Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nature ; 586(7830): 572-577, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32726802

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a respiratory disease called coronavirus disease 2019 (COVID-19), the spread of which has led to a pandemic. An effective preventive vaccine against this virus is urgently needed. As an essential step during infection, SARS-CoV-2 uses the receptor-binding domain (RBD) of the spike protein to engage with the receptor angiotensin-converting enzyme 2 (ACE2) on host cells1,2. Here we show that a recombinant vaccine that comprises residues 319-545 of the RBD of the spike protein induces a potent functional antibody response in immunized mice, rabbits and non-human primates (Macaca mulatta) as early as 7 or 14 days after the injection of a single vaccine dose. The sera from the immunized animals blocked the binding of the RBD to ACE2, which is expressed on the cell surface, and neutralized infection with a SARS-CoV-2 pseudovirus and live SARS-CoV-2 in vitro. Notably, vaccination also provided protection in non-human primates to an in vivo challenge with SARS-CoV-2. We found increased levels of RBD-specific antibodies in the sera of patients with COVID-19. We show that several immune pathways and CD4 T lymphocytes are involved in the induction of the vaccine antibody response. Our findings highlight the importance of the RBD domain in the design of SARS-CoV-2 vaccines and provide a rationale for the development of a protective vaccine through the induction of antibodies against the RBD domain.


Assuntos
Anticorpos Antivirais/imunologia , Betacoronavirus/imunologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Pandemias/prevenção & controle , Pneumonia Viral/imunologia , Pneumonia Viral/prevenção & controle , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , COVID-19 , Vacinas contra COVID-19 , Humanos , Macaca mulatta/imunologia , Macaca mulatta/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Animais , Modelos Moleculares , Domínios Proteicos , SARS-CoV-2 , Soro/imunologia , Baço/citologia , Baço/imunologia , Linfócitos T/imunologia , Vacinação
2.
J Transl Med ; 22(1): 257, 2024 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-38461288

RESUMO

BACKGROUND: Neural Tube Defects (NTDs) are congenital malformations of the central nervous system resulting from the incomplete closure of the neural tube during early embryonic development. Neuroinflammation refers to the inflammatory response in the nervous system, typically resulting from damage to neural tissue. Immune-related processes have been identified in NTDs, however, the detailed relationship and underlying mechanisms between neuroinflammation and NTDs remain largely unclear. In this study, we utilized integrated multi-omics analysis to explore the role of neuroinflammation in NTDs and identify potential prenatal diagnostic markers using a murine model. METHODS: Nine public datasets from Gene Expression Omnibus (GEO) and ArrayExpress were mined using integrated multi-omics analysis to characterize the molecular landscape associated with neuroinflammation in NTDs. Special attention was given to the involvement of macrophages in neuroinflammation within amniotic fluid, as well as the dynamics of macrophage polarization and their interactions with neural cells at single-cell resolution. We also used qPCR assay to validate the key TFs and candidate prenatal diagnostic genes identified through the integrated analysis in a retinoic acid-induced NTDs mouse model. RESULTS: Our analysis indicated that neuroinflammation is a critical pathological feature of NTDs, regulated both transcriptionally and epigenetically within central nervous system tissues. Key alterations in gene expression and pathways highlighted the crucial role of STATs molecules in the JAK-STAT signaling pathway in regulating NTDs-associated neuroinflammation. Furthermore, single-cell resolution analysis revealed significant polarization of macrophages and their interaction with neural cells in amniotic fluid, underscoring their central role in mediating neuroinflammation associated with NTDs. Finally, we identified a set of six potential prenatal diagnostic genes, including FABP7, CRMP1, SCG3, SLC16A10, RNASE6 and RNASE1, which were subsequently validated in a murine NTDs model, indicating their promise as prospective markers for prenatal diagnosis of NTDs. CONCLUSIONS: Our study emphasizes the pivotal role of neuroinflammation in the progression of NTDs and underlines the potential of specific inflammatory and neural markers as novel prenatal diagnostic tools. These findings provide important clues for further understanding the underlying mechanisms between neuroinflammation and NTDs, and offer valuable insights for the future development of prenatal diagnostics.


Assuntos
Multiômica , Defeitos do Tubo Neural , Gravidez , Feminino , Animais , Camundongos , Doenças Neuroinflamatórias , Estudos Prospectivos , Defeitos do Tubo Neural/diagnóstico , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/induzido quimicamente , Sistema Nervoso Central/patologia
3.
Mol Ther ; 30(2): 644-661, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-34547462

RESUMO

Preclinical and clinical studies have validated the antitumor effects of several oncolytic viruses (OVs). However, the efficacy of OVs is limited when they are administered as monotherapies. Combination therapy is a promising direction for oncolytic virotherapy in the future. A high dose of vitamin C (VitC) exerts anticancer effects by triggering the accretion of substantial amounts of reactive oxygen species (ROS). OVs can induce immunogenic tumor cell death and elicit an antitumor immune response. ROS play an important role in immunogenic cell death (ICD). This study aimed to explore whether high-dose VitC in combination with oncolytic adenoviruses (oAds) exhibited a synergistic antitumor effect. High-dose VitC synergized with oAds against tumor by enhancing immunogenic tumor cell death. Combination therapy with high-dose VitC and oAds significantly increased the number of T cells in the tumor microenvironment (TME) and promoted the activation of T cells. Furthermore, the antitumor effect of the combination therapy was CD8+ T cell dependent. In addition, combination therapy with high-dose VitC and oAds reprogramed the immunosuppressive TME. Our study provides a new strategy for combination therapy of OVs.


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Adenoviridae/genética , Humanos , Morte Celular Imunogênica , Neoplasias/terapia , Vírus Oncolíticos/fisiologia , Microambiente Tumoral
4.
PLoS Genet ; 16(11): e1009159, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33175846

RESUMO

Tumor metastasis is the major cause of poor prognosis and mortality in colorectal cancer (CRC). However, early diagnosis of highly metastatic CRC is currently difficult. In the present study, we screened for a novel biomarker, GDNF family receptor alpha 1 (GFRA1) based on the expression and methylation data in CRC patients from The Cancer Genome Altlas (TCGA), followed by further analysis of the correlation between the GFRA1 expression, methylation, and prognosis of patients. Our results show DNA hypomethylation-mediated upregulation of GFRA1 in invasive CRC, and it was found to be correlated with poor prognosis of CRC patients. Furthermore, GFRA1 methylation-modified sequences were found to have potential as methylation diagnostic markers of highly metastatic CRC. The targeted demethylation of GFRA1 by dCas9-TET1CD and gRNA promoted CRC metastasis in vivo and in vitro. Mechanistically, demethylation of GFRA1 induces epithelial-mesenchymal transition (EMT) by promoting AKT phosphorylation and increasing c-Jun expression in CRC cells. Collectively, our findings indicate that GFRA1 hypomethylation can promote CRC invasion via inducing EMT, and thus, GFRA1 methylation can be used as a biomarker for the early diagnosis of highly metastasis CRC.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Transição Epitelial-Mesenquimal/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Neoplasias Pulmonares/genética , Animais , Proliferação de Células/genética , Estudos de Coortes , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Biologia Computacional , Desmetilação do DNA , Metilação de DNA , Conjuntos de Dados como Assunto , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HT29 , Humanos , Neoplasias Pulmonares/secundário , Camundongos , Invasividade Neoplásica/genética , Fosforilação/genética , Prognóstico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA-Seq , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
6.
BMC Psychiatry ; 22(1): 629, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36167540

RESUMO

BACKGROUND: The pathophysiological mechanisms of aggression are manifold and they may closely interconnect. Current study aimed to determine the gut microbiota and its metabolites, and clarify their correlations with inflammation, oxidation, leaky gut and clinical profiles underlying aggression in schizophrenia (ScZ). METHODS: Serum and stool specimens from ScZ inpatients with (ScZ-Ag, 25 cases) and without aggression (NScZ-Ag, 25 cases) were collected. Systemic inflammation, oxidation and leaky gut biomarkers were determined by ELISA, gut microbiota by 16S rRNA sequencing, short-chain fatty acids (SCFAs) by gas chromatography-mass spectrometry analysis and neurotransmitters by liquid chromatograph mass spectrometry analysis. RESULTS: Significantly higher systemic pro-inflammation, pro-oxidation and leaky gut biomarkers were observed in ScZ-Ag than NScZ-Ag group (all P<0.001). Compared to NScZ-Ag group, the alpha-diversity and evenness of fecal bacterial community were much lower, the abundance of fecal genera Prevotella was significantly increased, while that Bacteroides, Faecalibacterium, Blautia, Bifidobacterium,Collinsella and Eubacterium_coprostanoligenes were remarkably reduced in ScZ-Ag group (all corrected P<0.001). Meanwhile, 6 SCFAs and 6 neurotransmitters were much lower in ScZ-Ag group (all P<0.05). Finally, a few strongly positive or negative correlations among altered gut microbiota, SCFAs, systemic pro-inflammation, leaky gut, pro-oxidation and aggression severity were detected. CONCLUSIONS: These results demonstrate that pro-inflammation, pro-oxidation and leaky gut phenotypes relating to enteric dysbacteriosis and microbial SCFAs feature the aggression onset or severity in ScZ individuals.


Assuntos
Microbioma Gastrointestinal , Esquizofrenia , Agressão , Biomarcadores , Citocinas , Ácidos Graxos Voláteis/análise , Ácidos Graxos Voláteis/metabolismo , Humanos , Inflamação , Pacientes Internados , RNA Ribossômico 16S/genética
7.
Carcinogenesis ; 41(2): 235-244, 2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-31802101

RESUMO

DNA methylation plays a crucial role in the pathogenesis of various diseases, including colorectal cancer (CRC). However, the global and temporal DNA methylation pattern during initiation and progression of colitis-associated cancer (CAC) are still unknown, including the potential therapeutic strategy of targeting methylation for CAC. In the present study, the global DNA methylation pattern was determined at different time points during CAC using DNA methylation sequencing, followed by the Starburst plot integrating alterations and potential functional prediction analysis. After demonstrating the regulatory role of DNA methyltransferases (DNMTs) on the expression of hub-genes in CRC cells, DNMT inhibitors were administered to treat CAC mice. Our results indicated that 811 genes were hypermethylated at different time points during initiation and progression of CAC. Genes that were downregulated and hypermethylated during CAC, including hub-genes BAD and inositol polyphosphate phosphatase-like 1 (INPPL1), were involved in MAPK signaling pathways, kit receptor signaling pathways, apoptosis and EGF/EGFR signaling pathways. Upregulated DNMTs (DNMT1, DNMT3A and DNMT3B) mediated downregulation and hypermethylation of BAD and INPPL1 in CAC and CRC cells. Low doses of DNMT inhibitors (decitabine (DAC) and azacitidine (AZA)) exerted efficient antitumor effects in CAC, accompanied with upregulation of BAD and INPPL1 expression, and apoptosis induction. In summary, the present study demonstrates the temporal DNA methylation pattern during CAC and provides a novel therapeutic strategy for treating this disease.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Colite/patologia , Neoplasias Colorretais/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Azacitidina/administração & dosagem , Azoximetano/toxicidade , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Linhagem Celular Tumoral , Colite/induzido quimicamente , Colite/diagnóstico por imagem , Colo/diagnóstico por imagem , Colo/efeitos dos fármacos , Colo/patologia , Colonoscopia , Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Decitabina/administração & dosagem , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta a Droga , Humanos , Mucosa Intestinal/diagnóstico por imagem , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Masculino , Camundongos , Terapia de Alvo Molecular/métodos , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/genética , Regulação para Cima , Proteína de Morte Celular Associada a bcl/genética
8.
J Cell Mol Med ; 24(7): 4341-4349, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32119762

