Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nutrients ; 13(3)2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33668212

ABSTRACT

Since alterations of the gut microbiota have been shown to play a major role in obesity, probiotics have attracted attention. Our aim was to identify probiotic candidates for the management of obesity using a combination of in vitro and in vivo approaches. We evaluated in vitro the ability of 23 strains to limit lipid accumulation in adipocytes and to enhance the secretion of satiety-promoting gut peptide in enteroendocrine cells. Following the in vitro screening, selected strains were further investigated in vivo, single, or as mixtures, using a murine model of diet-induced obesity. Strain Bifidobacterium longum PI10 administrated alone and the mixture of B. animalis subsp. lactis LA804 and Lactobacillus gasseri LA806 limited body weight gain and reduced obesity-associated metabolic dysfunction and inflammation. These protective effects were associated with changes in the hypothalamic gene expression of leptin and leptin receptor as well as with changes in the composition of gut microbiota and the profile of bile acids. This study provides crucial clues to identify new potential probiotics as effective therapeutic approaches in the management of obesity, while also providing some insights into their mechanisms of action.


Subject(s)
Adipocytes/microbiology , Enteroendocrine Cells/microbiology , Gastrointestinal Microbiome/physiology , Obesity/microbiology , Probiotics/pharmacology , Animals , Bile Acids and Salts/metabolism , Diet/adverse effects , Disease Models, Animal , Gastrointestinal Hormones/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Mice , Obesity/etiology , Obesity Management/methods , Receptors, Leptin/metabolism , Weight Gain/physiology
2.
Sci Transl Med ; 13(577)2021 01 20.
Article in English | MEDLINE | ID: mdl-33472955

ABSTRACT

Infections are a major complication of obesity, but the mechanisms responsible for impaired defense against microbes are not well understood. Here, we found that adipocyte progenitors were lost from the dermis during diet-induced obesity (DIO) in humans and mice. The loss of adipogenic fibroblasts from mice resulted in less antimicrobial peptide production and greatly increased susceptibility to Staphylococcus aureus infection. The decrease in adipocyte progenitors in DIO mice was explained by expression of transforming growth factor-ß (TGFß) by mature adipocytes that then inhibited adipocyte progenitors and the production of cathelicidin in vitro. Administration of a TGFß receptor inhibitor or a peroxisome proliferator-activated receptor-γ agonist reversed this inhibition in both cultured adipocyte progenitors and in mice and subsequently restored the capacity of obese mice to defend against S. aureus skin infection. Together, these results explain how obesity promotes dysfunction of the antimicrobial function of reactive dermal adipogenesis and identifies potential therapeutic targets to manage skin infection associated with obesity.


Subject(s)
Adipocytes/immunology , Anti-Infective Agents , Obesity/complications , Staphylococcal Infections/immunology , 3T3-L1 Cells , Adipocytes/microbiology , Animals , Anti-Infective Agents/pharmacology , Cell Differentiation , Diet , Diet, High-Fat , Mice , Mice, Inbred C57BL , PPAR gamma/agonists , Staphylococcal Infections/complications , Staphylococcus aureus , Transforming Growth Factor beta/antagonists & inhibitors
3.
Front Endocrinol (Lausanne) ; 11: 585923, 2020.
Article in English | MEDLINE | ID: mdl-33071987

ABSTRACT

Brucellosis is a prevalent global zoonotic infection but has far more impact in developing countries. The adipocytes are the most abundant cell type of adipose tissue and their secreted factors play an important role in several aspects of the innate and adaptive immune response. Here, we demonstrated the ability of Brucella abortus to infect and replicate in both adipocytes and its precursor cells (pre-adipocytes) derived from 3T3-L1 cell line. Additionally, infection of pre-adipocytes also inhibited adipogenesis in a mechanism independent of bacterial viability and dependent on lipidated outer membrane protein (L-Omp19). B. abortus infection was able to modulate the secretion of IL-6 and the matrix metalloproteases (MMPs) -2 and-9 in pre-adipocytes and adipocytes, and also modulated de transcription of adiponectin, leptin, and resistin in differentiated adipocytes. B. abortus-infected macrophages also modulate adipocyte differentiation involving a TNF-α dependent mechanism, thus suggesting a plausible interplay between B. abortus, adipocytes, and macrophages. In conclusion, B. abortus is able to alter adipogenesis process in adipocytes and its precursors directly after their infection, or merely their exposure to the B. abortus lipoproteins, and indirectly through soluble factors released by B. abortus-infected macrophages.


Subject(s)
Adipocytes/cytology , Adipogenesis , Brucellosis/complications , Cell Differentiation , Inflammation/immunology , Macrophages/immunology , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/metabolism , Adipocytes/microbiology , Animals , Brucella abortus/physiology , Cytokines/metabolism , Inflammation/metabolism , Inflammation/microbiology , Inflammation Mediators/metabolism , Lipoproteins/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mice
4.
PLoS Negl Trop Dis ; 14(10): e0008850, 2020 10.
Article in English | MEDLINE | ID: mdl-33075048

ABSTRACT

Leprosy is a chronic infectious disease caused by Mycobacterium leprae (M. leprae). In lepromatous leprosy (LL), skin macrophages, harboring extensive bacterial multiplication, gain a distinctive foamy appearance due to increased intracellular lipid load. To determine the mechanism by which M. leprae modifies the lipid homeostasis in host cells, an in vitro M. leprae infection system, using human macrophage precursor THP-1 cells and M. leprae prepared from the footpads of nude mice, was employed. RNA extracted from skin smear samples of patients was used to investigate host gene expressions before and after multidrug therapy (MDT). We found that a cluster of peroxisome proliferator-activated receptor (PPAR) target genes associated with adipocyte differentiation were strongly induced in M. leprae-infected THP-1 cells, with increased intracellular lipid accumulation. PPAR-δ and PPAR-γ expressions were induced by M. leprae infection in a bacterial load-dependent manner, and their proteins underwent nuclear translocalization after infection, indicating activation of PPAR signaling in host cells. Either PPAR-δ or PPAR-γ antagonist abolished the effect of M. leprae to modify host gene expressions and inhibited intracellular lipid accumulation in host cells. M. leprae-specific gene expressions were detected in the skin smear samples both before and after MDT, whereas PPAR target gene expressions were dramatically diminished after MDT. These results suggest that M. leprae infection activates host PPAR signaling to induce an array of adipocyte differentiation-associated genes, leading to accumulation of intracellular lipids to accommodate M. leprae parasitization. Certain PPAR target genes in skin lesions may serve as biomarkers for monitoring treatment efficacy.


Subject(s)
Foam Cells/microbiology , Leprosy/metabolism , Macrophages/microbiology , Mycobacterium leprae/physiology , PPAR delta/metabolism , PPAR gamma/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipocytes/microbiology , Animals , Cell Differentiation , Foam Cells/metabolism , Humans , Leprostatic Agents/therapeutic use , Leprosy/drug therapy , Leprosy/genetics , Leprosy/microbiology , Lipid Metabolism , Macrophages/metabolism , Mice , Mice, Nude , Mycobacterium leprae/drug effects , PPAR delta/genetics , PPAR gamma/genetics , Skin/metabolism , Skin/microbiology
5.
J Immunol ; 201(5): 1491-1499, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30037847

ABSTRACT

A link between obesity and periodontitis has been suggested because of compromised immune response and chronic inflammation in obese patients. In this study, we evaluated the anti-inflammatory properties of Kavain, an extract from Piper methysticum, on Porphyromonas gingivalis-induced inflammation in adipocytes with special focus on peroxisome proliferation-activated receptor γ coactivator α (PGC-1α) and related pathways. The 3T3-L1 mouse preadipocytes and primary adipocytes harvested from mouse adipose tissue were infected with P. gingivalis, and inflammation (TNF-α; adiponectin/adipokines), oxidative stress, and adipogenic marker (FAS, CEBPα, and PPAR-γ) expression were measured. Furthermore, effect of PGC-1α knockdown on Kavain action was evaluated. Results showed that P. gingivalis worsens adipocyte dysfunction through increase of TNF-α, IL-6, and iNOS and decrease of PGC-1α and adiponectin. Interestingly, although Kavain obliterated P. gingivalis-induced proinflammatory effects in wild-type cells, Kavain did not affect PGC-1α-deficient cells, strongly advocating for Kavain effects being mediated by PGC-1α. In vivo adipocytes challenged with i.p. injection of P. gingivalis alone or P. gingivalis and Kavain displayed the same phenotype as in vitro adipocytes. Altogether, our findings established anti-inflammatory and antioxidant effects of Kavain on adipocytes and emphasized protective action against P. gingivalis-induced adipogenesis. The use of compounds such as Kavain offer a portal to potential therapeutic approaches to counter chronic inflammation in obesity-related diseases.


Subject(s)
Adipocytes/immunology , Bacteroidaceae Infections/drug therapy , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/immunology , Porphyromonas gingivalis/immunology , Pyrones/pharmacology , Signal Transduction/drug effects , 3T3-L1 Cells , Adipocytes/microbiology , Adipocytes/pathology , Animals , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/pathology , Cytokines/genetics , Cytokines/immunology , Gene Knockdown Techniques , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Porphyromonas gingivalis/pathogenicity , Signal Transduction/genetics , Signal Transduction/immunology
6.
Nutrients ; 10(5)2018 May 19.
Article in English | MEDLINE | ID: mdl-29783731

ABSTRACT

The prevalence of obesity and associated metabolic disorders, including diabetes and cardiovascular disease, is rapidly becoming a severe global health problem. Recent reports have suggested that the alteration of the gut ecosystem through the consumption of probiotics and fermented foods, such as yogurt and Kimchi, can significantly impact obesity and Type 2 diabetes (T2D)-related biomarkers. In this study, we screened over 400 strains of lactic acid bacteria (LAB) that were isolated from fermented foods to identify potent anti-obesogenic and diabetic probiotics in vitro. Of the strains tested, Lactobacillus plantarum Ln4 (Ln4), which was obtained from napa cabbage kimchi, significantly reduced lipid accumulation and stimulated glucose uptake in 3T3-L1 adipocytes. Oral administration of Ln4 reduced weight gain and epididymal fat mass in mice fed on a high-fat diet (HFD). Total plasma triglyceride level was significantly lower in mice that were treated Ln4 as compared with mice fed HFD. The protein levels of adipokines such as C-reactive protein (CRP), insulin-like growth factor binding proteins-3 (IGFBP-3), and monocyte chemoattractant protein-1 (MCP-1) decreased in white adipose tissues of Ln4-treated mice. Furthermore, these mice exhibited a significant reduction of insulin resistance index (HOMA-IR) and the improvement of glucose tolerance (OGTT) and insulin response (ITT) following Ln4 administration. This was associated with changes in several hepatic gene expressions (increased mRNA levels of IRS2, Akt2, AMPK, LPL, and reduced CD36) that regulate glucose and lipid metabolism. Taken together, these results indicate that in vitro and in vivo Ln4 treatment attenuates diet-induced obesity and T2D biomarkers, highlighting the potential of Ln4 as a therapeutic probiotic agent for metabolic disorders.


Subject(s)
Blood Glucose/metabolism , Diet, High-Fat , Gastrointestinal Microbiome , Insulin Resistance , Lactobacillus plantarum/physiology , Lipids/blood , Liver/metabolism , Obesity/prevention & control , Probiotics/administration & dosage , RNA, Messenger/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/microbiology , Adipogenesis , Adipokines/blood , Animals , Biomarkers/blood , Disease Models, Animal , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Obesity/blood , Obesity/etiology , Obesity/microbiology , RNA, Messenger/genetics , Time Factors , Weight Gain
7.
Infect Immun ; 86(6)2018 06.
Article in English | MEDLINE | ID: mdl-29632245

ABSTRACT

Mycobacterium tuberculosis, a successful human pathogen, utilizes multiple carbon sources from the host but adapts to a fatty-acid-rich environment in vivo We sought to delineate the physiologic response of M. tuberculosis to a lipid-rich environment by using differentiated adipocytes as a model system. Global transcriptome profiling based on RNA sequencing was performed for bacilli from infected adipocytes and preadipocytes. Genes involved in de novo fatty acid synthesis were downregulated, while those predicted to be involved in triglyceride biosynthesis were upregulated, in bacilli isolated from adipocytes, indicating reliance on host-derived fatty acids. Transcription factor network analysis indicated suppression of IdeR-regulated genes, suggesting decreased iron uptake by M. tuberculosis in the adipocyte model. This suppression of iron uptake coincided with higher ferritin and iron levels in adipocytes than in preadipocytes. In accord with the role of iron in mediating oxidative stress, we observed upregulation of genes involved in mitigating oxidative stress in M. tuberculosis isolated from adipocytes. We provide evidence that oleic acid, a major host-derived fatty acid, helps reduce the bacterial cytoplasm, thereby providing a safe haven for an M. tuberculosis mutant that is sensitive to iron-mediated oxidative stress. Via an independent mechanism, host ferritin is also able to rescue the growth of this mutant. Our work highlights the inherent synergy between macronutrients and micronutrients of the host environment that converge to provide resilience to the pathogen. This complex synergy afforded by the adipocyte model of infection will aid in the identification of genes required by M. tuberculosis in a caseous host environment.


Subject(s)
Adipocytes/metabolism , Adipocytes/microbiology , Iron/metabolism , Mycobacterium tuberculosis/physiology , 3T3-L1 Cells , Animals , Humans , Lipid Metabolism , Mice , RAW 264.7 Cells
8.
Biochem Biophys Res Commun ; 495(1): 353-359, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29108997

ABSTRACT

Fatty acid-binding protein 4 (FABP4), a cytosolic lipid chaperone predominantly expressed in adipocytes and macrophages, modulates lipid fluxes, trafficking, signaling, and metabolism. Recent studies have demonstrated that FABP4 regulates metabolic and inflammatory pathways, and in mouse models its inhibition can improve type 2 diabetes mellitus and atherosclerosis. However, the role of FABP4 in bacterial infection, metabolic crosstalk between host and pathogen, and bacterial pathogenesis have not been studied. As an obligate intracellular pathogen, Chlamydia pneumoniae needs to obtain nutrients such as ATP and lipids from host cells. Here, we show that C. pneumoniae successfully infects and proliferates in murine adipocytes by inducing hormone sensitive lipase (HSL)-mediated lipolysis. Chemical inhibition or genetic manipulation of HSL significantly abrogated the intracellular growth of C. pneumoniae in adipocytes. Liberated free fatty acids were utilized to generate ATP via ß-oxidation, which C. pneumoniae usurped for its replication. Strikingly, chemical inhibition or genetic silencing of FABP4 significantly abrogated C. pneumoniae infection-induced lipolysis and mobilization of liberated FFAs, resulting in reduced bacterial growth in adipocytes. Collectively, these results demonstrate that C. pneumoniae exploits host FABP4 to facilitate fat mobilization and intracellular replication in adipocytes. This work uncovers a novel strategy used by intracellular pathogens for acquiring energy via hijacking of the host lipid metabolism pathway.


Subject(s)
Adipocytes/microbiology , Adipocytes/physiology , Chlamydophila pneumoniae/physiology , Fatty Acid-Binding Proteins/metabolism , Lipid Mobilization/physiology , Sterol Esterase/metabolism , 3T3-L1 Cells , Animals , Cell Proliferation/physiology , Chlamydophila pneumoniae/cytology , Mice
9.
Microbes Infect ; 20(2): 81-88, 2018 02.
Article in English | MEDLINE | ID: mdl-29109018

ABSTRACT

Tuberculosis (TB) remains as a major threat to human health worldwide despite of the availability of standardized antibiotic therapy. One of the characteristic of pathogenic Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis is its ability to persist in the host in a dormant state and develop latent infection without clinical signs of active disease. However, the mechanisms involved in bacterial persistence and the establishment of latency is not well understood. Adipose tissue is emerging as an important niche that favors actively replicating as well as dormant Mtb during acute and latent infection. This also suggests that Mtb can disseminate from the lungs to adipose tissue during aerosol infection and/or from adipose tissue to lungs during reactivation of latent infection. In this study, we report the interplay between key adipokine levels and the dynamics of Mtb pathogenesis in the lungs and adipose tissue using a rabbit model of pulmonary infection with two clinical isolates that produce divergent outcome in disease progression. Results show that markers of adipocyte physiology and function were significantly altered during Mtb infection and distinct patterns of adipokine expression were noted between adipose tissue and the lungs. Moreover, these markers were differentially expressed between active disease and latent infection. Thus, this study highlights the importance of targeting adipocyte function as potential target for developing better TB intervention strategies.


Subject(s)
Adipocytes/pathology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/microbiology , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/microbiology , Adiponectin/genetics , Adipose Tissue/metabolism , Adipose Tissue/microbiology , Adipose Tissue/pathology , Animals , Cytokines/genetics , Disease Models, Animal , Female , Host-Pathogen Interactions , Inflammation/metabolism , Latent Tuberculosis/microbiology , Lung/metabolism , Lung/microbiology , Mice , Mycobacterium tuberculosis/physiology , PPAR gamma/genetics , Rabbits , Receptors, Adiponectin/genetics , Signal Transduction
10.
PLoS Pathog ; 13(10): e1006676, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29040326

ABSTRACT

Mycobacterium tuberculosis (Mtb) primarily resides in the lung but can also persist in extrapulmonary sites. Macrophages are considered the prime cellular habitat in all tissues. Here we demonstrate that Mtb resides inside adipocytes of fat tissue where it expresses stress-related genes. Moreover, perigonadal fat of Mtb-infected mice disseminated the infection when transferred to uninfected animals. Adipose tissue harbors leukocytes in addition to adipocytes and other cell types and we observed that Mtb infection induces changes in adipose tissue biology depending on stage of infection. Mice infected via aerosol showed infiltration of inducible nitric oxide synthase (iNOS) or arginase 1 (Arg1)-negative F4/80+ cells, despite recruitment of CD3+, CD4+ and CD8+ T cells. Gene expression analysis of adipose tissue of aerosol Mtb-infected mice provided evidence for upregulated expression of genes associated with T cells and NK cells at 28 days post-infection. Strikingly, IFN-γ-producing NK cells and Mtb-specific CD8+ T cells were identified in perigonadal fat, specifically CD8+CD44-CD69+ and CD8+CD44-CD103+ subpopulations. Gene expression analysis of these cells revealed that they expressed IFN-γ and the lectin-like receptor Klrg1 and down-regulated CD27 and CD62L, consistent with an effector phenotype of Mtb-specific CD8+ T cells. Sorted NK cells expressed higher abundance of Klrg1 upon infection, as well. Our results reveal the ability of Mtb to persist in adipose tissue in a stressed state, and that NK cells and Mtb-specific CD8+ T cells infiltrate infected adipose tissue where they produce IFN-γ and assume an effector phenotype. We conclude that adipose tissue is a potential niche for Mtb and that due to infection CD8+ T cells and NK cells are attracted to this tissue.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/microbiology , Tuberculosis/immunology , Tuberculosis/microbiology , Virus Latency/immunology , Adipocytes/microbiology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Killer Cells, Natural/immunology , Mice , Mycobacterium tuberculosis/immunology
11.
Article in English | MEDLINE | ID: mdl-28567368

ABSTRACT

Adipose tissues were shown to host Mycobacterium tuberculosis which is persisting inside mature adipocytes. It remains unknown whether this holds true for Mycobacterium canettii, a rare representative of the M. tuberculosis complex responsible for lymphatic and pulmonary tuberculosis. Here, we infected primary murine white and brown pre-adipocytes and murine 3T3-L1 pre-adipocytes and mature adipocytes with M. canettii and M. tuberculosis as a positive control. Both mycobacteria were able to infect 18-22% of challenged primary murine pre-adipocytes; and to replicate within these cells during a 7-day experiment with the intracellular inoculums being significantly higher in brown than in white pre-adipocytes for M. canettii (p = 0.02) and M. tuberculosis (p = 0.03). Further in-vitro infection of 3T3-L1 mature adipocytes yielded 9% of infected cells by M. canettii and 17% of infected cells by M. tuberculosis (p = 0.001). Interestingly, M. canettii replicated and accumulated intra-cytosolic lipid inclusions within mature adipocytes over a 12-day experiment; while M. tuberculosis stopped replicating at day 3 post-infection. These results indicate that brown pre-adipocytes could be one of the potential targets for M. tuberculosis complex mycobacteria; and illustrate differential outcome of M. tuberculosis complex mycobacteria into adipose tissues. While white adipose tissue is an unlikely sanctuary for M. canettii, it is still an open question whether M. canettii and M. tuberculosis could persist in brown adipose tissues.


Subject(s)
Adipose Tissue/microbiology , Adipose Tissue/pathology , Mycobacterium/pathogenicity , Tuberculosis/microbiology , 3T3-L1 Cells , Adipocytes/microbiology , Adipocytes/pathology , Adipose Tissue/diagnostic imaging , Animals , Colony Count, Microbial , Disease Models, Animal , Mice , Mice, Inbred BALB C , Mycobacterium/classification , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/diagnostic imaging
12.
Pharmacol Res ; 119: 303-312, 2017 05.
Article in English | MEDLINE | ID: mdl-28249816

ABSTRACT

In obesity, gut microbiota LPS may translocate into the blood stream and then contribute to adipose tissue inflammation and oxidative stress, leading to insulin resistance. A causal link between periodontal infection, obesity and type 2 diabetes has also been suggested. We evaluated the ability of polyphenols from Antirhea borbonica medicinal plant to improve the inflammatory and redox status of 3T3-L1 adipocytes exposed to LPS of Porphyromonas gingivalis periodontopathogen or Escherichia coli enterobacteria. Our results show that LPS enhanced the production of Toll-like receptor-dependent MyD88 and NFκB signaling factors as well as IL-6, MCP-1, PAI-1 and resistin. Plant polyphenols reduced LPS pro-inflammatory action. Concomitantly, polyphenols increased the production of adiponectin and PPARγ, known as key anti-inflammatory and insulin-sensitizing mediators. Moreover, both LPS increased intracellular ROS levels and the expression of genes encoding ROS-producing enzymes including NOX2, NOX4 and iNOS. Plant polyphenols reversed these effects and up-regulated MnSOD and catalase antioxidant enzyme gene expression. Noticeably, preconditioning of cells with caffeic acid, chlorogenic acid or kaempferol identified among A. borbonica major polyphenols, led to similar protective properties. Altogether, these findings demonstrate the anti-inflammatory and antioxidant effects of A. borbonica polyphenols on adipocytes, in response to P. gingivalis or E. coli LPS. It will be of major interest to assess A. borbonica polyphenol benefits against obesity-related metabolic disorders such as insulin resistance in vivo.


Subject(s)
Adipocytes/drug effects , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Escherichia coli/immunology , Lipopolysaccharides/immunology , Polyphenols/pharmacology , Porphyromonas gingivalis/immunology , 3T3-L1 Cells , Adipocytes/immunology , Adipocytes/microbiology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Antioxidants/chemistry , Antioxidants/isolation & purification , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/microbiology , Mice , Oxidative Stress/drug effects , Plants, Medicinal/chemistry , Polyphenols/chemistry , Polyphenols/isolation & purification , Rubiaceae/chemistry
13.
Int J Mycobacteriol ; 5(1): 92-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26927997

ABSTRACT

During its persistence in the infected host, Mycobacterium tuberculosis (Mtb) accumulates host-derived fatty acids in intracytoplasmic lipid inclusions as triacylglycerols which serve primarily as carbon and energy reserves. The Mtb genome codes for more than 15 triacylglycerol synthases, 24 lipase/esterases, and seven cutinase-like proteins. Hence, we looked at the expression of the corresponding genes in intracellular bacilli persisting amidst the host triacylglycerols. We used the Mtb infected murine adipocyte model to ensure persistence and transcripts were quantified using real-time reverse transcriptase polymerase chain reaction. Dormancy and glyoxylate metabolism was confirmed by the upregulated expression of dosR and icl, respectively, by intra-adipocyte bacilli compared with in vitro growing bacilli. The study revealed that tgs1, tgs2, Rv3371, and mycolyltransferase Ag85A are the predominant triacylglycerol synthases, while lipF, lipH, lipJ, lipK, lipN, lipV, lipX, lipY, culp5, culp7, and culp6 are the predominant lipases/esterases used by Mtb for the storage and degradation of host-derived fat. Moreover, it was observed that many of these enzymes are used by Mtb during active replication rather than during nonreplicating persistence, indicating their probable function in cell wall synthesis.


Subject(s)
Adaptation, Physiological/genetics , Adipocytes/microbiology , Host-Pathogen Interactions , Lipid Metabolism/genetics , Macrophages/microbiology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Adipocytes/metabolism , Animals , Bacterial Proteins/genetics , DNA-Binding Proteins , Disease Models, Animal , Gene Expression Regulation, Bacterial , Genes, Bacterial , Glyoxylates/metabolism , Lipase/metabolism , Mice , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/growth & development , Protein Kinases/genetics , Real-Time Polymerase Chain Reaction
14.
Immunol Cell Biol ; 93(9): 789-95, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25801479

ABSTRACT

Mycobacterium tuberculosis can infect 'non-classical immune cells', which comprise a significant constituency of cells that reside outside of those defined as 'classical immune cells' from myeloid or lymphoid origin. Here we address the influence of specific 'non-classical immune cells' in host responses and their effects in controlling mycobacterial growth or enabling an environment conducive for bacilli persistence. The interaction of M. tuberculosis with epithelial cells, endothelial cells, fibroblasts, adipocytes, glia and neurons and downstream cellular responses that often dictate immune regulation and disease outcome are discussed. Functional integration and synergy between 'classical' and 'non-classical immune cells' are highlighted as critical for determining optimal immune outcomes that favour the host.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Adipocytes/immunology , Adipocytes/microbiology , Endothelial Cells/immunology , Endothelial Cells/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Fibroblasts/immunology , Fibroblasts/microbiology , Humans , Models, Immunological , Mycobacterium tuberculosis/physiology , Neuroglia/immunology , Neuroglia/microbiology , Neurons/immunology , Neurons/microbiology , Tuberculosis/microbiology
15.
Science ; 347(6217): 67-71, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25554785

ABSTRACT

Adipocytes have been suggested to be immunologically active, but their role in host defense is unclear. We observed rapid proliferation of preadipocytes and expansion of the dermal fat layer after infection of the skin by Staphylococcus aureus. Impaired adipogenesis resulted in increased infection as seen in Zfp423(nur12) mice or in mice given inhibitors of peroxisome proliferator-activated receptor γ. This host defense function was mediated through the production of cathelicidin antimicrobial peptide from adipocytes because cathelicidin expression was decreased by inhibition of adipogenesis, and adipocytes from Camp(-/-) mice lost the capacity to inhibit bacterial growth. Together, these findings show that the production of an antimicrobial peptide by adipocytes is an important element for protection against S. aureus infection of the skin.


Subject(s)
Adipocytes/immunology , Cathelicidins/immunology , Dermis/immunology , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/immunology , 3T3-L1 Cells , Adipocytes/microbiology , Adipogenesis/immunology , Animals , Antimicrobial Cationic Peptides/immunology , Cathelicidins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Dermis/microbiology , Host-Pathogen Interactions/immunology , Mice , Mice, Mutant Strains , Transcription Factors/genetics , Transcription Factors/immunology
16.
Microbes Infect ; 16(7): 571-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24819214

ABSTRACT

The adipocytes are one of the non-professional phagocytes postulated to be a haven for Mycobacterium tuberculosis during persistence in the human host. The adipocyte - M. tuberculosis interaction data available to date are ex vivo. The present study was primarily aimed to investigate M. tuberculosis infection of adipocytes in course of infection of mouse model. Using primary murine adipocytes, the study first confirmed the infection and immunomodulation of natural adipocytes by M. tuberculosis. The bacilli could be isolated form visceral, subcutaneous, peri renal and mesenteric adipose depots of immunocompetent mice infected with M. tuberculosis intravenously. The bacilli could be isolated from adipocytes and the stromal vascular fraction, even though the numbers were significantly higher in the latter. The bacterial burden in the adipose depots was comparable to those in lungs in the early phase of infection. But with time, the burden in the adipose depots was either decreased or kept under control, despite the increasing burden in the lungs. Infected mice treated with standard anti tubercular drugs, despite effective elimination of bacterial loads in the lungs, continued to harbour M. tuberculosis in adipose depots at loads similar to untreated mice in the late infection phase.


Subject(s)
Adipocytes/immunology , Adipocytes/microbiology , Antitubercular Agents/therapeutic use , Mycobacterium tuberculosis/growth & development , Tuberculosis/microbiology , Adipocytes/chemistry , Adipokines/analysis , Adipose Tissue/microbiology , Animals , Antitubercular Agents/pharmacology , Cells, Cultured , Cytokines/analysis , Disease Models, Animal , Host-Pathogen Interactions/immunology , Lipid Droplets , Mice , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/drug therapy
17.
PLoS One ; 7(7): e41079, 2012.
Article in English | MEDLINE | ID: mdl-22844426

ABSTRACT

BACKGROUND: Associations have been made between obesity and reduced intestinal numbers of members of the phylum Bacteroidetes, but there is no direct evidence of the role these bacteria play in obesity. Herein, the effects of Bacteroides uniformis CECT 7771 on obesity-related metabolic and immune alterations have been evaluated. METHODS AND FINDINGS: Adult (6-8 week) male wild-type C57BL-6 mice were fed a standard diet or a high-fat-diet HFD to induce obesity, supplemented or not with B. uniformis CECT 7771 for seven weeks. Animal weight was monitored and histologic, biochemical, immunocompetent cell functions, and features of the faecal microbiota were analysed after intervention. The oral administration of B. uniformis CECT 7771 reduced body weight gain, liver steatosis and liver cholesterol and triglyceride concentrations and increased small adipocyte numbers in HFD-fed mice. The strain also reduced serum cholesterol, triglyceride, glucose, insulin and leptin levels, and improved oral tolerance to glucose in HFD fed mice. The bacterial strain also reduced dietary fat absorption, as indicated by the reduced number of fat micelles detected in enterocytes. Moreover, B. uniformis CECT 7771 improved immune defence mechanisms, impaired in obesity. HFD-induced obesity led to a decrease in TNF-α production by peritoneal macrophages stimulated with LPS, conversely, the administration of B. uniformis CECT 7771 increased TNF-α production and phagocytosis. Administering this strain also increased TNF-α production by dendritic cells (DCs) in response to LPS stimulation, which was significantly reduced by HFD. B. uniformis CECT 7771 also restored the capacity of DCs to induce a T-cell proliferation response, which was impaired in obese mice. HFD induced marked changes in gut microbiota composition, which were partially restored by the intervention. CONCLUSIONS: Altogether, the findings indicate that administration of B. uniformis CECT 7771 ameliorates HFD-induced metabolic and immune dysfunction associated with intestinal dysbiosis in obese mice.


Subject(s)
Bacteroides/physiology , Diet, High-Fat/adverse effects , Obesity/immunology , Obesity/metabolism , Absorption , Adipocytes/microbiology , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/microbiology , Animals , Body Weight , Cell Size , Cytokines/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/microbiology , Enterocytes/metabolism , Enterocytes/microbiology , Fatty Liver/immunology , Fatty Liver/microbiology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Male , Metagenome , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/microbiology , Species Specificity
18.
Environ Entomol ; 41(1): 108-17, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22525065

ABSTRACT

Xylophagous insects derive nutrients from intractable substrates by producing or ingesting cellulolytic enzymes, or by maintaining associations with symbiotic microbes. Wood-boring cerambycid beetle larvae sometimes house maternally-transmitted endosymbiotic yeasts that are presumed to provide their hosts with nutritional benefits. These are thought to be absent from species in the large subfamily Lamiinae; nevertheless yeasts have been repeatedly isolated from the guts of neotropical lamiines. The objective of this study was to conduct transmission electron microscopy (TEM) studies of cerambycid larval midgut tissues to determine if gut yeasts were intracellular, or simply present in the gut lumen. Nine cerambycid larvae were harvested from two trees in the Brazil nut family (Lecythidaceae) in the rain forest of SE Peru; seven were identified using mtDNA sequence data and processed for TEM. Yeasts cultured from larval frass or exuvia, and identified with rDNA sequence data, were identical or similar to yeasts previously isolated from beetles. In TEM analyses yeast cells were found only in the gut lumens, sometimes associated with fragments of thick-walled xylem cells. Apparent bacteriocytes were found in either midgut or fat body tissue of three larval specimens, including two lamiines. This is the first report of a potential fat body symbiosis in a cerambycid beetle. Future studies of cerambycid symbiosis should distinguish the identities and potential roles of free-living organisms in the gut lumen from those of organisms harbored within gut epithelial or fat body tissue.


Subject(s)
Bacteria/isolation & purification , Coleoptera/cytology , Coleoptera/microbiology , Fat Body/microbiology , Yeasts/isolation & purification , Adipocytes/cytology , Adipocytes/microbiology , Animals , Bacteria/genetics , Bacterial Physiological Phenomena , Coleoptera/genetics , DNA, Fungal/genetics , DNA, Mitochondrial/genetics , DNA, Ribosomal/genetics , Electron Transport Complex IV/genetics , Fat Body/cytology , Feces/microbiology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/microbiology , Genes, Insect , Larva/cytology , Larva/genetics , Larva/microbiology , Microscopy, Electron, Transmission , Molecular Sequence Data , Peru , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Species Specificity , Symbiosis , Yeasts/classification , Yeasts/genetics , Yeasts/physiology
19.
Microbes Infect ; 13(14-15): 1242-51, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21813088

ABSTRACT

Mycobacterium tuberculosis (M. tb) takes advantage of various cell types, allowing it to remain in the host for long periods. Because adipocytes have been proposed as niches for dormant M. tb in the latent state, understanding the interaction of virulent M. tb with adipocytes is important. We compared changes in cytokine secretion from 3T3-L1 murine adipocytes infected with virulent M. tb H37Rv (V-M. tb) and attenuated M. tb H37Ra (A-M. tb) strains. Both strains maintained non-replicating states within adipocytes until 10 days post-infection. Adipocytes infected with V-M. tb secreted lower levels of pro-inflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-12p40, IL-6, and IL-17, and lower levels of nitric oxide than those infected with A-M. tb. In contrast, the anti-inflammatory cytokines, IL-10 and IL-4, were markedly induced in V-M. tb-infected adipocytes versus those infected with A-M. tb at an early time point. Heat-killed or formalin-fixed bacteria induced lower levels of cytokines and no difference was observed between strains. Moreover, V-M. tb induced a high level of necrosis versus A-M. tb in conjunction with increased levels of LHD. These results suggest that V-M. tb regulates cytokine expression in its favor, increasing cytokines necessary for immune evasion and decreasing those required for protective immunity.


Subject(s)
Adipocytes/immunology , Immune Evasion , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , 3T3-L1 Cells , Adipocytes/microbiology , Animals , Blotting, Western , Extracellular Space/chemistry , Formaldehyde , Hot Temperature , Humans , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-12 Subunit p40/biosynthesis , Interleukin-12 Subunit p40/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , L-Lactate Dehydrogenase/analysis , L-Lactate Dehydrogenase/metabolism , Mice , Microscopy, Fluorescence , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/microbiology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
20.
Antimicrob Agents Chemother ; 54(6): 2712-5, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20350948

ABSTRACT

Aerobic (5-day-old cultures) and nonreplicating (dormant) Mycobacterium tuberculosis (5-, 12-, and 19-day-old cultures) bacteria were treated with rifampin (R), moxifloxacin (MX), metronidazole (MZ), amikacin (AK), or capreomycin (CP) for 7, 14, and 21 days. R-MX-MZ-AK and R-MX-MZ-CP killed both aerobic and dormant bacilli in 21 days, as shown by lack of regrowth in solid and liquid media. R-MX-MZ-AK and R-MX-MZ-CP also caused a strong decrease of nonreplicating bacilli in 7 days in a cell-based dormancy model.


Subject(s)
Antitubercular Agents/administration & dosage , Latent Tuberculosis/drug therapy , Latent Tuberculosis/microbiology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , 3T3 Cells , Adipocytes/drug effects , Adipocytes/microbiology , Amikacin/administration & dosage , Animals , Aza Compounds/administration & dosage , Capreomycin/administration & dosage , Colony Count, Microbial , Drug Combinations , Drug Resistance, Bacterial , Fluoroquinolones , Humans , In Vitro Techniques , Metronidazole/administration & dosage , Mice , Microbial Sensitivity Tests , Models, Biological , Moxifloxacin , Quinolines/administration & dosage , Rifampin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...