Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Cell ; 167(2): 457-470.e13, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27667687

ABSTRACT

Activated macrophages undergo metabolic reprogramming, which drives their pro-inflammatory phenotype, but the mechanistic basis for this remains obscure. Here, we demonstrate that upon lipopolysaccharide (LPS) stimulation, macrophages shift from producing ATP by oxidative phosphorylation to glycolysis while also increasing succinate levels. We show that increased mitochondrial oxidation of succinate via succinate dehydrogenase (SDH) and an elevation of mitochondrial membrane potential combine to drive mitochondrial reactive oxygen species (ROS) production. RNA sequencing reveals that this combination induces a pro-inflammatory gene expression profile, while an inhibitor of succinate oxidation, dimethyl malonate (DMM), promotes an anti-inflammatory outcome. Blocking ROS production with rotenone by uncoupling mitochondria or by expressing the alternative oxidase (AOX) inhibits this inflammatory phenotype, with AOX protecting mice from LPS lethality. The metabolic alterations that occur upon activation of macrophages therefore repurpose mitochondria from ATP synthesis to ROS production in order to promote a pro-inflammatory state.


Subject(s)
Inflammation/immunology , Macrophage Activation , Macrophages/immunology , Mitochondria/enzymology , Succinate Dehydrogenase/metabolism , Succinic Acid/metabolism , Adenosine Triphosphate/metabolism , Animals , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Citric Acid Cycle , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/genetics , Interleukin-10/metabolism , Lipopolysaccharides/immunology , Macrophages/metabolism , Malonates/pharmacology , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Oxidation-Reduction/drug effects , Oxidative Phosphorylation/drug effects , Oxidoreductases/metabolism , Plant Proteins/metabolism , Reactive Oxygen Species/metabolism , Sequence Analysis, RNA , Succinate Dehydrogenase/genetics , Transcriptome
2.
Immunity ; 44(2): 368-79, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26885859

ABSTRACT

Humans that are heterozygous for the common S180L polymorphism in the Toll-like receptor (TLR) adaptor Mal (encoded by TIRAP) are protected from a number of infectious diseases, including tuberculosis (TB), whereas those homozygous for the allele are at increased risk. The reason for this difference in susceptibility is not clear. We report that Mal has a TLR-independent role in interferon-gamma (IFN-γ) receptor signaling. Mal-dependent IFN-γ receptor (IFNGR) signaling led to mitogen-activated protein kinase (MAPK) p38 phosphorylation and autophagy. IFN-γ signaling via Mal was required for phagosome maturation and killing of intracellular Mycobacterium tuberculosis (Mtb). The S180L polymorphism, and its murine equivalent S200L, reduced the affinity of Mal for the IFNGR, thereby compromising IFNGR signaling in macrophages and impairing responses to TB. Our findings highlight a role for Mal outside the TLR system and imply that genetic variation in TIRAP may be linked to other IFN-γ-related diseases including autoimmunity and cancer.


Subject(s)
Interferon-gamma/metabolism , Macrophages/physiology , Membrane Glycoproteins/metabolism , Mycobacterium tuberculosis/immunology , Receptors, Interleukin-1/metabolism , Tuberculosis, Pulmonary/immunology , Animals , Autophagy/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , HEK293 Cells , Humans , Immunity, Innate/genetics , MAP Kinase Signaling System/genetics , Macrophages/microbiology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Polymorphism, Genetic , Protein Binding/genetics , RNA, Small Interfering/genetics , Receptors, Interferon/metabolism , Receptors, Interleukin-1/genetics , Tuberculosis, Pulmonary/genetics , Interferon gamma Receptor
3.
Proc Natl Acad Sci U S A ; 113(48): E7778-E7787, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27856732

ABSTRACT

The parasite Trypanasoma brucei causes African trypanosomiasis, known as sleeping sickness in humans and nagana in domestic animals. These diseases are a major burden in the 36 sub-Saharan African countries where the tsetse fly vector is endemic. Untreated trypanosomiasis is fatal and the current treatments are stage-dependent and can be problematic during the meningoencephalitic stage, where no new therapies have been developed in recent years and the current drugs have a low therapeutic index. There is a need for more effective treatments and a better understanding of how these parasites evade the host immune response will help in this regard. The bloodstream form of T. brucei excretes significant amounts of aromatic ketoacids, including indolepyruvate, a transamination product of tryptophan. This study demonstrates that this process is essential in bloodstream forms, is mediated by a specialized isoform of cytoplasmic aminotransferase and, importantly, reveals an immunomodulatory role for indolepyruvate. Indolepyruvate prevents the LPS-induced glycolytic shift in macrophages. This effect is the result of an increase in the hydroxylation and degradation of the transcription factor hypoxia-inducible factor-1α (HIF-1α). The reduction in HIF-1α levels by indolepyruvate, following LPS or trypanosome activation, results in a decrease in production of the proinflammatory cytokine IL-1ß. These data demonstrate an important role for indolepyruvate in immune evasion by T. brucei.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunity, Innate , Macrophages/metabolism , Pyruvates/metabolism , Trypanosoma brucei brucei/immunology , Trypanosomiasis, African/immunology , Animals , Cell Line , Glycolysis , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immune Evasion , Indoles/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/parasitology , Lipopolysaccharides/pharmacology , Macrophages/parasitology , Mice, Inbred C57BL , Trypanosomiasis, African/parasitology
4.
J Immunol ; 194(3): 1252-60, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25548224

ABSTRACT

Activation of the noncanonical inflammasome, mediated by caspase-11, serves as an additional pathway for the production of the proinflammatory cytokines IL-1ß and IL-18. Noncanonical inflammasome activity occurs during host defense against Gram-negative bacteria and in models of acute septic shock. We propose that the noncanonical inflammasome is activated in mice during acute intestinal inflammation elicited by dextran sodium sulfate (DSS), a model of experimental colitis. We find that caspase-11(-/-) mice display enhanced susceptibility to DSS, because of impaired IL-18 production. The impaired IL-18 levels observed are shown to result in reduced intestinal epithelial cell proliferation and increased cell death. We also suggest that a novel type II IFN-dependent, type I IFN-TRIF-independent signaling pathway is required for in vivo caspase-11 production in intestinal epithelial cells during DSS colitis. Collectively, these data suggest that IFN-γ-mediated caspase-11 expression has a key role maintaining intestinal epithelial barrier integrity in vivo during experimentally induced acute colitis.


Subject(s)
Caspases/metabolism , Colitis/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Caspases/genetics , Caspases, Initiator , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Gene Expression , Genetic Predisposition to Disease , Immunohistochemistry , Interferon-gamma/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout , Phenotype , Signal Transduction
5.
J Biol Chem ; 289(10): 6429-6437, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24407287

ABSTRACT

The apoptosis-associated speck-like protein containing a caspase-activating recruitment domain (ASC) is an essential component of several inflammasomes, multiprotein complexes that regulate caspase-1 activation and inflammation. We report here an interaction between promyelocytic leukemia protein (PML) and ASC. We observed enhanced formation of ASC dimers in PML-deficient macrophages. These macrophages also display enhanced levels of ASC in the cytosol. Furthermore, IL-1ß production was markedly enhanced in these macrophages in response to both NLRP3 and AIM2 inflammasome activation and following bone marrow-derived macrophage infection with herpes simplex virus-1 (HSV-1) and Salmonella typhimurium. Collectively, our data indicate that PML limits ASC function, retaining ASC in the nucleus.


Subject(s)
Cytoskeletal Proteins/metabolism , Inflammasomes/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , CARD Signaling Adaptor Proteins , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoskeletal Proteins/genetics , Cytosol/metabolism , DNA-Binding Proteins , HEK293 Cells , Humans , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Multimerization , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
6.
Am J Physiol Gastrointest Liver Physiol ; 306(9): G769-78, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24603458

ABSTRACT

Toll-like receptors (TLRs) play a central role in the recognition and response to microbial pathogens and in the maintenance and function of the epithelial barrier integrity in the gut. The protein MyD88 adaptor-like (Mal/TIRAP) serves as a bridge between TLR2/TLR4- and MyD88-mediated signaling to orchestrate downstream inflammatory responses. Whereas MyD88 has an essential function in the maintenance of intestinal homeostasis, a role for Mal in this context is less well described. Colitis was induced in wild-type (WT) and Mal-deficient (Mal(-/-)) mice by administration of dextran sodium sulfate (DSS). Colitis-associated cancer was induced by DSS and azoxymethane (AOM) treatment. Chimeric mice were generated by total body gamma irradiation followed by transplantation of bone marrow cells. In the DSS model of colon epithelial injury, Mal(-/-) mice developed increased inflammation and severity of colitis relative to WT mice. Mal(-/-) mice demonstrated the presence of inflammatory cell infiltrates, increased crypt proliferation, and presence of neoformations. Furthermore, in the AOM/DSS model, Mal(-/-) mice had greater incidence of tumors. Mal(-/-) and WT bone marrow chimeras demonstrated that nonhematopoietic cell expression of Mal had an important protective role in the control of intestinal inflammation and inflammation-associated cancer. Mal is essential for the maintenance of intestinal homeostasis and expression of Mal in nonhematopoietic cells prevents chronic intestinal inflammation that may predispose to colon neoplasia.


Subject(s)
Colitis/etiology , Colon/metabolism , Colorectal Neoplasms/etiology , Membrane Glycoproteins/metabolism , Receptors, Interleukin-1/metabolism , Animals , Azoxymethane , Bone Marrow Transplantation , Caco-2 Cells , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Colitis/prevention & control , Colon/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Dextran Sulfate , Disease Models, Animal , Female , Homeostasis , Humans , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Severity of Illness Index , Time Factors , Transplantation Chimera
7.
J Immunol ; 188(11): 5706-12, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22544937

ABSTRACT

Preterm birth, the major cause of neonatal mortality in developed countries, is associated with intrauterine infections and inflammation, although the exact mechanisms underlying this event are unclear. In this study, we show that circulating fetal DNA, which is elevated in pregnancies complicated by preterm labor or preeclampsia, triggers an inflammatory reaction that results in spontaneous preterm birth. Fetal DNA activates NF-κB, shown by IκBα degradation in human PBMCs resulting in production of proinflammatory IL-6. We show that fetal resorption and preterm birth are rapidly induced in mice after i.p. injection of CpG or fetal DNA (300 µg/dam) on gestational day 10-14. In contrast, TLR9(-/-) mice were protected from these effects. Furthermore, this effect was blocked by oral administration of the TLR9 inhibitor chloroquine. Our data therefore provide a novel mechanism for preterm birth and preeclampsia, highlighting TLR9 as a potential therapeutic target for these common disorders of pregnancy.


Subject(s)
DNA/genetics , Fetal Death/immunology , Inflammation Mediators/physiology , Pre-Eclampsia/epidemiology , Premature Birth/epidemiology , Toll-Like Receptor 9/physiology , Adult , Animals , Cell Line, Tumor , Cells, Cultured , DNA/blood , Female , Fetal Death/genetics , Humans , Inflammation Mediators/adverse effects , Inflammation Mediators/blood , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Pregnancy , Toll-Like Receptor 9/biosynthesis , Toll-Like Receptor 9/deficiency
8.
Nucleic Acids Res ; 40(16): 8048-58, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684508

ABSTRACT

Fine-tuning of inflammatory responses by microRNAs (miRNAs) is complex, as they can both enhance and repress expression of pro-inflammatory mediators. In this study, we investigate inflammatory responses following global miRNA depletion, to better define the overall contribution of miRNAs to inflammation. We demonstrate that miRNAs positively regulate Toll-like receptor signaling using inducible Dicer1 deletion and global miRNA depletion. We establish an important contribution of miR-19b in this effect, which potentiates nuclear factor-κB (NF-κB) activity in human and mouse cells. Positive regulation of NF-κB signaling by miR-19b involves the coordinated suppression of a regulon of negative regulators of NF-κB signaling (including A20/Tnfaip3, Rnf11, Fbxl11/Kdm2a and Zbtb16). Transfection of miR-19b mimics exacerbated the inflammatory activation of rheumatoid arthritis primary fibroblast-like synoviocytes, demonstrating its physiological importance in the pathology of this disease. This study constitutes, to our knowledge, the first description of a miR-19 regulon that controls NF-κB signaling, and suggests that targeting this miRNA and linked family members could regulate the activity of NF-κB signaling in inflammation.


Subject(s)
MicroRNAs/metabolism , NF-kappa B/metabolism , Regulon , Signal Transduction , Animals , Bone Marrow Cells/metabolism , Cells, Cultured , Fibroblasts/metabolism , Humans , Inflammation/genetics , Inflammation Mediators/metabolism , Macrophages/metabolism , Mice , Synovial Membrane/cytology , Synovial Membrane/metabolism , Toll-Like Receptors/metabolism
9.
Front Nutr ; 11: 1346706, 2024.
Article in English | MEDLINE | ID: mdl-38425482

ABSTRACT

Introduction: Macrofungi, such as edible mushrooms, have been used as a valuable medical resource for millennia as a result of their antibacterial and immuno-modulatory components. Mushrooms contain dietary fibers known as ß-glucans, a class of polysaccharides previously linked to the induction of Trained Immunity. However, little is known about the ability of mushroom-derived ß-glucans to induce Trained Immunity. Methods & results: Using various powdered forms of the white button mushroom (Agaricus bisporus), we found that mouse macrophages pre-treated with whole mushroom powder (WMP) displayed enhanced responses to restimulation with TLR ligands, being particularly sensitive to Toll-like receptor (TLR)-2 stimulation using synthetic lipopeptides. This trained response was modest compared to training observed with yeast-derived ß-glucans and correlated with the amount of available ß-glucans in the WMP. Enriching for ß-glucans content using either a simulated in-vitro digestion or chemical fractionation retained and boosted the trained response with WMP, respectively. Importantly, both WMP and digested-WMP preparations retained ß-glucans as identified by nuclear magnetic resonance analysis and both displayed the capacity to train human monocytes and enhanced responses to restimulation. To determine if dietary incorporation of mushroom products can lead to Trained Immunity in myeloid cells in vivo, mice were given a regimen of WMP by oral gavage prior to sacrifice. Flow cytometric analysis of bone-marrow progenitors indicated alterations in hematopoietic stem and progenitor cells population dynamics, with shift toward myeloid-committed multi-potent progenitor cells. Mature bone marrow-derived macrophages derived from these mice displayed enhanced responses to restimulation, again particularly sensitive to TLR2. Discussion: Taken together, these data demonstrate that ß-glucans from common macrofungi can train innate immune cells and could point to novel ways of delivering bio-available ß-glucans for education of the innate immune system.

10.
iScience ; 27(3): 109030, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38361630

ABSTRACT

Fungal ß-glucans are major drivers of trained immunity which increases long-term protection against secondary infections. Heterogeneity in ß-glucan source, structure, and solubility alters interaction with the phagocytic receptor Dectin-1 and could impact strategies to improve trained immunity in humans. Using a panel of diverse ß-glucans, we describe the ability of a specific yeast-derived whole-glucan particle (WGP) to reprogram metabolism and thereby drive trained immunity in human monocyte-derived macrophages in vitro and mice bone marrow in vivo. Presentation of pure, non-soluble, non-aggregated WGPs led to the formation of the Dectin-1 phagocytic synapse with subsequent lysosomal mTOR activation, metabolic reprogramming, and epigenetic rewiring. Intraperitoneal or oral administration of WGP drove bone marrow myelopoiesis and improved mature macrophage responses, pointing to therapeutic and food-based strategies to drive trained immunity. Thus, the investment of a cell in a trained response relies on specific recognition of ß-glucans presented on intact microbial particles through stimulation of the Dectin-1 phagocytic response.

11.
Int J Clin Health Psychol ; 23(2): 100349, 2023.
Article in English | MEDLINE | ID: mdl-36605409

ABSTRACT

Moving towards a systems psychiatry paradigm embraces the inherent complex interactions across all levels from micro to macro and necessitates an integrated approach to treatment. Cortical 5-HT2A receptors are key primary targets for the effects of serotonergic psychedelics. However, the therapeutic mechanisms underlying psychedelic therapy are complex and traverse molecular, cellular, and network levels, under the influence of biofeedback signals from the periphery and the environment. At the interface between the individual and the environment, the gut microbiome, via the gut-brain axis, plays an important role in the unconscious parallel processing systems regulating host neurophysiology. While psychedelic and microbial signalling systems operate over different timescales, the microbiota-gut-brain (MGB) axis, as a convergence hub between multiple biofeedback systems may play a role in the preparatory phase, the acute administration phase, and the integration phase of psychedelic therapy. In keeping with an interconnected systems-based approach, this review will discuss the gut microbiome and mycobiome and pathways of the MGB axis, and then explore the potential interaction between psychedelic therapy and the MGB axis and how this might influence mechanism of action and treatment response. Finally, we will discuss the possible implications for a precision medicine-based psychedelic therapy paradigm.

12.
Mol Nutr Food Res ; 67(14): e2200845, 2023 07.
Article in English | MEDLINE | ID: mdl-37195234

ABSTRACT

SCOPE: Mushrooms are valued as an edible and medical resource for millennia. As macrofungi, they possess conserved molecular components recognized by innate immune cells like macrophages, yet unlike pathogenic fungi, they do not trigger the immune system in the same way. That these well-tolerated foods both avoid immuno-surveillance and have positive health benefits, highlights the dearth of information on the interactions of mushroom-derived products with the immune system. METHODS AND RESULTS: Using powders produced from the common white button mushroom, Agaricus bisporus, it is observed that pre-treatment of mouse and human macrophages with mushroom powders attenuates innate immune signaling triggered by microbial ligands like LPS and  ß-glucans, including NFκB activation and pro-inflammatory cytokine production. This effect of mushroom powders is observed at lower doses of TLR ligands, suggesting a model of competitive inhibition whereby mushroom compounds bind and occupy innate immune receptors, precluding activation by microbial stimuli. This effect is preserved following simulated digestion of the powders. Moreover, in vivo delivery of mushroom powders attenuates the development of colitis in a DSS-mouse model. CONCLUSION: This data highlights an important anti-inflammatory role for powdered A. bisporus mushrooms, which can be further utilized to develop complementary approaches to modulate chronic inflammation and disease.


Subject(s)
Agaricus , Humans , Ligands , Powders , Immunity, Innate
13.
Front Immunol ; 13: 840245, 2022.
Article in English | MEDLINE | ID: mdl-35464397

ABSTRACT

In recent decades, probiotic bacteria have become increasingly popular as a result of mounting scientific evidence to indicate their beneficial role in modulating human health. Although there is strong evidence associating various Lactobacillus probiotics to various health benefits, further research is needed, in particular to determine the various mechanisms by which probiotics may exert these effects and indeed to gauge inter-individual value one can expect from consuming these products. One must take into consideration the differences in individual and combination strains, and conditions which create difficulty in making direct comparisons. The aim of this paper is to review the current understanding of the means by which Lactobacillus species stand to benefit our gastrointestinal health.


Subject(s)
Lactobacillus , Probiotics , Bacteria , Gastrointestinal Tract/microbiology , Humans , Probiotics/therapeutic use
14.
Phytomedicine ; 105: 154353, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35932606

ABSTRACT

BACKGROUND: A traditionally prepared aqueous extract (= decoction) of Houttuynia cordata Thunb (Yu xing cao) (HC) is widely used in Traditional Chinese Medicine (TCM) to treat inflammatory disease. Previous chemical and biological studies on HC have mainly focused on organic extracts rather than the aqueous decoction, which is the traditional formulation. PURPOSE: The study aimed to investigate whether the chemical composition of HC aqueous decoction (HCD) varies with geographical sourcing, to investigate the mechanism of action of HCD, and to determine if chemical variation impacts on HCDs anti-inflammatory activity. METHOD: Sixteen samples of HC were purchased from Sichuan, Hubei and Anhui provinces in the People's Republic of China (PRC) and were prepared by the traditional decoction method to yield their corresponding HCDs. A Quality Control (QC) sample was prepared by combining individual HCD extracts. HCDs were analysed by Nuclear Magnetic Resonance (NMR) and High-Performance Liquid Chromatography-Mass Spectrometry (HPLC-MS). The anti-inflammatory activities associated with intestinal barrier function of HCD were studied by tumor necrosis factor-α (TNF-α) activated Caco-2 monolayers in vitro and in vivo using Dextran Sulfate Sodium (DSS)-induced murine colitis. Proteins involved in inflammation, mRNA levels, disease severity scores, and histology involved in intestinal inflammation were analysed. RESULTS: HCD samples exhibited different chemical fingerprints and three regional outliers were identified by Principal Component Analysis (PCA). Fifteen phytochemical metabolites were identified and quantified. HCD showed in vitro anti-inflammatory activity, enhancing zonula occludens-1 (ZO-1), occludin, interleukin (IL)-10 and decreasing IL-1ß, IL-6 and epidermal growth factor receptor (EGFR) via an EGFR-dependent mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase 1/2 (ERK 1/2) signaling pathway. This beneficial effect on intestinal inflammation was also seen in the in vivo colitis model at a molecular level in colonic tissues. CONCLUSION: This study shows that the test HCDs were chemically different, resulting in different levels of activity on intestinal barrier function and inflammation. Moreover, a "Daodi" product showed the greatest biological activity in this study, thus validating the importance of the "Daodi" quality material in TCM and supporting the traditional used of HCD for the treatment of inflammation.


Subject(s)
Colitis , Houttuynia , Animals , Anti-Inflammatory Agents , Caco-2 Cells , Dextran Sulfate , Disease Models, Animal , ErbB Receptors , Humans , Inflammation , MAP Kinase Signaling System , Medicine, Chinese Traditional , Mice , Mitogen-Activated Protein Kinase 3 , Mitogens , Plant Extracts , Signal Transduction
15.
Microorganisms ; 10(7)2022 Jul 09.
Article in English | MEDLINE | ID: mdl-35889102

ABSTRACT

The use of probiotics such as Lactobacillus and Bifidobacterium spp. as a therapeutic against inflammatory bowel disease (IBD) is of significant interest. Lactobacillus salivarus strain UCC118TM is a commensal that has been shown to possess probiotic properties in vitro and anti-infective properties in vivo. However, the usefulness of UCC118 TM as a therapeutic against colitis remains unclear. This study investigates the probiotic potential of Lactobacillus salivarius, UCC118™ in a mouse model of colitis. DSS-induced colitis was coupled with pre-treatment or post-treatment with UCC118TM by daily oral gavage. In the pre-treatment model of colitis, UCC118TM reduced the severity of the disease in the early stages. Improvement in disease severity was coupled with an upregulation of tissue IL-10 levels and increased expression of macrophage M2 markers. This anti-inflammatory activity of UCC118TM was further confirmed in vitro, using a model of LPS-treated bone marrow-derived macrophages. Taken together, these results suggest that UCC118TM may promote the resolution of inflammation. This was supported in a mouse model of established DSS-induced colitis whereby UCC118TM treatment accelerated recovery, as evidenced by weight, stool, histological markers and the recovery of microbiome-associated dysbiosis with an increased abundance of beneficial commensal species. These results demonstrate the potential of Lactobacillus salivarius UCC118TM as a probiotic-based therapeutic strategy to promote health through the upregulation of anti-inflammatory IL-10 and protect against dysbiosis during IBD.

16.
Nutrients ; 13(12)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34959809

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic inflammatory disease. The disease has a multifactorial aetiology, involving genetic, microbial as well as environmental factors. The disease pathogenesis operates at the host-microbe interface in the gut. The intestinal epithelium plays a central role in IBD disease pathogenesis. Apart from being a physical barrier, the epithelium acts as a node that integrates environmental, dietary, and microbial cues to calibrate host immune response and maintain homeostasis in the gut. IBD patients display microbial dysbiosis in the gut, combined with an increased barrier permeability that contributes to disease pathogenesis. Metabolites produced by microbes in the gut are dynamic indicators of diet, host, and microbial interplay in the gut. Microbial metabolites are actively absorbed or diffused across the intestinal lining to affect the host response in the intestine as well as at systemic sites via the engagement of cognate receptors. In this review, we summarize insights from metabolomics studies, uncovering the dynamic changes in gut metabolite profiles in IBD and their importance as potential diagnostic and prognostic biomarkers of disease. We focus on gut microbial metabolites as key regulators of the intestinal barrier and their role in the pathogenesis of IBD.


Subject(s)
Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/microbiology , Metabolomics , Biomarkers/metabolism , Humans , Permeability , Prognosis
17.
Cells ; 10(12)2021 11 30.
Article in English | MEDLINE | ID: mdl-34943865

ABSTRACT

Research on inflammatory bowel disease (IBD) has produced mounting evidence for the modulation of microRNAs (miRNAs) during pathogenesis. MiRNAs are small, non-coding RNAs that interfere with the translation of mRNAs. Their high stability in free circulation at various regions of the body allows researchers to utilise miRNAs as biomarkers and as a focus for potential treatments of IBD. Yet, their distinct regulatory roles at the gut epithelial barrier remain elusive due to the fact that there are several external and cellular factors contributing to gut permeability. This review focuses on how miRNAs may compromise two components of the gut epithelium that together form the initial physical barrier: the mucus layer and the intercellular epithelial junctions. Here, we summarise the impact of miRNAs on goblet cell secretion and mucin structure, along with the proper function of various junctional proteins involved in paracellular transport, cell adhesion and communication. Knowledge of how this elaborate network of cells at the gut epithelial barrier becomes compromised as a result of dysregulated miRNA expression, thereby contributing to the development of IBD, will support the generation of miRNA-associated biomarker panels and therapeutic strategies that detect and ameliorate gut permeability.


Subject(s)
Gastrointestinal Tract/pathology , Inflammatory Bowel Diseases/genetics , Intercellular Junctions/metabolism , MicroRNAs/metabolism , Mucus/metabolism , Animals , Humans , MicroRNAs/genetics , Permeability
18.
Front Cell Infect Microbiol ; 11: 622491, 2021.
Article in English | MEDLINE | ID: mdl-34350128

ABSTRACT

The metabolite-rich environment that is the intestinal lumen contains metabolic by-products deriving from microbial fermentation and host cell metabolism, with resident macrophages being constantly exposed to this metabolic flux. Succinate, lactate and itaconate are three metabolites secreted by primed macrophages due to a fragmented tri-carboxylic acid (TCA) cycle. Additionally, succinate and lactate are known by-products of microbial fermentation. How these metabolites impact biological functioning of resident macrophages particularly in response to bacterial infection remains poorly understood. We have investigated the potential influence of these metabolites on macrophage phagocytosis and clearance of Escherichia coli (E. coli) infection. Treatment of murine bone-marrow-derived macrophages (BMDMs) with succinate reduced numbers of intracellular E. coli early during infection, while lactate-treated BMDMs displayed no difference throughout the course of infection. Treatment of BMDMs with itaconate lead to higher levels of intracellular E. coli early in the infection with bacterial burden subsequently reduced at later time-points compared to untreated macrophages, indicative of enhanced engulfment and killing capabilities of macrophages in response to itaconate. Expression of engulfment mediators MARCKS, RhoB, and CDC42 were reduced or unchanged following succinate or lactate treatment and increased in itaconate-treated macrophages following E. coli infection. Nitric oxide (NO) levels varied while pro- and anti-inflammatory cytokines differed in secretory levels in all metabolite-treated macrophages post-infection with E. coli or in response to lipopolysaccharide (LPS) stimulation. Finally, the basal phenotypic profile of metabolite-treated macrophages was altered according to marker gene expression, describing how fluid macrophage phenotype can be in response to the microenvironment. Collectively, our data suggests that microbe- and host-derived metabolites can drive distinct macrophage functional phenotypes in response to infection, whereby succinate and itaconate regulate phagocytosis and bactericidal mechanisms, limiting the intracellular bacterial niche and impeding the pathogenesis of infection.


Subject(s)
Bacterial Infections , Escherichia coli , Animals , Lipopolysaccharides , Macrophages , Mice , Phagocytosis
19.
Cell Microbiol ; 11(5): 703-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19191786

ABSTRACT

Pathogen recognition and induction of immune responses are important for efficient elimination of infection. However, pathogens such as Listeria monocytogenes employ strategies to evade or modulate these defences, thus creating a more favourable environment that ensures their survival and pathogenesis. New insights into these strategies, particularly those targeting innate immunity, have recently emerged. L. monocytogenes is initially detected at the cell surface or in phagosomes by toll-like receptor 2 and in the cytosol by nuclear oligodimerization domain (NOD)-like receptors (NOD1, NOD2) and NALP3 and Ipaf. It carries out N-deacetylation of peptidoglycan to avoid this detection by toll-like receptor 2 and NOD-like receptors. L. monocytogenes modulates transcription of host immunity genes through modification of histones and chromatin remodelling. Furthermore, L. monocytogenes has recently been shown to avoid autophagy and induce apoptosis in immune effector cells. In this review we discuss some of these strategies, which have provided new insights into the interaction between L. monocytogenes and the immune response at a crucial stage of infection.


Subject(s)
Immunity, Innate , Listeriosis/immunology , Models, Immunological , Animals , Apoptosis , Autophagy , Chromatin Assembly and Disassembly , Histones/metabolism , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Lymphocytes/microbiology , Mice , Signal Transduction , Toll-Like Receptors/physiology
20.
Cell Rep ; 30(1): 124-136.e4, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914380

ABSTRACT

Increased glycolytic metabolism recently emerged as an essential process driving host defense against Mycobacterium tuberculosis (Mtb), but little is known about how this process is regulated during infection. Here, we observe repression of host glycolysis in Mtb-infected macrophages, which is dependent on sustained upregulation of anti-inflammatory microRNA-21 (miR-21) by proliferating mycobacteria. The dampening of glycolysis by miR-21 is mediated through targeting of phosphofructokinase muscle (PFK-M) isoform at the committed step of glycolysis, which facilitates bacterial growth by limiting pro-inflammatory mediators, chiefly interleukin-1ß (IL-1ß). Unlike other glycolytic genes, PFK-M expression and activity is repressed during Mtb infection through miR-21-mediated regulation, while other less-active isoenzymes dominate. Notably, interferon-γ (IFN-γ), which drives Mtb host defense, inhibits miR-21 expression, forcing an isoenzyme switch in the PFK complex, augmenting PFK-M expression and macrophage glycolysis. These findings place the targeting of PFK-M by miR-21 as a key node controlling macrophage immunometabolic function.


Subject(s)
Glycolysis , Host-Pathogen Interactions , Interleukin-1beta/metabolism , MicroRNAs/metabolism , Mycobacterium tuberculosis/physiology , Phosphofructokinase-1/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Base Sequence , Cell Proliferation , HEK293 Cells , Humans , Interferon-gamma/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , MicroRNAs/genetics , Phosphofructokinase-1/genetics , RAW 264.7 Cells , Tuberculosis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL