Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nat Commun ; 14(1): 8332, 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38097561

RESUMO

Microbiota have an important function in shaping and priming neonatal immunity, although the cellular and molecular mechanisms underlying these effects remain obscure. Here we report that prenatal antibiotic exposure causes significant elevation of group 2 innate lymphoid cells (ILC2s) in neonatal lungs, in both cell numbers and functionality. Downregulation of type 1 interferon signaling in ILC2s due to diminished production of microbiota-derived butyrate represents the underlying mechanism. Mice lacking butyrate receptor GPR41 (Gpr41-/-) or type 1 interferon receptor IFNAR1 (Ifnar1-/-) recapitulate the phenotype of neonatal ILC2s upon maternal antibiotic exposure. Furthermore, prenatal antibiotic exposure induces epigenetic changes in ILC2s and has a long-lasting deteriorative effect on allergic airway inflammation in adult offspring. Prenatal supplementation of butyrate ameliorates airway inflammation in adult mice born to antibiotic-exposed dams. These observations demonstrate an essential role for the microbiota in the control of type 2 innate immunity at the neonatal stage, which suggests a therapeutic window for treating asthma in early life.


Assuntos
Antibacterianos , Imunidade Inata , Interferon Tipo I , Linfócitos , Animais , Camundongos , Butiratos , Citocinas , Regulação para Baixo , Inflamação , Pulmão , Antibacterianos/efeitos adversos , Antibacterianos/farmacologia , Exposição Materna , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/metabolismo
2.
Gastroenterology ; 162(2): 590-603.e14, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34627860

RESUMO

BACKGROUND AND AIMS: Patients with pancreatic ductal adenocarcinoma (PDA) have not yet benefitted from the revolution in cancer immunotherapy due in large part to a dominantly immunosuppressive tumor microenvironment. MEK inhibition combined with autophagy inhibition leads to transient tumor responses in some patients with PDA. We examined the functional effects of combined MEK and autophagy inhibition on the PDA immune microenvironment and the synergy of combined inhibition of MEK and autophagy with CD40 agonism (aCD40) against PDA using immunocompetent model systems. METHODS: We implanted immunologically "cold" murine PDA cells orthotopically in wide type C57BL/6J mice. We administered combinations of inhibitors of MEK1/2, inhibitors of autophagy, and aCD40 and measured anticancer efficacy and immune sequelae using mass cytometry and multiplexed immunofluorescence imaging analysis to characterize the tumor microenvironment. We also used human and mouse PDA cell lines and human macrophages in vitro to perform functional assays to elucidate the cellular effects induced by the treatments. RESULTS: We find that coinhibition of MEK (using cobimetinib) and autophagy (using mefloquine), but not either treatment alone, activates the STING/type I interferon pathway in tumor cells that in turn activates paracrine tumor associated macrophages toward an immunogenic M1-like phenotype. This switch is further augmented by aCD40. Triple therapy (cobimetinib + mefloquine + aCD40) achieved cytotoxic T-cell activation in an immunologically "cold" mouse PDA model, leading to enhanced antitumor immunity. CONCLUSIONS: MEK and autophagy coinhibition coupled with aCD40 invokes immune repolarization and is an attractive therapeutic approach for PDA immunotherapy development.


Assuntos
Autofagia/imunologia , Azetidinas/farmacologia , Antígenos CD40/agonistas , Carcinoma Ductal Pancreático/imunologia , Mefloquina/farmacologia , Neoplasias Pancreáticas/imunologia , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/imunologia , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Humanos , Hidroxicloroquina/farmacologia , Imunoterapia , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/imunologia , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Macrófagos , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/imunologia , Camundongos , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/imunologia , Evasão Tumoral , Microambiente Tumoral/efeitos dos fármacos , Macrófagos Associados a Tumor/efeitos dos fármacos
4.
Clin Cancer Res ; 27(15): 4353-4366, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34011558

RESUMO

PURPOSE: Combining radiotherapy (RT) with DNA damage response inhibitors may lead to increased tumor cell death through radiosensitization. DNA-dependent protein kinase (DNA-PK) plays an important role in DNA double-strand break repair via the nonhomologous end joining (NHEJ) pathway. We hypothesized that in addition to a radiosensitizing effect from the combination of RT with AZD7648, a potent and specific inhibitor of DNA-PK, combination therapy may also lead to modulation of an anticancer immune response. EXPERIMENTAL DESIGN: AZD7648 and RT efficacy, as monotherapy and in combination, was investigated in fully immunocompetent mice in MC38, CT26, and B16-F10 models. Immunologic consequences were analyzed by gene expression and flow-cytometric analysis. RESULTS: AZD7648, when delivered in combination with RT, induced complete tumor regressions in a significant proportion of mice. The antitumor efficacy was dependent on the presence of CD8+ T cells but independent of NK cells. Analysis of the tumor microenvironment revealed a reduction in T-cell PD-1 expression, increased NK-cell granzyme B expression, and elevated type I IFN signaling in mice treated with the combination when compared with RT treatment alone. Blocking of the type I IFN receptor in vivo also demonstrated a critical role for type I IFN in tumor growth control following combined therapy. Finally, this combination was able to generate tumor antigen-specific immunologic memory capable of suppressing tumor growth following rechallenge. CONCLUSIONS: Blocking the NHEJ DNA repair pathway with AZD7648 in combination with RT leads to durable immune-mediated tumor control.


Assuntos
Linhagem Celular Tumoral/efeitos da radiação , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Interferon Tipo I/efeitos dos fármacos , Neoplasias/radioterapia , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Piranos/farmacologia , Radiossensibilizantes/farmacologia , Triazóis/farmacologia , Animais , Camundongos
5.
Lupus ; 30(5): 795-806, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33626969

RESUMO

OBJECTIVES: We aimed to identify transcriptional gene signatures predictive of clinical response, for pharmacodynamic evaluation, and to provide mechanistic insight into JNJ-55920839, a human IgG1κ neutralizing mAb targeting IFN-α/IFN-ω, in participants with systemic lupus erythematosus (SLE). METHODS: Blood samples were obtained from SLE participants at baseline and up to Day 130, who received six 10 mg/kg IV doses of JNJ-55920839/placebo every 2 weeks. Participants with mild-to-moderate SLE who achieved clinical responses using SLE Disease Activity Index 2000 Responder Index 4-point change were considered responders. Transcriptional signatures from longitudinally collected blood were generated by RNA-Seq; signatures were generated by microarray from baseline blood samples exposed in vitro to JNJ-55920839 versus untreated. RESULTS: Two gene signatures (IFN-I Signaling and Immunoglobulin Immune Response) exhibited pharmacodynamic changes among JNJ-55920839 responders. The Immunoglobulin signature, but not the IFN-I signature, was elevated at baseline in JNJ-55920839 responders. A gene cluster associated with neutrophil-mediated immunity was reduced at baseline in JNJ-55920839 responders, substantiated by lower neutrophil counts in responders. An IFN-I signature was suppressed by JNJ-55920839 in vitro treatment versus untreated blood to a greater extent in responders before in vivo dosing. CONCLUSIONS: These signatures may enable enrichment for treatment responders when using IFN-I-suppressing treatments in SLE.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Interferon-alfa/antagonistas & inibidores , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Administração Intravenosa , Adulto , Idoso , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/uso terapêutico , Biomarcadores/sangue , Estudos de Casos e Controles , Feminino , Humanos , Imunoglobulinas/genética , Imunoglobulinas/imunologia , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/genética , Interferon-alfa/genética , Interferon-alfa/imunologia , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/genética , Masculino , Pessoa de Meia-Idade , Placebos/administração & dosagem , Índice de Gravidade de Doença , Transcrição Gênica/genética , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Ustekinumab/administração & dosagem , Ustekinumab/farmacologia , Ustekinumab/uso terapêutico
6.
Cell Rep ; 34(1): 108586, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33406424

RESUMO

The cyclic GMP-AMP (cGAMP) synthase (cGAS) is a key DNA sensor that initiates STING-dependent signaling to produce type I interferons through synthesizing the secondary messenger 2'3'-cGAMP. In this study, we confirm previous studies showing that cGAS is located both in the cytoplasm and in the nucleus. Nuclear accumulation is observed when leptomycin B is used to block the exportin, CRM1 protein. As a result, leptomycin B impairs the production of interferons in response to DNA stimulation. We further identify a functional nuclear export signal (NES) in cGAS, 169LEKLKL174. Mutating this NES leads to the sequestration of cGAS within the nucleus and the loss of interferon response to cytosolic DNA treatment, and it further determines the key amino acid to L172. Collectively, our data demonstrate that the cytosolic DNA-sensing function of cGAS depends on its presence within the cytoplasm, which is warranted by a functional NES.


