Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.952
Filtrar
1.
J Ethnopharmacol ; 285: 114895, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34875348

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Datura metel L. has been used as an anesthetic in clinic for more than 1800 years in China, and the main efficacy of D. metel L. flower is relieving asthma and cough, relieving spasm and relieving pain. From 1978 to 1980, Datura metel L. was used as an anesthetic agent and occasionally cured psoriasis patients during anesthesia clinically, and our group confirmed that the effective portion is total withanolides (YWS). Moreover, the new drug "Datura metel L. capsule" composed of YWS has since been approved and used for the treatment of more than 3,000 psoriasis patients, with efficacy and cure rates greater than 90% and 65%. However, the immunological mechanism has not been elucidated. AIM OF THE STUDY: Nowadays, although total withanolides from Datura metel L. have a better clinical efficacy in the treatment of psoriasis, there is a lack of overall understanding of the mechanism of their treatment, especially about some immune cells and proteins closely related to psoriasis and their relationship in executive function and biological significance. This study focused on investigating the mechanism of psoriasis treatment by YWS and determined the biochemical processes in the treatment of psoriasis based on Treg/Th17 axis cell-mediated bidirectional immunoregulatory functions, which provides an important scientific basis for understanding the mechanism underlying the treatment of psoriasis by YWS. MATERIALS AND METHODS: The effects of YWS on the lesion pathology of IMQ-induced psoriasis mice and the underlying molecular mechanism were assessed directly using HE staining, the PASI score and the animal body mass. We also investigated the effects of YWS on the Treg/Th17 axis and their critical functions in psoriasis pathogenesis via molecular biological methods. Finally, we performed differential proteomics analysis on skin in IMQ-induced psoriasis mice to clarify the effect of YWS by incorporates mass spectrometry-bioinformatics and annotated the functions and pathways associated with the differential proteins through GO enrichment, KEGG pathway analysis and PPI networks analysis, respectively. RESULTS: YWS regulated the imbalance of the Treg/Th17 axis. And proteomic analysis showed that YWS up-regulated 46 and down-regulated 37 proteins. According to the bioinformatics analysis, the improvement of Treg/Th17 imbalance may be the key immunological mechanism of YWS in the treatment of psoriasis by up-regulating the butyrate metabolism pathway, down-regulating leukocyte migration, inhibiting the phagocytic function of natural killer cells, suppressing osteoclast differentiation and interfering with chemokine activity, and the critical proteins involved are Lyn, HMGCS2, ABAT, ITGß2, PRKCß, MMP9, NCF1, JUNß, and Hck. CONCLUSION: This research clarified that the improvement of the imbalance of the Treg/Th17 axis may be the key immunological mechanism of YWS in the treatment of psoriasis through metabolic pathways and influencing key proteins. The results not only expand the therapeutic targets and approaches for the treatment of psoriasis, which is a challenging and complex disease, but also deepens the understanding of the mechanism of YWS in the treatment of psoriasis and other important conditions to open up a new way of thinking for research on YWS in the treatment of psoriasis.


Asunto(s)
Imiquimod/toxicidad , Inflamación/tratamiento farmacológico , Psoriasis/tratamiento farmacológico , Witanólidos/uso terapéutico , Animales , Biología Computacional , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Inductores de Interferón/toxicidad , Masculino , Ratones , Ratones Endogámicos BALB C , Mapas de Interacción de Proteínas , Psoriasis/inducido químicamente , Distribución Aleatoria , Transducción de Señal , Regulación hacia Arriba/efectos de los fármacos , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
2.
Trends Immunol ; 42(12): 1054-1056, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34764015

RESUMEN

Type 1 interferon (IFN) is a major contributor to the pathogenesis of systemic lupus erythematosus (SLE). A landmark study by Caielli et al. now shows that erythrocytes from lupus patients that fail to switch from glycolysis to oxidative phosphorylation during differentiation retain their mitochondria. These mitochondria-containing erythrocytes represent a novel source of IFN when phagocytosed by macrophages.


Asunto(s)
Interferón Tipo I , Lupus Eritematoso Sistémico , Eritrocitos , Humanos , Inductores de Interferón/metabolismo , Interferón Tipo I/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Mitocondrias
3.
J Neurosci ; 41(48): 9971-9987, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34607967

RESUMEN

Human epidemiological studies implicate exposure to infection during gestation in the etiology of neurodevelopmental disorders. Animal models of maternal immune activation (MIA) have identified the maternal immune response as the critical link between maternal infection and aberrant offspring brain and behavior development. Here we evaluate neurodevelopment of male rhesus monkeys (Macaca mulatta) born to MIA-treated dams (n = 14) injected with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the same gestational time points (n = 10) or were untreated (n = 4). MIA-treated dams exhibited a strong immune response as indexed by transient increases in sickness behavior, temperature, and inflammatory cytokines. Although offspring born to control or MIA-treated dams did not differ on measures of physical growth and early developmental milestones, the MIA-treated animals exhibited subtle changes in cognitive development and deviated from species-typical brain growth trajectories. Longitudinal MRI revealed significant gray matter volume reductions in the prefrontal and frontal cortices of MIA-treated offspring at 6 months that persisted through the final time point at 45 months along with smaller frontal white matter volumes in MIA-treated animals at 36 and 45 months. These findings provide the first evidence of early postnatal changes in brain development in MIA-exposed nonhuman primates and establish a translationally relevant model system to explore the neurodevelopmental trajectory of risk associated with prenatal immune challenge from birth through late adolescence.SIGNIFICANCE STATEMENT Women exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder. Preclinical maternal immune activation (MIA) models have demonstrated that the effects of maternal infection on fetal brain development are mediated by maternal immune response. Since the majority of MIA models are conducted in rodents, the nonhuman primate provides a unique system to evaluate the MIA hypothesis in a species closely related to humans. Here we report the first longitudinal study conducted in a nonhuman primate MIA model. MIA-exposed offspring demonstrate subtle changes in cognitive development paired with marked reductions in frontal gray and white matter, further supporting the association between prenatal immune challenge and alterations in offspring neurodevelopment.


Asunto(s)
Encéfalo/patología , Modelos Animales de Enfermedad , Trastornos del Neurodesarrollo/etiología , Complicaciones Infecciosas del Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Animales , Femenino , Inductores de Interferón/toxicidad , Macaca mulatta , Masculino , Trastornos del Neurodesarrollo/patología , Neurogénesis/fisiología , Poli I-C/toxicidad , Embarazo , Complicaciones Infecciosas del Embarazo/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
4.
Int J Mol Sci ; 22(19)2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34639115

RESUMEN

Psoriasis is a chronic inflammatory skin disease. Recently, lysophosphatidic acid (LPA)/LPAR5 signaling has been reported to be involved in both NLRP3 inflammasome activation in macrophages and keratinocyte activation to produce inflammatory cytokines, contributing to psoriasis pathogenesis. However, the effect and molecular mechanisms of LPA/LPAR signaling in keratinocyte proliferation in psoriasis remain unclear. In this study, we investigated the effects of LPAR1/3 inhibition on imiquimod (IMQ)-induced psoriasis-like mice. Treatment with the LPAR1/3 antagonist, ki16425, alleviated skin symptoms in IMQ-induced psoriasis-like mouse models and decreased keratinocyte proliferation in the lesion. It also decreased LPA-induced cell proliferation and cell cycle progression via increased cyclin A2, cyclin D1, cyclin-dependent kinase (CDK)2, and CDK4 expression and decreased p27Kip1 expression in HaCaT cells. LPAR1 knockdown in HaCaT cells reduced LPA-induced proliferation, suppressed cyclin A2 and CDK2 expression, and restored p27Kip1 expression. LPA increased Rho-associated protein kinase 2 (ROCK2) expression and PI3K/AKT activation; moreover, the pharmacological inhibition of ROCK2 and PI3K/AKT signaling suppressed LPA-induced cell cycle progression. In conclusion, we demonstrated that LPAR1/3 antagonist alleviates IMQ-induced psoriasis-like symptoms in mice, and in particular, LPAR1 signaling is involved in cell cycle progression via ROCK2/PI3K/AKT pathways in keratinocytes.


Asunto(s)
Proliferación Celular , Regulación de la Expresión Génica/efectos de los fármacos , Imiquimod/toxicidad , Queratinocitos/citología , Lisofosfolípidos/farmacología , Psoriasis/tratamiento farmacológico , Animales , Apoptosis , Biomarcadores/metabolismo , Ciclo Celular , Células Cultivadas , Humanos , Inductores de Interferón/toxicidad , Queratinocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Psoriasis/inducido químicamente , Psoriasis/metabolismo , Psoriasis/patología , Receptores del Ácido Lisofosfatídico/genética , Receptores del Ácido Lisofosfatídico/metabolismo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
5.
Bull Exp Biol Med ; 171(6): 736-740, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34705177

RESUMEN

We studied the effect of tilorone on the dynamics of IFNα, IFNγ, and IL-1ß levels in the lung tissue and blood serum in relation to viral load in the lungs of BALB/c mice with pneumonia caused by influenza virus A/Aichi/2/68 (H3N2). Tilorone was administered per os in doses of 40, 150, and 540 µg per mouse 6, 30, and 78 h postinfection, which simulated the drug regimen used in the clinic for the treatment of influenza and acute respiratory viral infections in Russia and post-Soviet countries. Tilorone reduced viral load with the maximum amplitude (2-3 lg) after 1-2 administrations. The results of studying the dynamics of the cytokine levels in the infected animals in general support the previous hypothesis that, in repeated dosing, tilorone enhances the IFN response (compensates for its deficiency) at the early stages of acute respiratory viral infections and suppresses (damps) excessive production of IFN and proinflammatory cytokines at the later stages.


Asunto(s)
Antivirales/farmacología , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Inductores de Interferón/farmacología , Pulmón/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Tilorona/farmacología , Animales , Esquema de Medicación , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Subtipo H3N2 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Interferón-alfa/sangre , Interferón-alfa/inmunología , Interferón gamma/sangre , Interferón gamma/inmunología , Interleucina-1beta/sangre , Interleucina-1beta/inmunología , Pulmón/inmunología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/sangre , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Carga Viral/efectos de los fármacos
6.
J Leukoc Biol ; 110(6): 1047-1055, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34494306

RESUMEN

Tumor necrosis factor alpha (TNF) has been implicated in the pathogenesis of psoriasis and anti-TNF therapeutics are used in the treatment of psoriasis in the clinic. However, considerable proportion of patients fail to respond to anti-TNF treatment. Furthermore, anti-TNF therapy induces de novo development of psoriasis in some patients with other type of autoimmune disorders. Therefore, further understanding of the role of TNF-TNFR signaling in pathogenesis of psoriasis remains a critical to devise safer and more effective treatment. In this study, it is shown that in imiquimod-induced mouse psoriasis model, TNF receptor type 1 (TNFR1) deficiency inhibited the development of skin diseases. In sharp contrast, TNF receptor type 2 (TNFR2) deficiency led to more severe psoriasis that was associated with increased Th1 and Th17 responses and reduced number of CD4+ Foxp3+ regulatory T cells (Tregs). Importantly, adoptive transfer of WT Tregs was able to attenuate inflammatory responses in imiquimod-treated TNFR2-/- mice, suggestive of a role of malfunctioned Tregs in mice deficient in TNFR2. RNA sequencing data revealed that Tregs deficient in TNFR2 exhibited down-regulation of different biological processes linked to proliferative expansion. Taken together, our study clearly indicated that TNFR1 was pathogenic in mouse psoriasis. In contrast, through boosting the proliferative expansion of Tregs, TNFR2 was protective in this model. The data thus suggest that TNFR1-specific antagonist or TNFR2-specific agonist may be useful in the treatment of patients with psoriasis.


Asunto(s)
Imiquimod/toxicidad , Psoriasis/inducido químicamente , Psoriasis/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Inductores de Interferón/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Psoriasis/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Linfocitos T Reguladores/inmunología
7.
J Neurosci ; 41(32): 6954-6965, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34253630

RESUMEN

Episodic memory requires information to be stored and recalled in sequential order, and these processes are disrupted in schizophrenia. Hippocampal phase precession and theta sequences are thought to provide a biological mechanism for sequential ordering of experience at timescales suitable for plasticity. These phenomena have not previously been examined in any models of schizophrenia risk. Here, we examine these phenomena in a maternal immune activation (MIA) rodent model. We show that while individual pyramidal cells in the CA1 region continue to precess normally in MIA animals, the starting phase of precession as an animal enters a new place field is considerably more variable in MIA animals than in controls. A critical consequence of this change is a disorganization of the ordered representation of experience via theta sequences. These results provide the first evidence of a biological-level mechanism that, if it occurs in schizophrenia, may explain aspects of disorganized sequential processing that contribute to the cognitive symptoms of the disorder.SIGNIFICANCE STATEMENT Hippocampal phase precession and theta sequences have been proposed as biophysical mechanisms by which the sequential structure of cognition might be ordered. Disturbances of sequential processing have frequently been observed in schizophrenia. Here, we show for the first time that phase precession and theta sequences are disrupted in a maternal immune activation (MIA) model of schizophrenia risk. This is a result of greater variability in the starting phase of precession, indicating that the mechanisms that coordinate precession at the assembly level are disrupted. We propose that this disturbance in phase precession underlies some of the disorganized cognitive symptoms that occur in schizophrenia. These findings could have important preclinical significance for the identification and treatment of schizophrenia risk factors.


Asunto(s)
Hipocampo/fisiopatología , Memoria Episódica , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Esquizofrenia/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Inflamación/inducido químicamente , Inductores de Interferón/toxicidad , Masculino , Exposición Materna/efectos adversos , Poli I-C/toxicidad , Embarazo , Ratas Sprague-Dawley , Esquizofrenia/etiología
8.
J Neuroimmunol ; 357: 577620, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34062352

RESUMEN

Neuropsychiatric systemic lupus erythematosus (NPSLE) is the most serious and complicated clinical manifestation of lupus erythematosus. Cognitive dysfunction is the most common symptom of NPSLE. A variety of potential mechanisms or mediators related to the pathogenesis of NPSLE cognitive dysfunction have been proposed. However, the involvement of microglia CD40 has not been reported yet. This study aimed to investigate whether hippocampal microglia CD40 of MRL/MpJ-Faslpr (MRL/lpr) mice was involved in NPSLE cognitive dysfunction. This study found, using quantitative polymerase chain reaction, western blotting and immunohistochemistry, that hippocampal CD40 was aberrantly overexpressed in the MRL/lpr lupus mice. It also determined using flow cytometry and immunofluorescence that the aberrantly overexpressed CD40 was mainly derived from hippocampal microglia. The adeno-associated virus was used to inhibit microglia CD40 expression, and the brain damage and cognitive dysfunction of MRL/lpr mice improved. Also, imiquimod (IMQ)-induced lupus mice had the same NPSLE cognitive dysfunction, brain damage, and overexpressed hippocampal microglia CD40 as MRL/lpr mice. Therefore, IMQ-induced lupus mouse was proposed as one of the mouse models for studying NPSLE cognitive dysfunction for the first time in this study. The findings indicated that hippocampal microglia CD40 was involved in the development of NPSLE cognitive dysfunction, thus providing a novel research direction for the study of the pathogenesis of NPSLE.


Asunto(s)
Antígenos CD40/metabolismo , Disfunción Cognitiva/metabolismo , Hipocampo/metabolismo , Vasculitis por Lupus del Sistema Nervioso Central/metabolismo , Microglía/metabolismo , Animales , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Femenino , Imiquimod/farmacología , Inductores de Interferón/farmacología , Vasculitis por Lupus del Sistema Nervioso Central/complicaciones , Ratones , Ratones Endogámicos MRL lpr
9.
Mol Immunol ; 136: 110-117, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34098343

RESUMEN

Reticuloendothelial virus (REV) is widely found in many domestic poultry areas and results in severe immunosuppression of infected chickens. This increases the susceptibility to other pathogens, which causes economic losses to the poultry industry. The aim of our study was to determine whether polyinosinic-polycytidylic acid [Poly (I: C)] treatment could inhibit REV replication in chicken macrophage-like cell line, HD11. We found that Poly (I: C) treatment could markedly inhibit REV replication in HD11 from 24 to 48 h post infection (hpi). Additionally, Poly (I: C) treatment could switch HD11 from an inactive type into M1-like polarization from 24 to 48 hpi. Furthermore, Poly (I: C) treatment promoted interferon-ß secretion from HD11 post REV infection. Moreover, Toll-like receptor-3 (TLR-3) mRNA and protein levels in HD11 treated with Poly (I: C) were markedly increased compared to those of HD11 not treated with Poly (I: C). The above results suggested that Poly (I: C) treatment switches HD11 into M1-like polarization to secret more interferon-ß and activate TLR-3 signaling, which contributes to block REV replication. Our findings provide a theoretical reference for further studying the underlying pathogenic mechanism of REV and Poly (I: C) as a potential therapeutic intervention against REV infection.


Asunto(s)
Antivirales/farmacología , Inductores de Interferón/farmacología , Poli I-C/farmacología , Virus de la Reticuloendoteliosis Aviar/crecimiento & desarrollo , Receptor Toll-Like 3/metabolismo , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Pollos , Interferón beta/biosíntesis , Interferón beta/metabolismo , Macrófagos/inmunología , Macrófagos/virología , Virus de la Reticuloendoteliosis Aviar/efectos de los fármacos , Infecciones por Retroviridae/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Infecciones Tumorales por Virus/tratamiento farmacológico
10.
J Leukoc Biol ; 110(6): 1197-1208, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33988261

RESUMEN

Macrophage phagocytosis plays essential roles in antitumor immunity. CD47/SIRPα phagocytosis checkpoint blockade has demonstrated therapeutic potential in several hematopoietic cancers, but recent clinical studies reported very limited efficacy against solid malignancies. Here, we show that polyinosinic-polycytidylic acid (Poly(I:C)), a synthetic analog of double-stranded RNA, enhances the antitumor activity of CD47 blockade in colorectal cancer in vitro and in vivo. Poly(I:C) activation leads to a potent immune response characterized by the production of proinflammatory cytokines, especially IL-6. Stimulation with IL-6 promotes the PI3K signaling and cytoskeletal reorganization required for macrophage phagocytosis mediated by CD47 blockade. Our findings demonstrate the potential of Poly(I:C) to synergize the efficacy of CD47 blockade therapy and a novel role for IL-6 in macrophage phagocytosis, which provide new strategy for combinational cancer immunotherapy.


Asunto(s)
Neoplasias Colorrectales/inmunología , Inmunoterapia/métodos , Interleucina-6/biosíntesis , Macrófagos/inmunología , Poli I-C/farmacología , Animales , Antígeno CD47/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/inmunología , Inductores de Interferón/farmacología , Ratones Endogámicos C57BL , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología
11.
Viruses ; 13(3)2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33807534

RESUMEN

The mitochondrial antiviral-signaling protein (MAVS, also known as VISA, IPS-1, or CARDIF) plays an essential role in the type I interferon (IFN) response and in retinoic acid-inducible gene I (RIG-I) mediated antiviral innate immunity in mammals. In this study, the caprine MAVS gene (caMAVS, 1566 bp) was identified and cloned. The caMAVS shares the highest amino acid similarity (98.1%) with the predicted sheep MAVS. Confocal microscopy analysis of partial deletion mutants of caMAVS revealed that the transmembrane and the so-called Non-Characterized domains are indispensable for intracellular localization to mitochondria. Overexpression of caMAVS in caprine endometrial epithelial cells up-regulated the mRNA levels of caprine interferon-stimulated genes. We concluded that caprine MAVS mediates the activation of the type I IFN pathway. We further demonstrated that both the CARD-like domain and the transmembrane domain of caMAVS were essential for the activation of the IFN-ß promotor. The interaction between caMAVS and caprine RIG-I and the vital role of the CARD and NC domain in this interaction was demonstrated by co-immunoprecipitation. Upon infection with the Peste des Petits Ruminants Virus (PPRV, genus Morbillivirus), the level of MAVS was greatly reduced. This reduction was prevented by the addition of the proteasome inhibitor MG132. Moreover, we found that viral protein V could interact and colocalize with MAVS. Together, we identified caMAVS as a RIG-I interactive protein involved in the activation of type I IFN pathways in caprine cells and as a target for PPRV immune evasion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Inductores de Interferón/inmunología , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Animales , Chlorocebus aethiops , Células Epiteliales , Cabras , Células HEK293 , Humanos , Interferón Tipo I/inmunología , Células Vero
12.
Cancer Res ; 81(12): 3374-3386, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33837043

RESUMEN

Screening for sensitizers of cancer cells to TRAIL-mediated apoptosis identified a natural product of the 17ß-hydroxywithanolide (17-BHW) class, physachenolide C (PCC), as a promising hit. In this study, we show that PCC was also able to sensitize melanoma and renal carcinoma cells to apoptosis in response not only to TRAIL, but also to the synthetic polynucleotide poly I:C, a viral mimetic and immune activator, by reducing levels of antiapoptotic proteins cFLIP and Livin. Both death receptor and TLR3 signaling elicited subsequent increased assembly of a proapoptotic ripoptosome signaling complex. Administration of a combination of PCC and poly I:C in human M14 melanoma xenograft and a syngeneic B16 melanoma model provided significant therapeutic benefit as compared with individual agents. In addition, PCC enhanced melanoma cell death in response to activated human T cells in vitro and in vivo in a death ligand-dependent manner. Biochemical mechanism-of-action studies established bromo and extraterminal domain (BET) proteins as major cellular targets of PCC. Thus, by targeting of BET proteins to reduce antiapoptotic proteins and enhance caspase-8-dependent apoptosis of cancer cells, PCC represents a unique agent that can potentially be used in combination with various immunotherapeutic approaches to promote tumor regression and improve outcome. SIGNIFICANCE: These findings demonstrate that PCC selectively sensitizes cancer cells to immune-mediated cell death, potentially improving the efficacy of cancer immunotherapies. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3374/F1.large.jpg.


Asunto(s)
Productos Biológicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Inmunoterapia/métodos , Melanoma Experimental/tratamiento farmacológico , Poli I-C/farmacología , Factores de Transcripción/antagonistas & inhibidores , Witanólidos/farmacología , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Proliferación Celular , Quimioterapia Combinada , Femenino , Humanos , Inductores de Interferón/farmacología , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
JCI Insight ; 6(8)2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33884963

RESUMEN

It remains unresolved how retinal pigment epithelial cell metabolism is regulated following immune activation to maintain retinal homeostasis and retinal function. We exposed retinal pigment epithelium (RPE) to several stress signals, particularly Toll-like receptor stimulation, and uncovered an ability of RPE to adapt their metabolic preference on aerobic glycolysis or oxidative glucose metabolism in response to different immune stimuli. We have identified interleukin-33 (IL-33) as a key metabolic checkpoint that antagonizes the Warburg effect to ensure the functional stability of the RPE. The identification of IL-33 as a key regulator of mitochondrial metabolism suggests roles for the cytokine that go beyond its extracellular "alarmin" activities. IL-33 exerts control over mitochondrial respiration in RPE by facilitating oxidative pyruvate catabolism. We have also revealed that in the absence of IL-33, mitochondrial function declined and resultant bioenergetic switching was aligned with altered mitochondrial morphology. Our data not only shed new light on the molecular pathway of activation of mitochondrial respiration in RPE in response to immune stressors but also uncover a potentially novel role of nuclear intrinsic IL-33 as a metabolic checkpoint regulator.


Asunto(s)
Respiración de la Célula/fisiología , Glucólisis/fisiología , Interleucina-33/metabolismo , Mitocondrias/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular , Metabolismo Energético , Técnicas de Silenciamiento del Gen , Glucólisis/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Inductores de Interferón/farmacología , Interleucina-33/efectos de los fármacos , Interleucina-33/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Oxidantes/farmacología , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo , Poli I-C/farmacología , Cultivo Primario de Células , Ácido Pirúvico/metabolismo
14.
Cancer Immunol Immunother ; 70(11): 3081-3091, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33751208

RESUMEN

The nanoparticle complex of cholesteryl pullulan (CHP) and NY-ESO-1 antigen protein (CHP-NY-ESO-1) presents multiple epitope peptides to MHC class I and II pathways, leading to CD8+ and CD4+ T cell responses. Poly-ICLC is a synthetic, double-stranded RNA, an agonist of toll-like receptor (TLR)-3, and a cytoplasmic receptor of melanoma differentiation-associated gene (MDA)-5. It should be a suitable immune adjuvant of cancer vaccine to overcome the inhibitory tumor microenvironment. We conducted a phase 1 clinical trial of CHP-NY-ESO-1 with poly-ICLC in patients with advanced or recurrent esophageal cancer. CHP-NY-ESO-1/poly-ICLC (µg/mg) was administered at a dose of 200/0.5 or 200/1.0 (cohorts 1 and 2, respectively) every 2 weeks for a total of six doses. The primary endpoints were safety and immune response. The secondary endpoint was tumor response. In total, 16 patients were enrolled, and six patients in each cohort completed the trial. The most common adverse event (AE) was injection site skin reaction (86.7%). No grade 3 or higher drug-related AEs were observed. No tumor responses were observed, and three patients (30%) had stable disease. The immune response was comparable between the two cohorts, and all patients (100%) achieved antibody responses with a median of 2.5 vaccinations. Comparing CHP-NY-ESO-1 alone to the poly-ICLC combination, all patients in both groups exhibited antibody responses, but the titers were higher in the combination group. In a mouse model, adding anti-PD-1 antibody to the combination of CHP-NY-ESO-1/poly-ICLC suppressed the growth of NY-ESO-1-expressing tumors. Combining the vaccine with PD-1 blockade holds promise in human trials.


Asunto(s)
Antígenos de Neoplasias/uso terapéutico , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Carboximetilcelulosa de Sodio/análogos & derivados , Neoplasias Esofágicas/tratamiento farmacológico , Glucanos/uso terapéutico , Proteínas de la Membrana/uso terapéutico , Poli I-C/uso terapéutico , Polilisina/análogos & derivados , Adyuvantes Inmunológicos/uso terapéutico , Anciano , Anciano de 80 o más Años , Animales , Antígenos de Neoplasias/inmunología , Carboximetilcelulosa de Sodio/uso terapéutico , Neoplasias Esofágicas/inmunología , Femenino , Glucanos/inmunología , Humanos , Inductores de Interferón/inmunología , Inductores de Interferón/uso terapéutico , Masculino , Proteínas de la Membrana/inmunología , Ratones , Persona de Mediana Edad , Nanopartículas , Poli I-C/inmunología , Polilisina/inmunología , Polilisina/uso terapéutico
15.
Brain Res ; 1759: 147370, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33600830

RESUMEN

Genes and environment interact during intrauterine life, and potentially alter the developmental trajectory of the brain. This can result in life-long consequences on brain function. We have previously developed two transgenic mouse lines that suppress Gad1 expression in parvalbumin (PVALB) and neuropeptide Y (NPY) expressing interneuron populations using a bacterial artificial chromosome (BAC)-driven miRNA-based silencing technology. We were interested to assess if maternal immune activation (MIA), genetic interneuronal inhibition, and the combination of these two factors disrupt and result in long-term changes in neuroinflammatory gene expression, sterol biosynthesis, and acylcarnitine levels in the brain of maternally exposed offspring. Pregnant female WT mice were given a single intraperitoneal injection of saline or polyinosinic-polycytidilic acid [poly(I:C)] at E12.5. Brains of offspring were analyzed at postnatal day 90. We identified complex and persistent neuroinflammatory gene expression changes in the hippocampi of MIA-exposed offspring, as well in the hippocampi of Npy/Gad1 and Pvalb/Gad1 mice. In addition, both MIA and genetic inhibition altered the post-lanosterol sterol biosynthesis in the neocortex and disrupted the typical acylcarnitine profile. In conclusion, our findings suggest that both MIA and inhibition of interneuronal function have long-term consequences on critical homeostatic mechanisms of the brain, including immune function, sterol levels, and energy metabolism.


Asunto(s)
Mediadores de Inflamación/inmunología , Interneuronas/inmunología , Neuroinmunomodulación/fisiología , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/inmunología , Animales , Femenino , Glutamato Descarboxilasa/deficiencia , Glutamato Descarboxilasa/genética , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Hipocampo/metabolismo , Mediadores de Inflamación/metabolismo , Inductores de Interferón/toxicidad , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neocórtex/efectos de los fármacos , Neocórtex/inmunología , Neocórtex/metabolismo , Neuroinmunomodulación/efectos de los fármacos , Poli I-C/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo
16.
FEBS J ; 288(14): 4249-4266, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33452755

RESUMEN

Viral infection is a significant burden to health care worldwide. Statins, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibitors, are widely used as cholesterol-lowering drugs. Recently, long-term statin therapy was shown to reduce the antiviral immune response; however, the underlying molecular mechanisms are unclear. Here, we found that simvastatin decreased polyinosinic-polycytidylic acid [poly(I:C)]-induced expression of antiviral interferon (IFN)-ß and IFN-stimulated genes (ISGs) in the bronchoalveolar lavage fluid (BALF) and lungs of mice with high-fat diet-induced hyperlipidemia. Macrophages were the dominant cell type in the BALF of poly(I:C)-treated mice. We examined the effects of simvastatin in primary lung macrophages and found that simvastatin suppressed poly(I:C)-induced expression of IFN-ß and ISGs. We examined the molecular mechanisms of statin-mediated inhibition of antiviral gene expression using murine macrophage-like cell line, J774.1/JA-4. Simvastatin and pitavastatin decreased poly(I:C)-induced expression of IFN-ß and ISGs. Moreover, they repressed poly(I:C)-induced phosphorylation of IFN regulatory factor (IRF) 3 and signal transducers and activators of transcription (STAT) 1, which is involved in Janus kinase (JAK)/STAT signaling. Mevalonate and geranylgeranyl pyrophosphate (GGPP), but not cholesterol, counteracted the negative effect of statins on IFN-ß and ISG expression and phosphorylation of IRF3 and STAT1. The geranylgeranyltransferase inhibitor suppressed poly(I:C)-induced expression of IFN-ß and ISGs and phosphorylation of IRF3 and STAT1. These results suggest that statins suppressed the expression of IFN-ß and ISGs in poly(I:C)-treated hyperlipidemic mice and murine macrophages and that these effects occurred through the inhibition of IRF3 and JAK/STAT signaling in macrophages. Furthermore, GGPP recovered the statin-suppressed IRF3 and JAK/STAT signaling in poly(I:C)-treated macrophages.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hiperlipidemias/tratamiento farmacológico , Factores Reguladores del Interferón/metabolismo , Interferón beta/metabolismo , Macrófagos/efectos de los fármacos , Poli I-C/toxicidad , Animales , Hiperlipidemias/inducido químicamente , Hiperlipidemias/metabolismo , Hiperlipidemias/patología , Inductores de Interferón/toxicidad , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Interferón beta/genética , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/genética , Quinasas Janus/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
17.
Minerva Med ; 112(1): 144-152, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33104300

RESUMEN

The world is now entering its 9th month of combat against a pandemic of deadly pneumonia. Started out from China in December 2019, the disease has been declared as caused by infection with a so far unknown RNA Coronavirus of the respiratory family, then named severe acute respiratory syndrome coronavirus SARS-CoV-2. In the absence of a vaccine, and with scientists still struggling for an effective therapy, COVID-19 (the SARS-dependent syndrome) carries up to now, a death toll of more than 590,000 (July 18,2020) undermining jobs and finance of contemporary society in all continents. Social distancing, the only measure hitherto shown to restrain virus spread, has been progressively loosened from May 2020 in some countries, leaving us in the fear of repeat attacks from the unchecked virus. We discuss the problem and propose to tentatively boost the antivirus cell machinery by using lab-made viral mimics to engage cell receptors.


Asunto(s)
COVID-19/terapia , SARS-CoV-2 , COVID-19/complicaciones , COVID-19/epidemiología , Carboximetilcelulosa de Sodio/análogos & derivados , Carboximetilcelulosa de Sodio/uso terapéutico , Síndrome de Liberación de Citoquinas/etiología , Humanos , Inmunización Pasiva , Inductores de Interferón/uso terapéutico , Síndrome Mucocutáneo Linfonodular/etiología , Distanciamiento Físico , Poli I-C/uso terapéutico , Polilisina/análogos & derivados , Polilisina/uso terapéutico , Guías de Práctica Clínica como Asunto , ARN Bicatenario/efectos de los fármacos , ARN Viral/efectos de los fármacos , Recurrencia , SARS-CoV-2/clasificación , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Prevención Secundaria , Tratamiento Farmacológico de COVID-19 , Sueroterapia para COVID-19
18.
Methods Mol Biol ; 2239: 153-162, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33226618

RESUMEN

The discovery of induced pluripotent stem cells (iPSCs) allows for establishment of human embryonic stem-like cells from various adult human somatic cells (e.g., fibroblasts), without the need for destruction of human embryos. This provides an unprecedented opportunity where patient-specific iPSCs can be subsequently differentiated to many cell types, e.g., cardiac cells and neurons, so that we can use these iPSC-derived cells to study patient-specific disease mechanisms and conduct drug testing and screening. Critically, these cells have unlimited therapeutic potentials, and there are many ongoing clinical trials to investigate the regenerative potentials of these iPSC-derivatives in humans. However, the traditional iPSC reprogramming methods have problem of insertional mutagenesis because of use of the integrating viral vectors. While a number of advances have been made to mitigate this issue, including the use of chemicals, excisable and non-integrating vectors, and use of the modified mRNA, safety remains a concern. Both integrating and non-integrating methods also suffer from many other limitations including low efficiency, variability, and tumorigenicity. The non-integrating mRNA reprogramming is of high efficiency, but it is sensitive to reagents and need approaches to reduce the immunogenic reaction. An alternative non-integrating and safer way of generating iPSCs is via direct delivery of recombinant cell-penetrating reprogramming proteins into the cells to be reprogrammed, but reprogramming efficiency of the protein-based approach is extremely low compared to the conventional virus-based nuclear reprogramming. Herein, we describe detailed steps for efficient generation of human iPSCs by protein-based reprogramming in combination with stimulation of the Toll-like receptor 3 (TLR3) innate immune pathway.


Asunto(s)
Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Poli I-C/farmacología , Proteínas Recombinantes/metabolismo , Receptor Toll-Like 3/metabolismo , Técnicas de Cultivo de Célula/métodos , Permeabilidad de la Membrana Celular , Células Cultivadas , Criopreservación , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Inductores de Interferón/farmacología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Péptidos/química , Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Recombinantes/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal/genética , Receptor Toll-Like 3/genética
19.
Bioorg Med Chem Lett ; 30(24): 127613, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33075488

RESUMEN

Type I Interferon (IFN) signaling plays an important role in the immune defense system against virus infection and in the innate immune response, thus IFNs are widely used as anti-viral agents and treatment for immune disorder or cancer. However, there is a growing demand for novel small-molecule IFN inducer due to tolerance, toxicity, or short duration of action following direct administration of IFNs. In this study, we assessed arylpiperazine (ARP) as a new core skeleton of IFN inducer. To investigate structure-activity relationship, we designed and synthesized a series of ARP analogues and evaluated the ability to stimulate IFN response in THP-1 human monocyte cells. Compound 5i was identified as a potent type I IFN inducer as it significantly increased cytokine secretion and increased expression of various IFN-stimulating genes which are representative biomarkers of type I IFN pathway. Our results suggested a beneficial therapeutic potential of 5i as an anti-viral agent.


Asunto(s)
Inductores de Interferón/química , Inductores de Interferón/farmacología , Monocitos/efectos de los fármacos , Piperazinas/química , Piperazinas/farmacología , Diseño de Fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Inductores de Interferón/síntesis química , Interferón Tipo I/agonistas , Interferón Tipo I/inmunología , Monocitos/inmunología , Piperazinas/síntesis química , Células THP-1
20.
Int J Med Sci ; 17(12): 1803-1810, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32714083

RESUMEN

Since the end of 2019, a new type of coronavirus pneumonia (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has been spreading rapidly throughout the world. Previously, there were two outbreaks of severe coronavirus caused by different coronaviruses worldwide, namely Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and the Middle East Respiratory Syndrome Coronavirus (MERS-CoV). This article introduced the origin, virological characteristics and epidemiological overview of SARS-CoV-2, reviewed the currently known drugs that may prevent and treat coronavirus, explained the characteristics of the new coronavirus and provided novel information for the prevention and treatment of COVID-19.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/prevención & control , Amidas/farmacología , Amidas/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Betacoronavirus/aislamiento & purificación , Betacoronavirus/fisiología , COVID-19 , Cloroquina/análogos & derivados , Cloroquina/uso terapéutico , Clorpromazina/uso terapéutico , Coronavirus/genética , Infecciones por Coronavirus/genética , Ciclofilinas/antagonistas & inhibidores , Desarrollo de Medicamentos , Reposicionamiento de Medicamentos , Medicamentos Herbarios Chinos/uso terapéutico , Endocitosis/efectos de los fármacos , Humanos , Sueros Inmunes , Inductores de Interferón/uso terapéutico , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , Neumonía Viral/genética , Pirazinas/farmacología , Pirazinas/uso terapéutico , Resveratrol/farmacología , Resveratrol/uso terapéutico , SARS-CoV-2 , Vacunas Virales/uso terapéutico , Tratamiento Farmacológico de COVID-19
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...