Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.432
Filter
1.
Sci Rep ; 14(1): 18573, 2024 08 10.
Article in English | MEDLINE | ID: mdl-39127736

ABSTRACT

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.


Subject(s)
Immunity, Cellular , Leukocytes, Mononuclear , Pneumococcal Vaccines , Skin , Humans , Pneumococcal Vaccines/immunology , Pneumococcal Vaccines/administration & dosage , Male , Female , Skin/immunology , Skin/microbiology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Adult , Soil Microbiology , Cytokines/metabolism , Cytokines/blood , Gastrointestinal Microbiome/immunology , Middle Aged , Vaccination , Pneumococcal Infections/prevention & control , Pneumococcal Infections/immunology , Microbiota/immunology
2.
Front Cell Infect Microbiol ; 14: 1409464, 2024.
Article in English | MEDLINE | ID: mdl-39135638

ABSTRACT

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.


Subject(s)
Immunotherapy , Microbiota , Neoplasms , Tumor Microenvironment , Humans , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/microbiology , Immunotherapy/methods , Tumor Microenvironment/immunology , Microbiota/immunology , Animals , Gastrointestinal Microbiome/immunology
3.
Nat Commun ; 15(1): 7096, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154092

ABSTRACT

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.


Subject(s)
Gallium , Lacticaseibacillus rhamnosus , Microbiota , Pancreatic Neoplasms , Polyphenols , Probiotics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/microbiology , Animals , Probiotics/administration & dosage , Mice , Female , Humans , Lacticaseibacillus rhamnosus/immunology , Polyphenols/pharmacology , Microbiota/immunology , Microbiota/drug effects , Cell Line, Tumor , Immunotherapy/methods , Mice, Inbred C57BL , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , T-Lymphocytes, Cytotoxic/immunology
5.
Arch Dermatol Res ; 316(8): 495, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073596

ABSTRACT

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.


Subject(s)
Microbiota , Scalp , Humans , Microbiota/drug effects , Microbiota/immunology , Scalp/microbiology , Scalp Dermatoses/microbiology , Scalp Dermatoses/drug therapy , Scalp Dermatoses/therapy , Dysbiosis/microbiology , Dysbiosis/immunology , Folliculitis/microbiology , Folliculitis/diagnosis , Folliculitis/drug therapy , Folliculitis/therapy , Psoriasis/microbiology , Psoriasis/drug therapy , Psoriasis/immunology , Psoriasis/therapy , Dermatitis, Seborrheic/microbiology , Dermatitis, Seborrheic/drug therapy , Dermatitis, Seborrheic/therapy , Alopecia Areata/microbiology , Alopecia Areata/immunology , Alopecia Areata/therapy , Alopecia Areata/drug therapy , Dandruff/microbiology , Dandruff/drug therapy
7.
Front Immunol ; 15: 1352018, 2024.
Article in English | MEDLINE | ID: mdl-38989282

ABSTRACT

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.


Subject(s)
Metabolome , Polysaccharides , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Vaccines, Inactivated , Viral Vaccines , Animals , Swine , Porcine respiratory and reproductive syndrome virus/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/prevention & control , Viral Vaccines/immunology , Female , Vaccines, Inactivated/immunology , Antibodies, Viral/blood , Cytokines/metabolism , Microbiota/drug effects , Microbiota/immunology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Adjuvants, Immunologic
8.
Front Immunol ; 15: 1415565, 2024.
Article in English | MEDLINE | ID: mdl-38989285

ABSTRACT

How the microbiome regulates responses of systemic innate immune cells is unclear. In the present study, our purpose was to document a novel mechanism by which the microbiome mediates crosstalk with the systemic innate immune system. We have identified a family of microbiome Bacteroidota-derived lipopeptides-the serine-glycine (S/G) lipids, which are TLR2 ligands, access the systemic circulation, and regulate proinflammatory responses of splenic monocytes. To document the role of these lipids in regulating systemic immunity, we used oral gavage with an antibiotic to decrease the production of these lipids and administered exogenously purified lipids to increase the systemic level of these lipids. We found that decreasing systemic S/G lipids by decreasing microbiome Bacteroidota significantly enhanced splenic monocyte proinflammatory responses. Replenishing systemic levels of S/G lipids via exogenous administration returned splenic monocyte responses to control levels. Transcriptomic analysis demonstrated that S/G lipids regulate monocyte proinflammatory responses at the level of gene expression of a small set of upstream inhibitors of TLR and NF-κB pathways that include Trem2 and Irf4. Consistent with enhancement in proinflammatory cytokine responses, decreasing S/G lipids lowered gene expression of specific pathway inhibitors. Replenishing S/G lipids normalized gene expression of these inhibitors. In conclusion, our results suggest that microbiome-derived S/G lipids normally establish a level of buffered signaling activation necessary for well-regulated innate immune responses in systemic monocytes. By regulating gene expression of inflammatory pathway inhibitors such as Trem2, S/G lipids merit broader investigation into the potential dysfunction of other innate immune cells, such as microglia, in diseases such as Alzheimer's disease.


Subject(s)
Monocytes , Signal Transduction , Monocytes/immunology , Monocytes/metabolism , Monocytes/drug effects , Animals , Mice , Microbiota/immunology , Mice, Inbred C57BL , Immunity, Innate , Toll-Like Receptor 2/metabolism , Gene Expression Regulation/drug effects , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Lipopeptides/pharmacology , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , NF-kappa B/metabolism , Inflammation/immunology , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Male , Lipids , Spleen/immunology , Spleen/metabolism , Cytokines/metabolism , Female
9.
Front Immunol ; 15: 1389446, 2024.
Article in English | MEDLINE | ID: mdl-39034996

ABSTRACT

Microbiota has been closely related to human beings, whose role in tumor development has also been widely investigated. However, previous studies have mainly focused on the gut, oral, and/or skin microbiota. In recent years, the study of intratumoral microbiota has become a hot topic in tumor-concerning studies. Intratumoral microbiota plays an important role in the occurrence, development, and response to treatment of malignant tumors. In fact, increasing evidence has suggested that intratumoral microbiota is associated with malignant tumors in various ways, such as promoting the tumor development and affecting the efficacy of chemotherapy and immunotherapy. In this review, the impact of intratumoral microbiota on the immune microenvironment of malignant tumors has been analyzed, as well as its role in tumor immunotherapy, with the hope that it may contribute to the development of diagnostic tools and treatments for related tumors in the future.


Subject(s)
Immunotherapy , Microbiota , Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/microbiology , Immunotherapy/methods , Microbiota/immunology , Animals
10.
Adv Exp Med Biol ; 1449: 1-28, 2024.
Article in English | MEDLINE | ID: mdl-39060728

ABSTRACT

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.


Subject(s)
Microbiota , Probiotics , Humans , Infant , Infant, Newborn , Female , Microbiota/physiology , Microbiota/immunology , Pregnancy , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Child Development/physiology , Host Microbial Interactions/immunology , Host Microbial Interactions/physiology
11.
JCI Insight ; 9(16)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39024554

ABSTRACT

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.


Subject(s)
Anus Neoplasms , Carcinoma, Squamous Cell , Transcriptome , Humans , Anus Neoplasms/genetics , Anus Neoplasms/immunology , Anus Neoplasms/pathology , Anus Neoplasms/virology , Anus Neoplasms/microbiology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Microbiota/immunology , Male , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Papillomavirus Infections/immunology , Squamous Intraepithelial Lesions/genetics , Squamous Intraepithelial Lesions/pathology , Squamous Intraepithelial Lesions/virology , Female , Disease Progression , Middle Aged , HIV Infections/complications , HIV Infections/immunology
13.
Nat Rev Clin Oncol ; 21(8): 569-589, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38840029

ABSTRACT

Immunotherapy has revolutionized the treatment of cancer but continues to be constrained by limited response rates, acquired resistance, toxicities and high costs, which necessitates the development of new, innovative strategies. The discovery of a connection between the human microbiota and cancer dates back 4,000 years, when local infection was observed to result in tumour eradication in some individuals. However, the true oncological relevance of the intratumoural microbiota was not recognized until the turn of the twentieth century. The intratumoural microbiota can have pivotal roles in both the pathogenesis and treatment of cancer. In particular, intratumoural bacteria can either promote or inhibit cancer growth via remodelling of the tumour microenvironment. Over the past two decades, remarkable progress has been made preclinically in engineering bacteria as agents for cancer immunotherapy; some of these bacterial products have successfully reached the clinical stages of development. In this Review, we discuss the characteristics of intratumoural bacteria and their intricate interactions with the tumour microenvironment. We also describe the many strategies used to engineer bacteria for use in the treatment of cancer, summarizing contemporary data from completed and ongoing clinical trials. The work described herein highlights the potential of bacteria to transform the landscape of cancer therapy, bridging ancient wisdom with modern scientific innovation.


Subject(s)
Bacteria , Immunotherapy , Neoplasms , Tumor Microenvironment , Humans , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/microbiology , Immunotherapy/methods , Tumor Microenvironment/immunology , Bacteria/immunology , Microbiota/immunology
14.
Oncogene ; 43(31): 2389-2404, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38890429

ABSTRACT

The role of tumor-resident microbiota in modulating tumor immunity remains unclear. Here, we discovered an abundance of intra-tumoral bacteria, such us E.coli, residing and resulting in Colorectal cancer liver metastasis (CRLM). E.coli enhanced lactate production, which mediated M2 macrophage polarization by suppressing nuclear factor-κB -gene binding (NF-κB) signaling through retinoic acid-inducible gene 1 (RIG-I) lactylation. Lactylation of RIG-I suppressed recruitment of NF-κB to the Nlrp3 promoter in macrophages, thereby reducing its transcription. This loss of Nlrp3 affected the immunosuppressive activities of regulatory T cells (Tregs) and the antitumor activities of and CD8+ T cells. Small-molecule compound screening identified a RIG-I lactylation inhibitor that suppressed M2 polarization and sensitized CRLM to 5-fluorouracil (5-FU). Our findings suggest that tumor-resident microbiota may be a potential target for preventing and treating CRLM.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , NF-kappa B , Colorectal Neoplasms/pathology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/immunology , Liver Neoplasms/secondary , Liver Neoplasms/microbiology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Animals , Humans , Mice , NF-kappa B/metabolism , Microbiota/immunology , Macrophages/immunology , Macrophages/metabolism , Escherichia coli , T-Lymphocytes, Regulatory/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Cell Line, Tumor , Fluorouracil/pharmacology , Signal Transduction
15.
Int Immunopharmacol ; 137: 112537, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38909493

ABSTRACT

Colorectal cancer (CRC) has been one of the most common malignancies worldwide. Despite the advances in current therapies, the mortality rate of CRC remains high. Among them, immunotherapy has achieved satisfactory results in some CRC patients, however, how to expand the use of immunotherapy in CRC patients remains an urgent challenge. Surprisingly, the intratumoral microbiota has been found in multiple tumor tissues, including CRC. It has been demonstrated that the intratumoral microbiota is associated with the progression and treatment of CRC, and is able to enhance or decrease anti-tumor immune responses via different mechanisms as well as influence the immunotherapy efficacy, providing new potential therapeutic targets for CRC immunotherapy. In this review, we focus on the characteristics of the intratumoral microbiota, its roles in the genesis and development of CRC, its modulation of anti-tumor immune responses and immunotherapy, and propose potential applications of the intratumoral microbiota in CRC immunotherapy. Additionally, we propose possible directions for future research on the intratumoral microbiota related to CRC immunotherapy.


Subject(s)
Colorectal Neoplasms , Immunotherapy , Humans , Colorectal Neoplasms/immunology , Colorectal Neoplasms/therapy , Colorectal Neoplasms/microbiology , Immunotherapy/methods , Animals , Microbiota/immunology , Tumor Microenvironment/immunology , Gastrointestinal Microbiome/immunology
16.
Curr Allergy Asthma Rep ; 24(8): 415-423, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38904934

ABSTRACT

PURPOSE OF REVIEW: The human commensal microbiota is now widely accepted as a key regulator of human health and disease. The composition of the mucosal associated microbiota has been shown to play a critical role in the lung health. The role of the mucosal microbiota in the development and severity of allergy, asthma, and occupational lung disease is only beginning to take shape. However, advances in our understanding of these links have tremendous potential to led to new clinical interventions to reduce allergy, asthma, and occupational lung disease morbidity. RECENT FINDINGS: We review recent work describing the relationship and role of the commensal microbiota in the development of allergy, asthma, and occupational lung disease. Our review primarily focuses on occupational exposures and the effects of the microbiome, both in composition and function. Data generated from these studies may lead to the development of interventions targeted at establishing and maintaining a healthy microbiota. We also highlight the role of environmental exposures and the effects on the commensal microbial community and their potential association with occupational lung disease. This review explores the current research describing the role of the human microbiome in the regulation of pulmonary health and disease, with a specific focus on the role of the mucosal microbiota in the development of allergy, asthma, and occupational lung disease.


Subject(s)
Asthma , Hypersensitivity , Microbiota , Occupational Diseases , Humans , Microbiota/immunology , Asthma/immunology , Asthma/microbiology , Hypersensitivity/immunology , Hypersensitivity/microbiology , Occupational Diseases/microbiology , Occupational Diseases/immunology , Occupational Exposure/adverse effects , Lung Diseases/microbiology , Lung Diseases/immunology
17.
Immunity ; 57(7): 1648-1664.e9, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38876098

ABSTRACT

Allogeneic T cell expansion is the primary determinant of graft-versus-host disease (GVHD), and current dogma dictates that this is driven by histocompatibility antigen disparities between donor and recipient. This paradigm represents a closed genetic system within which donor T cells interact with peptide-major histocompatibility complexes (MHCs), though clonal interrogation remains challenging due to the sparseness of the T cell repertoire. We developed a Bayesian model using donor and recipient T cell receptor (TCR) frequencies in murine stem cell transplant systems to define limited common expansion of T cell clones across genetically identical donor-recipient pairs. A subset of donor CD4+ T cell clonotypes differentially expanded in identical recipients and were microbiota dependent. Microbiota-specific T cells augmented GVHD lethality and could target microbial antigens presented by gastrointestinal epithelium during an alloreactive response. The microbiota serves as a source of cognate antigens that contribute to clonotypic T cell expansion and the induction of GVHD independent of donor-recipient genetics.


Subject(s)
Graft vs Host Disease , Graft vs Host Disease/immunology , Graft vs Host Disease/microbiology , Animals , Mice , Mice, Inbred C57BL , CD4-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Microbiota/immunology , Clonal Selection, Antigen-Mediated , Transplantation, Homologous , Bayes Theorem , Stem Cell Transplantation/adverse effects , Mice, Inbred BALB C , Gastrointestinal Microbiome/immunology , Hematopoietic Stem Cell Transplantation/adverse effects
18.
PeerJ ; 12: e17415, 2024.
Article in English | MEDLINE | ID: mdl-38881859

ABSTRACT

Background: Cancer has surpassed infectious diseases and heart ailments, taking the top spot in the disease hierarchy. Cervical cancer is a significant concern for women due to high incidence and mortality rates, linked to the human papillomavirus (HPV). HPV infection leads to precancerous lesions progressing to cervical cancer. The cervix's external os, near the vagina, hosts various microorganisms. Evidence points to the link between vaginal microbiota and HPV-induced cervical cancer. Cervical cancer onset aligns with an imbalanced Th1/Th2 immune response, but the role of vaginal microbiota in modulating this imbalance is unclear. Methods: In this study, we collected vaginal samples from 99 HPV-infected patients across varying degrees of lesions, alongside control groups. These samples underwent bacterial DNA sequencing. Additionally, we employed Elisa kits to quantify the protein expression levels of Th1/Th2 cytokines IL2, IL12, IL5, IL13, and TNFa within the centrifuged supernatant of vaginal-cervical secretions from diverse research subjects. Subsequently, correlation analyses were conducted between inflammatory factors and vaginal microbiota. Results: Our findings highlighted a correlation between decreased Lactobacillus and increased Gardenerella presence with HPV-induced cervical cancer. Functionally, our predictive analysis revealed the predominant enrichment of the ABC transporter within the vaginal microbiota of cervical cancer patients. Notably, these microbiota alterations exhibited correlations with the production of Th1/Th2 cytokines, which are intimately tied to tumor immunity. Conclusions: This study suggests the potential involvement of vaginal microbiota in the progression of HPV-induced cervical cancer through Th1/Th2 cytokine regulation. This novel insight offers a fresh perspective for early cervical cancer diagnosis and future prevention strategies.


Subject(s)
Microbiota , Papillomavirus Infections , Uterine Cervical Neoplasms , Vagina , Humans , Female , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/microbiology , Uterine Cervical Neoplasms/pathology , Vagina/microbiology , Vagina/immunology , Vagina/virology , Microbiota/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Adult , Inflammation/immunology , Inflammation/microbiology , Middle Aged , Cytokines/metabolism , Cervix Uteri/microbiology , Cervix Uteri/immunology , Cervix Uteri/virology
19.
Front Immunol ; 15: 1401320, 2024.
Article in English | MEDLINE | ID: mdl-38835769

ABSTRACT

Host-microbe interactions are complex and ever-changing, especially during infections, which can significantly impact human physiology in both health and disease by influencing metabolic and immune functions. Infections caused by pathogens such as bacteria, viruses, fungi, and parasites are the leading cause of global mortality. Microbes have evolved various immune evasion strategies to survive within their hosts, which presents a multifaceted challenge for detection. Intracellular microbes, in particular, target specific cell types for survival and replication and are influenced by factors such as functional roles, nutrient availability, immune evasion, and replication opportunities. Identifying intracellular microbes can be difficult because of the limitations of traditional culture-based methods. However, advancements in integrated host microbiome single-cell genomics and transcriptomics provide a promising basis for personalized treatment strategies. Understanding host-microbiota interactions at the cellular level may elucidate disease mechanisms and microbial pathogenesis, leading to targeted therapies. This article focuses on how intracellular microbes reside in specific cell types, modulating functions through persistence strategies to evade host immunity and prolong colonization. An improved understanding of the persistent intracellular microbe-induced differential disease outcomes can enhance diagnostics, therapeutics, and preventive measures.


Subject(s)
Genomics , Single-Cell Analysis , Humans , Genomics/methods , Animals , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/genetics , Host Microbial Interactions/immunology , Host Microbial Interactions/genetics , Immune Evasion , Microbiota/immunology , Bacteria/genetics , Bacteria/immunology , Severity of Illness Index
20.
Front Immunol ; 15: 1385436, 2024.
Article in English | MEDLINE | ID: mdl-38919613

ABSTRACT

Periodontal disease is a chronic inflammatory condition that affects the supporting structures of the teeth, including the periodontal ligament and alveolar bone. Periodontal disease is due to an immune response that stimulates gingivitis and periodontitis, and its systemic consequences. This immune response is triggered by bacteria and may be modulated by environmental conditions such as smoking or systemic disease. Recent advances in single cell RNA-seq (scRNA-seq) and in vivo animal studies have provided new insight into the immune response triggered by bacteria that causes periodontitis and gingivitis. Dysbiosis, which constitutes a change in the bacterial composition of the microbiome, is a key factor in the initiation and progression of periodontitis. The host immune response to dysbiosis involves the activation of various cell types, including keratinocytes, stromal cells, neutrophils, monocytes/macrophages, dendritic cells and several lymphocyte subsets, which release pro-inflammatory cytokines and chemokines. Periodontal disease has been implicated in contributing to the pathogenesis of several systemic conditions, including diabetes, rheumatoid arthritis, cardiovascular disease and Alzheimer's disease. Understanding the complex interplay between the oral microbiome and the host immune response is critical for the development of new therapeutic strategies for the prevention and treatment of periodontitis and its systemic consequences.


Subject(s)
Alveolar Bone Loss , Dysbiosis , Periodontitis , Humans , Periodontitis/immunology , Periodontitis/microbiology , Animals , Alveolar Bone Loss/immunology , Alveolar Bone Loss/etiology , Alveolar Bone Loss/microbiology , Dysbiosis/immunology , Microbiota/immunology
SELECTION OF CITATIONS
SEARCH DETAIL