Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 793
Filtrar
1.
Int J Oncol ; 65(1)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38847233

RESUMEN

Several studies have indicated that the gut microbiome and tumor microbiota may affect tumors. Emerging metabolomics research illustrates the need to examine the variations in microbial metabolite composition between patients with cancer and healthy individuals. Microbial metabolites can impact the progression of tumors and the immune response by influencing a number of mechanisms, including modulation of the immune system, cancer or immune­related signaling pathways, epigenetic modification of proteins and DNA damage. Microbial metabolites can also alleviate side effects and drug resistance during chemotherapy and immunotherapy, while effectively activating the immune system to exert tumor immunotherapy. Nevertheless, the impact of microbial metabolites on tumor immunity can be both beneficial and harmful, potentially influenced by the concentration of the metabolites or the specific cancer type. The present review summarizes the roles of various microbial metabolites in different solid tumors, alongside their influence on tumor immunity and treatment. Additionally, clinical trials evaluating the therapeutic effects of microbial metabolites or related microbes on patients with cancer have been listed. In summary, studying microbial metabolites, which play a crucial role in the interaction between the microbiota and tumors, could lead to the identification of new supplementary treatments for cancer. This has the potential to improve the effectiveness of cancer treatment and enhance patient prognosis.


Asunto(s)
Progresión de la Enfermedad , Microbioma Gastrointestinal , Inmunoterapia , Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/inmunología , Neoplasias/microbiología , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Microambiente Tumoral/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoterapia/métodos , Pronóstico
2.
Front Immunol ; 15: 1367053, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38756775

RESUMEN

Background: With the worsening of the greenhouse effect, the correlation between the damp-heat environment (DH) and the incidence of various diseases has gained increasing attention. Previous studies have demonstrated that DH can lead to intestinal disorders, enteritis, and an up-regulation of NOD-like receptor protein 3 (NLRP3). However, the mechanism of NLRP3 in this process remains unclear. Methods: We established a DH animal model to observe the impact of a high temperature and humidity environment on the mice. We sequenced the 16S rRNA of mouse feces, and the RNA transcriptome of intestinal tissue, as well as the levels of cytokines including interferon (IFN)-γ and interleukin (IL)-4 in serum. Results: Our results indicate that the intestinal macrophage infiltration and the expression of inflammatory genes were increased in mice challenged with DH for 14 days, while the M2 macrophages were decreased in Nlrp3 -/- mice. The alpha diversity of intestinal bacteria in Nlrp3 -/- mice was significantly higher than that in control mice, including an up-regulation of the Firmicutes/Bacteroidetes ratio. Transcriptomic analysis revealed 307 differentially expressed genes were decreased in Nlrp3 -/- mice compared with control mice, which was related to humoral immune response, complement activation, phagocytic recognition, malaria and inflammatory bowel disease. The ratio of IFN-γ/IL-4 was decreased in control mice but increased in Nlrp3 -/- mice. Conclusions: Our study found that the inflammation induced by DH promotes Th2-mediated immunity via NLRP3, which is closely related to the disruption of intestinal flora.


Asunto(s)
Microbioma Gastrointestinal , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Células Th2 , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Ratones , Microbioma Gastrointestinal/inmunología , Células Th2/inmunología , Calor , Alarminas/inmunología , Alarminas/metabolismo , Ratones Endogámicos C57BL , Macrófagos/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad
3.
Adv Protein Chem Struct Biol ; 140: 381-417, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38762276

RESUMEN

Cardiovascular disease (CVD) and cancer are major contributors to global morbidity and mortality. This book chapter delves into the intricate relationship between the immune system and the pathogenesis of both cardiovascular and cancer diseases, exploring the roles of innate and adaptive immunities, immune regulation, and immunotherapy in these complex conditions. The innate immune system acts as the first line of defense against tissue damage and infection, with a significant impact on the initiation and progression of CVD and cancer. Endothelial dysfunction, a hallmark in CVD, shares commonalities with the tumor microenvironment in cancer, emphasizing the parallel involvement of the immune system in both conditions. The adaptive immune system, particularly T cells, contributes to prolonged inflammation in both CVD and cancer. Regulatory T cells and the intricate balance between different T cell subtypes influence disease progression, wound healing, and the outcomes of ischemic injury and cancer immunosurveillance. Dysregulation of immune homeostasis can lead to chronic inflammation, contributing to the development and progression of both CVD and cancer. Thus, immunotherapy emerged as a promising avenue for preventing and managing these diseases, with strategies targeting immune cell modulation, cytokine manipulation, immune checkpoint blockade, and tolerance induction. The impact of gut microbiota on CVD and cancer too is explored in this chapter, highlighting the role of gut leakiness, microbial metabolites, and the potential for microbiome-based interventions in cardiovascular and cancer immunotherapies. In conclusion, immunomodulatory strategies and immunotherapy hold promise in reshaping the landscape of cardiovascular and cancer health. Additionally, harnessing the gut microbiota for immune modulation presents a novel approach to prevent and manage these complex diseases, emphasizing the importance of personalized and precision medicine in healthcare. Ongoing research and clinical trials are expected to further elucidate the complex immunological underpinnings of CVD and cancer thereby refining these innovative approaches.


Asunto(s)
Enfermedades Cardiovasculares , Neoplasias , Humanos , Neoplasias/inmunología , Neoplasias/terapia , Enfermedades Cardiovasculares/inmunología , Inmunoterapia , Inmunidad Innata/inmunología , Microbioma Gastrointestinal/inmunología , Animales , Inmunidad Adaptativa/inmunología
4.
Aging Clin Exp Res ; 36(1): 117, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38780713

RESUMEN

Diet is one of the lifestyle factors that is most amenable to intervention, and has a substantial effect on the potential for successful aging and mitigation of the risk of disease. Good nutrition is a pillar of healthy aging, and a large body of evidence attests to the benefits of the Mediterranean diet on the quality of the aging process. The Mediterranean diet comprises a wide range of nutrients which, both individually and collectively, exert positive effects on immunity, in large part mediated by the gut microbiota. In this article, we review the effect of the Mediterranean diet on immunity, and how its beneficial effects are mediated by the gut microbiota. We review the effects of certain key components of the Mediterranean dietary pattern, including vitamins, zinc, selenium, and polyphenols. Overall, the existing body of evidence convincingly demonstrates that the Mediterreanean diet affects immune health by maintaining a healthy body weight and reducing the risk of metabolic and cardiovascular diseases; by reducing inflammation and by promoting a healthy gut microbiota profile.


Asunto(s)
Dieta Mediterránea , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/inmunología , Anciano , Envejecimiento/inmunología , Inmunidad/fisiología
5.
J Hematol Oncol ; 17(1): 33, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38745196

RESUMEN

The gut microbiota plays a critical role in the progression of human diseases, especially cancer. In recent decades, there has been accumulating evidence of the connections between the gut microbiota and cancer immunotherapy. Therefore, understanding the functional role of the gut microbiota in regulating immune responses to cancer immunotherapy is crucial for developing precision medicine. In this review, we extract insights from state-of-the-art research to decipher the complicated crosstalk among the gut microbiota, the systemic immune system, and immunotherapy in the context of cancer. Additionally, as the gut microbiota can account for immune-related adverse events, we discuss potential interventions to minimize these adverse effects and discuss the clinical application of five microbiota-targeted strategies that precisely increase the efficacy of cancer immunotherapy. Finally, as the gut microbiota holds promising potential as a target for precision cancer immunotherapeutics, we summarize current challenges and provide a general outlook on future directions in this field.


Asunto(s)
Microbioma Gastrointestinal , Inmunoterapia , Neoplasias , Humanos , Microbioma Gastrointestinal/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Inmunoterapia/métodos , Animales
6.
J Reprod Immunol ; 163: 104251, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38718429

RESUMEN

Recurrent pregnancy loss (RPL) is a troubling condition that affects couples worldwide. Despite extensive research efforts, many RPL cases remain unexplained, highlighting the need for novel approaches to unravel its underlying mechanisms. Recent advances in microbiome research have shed light on the potential role of the microbiome in reproductive health and outcomes. Based on a systematic literature research, this review aims to comprehensively explore the current understanding of the microbiome's involvement in RPL, focusing on the vaginal, endometrial, and gut microbiomes. Evidence from the available studies is examined to explain the relationship between the microbiome and RPL. Furthermore, we discuss the diagnostic potential of the microbiome, therapeutic interventions, and future directions in microbiome research for RPL. Understanding the complex interactions between the microbiome and reproductive health holds promise for developing targeted interventions to help patients today diagnosed as unexplained.


Asunto(s)
Aborto Habitual , Microbiota , Humanos , Aborto Habitual/microbiología , Aborto Habitual/inmunología , Aborto Habitual/diagnóstico , Femenino , Embarazo , Microbiota/inmunología , Microbioma Gastrointestinal/inmunología , Endometrio/microbiología , Endometrio/inmunología , Endometrio/patología , Vagina/microbiología , Vagina/inmunología
9.
Sci Immunol ; 9(95): eadi5374, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38758808

RESUMEN

The gut microbiota and tumor-associated macrophages (TAMs) affect tumor responses to anti-programmed cell death protein 1 (PD-1) immune checkpoint blockade. Reprogramming TAM by either blocking or deleting the macrophage receptor triggering receptor on myeloid cells 2 (TREM2) attenuates tumor growth, and lack of functional TREM2 enhances tumor elimination by anti-PD-1. Here, we found that anti-PD-1 treatment combined with TREM2 deficiency in mice induces proinflammatory programs in intestinal macrophages and a concomitant expansion of Ruminococcus gnavus in the gut microbiota. Gavage of wild-type mice with R. gnavus enhanced anti-PD-1-mediated tumor elimination, recapitulating the effect occurring in the absence of TREM2. A proinflammatory intestinal environment coincided with expansion, increased circulation, and migration of TNF-producing CD4+ T cells to the tumor bed. Thus, TREM2 remotely controls anti-PD-1 immune checkpoint blockade through modulation of the intestinal immune environment and microbiota, with R. gnavus emerging as a potential probiotic agent for increasing responsiveness to anti-PD-1.


Asunto(s)
Microbioma Gastrointestinal , Inmunoterapia , Macrófagos , Glicoproteínas de Membrana , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1 , Receptores Inmunológicos , Animales , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Ratones , Microbioma Gastrointestinal/inmunología , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Inmunoterapia/métodos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Macrófagos/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones Noqueados , Femenino , Intestinos/inmunología
10.
Int Immunopharmacol ; 133: 111877, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38608440

RESUMEN

The gut microbiome plays an important role in tumor growth by regulating immune cell function. However, the role of the gut microbiome-mediated monocytes in liver metastasis remains unclear. In this study, we found that fecal microbiome transplantation (FMT) from the stool of patients with liver metastasis (LM) significantly promoted liver metastasis compared with healthy donors (HD). Monocytes were upregulated in liver tissues by the CCL2/CCR2 axis in LM patients' stool transplanted mouse model. CCL2/CCR2 inhibition and monocyte depletion significantly suppress liver metastasis. FMT using LM patients' stool enhanced the plasma lipopolysaccharides (LPS) concentration. The LPS/TLR4 signaling pathway is crucial for gut microbiome-mediated liver metastasis. These results indicated that monocytes contribute to liver metastasis via the CCL2/CCR2 axis.


Asunto(s)
Quimiocina CCL2 , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Neoplasias Hepáticas , Monocitos , Receptores CCR2 , Receptor Toll-Like 4 , Microbioma Gastrointestinal/inmunología , Animales , Humanos , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/inmunología , Monocitos/inmunología , Quimiocina CCL2/metabolismo , Ratones , Receptores CCR2/metabolismo , Receptor Toll-Like 4/metabolismo , Masculino , Lipopolisacáridos/inmunología , Ratones Endogámicos C57BL , Femenino , Transducción de Señal , Línea Celular Tumoral , Hígado/patología , Hígado/inmunología , Hígado/metabolismo
11.
Clin Respir J ; 18(5): e13762, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38685799

RESUMEN

OBJECTIVE: This investigation aims to explore alterations in intestinal microecology and immune function among patients with advanced, unresectable lung adenocarcinoma undergoing different outcomes from immunotherapy. METHODS: A cohort of 30 patients diagnosed with advanced unresectable lung adenocarcinoma received sintilimab immunotherapy as a monotherapy. Post four treatment cycles, efficacy was assessed, leading to the segregation of patients into two distinct cohorts: those responsive to treatment and those nonresponsive. Analysis involved observing variations in the abundance, distribution, and composition of fecal intestinal microorganisms pretreatment and posttreatment via 16S rRNA gene sequencing. RESULTS: In this study involving 30 advanced lung adenocarcinoma patients, significant observations were made regarding the impact of immunotherapy on immune function and the gut microbiome composition. Patients were divided into treatment and control groups, revealing that immunotherapy led to a significant increase in CD4+ T cells and a decrease in CD8+ T cells among the treatment-responsive individuals, indicating an enhanced immune response. Furthermore, an in-depth analysis of the gut microbiome showed an increase in diversity and abundance of beneficial bacteria such as Faecalibacterium and Subdoligranulum in the treatment group. These findings highlight the dual effect of immunotherapy on modulating immune function and altering gut microbiome diversity, suggesting its potential therapeutic benefits in improving the health status of patients with advanced lung adenocarcinoma. CONCLUSION: The structuring of gut flora plays a pivotal role in augmenting the efficacy of anti-tumor immunotherapy, underscoring the interplay between intestinal microecology and immune response in cancer treatment outcomes.


Asunto(s)
Adenocarcinoma del Pulmón , Microbioma Gastrointestinal , Inmunoterapia , Neoplasias Pulmonares , Humanos , Microbioma Gastrointestinal/inmunología , Masculino , Femenino , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Persona de Mediana Edad , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/terapia , Adenocarcinoma del Pulmón/tratamiento farmacológico , Inmunoterapia/métodos , Anciano , ARN Ribosómico 16S/genética , Resultado del Tratamiento
12.
Dev Comp Immunol ; 156: 105176, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38582249

RESUMEN

Due to the ongoing global warming, the risk of heatwaves in the oceans is continuously increasing while our understanding of the physiological response of Litopenaeus vannamei under extreme temperature conditions remains limited. Therefore, this study aimed to evaluate the physiological responses of L. vannamei under heat stress. Our results indicated that as temperature rose, the structure of intestinal and hepatopancreatic tissues was damaged sequentially. Activity of immune-related enzymes (acid phosphatase/alkaline phosphatase) initially increased before decreased, while antioxidant enzymes (superoxide dismutase and glutathione-S transferase) activity and malondialdehyde content increased with rising temperature. In addition, the total antioxidant capacity decreased with rising temperature. With the rising temperature, there was a significant increase in the expression of caspase-3, heat shock protein 70, lipopolysaccharide-induced tumor necrosis factor-α, transcriptional enhanced associate domain and yorkie in intestinal and hepatopancreatic tissues. Following heat stress, the number of potentially beneficial bacteria (Rhodobacteraceae and Gemmonbacter) increased which maintain balance and promote vitamin synthesis. Intestinal transcriptome analysis revealed 852 differentially expressed genes in the heat stress group compared with the control group. KEGG functional annotation results showed that the endocrine system was the most abundant in Organismal systems followed by the immune system. These results indicated that heat stress leads to tissue damage in shrimp, however the shrimp may respond to stress through a coordinated interaction strategy of the endocrine system, immune system and gut microbiota. This study revealed the response mechanism of L. vannamei to acute heat stress and potentially provided a theoretical foundation for future research on shrimp environmental adaptations.


Asunto(s)
Microbioma Gastrointestinal , Respuesta al Choque Térmico , Penaeidae , Transcriptoma , Animales , Penaeidae/inmunología , Penaeidae/microbiología , Penaeidae/genética , Respuesta al Choque Térmico/genética , Respuesta al Choque Térmico/inmunología , Microbioma Gastrointestinal/inmunología , Intestinos/inmunología , Intestinos/microbiología , Sistema Inmunológico/metabolismo , Sistema Inmunológico/inmunología , Perfilación de la Expresión Génica , Hepatopáncreas/inmunología , Hepatopáncreas/metabolismo , Proteínas de Artrópodos/metabolismo , Proteínas de Artrópodos/genética , Antioxidantes/metabolismo
13.
Nature ; 629(8013): 901-909, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38658756

RESUMEN

The liver is the main gateway from the gut, and the unidirectional sinusoidal flow from portal to central veins constitutes heterogenous zones, including the periportal vein (PV) and the pericentral vein zones1-5. However, functional differences in the immune system in each zone remain poorly understood. Here intravital imaging revealed that inflammatory responses are suppressed in PV zones. Zone-specific single-cell transcriptomics detected a subset of immunosuppressive macrophages enriched in PV zones that express high levels of interleukin-10 and Marco, a scavenger receptor that sequesters pro-inflammatory pathogen-associated molecular patterns and damage-associated molecular patterns, and consequently suppress immune responses. Induction of Marco+ immunosuppressive macrophages depended on gut microbiota. In particular, a specific bacterial family, Odoribacteraceae, was identified to induce this macrophage subset through its postbiotic isoallolithocholic acid. Intestinal barrier leakage resulted in inflammation in PV zones, which was markedly augmented in Marco-deficient conditions. Chronic liver inflammatory diseases such as primary sclerosing cholangitis (PSC) and non-alcoholic steatohepatitis (NASH) showed decreased numbers of Marco+ macrophages. Functional ablation of Marco+ macrophages led to PSC-like inflammatory phenotypes related to colitis and exacerbated steatosis in NASH in animal experimental models. Collectively, commensal bacteria induce Marco+ immunosuppressive macrophages, which consequently limit excessive inflammation at the gateway of the liver. Failure of this self-limiting system promotes hepatic inflammatory disorders such as PSC and NASH.


Asunto(s)
Colangitis Esclerosante , Microbioma Gastrointestinal , Inflamación , Hígado , Macrófagos , Enfermedad del Hígado Graso no Alcohólico , Simbiosis , Animales , Femenino , Humanos , Masculino , Ratones , Bacteroidetes/metabolismo , Colangitis Esclerosante/inmunología , Colangitis Esclerosante/microbiología , Colangitis Esclerosante/patología , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Perfilación de la Expresión Génica , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Hígado/inmunología , Hígado/patología , Hígado/microbiología , Macrófagos/citología , Macrófagos/inmunología , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/patología , Vena Porta , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/metabolismo , Análisis de la Célula Individual , Simbiosis/inmunología
14.
Exp Hematol ; 133: 104211, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38527589

RESUMEN

Leukemias are a set of clonal hematopoietic malignant diseases that develop in the bone marrow. Several factors influence leukemia development and progression. Among these, the gut microbiota is a major factor influencing a wide array of its processes. The gut microbial composition is linked to the risk of tumor development and the host's ability to respond to treatment, mostly due to the immune-modulatory effects of their metabolites. Despite such strong evidence, its role in the development of hematologic malignancies still requires attention of investigators worldwide. In this review, we make an effort to discuss the role of host gut microbiota-immune crosstalk in leukemia development and progression. Additionally, we highlight certain recently developed strategies to modify the gut microbial composition that may help to overcome dysbiosis in leukemia patients in the near future.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Leucemia , Humanos , Disbiosis/inmunología , Disbiosis/complicaciones , Microbioma Gastrointestinal/inmunología , Leucemia/inmunología , Leucemia/microbiología , Leucemia/etiología , Animales
15.
Immunol Rev ; 323(1): 126-137, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38491842

RESUMEN

Group 3 innate lymphoid cells (ILC3s) are tissue-resident immune lymphocytes that critically regulate intestinal homeostasis, organogenesis, and immunity. ILC3s possess the capacity to "sense" the inflammatory environment within tissues, especially in the context of pathogen challenges that imprints durable non-antigen-specific changes in ILC3 function. As such, ILC3s become a new actor in the emerging field of trained innate immunity. Here, we summarize recent discoveries regarding ILC3 responses to bacterial challenges and the role these encounters play in triggering trained innate immunity. We further discuss how signaling events throughout ILC3 ontogeny potentially control the development and function of trained ILC3s. Finally, we highlight the open questions surrounding ILC3 "training" the answers to which may reveal new insights into innate immunity. Understanding the fundamental concepts behind trained innate immunity could potentially lead to the development of new strategies for improving immunity-based modulation therapies for inflammation, infectious diseases, and cancer.


Asunto(s)
Inmunidad Innata , Linfocitos , Transducción de Señal , Humanos , Animales , Linfocitos/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Homeostasis , Inflamación/inmunología , Microbioma Gastrointestinal/inmunología , Intestinos/inmunología
16.
Peptides ; 176: 171200, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38555054

RESUMEN

Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are peptide hormones produced by enteroendocrine cells in the small intestine. Despite being produced in the gut, the leveraging of their role in potentiating glucose-stimulated insulin secretion, also known as the incretin effect, has distracted from discernment of direct intestinal signaling circuits. Both preclinical and clinical evidence have highlighted a role for the incretins in inflammation. In this review, we highlight the discoveries of GLP-1 receptor (GLP-1R)+ natural (TCRαß and TCRγδ) and induced (TCRαß+CD4+ cells and TCRαß+CD8αß+) intraepithelial lymphocytes. Both endogenous signaling and pharmacological activation of GLP-1R impact local and systemic inflammation, the gut microbiota, whole-body metabolism, as well as the control of GLP-1 bioavailability. While GIPR signaling has been documented to impact hematopoiesis, the impact of these bone marrow-derived cells in gut immunology is not well understood. We uncover gaps in the literature of the evaluation of the impact of sex in these GLP-1R and GIP receptor (GIPR) signaling circuits and provide speculations of the maintenance roles these hormones play within the gut in the fasting-refeeding cycles. GLP-1R agonists and GLP-1R/GIPR agonists are widely used as treatments for diabetes and weight loss, respectively; however, their impact on gut homeostasis has not been fully explored. Advancing our understanding of the roles of GLP-1R and GIPR signaling within the gut at homeostasis as well as metabolic and inflammatory diseases may provide targets to improve disease management.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón , Inflamación , Receptores de la Hormona Gastrointestinal , Humanos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/genética , Receptores de la Hormona Gastrointestinal/metabolismo , Inflamación/metabolismo , Inflamación/inmunología , Animales , Inmunomodulación , Microbioma Gastrointestinal/inmunología , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Transducción de Señal
17.
Immunol Lett ; 267: 106853, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38513836

RESUMEN

BACKGROUND: Allergic asthma is a heterogeneous disease and new strategies are needed to prevent or treat this disease. Studies have shown that probiotic interventions are effective in preventing asthma. Here, we investigated the impact of Saccharomyces boulardii (S. boulardii) on ovalbumin (OVA)-induced allergic asthma in mice, as well as the underlying mechanisms. METHODS: First, we constructed a mouse asthma model using OVA and given S. boulardii intervention. Next, we measured N6-methyladenosine (m6A) levels in lung injury tissues. 16 s rRNA was employed to identify different gut microbiota in fecal samples. The analysis of differential metabolites in feces was performed by non-targeted metabolomics. Pearson correlation coefficient was utilized to analyze correlation between gut microbiota, metabolites and methyltransferase-like 3 (METTL3). Finally, we collected mouse feces treated by OVA and S. boulardii intervention for fecal microbiota transplantation (FMT) and interfered with METTL3. RESULTS: S. boulardii improved inflammation and oxidative stress and alleviated lung damage in asthmatic mice. In addition, S. boulardii regulated m6A modification levels in asthmatic mice. 16 s rRNA sequencing showed that S. boulardii remodeled gut microbiota homeostasis in asthmatic mice. Non-targeted metabolomics analysis showed S. boulardii restored metabolic homeostasis in asthmatic mice. There was a correlation between gut microbiota, differential metabolites, and METTL3 analyzed by Pearson correlation. Additionally, through FMT and interference of METTL3, we found that gut microbiota mediated the up-regulation of METTL3 by S. boulardii improved inflammation and oxidative stress in asthmatic mice, and alleviated lung injury. CONCLUSIONS: S. boulardii alleviated allergic asthma by restoring gut microbiota and metabolic homeostasis via up-regulation of METTL3 in an m6A-dependent manner.


Asunto(s)
Adenosina , Asma , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Homeostasis , Metiltransferasas , Probióticos , Saccharomyces boulardii , Regulación hacia Arriba , Animales , Asma/terapia , Asma/metabolismo , Asma/inmunología , Asma/etiología , Asma/microbiología , Metiltransferasas/metabolismo , Metiltransferasas/genética , Microbioma Gastrointestinal/inmunología , Ratones , Adenosina/metabolismo , Adenosina/análogos & derivados , Probióticos/administración & dosificación , Probióticos/uso terapéutico , Femenino , Trasplante de Microbiota Fecal , Ovalbúmina/inmunología , Ratones Endogámicos BALB C
18.
Eur J Immunol ; 54(6): e2350631, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38556632

RESUMEN

The intestinal barrier is mainly formed by a monolayer of epithelial cells, which forms a physical barrier to protect the gut tissues from external insults and provides a microenvironment for commensal bacteria to colonize while ensuring immune tolerance. Moreover, various immune cells are known to significantly contribute to intestinal barrier function by either directly interacting with epithelial cells or by producing immune mediators. Fulfilling this function of the gut barrier for mucosal homeostasis requires not only the intrinsic regulation of intestinal epithelial cells (IECs) but also constant communication with immune cells and gut microbes. The reciprocal interactions between IECs and immune cells modulate mucosal barrier integrity. Dysregulation of barrier function could lead to dysbiosis, inflammation, and tumorigenesis. In this overview, we provide an update on the characteristics and functions of IECs, and how they integrate their functions with tissue immune cells and gut microbiota to establish gut homeostasis.


Asunto(s)
Células Epiteliales , Microbioma Gastrointestinal , Homeostasis , Mucosa Intestinal , Humanos , Homeostasis/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Animales , Microbioma Gastrointestinal/inmunología , Células Epiteliales/inmunología , Comunicación Celular/inmunología , Tolerancia Inmunológica/inmunología
19.
Int Rev Immunol ; 43(4): 229-247, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38343353

RESUMEN

BACKGROUND: The gut microbiome plays a role in the development and progression of colorectal cancer (CRC). AIM AND OBJECTIVE: This review focuses on whether the gut microbiome is involved in the development and regulation of the host immune system. METHODS: The gut microbiome can influence the production and activity of immune cells and molecules that help to maintain the integrity of the intestinal barrier and prevent inflammation. Gut microbiota modulates the anti-cancer immune response. The gut microbiota can influence the function of immune cells, like T cells, that recognize and eliminate cancer cells. Gut microbiota can affect various aspects of cancer progression and the efficacy of various anti-cancer treatments. RESULTS: Gut microbiota provide promise as a potential biomarker to identify the effect of immunotherapy and as a target for modulation to improve the efficacy of immunotherapy in CRC treatment. CONCLUSION: The potential synergistic effect between the gut microbiome and anti-cancer treatment modalities provides an interest in developing strategies to modulate the gut microbiome to improve the efficacy of anti-cancer treatment.


This review focuses on the gut microbiome in the development and regulation of the host immune system. Gut microbiota provides potential biomarkers to identify the effect of immunotherapy and as a target for modulation of immunotherapy in the treatment of CRC. This provides potential synergistic effects between the gut microbiome and anti-cancer treatment modalities.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Humanos , Neoplasias Colorrectales/terapia , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Microbioma Gastrointestinal/inmunología , Animales , Inmunoterapia/métodos , Probióticos/uso terapéutico
20.
JAMA Dermatol ; 159(10): 1076-1084, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37647056

RESUMEN

Importance: The gut microbiome modulates the immune system and responses to immunotherapy in patients with late-stage melanoma. It is unknown whether fecal microbiota profiles differ between healthy individuals and patients with melanoma or if microbiota profiles differ among patients with different stages of melanoma. Defining gut microbiota profiles in individuals without melanoma and those with early-stage and late-stage melanoma may reveal features associated with disease progression. Objective: To characterize and compare gut microbiota profiles between healthy volunteers and patients with melanoma and between patients with early-stage and late-stage melanoma. Design, Setting, and Participants: This single-site case-control study took place at an academic comprehensive cancer center. Fecal samples were collected from systemic treatment-naive patients with stage I to IV melanoma from June 1, 2015, to January 31, 2019, and from healthy volunteers from June 1, 2021, to January 31, 2022. Patients were followed up for disease recurrence until November 30, 2021. Main Outcomes and Measures: Fecal microbiota was profiled by 16S ribosomal RNA sequencing. Clinical and pathologic characteristics, treatment, and disease recurrence were extracted from electronic medical records. Fecal microbiome diversity, taxonomic profiles and inferred functional profiles were compared between groups. Results: A total of 228 participants were enrolled (126 men [55.3%]; median age, 59 [range, 21-90] years), including 49 volunteers without melanoma, 38 patients with early-stage melanoma (29 with stage I or melanoma in situ and 9 with stage II), and 141 with late-stage melanoma (66 with stage III and 75 with stage IV). Community differences were observed between patients with melanoma and volunteers. Patients with melanoma had a higher relative abundance of Fusobacterium compared with controls on univariate analysis (0.19% vs 0.003%; P < .001), but this association was attenuated when adjusted for covariates (log2 fold change of 5.18 vs controls; P = .09). Microbiomes were distinct between patients with early-stage and late-stage melanoma. Early-stage melanoma had a higher alpha diversity (Inverse Simpson Index 14.6 [IQR, 9.8-23.0] vs 10.8 [IQR, 7.2-16.8]; P = .003), and a higher abundance of the genus Roseburia on univariate analysis (2.4% vs 1.2%; P < .001) though statistical significance was lost with covariate adjustment (log2 fold change of 0.86 vs controls; P = .13). Multiple functional pathways were differentially enriched between groups. No associations were observed between the microbial taxa and disease recurrence in patients with stage III melanoma treated with adjuvant immunotherapy. Conclusions and Relevance: The findings of this case-control study suggest that fecal microbiota profiles were significantly different among patients with melanoma and controls and between patients with early-stage and late-stage melanoma. Prospective investigations of the gut microbiome and changes that occur with disease progression may identify future microbial targets for intervention.


Asunto(s)
Microbioma Gastrointestinal , Melanoma , Masculino , Humanos , Persona de Mediana Edad , Microbioma Gastrointestinal/inmunología , Estudios Prospectivos , Estudios de Casos y Controles , Progresión de la Enfermedad , Melanoma Cutáneo Maligno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA