Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
FEBS J ; 286(16): 3164-3181, 2019 08.
Article in English | MEDLINE | ID: mdl-31034693

ABSTRACT

The Mycobacterium tuberculosis LipY protein, a prototype of the proline-glutamic acid (PE) family, exhibits a triacylglycerol (TAG) hydrolase activity that contributes to host cell lipid degradation and persistence of the bacilli. LipY is found either as a full-length intracytosolic form or as a mature extracellular form lacking the N-terminal PE domain. Even though the contribution of the extracellular form in TAG consumption has been partly elucidated, very little information is available regarding the potential interactions of either full-length LipY with the cytoplasmic membrane, or mature form LipY with the outer membrane. Herein, several LipY variants truncated in their N-terminal domain were produced and biochemically characterized in lipid-protein interaction assays, using the monomolecular film technique and FTIR. Comparison of the catalytic activities of these recombinant proteins showed that LipY∆149, corresponding to the extracellular form of LipY lacking the PE domain, is more active than the full-length protein. This confirms previous studies reporting that the PE domain negatively modulates the TAG hydrolase activity of LipY. Lipid-protein interaction studies indicate that the PE domain anchors LipY onto membrane lipids. Consistent with these findings, we show that LipY∆149 is loosely associated with the mycobacterial cell wall, and that this interaction is mediated by the sole lipase domain. Overall, our results bring new information regarding the molecular mechanisms by which LipY either binds and hydrolyses host cell lipids or degrades TAG, the major source of lipids within mycobacterial intracytosolic lipid inclusions.


Subject(s)
Bacterial Proteins/genetics , Carboxylic Ester Hydrolases/genetics , Lipid Metabolism/genetics , Membrane Lipids/genetics , Mycobacterium tuberculosis/genetics , Virulence Factors/genetics , Bacterial Proteins/metabolism , Carboxylic Ester Hydrolases/metabolism , Catalysis , Cell Wall/genetics , Cell Wall/metabolism , Lipase/genetics , Membrane Lipids/metabolism , Mycobacterium tuberculosis/metabolism , Protein Binding/genetics , Protein Domains/genetics , Triglycerides/genetics , Triglycerides/metabolism , Virulence Factors/metabolism
2.
Infect Immun ; 86(9)2018 09.
Article in English | MEDLINE | ID: mdl-29986895

ABSTRACT

Within tuberculous granulomas, a subpopulation of Mycobacterium tuberculosis resides inside foamy macrophages (FM) that contain abundant cytoplasmic lipid bodies (LB) filled with triacylglycerol (TAG). Upon fusion of LB with M. tuberculosis-containing phagosomes, TAG is hydrolyzed and reprocessed by the bacteria into their own lipids, which accumulate as intracytosolic lipid inclusions (ILI). This phenomenon is driven by many mycobacterial lipases, among which LipY participates in the hydrolysis of host and bacterial TAG. However, the functional contribution of LipY's PE domain to TAG hydrolysis remains unclear. Here, enzymatic studies were performed to compare the lipolytic activities of recombinant LipY and its truncated variant lacking the N-terminal PE domain, LipY(ΔPE). Complementarily, an FM model was used where bone marrow-derived mouse macrophages were infected with M. bovis BCG strains either overexpressing LipY or LipY(ΔPE) or carrying a lipY deletion mutation prior to being exposed to TAG-rich very-low-density lipoprotein (VLDL). Results indicate that truncation of the PE domain correlates with increased TAG hydrolase activity. Quantitative electron microscopy analyses showed that (i) in the presence of lipase inhibitors, large ILI (ILI+3) were not formed because of an absence of LB due to inhibition of VLDL-TAG hydrolysis or inhibition of LB-neutral lipid hydrolysis by mycobacterial lipases, (ii) ILI+3 profiles in the strain overexpressing LipY(ΔPE) were reduced, and (iii) the number of ILI+3 profiles in the ΔlipY mutant was reduced by 50%. Overall, these results delineate the role of LipY and its PE domain in host and mycobacterial lipid consumption and show that additional mycobacterial lipases take part in these processes.


Subject(s)
Bacterial Proteins/chemistry , Carboxylic Ester Hydrolases/chemistry , Lipid Metabolism , Macrophages/microbiology , Macrophages/physiology , Triglycerides/metabolism , Virulence Factors/chemistry , Animals , Bacterial Proteins/genetics , Carboxylic Ester Hydrolases/genetics , Catalytic Domain , Cells, Cultured , Female , Lipase/metabolism , Lipoproteins, VLDL/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mycobacterium bovis , Protein Structure, Tertiary , Tuberculosis/microbiology , Virulence Factors/genetics
3.
Food Funct ; 5(7): 1409-21, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24777447

ABSTRACT

CITREM is an emulsifier used in the food industry and contains citric acid esters of mono- and diglycerides (GCFE). It is generally recognized as safe but no publication on its digestibility under gastrointestinal conditions and impact on fat digestion was available. It was shown here that fatty acids are released from CITREM by gastric lipase, pancreatic lipase, pancreatic-lipase-related protein 2 and carboxyl ester hydrolase. A two-step in vitro digestion model mimicking lipolysis in the stomach and upper small intestine of term and preterm infants was then used to evaluate the digestibility of CITREM alone, CITREM-containing infant formula and fat emulsions, and isolated GCFE fractions. Overall, it was shown that fat digestion is not significantly changed by the presence of CITREM, and fatty acids contained in CITREM compounds are released to a large extent by lipases. Nevertheless, undigestible water-soluble compounds containing glycerol and citric acid units were identified, indicating that the ester bond between citric acid and glycerol is not fully hydrolyzed throughout the proposed digestion.


Subject(s)
Citrates/metabolism , Diglycerides/metabolism , Emulsifying Agents/metabolism , Esters/metabolism , Infant Formula/chemistry , Monoglycerides/metabolism , Carboxylesterase/metabolism , Digestion , Emulsions/chemistry , Fatty Acids/metabolism , Gastrointestinal Tract/enzymology , Humans , Infant , Lipase/metabolism , Lipolysis
4.
J Med Chem ; 55(22): 10204-19, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23095026

ABSTRACT

A new series of customizable diastereomeric cis- and trans-monocyclic enol-phosphonate analogs to Cyclophostin and Cyclipostins were synthesized. Their potencies and mechanisms of inhibition toward six representative lipolytic enzymes belonging to distinct lipase families were examined. With mammalian gastric and pancreatic lipases no inhibition occurred with any of the compounds tested. Conversely, Fusarium solani Cutinase and lipases from Mycobacterium tuberculosis (Rv0183 and LipY) were all fully inactivated. The best inhibitors displayed a cis conformation (H and OMe) and exhibited higher inhibitory activities than the lipase inhibitor Orlistat toward the same enzymes. Our results have revealed that chemical group at the γ-carbon of the phosphonate ring strongly impacts the inhibitory efficiency, leading to a significant improvement in selectivity toward a target lipase over another. The powerful and selective inhibition of microbial (fungal and mycobacterial) lipases suggests that these seven-membered monocyclic enol-phosphonates should provide useful leads for the development of novel and highly selective antimicrobial agents.


Subject(s)
Carboxylic Ester Hydrolases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Organophosphonates/chemistry , Organophosphorus Compounds/chemistry , Carboxylic Ester Hydrolases/metabolism , Fusarium/enzymology , Kinetics , Lipase/antagonists & inhibitors , Lipase/metabolism , Molecular Structure , Mycobacterium tuberculosis/enzymology , Structure-Activity Relationship
5.
PLoS One ; 7(9): e46493, 2012.
Article in English | MEDLINE | ID: mdl-23029536

ABSTRACT

Lipid metabolism plays an important role during the lifetime of Mycobacterium tuberculosis, the causative agent of tuberculosis. Although M. tuberculosis possesses numerous lipolytic enzymes, very few have been characterized yet at a biochemical/pharmacological level. This study was devoted to the M. tuberculosis lipolytic enzymes belonging to the Hormone-Sensitive Lipase (HSL) family, which encompasses twelve serine hydrolases closely related to the human HSL. Among them, nine were expressed, purified and biochemically characterized using a broad range of substrates. In vitro enzymatic inhibition studies using the recombinant HSL proteins, combined with mass spectrometry analyses, revealed the potent inhibitory activity of an oxadiazolone compound, named MmPPOX. In addition, we provide evidence that MmPPOX alters mycobacterial growth. Overall, these findings suggest that the M. tuberculosis HSL family displays important metabolic functions, thus opening the way to further investigations linking the involvement of these enzymes in mycobacterial growth.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Oxadiazoles/pharmacology , Sterol Esterase/antagonists & inhibitors , Anti-Bacterial Agents/chemistry , Bacterial Proteins/biosynthesis , Bacterial Proteins/chemistry , Enzyme Inhibitors/chemistry , Kinetics , Lactones/pharmacology , Molecular Weight , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/growth & development , Orlistat , Oxadiazoles/chemistry , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sterol Esterase/biosynthesis , Sterol Esterase/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...