Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Immunol ; 198(5): 2172-2181, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28100682

ABSTRACT

The gastrointestinal tract is continuously exposed to many environmental factors that influence intestinal epithelial cells and the underlying mucosal immune system. In this article, we demonstrate that dietary fiber and short chain fatty acids (SCFAs) induced the expression of the vitamin A-converting enzyme RALDH1 in intestinal epithelial cells in vivo and in vitro, respectively. Furthermore, our data showed that the expression levels of RALDH1 in small intestinal epithelial cells correlated with the activity of vitamin A-converting enzymes in mesenteric lymph node dendritic cells, along with increased numbers of intestinal regulatory T cells and a higher production of luminal IgA. Moreover, we show that the consumption of dietary fiber can alter the composition of SCFA-producing microbiota and SCFA production in the small intestines. In conclusion, our data illustrate that dietary adjustments affect small intestinal epithelial cells and can be used to modulate the mucosal immune system.


Subject(s)
Dendritic Cells/immunology , Diet , Epithelial Cells/immunology , Intestinal Mucosa/immunology , Isoenzymes/metabolism , Retinal Dehydrogenase/metabolism , T-Lymphocytes, Regulatory/immunology , Aldehyde Dehydrogenase 1 Family , Animals , Cells, Cultured , Fatty Acids, Volatile/metabolism , Immune Tolerance , Immunity, Mucosal , Immunoglobulin A/metabolism , Isoenzymes/genetics , Mice , Mice, Inbred C57BL , Microbiota , Receptors, G-Protein-Coupled/genetics , Receptors, Nicotinic/genetics , Retinal Dehydrogenase/genetics , Vitamin A/metabolism
2.
Trends Biochem Sci ; 39(11): 527-35, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25220377

ABSTRACT

An emerging theme in the regulation of cholesterol homeostasis is the role of the ubiquitin proteasome system (UPS), through which proteins are ubiquitylated and then degraded in response to specific signals. The UPS controls all aspects of cholesterol metabolism including its synthesis, uptake, and efflux. We review here recent work uncovering the ubiquitylation and degradation of key players in cholesterol homeostasis. This includes the low-density lipoprotein (LDL) receptor, transcription factors (sterol regulatory element binding proteins and liver X receptors), flux-controlling enzymes in cholesterol synthesis (3-hydroxy-3-methylglutaryl-CoA reductase and squalene monooxygenase), and cholesterol exporters (ATP-binding cassette transporters ABCA1 and ABCG1). We explore which E3 ligases are involved, and identify areas deserving of further research.


Subject(s)
Cholesterol/metabolism , Homeostasis , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , ATP-Binding Cassette Transporters/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/metabolism , Models, Biological , Receptors, LDL/metabolism , Squalene Monooxygenase/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
Eur J Immunol ; 45(1): 89-100, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25311225

ABSTRACT

The vitamin A metabolite retinoic acid (RA) has been reported to suppress Th1 responses and enhance Th2 responses. Here, we investigated whether differences in vitamin A metabolism could underlie the differences between C57BL/6 and BALB/c mice, which are reportedly seen as Th1 and Th2 responders, respectively. BALB/c mice were shown to have higher intestinal epithelial expression of RALDH1 (where RALDH is retinaldehyde dehydrogenase), and, consequently, higher RALDH activity in MLN-DCs, leading to an increased ability to induce IgA class switching in B cells. Furthermore, within BALB/c mice, induction of IgA secretion as well as increased accumulation of regulatory T cells (Treg) in the intestinal lamina propria was observed. Additionally, as BALB/c mice are more resistant to dextran sulphate sodium (DSS) induced colitis, mice that lacked vitamin A in their diet had a more severe form of DSS-induced colitis compared to control mice. Therefore, the level of RA production and consequently the degree of RA-mediated signaling is crucial for the efficiency of the mucosal immune system.


Subject(s)
Colitis/immunology , Immunity, Mucosal , Intestines/immunology , Isoenzymes/immunology , Mucous Membrane/immunology , Retinal Dehydrogenase/immunology , Vitamin A/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate , Gene Expression , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin Class Switching , Intestinal Mucosa/metabolism , Intestines/pathology , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucous Membrane/metabolism , Mucous Membrane/pathology , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Signal Transduction , Species Specificity , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology , Vitamin A/administration & dosage
5.
Cell Mol Immunol ; 19(2): 260-275, 2022 02.
Article in English | MEDLINE | ID: mdl-34983945

ABSTRACT

Conventional dendritic cells (cDCs) scan and integrate environmental cues in almost every tissue, including exogenous metabolic signals. While cDCs are critical in maintaining immune balance, their role in preserving energy homeostasis is unclear. Here, we showed that Batf3-deficient mice lacking conventional type 1 DCs (cDC1s) had increased body weight and adiposity during aging. This led to impaired energy expenditure and glucose tolerance, insulin resistance, dyslipidemia, and liver steatosis. cDC1 deficiency caused adipose tissue inflammation that was preceded by a paucity of NK1.1+ invariant NKT (iNKT) cells. Accordingly, among antigen-presenting cells, cDC1s exhibited notable induction of IFN-γ production by iNKT cells, which plays a metabolically protective role in lean adipose tissue. Flt3L treatment, which expands the dendritic cell (DC) compartment, mitigated diet-induced obesity and hyperlipidemia in a Batf3-dependent manner. This effect was partially mediated by NK1.1+ cells. These results reveal a new critical role for the cDC1-iNKT cell axis in the regulation of adipose tissue homeostasis.


Subject(s)
Natural Killer T-Cells , Obesity , Adipose Tissue , Animals , Dendritic Cells , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Microorganisms ; 9(2)2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33673117

ABSTRACT

Leishmania amazonensis parasites are etiological agents of cutaneous leishmaniasis in the New World. BALB/c mice are highly susceptible to L. amazonensis challenge due to their inability to mount parasite-dependent IFN-γ-mediated responses. Here, we analyzed the capacity of a single administration of the LiΔHSP70-II genetically-modified attenuated L. infantum line in preventing cutaneous leishmaniasis in mice challenged with L. amazonensis virulent parasites. In previous studies, this live attenuated vaccine has demonstrated to induce long-protection against murine leishmaniasis due to Old World Leishmania species. Vaccinated mice showed a reduction in the disease evolution due to L. amazonensis challenge, namely reduction in cutaneous lesions and parasite burdens. In contrast to control animals, after the challenge, protected mice showed anti-Leishmania IgG2a circulating antibodies accompanied to the induction of Leishmania-driven specific IFN-γ systemic response. An analysis performed in the lymph node draining the site of infection revealed an increase of the parasite-specific IFN-ϒ production by CD4+ and CD8+ T cells and a decrease in the secretion of IL-10 against leishmanial antigens. Since the immunity caused by the inoculation of this live vaccine generates protection against different forms of murine leishmaniasis, we postulate LiΔHSP70-II as a candidate for the development of human vaccines.

7.
Cell Rep ; 33(9): 108468, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33264612

ABSTRACT

Intracellular pathogens have evolved strategies to evade detection by cytotoxic CD8+ T lymphocytes (CTLs). Here, we ask whether Leishmania parasites trigger the SHP-1-FcRγ chain inhibitory axis to dampen antigen cross-presentation in dendritic cells expressing the C-type lectin receptor Mincle. We find increased cross-priming of CTLs in Leishmania-infected mice deficient for Mincle or with a selective loss of SHP-1 in CD11c+ cells. The latter also shows improved cross-presentation of cell-associated viral antigens. CTL activation in vitro reveals increased MHC class I-peptide complex expression in Mincle- or SHP-1-deficient CD11c+ cells. Neuraminidase treatment also boosts cross-presentation, suggesting that Leishmania triggers SHP-1-associated sialic-acid-binding receptors. Mechanistically, enhanced antigen processing correlates with reduced endosomal acidification in the absence of SHP-1. Finally, we demonstrate that SHP-1 inhibition improves CD11c+ cell-based vaccination against the parasite. Thus, SHP-1-mediated impairment of cross-presentation can be exploited by pathogens to evade CTLs, and SHP-1 inhibition improves CTL responses during vaccination.


Subject(s)
Antigen Presentation/immunology , Cross-Priming/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Animals , Leishmania , Mice
8.
Front Immunol ; 11: 590934, 2020.
Article in English | MEDLINE | ID: mdl-33362772

ABSTRACT

Unveiling the protective immune response to visceral leishmaniasis is critical for a rational design of vaccines aimed at reducing the impact caused by this fatal, if left untreated, vector-borne disease. In this study we sought to determine the role of the basic leucine zipper transcription factor ATF-like 3 (Batf3) in the evolution of infection with Leishmania infantum, the causative agent of human visceral leishmaniasis in the Mediterranean Basin and Latin America. For that, Batf3-deficient mice in C57BL/6 background were infected with an L. infantum strain expressing the luciferase gene. Bioluminescent imaging, as well as in vitro parasite titration, demonstrated that Batf3-deficient mice were unable to control hepatic parasitosis as opposed to wild-type C57BL/6 mice. The impaired microbicide capacities of L. infantum-infected macrophages from Batf3-deficient mice mainly correlated with a reduction of parasite-specific IFN-γ production. Our results reinforce the implication of Batf3 in the generation of type 1 immunity against infectious diseases.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Disease Resistance/immunology , Leishmania infantum , Leishmaniasis, Visceral/immunology , Repressor Proteins/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Bone Marrow/parasitology , Cytokines/immunology , Disease Models, Animal , Female , Leishmaniasis, Visceral/parasitology , Liver/parasitology , Mice, Inbred C57BL , Mice, Knockout , Nitrites/immunology , Repressor Proteins/genetics , Spleen/cytology , Spleen/parasitology , T-Lymphocytes/immunology
9.
Nat Metab ; 2(9): 974-988, 2020 09.
Article in English | MEDLINE | ID: mdl-32943786

ABSTRACT

Proinflammatory macrophages are key in the development of obesity. In addition, reactive oxygen species (ROS), which activate the Fgr tyrosine kinase, also contribute to obesity. Here we show that ablation of Fgr impairs proinflammatory macrophage polarization while preventing high-fat diet (HFD)-induced obesity in mice. Systemic ablation of Fgr increases lipolysis and liver fatty acid oxidation, thereby avoiding steatosis. Knockout of Fgr in bone marrow (BM)-derived cells is sufficient to protect against insulin resistance and liver steatosis following HFD feeding, while the transfer of Fgr-expressing BM-derived cells reverts protection from HFD feeding in Fgr-deficient hosts. Scavenging of mitochondrial peroxides is sufficient to prevent Fgr activation in BM-derived cells and HFD-induced obesity. Moreover, Fgr expression is higher in proinflammatory macrophages and correlates with obesity traits in both mice and humans. Thus, our findings reveal the mitochondrial ROS-Fgr kinase as a key regulatory axis in proinflammatory adipose tissue macrophage activation, diet-induced obesity, insulin resistance and liver steatosis.


Subject(s)
Diet, High-Fat , Inflammation/physiopathology , Macrophage Activation , Obesity/enzymology , Obesity/physiopathology , Proto-Oncogene Proteins/metabolism , src-Family Kinases/metabolism , Adipose Tissue, White/metabolism , Animals , Bone Marrow Cells/metabolism , Fatty Liver/genetics , Fatty Liver/physiopathology , Insulin Resistance , Interleukin-1beta/biosynthesis , Magnetic Resonance Imaging , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Liver/metabolism , Obesity/genetics , Proto-Oncogene Proteins/genetics , Reactive Oxygen Species/metabolism , src-Family Kinases/genetics
10.
Atherosclerosis ; 281: 137-142, 2019 02.
Article in English | MEDLINE | ID: mdl-30658189

ABSTRACT

BACKGROUND AND AIMS: Cholesterol is an essential lipid for cellular function and membrane integrity, and hence its cellular levels and distribution must be tightly regulated. Biosynthesis of cholesterol is ramped when its cellular levels are low. Herein, the ER-resident and rate-limiting enzymes 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) and squalene monooxygenase (SQLE) play a prominent role. We have recently reported that MARCH6, an E3 ubiquitin ligase, specifically promotes cholesterol-stimulated ubiquitylation and subsequent proteasomal degradation of SQLE, but not of HMGCR. To further delineate how post-translational regulation of SQLE and HMGCR is differentially achieved, we hypothesized that their sterol-dependent degradation machinery makes use of distinct E2 ubiquitin conjugating enzymes. METHODS: To study this possibility, we therefore used a CRISPR/Cas9-based approach to screen for ER-associated degradation (ERAD)-associated E2 enzymes that are essential for MARCH6-dependent degradation of SQLE. RESULTS: We report here the identification of UBE2J2 as the primary E2 ubiquitin conjugating enzyme essential for this process in mammalian cells, in contrast to UBE2G2, which is essential for sterol-stimulated degradation of HMGCR. We demonstrate that ablating UBE2J2 disturbs cholesterol-accelerated SQLE degradation in multiple human cell types, including cells of hepatic origin, and that the ability of UBE2J2 to support SQLE degradation critically depends on its enzymatic activity. CONCLUSIONS: Our findings establish UBE2J2 as an important partner of MARCH6 in cholesterol-stimulated degradation of SQLE, thereby contributing to the complex regulation of cellular cholesterol homeostasis.


Subject(s)
Cholesterol/biosynthesis , Hepatocytes/enzymology , Hydroxymethylglutaryl CoA Reductases/metabolism , Membrane Proteins/metabolism , Squalene Monooxygenase/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Enzyme Stability , HEK293 Cells , Hep G2 Cells , Humans , Membrane Proteins/genetics , Proteolysis , Time Factors , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
11.
PLoS One ; 12(2): e0172721, 2017.
Article in English | MEDLINE | ID: mdl-28231341

ABSTRACT

Cellular cholesterol metabolism is subject to tight regulation to maintain adequate levels of this central lipid molecule. Herein, the sterol-responsive Liver X Receptors (LXRs) play an important role owing to their ability to reduce cellular cholesterol load. In this context, identifying the full set of LXR-regulated genes will contribute to our understanding of their role in cholesterol metabolism. Using global transcriptional analysis we report here the identification of RNF145 as an LXR-regulated target gene. We demonstrate that RNF145 is regulated by LXRs in both human and mouse primary cells and cell lines, and in vivo in mice. Regulation of RNF145 by LXR depends on a functional LXR-element in its proximal promotor. Consistent with LXR-dependent regulation of Rnf145 we show that regulation is lost in macrophages and fibroblasts from Lxrαß(-/-) mice, and also in vivo in livers of Lxrα(-/-) mice treated with the LXR synthetic ligand T0901317. RNF145 is closely related to RNF139/TRC8, an E3 ligase implicated in control of SREBP processing. However, silencing of RNF145 in HepG2 or HeLa cells does not impair SREBP1/2 processing and sterol-responsive gene expression in these cells. Similar to TRC8, we demonstrate that RNF145 is localized to the ER and that it possesses intrinsic E3 ubiquitin ligase activity. In summary, we report the identification of RNF145 as an ER-resident E3 ubiquitin ligase that is transcriptionally controlled by LXR.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Liver X Receptors/genetics , Membrane Proteins/genetics , Transcription, Genetic , Animals , Cell Line , Cholesterol/metabolism , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver/drug effects , Liver/metabolism , Liver X Receptors/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic , Sulfonamides/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
12.
Mol Cell Biol ; 34(7): 1262-70, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24449766

ABSTRACT

The mevalonate pathway is used by cells to produce sterol and nonsterol metabolites and is subject to tight metabolic regulation. We recently reported that squalene monooxygenase (SM), an enzyme controlling a rate-limiting step in cholesterol biosynthesis, is subject to cholesterol-dependent proteasomal degradation. However, the E3-ubiquitin (E3) ligase mediating this effect was not established. Using a candidate approach, we identify the E3 ligase membrane-associated RING finger 6 (MARCH6, also known as TEB4) as the ligase controlling degradation of SM. We find that MARCH6 and SM physically interact, and consistent with MARCH6 acting as an E3 ligase, its overexpression reduces SM abundance in a RING-dependent manner. Reciprocally, knockdown of MARCH6 increases the level of SM protein and prevents its cholesterol-regulated degradation. Additionally, this increases cell-associated SM activity but is unexpectedly accompanied by increased flux upstream of SM. Prompted by this observation, we found that knockdown of MARCH6 also controls the level of 3-hydroxy-3-methyl-glutaryl coenzyme A reductase (HMGCR) in hepatocytes and model cell lines. In conclusion, MARCH6 controls abundance of both SM and HMGCR, establishing it as a major regulator of flux through the cholesterol synthesis pathway.


Subject(s)
Cholesterol/biosynthesis , Hydroxymethylglutaryl CoA Reductases/metabolism , Membrane Proteins/metabolism , Squalene Monooxygenase/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line , Enzyme Stability , Gene Knockdown Techniques , HEK293 Cells , Hep G2 Cells , Hepatocytes/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mevalonic Acid/metabolism , Models, Biological , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Squalene Monooxygenase/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL