Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
PLoS Pathog ; 14(2): e1006884, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29447249

RESUMO

Host restriction factors constitute a formidable barrier for viral replication to which many viruses have evolved counter-measures. Human SAMD9, a tumor suppressor and a restriction factor for poxviruses in cell lines, is antagonized by two classes of poxvirus proteins, represented by vaccinia virus (VACV) K1 and C7. A paralog of SAMD9, SAMD9L, is also encoded by some mammals, while only one of two paralogs is retained by others. Here, we show that SAMD9L functions similarly to SAMD9 as a restriction factor and that the two paralogs form a critical host barrier that poxviruses must overcome to establish infection. In mice, which naturally lack SAMD9, overcoming SAMD9L restriction with viral inhibitors is essential for poxvirus replication and pathogenesis. While a VACV deleted of both K1 and C7 (vK1L-C7L-) was restricted by mouse cells and highly attenuated in mice, its replication and virulence were completely restored in SAMD9L-/- mice. In humans, both SAMD9 and SAMD9L are poxvirus restriction factors, although the latter requires interferon induction in many cell types. While knockout of SAMD9 with Crispr-Cas9 was sufficient for abolishing the restriction for vK1L-C7L- in many human cells, knockout of both paralogs was required for abolishing the restriction in interferon-treated cells. Both paralogs are antagonized by VACV K1, C7 and C7 homologs from diverse mammalian poxviruses, but mouse SAMD9L is resistant to the C7 homolog encoded by a group of poxviruses with a narrow host range in ruminants, indicating that host species-specific difference in SAMD9/SAMD9L genes serves as a barrier for cross-species poxvirus transmission.


Assuntos
Especificidade de Hospedeiro/genética , Infecções por Poxviridae/genética , Poxviridae/genética , Poxviridae/patogenicidade , Proteínas/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Mamíferos , Camundongos , Camundongos Knockout , Células NIH 3T3 , Infecções por Poxviridae/transmissão , Infecções por Poxviridae/virologia , Proteínas/genética , Homologia de Sequência , Proteínas Supressoras de Tumor/genética , Vaccinia virus/genética , Vaccinia virus/patogenicidade , Células Vero
2.
Int J Mol Sci ; 21(17)2020 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-32887217

RESUMO

Invasive breast cancer is highly regulated by tumor-derived cytokines in tumor microenvironment. The development of drugs that specifically target cytokines are promising in breast cancer treatment. In this study, we reported that arctigenin, a bioactive compound from Arctium lappa L., could decrease tumor-promoting cytokines GM-CSF, MMP-3, MMP-9 and TSLP in breast cancer cells. Arctigenin not only inhibited the proliferation, but also the invasion and stemness of breast cancer cells via decreasing GM-CSF and TSLP. Mechanistically, arctigenin decreased the promoter activities of GM-CSF and TSLP via reducing the nuclear translocation of NF-κB p65 which is crucial for the transcription of GM-CSF and TSLP. Furthermore, arctigenin-induced depletion of GM-CSF and TSLP inhibited STAT3 phosphorylation and ß-catenin signaling resulting in decreased proliferation, invasion and stemness of breast cancer cells in vitro and in vivo. Our findings provide new insights into the mechanism by which tumor-promoting cytokines regulate breast cancer progression and suggest that arctigenin is a promising candidate for cytokine-targeted breast cancer therapy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Citocinas/metabolismo , Furanos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Lignanas/farmacologia , Fator de Transcrição STAT3/metabolismo , beta Catenina/metabolismo , Animais , Apoptose , Biomarcadores Tumorais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Proliferação de Células , Citocinas/genética , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fator de Transcrição STAT3/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/genética
4.
Immunology ; 157(1): 13-20, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30681737

