Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sensors (Basel) ; 24(8)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38676084

RESUMEN

The maturity of fruits and vegetables such as tomatoes significantly impacts indicators of their quality, such as taste, nutritional value, and shelf life, making maturity determination vital in agricultural production and the food processing industry. Tomatoes mature from the inside out, leading to an uneven ripening process inside and outside, and these situations make it very challenging to judge their maturity with the help of a single modality. In this paper, we propose a deep learning-assisted multimodal data fusion technique combining color imaging, spectroscopy, and haptic sensing for the maturity assessment of tomatoes. The method uses feature fusion to integrate feature information from images, near-infrared spectra, and haptic modalities into a unified feature set and then classifies the maturity of tomatoes through deep learning. Each modality independently extracts features, capturing the tomatoes' exterior color from color images, internal and surface spectral features linked to chemical compositions in the visible and near-infrared spectra (350 nm to 1100 nm), and physical firmness using haptic sensing. By combining preprocessed and extracted features from multiple modalities, data fusion creates a comprehensive representation of information from all three modalities using an eigenvector in an eigenspace suitable for tomato maturity assessment. Then, a fully connected neural network is constructed to process these fused data. This neural network model achieves 99.4% accuracy in tomato maturity classification, surpassing single-modal methods (color imaging: 94.2%; spectroscopy: 87.8%; haptics: 87.2%). For internal and external maturity unevenness, the classification accuracy reaches 94.4%, demonstrating effective results. A comparative analysis of performance between multimodal fusion and single-modal methods validates the stability and applicability of the multimodal fusion technique. These findings demonstrate the key benefits of multimodal fusion in terms of improving the accuracy of tomato ripening classification and provide a strong theoretical and practical basis for applying multimodal fusion technology to classify the quality and maturity of other fruits and vegetables. Utilizing deep learning (a fully connected neural network) for processing multimodal data provides a new and efficient non-destructive approach for the massive classification of agricultural and food products.


Asunto(s)
Frutas , Redes Neurales de la Computación , Solanum lycopersicum , Solanum lycopersicum/crecimiento & desarrollo , Solanum lycopersicum/fisiología , Frutas/crecimiento & desarrollo , Aprendizaje Profundo , Espectroscopía Infrarroja Corta/métodos , Color
2.
Nat Commun ; 15(1): 2820, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38561332

RESUMEN

RORγt+ group 3 innate lymphoid cells (ILC3s) are essential for intestinal homeostasis. Dysregulation of ILC3s has been found in the gut of patients with inflammatory bowel disease and colorectal cancer, yet the specific mechanisms still require more investigation. Here we observe increased ß-catenin in intestinal ILC3s from inflammatory bowel disease and colon cancer patients compared with healthy donors. In contrast to promoting RORγt expression in T cells, activation of Wnt/ß-catenin signaling in ILC3s suppresses RORγt expression, inhibits its proliferation and function, and leads to a deficiency of ILC3s and subsequent intestinal inflammation in mice. Activated ß-catenin and its interacting transcription factor, TCF-1, cannot directly suppress RORγt expression, but rather alters global chromatin accessibility and inhibits JunB expression, which is essential for RORγt expression in ILC3s. Together, our findings suggest that dysregulated Wnt/ß-catenin signaling impairs intestinal ILC3s through TCF-1/JunB/RORγt regulation, further disrupting intestinal homeostasis, and promoting inflammation and cancer.


Asunto(s)
Enfermedades Inflamatorias del Intestino , beta Catenina , Humanos , Ratones , Animales , beta Catenina/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Inmunidad Innata , Linfocitos/metabolismo , Vía de Señalización Wnt , Enfermedades Inflamatorias del Intestino/genética , Inflamación
3.
Mucosal Immunol ; 17(2): 257-271, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38340986

RESUMEN

Chemotherapy and radiotherapy frequently lead to intestinal damage. The mechanisms governing the repair or regeneration of intestinal damage are still not fully elucidated. Intraepithelial lymphocytes (IELs) are the primary immune cells residing in the intestinal epithelial layer. However, whether IELs are involved in intestinal epithelial injury repair remains unclear. Here, we found that IELs rapidly infiltrated the intestinal crypt region and are crucial for the recovery of the intestinal epithelium post-chemotherapy. Interestingly, IELs predominantly promoted intestinal regeneration by modulating the proliferation of transit-amplifying (TA) cells. Mechanistically, the expression of CD160 on IELs allows for interaction with herpes virus entry mediator (HVEM) on the intestinal epithelium, thereby activating downstream nuclear factor kappa (NF-κB) signaling and further promoting intestinal regeneration. Deficiency in either CD160 or HVEM resulted in reduced proliferation of intestinal progenitor cells, impaired intestinal damage repair, and increased mortality following chemotherapy. Remarkably, the adoptive transfer of CD160-sufficient IELs rescued the Rag1 deficient mice from chemotherapy-induced intestinal inflammation. Overall, our study underscores the critical role of IELs in intestinal regeneration and highlights the potential applications of targeting the CD160-HVEM axis for managing intestinal adverse events post-chemotherapy and radiotherapy.


Asunto(s)
Linfocitos Intraepiteliales , Receptores Inmunológicos , Animales , Ratones , Receptores Inmunológicos/metabolismo , Linfocitos Intraepiteliales/metabolismo , Transducción de Señal , Intestinos , Mucosa Intestinal/metabolismo , Regeneración
4.
Cell Rep Med ; 5(1): 101355, 2024 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-38194971

RESUMEN

Despite the encouraging efficacy of anti-PD-1/PD-L1 immunotherapy in microsatellite-instability-high/deficient mismatch repair (MSI-H/dMMR) advanced gastrointestinal cancer, many patients exhibit primary or acquired resistance. Using multi-omics approaches, we interrogate gut microbiome, blood metabolome, and cytokines/chemokines of patients with MSI-H/dMMR gastrointestinal cancer (N = 77) at baseline and during the treatment. We identify a number of microbes (e.g., Porphyromonadaceae) and metabolites (e.g., arginine) highly associated with primary resistance to immunotherapy. An independent validation cohort (N = 39) and mouse model are used to further confirm our findings. A predictive machine learning model for primary resistance is also built and achieves an accuracy of 0.79 on the external validation set. Furthermore, several microbes are pinpointed that gradually changed during the process of acquired resistance. In summary, our study demonstrates the essential role of gut microbiome in drug resistance, and this can be utilized as a preventative diagnosis tool and therapeutic target in the future.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Colorrectales , Microbioma Gastrointestinal , Neoplasias Gastrointestinales , Síndromes Neoplásicos Hereditarios , Animales , Ratones , Humanos , Ecosistema , Microbioma Gastrointestinal/genética , Multiómica , Mutación , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/terapia , Inmunoterapia , Repeticiones de Microsatélite
5.
FEBS J ; 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37994218

RESUMEN

RORγt+ group 3 innate lymphoid cells (ILC3s), the innate counterpart of Th17 cells, are enriched in the mucosal area and lymphoid tissues. ILC3s interact with a variety of cells through their effector molecules and play an important role in the host defense against a spectrum of infections. Recent studies suggest that the extensive crosstalk between ILC3s and adaptive immune cells, especially T cells, is essential for maintaining tissue homeostasis. Here we discuss recent advances in the crosstalk between ILC3s and adaptive immune responses in multiple tissues and diseases. Understanding how ILC3s engage with adaptive immune cells will enhance our comprehension of diseases and facilitate the identification of novel therapeutic targets.

6.
Clin Transl Med ; 13(7): e1312, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37381590

RESUMEN

BACKGROUND: Common treatments for metastatic/unresectable HER2-negative gastric cancer include chemotherapy, immune checkpoint inhibitor monotherapy and chemotherapy plus immune checkpoint inhibitor. However, significant drug resistance exists regardless of the treatment regimen. METHODS: Patients with metastatic/unresectable HER2-negative gastric/gastroesophageal junction adenocarcinoma were enrolled. All patients were divided into three groups according to the treatment regimen and were further divided into responders and non-responders according to efficacy evaluation. Metagenomics sequencing were performed to analyze gut microbiome signature of patients receiving different treatments at baseline and throughout treatment. RESULTS: One hundred seventeen patients with HER2-negative advanced gastric or gastroesophageal junction adenocarcinoma receiving chemotherapy alone, anti PD-1/PD-L1 immunotherapy alone or combined regimen were included in this study. Microbiome signatures related to clinical response are distinct among the three treatment groups. Among which, 14, 8 and 13 species were significantly different between responders and non-responders in immunotherapy, immunotherapy plus chemotherapy and chemotherapy group, respectively. Patients with higher relative abundance of Lactobacillus possessed higher microbiome diversity and significantly better response to anti-PD-1/PD-L1 immunotherapy and had a trend to achieve better progression-free survival. Another cohort of 101 patients has been used as an external validation set to confirm the stability and reliability of these findings. CONCLUSIONS: Gut microbiome affects response of treatments in HER2-negative advanced gastric cancer in a treatment-specific way, immunotherapy plus chemotherapy did not equal to a simple superposition of immunotherapy and chemotherapy. Lactobacillus is expected to become a novel choice as an adjuvant agent in promoting the efficacy of immunotherapy in gastric cancer.


Asunto(s)
Adenocarcinoma , Microbioma Gastrointestinal , Neoplasias Gástricas , Humanos , Microbioma Gastrointestinal/genética , Antígeno B7-H1/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Inhibidores de Puntos de Control Inmunológico , Reproducibilidad de los Resultados , Lactobacillus
7.
Adv Sci (Weinh) ; 9(30): e2202706, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36031409

RESUMEN

Emerging evidence emphasizes the functional impacts of host microbiome on the etiopathogenesis of autoimmune diseases, including rheumatoid arthritis (RA). However, there are limited mechanistic insights into the contribution of microbial biomolecules especially microbial peptides toward modulating immune homeostasis. Here, by mining the metagenomics data of tonsillar microbiome, a deficiency of the encoding genes of lantibiotic peptides salivaricins in RA patients is identified, which shows strong correlation with circulating immune cells. Evidence is provided that the salivaricins exert immunomodulatory effects in inhibiting T follicular helper (Tfh) cell differentiation and interleukin-21 (IL-21) production. Mechanically, salivaricins directly bind to and induce conformational changes of IL-6 and IL-21 receptors, thereby inhibiting the bindings of IL-6 and IL-21 to their receptors and suppressing the downstream signaling pathway. Finally, salivaricin administration exerts both prophylactic and therapeutic effects against experimental arthritis in a murine model of RA. Together, these results provide a mechanism link of microbial peptides-mediated immunomodulation.


Asunto(s)
Artritis Reumatoide , Bacteriocinas , Microbiota , Tonsila Palatina , Receptores de Interleucina-21 , Receptores de Interleucina-6 , Animales , Humanos , Ratones , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Bacteriocinas/uso terapéutico , Interleucina-6/metabolismo , Receptores de Interleucina-21/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Tonsila Palatina/microbiología , Receptores de Interleucina-6/metabolismo
8.
Sensors (Basel) ; 22(13)2022 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-35808359

RESUMEN

To study the dynamic changes of nutrient consumption and aflatoxin B1 (AFB1) accumulation in peanut kernels with fungal colonization, macro hyperspectral imaging technology combined with microscopic imaging was investigated. First, regression models to predict AFB1 contents from hyperspectral data ranging from 1000 to 2500 nm were developed and the results were compared before and after data normalization with Box-Cox transformation. The results indicated that the second-order derivative with a support vector regression (SVR) model using competitive adaptive reweighted sampling (CARS) achieved the best performance, with RC2 = 0.95 and RV2 = 0.93. Second, time-lapse microscopic images and spectroscopic data were captured and analyzed with scanning electron microscopy (SEM), transmission electron microscopy (TEM), and synchrotron radiation-Fourier transform infrared (SR-FTIR) microspectroscopy. The time-lapse data revealed the temporal patterns of nutrient loss and aflatoxin accumulation in peanut kernels. The combination of macro and micro imaging technologies proved to be an effective way to detect the interaction mechanism of toxigenic fungus infecting peanuts and to predict the accumulation of AFB1 quantitatively.


Asunto(s)
Aflatoxina B1 , Aflatoxinas , Aflatoxina B1/análisis , Aflatoxinas/análisis , Arachis/química , Arachis/microbiología , Contaminación de Alimentos/análisis , Análisis Espectral
9.
J Leukoc Biol ; 112(4): 931-938, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35657091

RESUMEN

According to the Correa model, the intestinal-type gastric cancer (GC) is preceded by premalignant lesions, including chronic gastritis, intestinal metaplasia and dysplasia. However, the dynamic change of innate and adaptive immune response during this process has not been studied comprehensively. In this study, we performed a comprehensive and trajectory analysis of circulating innate lymphoid cells (ILCs) and adaptive Th lymphocytes subtypes in patients spanning a cascade of gastric lesions. Increased circulating ILC2s frequency was found in the gastritis, premalignant stage and GC group, whereas further decreased ILC2s were detected in the GC group compared with the premalignant group. Moreover, ILC3s level was higher in both gastritis, premalignant lesion and GC stage, compared with healthy controls. Furthermore, up-regulated T follicular helper (Tfh) cell proportions were detected in the gastritis and premalignant process. In conclusion, by analyzing the circulating ILCs and Th cells frequency and the key cytokine production or immunoglobulin level, we demonstrated the potential involvement of ILC3 and Tfh in the gastric diseases. These findings will help to understand the immunologic mechanisms in both GC and the premalignant process and contribute to serve potential therapeutic targets to prevent the GC development.


Asunto(s)
Gastritis Atrófica , Gastritis , Lesiones Precancerosas , Citocinas , Gastritis Atrófica/patología , Humanos , Inmunidad Innata , Inmunoglobulinas , Linfocitos , Lesiones Precancerosas/patología
11.
Hepatology ; 76(4): 1013-1029, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35243668

RESUMEN

BACKGROUND AND AIMS: Innate lymphoid cells (ILCs) are tissue-resident lymphocytes that play critical roles in cytokine-mediated regulation of homeostasis and inflammation. However, relationships between their immune phenotypic characteristics and HCC remain largely unexplored. APPROACH AND RESULTS: We performed single-cell RNA sequencing analysis on sorted hepatic ILC cells from human patients with HCC and validated using flow cytometry, multiplex immunofluorescence staining, and functional experiments. Moreover, we applied selection strategies to enrich ILC populations in HCC samples to investigate the effects of B cells on the immune reaction of inducible T cell costimulator (ICOS)+ ILC2 cells. Dysregulation of ILCs was manifested by the changes in cell numbers or subset proportions in HCC. Seven subsets of 3433 ILCs were identified with unique properties, of which ICOS+ ILC2a were preferentially enriched in HCC and correlated with poor prognosis. Mechanistically, we report that B cells, particularly resting naïve B cells, have a previously unrecognized function that is involved in inflammatory differentiation of ILC2 cells. B cell-derived ICOSL signaling was responsible for exacerbating inflammation through the increased production of IL-13 in ICOS+ ILC2a cells. Heat shock protein 70 (HSP70) genes Heat Shock Protein Family A Member 1A (HSPA1A) and Heat Shock Protein Family A Member 1B (HSPA1B) were highly expressed in ILC2s in late-stage HCC, and targeting to ICOS and its downstream effector HSP70 in ILC2s suppressed tumor growth and remodeled the immunosuppressive tumor microenvironment. CONCLUSIONS: This in-depth understanding sheds light on B cell-driven innate type 2 inflammation and provides a potential strategy for HCC immunotherapy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/metabolismo , Citocinas/metabolismo , Proteínas HSP70 de Choque Térmico , Proteínas de Choque Térmico , Humanos , Inmunidad Innata , Inflamación/metabolismo , Interleucina-13/metabolismo , Neoplasias Hepáticas/metabolismo , Linfocitos , Fenotipo , Microambiente Tumoral
12.
Cell Rep ; 38(10): 110492, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35263601

RESUMEN

Immune checkpoint inhibitor (ICI) therapy is generating remarkable responses in individuals with cancer, but only a small portion of individuals with breast cancer respond well. Here we report that tumor-derived Jagged1 is a key regulator of the tumor immune microenvironment. Jagged1 promotes tumorigenesis in multiple spontaneous mammary tumor models. Through Jagged1-induced Notch activation, tumor cells increase expression and secretion of multiple cytokines to help recruit macrophages into the tumor microenvironment. Educated macrophages crosstalk with tumor-infiltrating T cells to inhibit T cell proliferation and tumoricidal activity. In individuals with triple-negative breast cancer, a high expression level of Jagged1 correlates with increased macrophage infiltration and decreased T cell activity. Co-administration of an ICI PD-1 antibody with a Notch inhibitor significantly inhibits tumor growth in breast cancer models. Our findings establish a distinct signaling cascade by which Jagged1 promotes adaptive immune evasion of tumor cells and provide several possible therapeutic targets.


Asunto(s)
Evasión Inmune , Neoplasias de la Mama Triple Negativas , Humanos , Macrófagos/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/metabolismo , Microambiente Tumoral
13.
Food Chem ; 382: 132340, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35139463

RESUMEN

The dynamics mechanisms regulating the growth and AFB1 production of Aspergillus flavus during its interactions with maize kernels remain unclear. In this study, shortwave infrared hyperspectral imaging (SWIR-HSI) and synchrotron radiation Fourier transform infrared (SR-FTIR) microspectroscopy were combined to investigate chemical and spatial-temporal changes in incremental damaged maize kernels induced by A. flavus infection at macroscopic and microscopic levels. SWIR-HSI was employed to extract spectral information of A. flavus growth and quantitatively detect AFB1 levels. Satisfactory full-spectrum models and simplified multispectral models were obtained respectively by partial least squares regression (PLSR) for three types of samples. Furthermore, SR-FTIR microspectroscopy coupled with two-dimensional correlation spectroscopy (2DCOS) was utilized to reveal the possible sequence of dynamic changes of nutrient loss and trace AFB1 in maize kernels. It exhibited new insights on how to quantify the spatio-temporal patterns of fungal infection and AFB1 accumulation on maize and provided theoretical basis for online sorting.


Asunto(s)
Aflatoxina B1 , Aspergillus flavus , Imágenes Hiperespectrales , Espectroscopía Infrarroja por Transformada de Fourier , Sincrotrones , Zea mays/química
14.
J Allergy Clin Immunol ; 149(6): 2091-2104, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34974065

RESUMEN

BACKGROUND: Group 2 innate lymphoid cells (ILC2s), the innate counterpart of TH2 cells, play a critical role in type 2 immune responses. However, the molecular regulatory mechanisms of ILC2s are still unclear. OBJECTIVE: The aim of this study was to explore the importance of signal transducer and activator of transcription 3 (STAT3) to ILC2 function in allergic lung inflammation. METHODS: Acute and chronic asthma models were established by intranasal administration of the protease allergen papain in VavicreStat3fl/fl, Il5tdtomato-creStat3fl/fl, and RorccreStat3fl/fl mice to verify the necessity of functional STAT3 for ILC2 allergic response. The intrinsic role of STAT3 in regulating ILC2 function was examined by generation of bone marrow chimera mice. The underlying mechanism was studied through confocal imaging, metabolomics analysis, and chromatin immunoprecipitation quantitative PCR. RESULTS: STAT3 is essential for ILC2 effector function and promotes ILC2-driven allergic inflammation in the lung. Mechanistically, the alarmin cytokine IL-33 induces a noncanonical STAT3 phosphorylation at serine 727 in ILC2s, leading to translocation of STAT3 into the mitochondria. Mitochondrial STAT3 further facilitates adenosine triphosphate synthesis to fuel the methionine cycle and generation of S-adenosylmethionine, which supports the epigenetic reprogramming of type 2 cytokines in ILC2s. STAT3 deficiency, inhibition of STAT3 mitochondrial translocation, or blockade of methionine metabolism markedly dampened the ILC2 allergic response and ameliorated allergic lung inflammation. CONCLUSION: The mitochondrial STAT3-methionine metabolism pathway is a key regulator that shapes ILC2 effector function through epigenetic regulation, and the related proteins or metabolites represent potential therapeutic targets for allergic lung inflammation.


Asunto(s)
Alveolitis Alérgica Extrínseca , Hipersensibilidad , Neumonía , Eosinofilia Pulmonar , Animales , Citocinas , Epigénesis Genética , Inmunidad Innata , Interleucina-33 , Pulmón , Linfocitos , Metionina , Ratones , Mitocondrias , Factor de Transcripción STAT3
15.
Adv Sci (Weinh) ; 9(6): e2103303, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35018740

RESUMEN

Innate lymphoid cells (ILCs) are crucial in orchestrating immunity and maintaining tissue homeostasis in various barrier tissues, but whether ILCs influence immune responses in the urinary tract remains poorly understood. Here, bladder-resident ILCs are comprehensively explored and identified their unique phenotypic and developmental characteristics. Notably, bladder-resident ILCs rapidly respond to uropathogenic Escherichia coli (UPEC) infection. It is found that ILC3 is necessary for early protection against UPEC infection in the bladder. Mechanistically, UPEC infection leads to interleukin (IL)-1ß production in the bladder via a MyD88-dependent pathway, which promotes ILC3 activation. ILC3-expressed IL-17A further recruits neutrophils and controls UPEC infection in the bladder. Together, these results demonstrate a critical role for bladder ILCs in the host defense against UPEC infection.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Inmunidad Innata/inmunología , Infecciones Urinarias/inmunología , Infecciones Urinarias/prevención & control , Escherichia coli Uropatógena/inmunología , Animales , Modelos Animales de Enfermedad , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Vejiga Urinaria/inmunología
16.
Cell Metab ; 33(5): 988-1000.e7, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33761313

RESUMEN

Recent studies in both mice and humans have suggested that gut microbiota could modulate tumor responsiveness to chemo- or immunotherapies. However, the underlying mechanism is not clear yet. Here, we found that gut microbial metabolites, especially butyrate, could promote the efficacy of oxaliplatin by modulating CD8+ T cell function in the tumor microenvironment. Butyrate treatment directly boosted the antitumor cytotoxic CD8+ T cell responses both in vitro and in vivo in an ID2-dependent manner by promoting the IL-12 signaling pathway. In humans, the oxaliplatin responder cancer patients exhibited a higher amount of serum butyrate than did non-responders, which could also increase ID2 expression and function of human CD8+ T cells. Together, our findings suggest that the gut microbial metabolite butyrate could promote antitumor therapeutic efficacy through the ID2-dependent regulation of CD8+ T cell immunity, indicating that gut microbial metabolites could be effective as a part of cancer therapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Metaboloma , Animales , Antibacterianos/farmacología , Antineoplásicos/uso terapéutico , Butiratos/sangre , Butiratos/farmacología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Línea Celular Tumoral , Humanos , Proteína 2 Inhibidora de la Diferenciación/deficiencia , Proteína 2 Inhibidora de la Diferenciación/genética , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Metaboloma/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/tratamiento farmacológico , Oxaliplatino/uso terapéutico , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral
17.
iScience ; 24(3): 102169, 2021 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-33665583

RESUMEN

Ulcerative colitis (UC) is a chronic inflammatory bowel disease, characterized by relapsing and remitting colon mucosal inflammation. For patients suffering from UC, a higher risk of colon cancer has been widely recognized. Here, we found that Elf4 -/- mice developed colon tumors with 3 cycles of dextran sulfate sodium salt (DSS) treatment alone. We further showed that ELF4 suppression was prevalent in both patients with UC and DSS-induced mice models, and this suppression was caused by promoter region methylation. ELF4, upon PARylation by PARP1, transcriptionally regulated multiple DNA damage repair machinery components. Consistently, ELF4 deficiency leads to more severe DNA damage both in vitro and in vivo. Oral administration of montmorillonite powder can prevent the reduction of ELF4 in DSS-induced colitis models and lower the risk of colon tumor development during azoxymethane (AOM) and DSS induced colitis-associated cancer (CAC). These data provided additional mechanism of CAC initiation and supported the "epigenetic priming model of tumor initiation".

19.
Nucleic Acids Res ; 49(1): 114-126, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33290558

RESUMEN

Histone modifications and their functional readout serve as an important mechanism for gene regulation. Lysine benzoylation (Kbz) on histones is a recently identified acylation mark associated with active transcription. However, it remains to be explored whether putative readers exist to recognize this epigenetic mark. Here, our systematic binding studies demonstrated that the DPF and YEATS, but not the Bromodomain family members, are readers for histone Kbz. Co-crystal structural analyses revealed a 'hydrophobic encapsulation' and a 'tip-sensor' mechanism for Kbz readout by DPF and YEATS, respectively. Moreover, the DPF and YEATS family members display subtle yet unique features to create somewhat flexible engagements of different acylation marks. For instance, YEATS2 but not the other YEATS proteins exhibits best preference for Kbz than lysine acetylation and crotonylation due to its wider 'tip-sensor' pocket. The levels of histone benzoylation in cultured cells or in mice are upregulated upon sodium benzoate treatment, highlighting its dynamic regulation. In summary, our work identifies the first readers for histone Kbz and reveals the molecular basis underlying Kbz recognition, thus paving the way for further functional dissections of histone benzoylation.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Epigenómica , Código de Histonas , Familia de Multigenes , Benzoato de Sodio/farmacología , Factores de Transcripción/metabolismo , Acilación , Secuencia de Aminoácidos , Animales , Línea Celular , Proteínas Cromosómicas no Histona/química , Cristalografía por Rayos X , Proteínas de Unión al ADN/química , Histona Acetiltransferasas/metabolismo , Histonas/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Lisina/química , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Unión Proteica , Conformación Proteica , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Factores de Transcripción/química
20.
STAR Protoc ; 1(3): 100218, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33377111

RESUMEN

Citrobacter rodentium is an extracellular enteric bacterial pathogen that induces both innate and adaptive immunity in mice, its natural host. Here, we detail the step-by-step procedure to evaluate the immune responses in a mouse model of C. rodentium infection. We describe the methods to establish infection, isolate group 3 innate lymphoid cells from lamina propria lymphocytes, and analyze their response. We also assess the response of T follicular helper cells and germinal center B cells. For complete details on the use and execution of this protocol, please refer to Guo et al. (2015), Kennedy and Hartland, (2018), and Wang et al. (2020).


Asunto(s)
Separación Celular/métodos , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/inmunología , Inmunidad Adaptativa/inmunología , Animales , Linfocitos B/inmunología , Citrobacter rodentium/inmunología , Citrobacter rodentium/patogenicidad , Colitis/inmunología , Colon/inmunología , Microbioma Gastrointestinal , Centro Germinal/inmunología , Inmunidad Innata/inmunología , Mucosa Intestinal/inmunología , Linfocitos/citología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...