Etiketter

Leta i den här bloggen

fredag 12 oktober 2018

Kolesterolin kuljetus (NPC1) endoplasmiseen retikulumiin ja NLRP3-inflammasomin aktivoituminen

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6168277/
Trafficking of cholesterol to the ER is required for NLRP3 inflammasome activation.
 Suomennosta abstraktista:
Solulipidit määräävät kalvointegriteetin ja fluiditeetin ja lisääntyvässä määrin  niitä  tunnistetaan  immuunivasteeseen vaikuttavina seikkoina.  Solun koleterolintarpeet täyttyvät biosynteesin ja  soluunotto-ohjelman avulla. Eräässä hyvin  mutkikkaassa tiessä, johon osallistuu  lysosomaalinen kolesterolin kuljettaja NPC1, steroli jakauttuu epätasaisesti intrasellulaarisissa aitioissa. Tutkijat kohdistivat NPC1 -kolesterolinkuljettajaan tutkimuksia farmakologisin ja geneettisin lähestymistavoin ja he saivat selville, että  kolesterolin kuljetuksen   blokeeraaminen myöhäisen endosomin (LE)--lysosomin kuljetustiessä  heikensi  NLRP3-inflammasomin aktivaatiota. 
 (NLRP3 on NOD -kaltaisia reseptoreita NLR , jolla on PYD, NACHT ja  LRR (leusiinitoisto) domeenit). 
Muuntunut kolesterolin lokalisoituminen plasmakalvossa  NPC1(-/-)  mallissa  poisti (kumosi)  TLR4- reseptorin  AKT/mTOR-signaloinnin.
Kuitenkin NLRP3-inflammasomin aktivoitumisen vajaakykyisyys johtui häriintyneestä  kolesterolin kuljetuksesta endoplasmiseen retikulumiin (ER)  ( eikä siis plasmakalvoon). 
Sen mukaan statiinin vaikuttama  akuutti kolesterolin vähenemä ER- kalvoista kumosi  caspaasi-1- aktiivisuuden  ja Il-1Beta-sekretion sekä   irrotti  NLRP3 -inflammasomin koostumuksestaan. 
Päinvastoin AIM2-inflammasomi suoritti koostumisen ja aktivaation progression ilman rajoitusta. 
Yhteenvetona  tutkimus selvittää Endoplasmisen retikulumin (ER) sterolipitoisuudet  metaboliseksi reostaatiksi  NLRP3-inflammasomin aktivaatiossa.
  • Abstract. Cellular lipids determine membrane integrity and fluidity and are being increasingly recognized to influence immune responses. Cellular cholesterol requirements are fulfilled through biosynthesis and uptake programs. In an intricate pathway involving the lysosomal cholesterol transporter NPC1, the sterol gets unequally distributed across intracellular compartments. By using pharmacological and genetic approaches targeting NPC1, we reveal that blockade of cholesterol trafficking through the late endosome–lysosome pathway blunts NLRP3 inflammasome activation.
  •  Altered cholesterol localization at the plasma membrane (PM) in Npc1−/− cells abrogated AKT–mTOR signaling by TLR4. However, the inability to activate the NLRP3 inflammasome was traced to perturbed cholesterol trafficking to the ER but not the PM. Accordingly, acute cholesterol depletion in the ER membranes by statins abrogated casp-1 activation and IL-1β secretion and ablated NLRP3 inflammasome assembly. By contrast, assembly and activation of the AIM2 inflammasome progressed unrestricted. Together, this study reveals ER sterol levels as a metabolic rheostat for the activation of the NLRP3 inflammasome.

Introduction

The inflammasome is a multiprotein complex that plays critical roles in infectious, inflammatory, and autoimmune diseases. The NLRP3 inflammasome is the most characterized inflammasome in terms of the diverse stimuli that are known to activate it. Activation of the NLRP3 inflammasome requires assembly of NLRP3 and caspase-1 (casp-1) bridged together through the adaptor protein ASC, wherein casp-1 undergoes autoproteolytic processing. Subsequently, active casp-1 cleaves precursor forms of cytokines interleukin (IL)–1β and IL-18, which can then be secreted (Man and Kanneganti, 2015; Hamilton et al., 2017). Casp-1 also cleaves gasdermin D (GSDMD), making its N-terminal pore-forming domain active, leading to cell rupture (Kayagaki et al., 2015; Shi et al., 2015). Distinct exogenous, endogenous, and environmental stimuli are known to activate the NLRP3 inflammasome, implying that these stimuli do not bind NLRP3 directly but likely converge on shared upstream pathways. The mechanistic details of NLRP3 activation remain ambiguous.

 Lipids are known to carry out diverse functions within cells, including being a major component of cell membranes, and as signaling messengers. Cholesterol is an essential lipid in mammalian cell membranes aiding varied functions, the most fundamental of which are membrane integrity and fluidity (Maxfield and Tabas, 2005). Levels of cholesterol in the cell are maintained through de novo synthesis in the ER, and uptake of low-density lipoproteins (LDLs) derived from dietary cholesterol. Excess free cholesterol can be toxic to cells; thus, sterol homeostasis needs to be integrated by a combination of cholesterol uptake, biosynthesis, and efflux programs. At the subcellular level, cholesterol follows an intricate pathway in cells (Ikonen, 2008). Exogenously obtained LDL bound to LDL receptor is internalized at the plasma membrane (PM) and is transported through the endocytic pathway to the late endosomes–lysosomes, where cholesterol esters within the LDL core are hydrolyzed by acid lipases. Unesterified or free cholesterol translocates through the lysosomal cholesterol transporter Niemann-Pick C1 (NPC1) to other cellular sites such as the PM and the ER. In the ER, cholesterol can be reesterified, permitting cytoplasmic storage in the form of lipid droplets.

Until recently, cholesterol has mostly been accepted to have an influence on immunity during pathological conditions such as in atherosclerosis (Fessler, 2016). However, evidence suggests that homeostatic lipid metabolism and trafficking directly regulate the inflammatory pathways in macrophages. For example, defective lipid trafficking in the absence of NPC1 leads to the lysosomal storage disorder Niemann-Pick disease (Platt et al., 2012). Mutations in the cholesterol efflux transporter, ABCA1, give rise to signs and symptoms of Tangier disease (Fasano et al., 2012). Similarly, perturbations in lipid metabolism contribute to several human pathologies including cardiovascular, obesity, and neurodegenerative diseases (Maxfield and Tabas, 2005). In addition to contributing to the pathogenesis of several diseases, cholesterol is also exploited by pathogens for their entry and proliferation within host cells. Several pathogens that lack the capacity for de novo sterol synthesis use cholesterol for their survival and replication by either increasing host lipid biosynthesis or redirecting cholesterol transport pathways (Coppens et al., 2000; Lauer et al., 2000; Carabeo et al., 2003; Kaul et al., 2004; Ilnytska et al., 2013). These studies suggest that reducing lipid synthesis may serve to limit nutrients available to pathogens, thus benefitting host cells. Conversely, host cells need lipids for mounting a robust immune response to infection through conserved pattern recognition receptors (Castrillo et al., 2003; York et al., 2015). Together, these studies lead to the hypothesis that lipid homeostasis is critical for an effective inflammatory response with implications for homeostatic lipid trafficking in both infectious and inflammatory diseases. Whether perturbations in homeostatic cholesterol trafficking pathway impact inflammasome activation remains unknown.

 In this study, by using pharmacological and genetic tools, we demonstrate that selective perturbation of the cellular cholesterol trafficking in macrophages ablates inflammasome activation. Mechanistically, perturbed sterol trafficking in Npc1 deficiency leads to two distinct effects: altered PM cholesterol levels resulted in inhibition of the AKT–mTOR pathway, while reduced cholesterol trafficking to the ER blunted NLRP3 inflammasome assembly. Accordingly, acute cholesterol depletion in the ER by statins decreased IL-1β secretion, which could be restored by supplementing with exogenous cholesterol. Our findings thus implicate sterol synthesis and distribution as critical factors influencing the activation of the inflammasome, thereby coupling lipid homeostasis to innate immune signaling.
---

Activation of inflammasome is characterized by cell swelling and subsequent osmotic lysis in the form of pyroptotic cell death following GSDMD cleavage (Kayagaki et al., 2015; Shi et al., 2015). In control cells primed with either LPS or Pam3, stimulation with ATP caused pyroptotic cell death. However, upon exposure to U18666a, cell death, and thus the secreted levels of cytosol-localized cell death marker, lactate dehydrogenase (LDH), were markedly diminished (Fig. 1, G–I). Overall these results demonstrate reduced casp-1 activation, IL-1β and IL-18 secretion, and decreased pyroptosis when cellular cholesterol trafficking is inhibited.
 ...
 We next performed experiments to determine the role of priming in our assays.  
Activation of the NLRP3 inflammasome requires two signals. 
(1) The first signal, which potentiates the NLRP3 inflammasome, is provided upon Toll-like receptor (TLR) ligation and results in the synthesis of pro–IL-1β and up-regulation of NLRP3 expression (Anand et al., 2011a). This first signal (also known as the priming step) is dependent on the transcription factor NF-κB (Bauernfeind et al., 2009; Hornung and Latz, 2010).
(2)  The second signal, which is more robustly provided by purinergic receptor P2X7 agonist ATP, results in the assembly of NLRP3, casp-1, and adaptor protein ASC to form a multiprotein complex (Pelegrin and Surprenant, 2006).
...
 A diverse array of signals is known to activate the NLRP3 inflammasome. 

In contrast, ligands that activate the NLRC4 and AIM2 inflammasomes are well defined. 

The NLRC4 inflammasome is triggered by the recognition of cytosolic flagellin and components of the bacterial secretion system by upstream NAIP family members (Amer et al., 2006; Kofoed and Vance, 2011; Zhao et al., 2011).
 AIM2 inflammasome, in contrast, is activated by recognition of double-stranded DNA of ∼80 bp in length (Jin et al., 2012).

Cholesterol supplementation restores inflammasome activation

Variation in the levels of cholesterol and other lipids within the cell are sensed by master transcriptional regulators SREBP1 and SREBP2, which, respectively regulate fatty acid and cholesterol biosynthesis (Horton et al., 2002). A feedback mechanism tightly regulates cholesterol de novo biosynthesis to maintain homeostatic levels. Cholesterol depletion triggers ER resident sterol cargo protein SCAP to escort SREBP2 to the Golgi, where the precursor is cleaved to activate cholesterogenic genes. By contrast, in cholesterol-replete cells, conformational change in SCAP promotes insulin-induced gene–mediated retention of SREBP2 in the ER.

Picture:
---
F) Homeostatic cellular cholesterol trafficking to the ER is required for optimal inflammasome activation. Exogenously obtained LDL bound to LDL-R is endocytosed at the PM. Cholesterol efflux from the late endosome–lysosome compartment to other cellular sites such as the PM and the ER is dependent on NPC1. (G) Blockade of the cholesterol transporter NPC1 leads to lysosomal cholesterol accumulation and a subsequent decrease in cholesterol pool in both the ER and the PM. This in turn leads to decreased phosphorylation of AKT and mTOR following TLR ligation, causing a reduction in SREBP2-dependent lipogenesis. Additionally, perturbation in the ER cholesterol levels leads to decreased association of NLRP3 and ASC, resulting in reduced active casp-1 levels and IL-1β secretion. Furthermore, inhibition of ER cholesterol levels by statins similarly blunts NLRP3 inflammasome activation. LDL-R, low-density lipoprotein receptor; mTOR, mammalian target of rapamycin; SREBP2, sterol regulatory element binding protein 2.