Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.389
Filter
1.
Front Immunol ; 15: 1352469, 2024.
Article in English | MEDLINE | ID: mdl-38711504

ABSTRACT

Vibriosis, caused by Vibrio, seriously affects the health of fish, shellfish, and shrimps, causing large economic losses. Teleosts are represent the first bony vertebrates with both innate and adaptive immune responses against pathogens. Aquatic animals encounter hydraulic pressure and more pathogens, compared to terrestrial animals. The skin is the first line of defense in fish, constituting the skin-associated lymphoid tissue (SALT), which belongs to the main mucosa-associated lymphoid tissues (MALT). However, little is known about the function of immunity related proteins in fish. Therefore, this study used iTRAQ (isobaric tags for relative and absolute quantitation) to compare the skin proteome between the resistant and susceptible families of Cynoglossus semilaevis. The protein integrin beta-2, the alpha-enolase isoform X1, subunit B of V-type proton ATPase, eukaryotic translation initiation factor 6, and ubiquitin-like protein ISG15, were highly expressed in the resistant family. The 16S sequencing of the skin tissues of the resistant and susceptible families showed significant differences in the microbial communities of the two families. The protein-microbial interaction identified ten proteins associated with skin microbes, including immunoglobulin heavy chain gene (IGH), B-cell lymphoma/leukemia 10 (BCL10) and pre-B-cell leukemia transcription factor 1 isoform X2 (PBX2). This study highlights the interaction between skin proteins and the microbial compositions of C. semilaevis and provides new insights into understanding aquaculture breeding research.


Subject(s)
Disease Resistance , Fish Diseases , Fish Proteins , Flatfishes , Microbiota , Skin , Vibrio Infections , Vibrio , Animals , Skin/immunology , Skin/microbiology , Skin/metabolism , Fish Diseases/immunology , Fish Diseases/microbiology , Disease Resistance/immunology , Vibrio Infections/immunology , Vibrio Infections/veterinary , Flatfishes/immunology , Flatfishes/microbiology , Microbiota/immunology , Vibrio/immunology , Fish Proteins/genetics , Fish Proteins/metabolism , Fish Proteins/immunology , Proteome , Proteomics/methods
4.
Medicine (Baltimore) ; 103(20): e38078, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758914

ABSTRACT

Microorganisms, including bacteria, viruses, and fungi, coexist in the human body, forming a symbiotic microbiota that plays a vital role in human health and disease. Intratumoral microbial components have been discovered in various tumor tissues and are closely linked to the occurrence, progression, and treatment results of cancer. The intratumoral microbiota can enhance antitumor immunity through mechanisms such as activating the stimulator of interferon genes signaling pathway, stimulating T and NK cells, promoting the formation of TLS, and facilitating antigen presentation. Conversely, the intratumoral microbiota might suppress antitumor immune responses by increasing reactive oxygen species levels, creating an anti-inflammatory environment, inducing T cell inactivation, and enhancing immune suppression, thereby promoting cancer progression. The impact of intratumoral microbiota on antitumor immunity varies based on microbial composition, interactions with cancer cells, and the cancer's current state. A deep understanding of the complex interactions between intratumoral microbiota and antitumor immunity holds the potential to bring new therapeutic strategies and targets to cancer immunotherapy.


Subject(s)
Immunotherapy , Microbiota , Neoplasms , Humans , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/microbiology , Immunotherapy/methods , Microbiota/immunology , Tumor Microenvironment/immunology
5.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673883

ABSTRACT

Respiratory infections are common in pregnancy with conflicting evidence supporting their association with neonatal congenital anomalies, especially during the first trimester. We profiled cytokine and chemokine systemic responses in 242 pregnant women and their newborns after SARS-CoV-2 infection, acquired in different trimesters. Also, we tested transplacental IgG passage and maternal vaginal-rectal microbiomes. IgG transplacental passage was evident, especially with infection acquired in the first trimester. G-CSF concentration-involved in immune cell recruitment-decreased in infected women compared to uninfected ones: a beneficial event for the reduction of inflammation but detrimental to ability to fight infections at birth. The later the infection was acquired, the higher the systemic concentration of IL-8, IP-10, and MCP-1, associated with COVID-19 disease severity. All infected women showed dysbiosis of vaginal and rectal microbiomes, compared to uninfected ones. Two newborns tested positive for SARS-CoV-2 within the first 48 h of life. Notably, their mothers had acute infection at delivery. Although respiratory infections in pregnancy are reported to affect babies' health, with SARS-CoV-2 acquired early during gestation this risk seems low because of the maternal immune response. The observed vaginal and rectal dysbiosis could be relevant for neonatal microbiome establishment, although in our series immediate neonatal outcomes were reassuring.


Subject(s)
COVID-19 , Dysbiosis , Pregnancy Complications, Infectious , SARS-CoV-2 , Vagina , Humans , Female , Pregnancy , COVID-19/immunology , Dysbiosis/immunology , Dysbiosis/microbiology , Adult , SARS-CoV-2/immunology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/virology , Vagina/microbiology , Vagina/immunology , Vagina/virology , Infant, Newborn , Cytokines/metabolism , Pregnancy Trimesters/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Microbiota/immunology
6.
J Invest Dermatol ; 144(5): 1001-1009, 2024 May.
Article in English | MEDLINE | ID: mdl-38573278

ABSTRACT

Whereas clinically apparent atopic dermatitis (AD) can be confirmed by validated diagnostic criteria, the preclinical phenotype of infants who eventually develop AD is less well-characterized. Analogous to unaffected or nonlesional skin in established AD, clinically normal-appearing skin in infants who will develop clinical AD has distinct changes. Prospective studies have revealed insights into this preclinical AD phenotype. In this study, we review the structural, immunologic, and microbiome nature of the preclinical AD phenotype. Determination of markers that predict the development of AD will facilitate targeting of interventions to prevent the development or reduce the severity of AD in infants.


Subject(s)
Dermatitis, Atopic , Skin , Humans , Infant , Biomarkers/analysis , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/diagnosis , Microbiota/immunology , Phenotype , Severity of Illness Index , Skin/microbiology , Skin/immunology , Skin/pathology
7.
Int Immunopharmacol ; 133: 112097, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38677092

ABSTRACT

BACKGROUNDS: The Gustave Roussy Immune (GRIm) score predicts survival outcomes in several cancers. However, the prognostic significance of the GRIm score in patients with malignant ascites has not yet been investigated. METHODS: Clinical samples were collected from a cohort of patients with malignant ascites secondary to hepatocellular carcinoma (HCC). We calculated serum GRIm (sGRIm) and ascites GRIm (aGRIm) scores and divided the samples into low and high GRIm score groups. Survival analysis was used to compare the prognostic significance of the sGRIm and aGRIm scores. 16S rRNA sequencing was performed to determine the profiles of the intratumoral microbiota in the groups. A fluorescent multiplex immunohistochemistry (mIHC) assay was used to detect the expression of different immune cells by labeling seven markers of malignant ascites. RESULTS: 155 patients with HCC and malignant ascites were enrolled in this study. Survival analysis revealed that the aGRIm score showed a superior prognostic significance compared to the sGRIm score. Microbial analysis demonstrated that the bacterial richness and diversity were higher in the low aGRIm score group than in the high aGRIm score group. LefSe analysis revealed that certain bacteria were correlated with high aGRIm scores. Fluorescent mIHC displayed the tumor microenvironment of malignant ascites and found that the density of CD8 + T cells was significantly higher in the low aGRIm score group than in the high aGRIm score group. CONCLUSIONS: Our present study identified a novel scoring system (aGRIm score) that can predict the survival outcome of patients with malignant ascites secondary to HCC.


Subject(s)
Ascites , Carcinoma, Hepatocellular , Liver Neoplasms , Microbiota , Humans , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Ascites/immunology , Ascites/microbiology , Female , Male , Middle Aged , Microbiota/immunology , Aged , Prognosis , Tumor Microenvironment/immunology , Adult , RNA, Ribosomal, 16S/genetics
8.
Fish Shellfish Immunol ; 149: 109527, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38561068

ABSTRACT

Skin mucus analysis has recently been used as a non-invasive method to evaluate for fish welfare. The present research study was conducted to examine the skin mucosal immunity and skin microbiota profiles of sturgeons infected with Citrobacter freundii. Our histology results showed that the thickness of the epidermal layer of skin remained thinner, and the number of mucous cells was significantly decreased in sturgeons after infection (p < 0.05). Total protein, alanine aminotransferase, aspartate aminotransferase, superoxide dismutase, and creatine kinase levels in the mucus showed biphasic pattern (decrease and then increase). Lactate dehydrogenase, lysozyme, and acid phosphatase activities in the mucus showed an increasing trend after infection. Furthermore, 16S rRNA sequencing also revealed that C. freundii infection also affected the diversity and community structure of the skin mucus microbiota. An increase in microbial diversity (p > 0.05) and a decrease in microbial abundance (p < 0.05) after infection were noted. The predominant bacterial phyla in the skin mucus were Proteobacteria, Fusobacteria, Bacteroidetes, Firmicutes, and Actinobacteria. Specifically, the relative abundance of Fusobacteria increased after infection. The predominant bacterial genera in the skin mucus were Cetobacterium, Pelomonas, Bradyrhizobium, Flavobacterium, and Pseudomonas. The relative abundance of Cetobacterium, Pseudomonas, and Flavobacterium increased after infection. Our current research findings will provide new insights into the theoretical basis for future research studies exploring the mechanism of sturgeon infection with C. freundii.


Subject(s)
Citrobacter freundii , Enterobacteriaceae Infections , Fish Diseases , Fishes , Immunity, Mucosal , Microbiota , Skin , Animals , Citrobacter freundii/immunology , Microbiota/immunology , Fish Diseases/immunology , Fish Diseases/microbiology , Skin/immunology , Skin/microbiology , Fishes/immunology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/veterinary , Enterobacteriaceae Infections/microbiology , Mucus/immunology , Mucus/microbiology , RNA, Ribosomal, 16S/genetics
9.
J Invest Dermatol ; 144(5): 969-977, 2024 May.
Article in English | MEDLINE | ID: mdl-38530677

ABSTRACT

Atopic dermatitis (AD) is a multifactorial, heterogeneous disease characterized by epidermal barrier dysfunction, immune system dysregulation, and skin microbiome alterations. Skin microbiome studies in AD have demonstrated that disease flares are associated with microbial shifts, particularly Staphylococcus aureus predominance. AD-associated S. aureus strains differ from those in healthy individuals across various genomic loci, including virulence factors, adhesion proteins, and proinflammatory molecules-which may contribute to complex microbiome barrier-immune system interactions in AD. Different microbially based treatments for AD have been explored, and their future therapeutic successes will depend on a deeper understanding of the potential microbial contributions to the disease.


Subject(s)
Dermatitis, Atopic , Microbiota , Skin , Staphylococcus aureus , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/immunology , Humans , Microbiota/immunology , Skin/microbiology , Skin/immunology , Staphylococcus aureus/immunology
10.
Nature ; 628(8006): 171-179, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38509360

ABSTRACT

The myriad microorganisms that live in close association with humans have diverse effects on physiology, yet the molecular bases for these impacts remain mostly unknown1-3. Classical pathogens often invade host tissues and modulate immune responses through interactions with human extracellular and secreted proteins (the 'exoproteome'). Commensal microorganisms may also facilitate niche colonization and shape host biology by engaging host exoproteins; however, direct exoproteome-microbiota interactions remain largely unexplored. Here we developed and validated a novel technology, BASEHIT, that enables proteome-scale assessment of human exoproteome-microbiome interactions. Using BASEHIT, we interrogated more than 1.7 million potential interactions between 519 human-associated bacterial strains from diverse phylogenies and tissues of origin and 3,324 human exoproteins. The resulting interactome revealed an extensive network of transkingdom connectivity consisting of thousands of previously undescribed host-microorganism interactions involving 383 strains and 651 host proteins. Specific binding patterns within this network implied underlying biological logic; for example, conspecific strains exhibited shared exoprotein-binding patterns, and individual tissue isolates uniquely bound tissue-specific exoproteins. Furthermore, we observed dozens of unique and often strain-specific interactions with potential roles in niche colonization, tissue remodelling and immunomodulation, and found that strains with differing host interaction profiles had divergent interactions with host cells in vitro and effects on the host immune system in vivo. Overall, these studies expose a previously unexplored landscape of molecular-level host-microbiota interactions that may underlie causal effects of indigenous microorganisms on human health and disease.


Subject(s)
Bacteria , Host Microbial Interactions , Microbiota , Phylogeny , Proteome , Symbiosis , Animals , Female , Humans , Mice , Bacteria/classification , Bacteria/immunology , Bacteria/metabolism , Bacteria/pathogenicity , Host Microbial Interactions/immunology , Host Microbial Interactions/physiology , Host Tropism , Microbiota/immunology , Microbiota/physiology , Organ Specificity , Protein Binding , Proteome/immunology , Proteome/metabolism , Reproducibility of Results
11.
Immunol Rev ; 323(1): 288-302, 2024 May.
Article in English | MEDLINE | ID: mdl-38445769

ABSTRACT

Maternal environmental exposures, particularly during gestation and lactation, significantly influence the immunological development and long-term immunity of offspring. Mammalian immune systems develop through crucial inputs from the environment, beginning in utero and continuing after birth. These critical developmental windows are essential for proper immune system development and, once closed, may not be reopened. This review focuses on the mechanisms by which maternal exposures, particularly to pathogens, diet, and microbiota, impact offspring immunity. Mechanisms driving maternal-offspring immune crosstalk include transfer of maternal antibodies, changes in the maternal microbiome and microbiota-derived metabolites, and transfer of immune cells and cytokines via the placenta and breastfeeding. We further discuss the role of transient maternal infections, which are common during pregnancy, in providing tissue-specific immune education to offspring. We propose a "maternal-driven immune education" hypothesis, which suggests that offspring can use maternal encounters that occur during a critical developmental window to develop optimal immune fitness against infection and inflammation.


Subject(s)
Maternal Exposure , Humans , Female , Pregnancy , Animals , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects/immunology , Immunity, Maternally-Acquired , Microbiota/immunology , Immune System/immunology , Immune System/growth & development , Maternal-Fetal Exchange/immunology , Placenta/immunology
12.
J Allergy Clin Immunol ; 153(5): 1215-1228, 2024 May.
Article in English | MEDLINE | ID: mdl-38341182

ABSTRACT

This is an exciting time to be conducting asthma research. The recent development of targeted asthma biologics has validated the power of basic research to discover new molecules amenable to therapeutic intervention. Advances in high-throughput sequencing are providing a wealth of "omics" data about genetic and epigenetic underpinnings of asthma, as well as about new cellular interacting networks and potential endotypes in asthma. Airway epithelial cells have emerged not only as key sensors of the outside environment but also as central drivers of dysregulated mucosal immune responses in asthma. Emerging data suggest that the airway epithelium in asthma remembers prior encounters with environmental exposures, resulting in potentially long-lasting changes in structure and metabolism that render asthmatic individuals susceptible to subsequent exposures. Here we summarize recent insights into asthma biology, focusing on studies using human cells or tissue that were published in the past 2 years. The studies are organized thematically into 6 content areas to draw connections and spur future research (on genetics and epigenetics, prenatal and early-life origins, microbiome, immune and inflammatory pathways, asthma endotypes and biomarkers, and lung structural alterations). We highlight recent studies of airway epithelial dysfunction and response to viral infections and conclude with a framework for considering how bidirectional interactions between alterations in airway structure and mucosal immunity can lead to sustained lung dysfunction in asthma.


Subject(s)
Asthma , Humans , Asthma/immunology , Asthma/genetics , Animals , Microbiota/immunology , Epigenesis, Genetic , Respiratory Mucosa/immunology , Biomarkers , Immunity, Mucosal
13.
J Invest Dermatol ; 144(5): 978-988, 2024 May.
Article in English | MEDLINE | ID: mdl-38363270

ABSTRACT

Chronic pruritus is a cardinal symptom of atopic dermatitis (AD). The mechanisms underlying atopic itch involve intricate crosstalk among skin, immune components, and neural components. In this review, we explore these mechanisms, focusing on key players and interactions that induce and exacerbate itch. We discuss the similarities and differences between pruritus and pain in patients with AD as well as the relationship between pruritus and factors such as sweat and the skin microbiome. Furthermore, we explore novel targets that could provide significant itch relief in these patients as well as exciting future research directions to better understand atopic pruritus in darker skin types.


Subject(s)
Dermatitis, Atopic , Pruritus , Skin , Humans , Dermatitis, Atopic/immunology , Dermatitis, Atopic/complications , Pruritus/immunology , Pruritus/etiology , Skin/pathology , Skin/immunology , Microbiota/immunology , Sweat , Chronic Disease , Animals
14.
Science ; 383(6678): 62-70, 2024 01 05.
Article in English | MEDLINE | ID: mdl-38175892

ABSTRACT

Immune checkpoint inhibitors can stimulate antitumor immunity but can also induce toxicities termed immune-related adverse events (irAEs). Colitis is a common and severe irAE that can lead to treatment discontinuation. Mechanistic understanding of gut irAEs has been hampered because robust colitis is not observed in laboratory mice treated with checkpoint inhibitors. We report here that this limitation can be overcome by using mice harboring the microbiota of wild-caught mice, which develop overt colitis following treatment with anti-CTLA-4 antibodies. Intestinal inflammation is driven by unrestrained activation of IFNγ-producing CD4+ T cells and depletion of peripherally induced regulatory T cells through Fcγ receptor signaling. Accordingly, anti-CTLA-4 nanobodies that lack an Fc domain can promote antitumor responses without triggering colitis. This work suggests a strategy for mitigating gut irAEs while preserving antitumor stimulating effects of CTLA-4 blockade.


Subject(s)
CD4-Positive T-Lymphocytes , Colitis , Immune Checkpoint Inhibitors , Lymphocyte Activation , Microbiota , Receptors, IgG , Animals , Mice , CD4-Positive T-Lymphocytes/immunology , Colitis/etiology , Colitis/microbiology , CTLA-4 Antigen/antagonists & inhibitors , Microbiota/immunology , Receptors, IgG/immunology , Immune Checkpoint Inhibitors/adverse effects , Mice, Inbred C57BL
15.
Prensa méd. argent ; 109(5): 182-192, 20230000.
Article in English | LILACS, BINACIS | ID: biblio-1523555

ABSTRACT

Las biopsias en el campo de la ortopedia se utilizan para guiar las opciones de diagnóstico y tratamiento para el proceso de la enfermedad que puede estar ocurriendo. La preparación de la piel de estas biopsias sigue la preparación estándar para un procedimiento quirúrgico, con el objetivo de disminuir la cantidad de microbiota que podría conducir a la contaminación del tejido de la biopsia e incluso a una posible infección. El tejido obtenido de la biopsia a menudo se somete a un studio histopatológico y cultivo. La tasa de contaminación bacteriana informada es aproximadamente inferior al 4%. Esta revisión cuestiona si las muestras de las biopsias se contaminan con la microbiota que permanece en la piel y cómo puede afectar el manejo. Además, qué técnicas o pasos pueden disminuir la tasa de contaminación al realizar una biopsia. Nuestra revisión bibliográfica identificó pocos estudios sobre la contaminación bacteriana de las biopsias. Identificamos diferentes factores implicados en el conocimiento de la microbiota de la piel: técnicas y soluciones de preparación de la piel, variación de la microbiota típica que coloniza la piel según la región anatómica, retención preoperatoria versus administración profiláctica de antibióticos y uso de diferentes hojas de bisturí para la piel superficial y para tejidos profundos, entre otros. Aunque no pudimos identificar ningún dato que proporcionara respuestas a nuestra pregunta original y cuantificar cada factor individualmente, la mayoría de los estudios en diferentes campos ortopédicos proporcionaron hallazgos significativos hasta cierto punto. Describimos algunas recomendaciones prácticas basadas en el consenso y la efectividad teórica para disminuir la tasa de contaminación. Se necesitan más investigaciones en el campo de la ortopedia que impliquen la contaminación por microbiota de la piel de una biopsia


Biopsies in the field of orthopaedics are used to guide diagnostics and treatment options for the disease process that may be occurring such as a tumor or infection. Skin preparation of these biopsies follows the standard skin preparation for a surgical procedure, with the aim to decrease the amount of microbiota that could lead to contamination of the tissue biopsy and even possible infection. The tissue obtained from the biopsy often undergoes pathology and culture. The reported bacterial contamination rate is roughly below 4%. This review questions how samples from the biopsies are getting contaminated by microbiota that remains on the skin and how it affects infection management. In addition, which techniques or steps can decrease the rate of contamination when performing a biopsy. Our review identified little to no data on investigating bacterial contamination of biopsies. In doing this, the review identified different factors implicated in skin microbiota awareness: skin preparation techniques and solutions, variation of typical microbiota that colonize the skin based on the anatomical region, preoperative withholding versus administrating antibiotics prophylactically and using different scalpel blades for superficial and deep incisions, among others. Although we failed to identify any data that provided answers to our original question and quantify each factor individually, most studies in different orthopaedic fields provided significant findings to some extent. We outline some practical recommendations based on consensus and theoretical effectiveness in decreasing the contamination rate. Further research entailing skin microbiota contamination of a biopsy is needed in the field of orthopaedics.


Subject(s)
Humans , Male , Female , Orthopedics , Bacterial Infections/prevention & control , Antisepsis/methods , Microbiota/immunology , Biopsy
17.
J Am Chem Soc ; 145(13): 7071-7074, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36952265

ABSTRACT

Some members of the human gut microbiota profoundly influence their host's physiology, health, and therapeutic responses, but the responsible molecules and mechanisms are largely unknown. As part of a project to identify immunomodulators produced by gut microbes, we analyzed the metabolome of Collinsella aerofaciens, an actinomycete that figures prominently in numerous association studies. The associations are typically positive correlations of C. aerofaciens with pro-inflammatory responses and undesirable outcomes, but an association with favorable responses to PD-1/PD-L1 cancer immunotherapy is a notable exception. A phenotypic assay-guided screen using dendritic cells (mBMDCs) and cytokine readouts identified the active compound, which was structurally characterized as a lysoglycoglycerolipid with an acetal-bearing ß-galactofuranose head group (CaLGL-1, 1). The structural assignment was confirmed through total synthesis. Assays with tlr2-/-, tlr4-/-, and wt mBMDCs revealed TLR2-dependent signaling. CaLGL-1 is produced by a conversion of a bacterially biosynthesized plasmalogen (CaPlsM, 3) to CaLGL-1 (1) in a low-pH environment.


Subject(s)
Actinobacteria , Toll-Like Receptor 2 , Humans , Hydrogen-Ion Concentration , Lipids , Microbiota/immunology , Dendritic Cells
18.
Semin Immunol ; 66: 101724, 2023 03.
Article in English | MEDLINE | ID: mdl-36758379

ABSTRACT

Innate effector cells are immune cells endowed with host protective features and cytotoxic functions. By sensing the tissue environment, innate cells have an important role in regulating the transition from homeostasis to inflammation and the establishment of pathological states, including the onset and development of cancer. The tumor microenvironment induces molecular and functional modifications in innate cells, dampening their capability to initiate and sustain anti-tumor immune responses. Emerging studies clearly showed a contribution of the microbiota in modulating the functions of innate cells in cancer. Commensal microorganisms can not only directly interact with innate cells in the tumor microenvironment but can also exert immunomodulatory features from non-tumor sites through the release of microbial products. The microbiota can mediate the priming of innate cells at mucosal tissues and determine the strength of immune responses mediated by such cells when they migrate to non-mucosal tissues, having an impact on cancer. Finally, several evidences reported a strong contribution of the microbiota in promoting innate immune responses during anti-cancer therapies leading to enhanced therapeutic efficacy. In this review, we considered the current knowledge on the role of the microbiota in shaping host innate immune responses in cancer.


Subject(s)
Immune System , Immunity, Innate , Immunotherapy , Microbiota , Neoplasms , Humans , Immune System/cytology , Immune System/immunology , Immunity, Innate/immunology , Microbiota/immunology , Neoplasms/immunology , Neoplasms/microbiology , Neoplasms/therapy , Tumor Microenvironment , Homeostasis , Animals
19.
Clin Rev Allergy Immunol ; 64(2): 161-178, 2023 Apr.
Article in English | MEDLINE | ID: mdl-35275333

ABSTRACT

The respiratory tract is home to a diverse microbial community whose influence on local and systemic immune responses is only beginning to be appreciated. Increasing reports have linked changes in this microbiome to a range of pulmonary and extrapulmonary disorders, including asthma, chronic obstructive pulmonary disease and rheumatoid arthritis. Central to many of these findings is the role of IL-17-type immunity as an important driver of inflammation. Despite the crucial role played by IL-17-mediated immune responses in protection against infection, overt Th17 cell responses have been implicated in the pathogenesis of several chronic inflammatory diseases. However, our knowledge of the influence of bacteria that commonly colonise the respiratory tract on IL-17-driven inflammatory responses remains sparse. In this article, we review the current knowledge on the role of specific members of the airway microbiota in the modulation of IL-17-type immunity and discuss how this line of research may support the testing of susceptible individuals and targeting of inflammation at its earliest stages in the hope of preventing the development of chronic disease.


Subject(s)
Inflammation , Interleukin-17 , Lung , Microbiota , Humans , Chronic Disease , Inflammation/immunology , Interleukin-17/immunology , Lung/immunology , Lung/microbiology , Microbiota/immunology , Respiratory System/immunology , Respiratory System/microbiology
20.
Front Cell Infect Microbiol ; 12: 958722, 2022.
Article in English | MEDLINE | ID: mdl-36569197

ABSTRACT

The prevalence of dental caries in the Mexican adult population aged 20 to 85 years is around 93.3%, and 50% in Mexican children and adolescents. Worldwide, it is the most common non-communicable disease. One of the main etiological factors for dental caries is the oral microbiome and changes in its structure and function, with an expansion of pathogenic bacteria like Streptococcus mutans. The exposed dental pulp tissue triggers an innate immune response to counteract this bacterial invasion. The relation between oral dysbiosis and innate immune responses remains unclear. We aimed to understand the relationship between innate immune response and the oral microbiota by quantifying the expression of Toll-like receptors (TLRs) and proinflammatory markers (cytokines and a chemokine) in dental pulp tissue, either exposed or not to carious dentin, and to correlate this information with the oral microbiome found in healthy teeth and those with moderate caries. RNA was purified from pulp tissue, subjected to RT-qPCR and analysed with the ΔΔCt method. Supragingival dental plaque of non-carious teeth and dentin of carious teeth were subjected to 16S targeted sequencing. Principal coordinate analysis, permutational multivariate ANOVA, and linear discriminant analysis were used to assess differences between non-carious and carious teeth. Correlations were assessed with Spearman´s test and corrected for multiple comparisons using the FDR method. The relative abundance (RA) of Lactobacillus, Actinomyces, Prevotella, and Mitsuokella was increased in carious teeth; while the RA of Haemophilus and Porphyromonas decreased. Olsenella and Parascardovia were only detected in carious teeth. Significant overexpression of interleukin 1 beta (IL1 ß), IL6, and CXCL8 was detected in pulp tissue exposed to carious dentin. IL1ß correlated positively with TLR2 and Actinomyces; yet negatively with Porphyromonas. These findings suggest that immune response of pulp tissue chronically exposed to cariogenic microbiome is triggered by proinflammatory cytokines IL1ß and IL6 and the chemokine CXCL8.


Subject(s)
Dental Caries , Dental Pulp , Microbiota , Adolescent , Adult , Child , Humans , Actinobacteria , Actinomyces , Cytokines/immunology , Dental Caries/immunology , Dental Caries/microbiology , Dental Pulp/immunology , Dental Pulp/microbiology , Dentin/metabolism , Dentin/microbiology , Interleukin-6/metabolism , Microbiota/genetics , Microbiota/immunology , Streptococcus mutans/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...