Assuntos
Citosol/metabolismo , DNA/metabolismo , Carioferinas/metabolismo , Sinais de Exportação Nuclear , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Sequência de Aminoácidos , Ácidos Graxos Insaturados/farmacologia , Células HEK293 , Células HeLa , Humanos , Imunidade Inata , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/metabolismo , Interferon beta/metabolismo , Carioferinas/efeitos dos fármacos , Carioferinas/imunologia , Proteínas de Membrana/metabolismo , Mutação , Nucleotidiltransferases/imunologia , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/imunologia , Transdução de Sinais , Células THP-1 , Proteína Exportina 1
7.
Viruses ; 12(10)2020 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-32992478

RESUMO

Influenza A virus is a major global pathogen of humans, and there is an unmet need for effective antivirals. Current antivirals against influenza A virus directly target the virus and are vulnerable to mutational resistance. Harnessing an effective host antiviral response is an attractive alternative. We show that brief exposure to low, non-toxic doses of thapsigargin (TG), an inhibitor of the sarcoplasmic/endoplasmic reticulum (ER) Ca2+ ATPase pump, promptly elicits an extended antiviral state that dramatically blocks influenza A virus production. Crucially, oral administration of TG protected mice against lethal virus infection and reduced virus titres in the lungs of treated mice. TG-induced ER stress unfolded protein response appears as a key driver responsible for activating a spectrum of host antiviral defences that include an enhanced type I/III interferon response. Our findings suggest that TG is potentially a viable host-centric antiviral for the treatment of influenza A virus infection without the inherent problem of drug resistance.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H3N8/crescimento & desenvolvimento , Tapsigargina/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Embrião de Galinha , Chlorocebus aethiops , Cães , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Influenza Humana/tratamento farmacológico , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/imunologia , Interferons/efeitos dos fármacos , Interferons/imunologia , Camundongos , Camundongos Endogâmicos BALB C , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , Suínos , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Células Vero , Interferon lambda
8.
J Virol ; 94(16)2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32461320

RESUMO

Butyrate is an abundant metabolite produced by gut microbiota. While butyrate is a known histone deacetylase inhibitor that activates expression of many genes involved in immune system pathways, its effects on virus infections and on the antiviral type I interferon (IFN) response have not been adequately investigated. We found that butyrate increases cellular infection with viruses relevant to human and animal health, including influenza virus, reovirus, HIV-1, human metapneumovirus, and vesicular stomatitis virus. Mechanistically, butyrate suppresses levels of specific antiviral IFN-stimulated gene (ISG) products, such as RIG-I and IFITM3, in human and mouse cells without inhibiting IFN-induced phosphorylation or nuclear translocation of the STAT1 and STAT2 transcription factors. Accordingly, we discovered that although butyrate globally increases baseline expression of more than 800 cellular genes, it strongly represses IFN-induced expression of 60% of ISGs and upregulates 3% of ISGs. Our findings reveal that there are differences in the IFN responsiveness of major subsets of ISGs depending on the presence of butyrate in the cell environment, and overall, they identify a new mechanism by which butyrate influences virus infection of cells.IMPORTANCE Butyrate is a lipid produced by intestinal bacteria. Here, we newly show that butyrate reprograms the innate antiviral immune response mediated by type I interferons (IFNs). Many of the antiviral genes induced by type I IFNs are repressed in the presence of butyrate, resulting in increased virus infection and replication. Our research demonstrates that metabolites produced by the gut microbiome, such as butyrate, can have complex effects on cellular physiology, including dampening of an inflammatory innate immune pathway resulting in a proviral cellular environment. Our work further suggests that butyrate could be broadly used as a tool to increase growth of virus stocks for research and for the generation of vaccines.


Assuntos
Butiratos/metabolismo , Expressão Gênica/efeitos dos fármacos , Interferon Tipo I/efeitos dos fármacos , Animais , Antivirais/farmacologia , Butiratos/farmacologia , Linhagem Celular , Expressão Gênica/genética , Humanos , Imunidade Inata/efeitos dos fármacos , Interferon Tipo I/metabolismo , Interferons/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Proteínas de Ligação a RNA/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Viroses , Replicação Viral/efeitos dos fármacos
9.
ACS Chem Biol ; 15(7): 1974-1986, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32459468

RESUMO

Immunosuppressants used to treat autoimmunity are often not curative and have many side effects. Our purpose was to identify therapeutics for autoimmunity of the skeletal muscle termed idiopathic inflammatory myopathies (myositis). Recent evidence shows that the pro-inflammatory type I interferons (IFN) and a downstream product major histocompatibility complex (MHC) class I are pathogenic in myositis. We conducted quantitative high-throughput screening on >4500 compounds, including all approved drugs, through a series of cell-based assays to identify those that inhibit the type I IFN-MHC class I pathway in muscle precursor cells (myoblasts). The primary screen utilized CRISPR/Cas9 genome-engineered human myoblasts containing a pro-luminescent reporter HiBit fused to the C-terminus of endogenous MHC class I. Active compounds were counter-screened for cytotoxicity and validated by MHC class I immunofluorescence, Western blot, and RT-qPCR. Actives included Janus kinase inhibitors, with the most potent being ruxolitinib, and epigenetic/transcriptional modulators like histone deacetylase inhibitors and the hypoxia-inducible factor 1 inhibitor echinomycin. Testing in animal models and clinical trials is necessary to translate these therapies to myositis patients. These robust assay technologies can be further utilized to interrogate the basic mechanisms of the type I IFN-MHC class I pathway, identify novel molecular probes, and elucidate possible environmental triggers that may lead to myositis.


Assuntos
Antígenos HLA-B/metabolismo , Fatores Imunológicos/farmacologia , Interferon Tipo I/metabolismo , Mioblastos/efeitos dos fármacos , Linhagem Celular , Antígenos HLA-B/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Humanos , Interferon Tipo I/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT1/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia
10.
Nat Commun ; 10(1): 5108, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31704921

RESUMO

Mounting evidence suggests that the tumor microenvironment is profoundly immunosuppressive. Thus, mitigating tumor immunosuppression is crucial for inducing sustained antitumor immunity. Whereas previous studies involved intratumoral injection, we report here an inhalable nanoparticle-immunotherapy system targeting pulmonary antigen presenting cells (APCs) to enhance anticancer immunity against lung metastases. Inhalation of phosphatidylserine coated liposome loaded with STING agonist cyclic guanosine monophosphate-adenosine monophosphate (NP-cGAMP) in mouse models of lung metastases enables rapid distribution of NP-cGAMP to both lungs and subsequent uptake by APCs without causing immunopathology. NP-cGAMP designed for enhanced cytosolic release of cGAMP stimulates STING signaling and type I interferons production in APCs, resulting in the pro-inflammatory tumor microenvironment in multifocal lung metastases. Furthermore, fractionated radiation delivered to one tumor-bearing lung synergizes with inhaled NP-cGAMP, eliciting systemic anticancer immunity, controlling metastases in both lungs, and conferring long-term survival in mice with lung metastases and with repeated tumor challenge.


Assuntos
Células Apresentadoras de Antígenos/efeitos dos fármacos , Imunoterapia , Neoplasias Pulmonares/secundário , Pulmão/efeitos dos fármacos , Melanoma Experimental/secundário , Proteínas de Membrana/agonistas , Nanopartículas , Nucleotídeos Cíclicos/farmacologia , Radioterapia , Administração por Inalação , Animais , Células Apresentadoras de Antígenos/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/imunologia , Lipossomos , Pulmão/imunologia , Pulmão/efeitos da radiação , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Nucleotídeos Cíclicos/administração & dosagem , Fosfatidilserinas
11.
Arthritis Rheumatol ; 71(1): 50-62, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30009417

RESUMO

OBJECTIVE: The production of class-switched high-affinity autoantibodies derived from organized germinal centers (GCs) is a hallmark of many autoimmune inflammatory diseases, including rheumatoid arthritis (RA). TANK-binding kinase 1 (TBK-1) is a serine/threonine kinase involved in the maturation of GC follicular helper T (Tfh) cells downstream of inducible costimulator signaling. We undertook this study to assess the therapeutic potential of TBK-1 inhibition using the small-molecule inhibitor WEHI-112 in antibody-dependent models of inflammatory arthritis. METHODS: Using the models of collagen-induced arthritis (CIA), antigen-induced arthritis (AIA), and K/BxN serum-transfer-induced arthritis (STIA), we determined the effectiveness of WEHI-112 at inhibiting clinical and histologic features of arthritis in C57BL/6 and DBA/1 mice. We used immunohistochemistry to characterize GC populations during CIA development, and we used enzyme-linked immunosorbent assays to determine levels of Ig autoantibodies in WEHI-112-treated mice compared to vehicle-treated mice. RESULTS: WEHI-112, a tool compound that is semiselective for TBK-1 but that also has activity against IKKε and JAK2, abolished TBK-1-dependent activation of interferon (IFN) regulatory factor 3 and inhibited type I IFN responses in vitro. In vivo, treatment with WEHI-112 selectively abrogated clinical and histologic features of established, antibody-dependent CIA, but had minimal effects on an antibody-independent model of AIA or on K/BxN STIA. In keeping with these findings, WEHI-112 reduced arthritogenic type II collagen-specific IgG1 and IgG2b antibody production. Furthermore, WEHI-112 altered the GC Tfh cell phenotype and GC B cell function in CIA. CONCLUSION: We report that TBK-1 inhibition using WEHI-112 abrogated antibody-dependent CIA. As WEHI-112 failed to inhibit non-antibody-driven joint inflammation, we conclude that the major effect of this compound was most likely the targeting of TBK-1-mediated mechanisms in the GC reaction. This approach may have therapeutic potential in RA and in other GC-associated autoantibody-driven inflammatory diseases.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Autoanticorpos/efeitos dos fármacos , Centro Germinativo/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Animais , Autoanticorpos/imunologia , Azetidinas/farmacologia , Colágeno Tipo II , Ciclobutanos/farmacologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Adjuvante de Freund , Centro Germinativo/imunologia , Imuno-Histoquímica , Fatores Imunológicos , Técnicas In Vitro , Fator Regulador 3 de Interferon/efeitos dos fármacos , Fator Regulador 3 de Interferon/imunologia , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/imunologia , Inibidores de Janus Quinases/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Morfolinas/farmacologia , Purinas , Pirazóis , Soroalbumina Bovina , Sulfonamidas/farmacologia , Linfócitos T Auxiliares-Indutores/imunologia
12.
Arterioscler Thromb Vasc Biol ; 38(9): 2148-2159, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30026273

RESUMO

Objective- Calcific aortic valve disease is the most prevalent valvulopathy in Western countries. An unanticipated pathogenetic clue involving IFN (interferon) was disclosed by the finding of constitutive type I IFN activity associated with aortic valve calcification in children with the atypical Singleton-Merten syndrome. On this basis, the role of type I IFN on inflammation and calcification in human aortic valve interstitial cells (AVIC) was examined. Approach and Results- IFN-α was weakly proinflammatory but potentiated lipopolysaccharide-mediated activation of NF (nuclear factor)-κB and the ensuing induction of proinflammatory molecules in human AVIC. Stimulation with IFN-α and in combination with lipopolysaccharide promoted osteoblast-like differentiation characterized by increased osteoblastic gene expression, BMP (bone morphogenetic protein)-2 secretion, and ectopic phosphatase activity. Sex differences were observed. Likewise, IFN-α treatment of human AVICs in osteogenic medium resulted in increased formation of calcific nodules. Strikingly, IFN-α-mediated calcification was significantly higher in AVICs from males, and was blocked by tofacitinib, a JAK (Janus kinase) inhibitor, and by a BMP antagonist. A female-specific protective mechanism involving the activation of PI3K-Akt (protein kinase B) pathways and cell survival was disclosed. Females exhibited higher levels of BCL2 in valve cells and tissues and lower annexin V staining on cell stimulation. Conclusions- IFN-α acts as a proinflammatory and pro-osteogenic cytokine in AVICs, its effects being potentiated by lipopolysaccharide. Results also uncovered sex differences with lower responses in female AVICs and sex-specific mechanisms involving apoptosis. Data point to JAK/STAT (signal transducer and activator of transcription) system as a potential therapeutic target for calcific aortic valve disease.


Assuntos
Estenose da Valva Aórtica/metabolismo , Valva Aórtica/patologia , Calcinose/metabolismo , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/metabolismo , Inibidores de Janus Quinases/farmacologia , Valva Aórtica/citologia , Valva Aórtica/metabolismo , Estenose da Valva Aórtica/patologia , Apoptose , Proteína Morfogenética Óssea 2/antagonistas & inibidores , Proteína Morfogenética Óssea 2/metabolismo , Calcinose/patologia , Diferenciação Celular , Células Cultivadas , Citocinas/metabolismo , Feminino , Humanos , Lipopolissacarídeos/farmacologia , Masculino , NF-kappa B/metabolismo , Osteoblastos/fisiologia , Piperidinas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinas/farmacologia , Pirróis/farmacologia , Fatores de Transcrição STAT/metabolismo , Fatores Sexuais , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
13.
Artigo em Inglês | MEDLINE | ID: mdl-29124042

RESUMO

E. fischeriana has long been used as a traditional Chinese medicine. Recent studies reported that some compounds of E. fischeriana exhibited antimicrobial and immune enhance activity. Innate immune system is essential for the immune surveillance of inner and outer threats, initial host defense responses and immune modulation. The role of natural drug compounds, including E. fischeriana, in innate immune regulation is largely unknown. Here we demonstrated that E. fischeriana compound Dpo is involved in antiviral signaling. The genome wide RNA-seq analysis revealed that the induction of ISGs by viral infection could be synergized by Dpo. Consistently, Dpo enhanced the antiviral immune responses and protected the mice from death during viral infection. Dpo however was not able to rescue STING deficient mice lethality caused by HSV-1 infection. The enhancement of ISG15 by Dpo was also impaired in STING, IRF3, IRF7, or ELF4 deficient cells, demonstrating that Dpo activates innate immune responses in a STING/IRFs/ELF4 dependent way. The STING/IRFs/ELF4 axis is therefore important for Dpo induced ISGs expression, and can be used by host to counteract infection.


Assuntos
Antivirais/farmacologia , Euphorbia/química , Imunidade Inata , Extratos Vegetais/farmacologia , Raízes de Plantas/química , Animais , Citocinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Herpes Simples/tratamento farmacológico , Herpesvirus Humano 1/efeitos dos fármacos , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Medicina Tradicional Chinesa , Proteínas de Membrana/metabolismo , Camundongos , Extratos Vegetais/química , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Ubiquitinas/metabolismo , Carga Viral
14.
Arthritis Rheumatol ; 69(11): 2124-2135, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28777892

RESUMO

OBJECTIVE: The role of plasmacytoid dendritic cells (PDCs) and type I interferons (IFNs) in rheumatoid arthritis (RA) remains a subject of controversy. This study was undertaken to explore the contribution of PDCs and type I IFNs to RA pathogenesis using various animal models of PDC depletion and to monitor the effect of localized PDC recruitment and activation on joint inflammation and bone damage. METHODS: Mice with K/BxN serum-induced arthritis, collagen-induced arthritis, and human tumor necrosis factor transgene insertion were studied. Symptoms were evaluated by visual scoring, quantification of paw swelling, determination of cytokine levels by enzyme-linked immunosorbent assay, and histologic analysis. Imiquimod-dependent therapeutic effects were monitored by transcriptome analysis (using quantitative reverse transcriptase-polymerase chain reaction) and flow cytometric analysis of the periarticular tissue. RESULTS: PDC-deficient mice showed exacerbation of inflammatory and arthritis symptoms after arthritogenic serum transfer. In contrast, enhancing PDC recruitment and activation to arthritic joints by topical application of the Toll-like receptor 7 (TLR-7) agonist imiquimod significantly ameliorated arthritis in various mouse models. Imiquimod induced an IFN signature and led to reduced infiltration of inflammatory cells. CONCLUSION: The therapeutic effects of imiquimod on joint inflammation and bone destruction are dependent on TLR-7 sensing by PDCs and type I IFN signaling. Our findings indicate that local recruitment and activation of PDCs represents an attractive therapeutic opportunity for RA patients.


Assuntos
Adjuvantes Imunológicos/farmacologia , Aminoquinolinas/farmacologia , Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Células Dendríticas/efeitos dos fármacos , Interferon Tipo I/efeitos dos fármacos , Animais , Artrite Experimental/genética , Artrite Reumatoide/genética , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Fator de Transcrição Ikaros/genética , Imiquimode , Interferon Tipo I/imunologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 7 Toll-Like/genética , Fator de Necrose Tumoral alfa/genética
16.
mBio ; 8(3)2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28465426

RESUMO

The ongoing concurrent outbreaks of Zika, Chikungunya, and dengue viruses in Latin America and the Caribbean highlight the need for development of broad-spectrum antiviral treatments. The type I interferon (IFN) system has evolved in vertebrates to generate tissue responses that actively block replication of multiple known and potentially zoonotic viruses. As such, its control and activation through pharmacological agents may represent a novel therapeutic strategy for simultaneously impairing growth of multiple virus types and rendering host populations resistant to virus spread. In light of this strategy's potential, we undertook a screen to identify novel interferon-activating small molecules. Here, we describe 1-(2-fluorophenyl)-2-(5-isopropyl-1,3,4-thiadiazol-2-yl)-1,2-dihydrochromeno[2,3-c]pyrrole-3,9-dione, which we termed AV-C. Treatment of human cells with AV-C activates innate and interferon-associated responses that strongly inhibit replication of Zika, Chikungunya, and dengue viruses. By utilizing genome editing, we investigated the host proteins essential to AV-C-induced cellular states. This showed that the compound requires a TRIF-dependent signaling cascade that culminates in IFN regulatory factor 3 (IRF3)-dependent expression and secretion of type I interferon to elicit antiviral responses. The other canonical IRF3-terminal adaptor proteins STING and IPS-1/MAVS were dispensable for AV-C-induced phenotypes. However, our work revealed an important inhibitory role for IPS-1/MAVS, but not TRIF, in flavivirus replication, implying that TRIF-directed viral evasion may not occur. Additionally, we show that in response to AV-C, primary human peripheral blood mononuclear cells secrete proinflammatory cytokines that are linked with establishment of adaptive immunity to viral pathogens. Ultimately, synthetic innate immune activators such as AV-C may serve multiple therapeutic purposes, including direct antimicrobial responses and facilitation of pathogen-directed adaptive immunity.IMPORTANCE The type I interferon system is part of the innate immune response that has evolved in vertebrates as a first line of broad-spectrum immunological defense against an unknowable diversity of microbial, especially viral, pathogens. Here, we characterize a novel small molecule that artificially activates this response and in so doing generates a cellular state antagonistic to growth of currently emerging viruses: Zika virus, Chikungunya virus, and dengue virus. We also show that this molecule is capable of eliciting cellular responses that are predictive of establishment of adaptive immunity. As such, this agent may represent a powerful and multipronged therapeutic tool to combat emerging and other viral diseases.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/agonistas , Antivirais/farmacologia , Benzopiranos/farmacologia , Vírus Chikungunya/fisiologia , Vírus da Dengue/fisiologia , Tiadiazóis/farmacologia , Replicação Viral , Zika virus/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Antivirais/química , Antivirais/isolamento & purificação , Benzopiranos/química , Benzopiranos/isolamento & purificação , Linhagem Celular , Febre de Chikungunya/tratamento farmacológico , Vírus Chikungunya/efeitos dos fármacos , Citocinas/biossíntese , Replicação do DNA/efeitos dos fármacos , Dengue/tratamento farmacológico , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/metabolismo , Descoberta de Drogas , Edição de Genes , Interações Hospedeiro-Patógeno , Humanos , Evasão da Resposta Imune , Imunidade Inata/efeitos dos fármacos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Tiadiazóis/química , Tiadiazóis/isolamento & purificação , Zika virus/efeitos dos fármacos
17.
Nat Med ; 23(6): 763-767, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28394328

RESUMO

Zika virus (ZIKV) infection of pregnant women can cause a wide range of congenital abnormalities, including microcephaly, in the infant, a condition now collectively known as congenital ZIKV syndrome. A vaccine to prevent or significantly attenuate viremia in pregnant women who are residents of or travelers to epidemic or endemic regions is needed to avert congenital ZIKV syndrome, and might also help to suppress epidemic transmission. Here we report on a live-attenuated vaccine candidate that contains a 10-nucleotide deletion in the 3' untranslated region of the ZIKV genome (10-del ZIKV). The 10-del ZIKV is highly attenuated, immunogenic, and protective in type 1 interferon receptor-deficient A129 mice. Crucially, a single dose of 10-del ZIKV induced sterilizing immunity with a saturated neutralizing antibody titer, which no longer increased after challenge with an epidemic ZIKV, and completely prevented viremia. The immunized mice also developed a robust T cell response. Intracranial inoculation of 1-d-old immunocompetent CD-1 mice with 1 × 104 infectious focus units (IFU) of 10-del ZIKV caused no mortality, whereas infections with 10 IFU of wild-type ZIKV were lethal. Mechanistically, the attenuated virulence of 10-del ZIKV may be due to decreased viral RNA synthesis and increased sensitivity to type-1-interferon inhibition. The attenuated 10-del ZIKV was incapable of infecting mosquitoes after oral feeding of spiked-blood meals, representing an additional safety feature. Collectively, the safety and efficacy results suggest that further development of this promising, live-attenuated ZIKV vaccine candidate is warranted.


Assuntos
Anticorpos Neutralizantes/efeitos dos fármacos , Imunogenicidade da Vacina , RNA Viral/efeitos dos fármacos , Vacinas Atenuadas/farmacologia , Replicação Viral/efeitos dos fármacos , Infecção por Zika virus/prevenção & controle , Zika virus/efeitos dos fármacos , Regiões 3' não Traduzidas/genética , Aedes , Animais , Anticorpos Neutralizantes/imunologia , Chlorocebus aethiops , Imunofluorescência , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/imunologia , Camundongos , Camundongos Knockout , Mosquitos Vetores/virologia , Mutação , RNA Viral/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Interferon/genética , Células Vero , Zika virus/genética , Zika virus/imunologia
18.
Rheumatology (Oxford) ; 56(10): 1662-1675, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28122959

RESUMO

Type I interferons (IFN-Is) are a group of molecules with pleiotropic effects on the immune system forming a crucial link between innate and adaptive immune responses. Apart from their important role in antiviral immunity, IFN-Is are increasingly recognized as key players in autoimmune CTDs such as SLE. Novel therapies that target IFN-I appear effective in SLE in early trials, but effectiveness is related to the presence of IFN-I biomarkers. IFN-I biomarkers may also act as positive or negative predictors of response to other biologics. Despite the high failure rate of clinical trials in SLE, subgroups of patients often respond better. Fully optimizing the potential of these agents is therefore likely to require stratification of patients using IFN-I and other biomarkers. This suggests the unified concept of type I IFN-mediated autoimmune diseases as a grouping including patients with a variety of different traditional diagnoses.


Assuntos
Doenças Autoimunes/imunologia , Interferon Tipo I/imunologia , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/tratamento farmacológico , Produtos Biológicos/imunologia , Biomarcadores/análise , Humanos , Interferon Tipo I/efeitos dos fármacos , Lúpus Eritematoso Sistêmico/diagnóstico , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/imunologia
19.
Nat Med ; 22(12): 1411-1420, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27841872

RESUMO

The cellular inhibitors of apoptosis (cIAP) 1 and 2 are amplified in about 3% of cancers and have been identified in multiple malignancies as being potential therapeutic targets as a result of their role in the evasion of apoptosis. Consequently, small-molecule IAP antagonists, such as LCL161, have entered clinical trials for their ability to induce tumor necrosis factor (TNF)-mediated apoptosis of cancer cells. However, cIAP1 and cIAP2 are recurrently homozygously deleted in multiple myeloma (MM), resulting in constitutive activation of the noncanonical nuclear factor (NF)-κB pathway. To our surprise, we observed robust in vivo anti-myeloma activity of LCL161 in a transgenic myeloma mouse model and in patients with relapsed-refractory MM, where the addition of cyclophosphamide resulted in a median progression-free-survival of 10 months. This effect was not a result of direct induction of tumor cell death, but rather of upregulation of tumor-cell-autonomous type I interferon (IFN) signaling and a strong inflammatory response that resulted in the activation of macrophages and dendritic cells, leading to phagocytosis of tumor cells. Treatment of a MM mouse model with LCL161 established long-term anti-tumor protection and induced regression in a fraction of the mice. Notably, combination of LCL161 with the immune-checkpoint inhibitor anti-PD1 was curative in all of the treated mice.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Tiazóis/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Ciclofosfamida/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Mieloma Múltiplo/imunologia , Recidiva Local de Neoplasia/imunologia , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Tiazóis/farmacologia
20.
Arthritis Rheumatol ; 68(11): 2686-2696, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27338297

RESUMO

OBJECTIVE: Increased expression of type I interferon (IFN) and a broad signature of type I IFN-induced gene transcripts are observed in patients with systemic lupus erythematosus (SLE) and other systemic autoimmune diseases. To identify disease-relevant triggers of the type I IFN pathway, this study sought to investigate whether endogenous virus-like genomic repeat elements, normally silent, are expressed in patients with systemic autoimmune disease, and whether these retroelements could activate an innate immune response and induce type I IFN. METHODS: Expression of type I IFN and long interspersed nuclear element 1 (LINE-1; L1) was studied by polymerase chain reaction, Western blotting, and immunohistochemistry in samples of kidney tissue from patients with lupus nephritis and minor salivary gland (MSG) tissue from patients with primary Sjögren's syndrome (SS). Induction of type I IFN by L1 was investigated by transfection of plasmacytoid dendritic cells (PDCs) or monocytes with an L1-encoding plasmid or L1 RNA. Involvement of innate immune pathways and altered L1 methylation were assessed. RESULTS: Levels of L1 messenger RNA transcripts were increased in lupus nephritis kidneys and in MSG tissue from patients with SS. Transcript expression correlated with the expression of type I IFN and L1 DNA demethylation. L1 open-reading frame 1/p40 protein and IFNß were expressed in MSG ductal epithelial cells and in lupus nephritis kidneys, and IFNα was detected in infiltrating PDCs. Transfection of PDCs or monocytes with L1-encoding DNA or RNA induced type I IFN. Inhibition of Toll-like receptor 7 (TLR-7)/TLR-8 reduced the induction of IFNα by L1 in PDCs, and an inhibitor of IKKε/TANK-binding kinase 1 abrogated the induction of type I IFN by L1 RNA in monocytes. CONCLUSION: L1 genomic repeat elements represent endogenous nucleic acid triggers of the type I IFN pathway in SLE and SS and may contribute to initiation or amplification of autoimmune disease.


Assuntos
Interferon Tipo I/imunologia , Elementos Nucleotídeos Longos e Dispersos/genética , Lúpus Eritematoso Sistêmico/genética , Síndrome de Sjogren/genética , Adulto , Idoso , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Western Blotting , Metilação de DNA , Células Dendríticas/imunologia , Feminino , Humanos , Quinase I-kappa B/antagonistas & inibidores , Imunidade Inata/imunologia , Imuno-Histoquímica , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/metabolismo , Interferon-alfa/efeitos dos fármacos , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Interferon beta/efeitos dos fármacos , Interferon beta/imunologia , Interferon beta/metabolismo , Rim/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Elementos Nucleotídeos Longos e Dispersos/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Nefrite Lúpica/imunologia , Nefrite Lúpica/metabolismo , Masculino , Pessoa de Meia-Idade , Monócitos/imunologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Glândulas Salivares Menores/metabolismo , Síndrome de Sjogren/imunologia , Síndrome de Sjogren/metabolismo , Receptor 7 Toll-Like/antagonistas & inibidores , Receptor 8 Toll-Like/antagonistas & inibidores , Receptor Toll-Like 9/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...