Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Immunol ; 20(11): 1339-1351, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37737309

RESUMO

Inhibitory immune receptors set thresholds for immune cell activation, and their deficiency predisposes a person to autoimmune responses. However, the agonists of inhibitory immune receptors remain largely unknown, representing untapped sources of treatments for autoimmune diseases. Here, we show that V-set and transmembrane domain-containing 1 (VSTM1) is an inhibitory receptor and that its binding by the competent ligand soluble galectin-1 (Gal1) is essential for maintaining neutrophil viability mediated by downregulated reactive oxygen species production. However, in patients with systemic lupus erythematosus (SLE), circulating Gal1 is oxidized and cannot be recognized by VSTM1, leading to increased intracellular reactive oxygen species levels and reduced neutrophil viability. Dysregulated neutrophil function or death contributes significantly to the pathogenesis of SLE by providing danger molecules and autoantigens that drive the production of inflammatory cytokines and the activation of autoreactive lymphocytes. Interestingly, serum levels of glutathione, an antioxidant able to convert oxidized Gal1 to its reduced form, were negatively correlated with SLE disease activity. Taken together, our findings reveal failed inhibitory Gal1/VSTM1 pathway activation in patients with SLE and provide important insights for the development of effective targeted therapies.


Assuntos
Lúpus Eritematoso Sistêmico , Neutrófilos , Humanos , Galectina 1 , Espécies Reativas de Oxigênio/metabolismo
2.
Cell Rep ; 42(2): 112097, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36800290

RESUMO

As one of the core components of the switching or sucrose non-fermentable (SWI/SNF) complex, SMARCC1 (BAF155, SRG3) plays essential roles in activation of late inflammatory genes in response to microbial challenge. However, little is known about the mechanism of how SMARCC1 regulates the inflammatory innate response. Via functional screening, we identify the nuclear E3 ubiquitin ligase RNF138 as a negative regulator in the inflammatory innate response and show that RNF138 interacts with SMARCC1 and mediates its K48-linked polyubiquitination at position Lys643 and proteasomal degradation. As a result, the catalytic activity of RNF138 fine-tunes the kinetics of late inflammatory gene transcription by inhibiting chromatin remodeling at SWI/SNF-regulated gene loci. Reduced RNF138 and increased SMARCC1 in monocytes of rheumatoid arthritis patients are observed. These results provide mechanistic insight into the interplay among nucleosome remodeling, inflammation, and ubiquitylation and underscore the important role of the E3 ubiquitin ligases in controlling the extent and duration of inflammatory responses.


Assuntos
Nucleossomos , Ubiquitina-Proteína Ligases , Humanos , Montagem e Desmontagem da Cromatina , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
3.
Sci Signal ; 15(765): eabo4356, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36538592

RESUMO

Histone deacetylases (HDACs) play important roles in immunity and inflammation. Through functional screening, we identified HDAC10 as an inhibitor of the type I interferon (IFN) response mediated by interferon regulatory factor 3 (IRF3). HDAC10 abundance was decreased in mouse macrophages in response to innate immune stimuli and was reduced in peripheral blood mononuclear cells (PBMCs) from patients with systemic lupus erythematosus (SLE) compared with that in PBMCs from healthy donors. Deficiency in HDAC10 in mouse embryonic fibroblasts and in mice promoted the expression of genes encoding type I IFNs and of IFN-stimulated genes (ISGs), leading to enhanced antiviral responses in vitro and in vivo. HDAC10 bound in a deacetylase-independent manner to IRF3 in uninfected cells to inhibit the phosphorylation of IRF3 at Ser396 by TANK-binding kinase 1 (TBK1). Upon viral infection, HDAC10 was targeted for autophagy-mediated degradation through its interaction with LC3-II. Consequently, IRF3 phosphorylation was increased, which resulted in enhanced type I IFN production and antiviral responses. Our findings identify a potential target for improving host defense responses against pathogen infection and for treating autoimmune disease.


