Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 316
Filtrar
1.
Front Immunol ; 12: 748423, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34691060

RESUMEN

The Type I Interferon family of cytokines all act through the same cell surface receptor and induce phosphorylation of the same subset of response regulators of the STAT family. Despite their shared receptor, different Type I Interferons have different functions during immune response to infection. In particular, they differ in the potency of their induced anti-viral and anti-proliferative responses in target cells. It remains not fully understood how these functional differences can arise in a ligand-specific manner both at the level of STAT phosphorylation and the downstream function. We use a minimal computational model of Type I Interferon signaling, focusing on Interferon-α and Interferon-ß. We validate the model with quantitative experimental data to identify the key determinants of specificity and functional plasticity in Type I Interferon signaling. We investigate different mechanisms of signal discrimination, and how multiple system components such as binding affinity, receptor expression levels and their variability, receptor internalization, short-term negative feedback by SOCS1 protein, and differential receptor expression play together to ensure ligand specificity on the level of STAT phosphorylation. Based on these results, we propose phenomenological functional mappings from STAT activation to downstream anti-viral and anti-proliferative activity to investigate differential signal processing steps downstream of STAT phosphorylation. We find that the negative feedback by the protein USP18, which enhances differences in signaling between Interferons via ligand-dependent refractoriness, can give rise to functional plasticity in Interferon-α and Interferon-ß signaling, and explore other factors that control functional plasticity. Beyond Type I Interferon signaling, our results have a broad applicability to questions of signaling specificity and functional plasticity in signaling systems with multiple ligands acting through a bottleneck of a small number of shared receptors.


Asunto(s)
Interferón-alfa/fisiología , Interferón beta/fisiología , Modelos Inmunológicos , Receptor Cross-Talk/fisiología , Receptor de Interferón alfa y beta/fisiología , Transducción de Señal/fisiología , Animales , Simulación por Computador , Dimerización , Retroalimentación Fisiológica , Femenino , Humanos , Concentración 50 Inhibidora , Cinética , Ligandos , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Mapeo de Interacción de Proteínas , Factores de Transcripción STAT/metabolismo , Bazo/citología , Proteína 1 Supresora de la Señalización de Citocinas/fisiología , Linfocitos T/inmunología , Ubiquitina Tiolesterasa
2.
J Virol ; 95(20): e0023121, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34379499

RESUMEN

The NS1 protein of the influenza A virus plays a critical role in regulating several biological processes in cells, including the type I interferon (IFN) response. We previously profiled the cellular factors that interact with the NS1 protein of influenza A virus and found that the NS1 protein interacts with proteins involved in RNA splicing/processing, cell cycle regulation, and protein targeting processes, including 14-3-3ε. Since 14-3-3ε plays an important role in retinoic acid-inducible gene I (RIG-I) translocation to mitochondrial antiviral-signaling protein (MAVS) to activate type I IFN expression, the interaction of the NS1 and 14-3-3ε proteins may prevent the RIG-I-mediated IFN response. In this study, we confirmed that the 14-3-3ε protein interacts with the N-terminal domain of the NS1 protein and that the NS1 protein inhibits RIG-I-mediated IFN-ß promoter activation in 14-3-3ε-overexpressing cells. In addition, our results showed that knocking down 14-3-3ε can reduce IFN-ß expression elicited by influenza A virus and enhance viral replication. Furthermore, we found that threonine in the 49th amino acid position of the NS1 protein plays a role in the interaction with 14-3-3ε. Influenza A virus expressing C terminus-truncated NS1 with a T49A mutation dramatically increases IFN-ß mRNA in infected cells and causes slower replication than that of virus without the T-to-A mutation. Collectively, this study demonstrates that 14-3-3ε is involved in influenza A virus-initiated IFN-ß expression and that the interaction of the NS1 protein and 14-3-3ε may be one of the mechanisms for inhibiting type I IFN activation during influenza A virus infection. IMPORTANCE Influenza A virus is an important human pathogen causing severe respiratory disease. The virus has evolved several strategies to dysregulate the innate immune response and facilitate its replication. We demonstrate that the NS1 protein of influenza A virus interacts with the cellular chaperone protein 14-3-3ε, which plays a critical role in retinoic acid-inducible gene I (RIG-I) translocation that induces type I interferon (IFN) expression, and that NS1 protein prevents RIG-I translocation to the mitochondrial membrane. The interaction site for 14-3-3ε is the RNA-binding domain (RBD) of the NS1 protein. Therefore, this research elucidates a novel mechanism by which the NS1 RBD mediates IFN-ß suppression to facilitate influenza A viral replication. Additionally, the findings reveal the antiviral role of 14-3-3ε during influenza A virus infection.


Asunto(s)
Proteínas 14-3-3/inmunología , Gripe Humana/inmunología , Interferón beta/metabolismo , Proteínas 14-3-3/metabolismo , Línea Celular Tumoral , Proteína 58 DEAD Box/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/inmunología , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Interferón beta/fisiología , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional , ARN Viral/genética , Receptores Inmunológicos/metabolismo , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/genética
3.
Vet Res ; 51(1): 135, 2020 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-33176874

RESUMEN

Duck enteritis virus (DEV) is a member of the Alphaherpesvirinae subfamily. The characteristics of some DEV genes have been reported. However, information regarding the DEV UL47 gene is limited. In this study, we identified the DEV UL47 gene encoding a late structural protein located in the nucleus of infected cells. We further found that two domains of DEV pUL47, amino acids (aa) 40 to 50 and 768 to 777, could function as nuclear localization sequence (NLS) to guide the nuclear localization of pUL47 and nuclear translocation of heterologous proteins, including enhanced green fluorescent protein (EGFP) and beta-galactosidase (ß-Gal). Moreover, pUL47 significantly inhibited polyriboinosinic:polyribocytidylic acid [poly(I:C)]-induced interferon beta (IFN-ß) production and downregulated interferon-stimulated gene (ISG) expression, such as Mx and oligoadenylate synthetase-like (OASL), by interacting with signal transducer and activator of transcription-1 (STAT1).


Asunto(s)
Patos , Interferón beta/fisiología , Mardivirus/fisiología , Enfermedad de Marek/virología , Enfermedades de las Aves de Corral/virología , Factor de Transcripción STAT1/fisiología , Proteínas Estructurales Virales/genética , Animales , Núcleo Celular/virología , Transducción de Señal
4.
Med Hypotheses ; 144: 110012, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32590324

RESUMEN

The most serious health issue today is the rapid outbreak of Coronavirus Disease 2019 (COVID-19). More than 6,973,427 confirmed cases were diagnosed in nearly 213 countries and territories around the world and two international conveyances, causing globally over 400,000 deaths. Epidemiology, risk factors, and clinical characteristics of COVID-19 patients have been identified, but the factors influencing the immune system against COVID-19 have not been well established. Upon infection or cell damage, high amounts of adenosine triphosphate (ATP) are released from damaged cells, which serve as mediators of inflammation through purinergic cell surface receptor signaling. As a protective mechanism to prevent excessive damage to host tissue, adenosine counteracts ATP's effects by adenosine receptor stimulation to suppress the pro-inflammatory response. Adenosine is seen as a major obstacle to the efficacy of immune therapies, and the adenosinergic axis components are critical therapeutic targets for cancer and microbial infections. Pharmacologic inhibitors or antibodies specific to adenosinergic pathway components or adenosine receptors in microbial and tumor therapy have shown efficacy in pre-clinical studies and are entering the clinical arena. In this review, we provide a novel hypothesis explaining the potential for improving the efficiency of innate and adaptive immune systems by targeting adenosinergic pathway components and adenosine A2A receptor signaling for the treatment of COVID-19.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Pandemias , Receptor de Adenosina A2A/fisiología , 5'-Nucleotidasa/metabolismo , Inmunidad Adaptativa/efectos de los fármacos , Antagonistas del Receptor de Adenosina A2/farmacología , Adenosina Trifosfato/metabolismo , Apirasa/metabolismo , COVID-19/epidemiología , COVID-19/inmunología , COVID-19/metabolismo , Proteínas Ligadas a GPI/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Interferón beta/fisiología , Modelos Inmunológicos , Terapia Molecular Dirigida , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , SARS-CoV-2/inmunología , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
5.
J Neurosci ; 40(11): 2357-2370, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32029532

RESUMEN

DNA damage and type I interferons (IFNs) contribute to inflammatory responses after traumatic brain injury (TBI). TBI-induced activation of microglia and peripherally-derived inflammatory macrophages may lead to tissue damage and neurological deficits. Here, we investigated the role of IFN-ß in secondary injury after TBI using a controlled cortical impact model in adult male IFN-ß-deficient (IFN-ß-/-) mice and assessed post-traumatic neuroinflammatory responses, neuropathology, and long-term functional recovery. TBI increased expression of DNA sensors cyclic GMP-AMP synthase and stimulator of interferon genes in wild-type (WT) mice. IFN-ß and other IFN-related and neuroinflammatory genes were also upregulated early and persistently after TBI. TBI increased expression of proinflammatory mediators in the cortex and hippocampus of WT mice, whereas levels were mitigated in IFN-ß-/- mice. Moreover, long-term microglia activation, motor, and cognitive function impairments were decreased in IFN-ß-/- TBI mice compared with their injured WT counterparts; improved neurological recovery was associated with reduced lesion volume and hippocampal neurodegeneration in IFN-ß-/- mice. Continuous central administration of a neutralizing antibody to the IFN-α/ß receptor (IFNAR) for 3 d, beginning 30 min post-injury, reversed early cognitive impairments in TBI mice and led to transient improvements in motor function. However, anti-IFNAR treatment did not improve long-term functional recovery or decrease TBI neuropathology at 28 d post-injury. In summary, TBI induces a robust neuroinflammatory response that is associated with increased expression of IFN-ß and other IFN-related genes. Inhibition of IFN-ß reduces post-traumatic neuroinflammation and neurodegeneration, resulting in improved neurological recovery. Thus, IFN-ß may be a potential therapeutic target for TBI.SIGNIFICANCE STATEMENT TBI frequently causes long-term neurological and psychiatric changes in head injury patients. TBI-induced secondary injury processes including persistent neuroinflammation evolve over time and can contribute to chronic neurological impairments. The present study demonstrates that TBI is followed by robust activation of type I IFN pathways, which have been implicated in microglial-associated neuroinflammation and chronic neurodegeneration. We examined the effects of genetic or pharmacological inhibition of IFN-ß, a key component of type I IFN mechanisms to address its role in TBI pathophysiology. Inhibition of IFN-ß signaling resulted in reduced neuroinflammation, attenuated neurobehavioral deficits, and limited tissue loss long after TBI. These preclinical findings suggest that IFN-ß may be a potential therapeutic target for TBI.


Asunto(s)
Daño Encefálico Crónico/fisiopatología , Lesiones Traumáticas del Encéfalo/fisiopatología , Interferón beta/fisiología , Degeneración Nerviosa/etiología , Animales , Daño Encefálico Crónico/etiología , Lesiones Traumáticas del Encéfalo/complicaciones , Corteza Cerebral/metabolismo , Conducta Exploratoria/fisiología , Regulación de la Expresión Génica , Hipocampo/metabolismo , Inflamación , Interferón beta/biosíntesis , Interferón beta/deficiencia , Interferón beta/genética , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/etiología , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Trastornos del Movimiento/etiología , Trastornos del Movimiento/fisiopatología , Distribución Aleatoria , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal , Regulación hacia Arriba
6.
JCI Insight ; 5(5)2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32053518

RESUMEN

Development of gastric cancer is often preceded by chronic inflammation, but the immune cellular mechanisms underlying this process are unclear. Here we demonstrated that an inflammasome molecule, absent in melanoma 2 (Aim2), was upregulated in patients with gastric cancer and in spasmolytic polypeptide-expressing metaplasia of chronically Helicobacter felis-infected stomachs in mice. However, we found that Aim2 was not necessary for inflammasome function during gastritis. In contrast, Aim2 deficiency led to an increase in gastric CD8+ T cell frequency, which exacerbated metaplasia. These gastric CD8+ T cells from Aim2-/- mice were found to have lost their homing receptor expression (sphingosine-1-phosphate receptor 1 [S1PR1] and CD62L), a feature of tissue-resident memory T cells. The process was not mediated by Aim2-dependent regulation of IFN-ß or by dendritic cell-intrinsic Aim2. Rather, Aim2 deficiency contributed to an increased production of CXCL16 by B cells, which could suppress S1PR1 and CD62L in CD8+ T cells. This study describes a potentially novel function of Aim2 that regulates CD8+ T cell infiltration and retention within chronically inflamed solid organ tissue. This function operates independent of the inflammasome, IFN-ß, or dendritic cells. We provide evidence that B cells can contribute to this mechanism via CXCL16.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proteínas de Unión al ADN/fisiología , Gastritis/patología , Interferón beta/fisiología , Animales , Quimiocina CXCL16/metabolismo , Proteínas de Unión al ADN/genética , Gastritis/inmunología , Gastritis/metabolismo , Memoria Inmunológica , Inmunofenotipificación , Metaplasia , Ratones , Ratones Noqueados
8.
Cytokine ; 132: 154725, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31153744

RESUMEN

BACKGROUND: We have previously shown that SLE BMSC have decreased proliferation, increased ROS, increased DNA damage and repair (DDR), a senescence associated secretory phenotype, and increased senescence-associated ß-galactosidase. We have also shown SLE BMSC produce increased amounts of interferon beta (IFNß), have increased mRNA for several genes induced by IFNß, and have a pro-inflammatory feedback loop mediated by a MAVS. To better understand the phenotype of SLE BMSC we conducted mRNA sequencing. METHODS: Patients fulfilling SLE classification criteria and age and sex matched healthy controls were recruited under an Institutional Review Board approved protocol. Bone marrow aspirates and peripheral blood samples were obtained. BMSC were isolated and grown in tissue culture. Early passage BMSC were harvested and mRNA samples were sent for RNAseq. Serum samples were assayed for IFNß by ELISA. RESULTS: On the basis of top differentially expressed genes between SLE and healthy controls, SLE patients with high levels of serum IFNß clustered together while SLE patients with low levels of IFNß clustered with healthy controls. Those genes differentially expressed in SLE patients generally belonged to known IFN pathways, and showed a strong overlap with the set of genes differentially expressed in IFNß high subjects, per se. Moreover, gene expression changes induced by treating healthy BMSC with exogenous IFNß were remarkably similar to gene expression differences in SLE IFNß high vs low BMSC. CONCLUSIONS: BMSCs from SLE patients are heterogeneous. A subgroup of SLE BMSC is distinguished from other SLE BMSC and from controls by increased levels of mRNAs induced by type I interferons. This subgroup of SLE patients had increased levels of IFNß in vivo.


Asunto(s)
Células de la Médula Ósea/metabolismo , Interferón beta/fisiología , Lupus Eritematoso Sistémico/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Cultivadas , Humanos , Interferón beta/sangre , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/genética , RNA-Seq
9.
Cancer Res ; 79(20): 5394-5406, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31431457

RESUMEN

Effective cancer immunotherapy depends on the robust activation of tumor-specific antigen-presenting cells (APC). Immune agonists encapsulated within nanoparticles (NP) can be delivered to tumor sites to generate powerful antitumor immune responses with minimal off-target dissemination. Systemic delivery enables widespread access to the microvasculature and draining to the APC-rich perivasculature. We developed an immuno-nanoparticle (immuno-NP) coloaded with cyclic diguanylate monophosphate, an agonist of the stimulator of interferon genes pathway, and monophosphoryl lipid A, and a Toll-like receptor 4 agonist, which synergize to produce high levels of type I IFNß. Using a murine model of metastatic triple-negative breast cancer, systemic delivery of these immuno-NPs resulted in significant therapeutic outcomes due to extensive upregulation of APCs and natural killer cells in the blood and tumor compared with control treatments. These results indicate that NPs can facilitate systemic delivery of multiple immune-potentiating cargoes for effective APC-driven local and systemic antitumor immunity. SIGNIFICANCE: Systemic administration of an immuno-nanoparticle in a murine breast tumor model drives a robust tumor site-specific APC response by delivering two synergistic immune-potentiating molecules, highlighting the potential of nanoparticles for immunotherapy.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , GMP Cíclico/análogos & derivados , Sistemas de Liberación de Medicamentos/métodos , Interferón beta/fisiología , Lípido A/análogos & derivados , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Nanocápsulas/administración & dosificación , Receptor Toll-Like 4/agonistas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , GMP Cíclico/administración & dosificación , GMP Cíclico/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Células Asesinas Naturales/inmunología , Lípido A/administración & dosificación , Lípido A/uso terapéutico , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microcirculación , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología
10.
Cells ; 8(8)2019 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-31426476

RESUMEN

Interferon (IFN) ß and Tumor Necrosis Factor (TNF) are key players in immunity against viruses. Compelling evidence has shown that the antiviral and inflammatory transcriptional response induced by IFNß is reprogrammed by crosstalk with TNF. IFNß mainly induces interferon-stimulated genes by the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway involving the canonical ISGF3 transcriptional complex, composed of STAT1, STAT2, and IRF9. The signaling pathways engaged downstream of the combination of IFNß and TNF remain elusive, but previous observations suggested the existence of a response independent of STAT1. Here, using genome-wide transcriptional analysis by RNASeq, we observed a broad antiviral and immunoregulatory response initiated in the absence of STAT1 upon IFNß and TNF costimulation. Additional stratification of this transcriptional response revealed that STAT2 and IRF9 mediate the expression of a wide spectrum of genes. While a subset of genes was regulated by the concerted action of STAT2 and IRF9, other gene sets were independently regulated by STAT2 or IRF9. Collectively, our data supports a model in which STAT2 and IRF9 act through non-canonical parallel pathways to regulate distinct pool of antiviral and immunoregulatory genes in conditions with elevated levels of both IFNß and TNF.


Asunto(s)
Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón beta/fisiología , Factor de Transcripción STAT2/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Células A549 , Humanos
11.
J Virol ; 93(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30867297

RESUMEN

Andes virus (ANDV) causes hantavirus pulmonary syndrome (HPS) and is the only hantavirus shown to spread person to person and cause a highly lethal HPS-like disease in Syrian hamsters. The unique ability of ANDV N protein to inhibit beta interferon (IFNß) induction may contribute to its virulence and spread. Here we analyzed IFNß regulation by ANDV N protein substituted with divergent residues from the nearly identical Maporal virus (MAPV) N protein. We found that MAPV N fails to inhibit IFNß signaling and that replacing ANDV residues 252 to 296 with a hypervariable domain (HVD) from MAPV N prevents IFNß regulation. In addition, changing ANDV residue S386 to the histidine present in MAPV N or the alanine present in other hantaviruses prevented ANDV N from regulating IFNß induction. In contrast, replacing serine with phosphoserine-mimetic aspartic acid (S386D) in ANDV N robustly inhibited interferon regulatory factor 3 (IRF3) phosphorylation and IFNß induction. Additionally, the MAPV N protein gained the ability to inhibit IRF3 phosphorylation and IFNß induction when ANDV HVD and H386D replaced MAPV residues. Mass spectroscopy analysis of N protein from ANDV-infected cells revealed that S386 is phosphorylated, newly classifying ANDV N as a phosphoprotein and phosphorylated S386 as a unique determinant of IFN regulation. In this context, the finding that the ANDV HVD is required for IFN regulation by S386 but dispensable for IFN regulation by D386 suggests a role for HVD in kinase recruitment and S386 phosphorylation. These findings delineate elements within the ANDV N protein that can be targeted to attenuate ANDV and suggest targeting cellular kinases as potential ANDV therapeutics.IMPORTANCE ANDV contains virulence determinants that uniquely permit it to spread person to person and cause highly lethal HPS in immunocompetent hamsters. We discovered that ANDV S386 and an ANDV-specific hypervariable domain permit ANDV N to inhibit IFN induction and that IFN regulation is directed by phosphomimetic S386D substitutions in ANDV N. In addition, MAPV N proteins containing D386 and ANDV HVD gained the ability to inhibit IFN induction. Validating these findings, mass spectroscopy analysis revealed that S386 of ANDV N protein is uniquely phosphorylated during ANDV infection. Collectively, these findings reveal new paradigms for ANDV N protein as a phosphoprotein and IFN pathway regulator and suggest new mechanisms for hantavirus regulation of cellular kinases and signaling pathways. Our findings define novel IFN-regulating virulence determinants of ANDV, identify residues that can be modified to attenuate ANDV for vaccine development, and suggest the potential for kinase inhibitors to therapeutically restrict ANDV replication.


Asunto(s)
Interferón beta/metabolismo , Proteínas de la Nucleocápside/metabolismo , Orthohantavirus/metabolismo , Animales , Chlorocebus aethiops , Células Endoteliales/virología , Células HEK293 , Orthohantavirus/patogenicidad , Infecciones por Hantavirus/virología , Interacciones Huésped-Patógeno , Humanos , Interferón beta/fisiología , Interferones/metabolismo , Interferones/fisiología , Proteínas de la Nucleocápside/fisiología , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Serina/metabolismo , Transducción de Señal , Células Vero , Virulencia , Factores de Virulencia/metabolismo , Replicación Viral
12.
J Neurosci Res ; 97(2): 116-127, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30320448

RESUMEN

Ischemic stroke is devastating and a major cause of morbidity and mortality worldwide. To date, only clot retrieval devices and/or intravenous tissue plasminogen activators (tPA) have been approved by the US-FDA for the treatment of acute ischemic stroke. Therefore, there is an urgent need to develop an effective treatment for stroke that can have limited shortcomings and broad spectrum of applications. Interferon-beta (IFN-ß), an endogenous cytokine and a key anti-inflammatory agent, contributes toward obviating deleterious stroke outcomes. Therefore, exploring the role of IFN-ß may be a promising alternative approach for stroke intervention in the future. In the present review, we have discussed about IFN-ß along with its different mechanistic roles in ischemic stroke. Furthermore, therapeutic approaches targeting the inflammatory cascade with IFN-ß therapy that may be helpful in improving stroke outcome are also discussed.


Asunto(s)
Isquemia Encefálica/fisiopatología , Interferón beta/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Isquemia Encefálica/metabolismo , Humanos , Interferón beta/clasificación , Interferón beta/metabolismo , Accidente Cerebrovascular/metabolismo
13.
Mol Immunol ; 93: 20-30, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29127843

RESUMEN

Multiple sclerosis (MS) and the corresponding animal model, experimental autoimmune encephalomyelitis (EAE), are chronic neuroinflammatory autoimmune diseases. Increased activation of CD4+T cells, especially the Th1 and Th17 subsets, is thought to play a causal role in this disease. IFN-ß is widely used in the treatment of MS and is found to decrease IL-17 and OPN production in MS patients and EAE mice. However, a definitive molecular mechanism has not yet been fully elucidated. In this study, we investigated the immunomodulatory effect of IFN-ß on the EAE model. We observed disease progression and determined the percentage of Th1/Th17 cells in the peripheral immune organs, brain, and spinal cord of mice. Furthermore, the levels of related cytokines and transcription factors were measured in splenocytes, and the effects of IFN-ß on Th17 differentiation were assessed in vitro. Compared to the control group, IFN-ß treatment significantly reduced the incidence of EAE and the associated pathological damage. Th1 and Th17 cells in IFN-ß-treated mice were significantly reduced, and the levels of cytokines, such as IFN-γ, IL-17, and OPN, were significantly decreased in splenocyte supernatants as well as the levels of corresponding transcription factors. IFN-ß inhibited downstream inflammatory cytokines through the inhibition of PI3K/AKT/NF-κB axis and p38, JNK-MAPK, as well as the regulation of mTOR complexes. Moreover, IFN-ß inhibited Th17 differentiation and neutralizing OPN antibodies offset the inhibitory effect of IFN-ß on Th17 cells. Meanwhile, IFN-ß influenced the acetylation of the Il17a and Opn gene promoters. The findings described herein provide novel evidence for the role of IFN-ß in Th17 differentiation partly through the inhibition of OPN.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Interferón beta/fisiología , Osteopontina/fisiología , Células Th17/efectos de los fármacos , Animales , Células Cultivadas , Citocinas/biosíntesis , Citocinas/genética , Regulación hacia Abajo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Inmunosupresores/farmacología , Interferón beta/farmacología , Interferón beta/uso terapéutico , Linfopoyesis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Osteopontina/antagonistas & inhibidores , Osteopontina/biosíntesis , Osteopontina/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Regiones Promotoras Genéticas/efectos de los fármacos , Distribución Aleatoria , Organismos Libres de Patógenos Específicos , Médula Espinal/química , Médula Espinal/patología , Especificidad del Receptor de Antígeno de Linfocitos T , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th17/inmunología , Células Th17/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
14.
Vet Immunol Immunopathol ; 191: 80-93, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28895871

RESUMEN

Type I interferons (IFNs) such as IFN-α, IFN-ß, IFN-ε, IFN-κ, and IFN-ω represent cytokines, which are deeply involved in the regulation and activation of innate and adaptive immune responses. They possess strong antiviral, antiproliferative, and immunomodulatory activities allowing their use in the therapy of different viral diseases, neoplasms, and immune-mediated disorders, respectively. Initially, treatment strategies were based on nonspecific inducers of type I IFNs, which were soon replaced by different recombinant proteins. Drugs with type I IFNs as active agents are currently used in the treatment of hepatitis B and C virus infection, lymphoma, myeloid leukemia, renal carcinoma, malignant melanoma, and multiple sclerosis in humans. In addition, recombinant feline IFN-ω has been approved for the treatment of canine parvovirus, feline leukemia virus, and feline immunodeficiency virus infections. However, the role of type I IFNs in the pathogenesis of canine diseases remains largely undetermined so far, even though some share pathogenic mechanisms and clinical features with their human counterparts. This review summarizes the present knowledge of type I IFNs and down-stream targets such as Mx and 2',5'-oligoadenylate synthetase proteins in the pathogenesis of infectious and immune-mediated canine diseases. Moreover, studies investigating the potential use of type I IFNs in the treatment of canine lymphomas, melanomas, sarcomas, and carcinomas, canine distemper virus, parvovirus, and papillomavirus infections as well as immune-mediated keratoconjunctivitis sicca and atopic dermatitis are presented. A separate chapter is dedicated to the therapeutic potential of IFN-λ, a type III IFN, in canine diseases. However, further future studies are still needed to unravel the exact functions of the different subtypes of type I IFNs and their target genes in healthy and diseased dogs and the full potential action of type I IFNs as treatment strategy.


Asunto(s)
Enfermedades de los Perros/tratamiento farmacológico , Interferón Tipo I/uso terapéutico , Animales , Enfermedades de los Perros/inmunología , Perros , Interferón Tipo I/fisiología , Interferón-alfa/fisiología , Interferón-alfa/uso terapéutico , Interferón beta/fisiología , Interferón beta/uso terapéutico , Interferón gamma/fisiología , Interferón gamma/uso terapéutico
15.
J Immunol ; 198(10): 4036-4045, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28411186

RESUMEN

Type I IFNs are a cytokine family essential for antiviral defense. More recently, type I IFNs were shown to be important during bacterial infections. In this article, we show that, in addition to known cytokine functions, IFN-ß is antimicrobial. Parts of the IFN-ß molecular surface (especially helix 4) are cationic and amphipathic, both classic characteristics of antimicrobial peptides, and we observed that IFN-ß can directly kill Staphylococcus aureus Further, a mutant S. aureus that is more sensitive to antimicrobial peptides was killed more efficiently by IFN-ß than was the wild-type S. aureus, and immunoblotting showed that IFN-ß interacts with the bacterial cell surface. To determine whether specific parts of IFN-ß are antimicrobial, we synthesized IFN-ß helix 4 and found that it is sufficient to permeate model prokaryotic membranes using synchrotron x-ray diffraction and that it is sufficient to kill S. aureus These results suggest that, in addition to its well-known signaling activity, IFN-ß may be directly antimicrobial and be part of a growing family of cytokines and chemokines, called kinocidins, that also have antimicrobial properties.


Asunto(s)
Antibacterianos/farmacología , Membrana Celular/efectos de los fármacos , Interferón beta/fisiología , Staphylococcus aureus/efectos de los fármacos , Animales , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Humanos , Interferón beta/química , Interferón beta/metabolismo , Interferón beta/farmacología , Ratones , Pruebas de Sensibilidad Microbiana , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Difracción de Rayos X
16.
Nucleic Acids Res ; 45(6): 3231-3241, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28100701

RESUMEN

Foreign and self-cytoplasmic DNA are recognized by numerous DNA sensor molecules leading to the production of type I interferons. Such DNA agonists should be degraded otherwise cells would be chronically stressed. Most human APOBEC3 cytidine deaminases can initiate catabolism of cytoplasmic mitochondrial DNA. Using the human myeloid cell line THP-1 with an interferon inducible APOBEC3A gene, we show that cytoplasmic DNA triggers interferon α and ß production through the RNA polymerase III transcription/RIG-I pathway leading to massive upregulation of APOBEC3A. By catalyzing C→U editing in single stranded DNA fragments, the enzyme prevents them from re-annealing so attenuating the danger signal. The price to pay is chromosomal DNA damage in the form of CG→TA mutations and double stranded DNA breaks which, in the context of chronic inflammation, could drive cells down the path toward cancer.


Asunto(s)
Citidina Desaminasa/biosíntesis , Roturas del ADN de Doble Cadena , ADN Mitocondrial/metabolismo , Línea Celular Tumoral , Cromosomas Humanos , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Citosol/metabolismo , Proteína 58 DEAD Box , ADN Mitocondrial/química , Humanos , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Interferón beta/fisiología , Proteínas/genética , Proteínas/metabolismo , ARN Polimerasa III/metabolismo , Receptores Inmunológicos , Transcripción Genética , Regulación hacia Arriba , Uracilo/metabolismo
17.
J Virol ; 91(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031360

RESUMEN

The DNA sensing pathway triggers innate immune responses against DNA virus infection, and NF-κB signaling plays a critical role in establishing innate immunity. We report here that the herpes simplex virus 1 (HSV-1) ubiquitin-specific protease (UL36USP), which is a deubiquitinase (DUB), antagonizes NF-κB activation, depending on its DUB activity. In this study, ectopically expressed UL36USP blocked promoter activation of beta interferon (IFN-ß) and NF-κB induced by cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING). UL36USP restricted NF-κB activation mediated by overexpression of STING, TANK-binding kinase 1, IκB kinase α (IKKα), and IKKß, but not p65. UL36USP was also shown to inhibit IFN-stimulatory DNA-induced IFN-ß and NF-κB activation under conditions of HSV-1 infection. Furthermore, UL36USP was demonstrated to deubiquitinate IκBα and restrict its degradation and, finally, abrogate NF-κB activation. More importantly, the recombinant HSV-1 lacking UL36USP DUB activity, denoted as C40A mutant HSV-1, failed to cleave polyubiquitin chains on IκBα. For the first time, UL36USP was shown to dampen NF-κB activation in the DNA sensing signal pathway to evade host antiviral innate immunity.IMPORTANCE It has been reported that double-stranded-DNA-mediated NF-κB activation is critical for host antiviral responses. Viruses have established various strategies to evade the innate immune system. The N terminus of the HSV-1 UL36 gene-encoded protein contains the DUB domain and is conserved across all herpesviruses. This study demonstrates that UL36USP abrogates NF-κB activation by cleaving polyubiquitin chains from IκBα and therefore restricts proteasome-dependent degradation of IκBα and that DUB activity is indispensable for this process. This study expands our understanding of the mechanisms utilized by HSV-1 to evade the host antiviral innate immune defense induced by NF-κB signaling.


Asunto(s)
Herpesvirus Humano 1/fisiología , FN-kappa B/metabolismo , Proteínas Virales/fisiología , Animales , Chlorocebus aethiops , Células HEK293 , Humanos , Interferón beta/fisiología , Proteolisis , Transducción de Señal , Ubiquitinación , Células Vero
18.
Int Rev Cell Mol Biol ; 326: 343-72, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27572132

RESUMEN

Interferon beta (IFNß) is a cytokine that is naturally produced by the immune system in response to biological and chemical stimuli. It signals by binding to the heterodimeric type I IFN receptor composed of the IFNAR1 and IFNAR2 chains, and regulates the expression of a plethora of genes by means of the classical JAK/STAT and other pathways. IFNß is pleiotropic in that it elicits antiviral, antiproliferative, and immunomodulatory activities on numerous cell types. The biological activities underpin the mechanisms by which the protein is used to treat various diseases such as hepatitis C infection and multiple sclerosis. Despite the success of IFNß therapy, the drug may evoke the production of antidrug antibodies that may reduce treatment efficiency. Immunogenicity is related to many factors: among them, structural properties, particularly aggregation, and T-cell and B-cell epitopes in the structure of IFNß, appear to be important. Knowledge of the structural properties of IFNß and its relation to immunogenicity may help scientists to develop safer and more effective forms. Several methods have been used to predict and reduce the immunogenicity of certain IFNß drug products. In this chapter, we review the current knowledge on IFNß from its structure, dynamic conformation, signaling pathway, and mechanism of action to its therapeutic effects. Immunogenicity and its relation to structural properties of IFNß are also discussed.


Asunto(s)
Interferón beta/metabolismo , Transducción de Señal , Animales , Antivirales , Humanos , Factores Inmunológicos , Interferón beta/farmacología , Interferón beta/fisiología , Interferón beta/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Estructura Terciaria de Proteína
19.
Yi Chuan ; 38(5): 411-7, 2016 05.
Artículo en Chino | MEDLINE | ID: mdl-27232489

RESUMEN

Rodents, including the nude mice with congenital aplasia of the thymus, cancer-resistant naked mole rat (Heterocephalus glaber) and blind mole rat (Spalax galili), are important model organisms that are widely used in biomedical research. The aging process is closely related to cancer incidence in mammals and the aging degree is positively correlated with the risk of cancer. Since rodents account for 40% of mammals, study of the unique antitumor mechanism in long-lived rodents is very important. Replicative senescence is anti-tumor mechanism that prevalently exist in rodents, however, unique anti-tumor mechanisms have been found in naked mole-rats and blind mole-rats. The cancer resistance of Spalax galili is mediated by cell-released IFN-ß which activates p53 and Rb signaling pathway and the cells undergoes concerted cell death while that of Heterocephalus glaber is mediated by high molecular weight hyaluronan (HMW-HA) which causes contact inhibition. In addition, highly expressed pro-cell-death and anti-inflammation related genes are found in the genome of both naked mole-rats and blind mole-rats. In this review, we summarize the anti-tumor mechanisms in both Heterocephalus glaber and Spalax galili, which may provide information for related research.


Asunto(s)
Ratas Topo/genética , Neoplasias/veterinaria , Enfermedades de los Roedores/genética , Spalax/genética , Animales , Senescencia Celular , Ácido Hialurónico/fisiología , Interferón beta/fisiología , Neoplasias/genética , Proteína de Retinoblastoma/fisiología , Proteína p53 Supresora de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA