Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Int J Mol Sci ; 24(3)2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36768437

ABSTRACT

In 2013, recognizing that Colorectal Cancer (CRC) is the second leading cause of death by cancer worldwide and that it was a neglected disease increasing rapidly in Mexico, the community of researchers at the Biomedicine Research Unit of the Facultad de Estudios Superiores Iztacala from the Universidad Nacional Autónoma de México (UNAM) established an intramural consortium that involves a multidisciplinary group of researchers, technicians, and postgraduate students to contribute to the understanding of this pathology in Mexico. This article is about the work developed by the Mexican Colorectal Cancer Research Consortium (MEX-CCRC): how the Consortium was created, its members, and its short- and long-term goals. Moreover, it is a narrative of the accomplishments of this project. Finally, we reflect on possible strategies against CRC in Mexico and contrast all the data presented with another international strategy to prevent and treat CRC. We believe that the Consortium's characteristics must be maintained to initiate a national strategy, and the reported data could be useful to establish future collaborations with other countries in Latin America and the world.


Subject(s)
Colorectal Neoplasms , Students , Humans , Mexico , Interdisciplinary Studies , Therapies, Investigational , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/therapy
2.
J Immunol ; 203(2): 532-543, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31142601

ABSTRACT

Gut lymphocytes and the microbiota establish a reciprocal relationship that impacts the host immune response. Class I-restricted T cell-associated molecule (CRTAM) is a cell adhesion molecule expressed by intraepithelial T cells and is required for their retention in the gut. In this study, we show that CRTAM expression affects gut microbiota composition under homeostatic conditions. Moreover, Crtam-/- mice infected with the intestinal pathogen Salmonella exhibit reduced Th17 responses, lower levels of inflammation, and reduced Salmonella burden, which is accompanied by expansion of other microbial taxa. Thus, CRTAM enhances susceptibility to Salmonella, likely by promoting the inflammatory response that promotes the pathogen's growth. We also found that the gut microbiota from wild-type mice, but not from Crtam-/- mice, induces CRTAM expression and Th17 responses in ex-germ-free mice during Salmonella infection. Our study demonstrates a reciprocal relationship between CRTAM expression and the gut microbiota, which ultimately impacts the host response to enteric pathogens.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunoglobulins/immunology , T-Lymphocytes/immunology , Animals , Female , Inflammation/immunology , Intestines/immunology , Male , Mice , Salmonella/immunology , Salmonella Infections/immunology , Th17 Cells/immunology
3.
Proc Natl Acad Sci U S A ; 113(47): 13462-13467, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821741

ABSTRACT

Infections with Gram-negative pathogens pose a serious threat to public health. This scenario is exacerbated by increases in antibiotic resistance and the limited availability of vaccines and therapeutic tools to combat these infections. Here, we report an immunization approach that targets siderophores, which are small molecules exported by enteric Gram-negative pathogens to acquire iron, an essential nutrient, in the host. Because siderophores are nonimmunogenic, we designed and synthesized conjugates of a native siderophore and the immunogenic carrier protein cholera toxin subunit B (CTB). Mice immunized with the CTB-siderophore conjugate developed anti-siderophore antibodies in the gut mucosa, and when mice were infected with the enteric pathogen Salmonella, they exhibited reduced intestinal colonization and reduced systemic dissemination of the pathogen. Moreover, analysis of the gut microbiota revealed that reduction of Salmonella colonization in the inflamed gut was accompanied by expansion of Lactobacillus spp., which are beneficial commensal organisms that thrive in similar locales as Enterobacteriaceae. Collectively, our results demonstrate that anti-siderophore antibodies inhibit Salmonella colonization. Because siderophore-mediated iron acquisition is a virulence trait shared by many bacterial and fungal pathogens, blocking microbial iron acquisition by siderophore-based immunization or other siderophore-targeted approaches may represent a novel strategy to prevent and ameliorate a broad range of infections.


Subject(s)
Enterobacteriaceae/growth & development , Enterobacteriaceae/immunology , Immunization , Siderophores/immunology , Animals , Antibody Formation , Colony Count, Microbial , Female , Gastrointestinal Microbiome , Immunity, Mucosal/immunology , Inflammation/pathology , Mice, Inbred C57BL , Siderophores/chemistry
4.
Trends Immunol ; 36(2): 112-20, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25582038

ABSTRACT

Pathogens have evolved clever strategies to evade and in some cases exploit the attacks of an activated immune system. Salmonella enterica is one such pathogen, exploiting multiple aspects of host defense to promote its replication in the host. Here we review recent findings on the mechanisms by which Salmonella establishes systemic and chronic infection, including strategies involving manipulation of innate immune signaling and inflammatory forms of cell death, as well as immune evasion by establishing residency in M2 macrophages. We also examine recent evidence showing that the oxidative environment and the high levels of antimicrobial proteins produced in response to localized Salmonella gastrointestinal infection enable the pathogen to successfully outcompete the resident gut microbiota.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity , Salmonella Infections/immunology , Salmonella/immunology , Animals , Humans , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/microbiology , Oxidative Stress , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Salmonella Infections/metabolism , Toll-Like Receptors/metabolism
5.
Infect Immun ; 84(9): 2639-52, 2016 09.
Article in English | MEDLINE | ID: mdl-27382022

ABSTRACT

Sodium phenylbutyrate (PBA) is a derivative of the short-chain fatty acid butyrate and is approved for treatment of urea cycle disorders and progressive familial intrahepatic cholestasis type 2. Previously known functions include histone deacetylase inhibitor, endoplasmic reticulum stress inhibitor, ammonia sink, and chemical chaperone. Here, we show that PBA has a previously undiscovered protective role in host mucosal defense during infection. Administration of PBA to Taconic mice resulted in the increase of intestinal Lactobacillales and segmented filamentous bacteria (SFB), as well as an increase of interleukin 17 (IL-17) production by intestinal cells. This effect was not observed in Jackson Laboratory mice, which are not colonized with SFB. Because previous studies showed that IL-17 plays a protective role during infection with mucosal pathogens, we hypothesized that Taconic mice treated with PBA would be more resistant to infection with Salmonella enterica serovar Typhimurium (S Typhimurium). By using the streptomycin-treated mouse model, we found that Taconic mice treated with PBA exhibited significantly lower S Typhimurium intestinal colonization and dissemination to the reticuloendothelial system, as well as lower levels of inflammation. The lower levels of S Typhimurium gut colonization and intestinal inflammation were not observed in Jackson Laboratory mice. Although PBA had no direct effect on bacterial replication, its administration reduced S Typhimurium epithelial cell invasion and lowered the induction of the proinflammatory cytokine IL-23 in macrophage-like cells. These effects likely contributed to the better outcome of infection in PBA-treated mice. Overall, our results suggest that PBA induces changes in the microbiota and in the mucosal immune response that can be beneficial to the host during infection with S Typhimurium and possibly other enteric pathogens.


Subject(s)
Phenylbutyrates/administration & dosage , Salmonella Infections, Animal/drug therapy , Salmonella typhimurium/drug effects , Animals , Disease Models, Animal , Immunity, Mucosal/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Interleukin-17/metabolism , Interleukin-23/metabolism , Intestines/immunology , Intestines/microbiology , Lactobacillales/drug effects , Lactobacillales/immunology , Lactobacillales/metabolism , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/immunology , Streptomycin/pharmacology
6.
J Immunol ; 190(3): 1201-9, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23284055

ABSTRACT

Salmonella infects and survives within B cells, but the mechanism used by the bacterium to promote its survival in these cells is unknown. In macrophages, flagellin secreted by Salmonella activates the Nod-like receptor (NLR) family CARD domain containing protein 4 (NLRC4) inflammasome, leading to the production of IL-1ß and pyroptosis of infected cells. In this study, we demonstrated that the NLRC4 inflammasome is functional in B cells; however, in Salmonella-infected B cells, IL-1ß secretion is prevented through the downregulation of NLRC4 expression. A functional Salmonella pathogenicity island 1 type III secretion system appears to be required for this process. Furthermore, infection induces Yap phosphorylation and promotes the interaction of Yap with Hck, thus preventing the transcriptional activation of NLRC4. The ability of Salmonella to inhibit IL-1ß production also prevents B cell death; thus, B cells represent an ideal niche in which Salmonella resides, thereby promoting its persistence and dissemination.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis/immunology , B-Lymphocytes/microbiology , Calcium-Binding Proteins/biosynthesis , Down-Regulation , Gene Expression Regulation , Immune Evasion/genetics , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Salmonella Infections, Animal/immunology , Salmonella typhimurium/physiology , Transcription, Genetic , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Calcium-Binding Proteins/genetics , Cell Cycle Proteins , Cytokines/metabolism , Cytotoxicity, Immunologic , Female , Flagellin/pharmacology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-hck/metabolism , Salmonella Infections, Animal/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Virulence , YAP-Signaling Proteins
7.
Microb Pathog ; 52(6): 367-74, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22475626

ABSTRACT

We have previously reported that Salmonella infects B cells and survives within endosomal-lysosomal compartments. However, the mechanisms used by Salmonella to enter B cells remain unknown. In this study, we have shown that Salmonella induces its own entry by the induction of localized ruffling, macropinocytosis, and spacious phagosome formation. These events were associated with the rearrangement of actin and microtubule networks. The Salmonella pathogenesis island 1 (SPI-1) was necessary to invade B cells. In contrast to macrophages, B cells were highly resistant to cell death induced by Salmonella. These data demonstrate the ability of Salmonella to infect these non-professional phagocytic cells, where the bacterium can find an ideal intracellular niche to support persistence and the possible dissemination of infection.


Subject(s)
B-Lymphocytes/microbiology , B-Lymphocytes/physiology , Host-Pathogen Interactions , Phagosomes/microbiology , Pinocytosis , Salmonella/pathogenicity , Actins/metabolism , Animals , Bacterial Proteins , Cells, Cultured , Female , Macrophages/microbiology , Macrophages/physiology , Mice , Mice, Inbred BALB C , Microtubules/metabolism , Salmonella/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
8.
Nat Commun ; 13(1): 3665, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35760817

ABSTRACT

Cell interactions determine phenotypes, and intercellular communication is shaped by cellular contexts such as disease state, organismal life stage, and tissue microenvironment. Single-cell technologies measure the molecules mediating cell-cell communication, and emerging computational tools can exploit these data to decipher intercellular communication. However, current methods either disregard cellular context or rely on simple pairwise comparisons between samples, thus limiting the ability to decipher complex cell-cell communication across multiple time points, levels of disease severity, or spatial contexts. Here we present Tensor-cell2cell, an unsupervised method using tensor decomposition, which deciphers context-driven intercellular communication by simultaneously accounting for multiple stages, states, or locations of the cells. To do so, Tensor-cell2cell uncovers context-driven patterns of communication associated with different phenotypic states and determined by unique combinations of cell types and ligand-receptor pairs. As such, Tensor-cell2cell robustly improves upon and extends the analytical capabilities of existing tools. We show Tensor-cell2cell can identify multiple modules associated with distinct communication processes (e.g., participating cell-cell and ligand-receptor pairs) linked to severities of Coronavirus Disease 2019 and to Autism Spectrum Disorder. Thus, we introduce an effective and easy-to-use strategy for understanding complex communication patterns across diverse conditions.


Subject(s)
Autism Spectrum Disorder , COVID-19 , Cell Communication , Humans , Ligands , Phenotype
9.
J Interferon Cytokine Res ; 39(4): 214-223, 2019 04.
Article in English | MEDLINE | ID: mdl-30855201

ABSTRACT

CCL28 is a mucosal chemokine that has been involved in various responses, including IgA production. We have analyzed its production in human tissues using a comprehensive microarray database. Its highest expression is in the salivary gland, indicating that it is an important component of saliva. It is also expressed in the trachea, bronchus, and in the mammary gland upon onset of lactation. We have also characterized a Ccl28-/- mouse that exhibits very low IgA levels in milk, and the IgA levels in feces are also reduced. These observations confirm a role for the CCL28/CCR10 chemokine axis in the recruitment of IgA plasmablasts to the lactating mammary gland. CCL28 is also expressed in the vomeronasal organ. We also detected olfactory defects (anosmia) in a Ccl28-/- mouse suggesting that CCL28 is involved in the function/development of olfaction. Importantly, Ccl28-/- mice are highly susceptible to Salmonella enterica serovar Typhimurium in an acute model of infection, indicating that CCL28 plays a major role in innate immunity against Salmonella in the gut. Finally, microbiome studies revealed modest differences in the gut microbiota between Ccl28-/- mice and their cohoused wild-type littermates. The latter observation suggests that under homeostatic conditions, CCL28 plays a limited role in shaping the gut microbiome.


Subject(s)
Chemokines, CC/immunology , Chemokines, CC/physiology , Immunity, Innate/immunology , Immunity, Mucosal/immunology , Immunoglobulin A/immunology , Salmonella Infections, Animal/immunology , Smell/physiology , Adaptive Immunity/immunology , Animals , Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Salmonella Infections, Animal/microbiology , Salmonella enterica/immunology
10.
Virulence ; 9(1): 1390-1402, 2018.
Article in English | MEDLINE | ID: mdl-30103648

ABSTRACT

B cells are a target of Salmonella infection, allowing bacteria survival without inducing pyroptosis. This event is due to downregulation of Nlrc4 expression and lack of inflammasome complex activation, which impairs the secretion of IL-1ß. YAP phosphorylation is required for downregulation of Nlrc4 in B cells during Salmonella infection; however, the microorganism's mechanisms underlying the inhibition of the NLRC4 inflammasome in B cells are not fully understood. Our findings demonstrate that the Salmonella effector SopB triggers a signaling cascade involving PI3K, PDK1 and mTORC2 that activates Akt with consequent phosphorylation of YAP. When we deleted sopB in Salmonella, infected B cells that lack Rictor, or inhibited the signaling cascade using a pharmacological approach, we were able to restore the function of the NLRC4 inflammasome in B cells and the ability to control the infection. Furthermore, B cells from infected mice exhibited activation of Akt and YAP phosphorylation, suggesting that Salmonella also triggers this pathway in vivo. In summary, our data demonstrate that the Salmonella effector inositide phosphate phosphatase SopB triggers the PI3K-Akt-YAP pathway to inhibit the NLRC4 inflammasome in B cells. This study provides further evidence that Salmonella triggers cellular mechanisms in B lymphocytes to manipulate the host environment by turning it into a survival niche to establish a successful infection.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , B-Lymphocytes/microbiology , Bacterial Proteins/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Bacterial Proteins/genetics , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Cell Cycle Proteins , Down-Regulation , Inflammasomes , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbial Viability , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , YAP-Signaling Proteins
11.
Cell Syst ; 6(5): 579-592.e4, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29778837

ABSTRACT

Group A Streptococcus (GAS) remains one of the top 10 deadliest human pathogens worldwide despite its sensitivity to penicillin. Although the most common GAS infection is pharyngitis (strep throat), it also causes life-threatening systemic infections. A series of complex networks between host and pathogen drive invasive infections, which have not been comprehensively mapped. Attempting to map these interactions, we examined organ-level protein dynamics using a mouse model of systemic GAS infection. We quantified over 11,000 proteins, defining organ-specific markers for all analyzed tissues. From this analysis, an atlas of dynamically regulated proteins and pathways was constructed. Through statistical methods, we narrowed organ-specific markers of infection to 34 from the defined atlas. We show these markers are trackable in blood of infected mice, and a subset has been observed in plasma samples from GAS-infected clinical patients. This proteomics-based strategy provides insight into host defense responses, establishes potentially useful targets for therapeutic intervention, and presents biomarkers for determining affected organs during bacterial infection.


Subject(s)
Host-Pathogen Interactions/immunology , Proteomics/methods , Streptococcal Infections/immunology , Animals , Bacterial Proteins/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , Pharyngitis/microbiology , Protein Interaction Maps/immunology , Proteome/metabolism , Sepsis/microbiology , Streptococcal Infections/microbiology , Streptococcus/genetics , Streptococcus/immunology , Streptococcus/pathogenicity , Streptococcus pyogenes/metabolism , Tandem Mass Spectrometry
12.
Nat Rev Immunol ; 16(3): 135-48, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26898110

ABSTRACT

The intestinal mucosa is a particularly dynamic environment in which the host constantly interacts with trillions of commensal microorganisms, known as the microbiota, and periodically interacts with pathogens of diverse nature. In this Review, we discuss how mucosal immunity is controlled in response to enteric bacterial pathogens, with a focus on the species that cause morbidity and mortality in humans. We explain how the microbiota can shape the immune response to pathogenic bacteria, and we detail innate and adaptive immune mechanisms that drive protective immunity against these pathogens. The vast diversity of the microbiota, pathogens and immune responses encountered in the intestines precludes discussion of all of the relevant players in this Review. Instead, we aim to provide a representative overview of how the intestinal immune system responds to pathogenic bacteria.


Subject(s)
Bacteria/immunology , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Humans , Immunity, Innate/immunology
13.
Immunol Lett ; 167(2): 131-40, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26292028

ABSTRACT

Salmonella persists for a long time in B cells; however, the mechanism(s) through which infected B cells avoid effector CD8 T cell responses has not been characterized. In this study, we show that Salmonella infects and survives within all B1 and B2 cell subpopulations. B cells are infected with a Salmonella typhimurium strain expressing an ovalbumin (OVA) peptide (SIINFEKL) to evaluate whether B cells process and present Salmonella antigens in the context of MHC-I molecules. Our data showed that OVA peptides are presented by MHC class I K(b)-restricted molecules and the presented antigen is generated through proteasomal degradation and vacuolar processing. In addition, Salmonella-infected B cells express co-stimulatory molecules such as CD40, CD80, and CD86 as well as inhibitory molecules such as PD-L1. Thus, the cross-presentation of Salmonella antigens and the expression of activation molecules suggest that infected B cells are able to prime and activate specific CD8(+) T cells. However, the Salmonella infection-stimulated expression of PD-L1 suggests that the PD-1/PD-L1 pathway may be involved in turning off the cytotoxic effector response during Salmonella persistent infection, thereby allowing B cells to become a reservoir for the bacteria.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B7-H1 Antigen/genetics , Gene Expression Regulation , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella/immunology , Animals , Antigen Presentation/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/microbiology , B-Lymphocytes/microbiology , B7-H1 Antigen/metabolism , Biological Transport , Cross-Priming/immunology , Disease Models, Animal , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Lymphocyte Activation/immunology , Mice , Salmonella typhimurium/immunology , Vacuoles/immunology , Vacuoles/metabolism
14.
Front Immunol ; 5: 586, 2014.
Article in English | MEDLINE | ID: mdl-25484884

ABSTRACT

Although B cells and antibodies are the central effectors of humoral immunity, B cells can also produce and secrete cytokines and present antigen to helper T cells. The uptake of antigen is mainly mediated by endocytosis; thus, antigens are often presented by MHC-II molecules. However, it is unclear if B cells can present these same antigens via MHC-I molecules. Recently, Salmonella bacteria were found to infect B cells, allowing possible antigen cross-processing that could generate bacterial peptides for antigen presentation via MHC-I molecules. Here, we will discuss available knowledge regarding Salmonella antigen presentation by infected B cell MHC-I molecules and subsequent inhibitory effects on CD8(+) T cells for bacterial evasion of cell-mediated immunity.

SELECTION OF CITATIONS
SEARCH DETAIL