RESUMO

Interleukin-35 (IL-35) is a recently identified heterodimeric cytokine in the IL-12 family. It consists of an IL-12 subunit α chain (P35) and IL-27 subunit Epstein-Barr virus-induced gene 3 (EBI3) ß chain. Unlike the other IL-12 family members, it signals through four unconventional receptors: IL-12Rß2-IL-27Rα, IL-12Rß2-IL-12Rß2, IL-12Rß2-GP130, and GP130-GP130. Interleukin-35 signaling is mainly carried out through the signal transducer and activator of transcription family of proteins. It is secreted not only by regulatory T (Treg) cells, but also by CD8+ Treg cells, activated dendritic cells and regulatory B cells. It exhibits immunosuppressive functions distinct from those of other members of the IL-12 family; these are mediated primarily by the inhibition of T helper type 17 cell differentiation and promotion of Treg cell proliferation. Interleukin-35 plays a critical role in several immune-associated diseases, such as autoimmune diseases and viral and bacterial infections, as well as in tumors. In this review, we summarize the structure and function of IL-35, describe its role in immune-related disorders, and discuss the mechanisms by which it regulates the development and progression of diseases, including inflammatory bowel disease, collagen-induced arthritis, allergic airway disease, hepatitis, and tumors. The recent research on IL-35, combined with improved techniques of studying receptors and signal transduction pathways, allows for consideration of IL-35 as a novel immunotherapy target.


Assuntos
Doenças do Sistema Imunitário/metabolismo , Imunoterapia/métodos , Subunidade p35 da Interleucina-12/metabolismo , Interleucinas/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Autoimunidade , Humanos , Subunidade p35 da Interleucina-12/genética , Interleucinas/genética , Ativação Linfocitária , Antígenos de Histocompatibilidade Menor/genética , Transdução de Sinais
5.
Immunology ; 157(3): 257-267, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31120548

RESUMO

Asthma is a chronic inflammatory disease that involves a variety of cytokines and cells. Interleukin-16 (IL-16) is highly expressed during allergic airway inflammation and is involved in its development. However, its specific mechanism of action remains unclear. In the present study, we used an animal model of ovalbumin (OVA)-induced allergic asthma with mice harboring an IL-16 gene deletion to investigate the role of this cytokine in asthma, in addition to its underlying mechanism. Increased IL-16 expression was observed during OVA-induced asthma in C57BL/6J mice. However, when OVA was used to induce asthma in IL-16-/- mice, a diminished inflammatory reaction, decreased bronchoalveolar lavage fluid (BALF) eosinophil numbers, and the suppression of OVA-specific IgE levels in the serum and BALF were observed. The results also demonstrated decreased levels of T helper type 2 (Th2) and Th17 cytokines upon OVA-induced asthma in IL-16-/- mice. Hence, we confirmed that IL-16 enhances the lung allergic inflammatory response and suggest a mechanism possibly associated with the up-regulation of IgE and the promotion of Th2 and Th17 cytokine production. This work explored the mechanism underlying the regulation of IL-16 in asthma and provides a new target for the clinical treatment of asthma.


Assuntos
Asma/metabolismo , Hiper-Reatividade Brônquica/metabolismo , Interleucina-16/metabolismo , Pulmão/metabolismo , Ovalbumina , Células Th17/metabolismo , Células Th2/metabolismo , Animais , Asma/imunologia , Asma/fisiopatologia , Asma/prevenção & controle , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/fisiopatologia , Hiper-Reatividade Brônquica/prevenção & controle , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Broncoconstrição , Modelos Animais de Doenças , Feminino , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Interleucina-16/deficiência , Interleucina-16/genética , Pulmão/imunologia , Pulmão/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Baço/imunologia , Baço/metabolismo , Células Th17/imunologia , Células Th2/imunologia
6.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2796-2807, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28802852

RESUMO

Autophagy extensively participates in immune responses and inflammatory diseases. Myeloid-derived suppressor cells (MDSCs) are derived from CD11b+Gr1+ cells under pathological conditions and play an immunosuppressive role in the pathogenesis of cancer and inflammatory diseases. However, the role of autophagy in regulating the accumulation and activity of MDSCs remains unknown. In the present study, we evaluated the effects and mechanisms of autophagy on regulating accumulation and activity of MDSCs. We first found that granulocytic MDSCs (G-MDSCs), but not monocytic MDSCs (M-MDSCs), were accumulated in mice challenged by lipopolysaccharide (LPS) and showed an elevated autophagy activity. Pharmacological inhibition of autophagy significantly enhanced accumulation of G-MDSCs in vivo and in vitro. Notably, inhibition of autophagy enhanced the immunosuppressive activity of G-MDSCs on M1 macrophage polarization by promoting reactive oxygen species (ROS) production. Inhibition of autophagy promotes the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in G-MDSCs, which is required for the accumulation and activity of MDSCs. In addition, in vivo pharmacological inhibition of autophagy significantly attenuated the condition of mice challenged by LPS. Thus, we conclude that inhibition of autophagy contributes to accumulation and immunosuppressive function of G-MDSCs by promoting the activation of STAT3 signaling, suggesting that autophagy may play a critical role in regulating accumulation and activity of MDSCs. Our study provides new insights into understanding the mechanisms of autophagy in regulating immune responses and pathogenesis of inflammatory diseases.


Assuntos
Autofagia/imunologia , Granulócitos/imunologia , Células Supressoras Mieloides/imunologia , Fator de Transcrição STAT3/imunologia , Choque Séptico/imunologia , Transdução de Sinais/imunologia , Animais , Autofagia/efeitos dos fármacos , Granulócitos/patologia , Lipopolissacarídeos/toxicidade , Camundongos , Células Supressoras Mieloides/patologia , Choque Séptico/induzido quimicamente , Choque Séptico/patologia , Transdução de Sinais/efeitos dos fármacos
7.
Immunol Invest ; 46(3): 263-273, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27967259

RESUMO

Plasmacytoid dendritic cells (pDCs) exert dual roles in immune responses through inducing inflammation and maintaining immune tolerance. A switch of pDC phenotype from pro-inflammation to tolerance has therapeutic promise in the treatment of autoimmune diseases. Vinpocetine, a vasoactive vinca alkaloid extracted from the periwinkle plant, has recently emerged as an immunomodulatory agent. In this study, we evaluated the effect of vinpocetine on phenotype of pDCs isolated from C57BL/6 mice and explored its possible mechanism. Our data showed that vinpocetine significantly downregulated the expression of CD40, CD80, and CD86 on pDCs and increased the expression of translocator protein (TSPO), the specific receptor of vinpocetine, in pDCs. Vinpocetine significantly inhibited the Toll-like receptor 9 signaling pathway and reduced the secretion of related cytokines in pDCs through TSPO. Furthermore, viability of pDCs was significantly promoted by vinpocetine. These findings imply that vinpocetine serves as an immunomodulatory agent for pDCs and may be applied for the treatment of pDCs-related autoimmune diseases.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Células Dendríticas/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Alcaloides de Vinca/farmacologia , Vinca/imunologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Ilhas de CpG/genética , Citocinas/metabolismo , Células Dendríticas/imunologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/imunologia , Receptores de GABA/genética , Receptores de GABA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor Toll-Like 9/metabolismo
8.
Mol Cell Biochem ; 406(1-2): 293-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25981534

RESUMO

The metalloprotease lethal factor (LF) from Bacillus anthracis plays a vital role in anthrax toxin action, and thus becomes a target for anti-anthrax therapy. Following the guidelines based on existing metalloprotease inhibitors, we designed a 'first-generation' LF inhibitor R9LF-1. This inhibitor was shown to be very stable by itself in a wide range of pH and temperature and able to inhibit LF activity in vitro. However, as we reported previously in the presence of LF, this inhibitor was degraded to a small molecular weight species, resulting in a significantly decreased ability to protect MAPKK from cleavage by LF as well as to protect murine macrophages from lethal toxin. In order to elucidate this unusual phenomenon to build solid basis for high-efficiency LF inhibitor development, we performed extensive research to study the effect of LF on its peptide-based inhibitor. Effects of temperature and incubation period of time on generation of the smaller peptide (short version R9LF-1) by LF as well as its catalytic domain were analyzed. We found that LF degraded R9LF-1 with maximum efficiency in the pH range of 7.0-8.5, which correlates well with the range of LF enzymatic activity with its native substrate. The degradation showed a deviation from normal hyperbolic kinetics but a similarity to the kinetics profile of an enzyme-catalyzed reaction with positive cooperativity. The short version R9LF-1 had decreased inhibitory activity toward LF; surprisingly, BIAcore results suggested a better affinity for its binding to LF. In addition, R9LF-1 was not hydrolyzed by other common proteases, such as chymotrypsin and pepsin, suggesting hydrolysis of the bond between amino acid and hydroxamate groups is unique to LF. This study calls for caution when designing peptide-based LF inhibitors and when interpreting effects of these types of inhibitors.


Assuntos
Antígenos de Bactérias/química , Toxinas Bacterianas/química , Metaloproteases/química , Oligopeptídeos/química , Inibidores de Proteases/química , Quimotripsina/química , Cinética , Ligação Proteica , Proteólise
9.
J Med Virol ; 86(10): 1780-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24699993

RESUMO

The signal peptide Ag85B of Bacillus Chalmette-Guerin (BCG) was used to construct a recombinant plasmid of BCG. The BCG-Ag85B gene and fused EBV LMP2A and BZLF1 genes were amplified and successively inserted into the Escherichia coli-BCG shuttle-vector pMV261. The recombinant plasmids were then amplified in E. coli DH5α and transformed into competent BCG. The expression of BZLF1 and LMP2A fusion proteins in recombinant-BCG (rBCG) was shown by Western blot. After the injection of recombinant-BCG into mice, antibodies against the fusion protein BZLF1 and LMP2A were measured by ELISA, and the cellular immune effects were determined by the lactate dehydrogenate (LDH) release assays. The results confirmed that the cloned genes of BCG-Ag85B and Z2A were correctly inserted into the vector pMV261. The recombinant plasmid pMVZ2A expressed Z2A in BCG effectively after transformation. The rBCG proteins were recognized by the BZLF1 (LMP2A) antibody. An ELISA demonstrated that rBCG could stimulate the generation of antibody against the fusion protein. The fusion gene was constructed successfully, and the rBCG induced humoral and cellular immune response in mice.


Assuntos
Vetores Genéticos , Mycobacterium bovis/genética , Transativadores/imunologia , Proteínas da Matriz Viral/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Transativadores/genética , Vacinação/métodos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas da Matriz Viral/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
10.
Immun Inflamm Dis ; 10(7): e643, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35759238

RESUMO

INTRODUCTION: Acute liver inflammatory reactions contribute to many health problems; thus, it is critical to understand the underlying pathogenic mechanisms of acute hepatitis. In this study, an experimental in vivo model of concanavalin A (ConA)-induced hepatitis was used. MATERIALS AND METHODS: C57BL/6 (wild-type, WT) or inducible nitric oxide synthase-deficient (iNOS-/- ) mice were injected with PBS or 15 mg/kg ConA via tail vein. Detection of liver injury by histological examination and apoptosis, and flow cytometry to detect the effect of immune cells on liver injury. RESULTS: iNOS-/-  mice had lower levels of the liver enzymes aspartate aminotransferase and alanine aminotransferase, suggesting that they were protected against ConA-induced pathological liver injury and that iNOS participated in the regulation of hepatitis. Furthermore, iNOS deficiency was found to lower CD86 expression and suppressed the messenger RNA levels of inflammatory factors in the liver. In vitro experiments also demonstrated that iNOS deficiency suppressed the sequential phosphorylation of the mitogen-activated protein kinase pathway cascade, thereby inhibiting the M1 polarization of macrophages and consequently suppressing the transcription of inflammation factors. CONCLUSION: iNOS may contribute to ConA-induced inflammation by promoting the activation of proinflammatory macrophages.


Assuntos
Hepatite , Animais , Concanavalina A/metabolismo , Concanavalina A/toxicidade , Hepatite/etiologia , Hepatite/metabolismo , Hepatite/patologia , Inflamação , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo
11.
Artigo em Zh | MEDLINE | ID: mdl-33441222

RESUMO

Objective To study the effect of CD11b agonist leukadherin-1 (LA1) on Toll-like receptor 7 (TLR7)- and TLR9-induced activation of mouse bone marrow-derived dendritic cells (BMDCs) and its specific mechanism. Methods BMDCs were successfully induced and the concentrations of LA1 used in the study were determined by CCK-8 assay and annexin V-FITC/PI double staining. BMDCs were treated with LA1 for 2 hours followed by stimulation of TLR7 agonist R837 and TLR9 agonist CpG1826. The expression of BMDCs surface markers CD40, CD86 and MHC-II were detected by flow cytometry; IL-6, IL-12p40 and tumor necrosis factor α (TNF-α) in the cell culture supernatant were detected by ELISA; the phosphorylation of NF-κB p65 in BMDCs was detected by Western blotting. Results LA1 concentration below 20 µmol/L had no effect on the viability and apoptosis of BMDCs. LA1 pretreatment significantly inhibited R837- and CpG 1826-induced expression of CD40, CD86 and MHC-II , and the secretion of IL-6, IL-12p40 and TNF-α in BMDCs. Moreover, LA1 pretreatment significantly inhibited the phosphorylation of NF-κB p65 activated by R837 and CpG1826 in BMDCs. Conclusion CD11b agonist LA1 can significantly inhibit the activation of TLR7 and TLR9 in BMDCs by blocking the NF-κB p65 signaling pathway.


Assuntos
NF-kappa B , Receptor 7 Toll-Like , Animais , Benzoatos , Medula Óssea , Células Dendríticas , Glicoproteínas de Membrana , Camundongos , Tioidantoínas , Receptor Toll-Like 9
12.
Biomed Res Int ; 2020: 1893434, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32337226

RESUMO

Gastric cancer is among the most common malignant tumors of the digestive tract. Establishing a robust and reliable animal model is the foundation for studying the pathogenesis of cancer. The present study established a mouse model of gastric carcinoma by inoculating immunocompetent mice with MKN45 cells using microcarrier. Sixty male C57BL/6 mice were randomly divided into three groups: a 2D group, an empty carrier group, and a 3D group, according to the coculture system of MKN45 and the microcarrier. The mouse models were established by hypodermic injection. Time to develop tumor, rate of tumor formation, and pathological features were observed in each group. In the 3D group, the tumorigenesis time was short, while the rate of tumor formation was high (75%). There was no detectable tumor formation in either the 2D or the empty carrier group. Both H&E and immunohistochemical staining of the tumor xenograft showed characteristic evidence of human gastric neoplasms. The present study successfully established a human gastric carcinoma model in immunocompetent mice, which provides a novel and valuable animal model for the cancer research and development of anticancer drugs.


Assuntos
Técnicas de Cultura de Células , Neoplasias Experimentais , Neoplasias Gástricas , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
14.
Exp Ther Med ; 19(2): 1370-1378, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32010311

RESUMO

Atherosclerosis (AS) is currently the leading cause of mortality worldwide, with the development of new strategies to prevent the formation and rupture of atherosclerotic plaques being a paramount area of research. Amounting evidence suggests autophagy has an important role in the pathogenesis of AS and may be a potential therapeutic target. In this study, the effect of SBI-0206965(6965), a novel inhibitor of autophagy, was tested on the development of AS in apolipoprotein E deficient (ApoE-/-) mice. Systemic application of 6965 was found to aggravate AS, with increased plaque size and decreased plaque stability in comparison with the control. Of note, it was observed that 6965 decreased the proportion of myeloid-derived suppressor cells (MDSCs). Further investigation demonstrated MDSCs markedly alleviated AS in ApoE-/- mice; while 6965 reduced the viability and promoted apoptosis of MDSCs in vitro. This is the first study describing an association between autophagy and MDSCs in AS models, providing a novel mechanism to potentially target in the management of this condition.

15.
Biochim Biophys Acta Mol Basis Dis ; 1866(1): 165554, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31513833

RESUMO

Activation of interferon (IFN)-I signaling in B cells contributes to the pathogenesis of systemic lupus erythematosus (SLE). Recent studies have shown that myeloid-derived suppressor cells (MDSCs) significantly expand in SLE patients and lupus-prone MRL/lpr mice and contribute to the pathogenesis of SLE. However, the role of SLE-derived MDSCs in regulating IFN-I signaling activation of B cells remains unknown. Here, we demonstrate that expansions of MDSCs, including granulocyte (G)-MDSCs and monocytic (M)-MDSCs, during the progression of SLE were correlated with the IFN-I signature of B cells. Interestingly, G-MDSCs from MRL/lpr mice, but not M-MDSCs, could significantly promote IFN-I signaling activation of B cells and contribute to the pathogenesis of SLE. Mechanistically, we identified that the long non-coding RNA NEAT1 was over-expressed in G-MDSCs from MRL/lpr mice and could induce the promotion of G-MDSCs on IFN-I signaling activation of B cells through B cell-activating factor (BAFF) secretion. Importantly, NEAT1 deficiency significantly attenuated the lupus symptoms in pristane-induced lupus mice. In addition, there was a positive correlation between NEAT1 and BAFF with the IFN signature in SLE patients. In conclusion, G-MDSCs may contribute to the IFN signature in SLE B cells through the NEAT1-BAFF axis, highlighting G-MDSCs as a potential therapeutic target to treat SLE.


Assuntos
Fator Ativador de Células B/metabolismo , Linfócitos B/metabolismo , Interferon Tipo I/metabolismo , Lúpus Eritematoso Sistêmico/metabolismo , Células Supressoras Mieloides/metabolismo , RNA Longo não Codificante/metabolismo , Transdução de Sinais/fisiologia , Animais , Citocinas/metabolismo , Progressão da Doença , Feminino , Granulócitos/metabolismo , Granulócitos/patologia , Humanos , Rim/metabolismo , Rim/patologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Células Supressoras Mieloides/patologia
16.
Am J Transl Res ; 11(4): 2350-2358, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31105841

RESUMO

Transplantation of adult human hepatic stem/progenitor cells (hHSPCs) has been considered as an alternative therapy, replacing donor liver transplantation to treat liver cirrhosis. This study assessed the antifibrotic effects of hHSPCs in mice with fibrosis induced by carbon tetrachloride (CCl4) and examined the actions of hHSPCs on the fibrogenic activity of human hepatic stellate cells (HSCs) in a coculture system. Isolated hHSPCs expressed stem/progenitor cell phenotypic markers. Mice were given CCl4 (twice weekly for 7 weeks) and hHSPC transplantation weekly. CCl4 induced advanced fibrosis (bridging fibrosis and cirrhosis) in mice, which was prevented by hHSPC transplantation. The liver of hHSPC-transplanted mice showed only occasional short septa and focal parenchymal fibrosis, and a 50% reduction in hepatic collagen, assessed by Sirius red stain histomorphometry. Moreover, the proteins for α-smooth muscle actin (α-SMA) and collagen I were decreased. While α-SMA, collagen α1(I), and tissue inhibitor of metalloproproteinase-1 mRNAs were decreased, matrix metalloproteinase (MMP)-1 mRNA was increased, consistent with decreased fibrogenesis. MMP-2 and transforming growth factor-ß were not affected. Alanine aminotransferase and aspartate aminotransferase were lower, suggesting improvement of liver function/damage. In coculture, hHSPCs elicited changes of α-SMA and fibrogenic molecules in HSCs similar to those observed in vivo, providing evidence for a functional link between hHSPCs and HSCs. A decreased HSC proliferation was noted. Thus, transplantation of hHSPCs prevents histogenesis of advanced liver fibrosis caused by CCl4. hHSPCs mediate downregulation of HSC activation coincident with modulation of fibrogenic molecule expression, leading to suppression of fibrogenesis both in vivo and in vitro.

17.
Biochim Biophys Acta Mol Basis Dis ; 1865(3): 535-546, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30557700

RESUMO

Myeloid-derived suppressor cells (MDSCs) play an immunosuppressive role in the pathogenesis of inflammatory diseases. CD180, a TLR-like protein, can regulate the proliferation and activation of immune cells. However, the roles of CD180 in regulating the accumulation and function of MDSCs have not been investigated. Here, we found that, compared with non-treated controls, the expression of CD180 was significantly elevated in MDSCs, especially granulocytic MDSCs (G-MDSCs), from mice challenged with lipopolysaccharide (LPS). Ligation of CD180 by the anti-CD180 antibody not only blocked the expansion of MDSCs by preventing the phosphorylation of signal transducer and activator of transcription 3 (STAT3), but also reduced the immunosuppressive activity of MDSCs on M1 macrophage polarization through inhibition of Arg-1 expression in vitro. In vivo studies showed that injection of anti-CD180 antibody significantly aggravated pathological lesions in mice challenged with LPS. Furthermore, injection of anti-CD180 antibody inhibited the accumulation of G-MDSCs in mice challenged with LPS and reduced the immunosuppressive activity of G-MDSCs on M1 macrophage polarization. Based on these findings, we conclude that ligation of CD180 contributes to the pathogenesis of endotoxic shock by inhibiting the accumulation and immunosuppressive activity of G-MDSCs, thus providing insight into the function of CD180 in inflammatory diseases.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD/imunologia , Células Supressoras Mieloides/imunologia , Fator de Transcrição STAT3/fisiologia , Choque Séptico/induzido quimicamente , Choque Séptico/imunologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/citologia , Células Supressoras Mieloides/efeitos dos fármacos , Ligação Proteica , Choque Séptico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
18.
Am J Transl Res ; 11(5): 3029-3038, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31217872

RESUMO

Immune-mediated liver injury plays a crucial role in the pathogenesis of liver diseases, which can result from viral infections, autoimmunity, alcohol intake, and drug use. Concanavalin A (Con A)-induced hepatitis is a well-characterized murine model with similar pathophysiology to that of human viral and autoimmune hepatitis. Capsaicin, a selective agonist of the transient potential vanilloid subfamily member 1 (TRPV1) receptor, exhibits anti-inflammatory effects on various causes of inflammation. In the present study, we investigated the effect of capsaicin on Con A-induced hepatitis. Capsaicin (1 mg/kg body weight) was administered by intraperitoneal injection, after which (30 minutes), the mice were challenged intravenously with Con A (20 µg/g body weight). We collected serum for plasma transaminase analysis. Pro-inflammatory cytokine levels and hepatocyte apoptosis were assayed by ELISA and TUNEL, respectively. Liver samples were collected for real-time PCR, hematoxylin and eosin staining, and measuring oxidative stress and myeloperoxidase levels. Activation of splenocytes and hepatic mononuclear cells was analyzed by flow cytometry. Compared with control, the capsaicin-treated group showed significantly decreased aminotransferase levels and markedly prolonged mouse survival. Capsaicin pretreatment also attenuated hepatocyte apoptosis and oxidative stress. Furthermore, tumor necrosis factor-α and interferon-γ levels in serum and liver were significantly suppressed, while the percentage of myeloid-derived suppressor cells increased after capsaicin pretreatment. Our findings indicate that capsaicin pretreatment protects mice from Con A-induced hepatic damage and is partially involved in inhibiting hepatocyte apoptosis, oxidative stress, and inflammatory mediators as well as regulating activation and recruitment of intrahepatic leukocytes.

19.
Front Immunol ; 10: 215, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30809230

RESUMO

Dysregulation of macrophage has been demonstrated to contribute to aberrant immune responses and inflammatory diseases. CD11b, expressed on macrophages, plays a critical role in regulating pathogen recognition, phagocytosis, and cell survival. In the present study, we explored the effect of leukadherin-1 (LA1), an agonist of CD11b, on regulating LPS-induced pro-inflammatory response in macrophages and endotoxic shock. Intriguingly, we found that LA1 could significantly reduce mortalities of mice and alleviated pathological injury of liver and lung in endotoxic shock. In vivo studies showed that LA1-induced activation of CD11b significantly inhibited the LPS-induced pro-inflammatory response in macrophages of mice. Moreover, LA1-induced activation of CD11b significantly inhibited LPS/IFN-γ-induced pro-inflammatory response in macrophages by inhibiting MAPKs and NF-κB signaling pathways in vitro. Furthermore, the mice injected with LA1-treated BMDMs showed fewer pathological lesions than those injected with vehicle-treated BMDMs in endotoxic shock. In addition, we found that activation of TLR4 by LPS could endocytose CD11b and activation of CD11b by LA1 could endocytose TLR4 in vitro and in vivo, subsequently blocking the binding of LPS with TLR4. Based on these findings, we concluded that LA1-induced activation of CD11b negatively regulates LPS-induced pro-inflammatory response in macrophages and subsequently protects mice from endotoxin shock by partially blocking LPS-TLR4 interaction. Our study provides a new insight into the role of CD11b in the pathogenesis of inflammatory diseases.


Assuntos
Antígenos CD1/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Choque Séptico/etiologia , Choque Séptico/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Benzoatos/farmacologia , Biomarcadores , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Endocitose , Feminino , Estimativa de Kaplan-Meier , Lipopolissacarídeos/imunologia , Hepatopatias/etiologia , Hepatopatias/patologia , Ativação de Macrófagos/efeitos dos fármacos , Camundongos , Modelos Biológicos , Mortalidade , Choque Séptico/complicações , Choque Séptico/mortalidade , Tioidantoínas/farmacologia
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 34(11): 961-968, 2018 Nov.
Artigo em Zh | MEDLINE | ID: mdl-30591103

RESUMO

Objective To study the effect of CD11b agonist leukadherin-1 (LA1) on the aggregation and immunosuppressive function of myeloid-derived suppressor cells (MDSCs) and its therapeutic effect on the condition of endotoxic shock mice. Methods The percentages of MDSCs , granulocytic myeloid-derived suppressor cells(G-MDSCs)and monocytic myeloid-derived suppressor cells(M-MDSCs)in spleen were detected by flow cytometry, after C57BL/6 female mice were injected of LA1 to activate through abdominal cavity for 12 hours and 48 hours. MDSCs were induced from the femur and tibia of C57BL/6 female mice in vitro. The expression levels of immunosuppressive related factors, such as interleukin 10 (IL-10), NADPH oxidase 1 (NOX1) and inducible nitric oxide synthase (iNOS) , were detected by real time quantitative PCR. C57BL/6 female mice were randomly divided into PBS group, LA1 group, PBS combined LPS group and LA1 combined LPS group. Flow cytometry was utilized to detect the ratio changes of MDSCs, G-MDSCs and M-MDSCs as well as the expression of CD86 and CD40 in macrophage, hematoxylin-eosin staining of lung and liver was utilized to detect the pathological injury, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling(TUNEL)was used to detect the apoptosis of pneumonocyte and hepatocyte and mortality analysis was reflected the severity of the disease. Based on the above indicators, we analyzed the effects of LA1 on the aggregation of MDSCs and the condition of mice in endotoxic shock. Results The ratio of MDSCs was increased by LA1 treatment for 12 and 48 hours. Further analysis of the proportions of G-MDSCs showed that LA1 treatment for 12 hours increased the proportions of G-MDSCs compared with the control group. In vitro, mRNA levels of IL-10, NOX1 and iNOS were increased after LA1 treatment in MDSCs. In vivo experiments, compared with the PBS combined LPS group, the proportions of MDSCs and G-MDSCs in LA1 combined LPS group were increased, the injuries of liver and lung were alleviated, the mortalities were reduced, and the activations of macrophage were decreased. Conclusion The activation of CD11b by LA1 alleviates endotoxin shock by promoting the aggregation of MDSCs and the expression of immunosuppressive related factors.


Assuntos
Benzoatos/farmacologia , Antígeno CD11b/agonistas , Células Supressoras Mieloides/citologia , Choque Séptico/tratamento farmacológico , Tioidantoínas/farmacologia , Animais , Feminino , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidase 1/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Distribuição Aleatória , Baço/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA