Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.514
Filtrar
1.
Nature ; 613(7942): 187-194, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36544021

RESUMEN

R-loops are RNA-DNA-hybrid-containing nucleic acids with important cellular roles. Deregulation of R-loop dynamics can lead to DNA damage and genome instability1, which has been linked to the action of endonucleases such as XPG2-4. However, the mechanisms and cellular consequences of such processing have remained unclear. Here we identify a new population of RNA-DNA hybrids in the cytoplasm that are R-loop-processing products. When nuclear R-loops were perturbed by depleting the RNA-DNA helicase senataxin (SETX) or the breast cancer gene BRCA1 (refs. 5-7), we observed XPG- and XPF-dependent cytoplasmic hybrid formation. We identify their source as a subset of stable, overlapping nuclear hybrids with a specific nucleotide signature. Cytoplasmic hybrids bind to the pattern recognition receptors cGAS and TLR3 (ref. 8), activating IRF3 and inducing apoptosis. Excised hybrids and an R-loop-induced innate immune response were also observed in SETX-mutated cells from patients with ataxia oculomotor apraxia type 2 (ref. 9) and in BRCA1-mutated cancer cells10. These findings establish RNA-DNA hybrids as immunogenic species that aberrantly accumulate in the cytoplasm after R-loop processing, linking R-loop accumulation to cell death through the innate immune response. Aberrant R-loop processing and subsequent innate immune activation may contribute to many diseases, such as neurodegeneration and cancer.


Asunto(s)
Citoplasma , ADN , Reconocimiento de Inmunidad Innata , Ácidos Nucleicos Heterodúplex , Estructuras R-Loop , ARN , Humanos , Apoptosis , Citoplasma/inmunología , Citoplasma/metabolismo , ADN/química , ADN/inmunología , ADN Helicasas/genética , ADN Helicasas/metabolismo , Genes BRCA1 , Enzimas Multifuncionales/genética , Enzimas Multifuncionales/metabolismo , Mutación , Neoplasias , Ácidos Nucleicos Heterodúplex/química , Ácidos Nucleicos Heterodúplex/inmunología , Estructuras R-Loop/inmunología , ARN/química , ARN/inmunología , ARN Helicasas/genética , ARN Helicasas/metabolismo , Ataxias Espinocerebelosas/genética
2.
J Clin Invest ; 132(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35040435

RESUMEN

Inborn errors of nucleic acid metabolism often cause aberrant activation of nucleic acid sensing pathways, leading to autoimmune or autoinflammatory diseases. The SKIV2L RNA exosome is cytoplasmic RNA degradation machinery that was thought to be essential for preventing the self-RNA-mediated interferon (IFN) response. Here, we demonstrate the physiological function of SKIV2L in mammals. We found that Skiv2l deficiency in mice disrupted epidermal and T cell homeostasis in a cell-intrinsic manner independently of IFN. Skiv2l-deficient mice developed skin inflammation and hair abnormality, which were also observed in a SKIV2L-deficient patient. Epidermis-specific deletion of Skiv2l caused hyperproliferation of keratinocytes and disrupted epidermal stratification, leading to impaired skin barrier with no appreciable IFN activation. Moreover, Skiv2l-deficient T cells were chronically hyperactivated and these T cells attacked lesional skin as well as hair follicles. Mechanistically, SKIV2L loss activated the mTORC1 pathway in both keratinocytes and T cells. Both systemic and topical rapamycin treatment of Skiv2l-deficient mice ameliorated epidermal hyperplasia and skin inflammation. Together, we demonstrate that mTORC1, a classical nutrient sensor, also senses cytoplasmic RNA quality control failure and drives autoinflammatory disease. We also propose SKIV2L-associated trichohepatoenteric syndrome (THES) as a new mTORopathy for which sirolimus may be a promising therapy.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Citoplasma/inmunología , Diarrea Infantil/inmunología , Retardo del Crecimiento Fetal/inmunología , Enfermedades del Cabello/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Estabilidad del ARN/inmunología , ARN/inmunología , Animales , Enfermedades Autoinmunes/genética , Citoplasma/genética , ADN Helicasas/deficiencia , ADN Helicasas/inmunología , Diarrea Infantil/genética , Facies , Retardo del Crecimiento Fetal/genética , Enfermedades del Cabello/genética , Inflamación/genética , Inflamación/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Noqueados , ARN/genética , Estabilidad del ARN/genética
3.
Cell Death Differ ; 29(6): 1267-1282, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34916628

RESUMEN

Cytoplasmic recognition of microbial lipopolysaccharides (LPS) in human cells is elicited by the caspase-4 and caspase-5 noncanonical inflammasomes, which induce a form of inflammatory cell death termed pyroptosis. Here we show that LPS-mediated activation of caspase-4 also induces a stress response promoting cellular senescence, which is dependent on the caspase-4 substrate gasdermin-D and the tumor suppressor p53. Furthermore, we found that the caspase-4 noncanonical inflammasome is induced and assembled in response to oncogenic RAS signaling during oncogene-induced senescence (OIS). Moreover, targeting caspase-4 expression in OIS showed its critical role in the senescence-associated secretory phenotype and the cell cycle arrest induced in cellular senescence. Finally, we observed that caspase-4 induction occurs in vivo in mouse models of tumor suppression and ageing. Altogether, we are showing that cellular senescence is induced by cytoplasmic LPS recognition by the noncanonical inflammasome and that this pathway is conserved in the cellular response to oncogenic stress.


Asunto(s)
Caspasas Iniciadoras , Inflamasomas , Animales , Caspasas Iniciadoras/inmunología , Senescencia Celular/inmunología , Citoplasma/inmunología , Humanos , Inmunidad Innata , Inflamasomas/inmunología , Lipopolisacáridos/farmacología , Ratones
4.
Front Immunol ; 12: 771277, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868031

RESUMEN

Histone H2A is a nuclear molecule tightly associated in the form of the nucleosome. Our previous studies have demonstrated the antibacterial property of piscine H2A variants against gram-negative bacteria Edwardsiella piscicida and Gram-positive bacteria Streptococcus agalactiae. In this study, we show the function and mechanism of piscine H2A in the negative regulation of RLR signaling pathway and host innate immune response against spring viremia of carp virus (SVCV) infection. SVCV infection significantly inhibits the expression of histone H2A during an early stage of infection, but induces the expression of histone H2A during the late stage of infection such as at 48 and 72 hpi. Under normal physiological conditions, histone H2A is nuclear-localized. However, SVCV infection promotes the migration of histone H2A from the nucleus to the cytoplasm. The in vivo studies revealed that histone H2A overexpression led to the increased expression of SVCV gene and decreased survival rate. The overexpression of histone H2A also significantly impaired the expression levels of those genes involved in RLR antiviral signaling pathway. Furthermore, histone H2A targeted TBK1 and IRF3 to promote their protein degradation via the lysosomal pathway and impair the formation of TBK1-IRF3 functional complex. Importantly, histone H2A completely abolished TBK1-mediated antiviral activity and enormously impaired the protein expression of IRF3, especially nuclear IRF3. Further analysis demonstrated that the inhibition of histone H2A nuclear/cytoplasmic trafficking could relieve the protein degradation of TBK1 and IRF3, and blocked the negative regulation of histone H2A on the SVCV infection. Collectively, our results suggest that histone H2A nuclear/cytoplasmic trafficking is essential for negative regulation of RLR signaling pathway and antiviral immune response in response to SVCV infection.


Asunto(s)
Histonas/inmunología , Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/inmunología , Lisosomas/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Rhabdoviridae/inmunología , Proteínas de Pez Cebra/inmunología , Pez Cebra/inmunología , Animales , Línea Celular , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Citoplasma/inmunología , Citoplasma/metabolismo , Regulación de la Expresión Génica/inmunología , Histonas/genética , Histonas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Larva/inmunología , Larva/metabolismo , Larva/virología , Lisosomas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas/inmunología , Proteolisis , Rhabdoviridae/fisiología , Pez Cebra/metabolismo , Pez Cebra/virología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
PLoS Pathog ; 17(11): e1010070, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34788350

RESUMEN

Nuclear scaffold attachment factor A (SAFA) is a novel RNA sensor involved in sensing viral RNA in the nucleus and mediating antiviral immunity. Severe fever with thrombocytopenia syndrome virus (SFTSV) is a bunyavirus that causes SFTS with a high fatality rate of up to 30%. It remains elusive whether and how cytoplasmic SFTSV can be sensed by the RNA sensor SAFA. Here, we demonstrated that SAFA was able to detect SFTSV infection and mediate antiviral interferon and inflammatory responses. Transcription and expression levels of SAFA were strikingly upregulated under SFTSV infection. SAFA was retained in the cytoplasm by interaction with SFTSV nucleocapsid protein (NP). Importantly, SFTSV genomic RNA was recognized by cytoplasmic SAFA, which recruited and promoted activation of the STING-TBK1 signaling axis against SFTSV infection. Of note, the nuclear localization signal (NLS) domain of SAFA was important for interaction with SFTSV NP and recognition of SFTSV RNA in the cytoplasm. In conclusion, our study reveals a novel antiviral mechanism in which SAFA functions as a novel cytoplasmic RNA sensor that directly recognizes RNA virus SFTSV and mediates an antiviral response.


Asunto(s)
Antivirales/metabolismo , Infecciones por Bunyaviridae/inmunología , Citoplasma/inmunología , Inmunidad Innata/inmunología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Phlebovirus/inmunología , Infecciones por Bunyaviridae/metabolismo , Infecciones por Bunyaviridae/virología , Citoplasma/virología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Proteínas Asociadas a Matriz Nuclear/genética
6.
Signal Transduct Target Ther ; 6(1): 382, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732709

RESUMEN

The global coronavirus disease 2019 (COVID-19) pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a positive-sense RNA virus. How the host immune system senses and responds to SARS-CoV-2 infection remain largely unresolved. Here, we report that SARS-CoV-2 infection activates the innate immune response through the cytosolic DNA sensing cGAS-STING pathway. SARS-CoV-2 infection induces the cellular level of 2'3'-cGAMP associated with STING activation. cGAS recognizes chromatin DNA shuttled from the nucleus as a result of cell-to-cell fusion upon SARS-CoV-2 infection. We further demonstrate that the expression of spike protein from SARS-CoV-2 and ACE2 from host cells is sufficient to trigger cytoplasmic chromatin upon cell fusion. Furthermore, cytoplasmic chromatin-cGAS-STING pathway, but not MAVS-mediated viral RNA sensing pathway, contributes to interferon and pro-inflammatory gene expression upon cell fusion. Finally, we show that cGAS is required for host antiviral responses against SARS-CoV-2, and a STING-activating compound potently inhibits viral replication. Together, our study reported a previously unappreciated mechanism by which the host innate immune system responds to SARS-CoV-2 infection, mediated by cytoplasmic chromatin from the infected cells. Targeting the cytoplasmic chromatin-cGAS-STING pathway may offer novel therapeutic opportunities in treating COVID-19. In addition, these findings extend our knowledge in host defense against viral infection by showing that host cells' self-nucleic acids can be employed as a "danger signal" to alarm the immune system.


Asunto(s)
COVID-19/inmunología , Cromatina/inmunología , Citoplasma/inmunología , Inmunidad Innata , Nucleotidiltransferasas/inmunología , SARS-CoV-2/inmunología , Animales , COVID-19/genética , Cromatina/genética , Citoplasma/genética , Modelos Animales de Enfermedad , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Transgénicos , Nucleotidiltransferasas/genética , SARS-CoV-2/genética
7.
J Neurosci ; 41(45): 9286-9307, 2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34593604

RESUMEN

Adult-onset neurodegenerative diseases are often accompanied by evidence of a chronic inflammation that includes activation of microglial cells and altered levels of brain cytokines. Aspects of this response are likely secondary reactions to neurodegeneration, but for many illnesses the inflammation may itself be an early and even causative disease event. In such cases, the inflammation is referred to as "sterile" as it occurs in the absence of an actual bacterial or viral pathogen. A potent trigger of sterile inflammation in CNS microglia has been shown to be the presence of DNA in the cytoplasm (cytoDNA) induced either by direct DNA damage or by inhibited DNA repair. We have shown that cytoDNA comes from the cell nucleus as a result of insufficient DNA damage repair. Using wild-type and Atm-/- mouse microglia, we extend these observations here by showing that its genomic origins are not random, but rather are heavily biased toward transcriptionally inactive, intergenic regions, in particular repetitive elements and AT-rich sequences. Once released from the genome, in both males and females, we show that cytoDNA is actively exported to the cytoplasm by a CRM1-dependent mechanism. In the cytoplasm, it is degraded either by a cytosolic exonuclease, Trex1, or an autophagy pathway that ends with degradation in the lysosome. Blocking the accumulation of cytoDNA prevents the emergence of the sterile inflammation reaction. These findings offer new insights into the emergence of sterile inflammation and offer novel approaches that may be of use in combatting a wide range of neurodegenerative conditions.SIGNIFICANCE STATEMENT Sterile inflammation describes a state where the defenses of the immune system are activated in the absence of a true pathogen. A potent trigger of this unorthodox response is the presence of DNA in the cytoplasm, which immune cells interpret as an invading virus or pathogen. We show that when DNA damage increases, fragments of the cell's own genome are actively exported to the cytoplasm where they are normally degraded. If this degradation is incomplete an immune reaction is triggered. Both age and stress increase DNA damage, and as age-related neurodegenerative diseases are frequently accompanied by a chronic low-level inflammation, strategies that reduce the induction of cytoplasmic DNA or speed its clearance become attractive therapeutic targets.


Asunto(s)
Citoplasma/inmunología , Daño del ADN/inmunología , ADN/inmunología , Inflamación/inmunología , Secuencias Repetitivas de Ácidos Nucleicos/inmunología , Animales , Citoplasma/metabolismo , ADN/metabolismo , Reparación del ADN , Femenino , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Microglía/metabolismo
8.
Pathol Res Pract ; 224: 153527, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34167064

RESUMEN

ß-catenin (ßcat) is an important downstream effector in the Wnt signaling pathway and plays important roles in the development and progression of many cancers including melanoma. ßcat expression is regulated by GSK-3ß-mediated phosphorylation at positions 33, 37 and 41. In normal cells, phosphorylation at these sites triggers proteasomal degradation, which prevents accumulation of free cytoplasmic ßcat. In cancer cells, stabilized ß-catenin translocates into the nucleus, where it associates with TCF/Lef proteins to activate transcription of genes that promote tumorigenesis and metastasis, including PD-L1. It has been suggested that nuclear phospho-ßcat (pßcat) staining may be diagnostically useful in differentiating primary from metastatic melanoma. Also, a pßcat peptide (residues 30-39, with only S33 phosphorylated) is naturally presented by melanoma cells as a T-cell target. We evaluated expression of pS33-ßcat in primary and metastatic melanomas by immunohistochemistry and found its expression varied widely but was most commonly cytoplasmic. Nuclear staining was identified in only 18% of metastatic melanomas. Staining with antibodies to pS33-ßcat and pS33/37/T41-ßcat was most intense in mitotic melanoma cells; however, pS33-ßcat intensity was not significantly associated with AJCC stage, tumor location, BRAF mutation status, or immune infiltrates. Yet, PD-L1 and PD-L2 expression by tumor cells were significantly higher in tumors with high pS33-ßcat expression. The low rate of nuclear pS33-ßcat expression suggests that pS33-ßcat may have limited utility for identifying metastatic melanomas. However, high expression in dividing cells and strong associations with PD-L1 and PD-L2 expression may inform future personalized therapies for tumors with high pS33-ßcat expression.


Asunto(s)
Antígeno B7-H1/metabolismo , Melanoma/metabolismo , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Neoplasias Cutáneas/metabolismo , beta Catenina/metabolismo , Núcleo Celular/metabolismo , Citoplasma/inmunología , Citoplasma/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunohistoquímica/métodos , Melanoma/genética , Mutación/genética , Neoplasias Cutáneas/genética , Melanoma Cutáneo Maligno
9.
Front Immunol ; 12: 657393, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054821

RESUMEN

Human neutrophils express two unique antibody receptors for IgG, the FcγRIIa and the FcγRIIIb. FcγRIIa contains an immunoreceptor tyrosine-based activation motif (ITAM) sequence within its cytoplasmic tail, which is important for initiating signaling. In contrast, FcγRIIIb is a glycosylphosphatidylinositol (GPI)-linked receptor with no cytoplasmic tail. Although, the initial signaling mechanism for FcγRIIIb remains unknown, it is clear that both receptors are capable of initiating distinct neutrophil cellular functions. For example, FcγRIIa is known to induce an increase in L-selectin expression and efficient phagocytosis, while FcγRIIIb does not promote these responses. In contrast, FcγRIIIb has been reported to induce actin polymerization, activation of ß1 integrins, and formation of neutrophils extracellular traps (NET) much more efficiently than FcγRIIa. Another function where these receptors seem to act differently is the increase of cytoplasmic calcium concentration. It has been known for a long time that FcγRIIa induces production of inositol triphosphate (IP3) to release calcium from intracellular stores, while FcγRIIIb does not use this phospholipid. Thus, the mechanism for FcγRIIIb-mediated calcium rise remains unknown. Transient Receptor Potential Melastatin 2 (TRPM2) is a calcium permeable channel expressed in many cell types including vascular smooth cells, endothelial cells and leukocytes. TRPM2 can be activated by protein kinase C (PKC) and by oxidative stress. Because we previously found that FcγRIIIb stimulation leading to NET formation involves PKC activation and reactive oxygen species (ROS) production, in this report we explored whether TRPM2 is activated via FcγRIIIb and mediates calcium rise in human neutrophils. Calcium rise was monitored after Fcγ receptors were stimulated by specific monoclonal antibodies in Fura-2-loaded neutrophils. The bacterial peptide fMLF and FcγRIIa induced a calcium rise coming initially from internal pools. In contrast, FcγRIIIb caused a calcium rise by inducing calcium entry from the extracellular medium. In addition, in the presence of 2-aminoethoxydiphenyl borate (2-APB) or of clotrimazole, two inhibitors of TRPM2, FcγRIIIb-induced calcium rise was blocked. fMLF- or FcγRIIa-induced calcium rise was not affected by these inhibitors. These data suggest for the first time that FcγRIIIb aggregation activates TRPM2, to induce an increase in cytoplasmic calcium concentration through calcium internalization in human neutrophils.


Asunto(s)
Calcio/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de IgG/metabolismo , Canales Catiónicos TRPM/metabolismo , Señalización del Calcio , Citoplasma/inmunología , Citoplasma/metabolismo , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Trampas Extracelulares/metabolismo , Técnica del Anticuerpo Fluorescente , Proteínas Ligadas a GPI/metabolismo , Humanos , Modelos Biológicos , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Fagocitosis/inmunología , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
10.
Anticancer Res ; 41(5): 2543-2552, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33952482

RESUMEN

BACKGROUND/AIM: Maspin is a tumor-suppressor protein expressed in >90% of pancreatic ductal adenocarcinoma (PDAC) cases. We aimed to assess the prognostic value of subcellular localization of maspin. PATIENTS AND METHODS: Ninety-two resected PDAC specimens were immunohistochemically analyzed. Cytoplasmic-only expression observed in >10% of the tumor was defined as maspin-positive. RESULTS: The maspin-positive status (21.7%) was inversely correlated with well-differentiated histological type and indicated a shorter recurrence-free survival (RFS) and overall survival (OS). Cox's multivariate analysis showed that maspin-positive status was an independent factor for shorter RFS and OS. Maspin was localized to cytoplasm in AsPC-1 cells, but to both nucleus and cytoplasm in BxPC-3 cells. In AsPC-1 cells, cell invasion was significantly reduced in response to maspin suppression via transfection with siRNA targeting maspin, whereas no reduction was observed in BxPC-3 cells. CONCLUSION: Cytoplasmic-only expression of maspin could be an independent unfavorable prognostic indicator for patients with PDAC.


Asunto(s)
Adenocarcinoma/genética , Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/genética , Serpinas/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Anciano , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/genética , Citoplasma/efectos de los fármacos , Citoplasma/inmunología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/inmunología , Serpinas/inmunología
11.
J Exp Med ; 218(7)2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-33970189

RESUMEN

Because of their common signaling molecules, the main T cell receptor (TCR) signaling cascades in CD4+ and CD8+ T cells are considered qualitatively identical. Herein, we show that TCR signaling in CD8+ T cells is qualitatively different from that in CD4+ T cells, since CD8α ignites another cardinal signaling cascade involving phospholipase C ß4 (PLCß4). TCR-mediated responses were severely impaired in PLCß4-deficient CD8+ T cells, whereas those in CD4+ T cells were intact. PLCß4-deficient CD8+ T cells showed perturbed activation of peripheral TCR signaling pathways downstream of IP3 generation. Binding of PLCß4 to the cytoplasmic tail of CD8α was important for CD8+ T cell activation. Furthermore, GNAQ interacted with PLCß4, mediated double phosphorylation on threonine 886 and serine 890 positions of PLCß4, and activated CD8+ T cells in a PLCß4-dependent fashion. PLCß4-deficient mice exhibited defective antiparasitic host defense and antitumor immune responses. Altogether, PLCß4 differentiates TCR signaling in CD4+ and CD8+ T cells and selectively promotes CD8+ T cell-dependent adaptive immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Fosfolipasa C beta/inmunología , Transducción de Señal/inmunología , Animales , Línea Celular , Citoplasma/inmunología , Células HEK293 , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Fosforilación/inmunología , Receptores de Antígenos de Linfocitos T/inmunología
12.
Immunity ; 54(4): 632-647.e9, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33667382

RESUMEN

Aging is associated with DNA accumulation and increased homeostatic proliferation of circulating T cells. Although these attributes are associated with aging-related autoimmunity, their direct contributions remain unclear. Conventionally, KU complex, the regulatory subunit of DNA-dependent protein kinase (DNA-PK), together with the catalytic subunit of DNA-PK (DNA-PKcs), mediates DNA damage repair in the nucleus. Here, we found KU complex abundantly expressed in the cytoplasm, where it recognized accumulated cytoplasmic DNA in aged human and mouse CD4+ T cells. This process enhanced T cell activation and pathology of experimental autoimmune encephalomyelitis (EAE) in aged mice. Mechanistically, KU-mediated DNA sensing facilitated DNA-PKcs recruitment and phosphorylation of the kinase ZAK. This activated AKT and mTOR pathways, promoting CD4+ T cell proliferation and activation. We developed a specific ZAK inhibitor, which dampened EAE pathology in aged mice. Overall, these findings demonstrate a KU-mediated cytoplasmic DNA-sensing pathway in CD4+ T cells that potentiates aging-related autoimmunity.


Asunto(s)
Envejecimiento/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Citoplasma/inmunología , Proteína Quinasa Activada por ADN/inmunología , ADN/inmunología , Inflamación/inmunología , Animales , Línea Celular , Línea Celular Tumoral , Núcleo Celular/inmunología , Proliferación Celular/fisiología , Reparación del ADN/inmunología , Células HEK293 , Humanos , Células Jurkat , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Células U937
13.
Int J Biol Macromol ; 172: 550-559, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33465362

RESUMEN

The crude polysaccharide was extracted from A. asphodeloides rhizomes and further purified to produce two fractions F1 (50.0%) and F2 (19.6%). The chemical constitutions of the polysaccharides were neutral sugars (51.4%-89.7%), uronic acids (1.0%-30.2%) and sulfate esters (3.4%-8.1%), with various ratios of monosaccharides including rhamnose (1.4%-6.1%), arabinose (7.1%-21.2%), xylose (0.2%-4.8%), mannose (39.9%-79.0%), glucose (6.0%-11.1%) and galactose (2.6%-22.0%). The molecular properties of the polysaccharides were investigated by the HPSEC-UV-MALLS-RI system, revealing the Mw 130.0 × 103-576.5 × 103 g/moL, Rg 87.6-382.6 nm and SVg 0.3-54.3 cm3/g. The polysaccharides stimulated RAW264.7 cells to produce considerable amounts of NO and up-regulate the expression of TNF-α, IL-1 and COX-2 genes. Polysaccharides exhibited the growth inhibitory effects on cancer cells lines of AGS, MKN-28 and MKN-45, in which F2 fraction exhibited prominent bioactivities. The AGS cells treated with F2 experienced condensed cytoplasm, shrinkage of nucleus and chromatin marginalization with the highest number of cells at early-stage apoptosis reaching 54.6%. The inhibitory effect of F2 polysaccharide on AGS cells was through MAPKs and STAT3 signaling pathways. The backbone of the F2 was mainly linked by (1 â†’ 4)-linked mannopyranosyl and (1 â†’ 3)-linked galactopyranosyl. Taken together, the polysaccharide from A. asphodeloides rhizomes could be utilized as medicinal, pharmacological and functional food ingredients.


Asunto(s)
Anemarrhena/química , Regulación de la Expresión Génica/efectos de los fármacos , Factores Inmunológicos/farmacología , Polisacáridos/farmacología , Rizoma/química , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Secuencia de Carbohidratos , Línea Celular Tumoral , Cromatina/química , Cromatina/efectos de los fármacos , Cromatina/inmunología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/inmunología , Citoplasma/efectos de los fármacos , Citoplasma/inmunología , Citoplasma/patología , Factores Inmunológicos/química , Factores Inmunológicos/aislamiento & purificación , Interleucina-1/genética , Interleucina-1/inmunología , Ratones , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/inmunología , Monosacáridos/química , Monosacáridos/aislamiento & purificación , Óxido Nítrico/biosíntesis , Polisacáridos/química , Polisacáridos/aislamiento & purificación , Células RAW 264.7 , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Ácidos Urónicos/química , Ácidos Urónicos/aislamiento & purificación
14.
Mol Immunol ; 129: 21-31, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33260037

RESUMEN

The Atlantic cod immune system deviates from antigen presentation processes seen in other vertebrates in that it lacks the necessary genes for exogenous antigen presentation (i.e., MHC-II and li) and a key MHC-II interacting molecule necessary for T-helper cell function (i.e., CD4), while possessing an expanded repertoire of MHC-I genes that facilitate endogenous antigen presentation. These observations, combined with the identification of putative endosomal sorting signals in MHC-I cytoplasmic tails, have led to speculation that cod rely on cross-presentation of exogenous antigens to elicit cytotoxic T-lymphocyte responses against extracellular threats. In light of this suggestion, we investigated MHC-I transcriptional profiles and endosomal sorting signals in a closely related gadoid species, the haddock. Analysis of transcripts from one individual identified 13 unique MHC-I molecules, including two non-classical molecules as determined by the level of conservation at their peptide anchoring sites. This suggests that like the cod, the haddock has an expanded MHC-I repertoire. Analysis of haddock MHC-I cytoplasmic tail sequences revealed that the dileucine- and tyrosine-based endosomal signaling motifs, that are suggested to facilitate cross-presentation in cod, were absent. Closer examination of the cod signaling motifs, including their relative position in the cytoplasmic tail region, indicates that these motifs might be non-functional, further supporting the need for functional studies to assess cross-presentation. Finally, in silico analysis and in vitro N-type de-glycosylation experiments demonstrate that haddock and cod beta-2-microglobulin (ß2M) are glycosylated at the same NQT sequon. Interestingly, whole genome tBLASTn searches also revealed that putative ß2 M glycosylation sites appear frequently within the Gadiformes lineage, as the predictive NQT and other N-X-S/T sequons were located in ß2M orthologues from 19 of the 25 additional gadoid genomes analyzed. Though the exact significance of ß2M glycosylation has yet to be elucidated, phylogenetic comparisons predict that the same NQT glycosylation sequence occurs in 13 additional species comprising four different orders of Actinopterygii (Gymnotiformes, Esociformes, Beryciformes and Perciformes). This suggests either that this feature has arisen independently in multiple lineages or that it comes from a common ancestor and has been lost or modified in many species. Together, the analysis of gadoid MHC-I genes and ß2M molecules highlights the challenges in generalizing immune system paradigms across the most diverse vertebrate lineage (i.e., fish) and between fish and more well-studied mammals.


Asunto(s)
Presentación de Antígeno/genética , Antígenos/genética , Reactividad Cruzada/genética , Gadus morhua/genética , Microglobulina beta-2/genética , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/inmunología , Antígenos/inmunología , Reactividad Cruzada/inmunología , Citoplasma/genética , Citoplasma/inmunología , Endosomas/genética , Endosomas/inmunología , Gadus morhua/inmunología , Genoma/genética , Genoma/inmunología , Glicosilación , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Alineación de Secuencia , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Transcripción Genética/genética , Transcripción Genética/inmunología , Microglobulina beta-2/inmunología
15.
Front Immunol ; 12: 780900, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095855

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in virtually all tissues; they have potent self-renewal capacity and differentiate into multiple cell types. For many reasons, these cells are a promising therapeutic alternative to treat patients with severe COVID-19 and pulmonary post-COVID sequelae. These cells are not only essential for tissue regeneration; they can also alter the pulmonary environment through the paracrine secretion of several mediators. They can control or promote inflammation, induce other stem cells differentiation, restrain the virus load, and much more. In this work, we performed single-cell RNA-seq data analysis of MSCs in bronchoalveolar lavage samples from control individuals and COVID-19 patients with mild and severe clinical conditions. When we compared samples from mild cases with control individuals, most genes transcriptionally upregulated in COVID-19 were involved in cell proliferation. However, a new set of genes with distinct biological functions was upregulated when we compared severely affected with mild COVID-19 patients. In this analysis, the cells upregulated genes related to cell dispersion/migration and induced the γ-activated sequence (GAS) genes, probably triggered by IFNGR1 and IFNGR2. Then, IRF-1 was upregulated, one of the GAS target genes, leading to the interferon-stimulated response (ISR) and the overexpression of many signature target genes. The MSCs also upregulated genes involved in the mesenchymal-epithelial transition, virus control, cell chemotaxis, and used the cytoplasmic RNA danger sensors RIG-1, MDA5, and PKR. In a non-comparative analysis, we observed that MSCs from severe cases do not express many NF-κB upstream receptors, such as Toll-like (TLRs) TLR-3, -7, and -8; tumor necrosis factor (TNFR1 or TNFR2), RANK, CD40, and IL-1R1. Indeed, many NF-κB inhibitors were upregulated, including PPP2CB, OPTN, NFKBIA, and FHL2, suggesting that MSCs do not play a role in the "cytokine storm" observed. Therefore, lung MSCs in COVID-19 sense immune danger and act protectively in concert with the pulmonary environment, confirming their therapeutic potential in cell-based therapy for COVID-19. The transcription of MSCs senescence markers is discussed.


Asunto(s)
COVID-19/inmunología , Proliferación Celular/fisiología , Inflamación/inmunología , Pulmón/inmunología , Células Madre Mesenquimatosas/inmunología , Regeneración/inmunología , Adulto , COVID-19/metabolismo , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Citoplasma/inmunología , Transición Epitelial-Mesenquimal/inmunología , Humanos , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , SARS-CoV-2/inmunología , Regulación hacia Arriba/inmunología , Adulto Joven
16.
Front Immunol ; 12: 763044, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35087515

RESUMEN

Cytolytic T cell responses are predicted to be biased towards membrane proteins. The peptide-binding grooves of most alleles of histocompatibility complex class I (MHC-I) are relatively hydrophobic, therefore peptide fragments derived from human transmembrane helices (TMHs) are predicted to be presented more often as would be expected based on their abundance in the proteome. However, the physiological reason of why membrane proteins might be over-presented is unclear. In this study, we show that the predicted over-presentation of TMH-derived peptides is general, as it is predicted for bacteria and viruses and for both MHC-I and MHC-II, and confirmed by re-analysis of epitope databases. Moreover, we show that TMHs are evolutionarily more conserved, because single nucleotide polymorphisms (SNPs) are present relatively less frequently in TMH-coding chromosomal regions compared to regions coding for extracellular and cytoplasmic protein regions. Thus, our findings suggest that both cytolytic and helper T cells are more tuned to respond to membrane proteins, because these are evolutionary more conserved. We speculate that TMHs are less prone to mutations that enable pathogens to evade T cell responses.


Asunto(s)
Presentación de Antígeno/genética , Epítopos de Linfocito T/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase I/genética , Proteínas de la Membrana/genética , Estructura Secundaria de Proteína/genética , Alelos , Presentación de Antígeno/inmunología , Cromosomas/genética , Cromosomas/inmunología , Citoplasma/genética , Citoplasma/inmunología , Epítopos de Linfocito T/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Proteínas de la Membrana/inmunología , Péptidos/genética , Péptidos/inmunología , Polimorfismo de Nucleótido Simple/genética , Polimorfismo de Nucleótido Simple/inmunología , Linfocitos T Colaboradores-Inductores/inmunología
17.
Am J Physiol Cell Physiol ; 320(2): C195-C215, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264078

RESUMEN

Single-domain antibodies, derived from camelid heavy antibodies (nanobodies) or shark variable new antigen receptors, have attracted increasing attention in recent years due to their extremely versatile nature and the opportunities they offer for downstream modification. Discovered more than three decades ago, these 120-amino acid (∼15-kDa) antibody fragments are known to bind their target with high specificity and affinity. Key features of nanobodies that make them very attractive include their single-domain nature, small size, and affordable high-level expression in prokaryotes, and their cDNAs are routinely obtained in the process of their isolation. This facilitates and stimulates new experimental approaches. Hence, it allows researchers to formulate new answers to complex biomedical questions. Through elementary PCR-based technologies and chemical modification strategies, their primary structure can be altered almost at leisure while retaining their specificity and biological activity, transforming them into highly tailored tools that meet the increasing demands of current-day biomedical research. In this review, various aspects of camelid nanobodies are expounded, including intracellular delivery in recombinant format for manipulation of, i.e., cytoplasmic targets, their derivatization to improve nanobody orientation as a capturing device, approaches to reversibly bind their target, their potential as protein-silencing devices in cells, the development of strategies to transfer nanobodies through the blood-brain barrier and their application in CAR-T experimentation. We also discuss some of their disadvantages and conclude with future prospects.


Asunto(s)
Membrana Celular/metabolismo , Citoplasma/metabolismo , Inmunoterapia Adoptiva/métodos , Anticuerpos de Dominio Único/metabolismo , Animales , Anticuerpos/inmunología , Anticuerpos/metabolismo , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Membrana Celular/inmunología , Citoplasma/inmunología , Humanos , Inmunoterapia Adoptiva/tendencias , Espacio Intracelular/inmunología , Espacio Intracelular/metabolismo , Proteínas/fisiología , Anticuerpos de Dominio Único/inmunología
18.
Front Immunol ; 11: 543444, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329509

RESUMEN

Host antiviral factor interferon-induced transmembrane proteins (IFITMs) are a kind of small-molecule transmembrane proteins induced by interferon. Their broad-spectrum antiviral activity and unique ability to inhibit viral invasion have made them a hot molecule in antiviral research in recent years. Since the first demonstration of their natural ability to resist viral infection in 1996, IFITMs have been reported to limit a variety of viral infections, including some major pathogens that seriously endanger human health and social stability, such as influenza A, Ebol, severe acute respiratory syndrome, AIDS, and Zika viruses, etc. Studies show that IFITMs mainly exert antiviral activity during virus entry, specifically interfering with the fusion of the envelope and the endosome membrane or forming fusion micropores to block the virus from entering the cytoplasm. However, their specific mechanism is still unclear. This article mainly reviews the research progress in the structure, evolution, function, and mechanism of IFITMs, which may provide a theoretical basis for clarifying the molecular mechanism of interaction between the molecules and viruses and the research and development of new antiviral drugs based on IFITMs.


Asunto(s)
Citoplasma/inmunología , Inmunidad Innata , Proteínas de la Membrana/inmunología , Virosis/inmunología , Internalización del Virus , Virus/inmunología , Animales , Antivirales/uso terapéutico , Citoplasma/virología , Humanos , Virosis/tratamiento farmacológico
19.
Ann Clin Transl Neurol ; 7(11): 2231-2242, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33009713

RESUMEN

OBJECTIVE: Identify the subcellular location and potential binding partners of two cerebellar degeneration-related proteins, CDR2L and CDR2, associated with anti-Yo-mediated paraneoplastic cerebellar degeneration. METHODS: Cancer cells, rat Purkinje neuron cultures, and human cerebellar sections were exposed to cerebrospinal fluid and serum from patients with paraneoplastic cerebellar degeneration with Yo antibodies and with several antibodies against CDR2L and CDR2. We used mass spectrometry-based proteomics, super-resolution microscopy, proximity ligation assay, and co-immunoprecipitation to verify the antibodies and to identify potential binding partners. RESULTS: We confirmed the CDR2L specificity of Yo antibodies by mass spectrometry-based proteomics and found that CDR2L localized to the cytoplasm and CDR2 to the nucleus. CDR2L co-localized with the 40S ribosomal protein S6, while CDR2 co-localized with the nuclear speckle proteins SON, eukaryotic initiation factor 4A-III, and serine/arginine-rich splicing factor 2. INTERPRETATION: We showed that Yo antibodies specifically bind to CDR2L in Purkinje neurons of PCD patients where they potentially interfere with the function of the ribosomal machinery resulting in disrupted mRNA translation and/or protein synthesis. Our findings demonstrating that CDR2L interacts with ribosomal proteins and CDR2 with nuclear speckle proteins is an important step toward understanding PCD pathogenesis.


Asunto(s)
Autoanticuerpos , Autoantígenos , Núcleo Celular , Citoplasma , Proteínas del Tejido Nervioso , Degeneración Cerebelosa Paraneoplásica , Biosíntesis de Proteínas , Células de Purkinje , Animales , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Autoantígenos/inmunología , Autoantígenos/metabolismo , Línea Celular Tumoral , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Citoplasma/inmunología , Citoplasma/metabolismo , Humanos , Inmunoprecipitación , Espectrometría de Masas , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Degeneración Cerebelosa Paraneoplásica/inmunología , Degeneración Cerebelosa Paraneoplásica/metabolismo , Biosíntesis de Proteínas/fisiología , Proteómica , Células de Purkinje/inmunología , Células de Purkinje/metabolismo , Ratas
20.
Sci Rep ; 10(1): 15290, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943718

RESUMEN

Autophagy, a degradation system, works to maintain cellular homeostasis. However, as the impact of Hepatitis C virus (HCV) infection on hepatocyte autophagy and its effect on HCV replication remain unclear, we examined them. HCV infection suppressed late-stage autophagy and increased Rubicon. siRNA-mediated knockdown of Rubicon promoted autophagy in HCV-infected cells. In Huh-7 cells harbouring the HCV replicon, Rubicon knockdown downregulated the expression of type 1 interferon (IFN)-related genes and upregulated HCV replication. Rubicon overexpression or administration of bafilomycin A1 or chloroquine, an inhibitor of late-stage autophagy, suppressed autophagy and activated the type 1 IFN pathway. On the other hand, Atg7 knockout suppressed early-stage autophagy and did not activate the type 1 IFN pathway. In livers of humanized liver chimeric mice, HCV infection increased Rubicon and enhanced type 1 IFN signalling. Elimination of HCV in the mice reduced the increase in Rubicon due to HCV infection. The expression levels of Rubicon and IFN-stimulated genes in chronic hepatitis C patients were higher than those in non-B, non-C hepatitis patients. HCV infection increased Rubicon and suppressed hepatocyte autophagy, leading to activation of the intracellular immune response. Rubicon induction is involved in HCV replication via activation of the intracellular immune response.


Asunto(s)
Proteínas Relacionadas con la Autofagia/inmunología , Autofagia/inmunología , Hepacivirus/inmunología , Hepatitis C Crónica/inmunología , Inmunidad Innata/inmunología , Animales , Línea Celular Tumoral , Citoplasma/inmunología , Hepatocitos/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferón Tipo I/inmunología , Ratones , Replicón/inmunología , Transducción de Señal/inmunología , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...