Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Immunity ; 56(4): 753-767.e8, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37001519

RESUMEN

Intracellular sensing of lipopolysaccharide (LPS) by murine caspase-11 or human caspase-4 initiates a protease cascade, termed the non-canonical inflammasome, that results in gasdermin D (GSDMD) processing and subsequent NLRP3 inflammasome activation. In an effort aimed at identifying additional sensors for intracellular LPS by biochemical screening, we identified the nuclear orphan receptor Nur77 as an LPS-binding protein in macrophage lysates. Nr4a1-/- macrophages exhibited impaired activation of the NLRP3 inflammasome, but not caspase-11, in response to LPS. Biochemical mapping revealed that Nur77 bound LPS directly through a domain in its C terminus. Yeast two-hybrid assays identified NLRP3 as a binding partner for Nur77. The association between Nur77 and NLRP3 required the presence of LPS and dsDNA. The source of dsDNA was the mitochondria, requiring the formation of gasdermin-D pores. In vivo, Nur77 deficiency ameliorated host response to endotoxins. Thus, Nur77 functions as an intracellular LPS sensor, binding mitochondrial DNA and LPS to activate the non-canonical NLRP3 inflammasome.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Animales , Humanos , Ratones , Caspasa 1/metabolismo , Caspasas/metabolismo , Gasderminas , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo
2.
Cell ; 171(1): 201-216.e18, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28844693

RESUMEN

An emerging family of innate lymphoid cells (termed ILCs) has an essential role in the initiation and regulation of inflammation. However, it is still unclear how ILCs are regulated in the duration of intestinal inflammation. Here, we identify a regulatory subpopulation of ILCs (called ILCregs) that exists in the gut and harbors a unique gene identity that is distinct from that of ILCs or regulatory T cells (Tregs). During inflammatory stimulation, ILCregs can be induced in the intestine and suppress the activation of ILC1s and ILC3s via secretion of IL-10, leading to protection against innate intestinal inflammation. Moreover, TGF-ß1 is induced by ILCregs during the innate intestinal inflammation, and autocrine TGF-ß1 sustains the maintenance and expansion of ILCregs. Therefore, ILCregs play an inhibitory role in the innate immune response, favoring the resolution of intestinal inflammation.


Asunto(s)
Colitis/inmunología , Inmunidad Innata , Linfocitos/citología , Linfocitos/inmunología , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Animales , Linfocitos B/inmunología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Organismos Libres de Patógenos Específicos , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta1/inmunología
3.
Nat Immunol ; 19(2): 141-150, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29292386

RESUMEN

Cyclic diadenylate monophosphate (c-di-AMP) is secreted by bacteria as a secondary messenger. How immune cells detect c-di-AMP and initiate anti-bacterial immunity remains unknown. We found that the endoplasmic reticulum (ER) membrane adaptor ERAdP acts as a direct sensor for c-di-AMP. ERAdP-deficient mice were highly susceptible to Listeria monocytogenes infection and exhibited reduced pro-inflammatory cytokines. Mechanistically, c-di-AMP bound to the C-terminal domain of ERAdP, which in turn led to dimerization of ERAdP, resulting in association with and activation of the kinase TAK1. TAK1 activation consequently initiated activation of the transcription factor NF-κB to induce the production of pro-inflammatory cytokines in innate immune cells. Moreover, double-knockout of ERAdP and TAK1 resulted in heightened susceptibility to L. monocytogenes infection. Thus, ERAdP-mediated production of pro-inflammatory cytokines is critical for controlling bacterial infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Fosfatos de Dinucleósidos/inmunología , Inmunidad Innata/inmunología , Listeriosis/inmunología , Proteínas de la Membrana/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sistemas de Mensajero Secundario/inmunología
4.
Nat Immunol ; 17(4): 369-78, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26829768

RESUMEN

Cyclic GMP-AMP synthase (cGAS) senses cytosolic DNA during viral infection and catalyzes synthesis of the dinucleotide cGAMP, which activates the adaptor STING to initiate antiviral responses. Here we found that deficiency in the carboxypeptidase CCP5 or CCP6 led to susceptibility to DNA viruses. CCP5 and CCP6 were required for activation of the transcription factor IRF3 and interferons. Polyglutamylation of cGAS by the enzyme TTLL6 impeded its DNA-binding ability, whereas TTLL4-mediated monoglutamylation of cGAS blocked its synthase activity. Conversely, CCP6 removed the polyglutamylation of cGAS, whereas CCP5 hydrolyzed the monoglutamylation of cGAS, which together led to the activation of cGAS. Therefore, glutamylation and deglutamylation of cGAS tightly modulate immune responses to infection with DNA viruses.


Asunto(s)
Carboxipeptidasas/genética , Infecciones por Virus ADN/metabolismo , ADN Viral/inmunología , Nucleotidiltransferasas/metabolismo , Péptido Sintasas/metabolismo , Animales , Citosol , Virus ADN/genética , Técnica del Anticuerpo Fluorescente , Herpes Simple/metabolismo , Inmunoprecipitación , Factor 3 Regulador del Interferón/inmunología , Interferones/inmunología , Ratones , Ratones Noqueados , Nucleótidos Cíclicos/biosíntesis , Nucleotidiltransferasas/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simplexvirus/genética , Vaccinia/metabolismo , Virus Vaccinia/genética
5.
Nat Immunol ; 16(4): 366-75, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25729924

RESUMEN

Neutrophils express Toll-like receptors (TLRs) for the recognition of conserved bacterial elements to initiate antimicrobial responses. However, whether other cytosolic DNA sensors are expressed by neutrophils remains elusive. Here we found constitutive expression of the transcription factor Sox2 in the cytoplasm of mouse and human neutrophils. Neutrophil-specific Sox2 deficiency exacerbated bacterial infection. Sox2 directly recognized microbial DNA through its high-mobility-group (HMG) domain. Upon challenge with bacterial DNA, Sox2 dimerization was needed to activate a complex of the kinase TAK1 and its binding partner TAB2, which led to activation of the transcription factors NF-κB and AP-1 in neutrophils. Deficiency in TAK1 or TAB2 impaired Sox2-mediated antibacterial immunity. Overall, we reveal a previously unrecognized role for Sox2 as a cytosolic sequence-specific DNA sensor in neutrophils, which might provide potential therapeutic strategies for the treatment of infectious diseases.


Asunto(s)
ADN Bacteriano/inmunología , Inmunidad Innata , Listeriosis/inmunología , Neutrófilos/inmunología , Factores de Transcripción SOXB1/inmunología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Citoplasma/inmunología , Citoplasma/microbiología , Regulación de la Expresión Génica , Humanos , Listeria monocytogenes/inmunología , Listeriosis/genética , Listeriosis/microbiología , Listeriosis/mortalidad , Quinasas Quinasa Quinasa PAM/deficiencia , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Ratones , FN-kappa B/genética , FN-kappa B/inmunología , Neutrófilos/microbiología , Multimerización de Proteína , Factores de Transcripción SOXB1/genética , Transducción de Señal , Análisis de Supervivencia , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/inmunología
6.
Immunity ; 48(4): 688-701.e7, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29625897

RESUMEN

Disrupting the balance between self-renewal and differentiation of hematopoietic stem cells (HSCs) leads to bone marrow failure or hematologic malignancy. However, how HSCs sustain their quiescent state and avoid type I interferon (IFN)-mediated exhaustion remains elusive. Here we defined a circular RNA that we named cia-cGAS that was highly expressed in the nucleus of long-term (LT)-HSCs. Cia-cGAS deficiency in mice caused elevated expression of type I IFNs in bone marrow and led to decreased numbers of dormant LT-HSCs. Under homeostatic conditions, cia-cGAS bound DNA sensor cGAS in the nucleus to block its synthase activity, thereby protecting dormant LT-HSCs from cGAS-mediated exhaustion. Moreover, cia-cGAS harbored a stronger binding affinity to cGAS than self-DNA did and consequently suppressed cGAS-mediated production of type I IFNs in LT-HSCs. Our findings reveal a mechanism by which cia-cGAS inhibits nuclear cGAS by blocking its enzymatic activity and preventing cGAS from recognizing self-DNA to maintain host homeostasis.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Interferón Tipo I/metabolismo , Nucleotidiltransferasas/metabolismo , ARN/metabolismo , Animales , Médula Ósea/metabolismo , Comunicación Celular , Línea Celular , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Conformación de Ácido Nucleico , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/genética , ARN/genética , Interferencia de ARN , ARN Circular , ARN Interferente Pequeño/genética
7.
Mol Cell ; 74(2): 363-377.e5, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30879902

RESUMEN

In eukaryotic cells, RNA-binding proteins (RBPs) interact with RNAs to form ribonucleoprotein complexes (RNA granules) that have long been thought to regulate RNA fate or activity. Emerging evidence suggests that some RBPs not only bind RNA but also possess enzymatic activity related to ubiquitin regulation, raising important questions of whether these RBP-formed RNA granules regulate ubiquitin signaling and related biological functions. Here, we show that Drosophila Otu binds RNAs and coalesces to membrane-less biomolecular condensates via its intrinsically disordered low-complexity domain, and coalescence represents a functional state for Otu exerting deubiquitinase activity. Notably, coalescence-mediated enzymatic activity of Otu is positively regulated by its bound RNAs and co-partner Bam. Further genetic analysis reveals that the Otu/Bam deubiquitinase complex and dTraf6 constitute a feedback loop to maintain intestinal immune homeostasis during aging, thereby controlling longevity. Thus, regulated biomolecular condensates may represent a mechanism that controls dynamic enzymatic activities and related biological processes.


Asunto(s)
Proteínas de Drosophila/genética , Longevidad/genética , Factor 6 Asociado a Receptor de TNF/genética , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Enzimas Desubicuitinizantes , Drosophila/genética , Longevidad/fisiología , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/genética , Ubiquitina/genética
8.
Nat Chem Biol ; 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38413746

RESUMEN

Intracellular recognition of lipopolysaccharide (LPS) by mouse caspase-11 or human caspase-4 is a vital event for the activation of the noncanonical inflammasome. Whether negative regulators are involved in intracellular LPS sensing is still elusive. Here we show that adipose triglyceride lipase (ATGL) is a negative regulator of the noncanonical inflammasome. Through screening for genes participating in the noncanonical inflammasome, ATGL is identified as a negative player for intracellular LPS signaling. ATGL binds LPS and catalyzes the removal of the acylated side chains that contain ester bonds. LPS with under-acylated side chains no longer activates the inflammatory caspases. Cells with ATGL deficiency exhibit enhanced immune responses when encountering intracellular LPS, including an elevated secretion of interleukin-1ß, decreased cell viability and increased cell cytotoxicity. Moreover, ATGL-deficient mice show exacerbated responses to endotoxin challenges. Our results uncover that ATGL degrades cytosolic LPS to suppress noncanonical inflammasome activation.

9.
EMBO J ; 40(18): e108249, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34296442

RESUMEN

SARS-CoV-2 is an emerging coronavirus that causes dysfunctions in multiple human cells and tissues. Studies have looked at the entry of SARS-CoV-2 into host cells mediated by the viral spike protein and human receptor ACE2. However, less is known about the cellular immune responses triggered by SARS-CoV-2 viral proteins. Here, we show that the nucleocapsid of SARS-CoV-2 inhibits host pyroptosis by blocking Gasdermin D (GSDMD) cleavage. SARS-CoV-2-infected monocytes show enhanced cellular interleukin-1ß (IL-1ß) expression, but reduced IL-1ß secretion. While SARS-CoV-2 infection promotes activation of the NLRP3 inflammasome and caspase-1, GSDMD cleavage and pyroptosis are inhibited in infected human monocytes. SARS-CoV-2 nucleocapsid protein associates with GSDMD in cells and inhibits GSDMD cleavage in vitro and in vivo. The nucleocapsid binds the GSDMD linker region and hinders GSDMD processing by caspase-1. These insights into how SARS-CoV-2 antagonizes cellular inflammatory responses may open new avenues for treating COVID-19 in the future.


Asunto(s)
COVID-19/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Nucleocápside/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Piroptosis/fisiología , SARS-CoV-2/metabolismo , Enzima Convertidora de Angiotensina 2/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/inmunología , COVID-19/patología , COVID-19/virología , Caspasa 1/inmunología , Caspasa 1/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Ratones , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Unión a Fosfato/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células THP-1
10.
Immunity ; 45(1): 131-44, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27421702

RESUMEN

Natural killer (NK) cells and non-cytotoxic interferon-γ (IFN-γ)-producing group I innate lymphoid cells (ILC1s) produce large amounts of IFN-γ and cause activation of innate and adaptive immunity. However, how NKs and ILC1s are primed during infection remains elusive. Here we have shown that a lymphocyte subpopulation natural killer-like B (NKB) cells existed in spleen and mesenteric lymph nodes (MLNs). NKBs had unique features that differed from T and B cells, and produced interleukin-18 (IL-18) and IL-12 at an early phase of infection. NKB cells played a critical role in eradication of microbial infection via secretion of IL-18 and IL-12. Moreover, IL-18 deficiency abrogated the antibacterial effect of NKBs. Upon bacterial challenge, NKB precursors (NKBPs) rapidly differentiated to NKBs that activated NKs and ILC1s against microbial infection. Our findings suggest that NKBs might be exploited to develop effective therapies for treatment of infectious diseases.


Asunto(s)
Linfocitos B/inmunología , Infecciones/inmunología , Células Asesinas Naturales/inmunología , Ganglios Linfáticos/inmunología , Subgrupos Linfocitarios/inmunología , Bazo/inmunología , Animales , Linfocitos B/microbiología , Diferenciación Celular , Células Cultivadas , Humanos , Inmunidad Innata , Infecciones/terapia , Interleucina-12/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Células Asesinas Naturales/microbiología , Activación de Linfocitos , Subgrupos Linfocitarios/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Precursoras de Linfocitos B/inmunología
12.
EMBO J ; 37(8)2018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29535137

RESUMEN

Divergent long noncoding RNAs (lncRNAs) represent a major lncRNA biotype in mouse and human genomes. The biological and molecular functions of the divergent lncRNAs remain largely unknown. Here, we show that lncKdm2b, a divergent lncRNA for Kdm2b gene, is conserved among five mammalian species and highly expressed in embryonic stem cells (ESCs) and early embryos. LncKdm2b knockout impairs ESC self-renewal and causes early embryonic lethality. LncKdm2b can activate Zbtb3 by promoting the assembly and ATPase activity of Snf2-related CREBBP activator protein (SRCAP) complex in trans Zbtb3 potentiates the ESC self-renewal in a Nanog-dependent manner. Finally, Zbtb3 deficiency impairs the ESC self-renewal and early embryonic development. Therefore, our findings reveal that lncRNAs may represent an additional layer of the regulation of ESC self-renewal and early embryogenesis.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Madre Embrionarias/metabolismo , ARN Largo no Codificante/genética , Animales , Desarrollo Embrionario , Humanos , Ratones Noqueados
13.
Mediators Inflamm ; 2022: 2408598, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35677735

RESUMEN

Background: Cardiac and brain inflammation can lead to a host of deleterious health effects. Our formal experimental research showed that Ginkgo Biloba Extract (GBE) contributed to the reduction of inflammation in mice with myocardial infarction along with depression. This study is aimed at expanding on these findings via analysis of the cardiac and brain inflammation, which was prevented by GBE in rats suffering with a high-fat diet (HFD) combined with unpredictable chronic mild stress (UCMS). Methods: Fifty male Wistar rats were randomly divided into 5 groups treated with normal diet, UCMS, HFD, HFD+UCMS, or HFD+UCMS+GBE respectively. Rats treated with HFD were fed a high-fat diet for 10 or 13 weeks. Rats treated with UCMS were exposed to 8 types of chronic physical and psychological stressors for 10 or 13 weeks. The HFD+UCMS+GBE group was given GBE via intragastric gavage for 8 consecutive weeks. Sucrose preference was established for the assessment of depressive behaviors. The heart function was evaluated by echocardiography. The rats were terminated at the end of the 10th or 13th week. The blood was used for detecting low-density lipoprotein cholesterol (LDL-c) and total cholesterol (TCHO) by the kit instructions; Helper T Lymphocytes (TH cells, CD3+CD4+) by flow cytometry; and Interleukin- (IL-) 1ß, IL-37, IL-38, NT-proBNP, hs-cTNI, and Ischemia-modified albumin (IMA) by enzyme-linked immunosorbent assay (ELISA). The cardiac tissues were used for detecting IL-1ß, nuclear factor kappa B (NF-κB), inhibitor molecule protein (IκB), and IL-1 receptor (IL-1R) by ELISA and P65, P-P65, IκB, and phosphorylated inhibitor molecule protein α (P-IκBα) for western blotting. Cortex tissues were used for detecting 8-iso-prostaglandinF2α (8-iso-PGF2α) by ELISA. Oil Red staining was carried out to evaluate the lipid deposits in the rats' aortic arteries. Sirius Red staining was performed to display collagen fibers in the arteries. Hematoxylin and Eosin (HE) staining was applied to reveal pathological changes to arteries and cardiac tissue. Immunohistochemical staining was employed to assess the distribution of inflammatory cytokine IL-1ß in arteries and cardiac tissues. Transmission Electron Microscopy (TEM) was performed to observe the ultrastructure of hippocampal cornu ammonis (CA)1 (CA1) neurons. Results: In the rats with HFD+UCMS+GBE, over 13 weeks, GBE exerted a protective role of both the heart and brain, by attenuating cardiac inflammation and brain oxidative stress. Levels of Helper T lymphocytes and serum anti-inflammatory cytokines involving IL-37 and IL-38 were all elevated, and the depressive behaviors of HFD+UCMS rats were attenuated by GBE. This protective role was accomplished via inhibition of the canonical NF-κB signaling pathway, through downregulation of the expressions of P-P65 and P-IκB-α in the heart, hippocampus, cortex, and hypothalamus. Conclusions: This study suggests that GBE poses a protective role from the various pathologies associated with high-fat diets, unpredictable chronic mild stress, and depression, possibly via improving peripheral immunity and reducing cardiac and brain inflammation.


Asunto(s)
Encefalitis , FN-kappa B , Animales , Biomarcadores , Colesterol , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Ginkgo biloba/química , Ginkgo biloba/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Ratones , FN-kappa B/metabolismo , Extractos Vegetales , Ratas , Ratas Wistar , Albúmina Sérica , Estrés Psicológico/tratamiento farmacológico
15.
EMBO J ; 32(20): 2685-96, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23974797

RESUMEN

Autophagy degrades cytoplasmic proteins and organelles to recycle cellular components that are required for cell survival and tissue homeostasis. However, it is not clear how autophagy is regulated in mammalian cells. WASH (Wiskott-Aldrich syndrome protein (WASP) and SCAR homologue) plays an essential role in endosomal sorting through facilitating tubule fission via Arp2/3 activation. Here, we demonstrate a novel function of WASH in modulation of autophagy. We show that WASH deficiency causes early embryonic lethality and extensive autophagy of mouse embryos. WASH inhibits vacuolar protein sorting (Vps)34 kinase activity and autophagy induction. We identified that WASH is a new interactor of Beclin 1. Beclin 1 is ubiquitinated at lysine 437 through lysine 63 linkage in cells undergoing autophagy. Ambra1 is an E3 ligase for lysine 63-linked ubiquitination of Beclin 1 that is required for starvation-induced autophagy. The lysine 437 ubiquitination of Beclin 1 enhances the association with Vps34 to promote Vps34 activity. WASH can suppress Beclin 1 ubiquitination to inactivate Vps34 activity leading to suppression of autophagy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/genética , Pérdida del Embrión/genética , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/fisiología , Ubiquitinación/genética , Proteínas de Transporte Vesicular/fisiología , Animales , Beclina-1 , Células Cultivadas , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Regulación hacia Abajo/genética , Embrión de Mamíferos/metabolismo , Genes Letales/fisiología , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Transporte Vesicular/genética
16.
Cell Mol Life Sci ; 72(9): 1699-713, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25572296

RESUMEN

Autophagy is an evolutionarily conserved process that degrades cytoplasmic components, thus contributing to cell survival and tissue homeostasis. Recent studies have demonstrated that autophagy maintains stem cells in relatively undifferentiated states (stemness) and also contributes to differentiation processes. Autophagy likewise plays a crucial role in somatic cell reprogramming, a finely regulated process that resets differentiated cells to a pluripotent state and that requires comprehensive alterations in transcriptional activities and epigenetic signatures. Autophagy assists in manifesting the functional consequences that arise from these alterations by modifying cellular protein expression profiles. The role of autophagy appears to be particularly relevant for early phases of cell reprogramming during the generation of induced pluripotent stems cells (iPSCs). In this review, we provide an overview of the core molecular machinery that constitutes the autophagic degradation system, describe the roles of autophagy in maintenance, self-renewal, and differentiation of stem cells, and discuss the autophagic process and its regulation during cell reprogramming.


Asunto(s)
Autofagia , Reprogramación Celular , Células Madre/citología , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre/metabolismo
17.
J Biol Chem ; 289(25): 17647-57, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24817116

RESUMEN

Natural killer (NK) cell activation is well orchestrated by a wide array of NK cell receptor repertoire. T-cell immunoglobulin and ITIM domain (TIGIT) receptor was recently defined as an inhibitory receptor that is expressed on NK cells and T cells. TIGIT receptor/poliovirus receptor (PVR) ligand engagement signaling inhibits cytotoxicity mediated by NK and CD8(+) T cells. However, it is unclear how TIGIT/PVR signaling regulates cytokine secretion in NK cells. Here we show that TIGIT/PVR engagement suppresses interferon-γ (IFN-γ) production of NK cells. TIGIT transgenic NK cells generate less IFN-γ undergoing TIGIT/PVR ligation. Moreover, TIGIT knock-out NK cells produce much more IFN-γ. TIGIT/PVR ligation signaling mediates suppression of IFN-γ production via the NF-κB pathway. We identified a novel adaptor ß-arrestin 2 that associates with phosphorylated TIGIT for further recruitment of SHIP1 (SH2-containing inositol phosphatase 1) through the ITT-like motif. Importantly, SHIP1, but not other phosphatases, impairs the TNF receptor-associated factor 6 (TRAF6) autoubiquitination to abolish NF-κB activation, leading to suppression of IFN-γ production in NK cells.


Asunto(s)
Arrestinas/metabolismo , Interferón gamma/biosíntesis , Células Asesinas Naturales/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Virales/metabolismo , Transducción de Señal/fisiología , Animales , Arrestinas/genética , Arrestinas/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Humanos , Recubrimiento Inmunológico/fisiología , Inositol Polifosfato 5-Fosfatasas , Interferón gamma/genética , Interferón gamma/inmunología , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Ratones , Ratones Noqueados , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/inmunología , Monoéster Fosfórico Hidrolasas/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores Virales/genética , Receptores Virales/inmunología , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Arrestina beta 2 , beta-Arrestinas
18.
Stem Cells ; 32(3): 623-35, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24167073

RESUMEN

Self-renewal and differentiation are the hallmarks of embryonic stem cells (ESCs). However, it is largely unknown about how the pluripotency is regulated. Here we demonstrate that Pcid2 is required for the maintenance of self-renewal both in mouse and human ESCs. Pcid2 plays a critical role in suppression of ESC differentiation. Pcid2 deficiency causes early embryonic lethality before the blastocyst stage. Pcid2 associates with EID1 and is present in the CBP/p300-EID1 complex in the ESCs. We show that MDM2 is an E3 ligase for K48-linked EID1 ubiquitination for its degradation. For the maintenance of self-renewal, Pcid2 binds to EID1 to impede the association with MDM2. Then EID1 is not degraded to sustain its stability to block the HAT activity of CBP/p300, leading to suppression of the developmental gene expression. Collectively, Pcid2 is present in the CBP/p300-EID1 complex to control the switch balance of mouse and human ESCs through modulation of EID1 degradation.


Asunto(s)
Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/metabolismo , Animales , Unión Competitiva , Proteínas de Ciclo Celular , Diferenciación Celular , Proliferación Celular , Eliminación de Gen , Humanos , Lisina/metabolismo , Ratones , Unión Proteica , Estabilidad Proteica , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Factores de Transcripción p300-CBP/metabolismo
19.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 4): 1094-103, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24699653

RESUMEN

VgrG proteins form the spike of the type VI secretion system (T6SS) syringe-like complex. VgrG3 of Vibrio cholerae degrades the peptidoglycan cell wall of rival bacteria via its C-terminal region (VgrG3C) through its muramidase activity. VgrG3C consists of a peptidoglycan-binding domain (VgrG3C(PGB)) and a putative catalytic domain (VgrG3C(CD)), and its activity can be inhibited by its immunity protein partner TsiV3. Here, the crystal structure of V. cholerae VgrG3C(CD) in complex with TsiV3 is presented at 2.3 Šresolution. VgrG3C(CD) adopts a chitosanase fold. A dimer of TsiV3 is bound in the deep active-site groove of VgrG3C(CD), occluding substrate binding and distorting the conformation of the catalytic dyad. Gln91 and Arg92 of TsiV3 are located in the centre of the interface and are important for recognition of VgrG3C. Mutation of these residues destabilized the complex and abolished the inhibitory activity of TsiV3 against VgrG3C toxicity in cells. Disruption of TsiV3 dimerization also weakened the complex and impaired the inhibitory activity. These structural, biochemical and functional data define the molecular mechanism underlying the self-protection of V. cholerae and expand the understanding of the role of T6SS in bacterial competition.


Asunto(s)
Proteínas Bacterianas/química , Sistemas de Secreción Bacterianos , Vibrio cholerae/química , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
20.
Nat Metab ; 6(5): 947-962, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38769396

RESUMEN

Polycystic ovary syndrome (PCOS), an endocrine disorder afflicting 6-20% of women of reproductive age globally, has been linked to alterations in the gut microbiome. We previously showed that in PCOS, elevation of Bacteroides vulgatus in the gut microbiome was associated with altered bile acid metabolism. Here we show that B. vulgatus also induces a PCOS-like phenotype in female mice via an alternate mechanism independent of bile acids. We find that B. vulgatus contributes to PCOS-like symptoms through its metabolite agmatine, which is derived from arginine by arginine decarboxylase. Mechanistically, agmatine activates the farnesoid X receptor (FXR) pathway to subsequently inhibit glucagon-like peptide-1 (GLP-1) secretion by L cells, which leads to insulin resistance and ovarian dysfunction. Critically, the GLP-1 receptor agonist liraglutide and the arginine decarboxylase inhibitor difluoromethylarginine ameliorate ovarian dysfunction in a PCOS-like mouse model. These findings reveal that agmatine-FXR-GLP-1 signalling contributes to ovarian dysfunction, presenting a potential therapeutic target for PCOS management.


Asunto(s)
Agmatina , Microbioma Gastrointestinal , Síndrome del Ovario Poliquístico , Receptores Citoplasmáticos y Nucleares , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/metabolismo , Animales , Femenino , Ratones , Agmatina/farmacología , Agmatina/metabolismo , Agmatina/uso terapéutico , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Péptido 1 Similar al Glucagón/metabolismo , Transducción de Señal/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a la Insulina , Bacteroides/efectos de los fármacos , Humanos , Carboxiliasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA