Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.421
Filtrar
1.
Arch Microbiol ; 206(10): 410, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39302484

RESUMO

Atopic dermatitis (AD) is a common and recurrent skin disease characterized by skin barrier dysfunction, inflammation and chronic pruritus, with wide heterogeneity in terms of age of onset, clinical course and persistence over the lifespan. Although the pathogenesis of the disease are unclear, epidermal barrier dysfunction, immune and microbial dysregulation, and environmental factors are known to be critical etiologies in AD pathology. The skin microbiota represents an ecosystem consisting of numerous microbial species that interact with each other as well as host epithelial cells and immune cells. Although the skin microbiota benefits the host by supporting the basic functions of the skin and preventing the colonization of pathogens, disruption of the microbial balance (dysbiosis) can cause skin diseases such as AD. Although AD is a dermatological disease, recent evidence has shown that changes in microbiota composition in the skin and intestine contribute to the pathogenesis of AD. Environmental factors that contribute to skin barrier dysfunction and microbial dysbiosis in AD include allergens, diet, irritants, air pollution, epigenetics and microbial exposure. Knowing the microbial combination of intestin, as well as the genetic and epigenetic determinants associated with the development of autoantibodies, may help elucidate the pathophysiology of the disease. The skin of patients with AD is characterized by microbial dysbiosis as a result of reduced microbial diversity and overgrowth of the pathogens such as Staphylococcus aureus. Recent studies have revealed the importance of building a strong immune response against microorganisms during childhood and new mechanisms of microbial community dynamics in modulating the skin microbiome. Numerous microorganisms are reported to modulate host response through communication with keratinocytes, specific immune cells and adipocytes to improve skin health and barrier function. This growing insight into bioactive substances in the skin microbiota has led to novel biotherapeutic approaches targeting the skin surface for the treatment of AD. This review will provide an updated overview of the skin microbiota in AD and its complex interaction with immune response mechanisms, as well as explore possible underlying mechanisms in the pathogenesis of AD and provide insights into new therapeutic developments for the treatment of AD. It also focuses on restoring skin microbial homeostasis, aiming to reduce inflammation by repairing the skin barrier.


Assuntos
Dermatite Atópica , Disbiose , Pele , Staphylococcus aureus , Dermatite Atópica/microbiologia , Dermatite Atópica/imunologia , Humanos , Staphylococcus aureus/imunologia , Staphylococcus aureus/fisiologia , Pele/microbiologia , Pele/imunologia , Pele/patologia , Disbiose/microbiologia , Disbiose/imunologia , Microbiota/imunologia , Animais , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia
2.
Immunity ; 57(9): 1995-1999, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39260349

RESUMO

The immune system has a vital, albeit complex, relationship with the microbes residing within us, one that we are only beginning to understand. We asked investigators what they felt were the fundamental challenges we currently face in unraveling the impacts of microbes and their metabolites on host immunity and to discuss key opportunities toward achieving future insights and innovation.


Assuntos
Imunidade , Animais , Humanos , Bactérias/imunologia , Bactérias/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Microbiota/imunologia
3.
Nat Plants ; 10(9): 1363-1376, 2024 09.
Artigo em Inglês | MEDLINE | ID: mdl-39242981

RESUMO

Over the past three decades, researchers have isolated plant mutants that show constitutively activated defence responses in the absence of pathogen infection. These mutants are called autoimmune mutants and are typically dwarf and/or bearing chlorotic/necrotic lesions. Here, from a genetic screen for Arabidopsis genes involved in maintaining a normal leaf microbiota, we identified TIP GROWTH DEFECTIVE 1 (TIP1), which encodes an S-acyltransferase, as a key player in guarding leaves against abnormal microbiota level and composition under high-humidity conditions. The tip1 mutant has several characteristic phenotypes of classical autoimmune mutants, including a dwarf stature, showing lesions, and having a high basal level of defence gene expression. Gnotobiotic experiments revealed that the autoimmune phenotypes of the tip1 mutant are largely dependent on the presence of microbiota as axenic tip1 plants have markedly reduced autoimmune phenotypes. We found that the microbiota dependency of autoimmune phenotypes is shared by several 'lesion mimic'-type autoimmune mutants in Arabidopsis. It is worth noting that autoimmune phenotypes caused by mutations in two Nucleotide-Binding, Leucine-Rich Repeat (NLR) genes do not require the presence of microbiota and can even be partially alleviated by microbiota. Our results therefore suggest the existence of at least two classes of autoimmunity (microbiota-dependent versus microbiota-independent) in plants. The observed interplay between autoimmunity and microbiota in the lesion mimic class of autoimmunity is reminiscent of the interactions between autoimmunity and dysbiosis in the animal kingdom. These parallels highlight the intricate relationship between host immunity and microbial communities across various biological systems.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Autoimunidade , Microbiota , Folhas de Planta , Arabidopsis/microbiologia , Arabidopsis/imunologia , Arabidopsis/genética , Folhas de Planta/microbiologia , Folhas de Planta/imunologia , Folhas de Planta/genética , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Microbiota/imunologia , Mutação , Imunidade Vegetal/genética , Aciltransferases/genética , Fenótipo
5.
Sci Rep ; 14(1): 20813, 2024 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-39242814

RESUMO

During pregnancy the immune system needs to maintain immune tolerance of the foetus while also responding to infection, which can cause premature activation of the inflammatory pathways leading to the onset of labour and preterm birth. The vaginal microbiome is an important modifier of preterm birth risk, with Lactobacillus dominance during pregnancy associated with term delivery while high microbial diversity is associated with an increased risk of preterm birth. Glycans on glycoproteins along the lower female reproductive tract are fundamental to microbiota-host interactions and the mediation of inflammatory responses. However, the specific glycan epitopes involved in these processes are not well understood. To address this, we conducted glycomic analyses of cervicovaginal fluid (CVF) from 36 pregnant women at high risk of preterm birth and 4 non-pregnant women. Our analysis of N- and O-glycans revealed a rich CVF glycome. While O-glycans were shown to be the main carriers of ABO blood group epitopes, the main features of N-glycans were the presence of abundant paucimannose and high mannose glycans, and a remarkable diversity of complex bi-, tri-, and tetra-antennary glycans decorated with fucose and sialic acid. We identified immuno-regulatory epitopes, such as Lewis antigens, and found that fucosylation was negatively correlated to pro-inflammatory factors, such as IL-1ß, MMP-8, C3a and C5a, while glycans with only sialylated antennae were mainly positively correlated to those. Similarly, paucimannose glycans showed a positive correlation to pro-inflammatory factors. We revealed a high abundance of glycans which have previously been identified as hallmarks of cancer and viral glycosylation, such as Man8 and Man9 high mannose glycans. Although each pregnant woman had a unique glycomic profile, longitudinal studies showed that the main glycosylation features were consistent throughout pregnancy in women who delivered at term, whereas women who experienced extreme preterm birth exhibited sharp changes in the CVF glycome shortly before delivery. These findings shed light on the processes underlying the role of glycosylation in maintaining a healthy vaginal microbiome and associated host immune responses. In addition, these discoveries facilitate our understanding of the lower female reproductive tract which has broad implications for women's health.


Assuntos
Epitopos , Glicômica , Polissacarídeos , Nascimento Prematuro , Vagina , Humanos , Feminino , Nascimento Prematuro/imunologia , Nascimento Prematuro/metabolismo , Gravidez , Glicosilação , Vagina/imunologia , Vagina/metabolismo , Vagina/microbiologia , Adulto , Epitopos/imunologia , Polissacarídeos/metabolismo , Polissacarídeos/imunologia , Colo do Útero/imunologia , Colo do Útero/metabolismo , Líquidos Corporais/imunologia , Líquidos Corporais/metabolismo , Microbiota/imunologia
6.
Front Immunol ; 15: 1423232, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39267734

RESUMO

Over the last decades, extracellular vesicles (EVs) have become increasingly popular for their roles in various pathologies, including cancer and neurological and immunological disorders. EVs have been considered for a long time as a means for normal cells to get rid of molecules it no longer needs. It is now well established that EVs play their biological roles also following uptake or by the interaction of EV surface proteins with cellular receptors and membranes. In this review, we summarize the current status of EV production and secretion in glioblastoma, the most aggressive type of glioma associated with high mortality. The main purpose is to shed light on the EVs as a universal mediator of interkingdom and intrakingdom communication in the context of tumor microenvironment heterogeneity. We focus on the immunomodulatory EV functions in glioblastoma-immune cross-talk to enhance immune escape and reprogram tumor-infiltrating immune cells. We critically examine the evidence that GBM-, immune cell-, and microbiome-derived EVs impact local tumor microenvironment and host immune responses, and can enter the circulatory system to disseminate and drive premetastatic niche formation in distant organs. Taking into account the current state of the art in intratumoral microbiome studies, we discuss the emerging role of bacterial EV in glioblastoma and its response to current and future therapies including immunotherapies.


Assuntos
Neoplasias Encefálicas , Vesículas Extracelulares , Glioblastoma , Microambiente Tumoral , Humanos , Microambiente Tumoral/imunologia , Glioblastoma/imunologia , Glioblastoma/patologia , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Animais , Evasão Tumoral , Comunicação Celular/imunologia , Imunoterapia/métodos , Microbiota/imunologia
7.
JCI Insight ; 9(18)2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39163137

RESUMO

Palatine tonsils are the only air-contacted lymphoid organs that constantly engage in crosstalk with commensal microorganisms and serve as the first handling sites against microbial antigens. While tonsil inflammations have been implicated in various autoimmune diseases, including rheumatoid arthritis (RA), the precise role of tonsillar microbiota in autoimmune pathogenesis remains inadequately characterized. In this study, we profiled the tonsillar microbiota and identified a notable dysbiosis in patients with RA, particularly within the Streptococcus genus. Specifically, patients with RA exhibited an enrichment of pathogenic Streptococcus species, including S. pyogenes, S. dysgalactiae, and S. agalactiae. Colonization with these bacteria significantly exacerbated arthritis severity and increased autoimmune responses in collagen-induced arthritis (CIA). Furthermore, immunization with peptides derived from these pathogenic Streptococcus species directly induced experimental arthritis. Conversely, patients with RA demonstrated a marked deficiency in commensal Streptococcus members, notably S. salivarius. Treatment of CIA mice with S. salivarius attenuated the progression of arthritis and downregulated autoimmune responses. These findings highlight a pathogenic link of tonsillar microbiota with RA, shedding light on their contribution to autoimmunity.


Assuntos
Artrite Experimental , Artrite Reumatoide , Microbiota , Tonsila Palatina , Streptococcus , Artrite Reumatoide/imunologia , Artrite Reumatoide/microbiologia , Animais , Tonsila Palatina/microbiologia , Tonsila Palatina/imunologia , Humanos , Camundongos , Artrite Experimental/imunologia , Artrite Experimental/microbiologia , Microbiota/imunologia , Streptococcus/imunologia , Masculino , Feminino , Disbiose/imunologia , Disbiose/microbiologia , Autoimunidade/imunologia , Pessoa de Meia-Idade , Camundongos Endogâmicos DBA
8.
PLoS Pathog ; 20(8): e1012056, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39208402

RESUMO

The Staphylococcus sp. are a dominant part of the human skin microbiome and present across the body. Staphylococcus epidermidis is a ubiquitous skin commensal, while S. aureus is thought to colonize at least 30% of the population. S. aureus are not only colonizers but a leading cause of skin and soft tissue infections and a critical healthcare concern. To understand how healthy human skin may differentiate commensal bacteria, such as S. epidermidis, from the potential pathogen methicillin-resistant S. aureus (MRSA), we use ex vivo human skin models that allow us to study this host-bacterial interaction in the most clinically relevant environment. Our work highlights the role of the outer stratum corneum as a protective physical barrier against invasion by colonizing Staphylococci. We show how the structural cells of the skin can internalize and respond to different Staphylococci with increasing sensitivity. In intact human skin, a discriminatory IL-1ß response was identified, while disruption of the protective stratum corneum triggered an increased and more diverse immune response. We identified and localized tissue resident Langerhans cells (LCs) as a potential source of IL-1ß and go on to show a dose-dependent response of MUTZ-LCs to S. aureus but not S. epidermidis. This suggests an important role of LCs in sensing and discriminating between bacteria in healthy human skin, particularly in intact skin and provides a detailed snapshot of how human skin differentiates between friend and potential foe. With the rise in antibiotic resistance, understanding the innate immune response of healthy skin may help us find ways to enhance or manipulate these natural defenses to prevent invasive infection.


Assuntos
Interleucina-1beta , Pele , Staphylococcus aureus , Staphylococcus epidermidis , Humanos , Interleucina-1beta/metabolismo , Pele/microbiologia , Pele/imunologia , Staphylococcus aureus/imunologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Células de Langerhans/imunologia , Células de Langerhans/microbiologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/imunologia , Microbiota/imunologia
9.
Int Immunopharmacol ; 141: 112984, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-39173404

RESUMO

BACKGROUND: The nasopharynx serves as a crucial niche for the microbiome of the upper respiratory tract. However, the association between the intratumoral microbiota and host systemic inflammation and immune status in nasopharyngeal carcinoma (NPC) remain uncertain. METHODS: We performed 5R 16S rDNA sequencing on NPC tissue samples, followed by diversity analysis, LEfSe differential analysis, and KEGG functional prediction. The analyses were based on indices such as AISI, SIRI, PAR, PLR, and NAR. Correlation analyses between microbes and these indices were performed to identify microbes associated with inflammation and immune status. Additionally, regression analysis based on tumor TNM stage was performed to identify key microbes linked to tumor progression. The head and neck squamous cell carcinoma (HNSC) transcriptome and the paired HNSC microbiome data from TCGA were utilized to validate the analyses. RESULTS: The Proteobacteria, Actinobacteria, Firmicutes, and Bacteroidetes were the most enriched phyla in NPC tissues. Microbes within these phyla demonstrated high sensitivity to changes in host systemic inflammation and immune status. Proteobacteria and Firmicutes showed significant differences between inflammation groups. Actinobacteria varied specifically with platelet-related inflammatory indices, and Bacteroidetes genera exhibited significant differences between NAR groups. Corynebacterium and Brevundimonas significantly impacted the T stage of tumors, with a high load of Corynebacterium within tumors associated with a better prognosis CONCLUSION: Our analysis indicates that Proteobacteria play a crucial role in the inflammatory state of NPC, while Bacteroidetes are more sensitive to the tumor immune status.


Assuntos
Inflamação , Microbiota , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Humanos , Carcinoma Nasofaríngeo/imunologia , Carcinoma Nasofaríngeo/microbiologia , Neoplasias Nasofaríngeas/imunologia , Neoplasias Nasofaríngeas/microbiologia , Microbiota/imunologia , Masculino , Inflamação/imunologia , Inflamação/microbiologia , Feminino , Pessoa de Meia-Idade , Adulto , RNA Ribossômico 16S/genética , Bactérias/classificação , Bactérias/imunologia , Bactérias/genética , Bactérias/isolamento & purificação , Idoso
10.
Autoimmun Rev ; 23(9): 103607, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39187222

RESUMO

Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by widespread inflammation affecting various organs. This review discusses the role of oxidative stress and gut microbiota in the pathogenesis of SLE and evaluates the therapeutic potential of intravenous immunoglobulins (IVIg). Oxidative stress contributes to SLE by causing impairment in the function of mitochondria, resulting in reactive oxygen species production, which triggers autoantigenicity and proinflammatory cytokines. Gut microbiota also plays a significant role in SLE. Dysbiosis has been associated to disease's onset and progression. Moreover, dysbiosis exacerbates SLE symptoms and influences systemic immunity, leading to a breakdown in bacterial tolerance and an increase in inflammatory responses. High-dose IVIg has emerged as a promising treatment for refractory cases of SLE. The beneficial effects of IVIg are partly due to its antioxidant property, reducing oxidative stress markers and modulating the immune responses. Additionally, IVIg can normalize the gut flora, as demonstrated in a case of severe intestinal pseudo-obstruction. In summary, both oxidative stress and dysregulation of microbiota are pivotal in the pathogenesis of SLE. The use of IVIg may improve the disease's outcome. Future research should be directed to elucidating the precise mechanisms by which oxidative stress and microbiota are linked with autoimmunity in SLE in developing targeted therapies.


Assuntos
Disbiose , Microbioma Gastrointestinal , Imunoglobulinas Intravenosas , Lúpus Eritematoso Sistêmico , Estresse Oxidativo , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Imunoglobulinas Intravenosas/uso terapêutico , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Disbiose/imunologia , Animais , Microbiota/imunologia , Microbiota/efeitos dos fármacos
11.
Transpl Immunol ; 86: 102102, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39094907

RESUMO

This comprehensive review delves into the intricate dynamics between the immune system and bacterial infections in organ transplant recipients. Its primary objective is to fill existing knowledge gaps while critically assessing the strengths and weaknesses of current research. The paper accentuates the delicate balance that must be struck between preventing graft rejection through immunosuppression and maintaining robust immunity against bacterial threats. In this context, personalized medicine emerges as a transformative concept, offering the potential to revolutionize clinical outcomes by tailoring immunosuppressive regimens and vaccination strategies to the unique profiles of transplant recipients. By emphasizing the pivotal role of continuous monitoring, the review underscores the necessity for vigilant surveillance of transplant recipients to detect bacterial infections and associated immune responses early, thereby reducing the risk of severe infections and ultimately improving patient outcomes. Furthermore, the study highlights the significance of the host microbiome in shaping immune responses, suggesting that interventions targeting the microbiome hold promise for enhancing bacterial immunity in transplant recipients, both in research and clinical practice. In terms of future research directions, the review advocates for large-scale, longitudinal studies encompassing diverse patient cohorts to provide more comprehensive insights into post-transplant immune responses. It also advocates integrating multi-omics approaches, including genomics, transcriptomics, proteomics, and microbiome data, to understand immune responses and their underlying mechanisms. In conclusion, this review significantly enriches our understanding of immune responses in transplant recipients. It paves the way for more effective and personalized approaches to managing infections in this complex setting.


Assuntos
Infecções Bacterianas , Transplante de Órgãos , Transplantados , Humanos , Infecções Bacterianas/imunologia , Transplante de Órgãos/efeitos adversos , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Animais , Microbiota/imunologia , Medicina de Precisão
12.
Curr Opin Immunol ; 89: 102452, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39180941

RESUMO

Microbiota-derived antigens play a critical role in the development of both the mucosal and systemic B cell repertoires; however, how glycan epitopes promote B cell repertoire selection is only recently being understood. The production of glycan-derived antigens by individual microbes within a host can be dynamic and influenced by interactions within other members of microbial communities, the composition of diet, and host-derived contents, including those of the mucosal immune system. The size and complexity of the emerging neonatal B cell repertoire are paralleled by the acquisition of a diverse microbiota from maternal and environmental sources, which is now appreciated to exert long-lasting influences on the nascent B cell repertoire.


Assuntos
Microbiota , Animais , Camundongos , Microbiota/imunologia , Linfócitos B/imunologia , Polissacarídeos/imunologia , Polissacarídeos/metabolismo , Humanos , Imunidade nas Mucosas
13.
Sci Rep ; 14(1): 18573, 2024 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-39127736

RESUMO

A resilient immune system is characterized by its capacity to respond appropriately to challenges, such as infections, and it is crucial in vaccine response. Here we report a paired randomized intervention-control trial in which we evaluated the effect of microbially rich soil on immune resilience and pneumococcal vaccine response. Twenty-five age and sex matched pairs of volunteers were randomized to intervention and control groups. The intervention group rubbed hands three times a day in microbially rich soil until participants received a pneumococcal vaccine on day 14. Vaccine response, skin and gut bacteriome and blood cytokine levels were analyzed on days 0, 14 and 35. Peripheral blood mononuclear cells (PBMCs) were stimulated with vaccine components and autoclaved soil for cytokine production. Commensal bacterial community shifted only in the intervention group during the 14-day intervention period. When PBMCs collected on day 14 before the vaccination were stimulated with the vaccine components, IFN-y production increased in the intervention but not in the control group. On day 35, vaccination induced a robust antibody response in both groups. In parallel, gut bacterial community was associated with TGF-ß plasma levels and TGF-ß decrease in plasma was lower in the intervention group. The results indicate that exposure to microbially rich soil can modulate the cell-mediated immunity to components in pneumococcal vaccine.


Assuntos
Imunidade Celular , Leucócitos Mononucleares , Vacinas Pneumocócicas , Pele , Humanos , Vacinas Pneumocócicas/imunologia , Vacinas Pneumocócicas/administração & dosagem , Masculino , Feminino , Pele/imunologia , Pele/microbiologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Adulto , Microbiologia do Solo , Citocinas/metabolismo , Citocinas/sangue , Microbioma Gastrointestinal/imunologia , Pessoa de Meia-Idade , Vacinação , Infecções Pneumocócicas/prevenção & controle , Infecções Pneumocócicas/imunologia , Microbiota/imunologia
14.
Nat Commun ; 15(1): 7096, 2024 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-39154092

RESUMO

The intratumor microbiome imbalance in pancreatic cancer promotes a tolerogenic immune response and triggers immunotherapy resistance. Here we show that Lactobacillus rhamnosus GG probiotics, outfitted with a gallium-polyphenol network (LGG@Ga-poly), bolster immunotherapy in pancreatic cancer by modulating microbiota-immune interactions. Upon oral administration, LGG@Ga-poly targets pancreatic tumors specifically, and selectively eradicates tumor-promoting Proteobacteria and microbiota-derived lipopolysaccharides through a gallium-facilitated disruption of bacterial iron respiration. This elimination of intratumor microbiota impedes the activation of tumoral Toll-like receptors, thus reducing immunosuppressive PD-L1 and interleukin-1ß expression by tumor cells, diminishing immunotolerant myeloid populations, and improving the infiltration of cytotoxic T lymphocytes in tumors. Moreover, LGG@Ga-poly hampers pancreatic tumor growth in both preventive and therapeutic contexts, and amplifies the antitumor efficacy of immune checkpoint blockade in preclinical cancer models in female mice. Overall, we offer evidence that thoughtfully designed biomaterials targeting intratumor microbiota can efficaciously augment immunotherapy for the challenging pancreatic cancer.


Assuntos
Gálio , Lacticaseibacillus rhamnosus , Microbiota , Neoplasias Pancreáticas , Polifenóis , Probióticos , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/microbiologia , Animais , Probióticos/administração & dosagem , Camundongos , Feminino , Humanos , Lacticaseibacillus rhamnosus/imunologia , Polifenóis/farmacologia , Microbiota/imunologia , Microbiota/efeitos dos fármacos , Linhagem Celular Tumoral , Imunoterapia/métodos , Camundongos Endogâmicos C57BL , Antígeno B7-H1/metabolismo , Antígeno B7-H1/imunologia , Linfócitos T Citotóxicos/imunologia
15.
Front Cell Infect Microbiol ; 14: 1409464, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39135638

RESUMO

Over the past decade, advancements in high-throughput sequencing technologies have led to a qualitative leap in our understanding of the role of the microbiota in human diseases, particularly in oncology. Despite the low biomass of the intratumoral microbiota, it remains a crucial component of the tumor immune microenvironment, displaying significant heterogeneity across different tumor tissues and individual patients. Although immunotherapy has emerged a major strategy for treating tumors, patient responses to these treatments vary widely. Increasing evidence suggests that interactions between the intratumoral microbiota and the immune system can modulate host tumor immune responses, thereby influencing the effectiveness of immunotherapy. Therefore, it is critical to gain a deep understanding of how the intratumoral microbiota shapes and regulates the tumor immune microenvironment. Here, we summarize the latest advancements on the role of the intratumoral microbiota in cancer immunity, exploring the potential mechanisms through which immune functions are influenced by intratumoral microbiota within and outside the gut barrier. We also discuss the impact of the intratumoral microbiota on the response to cancer immunotherapy and its clinical applications, highlighting future research directions and challenges in this field. We anticipate that the valuable insights into the interactions between cancer immunity and the intratumoral microbiota provided in this review will foster the development of microbiota-based tumor therapies.


Assuntos
Imunoterapia , Microbiota , Neoplasias , Microambiente Tumoral , Humanos , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/microbiologia , Imunoterapia/métodos , Microambiente Tumoral/imunologia , Microbiota/imunologia , Animais , Microbioma Gastrointestinal/imunologia
16.
J Reprod Immunol ; 165: 104298, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39002425

RESUMO

BACKGROUND: This study aims to conduct a preliminary exploration of the correlation between the oral microbiota of full-term pregnant women and both local placental immunity and the systemic immune system of the mother. METHODS: A total of 26 pregnant women participated in this study, with samples collected from oral swabs, placental tissue, and peripheral venous blood. High-throughput sequencing was used to examine the oral microbial community. Flow cytometry was employed to assess immune cells in placental tissue and peripheral venous blood. ELISA and Luminex liquid bead chip technology were utilized to detect cytokines in both placental tissue and peripheral venous blood. RESULTS: In placental tissue, The oral microbial community is primarily negatively correlated with placental CD3+CD4+CD8+T cells and positively correlated with placental IL-5. In the peripheral blood, The oral microbial community is primarily positively correlated with maternal systemic immune parameters, including CD3+CD4+ T cells and the CD4+/CD8+ ratio, as well as positively correlated with peripheral IL-18. CONCLUSIONS: The oral microbiota of full-term pregnant women participates in the regulatory function of the maternal immune system. Meanwhile, the oral microbial community may also be an important factor mediating local immune regulation in the placenta.


Assuntos
Microbiota , Placenta , Humanos , Feminino , Gravidez , Microbiota/imunologia , Adulto , Placenta/imunologia , Placenta/microbiologia , Boca/microbiologia , Boca/imunologia , Interleucina-5/imunologia , Interleucina-5/metabolismo
17.
JCI Insight ; 9(16)2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39024554

RESUMO

Anal squamous cell carcinoma (ASCC) is a rare gastrointestinal malignancy linked to high-risk human papillomavirus (HPV) infection, which develops from precursor lesions like low-grade squamous intraepithelial lesions and high-grade squamous intraepithelial lesions (HGSILs). ASCC incidence varies across populations and poses increased risk for people living with HIV. Our investigation focused on transcriptomic and metatranscriptomic changes from squamous intraepithelial lesions to ASCC. Metatranscriptomic analysis highlighted specific bacterial species (e.g., Fusobacterium nucleatum, Bacteroides fragilis) more prevalent in ASCC than precancerous lesions. These species correlated with gene-encoding enzymes (Acca, glyQ, eno, pgk, por) and oncoproteins (FadA, dnaK), presenting potential diagnostic or treatment markers. Unsupervised transcriptomic analysis identified distinct sample clusters reflecting histological diagnosis, immune infiltrate, HIV/HPV status, and pathway activities, recapitulating anal cancer progression's natural history. Our study unveiled molecular mechanisms in anal cancer progression, aiding in stratifying HGSIL cases based on low or high risk of progression to malignancy.


Assuntos
Neoplasias do Ânus , Carcinoma de Células Escamosas , Transcriptoma , Humanos , Neoplasias do Ânus/genética , Neoplasias do Ânus/imunologia , Neoplasias do Ânus/patologia , Neoplasias do Ânus/virologia , Neoplasias do Ânus/microbiologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/microbiologia , Carcinoma de Células Escamosas/patologia , Microbiota/imunologia , Masculino , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/virologia , Infecções por Papillomavirus/imunologia , Lesões Intraepiteliais Escamosas/genética , Lesões Intraepiteliais Escamosas/patologia , Lesões Intraepiteliais Escamosas/virologia , Feminino , Progressão da Doença , Pessoa de Meia-Idade , Infecções por HIV/complicações , Infecções por HIV/imunologia
18.
Adv Exp Med Biol ; 1449: 1-28, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39060728

RESUMO

Given that the host-microbe interaction is shaped by the immune system response, it is important to understand the key immune system-microbiota relationship during the period from conception to the first years of life. The present work summarizes the available evidence concerning human reproductive microbiota, and also, the microbial colonization during early life, focusing on the potential impact on infant development and health outcomes. Furthermore, we conclude that some dietary strategies including specific probiotics and other-biotics could become potentially valuable tools to modulate the maternal-neonatal microbiota during this early critical window of opportunity for targeted health outcomes throughout the entire lifespan.


Assuntos
Microbiota , Probióticos , Humanos , Lactente , Recém-Nascido , Feminino , Microbiota/fisiologia , Microbiota/imunologia , Gravidez , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Desenvolvimento Infantil/fisiologia , Interações entre Hospedeiro e Microrganismos/imunologia , Interações entre Hospedeiro e Microrganismos/fisiologia
19.
Arch Dermatol Res ; 316(8): 495, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39073596

RESUMO

The scalp microbiome represents an array of microorganisms important in maintaining scalp homeostasis and mediating inflammation. Scalp microbial dysregulation has been implicated in dermatologic conditions including alopecia areata (AA), dandruff/seborrheic dermatitis (D/SD), scalp psoriasis (SP) and folliculitis decalvans (FD). Understanding the impact of scalp microbial dysbiosis gives insight on disease pathophysiology and guides therapeutic decision making. Herein we review the scalp microbiome and its functional role in scalp conditions by analysis of metagenomic medical literature in alopecia, D/SD, SP, and other dermatologic disease.Increased abundance of Malassezia, Staphylococcus, and Brevibacterium was associated with SD compared to healthy controls. A higher proportion of Corynebacterium, actinobacteria, and firmicutes are present in AA patients, and lower proportions of Staphylococcus caprae are associated with worse clinical outcomes. Decreased prevalence of actinobacteria and Propionibacterium and increased firmicutes, staphylococcus, and streptococcus are associated with scalp psoriasis. Studies of central centrifugal cicatricial alopecia (CCCA) suggest scalp microbial composition contributes to CCCA's pro-inflammatory status. The most common organisms associated with FD include methicillin-resistant S. aureus and S. lugdunensis. Antifungals have been a mainstay treatment for these diseases, while other alternatives including coconut oils and shampoos with heat-killed probiotics have shown considerable potential efficacy by replenishing the scalp microbiome.


Assuntos
Microbiota , Couro Cabeludo , Humanos , Microbiota/efeitos dos fármacos , Microbiota/imunologia , Couro Cabeludo/microbiologia , Dermatoses do Couro Cabeludo/microbiologia , Dermatoses do Couro Cabeludo/tratamento farmacológico , Dermatoses do Couro Cabeludo/terapia , Disbiose/microbiologia , Disbiose/imunologia , Foliculite/microbiologia , Foliculite/diagnóstico , Foliculite/tratamento farmacológico , Foliculite/terapia , Psoríase/microbiologia , Psoríase/tratamento farmacológico , Psoríase/imunologia , Psoríase/terapia , Dermatite Seborreica/microbiologia , Dermatite Seborreica/tratamento farmacológico , Dermatite Seborreica/terapia , Alopecia em Áreas/microbiologia , Alopecia em Áreas/imunologia , Alopecia em Áreas/terapia , Alopecia em Áreas/tratamento farmacológico , Caspa/microbiologia , Caspa/tratamento farmacológico
20.
Front Immunol ; 15: 1352018, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38989282

RESUMO

In this study, we investigated how Radix pseudostellariae polysaccharide (RPP) enhances the immune response of the inactivated porcine reproductive and respiratory syndrome virus (PRRSV) vaccine through interactions with the microbiome and metabolome. We pretreated sows with 10 mg/kg body weight of RPP via drinking water for 7 days prior to intramuscular injection of the PRRSV vaccine. This significantly increased the concentrations of PRRSV GP5 protein antibody, interleukin (IL)-2, IL-4, IL-10, and interferon (IFN)-γ. Oral administration of RPP also significantly improved the abundance of beneficial bacteria in the stool, such as Parabacteroides distasonis, Prevotella_copri, Eubacterium_sp., and Clostridium_sp._CAG:226, and decreased the levels of potentially pathogenic bacteria, such as Paraeggerthella and [Clostridium] innocuum, compared to the vaccine alone. These bacterial changes were confirmed using quantitative real-time polymerase chain reaction (Q-PCR). Moreover, RPP treatment significantly increased the blood concentrations of L-theanine, taurodeoxycholic acid (TDCA), and N-arachidonoyl proline, and decreased the levels of L-glutamine, oclacitinib, lipoxin C4, and leukotriene C5 in sows after immunization (p< 0.05). The concentrations of various blood metabolites were validated using sandwich enzyme-linked immunosorbent assay (ELISA), confirming the accuracy of the metabolomics data. Intriguingly, the integration of microbiome and metabolome analyses highlighted the significance of Prevotella_copri and TDCA. We consequently developed a mouse immunity model using GP5 protein and discovered that oral administration of RPP significantly enhanced the levels of GP5 protein antibodies, IL-2, IL-4, IL-10, and IFN-γ in mouse serum. It also increased the number of CD3+ and CD3+CD4+ cells in the spleen. Additionally, Prevotella_copri was administered into the large intestine via the anus for 7 days prior to the intramuscular injection of the PRRSV GP5 protein. The results demonstrated a significant increase in TDCA and GP5 antibody concentration in the mouse serum, indicating that RPP modulates Prevotella_copri to elevate its metabolite TDCA, thereby enhancing the GP5 antibody level. In conclusion, oral administration of 10 mg/kg RPP optimizes gut flora diversity and blood metabolites, particularly Prevotella_copri and TDCA, thereby improving the immune response to the inactivated PRRSV vaccine.


Assuntos
Metaboloma , Polissacarídeos , Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Vacinas de Produtos Inativados , Vacinas Virais , Animais , Suínos , Vírus da Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Vacinas Virais/imunologia , Feminino , Vacinas de Produtos Inativados/imunologia , Anticorpos Antivirais/sangue , Citocinas/metabolismo , Microbiota/efeitos dos fármacos , Microbiota/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Adjuvantes Imunológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...