Assuntos
Fator Regulador 3 de Interferon , Interferon Tipo I , Animais , Camundongos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Leucócitos Mononucleares/metabolismo , Fibroblastos/metabolismo , Imunidade Inata , Fosforilação , Interferon Tipo I/metabolismo , Antivirais , Autofagia
4.
J Autoimmun ; 130: 102840, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35598407

RESUMO

Cell death is important in the elimination of damaged cells such as virus-infected cells and also is closely involved in the pathogenesis of autoimmune diseases such as systemic lupus erythematosus (SLE). The retinoic acid-inducible gene-I (RIG-I), one cytosolic RNA innate sensor, can trigger antiviral innate response by inducing production of type I interferons (IFN-I). However, the function of RIG-I, once translocated from cytoplasm to nucleus at the late stage of viral infection when IFN-I production is almost terminated, remains poorly understood. Here, we reported that RIG-I is accumulated in the nucleus of macrophages and fibroblasts after virus infection, and nuclear RIG-I is present in peripheral blood mononuclear cells (PBMCs) from SLE patients. We found that nuclear RIG-I interacts with the first 20 amino acids of apurinic/apyrimidinic endodeoxyribonuclease 1 (APEX1) and attenuates the anti-apoptotic properties of APEX1, therefore promoting apoptosis of virus-infected cells to suppress viral infection through an IFN-I-independent way at the late stage of viral infection. Together, our findings reveal a non-canonical role of nuclear RIG-I in the induction of cellular apoptosis, besides its activation of IFN-I expression as the cytosolic innate sensor. This study provides new insight to the regulation of infection, IFN-I and autoimmune diseases by nuclear RIG-I-APEX1 interaction.


Assuntos
Doenças Autoimunes , Leucócitos Mononucleares , Apoptose , Proteína DEAD-box 58/genética , Humanos , Receptores Imunológicos
5.
J Autoimmun ; 129: 102828, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35429914

RESUMO

Heterogeneous nuclear ribonucleoproteins (hnRNPs), a family of RNA-binding proteins, play important roles in various biological processes. However, the roles of hnRNPs members in immunity and inflammation remain to be fully understood. By a functional screening for hnRNPs members in LPS-stimulated macrophage inflammatory response, we identified hnRNP UL1 as a negative regulator of NF-κB-mediated inflammation. hnRNP UL1 constrains NF-κB-triggered transcriptional expression of pro-inflammatory cytokines in response to innate stimuli. Perturbation of hnRNP UL1 enhanced pro-inflammatory cytokine production in macrophages. In vivo deficiency of hnRNP UL1 increased the pro-inflammatory cytokine production once challenged with LPS. Accordingly, the expression of hnRNP UL1 decreased in peripheral blood mononuclear cells of rheumatoid arthritis patients. Mechanistically, hnRNP UL1 competes with NF-κB to bind κB sites to constrain the magnitude and duration of inflammatory response. Meanwhile, the broadly and dynamically binding of hnRNP UL1 on the target genes' promoter during inflammatory response is unraveled. Our study adds new insight into the functions of hnRNPs in NF-κB-mediated inflammation, proposing a potential therapeutic strategy for controlling inflammatory autoimmune diseases.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas , NF-kappa B , Proteínas Nucleares , Fatores de Transcrição , Citocinas/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Inflamação/genética , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/metabolismo
6.
Cell Rep ; 37(5): 109926, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731629

RESUMO

Interferon regulatory factor 3 (IRF3) is an essential transductor for initiation of many immune responses. Here, we show that lncRNA-ISIR directly binds IRF3 to promote its phosphorylation, dimerization, and nuclear translocation, along with enhanced target gene productions. In vivo lncRNA-ISIR deficiency results in reduced IFN production, uncontrolled viral replication, and increased mortality. The human homolog, AK131315, also binds IRF3 and promotes its activation. More important, AK131315 expression is positively correlated with type I interferon (IFN-I) level and severity in patients with lupus. Mechanistically, in resting cells, IRF3 is bound to suppressor protein Flightless-1 (Fli-1), which keeps its inactive state. Upon infection, IFN-I-induced lncRNA-ISIR binds IRF3 at DNA-binding domain in cytoplasm and removes Fli-1's association from IRF3, consequently facilitating IRF3 activation. Our results demonstrate that IFN-I-inducible lncRNA-ISIR feedback strengthens IRF3 activation by removing suppressive Fli-1 in immune responses, revealing a method of lncRNA-mediated modulation of transcription factor (TF) activation.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Macrófagos Peritoneais/metabolismo , RNA Longo não Codificante/metabolismo , Estomatite Vesicular/metabolismo , Animais , Estudos de Casos e Controles , Chlorocebus aethiops , Modelos Animais de Doenças , Inativação Gênica , Células HEK293 , Humanos , Fator Regulador 3 de Interferon/genética , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Proto-Oncogênica c-fli-1/genética , Proteína Proto-Oncogênica c-fli-1/metabolismo , Células RAW 264.7 , RNA Longo não Codificante/genética , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Células Vero , Estomatite Vesicular/genética , Estomatite Vesicular/imunologia , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/imunologia , Vírus da Estomatite Vesicular Indiana/patogenicidade
7.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34607953

RESUMO

Chemokine production by epithelial cells is important for neutrophil recruitment during viral infection, the appropriate regulation of which is critical for restraining inflammation and attenuating subsequent tissue damage. Epithelial cell expression of long noncoding RNAs (lncRNAs), RNA-binding proteins, and their functional interactions during viral infection and inflammation remain to be fully understood. Here, we identified an inducible lncRNA in the Cxcl2 gene locus, lnc-Cxcl2, which could selectively inhibit Cxcl2 expression in mouse lung epithelial cells but not in macrophages. lnc-Cxcl2-deficient mice exhibited increased Cxcl2 expression, enhanced neutrophils recruitment, and more severe inflammation in the lung after influenza virus infection. Mechanistically, nucleus-localized lnc-Cxcl2 bound to Cxcl2 promoter, recruited a ribonucleoprotein La, which inhibited the chromatin accessibility of chemokine promoters, and consequently inhibited Cxcl2 transcription in cis However, unlike mouse lnc-Cxcl2, human lnc-CXCL2-4-1 inhibited multiple immune cytokine expressions including chemokines in human lung epithelial cells. Together, our results demonstrate a self-protecting mechanism within epithelial cells to restrain chemokine and neutrophil-mediated inflammation, providing clues for better understanding chemokine regulation and epithelial cell function in lung viral infection.


Assuntos
Quimiocina CXCL2/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , RNA Longo não Codificante/genética , Células A549 , Animais , Linhagem Celular Tumoral , Quimiocina CXCL2/metabolismo , Cromatina/metabolismo , Células Epiteliais/metabolismo , Células HEK293 , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Humanos , Inflamação/prevenção & controle , Mediadores da Inflamação , Vírus da Influenza A/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Pneumonia/imunologia , Pneumonia/patologia , Pneumonia/virologia , Regiões Promotoras Genéticas/genética , Células RAW 264.7 , Estomatite Vesicular/imunologia , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/imunologia
9.
Phys Rev Lett ; 125(17): 177203, 2020 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-33156649

RESUMO

We show that a conical magnetic field H=(1,1,1)H can be used to tune the topological order and hence, anyon excitations of the Z_{2} quantum spin liquid in the isotropic antiferromagnetic Kitaev model. A novel topological order, featured with Chern number C=4 and Abelian anyon excitations, is induced in a narrow range of intermediate fields H_{c1}≤H≤H_{c2}. On the other hand, the C=1 Ising-topological order with non-Abelian anyon excitations, as previously known to be present at small fields, is found here to survive up to H_{c1}. The results are obtained by developing and applying a Z_{2} mean field theory that works at finite fields and is asymptotically exact in the zero field limit and the associated variational quantum Monte Carlo.

10.
Proc Natl Acad Sci U S A ; 117(38): 23695-23706, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32907941

RESUMO

Long noncoding RNAs (lncRNAs) involved in the regulation of antiviral innate immune responses need to be further identified. By functionally screening the lncRNAs in macrophages, here we identified lncRNA Malat1, abundant in the nucleus but significantly down-regulated after viral infection, as a negative regulator of antiviral type I IFN (IFN-I) production. Malat1 directly bound to the transactive response DNA-binding protein (TDP43) in the nucleus and prevented activation of TDP43 by blocking the activated caspase-3-mediated TDP43 cleavage to TDP35. The cleaved TDP35 increased the nuclear IRF3 protein level by binding and degrading Rbck1 pre-mRNA to prevent IRF3 proteasomal degradation upon viral infection, thus selectively promoting antiviral IFN-I production. Deficiency of Malat1 enhanced antiviral innate responses in vivo, accompanying the increased IFN-I production and reduced viral burden. Importantly, the reduced MALAT1, augmented IRF3, and increased IFNA mRNA were found in peripheral blood mononuclear cells (PBMCs) from systemic lupus erythematosus (SLE) patients. Therefore, the down-regulation of MALAT1 in virus-infected cells or in human cells from autoimmune diseases will increase host resistance against viral infection or lead to autoinflammatory interferonopathies via the increased type I IFN production. Our results demonstrate that the nuclear Malat1 suppresses antiviral innate responses by targeting TDP43 activation via RNA-RBP interactive network, adding insight to the molecular regulation of innate responses and autoimmune pathogenesis.


Assuntos
Proteínas de Ligação a DNA , Imunidade Inata/imunologia , Fator Regulador 3 de Interferon , RNA Longo não Codificante , Adolescente , Adulto , Animais , Antivirais/imunologia , Antivirais/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Feminino , Humanos , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Leucócitos Mononucleares/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , RNA Longo não Codificante/imunologia , RNA Longo não Codificante/metabolismo , Viroses/imunologia , Adulto Jovem
11.
Cell Mol Immunol ; 17(8): 865-874, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31076723

RESUMO

Upon recognition of dsRNA, toll-like receptor 3 (TLR3) recruits the adaptor protein TRIF to activate IRF3 and NF-κB signaling, initiating innate immune responses. The ubiquitination of TLR3 downstream signaling molecules and their roles in the innate response have been discovered; however, whether TLR3 itself is ubiquitinated and then functionally involved remains to be elucidated. By immunoprecipitating TLR3-binding proteins in macrophages, we identified ring finger protein 170 (RNF170) as a TLR3-binding E3 ligase. RNF170 mediated the K48-linked polyubiquitination of K766 in the TIR domain of TLR3 and promoted the degradation of TLR3 through the proteasome pathway. The genetic ablation of RNF170 selectively augmented TLR3-triggered innate immune responses both in vitro and in vivo. Our results reveal a novel role for RNF170 in selectively inhibiting TLR3-triggered innate immune responses by promoting TLR3 degradation.


Assuntos
Imunidade Inata , Proteólise , Receptor 3 Toll-Like/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Feminino , Células HEK293 , Humanos , Lisina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Domínios Proteicos , Células RAW 264.7 , Transdução de Sinais , Ubiquitina-Proteína Ligases/química , Ubiquitinação
12.
Nat Immunol ; 20(7): 812-823, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31036902

RESUMO

The helicase RIG-I initiates an antiviral immune response after recognition of pathogenic RNA. TRIM25, an E3 ubiquitin ligase, mediates K63-linked ubiquitination of RIG-I, which is crucial for RIG-I downstream signaling and the antiviral innate immune response. The components and mode of the RIG-I-initiated innate signaling remain to be fully understood. Here we identify a novel long noncoding RNA (Lnczc3h7a) that binds to TRIM25 and promotes RIG-I-mediated antiviral innate immune responses. Depletion of Lnczc3h7a impairs RIG-I signaling and the antiviral innate response to RNA viruses in vitro and in vivo. Mechanistically, Lnczc3h7a binds to both TRIM25 and activated RIG-I, serving as a molecular scaffold for stabilization of the RIG-I-TRIM25 complex at the early stage of viral infection. Lnczc3h7a facilitates TRIM25-mediated K63-linked ubiquitination of RIG-I and thus promotes downstream signaling transduction. Our findings reveal that host RNAs can enhance the response of innate immune sensors to foreign RNAs, ensuring effective antiviral defense.


Assuntos
Proteína DEAD-box 58/genética , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata/genética , RNA Longo não Codificante/genética , Fatores de Transcrição/genética , Animais , Linhagem Celular , Humanos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Camundongos , Modelos Biológicos , Interferência de RNA , Vírus de RNA/imunologia , Transdução de Sinais , Viroses/genética , Viroses/imunologia , Viroses/metabolismo , Viroses/virologia
13.
Cell ; 173(4): 906-919.e13, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29706547

RESUMO

The innate RNA sensor RIG-I is critical in the initiation of antiviral type I interferons (IFNs) production upon recognition of "non-self" viral RNAs. Here, we identify a host-derived, IFN-inducible long noncoding RNA, lnc-Lsm3b, that can compete with viral RNAs in the binding of RIG-I monomers and feedback inactivate the RIG-I innate function at late stage of innate response. Mechanistically, binding of lnc-Lsm3b restricts RIG-I protein's conformational shift and prevents downstream signaling, thereby terminating type I IFNs production. Multivalent structural motifs and long-stem structure are critical features of lnc-Lsm3b for RIG-I binding and inhibition. These data reveal a non-canonical self-recognition mode in the regulation of immune response and demonstrate an important role of an inducible "self" lncRNA acting as a potent molecular decoy actively saturating RIG-I binding sites to restrict the duration of "non-self" RNA-induced innate immune response and maintaining immune homeostasis, with potential utility in inflammatory disease management.


Assuntos
Proteína DEAD-box 58/metabolismo , Imunidade Inata , RNA Longo não Codificante/metabolismo , Animais , Células HEK293 , Humanos , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Células RAW 264.7 , Interferência de RNA , RNA de Cadeia Dupla/metabolismo , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Vesiculovirus/patogenicidade
14.
Nat Immunol ; 19(1): 41-52, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29242538

RESUMO

Prolonged activation of interferon-STAT1 signaling is closely related to inflammatory autoimmune disorders, and therefore the identification of negative regulators of these pathways is important. Through high-content screening of 115 mouse RING-domain E3 ligases, we identified the E3 ubiquitin ligase RNF2 as a potent inhibitor of interferon-dependent antiviral responses. RNF2 deficiency substantially enhanced interferon-stimulated gene (ISG) expression and antiviral responses. Mechanistically, nuclear RNF2 directly bound to STAT1 after interferon stimulation and increased K33-linked polyubiquitination of the DNA-binding domain of STAT1 at position K379, in addition to promoting the disassociation of STAT1/STAT2 from DNA and consequently suppressing ISG transcription. Our study provides insight into the regulation of interferon-dependent responses via a previously unrecognized post-translational modification of STAT1 in the nucleus.


Assuntos
DNA/metabolismo , Interferon Tipo I/farmacologia , Lisina/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Fator de Transcrição STAT1/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antivirais/farmacologia , Linhagem Celular , Expressão Gênica/efeitos dos fármacos , Lisina/genética , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Complexo Repressor Polycomb 1/genética , Ligação Proteica/efeitos dos fármacos , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT2/genética , Fator de Transcrição STAT2/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/efeitos dos fármacos , Estomatite Vesicular/genética , Estomatite Vesicular/prevenção & controle , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/fisiologia
15.
Nat Commun ; 8: 15818, 2017 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-28598443

RESUMO

Regulatory T (Treg) cells are important for the maintenance of immune homoeostasis and prevention of autoimmune diseases. Epigenetic modifications have been reported to modulate autoimmunity by altering Treg cell fate. Here we show that the H3K4 methyltransferase Ash1l facilitates TGF-ß-induced Treg cell polarization in vitro and protects mice from T cell-mediated colitis in vivo. Ash1l upregulates Smad3 expression by directly targeting Smad3 promoter to increase local H3K4 trimethylation. Furthermore, we identify an lncRNA, namely lnc-Smad3, which interacts with the histone deacetylase HDAC1 and silences Smad3 transcription. After TGF-ß stimulation, activated Smad3 suppresses lnc-Smad3 transcription, thereby recovering the Smad3 promoter accessibility to Ash1l. By revealing the opposite regulatory functions of Ash1l and lnc-Smad3 in Smad3 expression, our data provide insights for the epigenetic control of Treg cell fate to potentially aid in the development of therapeutic intervention for autoimmune diseases.


Assuntos
Autoimunidade , Polaridade Celular , Histona-Lisina N-Metiltransferase/imunologia , RNA Longo não Codificante/genética , Proteína Smad3/genética , Linfócitos T Reguladores/citologia , Linfócitos T/imunologia , Adulto , Idoso , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Epigênese Genética , Regulação da Expressão Gênica , Inativação Gênica , Histona Desacetilase 1/genética , Histona Desacetilase 1/imunologia , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Histonas/imunologia , Humanos , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Regiões Promotoras Genéticas , RNA Longo não Codificante/imunologia , Proteína Smad3/imunologia , Linfócitos T/citologia , Linfócitos T Reguladores/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Fator de Crescimento Transformador beta/imunologia
16.
Mater Sci Eng C Mater Biol Appl ; 75: 182-190, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28415453

RESUMO

Baicalin (BAI) has been reported to exert antitumor effects. However, BAI has limited water solubility, non-specific tumor targeting, and low bioavailability, which severely limited its clinical application. The aim of this study was to develop folic acid (FA) covalently conjugated-polyamidoamine (PAMAM) dendrimers (PAMAM-FA) as carrier systems for improvement of water solubility and tumor-specificity of BAI, and study the role of generation on the physiochemical properties and biological effects of PAMAM-FA/BAI complexes. In this work, four generations of PAMAM-FA were synthesized to entrap BAI. The average sizes of G3-FA/BAI, G4-FA/BAI, G5-FA/BAI, and G6-FA/BAI complexes were 174.4nm, 184.5nm, 258.8nm, and 247.5nm, respectively, and the zeta potentials of four PAMAM-FA/BAI complexes were -2.9mV, -6.6mV, -9.3mV, -9.0mV, respectively. The entrapment efficiencies of four PAMAM-FA/BAI complexes were 91.1%, 53.5%, 80.3%, and 91.9%, respectively, and the drug loading of PAMAM-FA/BAI complexes were about 22%. The formed PAMAM-FA/BAI complexes allowed sustained release of BAI in acidic PBS (pH5.4). In cellular uptake assay, PAMAM-FA/BAI complexes demonstrated increased drug uptake level in folate receptor (FR)-positive Hela cancer cells than FR-negative A549 cells, and the cellular uptake efficiency of PAMAM-FA is closely related with the generation of PAMAM. The MTT assay results showed that PAMAM-FA/BAI complexes demonstrated enhanced toxicity against Hela cells than non-FA-modified PAMAM/BAI complexes, and the G6-FA/BAI demonstrated the best inhibition efficiency. The cell cycle and cell apoptosis analysis further demonstrated the tumor-specific therapeutic efficacy of PAMAM-FA/BAI. These results suggested that the PAMAM-FA have the potential for targeted delivery of BAI into cancer cells to enhance its anti-tumor efficacy.


Assuntos
Dendrímeros , Sistemas de Liberação de Medicamentos/métodos , Flavonoides , Ácido Fólico , Neoplasias/tratamento farmacológico , Poliaminas , Células A549 , Dendrímeros/química , Dendrímeros/farmacocinética , Dendrímeros/farmacologia , Flavonoides/química , Flavonoides/farmacocinética , Flavonoides/farmacologia , Ácido Fólico/química , Ácido Fólico/farmacocinética , Ácido Fólico/farmacologia , Células HeLa , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Poliaminas/química , Poliaminas/farmacocinética , Poliaminas/farmacologia
17.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 33(6): 531-534, 2017 Jun 08.
Artigo em Chinês | MEDLINE | ID: mdl-29931903

RESUMO

OBJECTIVE: To study the role and mechanism of myocardial apoptosis after short-term and long-term exercise preconditioning. METHODS: Forty-eight male SD rats were randomly divided into control group (C), exhaust group (E), short-exercise preconditioning (S-EP) and long-term exercise preconditioning group (L-EP). Short-term and long-term exercise preconditioning were conducted for 3 days and 3 weeks of repeated intermittent swimming training program. The changes of myocardial cells were observed under light microscope. The serum levels of ischemia-modified albumin(IMA) and creatine kinase-isoenzyme(CK-MB) were detected by ELISA. Real time fluorescence quantitative PCR and Western blot were used to detect the expressions of tumor necrosis factor-α(TNF-α),Caspase-8, Caspase-3 genes and proteins in myocardial tissue. The apoptosis of cardiomyocytes was observed by TUNEL method. RESULTS: Compared with group C, group E had serious myocardial injury. The levels of serum IMA, CK-MB and the expressions of TNF-α, Caspase-8 and Caspase-3 in myocardium were increased (P<0.05). Compared with group E, serum CK-MB and TNF-α and Caspase-8 mRNA in S-EP group were significantly lower than those in group E (P<0.05), but there was no significant difference in serum IMA and Caspase-3 mRNA and protein (P>0.05). The levels of serum IMA, CK-MB and TNF-α, Caspase-8 and Caspase-3 mRNA in L-EP group were significantly lower than those in control group (P<0.05). The apoptosis of cardiomyocytes in group E was obvious. Short-term and long-term exercise preconditioning could inhibit apoptosis. Compared with S-EP group, the apoptosis of L-EP group was significantly decreased. CONCLUSIONS: Short-term and long-term exercise preconditioning can reduce myocardial injury after exhaustive exercise, but short-term exercise preconditioning does not alter the expression of Caspase protease. Long-term exercise preconditioning significantly inhibits Caspase-8, 3 mRNA expression and reduces protein synthesis. The inhibitive effects of long-term exercise preconditioning on myocardial cell apoptosis were stronger than those of short-term exercise preconditioning.


Assuntos
Traumatismo por Reperfusão Miocárdica/terapia , Miocárdio/patologia , Miócitos Cardíacos/citologia , Condicionamento Físico Animal , Animais , Apoptose , Biomarcadores , Caspase 3/metabolismo , Caspase 8/metabolismo , Creatina Quinase Forma MB/sangue , Masculino , Ratos , Ratos Sprague-Dawley , Albumina Sérica/análise , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
18.
Mol Immunol ; 80: 11-16, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27788393

RESUMO

Macrophages are innate immune cells that are important contributors to age-related functional impairment of the immune system. During the cell aging process, microRNAs are differentially expressed and participate in the regulation of aging-related immune responses. However, the role of aging-associated changes in miRNA expression in macrophages remains unclear. Here, we found that miR-142-3p expression is downregulated 50% in peritoneal macrophages from aged mice compared with young mice and is not upregulated by cell treatment with lipopolysaccharide (LPS), CpG, or polyinosinic-polycytidylic acid. Serum levels of miR-142-3p are also lower in aged mice than in young mice by q-PCR. Luciferase reporter analysis showed that IL-6 is a target of miR-142-3p in macrophages. In addition, the histone deacetylase inhibitor trichostatin A increased miR-142-3p expression by more than 3-fold in LPS-treated macrophages from aged mice compared with young mice, which in turn suppressed LPS-stimulated IL-6 production, suggesting that inhibition of miR-142-3p by histone deacetylation may be involved in the lack of response to LPS stimulation in macrophages of aged mice. These findings suggest that downregulation of miR-142-3p in macrophages of aged mice might contribute to IL-6-associated aging disorders and that epigenetic modification might be involved in age-related inflammatory diseases.


Assuntos
Envelhecimento/genética , Envelhecimento/imunologia , Interleucina-6/biossíntese , Macrófagos/imunologia , MicroRNAs/biossíntese , Animais , Regulação para Baixo , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Interleucina-6/imunologia , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/imunologia , Poli I-C/imunologia , Reação em Cadeia da Polimerase em Tempo Real
19.
Proc Natl Acad Sci U S A ; 113(34): 9581-6, 2016 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-27506794

RESUMO

The activation of retinoic acid-inducible gene 1 (RIG-I), a cytoplasmic innate sensor for viral RNA, is tightly regulated to maintain immune homeostasis properly and prevent excessive inflammatory reactions other than initiation of antiviral innate response to eliminate RNA virus effectively. Posttranslational modifications, particularly ubiquitination, are crucial for regulation of RIG-I activity. Increasing evidence suggests that E3 ligases play important roles in various cellular processes, including cell proliferation and antiviral innate signaling. Here we identify that E3 ubiquitin ligase RING finger protein 122 (RNF122) directly interacts with mouse RIG-I through MS screening of RIG-I-interacting proteins in RNA virus-infected cells. The transmembrane domain of RNF122 associates with the caspase activation and recruitment domains (CARDs) of RIG-I; this interaction effectively triggers RING finger domain of RNF122 to deliver the Lys-48-linked ubiquitin to the Lys115 and Lys146 residues of RIG-I CARDs and promotes RIG-I degradation, resulting in a marked inhibition of RIG-I downstream signaling. RNF122 is widely expressed in various immune cells, with preferential expression in macrophages. Deficiency of RNF122 selectively increases RIG-I-triggered production of type I IFNs and proinflammatory cytokines in macrophages. RNF122-deficient mice exhibit more resistance against lethal RNA virus infection, with increased production of type I IFNs. Thus, we demonstrate that RNF122 acts as a selective negative regulator of RIG-I-triggered antiviral innate response by targeting CARDs of RIG-I and mediating proteasomal degradation of RIG-I. Our study outlines a way for E3 ligase to regulate innate sensor RIG-I for the control of antiviral innate immunity.


Assuntos
Imunidade Inata , Interferon Tipo I/imunologia , Macrófagos/imunologia , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Ubiquitina-Proteína Ligases/genética , Animais , Regulação da Expressão Gênica , Interferon Tipo I/antagonistas & inibidores , Interferon Tipo I/biossíntese , Macrófagos/virologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteólise , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Superfície Celular , Vírus Sendai/crescimento & desenvolvimento , Vírus Sendai/imunologia , Transdução de Sinais , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/deficiência , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/imunologia
20.
Age (Dordr) ; 37(5): 97, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26371058

RESUMO

Aging is the natural process of decline in physiological structure and function of various molecules, cells, tissues, and organs. Growing evidence indicates that increased immune genetic diversity and dysfunction of immune system cause aging-related pathophysiological process with the growth of age. In the present study, we observed that LPS-induced higher activation of cyclooxygenase (COX)-2 promoter is associated with the upregulated binding activity of nuclear factor kappa B (NF-κB) in peritoneal macrophages of aged mice than young ones. Additionally, COX-2 is a direct target of miR-101b and miR-26b in the macrophages. Significant upregulation of miR-101b and miR-26b effectively prevented LPS-induced excessive expression of COX-2 in the young mice. Because these negative regulatory factors were unresponsive to LPS stimulation, the levels of COX-2 were markedly higher in the macrophages of aged mice. Further study showed that NF-κB activation contributed to the increase in the expression of miR-101b and miR-26b in the LPS-stimulated macrophages of young mice, but not aged ones. Moreover, histone deacetylase (HDAC) inhibitor trichostatin A (TSA) upregulated expression of miR-101b and miR-26b in the aged mouse macrophages only, but not the young cells. This demonstrated that HDAC suppressed the expression of miR-101b and miR-26b in the LPS-treated macrophages of aged mice and contributed to the aging process. TSA-induced increased expression of miR-101b and miR-26b could further suppress COX-2 expression. These findings provide novel evidence on the regulation of immune senescence and miR-101b and miR-26b, which might be promising targets in treating aged-related inflammatory diseases. Epigenetic regulation of the microRNAs (miRNAs) provides an important evidence for the treatment of innate inflammatory disease with HDAC inhibitors in elderly.


Assuntos
Envelhecimento/genética , Ciclo-Oxigenase 2/genética , Regulação da Expressão Gênica , Inflamação/genética , Macrófagos/metabolismo , MicroRNAs/genética , Envelhecimento/metabolismo , Animais , Ciclo-Oxigenase 2/biossíntese , Modelos Animais de Doenças , Inflamação/induzido quimicamente , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , MicroRNAs/biossíntese , RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...