Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 209(7): 1401-1413, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36038289

RESUMO

Myeloid-derived suppressor cells (MDSCs) are pathologically activated neutrophils and monocytes with potent immunosuppressive activity that regulate immune responses in the tumor microenvironment. We identified a novel long noncoding RNA (lncRNA), named as lnc57Rik, in the MDSCs that controls their immunosuppressive functions. Lnc57Rik was induced in in vitro and in vivo inflammatory settings and upregulated the genes related to MDSC-mediated immunosuppression, including Arg-1, NOS2, NOX2, and COX2 Furthermore, Lnc57Rik can not only bind with the C/EBPß isoform liver-enriched activator protein to activate C/EBPß but also with the methyltransferase WD repeat-containing protein 5 that enables the enrichment of histone H3 trimethylated lysine 4 marks on the promoter regions of Arg-1, NOS2, NOX2, and COX2, eventually resulting in their transcriptional activation. Furthermore, the conserved human lnc57Rik has a similar function as murine lnc57Rik Taken together, upregulation of lnc57Rik in the tumor microenvironment promotes the immunosuppressive function of MDSCs.


Assuntos
Células Supressoras Mieloides , Neoplasias , RNA Longo não Codificante , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Microambiente Tumoral/genética
2.
J Immunol ; 204(12): 3400-3415, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32358019

RESUMO

Mixed-lineage leukemia 1 (MLL1), which exerts its H3K4 methyltransferase activity by interacting with WDR5, ASH2L, and RBBP5, plays a pivotal role in regulating hematopoietic stem cell homeostasis. Disrupting the integrity of MLL1-complex has been reported to be associated with acute leukemia. However, the exact role of MLL1-complex in myeloid cells is unknown. In this study, microarray analysis revealed that the core components of the Mll1-complex, Wdr5, Ash2l, and Mll1, were concurrently downregulated by tumor-secreted factors as well as GM-CSF + IL-6 during the accumulation and activation of murine myeloid-derived suppressor cells (MDSCs). These changes were further validated by quantitative RT-PCR and Western blotting both in vitro and in vivo. The expression levels of WDR5 and ASH2L were also significantly decreased in bone marrow MDSCs of lung cancer patients compared with that of healthy controls. Functionally, ectopic expression of Wdr5, Ash2l, and Mll1 (C terminus) reversed the accumulation and function of GM-CSF + IL-6-induced as well as tumor-cocultured polymorphonuclear MDSCs (PMN-MDSCs) by promoting them to differentiate into mature neutrophil-like cells. Mechanistically, GM-CSF + IL-6-activated Stat3 and Cebpß synergistically induced the expression of miR-21a, miR-21b, and miR-181b, and thus inhibited the expression of Wdr5, Ash2l, and Mll1 by targeting to their 3' untranslated regions, respectively. Furthermore, knockdown of these microRNAs also suppressed the expansion and function of GM-CSF + IL-6-induced PMN-MDSCs. Taken together, our findings indicate that the Stat3/Cebpß-miR-21a/b/181b-Mll1-complex axis may play a critical role in PMN-MDSC expansion, activation, and differentiation, and this axis may provide an effectively immunological therapeutic approach for patients with cancer or other immunological diseases.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Homeostase/fisiologia , Tolerância Imunológica/fisiologia , Leucemia Aguda Bifenotípica/metabolismo , MicroRNAs/metabolismo , Células Supressoras Mieloides/metabolismo , Fator de Transcrição STAT3/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Diferenciação Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/genética , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Células HEK293 , Humanos , Interleucina-6/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
3.
J Immunol ; 200(8): 2603-2614, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29531162

RESUMO

Myeloid-derived suppressor cells (MDSCs) are major regulators of immune responses in cancer. Both C/EBP homologous protein (CHOP) and C/EBPß play a critical role in regulating immunosuppressive function of MDSCs. In this study, we identified a novel long noncoding RNA termed as lnc-chop in MDSCs, which may interact with CHOP and the C/EBPß isoform liver-enriched inhibitory protein. The binding of lnc-chop with both CHOP and the C/EBPß isoform liver-enriched inhibitory protein promoted the activation of C/EBPß and upregulated the expression of arginase-1, NO synthase 2, NADPH oxidase 2, and cyclooxygenase-2, which are related to the immunosuppressive function of MDSCs in inflammatory and tumor environments. Additionally, lnc-chop also promoted the enrichment of H3K4me3 on the promoter region of arginase-1, NO synthase 2, NADPH oxidase 2, and cyclooxygenase-2. These findings suggest an important role of lnc-chop in controlling immunosuppressive function of MDSCs in the tumor environment.


Assuntos
Células Supressoras Mieloides/patologia , Neoplasias/genética , Neoplasias/patologia , RNA Longo não Codificante/genética , Fator de Transcrição CHOP/genética , Animais , Arginase/genética , Proteína beta Intensificadora de Ligação a CCAAT/genética , Linhagem Celular , Ciclo-Oxigenase 2/genética , Células HEK293 , Humanos , Terapia de Imunossupressão/métodos , Inflamação/genética , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidase 2/genética , Óxido Nítrico Sintase Tipo II/genética , Regiões Promotoras Genéticas/genética , Microambiente Tumoral/genética
4.
J Immunol ; 198(7): 2712-2720, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28193828

RESUMO

STAT3 plays a critical role in myeloid-derived suppressor cell (MDSC) accumulation and activation. Most studies have probed underlying mechanisms of STAT3 activation. However, epigenetic events involved in STAT3 activation are poorly understood. In this study, we identified several epigenetic-associated proteins such as p66a (Gatad2a), a novel protein transcriptional repressor that might interact with STAT3 in functional MDSCs, by using immunoprecipitation and mass spectrometry. p66a could regulate the phosphorylation and ubiquitination of STAT3. Silencing p66a promoted not only phosphorylation but also K63 ubiquitination of STAT3 in the activated MDSCs. Interestingly, p66a expression was significantly suppressed by IL-6 both in vitro and in vivo during MDSC activation, suggesting that p66a is involved in IL-6-mediated differentiation of MDSCs. Indeed, silencing p66a could promote MDSC accumulation, differentiation, and activation. Tumors in mice injected with p66a small interfering RNA-transfected MDSCs also grew faster, whereas tumors in mice injected with p66a-transfected MDSCs were smaller as compared with the control. Thus, our data demonstrate that p66a may physically interact with STAT3 to suppress its activity through posttranslational modification, which reveals a novel regulatory mechanism controlling STAT3 activation during myeloid cell differentiation.


Assuntos
Epigênese Genética/imunologia , Células Supressoras Mieloides/metabolismo , Neoplasias Experimentais/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Diferenciação Celular/imunologia , Citometria de Fluxo , Regulação da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Imunoprecipitação , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/imunologia , Neoplasias Experimentais/imunologia , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT3/imunologia , Ubiquitinação
5.
J Theor Biol ; 380: 24-39, 2015 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-26002420

RESUMO

Histone modifications (HMs) play an important role in controlling eukaryotic gene expression and next generation sequencing (NGS) has greatly advanced the research on this topic with generating many high-resolution maps for HMs. Here, we use these maps to analyze the relationship between HMs and transcription. By incorporating various segments of genes into analysis without restricting the scope only in the promoter region, we have collected more comprehensive data and captured some details of this process. A position effect of gene regions has been revealed and it can even inverse the property of some HMs from activating to repressing genes such as the cases of H3K4me3, H3K36me3 and H3K14ac. Especially H3K36me3, its dual character on gene transcription makes it able to serve as a criterion to distinguish high and low expressed genes. We also study the general property of different HMs based on the comprehensive data. Using exploratory factor analysis (EFA), we have extracted 4 latent structures underlying the HMs, which are able to represent their activating and repressing effects concisely. These 4 factors have fine properties in the aspects of distinguishing high and low expressed genes, predicting transcription level and identifying genes with unique attributes such as stable RNA generating genes found to have a close relationship with lifespan of organisms here. In summary, while the position effect associated HM peculiarities demonstrate some details of the complex HM regulation network divergently, the common factors catch the nature of the network convergently. This deepens our understanding on the HM-transcription relationship.


Assuntos
Histonas/fisiologia , Transcrição Gênica/fisiologia , Regulação da Expressão Gênica , Histonas/genética
6.
J Immunol ; 190(5): 2437-46, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23355742

RESUMO

Tumors use a wide array of immunosuppressive strategies, such as reducing the longevity and survival of dendritic cells (DCs), to diminish immune responses and limit the effect of immunotherapy. In this study, we found that tumors upregulate the expression of multiple microRNAs (miRNAs), such as miR-16-1, miR-22, miR-155, and miR-503. These tumor-associated miRNAs influenced the survival and longevity of DCs by affecting the expression of multiple molecules that are associated with apoptotic signaling pathways. Specifically, miR-22 targeted YWHAZ to interrupt the PI3K/Akt and MAPK signaling pathways, and miR-503 downregulated Bcl2 expression. The result of the increased expression of miR-22 and miR-503 in the tumor-associated DCs was their reduced survival and longevity. Thus, tumor-associated miRNAs can target multiple intracellular signaling molecules to cause the apoptosis of DCs in the tumor environment. Use of miR-22 and miR-503 as inhibitors may therefore represent a new strategy to improve DC-based immunotherapies against tumors.


Assuntos
Proteínas 14-3-3/genética , Sobrevivência Celular/genética , Células Dendríticas/metabolismo , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transdução de Sinais/imunologia , Proteínas 14-3-3/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/imunologia , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/imunologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , RNA Interferente Pequeno/genética , Transfecção
7.
Gastro Hep Adv ; 3(6): 855-870, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39280926

RESUMO

Colorectal cancer (CRC) is one of the most common cancers worldwide. The occurrence and development of CRC are related to multiple risk factors such as gut microbiota. Indeed, gut microbiota plays an important role in the different phases of colorectal cancers (CRCs) from oncogenesis to metastasis. Some specific bacteria such as Fusobacterium nucleatum (F. nucleatum) associated with CRCs have been found. However, recently identified bile acid and tryptophan metabolites as well as short chain fatty acids (SCFAs), which are derived from gut microbiota, can also exert effects on the CRCs such as that SCFAs directly inhibit CRC growth. Importantly these metabolites also modulate immune responses to affect CRCs. They not only act as tumor inhibiting factor(s) but also promotor(s) in the occurrence, development, and metastasis of CRCs. While gut microbiota metabolites (GMMs) inhibit immunity against CRCs, some of them also improve immune responses to CRCs. Notably, GMMs also potentially affect the shaping of immune-privileged metastatic niches through direct roles or immune cells such as macrophages and myeloid-derived suppressive cells. These findings offer new insights for clinical application of gut microbiota in precise and personalized treatments of CRCs. Here, we will mainly discuss direct and indirect (via immune cells) effects of GMMs, especially SCFAs, bile acid and tryptophan metabolites on the occurrence, development and metastasis of CRCs.

8.
Cancer Lett ; 582: 216582, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-38065401

RESUMO

Tumor cells can evade immune surveillance by expressing immune checkpoint molecule ligands, resulting in effective immune cell inactivation. Immune checkpoint blockades (ICBs) have dramatically improved survival of patients with multiple types of cancers. However, responses to ICB immunotherapy are heterogeneous with lower patient response rates. The advances have established that the gut microbiota can be as a promising target to overcome resistance to ICB immunotherapy. Furthermore, some bacterial species have shown to promote improved responses to ICBs. However, gut microbiota is critical in maintaining gut and systemic immune homeostasis. It not only promotes differentiation and function of immunosuppressive immune cells but also inhibits inflammatory cells via gut microbiota derived products such as short chain fatty acids (SCFAs), tryptophan (Trp) and bile acid (BA) metabolites, which play an important role in tumor immunity. Since the gut microbiota can either inhibit or enhance immune against tumor, it should be a double-edged sword in ICBs against tumor. In this review, we discuss the effects of gut microbiota on immune cells and also tumor cells, especially enhances of gut microbiota on ICB immunotherapy. These discussions can hopefully promote the development of ICB immunotherapy.


Assuntos
Microbioma Gastrointestinal , Neoplasias , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Neoplasias/tratamento farmacológico
9.
Front Immunol ; 15: 1331518, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39229258

RESUMO

One of major breakthroughs in immunotherapy against tumor is from blocking immune checkpoint molecules on tumor and reactive T cells. The development of CTLA-4 and PD-1 blockage antibodies has triggered to search for additional effective therapeutic strategies. This causes recent findings that blocking the interaction of checkpoint molecule NKG2A in NK and CD8 T cells with HLA-E in tumors is effective in defensing tumors. Interestingly, gut microbiota also affects this immune checkpoint immunotherapy against tumor. Gut microbiota such as bacteria can contribute to the regulation of host immune response and homeostasis. They not only promote the differentiation and function of immunosuppressive cells but also the inflammatory cells through the metabolites such as tryptophan (Trp) and bile acid (BA) metabolites as well as short chain fatty acids (SCFAs). These gut microbiota metabolites (GMMs) educated immune cells can affect the differentiation and function of effective CD8 and NK cells. Notably, these metabolites also directly affect the activity of CD8 and NK cells. Furthermore, the expression of CD94/NKG2A in the immune cells and/or their ligand HLA-E in the tumor cells is also regulated by gut microbiota associated immune factors. These findings offer new insights for the clinical application of gut microbiota in precise and/or personalized treatments of tumors. In this review, we will discuss the impacts of GMMs and GMM educated immune cells on the activity of effective CD8 and NK cells and the expression of CD94/NKG2A in immune cells and/or their ligand HLA-E in tumor cells.


Assuntos
Microbioma Gastrointestinal , Antígenos HLA-E , Imunoterapia , Subfamília C de Receptores Semelhantes a Lectina de Células NK , Neoplasias , Humanos , Microbioma Gastrointestinal/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/metabolismo , Imunoterapia/métodos , Animais , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Inibidores de Checkpoint Imunológico/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo
10.
Int J Oncol ; 64(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38551162

RESUMO

Fungi inhabit different anatomic sites in the human body. Advances in omics analyses of host­microbiome interactions have tremendously improved our understanding of the effects of fungi on human health and diseases such as tumors. Due to the significant enrichment of specific fungi in patients with malignant tumors, the associations between fungi and human cancer have attracted an increasing attention in recent years. Indeed, cancer type­specific fungal profiles have been found in different tumor tissues. Importantly, fungi also influence tumorigenesis through multiple factors, such as host immunity and bioactive metabolites. Microbiome interactions, host factors and fungal genetic and epigenetic factors could be involved in fungal enrichment in tumor tissues and/or in the conversion from a commensal fungus to a pathogenic fungus. Exploration of the interactions of fungi with the bacterial microbiome and the host may enable them to be a target for cancer diagnosis and treatment. In the present review, the associations between fungi and human cancer, cancer type­specific fungal profiles and the mechanisms by which fungi cause tumorigenesis were discussed. In addition, possible factors that can lead to the enrichment of fungi in tumor tissues and/or the conversion of commensal fungi to pathogenic fungi, as well as potential therapeutic and preventive strategies for tumors based on intratumoral fungi were summarized.


Assuntos
Neoplasias , Simbiose , Humanos , Fungos/genética , Bactérias , Carcinogênese/genética
11.
Immunohorizons ; 8(1): 35-46, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38189743

RESUMO

Inflammasome NLRC4 (NLR family CARD domain containing 4) can protect mucosal barriers such as intestine from invading bacterial pathogens. However, it was incompletely clear how NLRC4 was activated in intestinal epithelial cells. In this study, we demonstrated that LNCGM1082 could mediate the activation of NLRC4 via binding NLRC4 with protein kinase C (PKC)δ. LNCGM1082 knockout (KO) mice had reduced resistance against Salmonella Typhimurium infection, as well as impaired expulsion of infected gut epithelial cells and release of IL-18 upon exposure to S. Typhimurium. Similar to NLRC4 KO and PKCδ knockdown gut organoids, there also was impaired expulsion of gut epithelial cells and release of IL-18 in LNCGM1082 KO gut organoids. Furthermore, there also was reduced activation of caspase-1 and caspase-8 in these LNCGM1082 KO, NLRC4 KO, and PKCδ knockdown gut organoids upon exposure to S. Typhimurium. Our results show that LNCGM1082 in the ICEs plays a critical role in mediating activation of NLRC4 through binding NLRC4 and PKCδ and promoting expulsion of infected epithelial cells and release of IL-18 upon exposure to bacteria such as S. Typhimurium.


Assuntos
Células Epiteliais , Interleucina-18 , Animais , Camundongos , Interleucina-18/genética , Inflamassomos , Camundongos Knockout
12.
Cancer Lett ; 598: 217115, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39025428

RESUMO

Gut microbiota contributes to the homeostasis of immune system and is related to various diseases such as tumorigenesis. Ferroptosis, a new type of cell death, is also involved in the disease pathogenesis. Recent studies have found the correlations of gut microbiota mediated ferroptosis and immune cell death. Gut microbiota derived immunosuppressive metabolites, which can promote differentiation and function of immune cells, tend to inhibit ferroptosis through their receptors, whereas inflammatory metabolites from gut microbiota also affect the differentiation and function of immune cells and their ferroptosis. Thus, it is possible for gut microbiota to regulate immune cell ferroptosis. Indeed, gut microbiota metabolite receptor aryl hydrocarbon receptor (AhR) can affect ferroptosis of intestinal intraepithelial lymphocytes, leading to disease pathogenesis. Since immune cell ferroptosis in tumor microenvironment (TME) affects the occurrence and development of tumor, the modulation of gut microbiota in these cell ferroptosis might influence on the tumorigenesis, and also immunotherapy against tumors. Here we will summarize the recent advance of ferroptosis mediated by gut microbiota metabolites, which potentially acts as regulator(s) on immune cells in TME for therapy against tumor.


Assuntos
Ferroptose , Microbioma Gastrointestinal , Imunoterapia , Neoplasias , Microambiente Tumoral , Ferroptose/imunologia , Humanos , Microbioma Gastrointestinal/imunologia , Imunoterapia/métodos , Microambiente Tumoral/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/microbiologia , Animais , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores de Hidrocarboneto Arílico/imunologia , Carcinogênese/imunologia
13.
Int J Biol Sci ; 20(7): 2476-2490, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38725863

RESUMO

Peristaltic movements in gut are essential to propel ingested materials through the gastrointestinal tract. Intestinal resident macrophages play an important role in this physiological function through protecting enteric neurons. However, it is incompletely clear how individuals maintain the homeostasis of gut motility. Here we found that NLRP3 is a critical factor in controlling loss of muscularis resident macrophages (MMs), and demonstrate that MMs are involved in the homeostasis of excitatory neurons such as choline acetyltransferase (ChAT)+ and vesicular glutamate transporter 2 (VGLUT2)+ but not inhibitory neuronal nitric oxide synthase (nNOS)+ neurons. NLRP3 knockout (KO) mice had enhanced gut motility and increased neurons, especially excitatory ChAT+ and VGLUT2+ neurons. Single cell analyses showed that there had increased resident macrophages, especially MMs in NLRP3 KO mice. The MM proportion in the resident macrophages was markedly higher than those in wild-type (WT) or caspase 1/11 KO mice. Deletion of the MMs and transplantation of the NLRP3 KO bone marrow cells showed that survival of the gut excitatory ChAT+ and VGLUT2+ neurons was dependent on the MMs. Gut microbiota metabolites ß-hydroxybutyrate (BHB) could promote gut motility through protecting MMs from pyroptosis. Thus, our data suggest that MMs regulated by NLRP3 maintain the homeostasis of excitatory neurons.


Assuntos
Motilidade Gastrointestinal , Macrófagos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Neurônios , Camundongos , Trato Gastrointestinal/citologia , Trato Gastrointestinal/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Masculino , Feminino , Animais , Camundongos Knockout , Plexo Mientérico/citologia , Plexo Mientérico/metabolismo , Colo/citologia , Colo/metabolismo , Mucosa/citologia , Mucosa/metabolismo
14.
Gut Microbes ; 16(1): 2377576, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39068517

RESUMO

The global incidence and prevalence of inflammatory bowel disease (IBD) are gradually increasing. A high-fat diet (HFD) is known to disrupt intestinal homeostasis and aggravate IBD, yet the underlying mechanisms remain largely undefined. Here, a positive correlation between dietary fat intake and disease severity in both IBD patients and murine colitis models is observed. A HFD induces a significant decrease in indole-3-acetic acid (IAA) and leads to intestinal barrier damage. Furthermore, IAA supplementation enhances intestinal mucin sulfation and effectively alleviates colitis. Mechanistically, IAA upregulates key molecules involved in mucin sulfation, including 3'-phosphoadenosine 5'-phosphosulfate synthase 2 (Papss2) and solute carrier family 35 member B3 (Slc35b3), the synthesis enzyme and the transferase of 3'-phosphoadenosine-5'-phosphosulfate (PAPS), via the aryl hydrocarbon receptor (AHR). More importantly, AHR can directly bind to the transcription start site of Papss2. Oral administration of Lactobacillus reuteri, which can produce IAA, contributes to protecting against colitis and promoting mucin sulfation, while the modified L. reuteri strain lacking the iaaM gene (LactobacillusΔiaaM) and the ability to produce IAA fail to exhibit such effects. Overall, IAA enhances intestinal mucin sulfation through the AHR-Papss2-Slc35b3 pathway, contributing to the protection of intestinal homfeostasis.


A HFD can lead to the development of colitis by disrupting tryptophan metabolism in the gut microbiome and lowering levels of IAA. Supplementation with IAA has been shown to alleviate colitis in mice and improve intestinal barrier function. It is believed that IAA may activate the AHR to upregulate the expression of Papss2 and Slc35b3, promoting sulfation modification of mucins and protecting the intestinal barrier. HFD, high-fat diet; AHR, aryl hydrocarbon receptor; IAA, indole-3-acetic acid; Papss2, 3'-phosphoadenosine 5'-phosphosulfate synthase 2; Slc35b3, solute carrier family 35 member B3.


Assuntos
Microbioma Gastrointestinal , Homeostase , Ácidos Indolacéticos , Mucosa Intestinal , Mucinas , Animais , Humanos , Camundongos , Microbioma Gastrointestinal/efeitos dos fármacos , Mucinas/metabolismo , Ácidos Indolacéticos/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Camundongos Endogâmicos C57BL , Colite/microbiologia , Colite/metabolismo , Colite/induzido quimicamente , Limosilactobacillus reuteri/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Dieta Hiperlipídica/efeitos adversos , Masculino , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Modelos Animais de Doenças
15.
Microbiome ; 12(1): 4, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38172943

RESUMO

BACKGROUND: The overgrowth of Desulfovibrio, an inflammation promoting flagellated bacteria, has been found in ulcerative colitis (UC) patients. However, the molecular mechanism in promoting colitis remains unestablished. METHODS: The relative abundance Desulfovibrio vulgaris (D. vulgaris) in stool samples of UC patients was detected. Mice were treated with dextran sulfate sodium to induce colitis with or without administration of D. vulgaris or D. vulgaris flagellin (DVF), and the severity of colitis and the leucine-rich repeat containing 19 (LRRC19) signaling were assessed. The interaction between DVF and LRRC19 was identified by surface plasmon resonance and intestinal organoid culture. Lrrc19-/- and Tlr5-/- mice were used to investigate the indispensable role of LRRC19. Finally, the blockade of DVF-LRRC19 interaction was selected through virtual screening and the efficacy in colitis was assessed. RESULTS: D. vulgaris was enriched in fecal samples of UC patients and was correlated with the disease severity. D. vulgaris or DVF treatment significantly exacerbated colitis in germ-free mice and conventional mice. Mechanistically, DVF could interact with LRRC19 (rather than TLR5) in colitis mice and organoids, and then induce the production of pro-inflammatory cytokines. Lrrc19 knockdown blunted the severity of colitis. Furthermore, typhaneoside, a blockade of binding interfaces, blocked DVF-LRRC19 interaction and dramatically ameliorated DVF-induced colitis. CONCLUSIONS: D. vulgaris could promote colitis through DVF-LRRC19 interaction. Targeting DVF-LRRC19 interaction might be a new therapeutic strategy for UC therapy. Video Abstract.


Assuntos
Colite Ulcerativa , Colite , Desulfovibrio vulgaris , Humanos , Camundongos , Animais , Receptor 5 Toll-Like/metabolismo , Receptor 5 Toll-Like/uso terapêutico , Desulfovibrio vulgaris/metabolismo , Colite/induzido quimicamente , Colite/metabolismo , Colite Ulcerativa/microbiologia , Inflamação/metabolismo , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Colo/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/uso terapêutico
16.
Biochem Biophys Res Commun ; 434(1): 28-34, 2013 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-23545260

RESUMO

Toll-like receptors (TLRs) are important initiators in innate immune responses against pathogenic microbes such as viruses, intracellular bacteria or parasites. Although the innate immune system is designed to fight infectious pathogens, excessive activation of TLR signaling may lead to unwarranted inflammation with hazardous outcomes. Mechanisms of restraining excessive inflammation and controlling homeostasis for innate immunity are the focus of intense study. Here we showed that LRRC33, a novel member of leucine-rich repeat (LRR) protein family, plays a critical role in desensitizing TLR signaling. LRRC33 is TLR homolog that contains 17 putative LRRs in the extracellular region but lacks a cytoplasmic Toll/IL-1 receptor (TIR) domain. Expression of LRRC33 appears to be ubiquitous with high level of expression found in bone marrow, thymus, liver, lung, intestine and spleen. The LRRs of LRRC33 is required for the interaction with TLR and its inhibitory effect on NF-κB and AP-1 activation as well as cytokine production. Our study sheds new insight into the TLR signaling and inflammatory response in development and human diseases.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Imunidade Inata , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas/química , Proteínas/fisiologia , Receptores Toll-Like/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Células HEK293 , Humanos , Imunidade Inata/genética , Proteínas de Ligação a TGF-beta Latente , Proteínas de Repetições Ricas em Leucina , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , NF-kappa B/genética , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Proteínas/genética , Proteínas Modificadoras da Atividade de Receptores , Sequências Repetitivas de Aminoácidos/genética , Sequências Repetitivas de Aminoácidos/imunologia , Receptores Toll-Like/genética , Receptores Toll-Like/fisiologia , Células U937
17.
Biochem Biophys Res Commun ; 434(4): 760-6, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23583404

RESUMO

Activation of ß-catenin and PI3K pathways are crucial for the oncogenesis of colorectal cancer (CRC). It remains controversial whether these two pathways function independently or cooperatively in the development and progression of CRC. We showed previously that ß-catenin inhibited NF-κB activation by interacting with p65 and this inhibitory interaction involved an unidentified cellular protein. In this study, we found that the PI3K effect on NF-κB activity is dependent on the level of ß-catenin in CRC cells. PI3K promoted NF-κB activity in the ß-catenin-low RKO cells; whereas it inhibited NF-κB activity in the ß-catenin-high HCT116, DLD-1, and SW480 cells. We showed that PI3K is required for the physical interaction and functional inhibition of NF-κB by ß-catenin. Inhibition of PI3K released NF-κB suppression in ß-catenin-high CRC cells, which conferred these cells with susceptibility to TNFα- and Fas-induced apoptosis. This is consistent with the observation showing that the level of ß-catenin and activated Akt are both inversely correlated with the expression of Fas, a downstream target of NF-κB, in CRC specimens. Mechanistically, the PI3K subunit p85 formed a complex with ß-catenin and NF-κB. Inhibition of PI3K disrupted the complex formation, leading to NF-κB activation. Our study not only provides new insight into the cross-talk among PI3K, ß-catenin and NF-κB signaling pathways but also indicates that targeting PI3K may yield therapeutic efficacy in treating ß-catenin-high CRC.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/metabolismo , beta Catenina/metabolismo , Androstadienos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Cromonas/farmacologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Células HCT116 , Humanos , Microscopia de Fluorescência , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição RelA/genética , Fator de Necrose Tumoral alfa/farmacologia , Wortmanina , beta Catenina/genética , Receptor fas/metabolismo
18.
J Immunol ; 186(8): 4716-24, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21383238

RESUMO

Myeloid-derived suppressor cells (MDSCs) were one of the major components of the immune suppressive network. STAT3 has an important role in regulating the suppressive potential of MDSCs. In this study, we found that the expression of STAT3 could be modulated by both miR-17-5p and miR-20a. The transfection of miR-17-5p or miR-20a remarkably reduces the expression of reactive oxygen species and the production of H(2)O(2), which are regulated by STAT3. MDSCs transfected with miR-17-5p or miR-20a are less able to suppress Ag-specific CD4 and CD8 T cells. Importantly, both miR-17-5p and miR-20a alleviate the suppressive function of MDSCs in vivo. The expression of miR-17-5p and miR-20a in tumor-associated MDSCs was found to be lower than in Gr1(+)CD11b(+) cells isolated from the spleens of disease-free mice. Tumor-associated factor downregulates the expression of both miR-17-5p and miR-20a. The modulation of miR-17-5p and miR-20a expression may be important for the process by which patients with a tumor can overcome the immune tolerance mediated by MDSCs. Our results suggest that miR-17-5p and miR-20a could potentially be used for immunotherapy against diseases, especially cancer, by blocking STAT3 expression.


Assuntos
MicroRNAs/genética , Células Mieloides/imunologia , Neoplasias Experimentais/imunologia , Fator de Transcrição STAT3/genética , Regiões 3' não Traduzidas/genética , Animais , Ligação Competitiva , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Regulação da Expressão Gênica , Células HEK293 , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , MicroRNAs/metabolismo , Mutação , Células Mieloides/metabolismo , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
19.
Front Immunol ; 14: 1127743, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37256134

RESUMO

Bile acids (BAs) as cholesterol-derived molecules play an essential role in some physiological processes such as nutrient absorption, glucose homeostasis and regulation of energy expenditure. They are synthesized in the liver as primary BAs such as cholic acid (CA), chenodeoxycholic acid (CDCA) and conjugated forms. A variety of secondary BAs such as deoxycholic acid (DCA) and lithocholic acid (LCA) and their derivatives is synthesized in the intestine through the involvement of various microorganisms. In addition to essential physiological functions, BAs and their metabolites are also involved in the differentiation and functions of innate and adaptive immune cells such as macrophages (Macs), dendritic cells (DCs), myeloid derived suppressive cells (MDSCs), regulatory T cells (Treg), Breg cells, T helper (Th)17 cells, CD4 Th1 and Th2 cells, CD8 cells, B cells and NKT cells. Dysregulation of the BAs and their metabolites also affects development of some diseases such as inflammatory bowel diseases. We here summarize recent advances in how BAs and their metabolites maintain gut and systemic homeostasis, including the metabolism of the BAs and their derivatives, the role of BAs and their metabolites in the differentiation and function of immune cells, and the effects of BAs and their metabolites on immune-associated disorders.


Assuntos
Ácidos e Sais Biliares , Microbioma Gastrointestinal , Ácidos e Sais Biliares/metabolismo , Ácido Cólico/metabolismo , Ácido Cólico/farmacologia , Fígado/metabolismo , Homeostase
20.
Cancer Lett ; 569: 216302, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37451425

RESUMO

A variety of bacteria, viruses, fungi, protists, archaea and protozoa coexists within the mammalian gastrointestinal (GI) tract such as that fungi are detectable in all intestinal and colon segments in almost all healthy adults. Although fungi can cause infectious diseases, they are also related to gut and systemic homeostasis. Importantly, through transformation of different forms such as from yeast to hyphae, interaction among gut microbiota such as fungal and bacterial interaction, host factors such as immune and host derived factors, and fungus genetic and epigenetic factors, fungi can be transformed from commensal into pathogenic lifestyles. Recent studies have shown that fungi play a significant role in the occurrence and development of tumors such as colorectal cancer. Indeed, evidences have shown that multiple species of different fungi exist in different tumors. Studies have also demonstrated that fungi are related to the occurrence and development of tumors, and also survival of patients. Here we summarize recent advances in the transformation of fungi from commensal into pathogenic lifestyles, and the effects of gut pathogenic fungi on the occurrence and development of tumors such as colorectal and pancreatic cancers.


Assuntos
Microbioma Gastrointestinal , Micobioma , Neoplasias , Adulto , Animais , Humanos , Fungos , Trato Gastrointestinal , Bactérias/genética , Mamíferos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA