsRdRp and CSpV1-dsCA. Both CSpV1-dsRdRp and CSpV1-dsCA can be delivered into infected cells as previously reported. We found that cells transfected with in vitro transcribed CSpV1-dsCA or CSpV1-dsRdRp displayed an increased level of U90926, suggesting that CSpV1 is involved in the upregulation of U90926 during Cryptosporidium infection. Our study highlights a new strategy by Cryptosporidium to hijack a host lncRNA to suppress epithelial cell-autonomous antiparasitic defense and allow for a robust infection.


Asunto(s)
Antiinfecciosos , Criptosporidiosis , Cryptosporidium parvum , Cryptosporidium , ARN Largo no Codificante , Niño , Humanos , Animales , Ratones , Antiparasitarios , Cryptosporidium parvum/genética , ARN Largo no Codificante/genética , Criptosporidiosis/genética , Cryptosporidium/genética , Células Epiteliales
3.
Nat Commun ; 14(1): 1456, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36928642

RESUMEN

Cryptosporidium infects gastrointestinal epithelium and is a leading cause of infectious diarrhea and diarrheal-related death in children worldwide. There are no vaccines and no fully effective therapy available for the infection. Type II and III interferon (IFN) responses are important determinants of susceptibility to infection but the role for type I IFN response remains obscure. Cryptosporidium parvum virus 1 (CSpV1) is a double-stranded RNA (dsRNA) virus harbored by Cryptosporidium spp. Here we show that intestinal epithelial conditional Ifnar1-/- mice (deficient in type I IFN receptor) are resistant to C. parvum infection. CSpV1-dsRNAs are delivered into host cells and trigger type I IFN response in infected cells. Whereas C. parvum infection attenuates epithelial response to IFN-γ, loss of type I IFN signaling or inhibition of CSpV1-dsRNA delivery can restore IFN-γ-mediated protective response. Our findings demonstrate that type I IFN signaling in intestinal epithelial cells is detrimental to intestinal anti-C. parvum defense and Cryptosporidium uses CSpV1 to activate type I IFN signaling to evade epithelial antiparasitic response.


Asunto(s)
Criptosporidiosis , Cryptosporidium parvum , Interacciones Huésped-Parásitos , Interferón Tipo I , Animales , Ratones , Antiparasitarios/metabolismo , Antiparasitarios/farmacología , Criptosporidiosis/etiología , Criptosporidiosis/parasitología , Criptosporidiosis/virología , Cryptosporidium/patogenicidad , Cryptosporidium/virología , Cryptosporidium parvum/patogenicidad , Cryptosporidium parvum/virología , Interacciones Huésped-Parásitos/genética , Interferón Tipo I/metabolismo , Interferón Tipo I/farmacología , Virus ARN Bicatenario/metabolismo
4.
Front Immunol ; 13: 863957, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35464447

RESUMEN

The cells of the intestinal epithelium establish the frontline for host defense against pathogens in the gastrointestinal tract and play a vital role in the initiation of the immune response. Increasing evidence supports the role of long non-coding RNAs (lncRNAs) as critical regulators of diverse cellular processes, however, their role in antimicrobial host defense is incompletely understood. In this study, we provide evidence that the lncRNA Nostrill is upregulated in the intestinal epithelium following infection by Cryptosporidium parvum, a globally prevalent apicomplexan parasite that causes significant diarrheal disease and an important opportunistic pathogen in the immunocompromised and AIDS patients. Induction of Nostrill in infected intestinal epithelial cells was triggered by NF-κB signaling and was observed to enhance epithelial defense by decreasing parasitic infection burden. Nostrill participates in the transcriptional regulation of C. parvum-induced Irf7 expression through interactions with NF-κB p65, and induction of Nostrill promotes epigenetic histone modifications and occupancy of RNA polymerase II at the Irf7 promoter. Our data suggest that the induction of Nostrill promotes antiparasitic defense against C. parvum and enhances intestinal epithelial antimicrobial defense through contributions to transcriptional regulation of immune-related genes, such as Irf7.


Asunto(s)
Antiinfecciosos , Criptosporidiosis , Cryptosporidium parvum , Cryptosporidium , ARN Largo no Codificante , Criptosporidiosis/genética , Criptosporidiosis/parasitología , Cryptosporidium/genética , Cryptosporidium/metabolismo , Cryptosporidium parvum/genética , Humanos , FN-kappa B/metabolismo , ARN Largo no Codificante/genética
5.
mBio ; 12(5): e0212721, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34488445

RESUMEN

Interferon (IFN) signaling is key to mucosal immunity in the gastrointestinal tract, but cellular regulatory elements that determine interferon gamma (IFN-γ)-mediated antimicrobial defense in intestinal epithelial cells are not fully understood. We report here that a long noncoding RNA (lncRNA), GenBank accession no. XR_001779380, was increased in abundance in murine intestinal epithelial cells following infection by Cryptosporidium, an important opportunistic pathogen in AIDS patients and a common cause of diarrhea in young children. Expression of XR_001779380 in infected intestinal epithelial cells was triggered by TLR4/NF-κB/Cdc42 signaling and epithelial-specific transcription factor Elf3. XR_001779380 primed epithelial cells for IFN-γ-mediated gene transcription through facilitating Stat1/Swi/Snf-associated chromatin remodeling. Interactions between XR_001779380 and Prdm1, which is expressed in neonatal but not adult intestinal epithelium, attenuated Stat1/Swi/Snf-associated chromatin remodeling induced by IFN-γ, contributing to suppression of IFN-γ-mediated epithelial defense in neonatal intestine. Our data demonstrate that XR_001779380 is an important regulator in IFN-γ-mediated gene transcription and age-associated intestinal epithelial antimicrobial defense. IMPORTANCE Epithelial cells along the mucosal surface provide the front line of defense against luminal pathogen infection in the gastrointestinal tract. These epithelial cells represent an integral component of a highly regulated communication network that can transmit essential signals to cells in the underlying intestinal mucosa that, in turn, serve as targets of mucosal immune mediators. LncRNAs are recently identified long noncoding transcripts that can regulate gene transcription through their interactions with other effect molecules. In this study, we demonstrated that lncRNA XR_001779380 was upregulated in murine intestinal epithelial cells following infection by a mucosal protozoan parasite Cryptosporidium. Expression of XR_001779380 in infected cells primed host epithelial cells for IFN-γ-mediated gene transcription, relevant to age-dependent intestinal antimicrobial defense. Our data provide new mechanistic insights into how intestinal epithelial cells orchestrate intestinal mucosal defense against microbial infection.


Asunto(s)
Criptosporidiosis/inmunología , Cryptosporidium parvum/fisiología , Interferón gamma/inmunología , Mucosa Intestinal/inmunología , ARN Largo no Codificante/inmunología , Factores de Edad , Animales , Criptosporidiosis/genética , Criptosporidiosis/parasitología , Cryptosporidium parvum/genética , Células Epiteliales/inmunología , Células Epiteliales/parasitología , Humanos , Inmunidad Mucosa , Interferón gamma/genética , Mucosa Intestinal/parasitología , Ratones , FN-kappa B/genética , FN-kappa B/inmunología , ARN Largo no Codificante/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
6.
Front Immunol ; 12: 705232, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34295340

RESUMEN

Increasing evidence supports that N6-methyladenosine (m6A) mRNA modification may play an important role in regulating immune responses. Intestinal epithelial cells orchestrate gastrointestinal mucosal innate defense to microbial infection, but underlying mechanisms are still not fully understood. In this study, we present data demonstrating significant alterations in the topology of host m6A mRNA methylome in intestinal epithelial cells following infection by Cryptosporidium parvum, a coccidian parasite that infects the gastrointestinal epithelium and causes a self-limited disease in immunocompetent individuals but a life-threatening diarrheal disease in AIDS patients. Altered m6A methylation in mRNAs in intestinal epithelial cells following C. parvum infection is associated with downregulation of alpha-ketoglutarate-dependent dioxygenase alkB homolog 5 and the fat mass and obesity-associated protein with the involvement of NF-кB signaling. Functionally, m6A methylation statuses influence intestinal epithelial innate defense against C. parvum infection. Specifically, expression levels of immune-related genes, such as the immunity-related GTPase family M member 2 and interferon gamma induced GTPase, are increased in infected cells with a decreased m6A mRNA methylation. Our data support that intestinal epithelial cells display significant alterations in the topology of their m6A mRNA methylome in response to C. parvum infection with the involvement of activation of the NF-кB signaling pathway, a process that modulates expression of specific immune-related genes and contributes to fine regulation of epithelial antimicrobial defense.


Asunto(s)
Adenosina/análogos & derivados , Criptosporidiosis/inmunología , Cryptosporidium parvum/inmunología , Epitelio/inmunología , Inmunidad Innata , Mucosa Intestinal/inmunología , Procesamiento Postranscripcional del ARN , ARN Mensajero/inmunología , Adenosina/fisiología , Desmetilasa de ARN, Homólogo 5 de AlkB/antagonistas & inhibidores , Desmetilasa de ARN, Homólogo 5 de AlkB/biosíntesis , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/biosíntesis , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Animales , Sistemas CRISPR-Cas , GTP Fosfohidrolasas/biosíntesis , GTP Fosfohidrolasas/genética , Proteínas de Unión al GTP/biosíntesis , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica/inmunología , Humanos , Mucosa Intestinal/citología , Metilación , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética
7.
Microorganisms ; 9(1)2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33445463

RESUMEN

Cryptosporidium is a genus of protozoan parasites that infect the gastrointestinal epithelium of a variety of vertebrate hosts. Intestinal epithelial cells are the first line of defense and play a critical role in orchestrating host immunity against Cryptosporidium infection. To counteract host defense response, Cryptosporidium has developed strategies of immune evasion to promote parasitic replication and survival within epithelial cells, but the underlying mechanisms are largely unclear. Using various models of intestinal cryptosporidiosis, we found that Cryptosporidium infection caused suppression of mitogen-activated protein kinase (MAPK) signaling in infected murine intestinal epithelial cells. Whereas expression levels of most genes encoding the key components of the MAPK signaling pathway were not changed in infected intestinal epithelial cells, we detected a significant downregulation of p38/Mapk, MAP kinase-activated protein kinase 2 (Mk2), and Mk3 genes in infected host cells. Suppression of MAPK signaling was associated with an impaired intestinal epithelial defense against C. parvum infection. Our data suggest that cryptosporidial infection may suppress intestinal epithelial cell MAPK signaling associated with the evasion of host antimicrobial defense.

8.
PLoS Pathog ; 17(1): e1009241, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33481946

RESUMEN

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.


Asunto(s)
Criptosporidiosis/inmunología , Cryptosporidium/inmunología , Interferón Tipo I/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal , Animales , Animales Recién Nacidos , Criptosporidiosis/parasitología , Diarrea/inmunología , Diarrea/parasitología , Células Epiteliales/parasitología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/parasitología , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Intestinos/parasitología , Ratones , Transcripción Genética , Regulación hacia Arriba
9.
J Neuroinflammation ; 18(1): 16, 2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407594

RESUMEN

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.


Asunto(s)
Microglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , ARN Largo no Codificante/biosíntesis , Transcripción Genética/fisiología , Animales , Línea Celular , Células Cultivadas , Femenino , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/patología , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Óxido Nítrico Sintasa de Tipo II/genética , ARN Largo no Codificante/genética , Transcripción Genética/efectos de los fármacos
10.
Oncol Lett ; 20(3): 2191-2198, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32782536

RESUMEN

N6-methyladenosine (m6A) RNA modification regulates multiple biological functions. Methyltransferase like 3 (METTL3), one of the major N6-methyltransferases, is highly expressed in gastric cancer, but its potential role in disease is unclear. The current study knocked out METTL3 (METTL3-KO) in human gastric cancer AGS cells using CRISPR/Cas9. METTL3-KO AGS cells exhibited decreased m6A methylation levels. A significant inhibition of cell proliferation was observed in METTL3-KO AGS cells. Silencing METTL3 in AGS cells altered the expression profile of many effector molecules that were previously demonstrated to serve key roles in AGS cell proliferation, including the suppressor of cytokine signaling (SOCS) family of proteins. The results further demonstrated that SOCS2 upregulation in METTL3-KO AGS cells was associated with a decreased RNA decay rate. Furthermore, SOCS2 KO or SOCS2 overexpression caused a significant increase and decrease in AGS cell proliferation, respectively. The current data suggested that METTL3-KO in gastric cancer cells resulted in the suppression of cell proliferation by inducing SOCS2, suggesting a potential role of elevated METTL3 expression in gastric cancer progression.

11.
Infect Immun ; 87(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30642905

RESUMEN

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.


Asunto(s)
Criptosporidiosis/inmunología , Criptosporidiosis/parasitología , Cryptosporidium parvum/fisiología , Células Epiteliales/inmunología , Exosomas/inmunología , Intestinos/inmunología , Bazo/citología , Animales , Criptosporidiosis/genética , Células Epiteliales/parasitología , Exosomas/genética , Femenino , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/inmunología , Humanos , Intestinos/parasitología , Hígado/inmunología , Hígado/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/inmunología , Bazo/inmunología , Bazo/parasitología
12.
J Immunol ; 201(12): 3630-3640, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30446564

RESUMEN

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.


Asunto(s)
Criptosporidiosis/genética , Cryptosporidium parvum/fisiología , Mucosa Intestinal/fisiología , ARN Largo no Codificante/genética , Animales , Antiinfecciosos , Línea Celular , Criptosporidiosis/inmunología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Inmunidad/genética , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo
13.
J Infect Dis ; 218(8): 1336-1347, 2018 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-30052999

RESUMEN

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.


Asunto(s)
Cryptosporidium parvum/metabolismo , Células Epiteliales/parasitología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mucosa Intestinal/citología , ARN Protozoario/farmacología , Animales , Línea Celular , Cryptosporidium parvum/genética , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Activación Transcripcional
14.
Int J Parasitol ; 48(6): 423-431, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29438669

RESUMEN

Intestinal infection by Cryptosporidium parvum causes significant alterations in the gene expression profile in host epithelial cells. Previous 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 human intestinal cryptosporidiosis, we report here that trans-suppression of the cadherin 3 (CDH3) and lysyl oxidase like 4 (LOXL4) genes in human intestinal epithelial cells following C. parvum infection involves host delivery of the Cdg7_FLc_1000 RNA, a C. parvum RNA that has been previously demonstrated to be delivered into the nuclei of infected host cells. Downregulation of CDH3 and LOXL4 genes was detected in host epithelial cells following C. parvum infection or in cells expressing the parasite Cdg7_FLc_1000 RNA. Knockdown of Cdg7_FLc_1000 attenuated the trans-suppression of CDH3 and LOXL4 genes in host cells induced by infection. Interestingly, Cdg7_FLc_1000 was detected to be recruited to the promoter regions of both CDH3 and LOXL4 gene loci in host cells following C. parvum infection. Host delivery of Cdg7_FLc_1000 promoted the PH domain zinc finger protein 1 (PRDM1)-mediated H3K9 methylation associated with trans-suppression in the CDH3 gene locus, but not the LOXL4 gene. Therefore, our data suggest that host delivery of Cdg7_FLc_1000 causes CDH3 trans-suppression in human intestinal epithelial cells following C. parvum infection through PRDM1-mediated H3K9 methylation in the CDH3 gene locus, whereas Cdg7_FLc_1000 induces trans-suppression of the host LOXL4 gene through H3K9/H3K27 methylation-independent mechanisms.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Aminoácido Oxidorreductasas/metabolismo , Cadherinas/metabolismo , Cryptosporidium parvum/fisiología , Proteínas Protozoarias/farmacología , ARN Protozoario/metabolismo , Aminoácido Oxidorreductasas/genética , Cadherinas/genética , Línea Celular , Cryptosporidium parvum/genética , Regulación hacia Abajo , Células Epiteliales/metabolismo , Células Epiteliales/parasitología , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Proteína-Lisina 6-Oxidasa , Proteínas Protozoarias/metabolismo
15.
Vet Parasitol ; 251: 27-33, 2018 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-29426472

RESUMEN

Intestinal infection by the zoonotic protozoan, Cryptosporidium parvum, causes significant alterations in the gene expression profile in host epithelial cells. The molecular mechanisms of how C. parvum may modulate host cell gene transcription and the pathological significance of such alterations are largely unclear. Previous studies demonstrate that a panel of parasite RNA transcripts 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 impact of host delivery of C. parvum Cdg2_FLc_0220 RNA transcript on host gene expression profile. We found that alterations in host gene expression profile following C. parvum infection were partially associated with the nuclear delivery of Cdg2_FLc_0220. Specifically, we identified a total of 46 overlapping upregulated genes and 8 overlapping downregulated genes in infected cells and cells transfected with Full-Cdg2_FLc_0220. Trans-suppression of the DAZ interacting zinc finger protein 1 like (DZIP1L) gene, the top overlapping downregulated gene in host cells following C. parvum infection and cells transfected with Full-Cdg2_FLc_0220, was mediated by G9a, independent of PRDM1. Cdg2_FLc_0220-mediated trans-suppression of the DZIP1L gene was independent of H3K9 and H3K27 methylation. Data from this study provide additional evidence that delivery of C. parvum Cdg2_FLc_0220 RNA transcript in infected epithelial cells modulates the transcription of host genes, contributing to the alterations in the gene expression profile in host epithelial cells during C. parvum infection.


Asunto(s)
Cryptosporidium parvum/genética , Interacciones Huésped-Patógeno/genética , Mucosa Intestinal/parasitología , ARN Protozoario/genética , Transcripción Genética , Animales , Línea Celular Tumoral , Criptosporidiosis/parasitología , Cryptosporidium parvum/fisiología , Regulación de la Expresión Génica , Humanos , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Transcriptoma , Regulación hacia Arriba
16.
Parasitol Res ; 117(3): 831-840, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29374323

RESUMEN

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.


Asunto(s)
Criptosporidiosis/genética , Cryptosporidium parvum , Mucosa Intestinal/parasitología , ARN Protozoario/fisiología , beta-Defensinas/genética , Animales , Línea Celular , Criptosporidiosis/inmunología , Criptosporidiosis/parasitología , Cryptosporidium parvum/genética , Regulación de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Ratones
17.
J Infect Dis ; 217(1): 122-133, 2017 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-28961856

RESUMEN

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.


Asunto(s)
Movimiento Celular , Criptosporidiosis/patología , Cryptosporidium parvum/patogenicidad , Regulación hacia Abajo , Células Epiteliales/fisiología , ARN Protozoario/metabolismo , Esfingomielina Fosfodiesterasa/biosíntesis , Animales , Línea Celular , Modelos Animales de Enfermedad , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Enfermedades Intestinales/patología , Metilación , Ratones , Procesamiento Proteico-Postraduccional
18.
Cell Microbiol ; 19(11)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28655069

RESUMEN

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.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/biosíntesis , Cryptosporidium parvum/genética , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/biosíntesis , Interleucina-33/biosíntesis , Mucosa Intestinal/parasitología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/biosíntesis , Transcripción Genética/genética , Sistema de Transporte de Aminoácidos y+/genética , Animales , Línea Celular , Criptosporidiosis/parasitología , Criptosporidiosis/patología , Cryptosporidium parvum/patogenicidad , Epigénesis Genética , Células Epiteliales/parasitología , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Proteínas del Choque Térmico HSP72/genética , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Interleucina-33/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Metilación , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Protozoario/genética , ARN Interferente Pequeño/genética
19.
FASEB J ; 31(3): 1215-1225, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27979905

RESUMEN

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.


Asunto(s)
Activación de Macrófagos/genética , Macrófagos/inmunología , FN-kappa B/genética , ARN Largo no Codificante/genética , Animales , Línea Celular , Ensamble y Desensamble de Cromatina , Proteína HMGB1/metabolismo , Histonas/metabolismo , Ratones , FN-kappa B/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética
20.
J Infect Dis ; 215(4): 636-643, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28007919

RESUMEN

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.


Asunto(s)
Criptosporidiosis/genética , Células Epiteliales/parasitología , Interacciones Huésped-Patógeno/genética , ARN Protozoario/genética , Transcripción Genética , Línea Celular , Cryptosporidium parvum/patogenicidad , Células Epiteliales/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Intestinos/citología , Intestinos/parasitología , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...