RESUMO

SARI (Suppressor of AP-1, regulated by IFN-ß) is known to play an important role in some systemic disease processes such an inflammatory conditions and cancer. We hypothesize that SARI may also play a role in ocular diseases involving inflammation and neovascularization. To explore our hypothesis, further, we investigated an endotoxin-induced uveitis (EIU) and experimental argon laser-induced choroidal neovascularization (CNV) model in SARI wild-type (SARIWT ) and SARI-deficient (SARI-/- ) mice. Through imaging, morphological and immunohistochemical (IHC) studies, we found that SARI deficiency exacerbated the growth of CNV. More VEGF-positive cells were presented in the retina of SARI-/- mice with CNV. Compared to SARIWT  mice, more inflammatory cells infiltrated the ocular anterior segment and posterior segments in SARI-/- mice with EIU. Collectively, the results point to a potential dual functional role of SARI in inflammatory ocular diseases, suggesting that SARI could be a potential therapy target for ocular inflammation and neovascularization.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/genética , Neovascularização de Coroide/genética , Inflamação/genética , Retina/metabolismo , Uveíte/genética , Animais , Neovascularização de Coroide/patologia , Citocinas/genética , Modelos Animais de Doenças , Endotoxinas/toxicidade , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Lasers de Gás/efeitos adversos , Camundongos , Camundongos Knockout , Retina/patologia , Retina/efeitos da radiação , Uveíte/induzido quimicamente , Uveíte/patologia , Fator A de Crescimento do Endotélio Vascular/genética
9.
J Cell Mol Med ; 24(23): 13837-13852, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33085209

RESUMO

Pernicious placenta previa with placenta percreta (PP) is a catastrophic condition during pregnancy. However, the underlying pathogenesis remains unclear. In the present study, the placental tissues of normal cases and PP tissues of pernicious placenta previa cases were collected to determine the expression profile of protein-coding genes, miRNAs, and lncRNAs through sequencing. Weighted gene co-expression network analysis (WGCNA), accompanied by miRNA target prediction and correlation analysis, were employed to select potential hub protein-coding genes and lncRNAs. The expression levels of selected protein-coding genes, Wnt5A and MAPK13, were determined by quantitative PCR and immunohistochemical staining, and lncRNA PTCHD1-AS and PAPPA-AS1 expression levels were determined by quantitative PCR and fluorescence in situ hybridization. The results indicated that 790 protein-coding genes, 382 miRNAs, and 541 lncRNAs were dysregulated in PP tissues, compared with normal tissues. WGCNA identified coding genes in the module (ME) black and ME turquoise modules that may be involved in the pathogenesis of PP. The selected potential hub protein-coding genes, Wnt5A and MAPK13, were down-regulated in PP tissues, and their expression levels were positively correlated with the expression levels of PTCHD1-AS and PAPPA-AS1. Further analysis demonstrated that PTCHD1-AS and PAPPA-AS1 regulated Wnt5A and MAPK13 expression by interacting with specific miRNAs. Collectively, our results provided multi-omics data to better understand the pathogenesis of PP and help identify predictive biomarkers and therapeutic targets for PP.


Assuntos
Biomarcadores , Suscetibilidade a Doenças , Genômica , Placenta Acreta/etiologia , Placenta Acreta/metabolismo , Proteômica , Adulto , Biologia Computacional/métodos , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Genômica/métodos , Humanos , Imuno-Histoquímica , Placenta Acreta/diagnóstico por imagem , Placenta Acreta/patologia , Gravidez , Proteômica/métodos , Transdução de Sinais , Tomografia Computadorizada por Raios X
10.
J Cell Mol Med ; 24(1): 189-201, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31578820

RESUMO

SARI (suppressor of AP-1, regulated by IFN) impaired tumour growth by promoting apoptosis and inhibiting cell proliferation and tumour angiogenesis in various cancers. However, the role of SARI in regulating tumour-associated inflammation microenvironment is still elusive. In our study, the colitis-dependent and -independent primary model were established in SARI deficiency mice and immuno-reconstructive mice to investigate the functional role of SARI in regulating tumour-associated inflammation microenvironment and primary colon cancer formation. The results have shown that SARI deficiency promotes colitis-associated cancer (CAC) development only in the presence of colon inflammation. SARI inhibited tumour-associated macrophages (TAM) infiltration in colon tissues, and SARI deficiency in bone marrow cells has no observed role in the promotion of intestinal tumorigenesis. Mechanism investigations indicated that SARI down-regulates p-STAT1 and STAT1 expression in colon cancer cells, following inhibition of MCP-1/CCR2 axis activation during CAC development. Inverse correlations between SARI expression and macrophage infiltration, MCP-1 expression and p-STAT1 expression were also demonstrated in colon malignant tissues. Collectively, our results prove the inhibition role of SARI in colon cancer formation through regulating TAM infiltration.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/fisiologia , Quimiocina CCL2/metabolismo , Neoplasias Associadas a Colite/prevenção & controle , Colite/complicações , Neoplasias do Colo/prevenção & controle , Inflamação/fisiopatologia , Macrófagos Associados a Tumor/imunologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Quimiocina CCL2/genética , Neoplasias Associadas a Colite/etiologia , Neoplasias Associadas a Colite/metabolismo , Neoplasias Associadas a Colite/patologia , Neoplasias do Colo/etiologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Receptores CCR2/genética , Receptores CCR2/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
11.
BMC Cancer ; 20(1): 42, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31952506

RESUMO

Following publication of the original article [1], the authors reported an error in Fig 5 of this article, graphs presenting FCM and immunofluorescent for CD4T, CD8T and NK cell of the Control Groups (LL2, LL2-irradation, MCS-irradiation) were inadvertently duplicated from another parallel experiment.

12.
Mol Ther ; 27(1): 244-260, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30527756

RESUMO

Immunotherapy based on the immune checkpoint blockade has emerged as the most promising approach for cancer therapy. However, the proportion of colorectal cancer patients who benefit from immunotherapy is small due to the immunosuppressive tumor microenvironment. Hence, combination immunotherapy is an ideal strategy to overcome this limitation. In this study, we developed a novel combination of CSF-1R (colony-stimulating factor 1 receptor) inhibitor (PLX3397), oncolytic viruses, and anti-PD-1 antibody. Our results demonstrated that the triple treatment synergistically conferred significant tumor control and prolonged the survival of mouse models of colon cancer. Approximately 43% and 82% of mice bearing the CT26 and MC38 tumor, respectively, survived long term following the triple treatment. This combination therapy reprogrammed the immunosuppressive tumor microenvironment toward a CD8+ T cell-biased anti-tumor immunity by increasing T cell infiltration in the tumor and augmenting anti-tumor CD8+ T cell function. Our results provide a robust strategy for clinical combination therapy.


Assuntos
Vírus Oncolíticos/fisiologia , Receptor de Morte Celular Programada 1/imunologia , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Aminopiridinas/farmacologia , Animais , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Neoplasias do Colo/metabolismo , Feminino , Citometria de Fluxo , Humanos , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vírus Oncolíticos/genética , Pirróis/farmacologia , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
13.
J Cell Mol Med ; 23(6): 4127-4138, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30920116

RESUMO

Long non-coding RNAs (lncRNAs) have potential applications in clinical diagnosis and targeted cancer therapies. However, the expression profile of lncRNAs in colorectal cancer (CRC) initiation is still unclear. In this study, the expression profiles of lncRNAs and mRNAs were determined by microarray at specific tumour stages in an AOM/DSS-induced primary colon cancer model. The temporal expression of lncRNAs was analysed by K-means clustering. Additionally, weighted correlation network analysis (WGCNA) and gene ontology analysis were performed to construct co-expression networks and establish functions of the identified lncRNAs and mRNAs. Our results suggested that 4307 lncRNAs and 5798 mRNAs are deregulated during CRC initiation. These differential expression genes (DEGs) exhibited a clear correlation with the differential stage of tumour initiation. WGCNA results suggested that a series of hub lncRNAs are involved in regulating cell stemness, colon inflammation, oxidative stress response and cell death at each stage. Among them, lncRNA H19 was up-regulated in colon tumours and correlated with poor patient prognosis. Collectively, we have been the first to demonstrate the temporal expression and function of lncRNAs in CRC initiation. These results provide novel diagnosis and therapy targets for CRC.


Assuntos
Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica/genética , RNA Longo não Codificante/genética , Animais , Morte Celular/genética , Transformação Celular Neoplásica/genética , Perfilação da Expressão Gênica/métodos , Ontologia Genética , Redes Reguladoras de Genes/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estadiamento de Neoplasias/métodos , Estresse Oxidativo/genética , RNA Mensageiro/genética , Regulação para Cima/genética
14.
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
15.
J Cell Mol Med ; 22(12): 6190-6201, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30255547

RESUMO

Hepatocellular carcinoma (HCC) is one of the most common and deadly malignant tumors in the world, especially in China. Follistatin-like protein 5 (FSTL5) is a member of the FSTL family, which is involved in cell proliferation, migration, differentiation, and embryo development. We aimed to investigate the function and underlying mechanism of FSTL5 in HCC. FSTL5 expression was determined by immunohistochemistry staining in a liver cancer tissue microarray (TMA) and the correlation between FSTL5 and the prognosis of HCC patients was analysed. Further proliferation assay, colony formation assay, flow cytometry, and xenograft tumor model were performed to investigate the bioeffects of FSTL5 in HCC in vitro and in vivo. We found that FSTL5 expression was downregulated in HCC tissues and positively correlated with the prognosis of patients with HCC at tumor node metastasis stage I/II. Overexpression of FSTL5 efficiently impaired HCC growth both in vivo and in vitro with an exogenous manner. Mechanistic investigation demonstrated that FSTL5 promoted HCC cell apoptosis in a caspase-dependent manner and regulated Bcl-2 family proteins. These results indicate that FSTL5 may be a potential novel target for HCC treatment, and a biomarker for tumor prognosis.


Assuntos
Carcinoma Hepatocelular/genética , Proteínas Relacionadas à Folistatina/genética , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Idoso , Animais , Apoptose/genética , Carcinoma Hepatocelular/patologia , Caspases/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Xenoenxertos , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Prognóstico
16.
J Cell Mol Med ; 22(2): 1014-1025, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29193791

RESUMO

Interleukin-35 (IL-35), a member of the IL-12 family, functions as a new anti-inflammatory factor involved in arthritis, psoriasis, inflammatory bowel disease (IBD) and other immune diseases. Although IL-35 can significantly prevent the development of inflammation in many diseases, there have been no early studies accounting for the role of IL-35 recombinant protein in IBD and psoriasis. In this study, we assessed the therapeutic potential of IL-35 recombinant protein in three well-known mouse models: the dextransulfate sodium (DSS)-induced colitis mouse model, the keratin14 (K14)-vascular endothelial growth factor A (VEGF-A)-transgenic (Tg) psoriasis mouse model and the imiquimod (IMQ)-induced psoriasis mouse model. Our results indicated that IL-35 recombinant protein can slow down the pathologic process in DSS-induced acute colitis mouse model by decreasing the infiltrations of macrophages, CD4+ T and CD8+ T cells and by promoting the infiltration of Treg cells. Further analysis demonstrated that IL-35 recombinant protein may regulate inflammation through promoting the secretion of IL-10 and inhibiting the expression of pro-inflammatory cytokines such as IL-6, TNF-α and IL-17 in acute colitis model. In addition, lower dose of IL-35 recombinant protein could achieve long-term treatment effects as TNF-α monoclonal antibody did in the psoriasis mouse. In summary, the remarkable therapeutic effects of IL-35 recombinant protein in acute colitis and psoriasis mouse models indicated that IL-35 recombinant protein had a variety of anti-inflammatory effects and was expected to become an effective candidate drug for the treatment of inflammatory diseases.


Assuntos
Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/imunologia , Interleucinas/uso terapêutico , Linfócitos/metabolismo , Psoríase/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Doença Aguda , Animais , Colite/complicações , Colite/tratamento farmacológico , Colite/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Feminino , Humanos , Imiquimode/farmacologia , Imiquimode/uso terapêutico , Doenças Inflamatórias Intestinais/complicações , Doenças Inflamatórias Intestinais/patologia , Interleucinas/farmacologia , Linfócitos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Psoríase/complicações , Psoríase/imunologia , Proteínas Recombinantes/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Stem Cells ; 35(9): 2060-2070, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28514506

RESUMO

Autologous adipose tissue or adipose tissue with additive adipose-derived mesenchymal stem cells (ADSCs) is used in the breast reconstruction of breast cancer patients who undergo mastectomy. ADSCs play an important role in the angiogenesis and adipogenesis, which make it much better than other materials. However, ADSCs may promote residual tumor cells to proliferate or metastasize, and the mechanism is still not fully understood. In this study, we demonstrated that human ADSCs (hADSCs) could facilitate tumor cells growth after co-injection with MCF7 and ZR-75-30 breast cancer cells (BCCs) by promoting angiogenesis, but hADSCs showed limited effect on the growth of MDA-MB-231 BCCs. Intriguingly, compared with ZR-75-30 tumor cells, MCF7 tumor cells were more potentially promoted by hADSCs in the aspects of angiogenesis and proliferation. Consistent with this, cytokine and angiogenesis array analyses showed that after co-injection with hADSCs, the CXCL1 and CXCL8 concentration were significantly increased in MCF7 tumor, but only moderately increased in ZR-75-30 tumor and did not increase in MDA-MB-231 tumor. Furthermore, we found that CXCL1/8 were mainly derived from hADSCs and could increase the migration and tube formation of human umbilical vein endothelial cells (HUVECs) by signaling via their receptors CXCR1 and CXCR2. A CXCR1/2-specific antagonist (SCH527123) attenuated the angiogenesis and tumor growth in vivo. Our findings suggest that CXCL1/8 secreted by hADSCs could promote breast cancer angiogenesis and therefore provide better understanding of safety concerns regarding the clinical application of hADSCs and suggestion in further novel therapeutic options. Stem Cells 2017;35:2060-2070.


Assuntos
Tecido Adiposo/patologia , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Quimiocina CXCL1/metabolismo , Interleucina-8/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neovascularização Patológica/patologia , Animais , Benzamidas/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Ciclobutanos/farmacologia , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Nus , Neovascularização Fisiológica/efeitos dos fármacos , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
19.
J Immunol ; 197(6): 2131-44, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27527600

RESUMO

IL-35 downregulates Th17 cell development and suppresses certain types of autoimmune inflammation such as collagen-induced arthritis and experimental autoimmune uveitis. Psoriasis is thought to be initiated by abnormal interactions between cutaneous keratinocytes and systemic immune cells. However, the role of IL-35 in psoriasis remains unclear. In this study, we assessed IL-35 in three well-known psoriasis models: a human keratinocyte cell line (HaCaT), a keratin 14 (K14)-vascular endothelial growth factor A (VEGF-A)-transgenic (Tg) mouse model, and an imiquimod-induced psoriasis mouse model. First, we found that IL-35 suppressed the expression of IL-6, CXCL8, and S100A7, which are highly upregulated by a mixture of five proinflammatory cytokines in HaCaT. Second, a plasmid coding for the human IL-35 sequence coated with cationic liposomes showed potent immunosuppressive effects on K14-VEGF-A-Tg and imiquimod-induced psoriasis mouse models. In the K14-VEGF-A-Tg model, our results showed that several types of proinflammatory cytokines were significantly reduced, whereas IL-10 was remarkably induced by IL-35. Compared with pcDNA3.1, there was a small number of CD4(+)IL-17(+) T cells and a large number of CD4(+)IL-10(+) and CD4(+)CD25(+)Foxp3(+) T cells in the IL-35 group. Most importantly, we found that IL-35 decreased the total number of macrophages and ratio of M1/M2 macrophages, which has not been reported previously. In addition, compared with dexamethasone, IL-35 showed long-term therapeutic efficacy. In summary, our results strongly indicate that IL-35 plays a potent immunosuppressive role in psoriasis. Thus, IL-35 has potential for development as a new therapeutic strategy for patients with chronic psoriasis and other cutaneous inflammatory diseases.


Assuntos
Citocinas/biossíntese , Inflamação/tratamento farmacológico , Interleucinas/farmacologia , Macrófagos/efeitos dos fármacos , Psoríase/tratamento farmacológico , Animais , Células Cultivadas , Dexametasona/farmacologia , Humanos , Inflamação/imunologia , Interleucina-6/biossíntese , Interleucina-8/biossíntese , Interleucinas/uso terapêutico , Queratinócitos/metabolismo , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Psoríase/imunologia , Proteína A7 Ligante de Cálcio S100 , Proteínas S100/biossíntese , Células Th17/efeitos dos fármacos , Células Th17/fisiologia
20.
J Cell Mol Med ; 21(8): 1555-1571, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28332318

RESUMO

Elevated vascular endothelial growth factor (VEGF) and complement activation are implicated in the pathogenesis of different ocular diseases. The objective of this study was to investigate the hypothesis that dual inhibition of both VEGF and complement activation would confer better protection against ocular inflammation and neovascularization. In this study, we engineered a secreted chimeric VEGF inhibitor domain (VID), a complement inhibitor domain (CID) and a dual inhibitor (ACVP1). Vectors expressing these three inhibitors were constructed and packaged into AAV2 (sextY-F) particles. The expression and secretion of the proteins were validated by Western blot. The effects of these inhibitors expressed from AAV2 vectors were examined in endotoxin-induced uveitis (EIU), experimental autoimmune uveoretinitis (EAU) and choroidal neovascularization (CNV) mouse models. The AAV2 vectors expressing the CID- and ACVP1-attenuated inflammation in EIU and EAU model, whereas the vector expressing VID showed improved retinal structure damaged by EAU, but not affect the infiltration of inflammatory cells in EAU or EIU eyes. Both VID and CID vectors improved laser-induced retinal and choroid/RPE injuries and CNV, whereas ACVP1 vector provided significantly better protection. Our results suggest that gene therapy targeting VEGF and complement components could provide an innovative and long-term strategy for ocular inflammatory and neovascular diseases.


Assuntos
Neovascularização de Coroide/terapia , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/metabolismo , Retinite/terapia , Uveíte/terapia , Animais , Doenças Autoimunes , Corioide/imunologia , Corioide/patologia , Neovascularização de Coroide/genética , Neovascularização de Coroide/imunologia , Neovascularização de Coroide/patologia , Dependovirus/metabolismo , Endotoxinas , Expressão Gênica , Genes Reporter , Vetores Genéticos/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Domínios Proteicos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Retina/imunologia , Retina/patologia , Retinite/genética , Retinite/imunologia , Retinite/patologia , Uveíte/induzido quimicamente , Uveíte/genética , Uveíte/imunologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA