Reactive microglia enhance the transmission of exosomal alpha-synuclein via toll-like receptor 2

Brain ◽  
2021 ◽  
Author(s):  
Yun Xia ◽  
Guoxin Zhang ◽  
Liang Kou ◽  
Sijia Yin ◽  
Chao Han ◽  
...  

Abstract Increasing evidence suggests that microglial activation is strongly linked to the initiation and progression of Parkinson’s disease (PD). Cell-to-cell propagation of α-synuclein (α-syn) pathology is a highlighted feature of PD, and the focus of such research has been primarily on neurons. However, recent studies as well as the data contained herein suggest that microglia, the primary phagocytes in the brain, play a direct role in the spread of α-syn pathology. Recent data revealed that plasma exosomes derived from PD patients (PD-EXO) carry pathological α-syn and target microglia preferentially. Hence, PD-EXO is likely a key tool for investigating the role of microglia in α-syn transmission. We showed that intrastriatal injection of PD-EXO resulted in the propagation of exosomal α-syn from microglia to neurons following microglia activation. Toll-like receptor 2 (TLR2) in microglia was activated by exosomal α-syn and acted as a crucial mediator of PD-EXO-induced microglial activation. Additionally, partial microglia depletion resulted in a significant decrease of exogenous α-syn in the substantia nigra (SN). Furthermore, exosomal α-syn internalization was initiated by binding to TLR2 of microglia. Excessive α-syn phagocytosis may induce the inflammatory responses of microglia and provide the seed for microglia-to-neuron transmission. Consistently, TLR2 silencing in microglia mitigated α-syn pathology in vivo. Overall, the present data support the idea that the interaction of exosomal α-syn and microglial TLR2 contribute to excessive α-syn phagocytosis and microglial activation, which lead to the further propagation and spread of α-syn pathology, thereby highlighting the pivotal roles of reactive microglia in α-syn transmission.

Shock ◽  
2000 ◽  
Vol 14 (3) ◽  
pp. 361-365 ◽  
Author(s):  
Shubing Liu ◽  
Neil A. Salyapongse ◽  
David A. Geller ◽  
Yoram Vodovotz ◽  
Timothy R. Billiar

2013 ◽  
Vol 81 (9) ◽  
pp. 3479-3489 ◽  
Author(s):  
Robert B. Clark ◽  
Jorge L. Cervantes ◽  
Mark W. Maciejewski ◽  
Vahid Farrokhi ◽  
Reza Nemati ◽  
...  

ABSTRACTThe total cellular lipids ofPorphyromas gingivalis, a known periodontal pathogen, were previously shown to promote dendritic cell activation and inhibition of osteoblasts through engagement of Toll-like receptor 2 (TLR2). The purpose of the present investigation was to fractionate all lipids ofP. gingivalisand define which lipid classes account for the TLR2 engagement, based on bothin vitrohuman cell assays andin vivostudies in mice. Specific serine-containing lipids ofP. gingivalis, called lipid 654 and lipid 430, were identified in specific high-performance liquid chromatography fractions as the TLR2-activating lipids. The structures of these lipids were defined using tandem mass spectrometry and nuclear magnetic resonance methods.In vitro, both lipid 654 and lipid 430 activated TLR2-expressing HEK cells, and this activation was inhibited by anti-TLR2 antibody. In contrast, TLR4-expressing HEK cells failed to be activated by either lipid 654 or lipid 430. Wild-type (WT) or TLR2-deficient (TLR2−/−) mice were injected with either lipid 654 or lipid 430, and the effects on serum levels of the chemokine CCL2 were measured 4 h later. Administration of either lipid 654 or lipid 430 to WT mice resulted in a significant increase in serum CCL2 levels; in contrast, the administration of lipid 654 or lipid 430 to TLR2−/−mice resulted in no increase in serum CCL2. These results thus identify a new class of TLR2 ligands that are produced byP. gingivalisthat likely play a significant role in mediating inflammatory responses both at periodontal sites and, potentially, in other tissues where these lipids might accumulate.


2015 ◽  
Vol 5 (1) ◽  
Author(s):  
Fu Gao ◽  
Chaoxiong Zhang ◽  
Chuanfeng Zhou ◽  
Weimin Sun ◽  
Xin Liu ◽  
...  

2019 ◽  
Author(s):  
Shen-Hsing Hsu ◽  
Ming-Yang Chang ◽  
Yi-Ching Ko ◽  
Li-Feng Chou ◽  
Ya-Chung Tian ◽  
...  

AbstractLeptospirosis is an overlooked zoonotic disease caused by pathogenic Leptospira. The kidney is the major organ infected by Leptospira which causes tubulointerstitial nephritis. Leptospira outer membrane components contain several virulence factors that play important roles in the pathogenesis of leptospirosis. Among them, OmpA-like protein Loa22 is essential for leptospiral virulence. However, the pathogenic mechanisms of tubulointerstitial nephritis involving this virulence factor are still unclear and need further investigation. In this study, pull-down assays suggested that Toll-like receptor 2 (TLR2) proteins interacted with Loa22 from Leptospira outer membrane extractions. Combination of Atomic force microscopy (AFM) and side-directed mutagenesis suggested that Loa22 exhibited high affinity for Leptospira peptidoglycan (LPGN) and the residues of Loa22 were involved in LPGN interaction. Mutation of two key residues within the OmpA-like domain of Loa22, Asp122 and Arg143, significantly attenuated their relative affinities for LPGN indicating that these two residues were responsible for LPGN binding. Thus Loa22 OmpA domain was responsible for interacting with LPGN and the two indicated residues may participate in binding to LPGN. Recombinant Loa22 (rLoa22) protein was further complexed with LPGN and incubated with HEK293-TLR2 cells to monitor inflammatory responses. Inflammatory responses were provoked by rLoa22-LPGN complexes, but not rLoa22 alone, involved CXCL8/IL8, hCCL2/MCP-1, and hTNF-α activation. Confocal microscopy further identified the co-localization of Loa22-LPGN complexes and TLR2 receptors on HEK293-TLR2 cell surface. The affinity between Loa22-LPGN complexes and TLR2 were further confirmed and measured by AFM and ELISA. Downstream signals from TLR2 including p38, ERK, and JNK were observed by western blotting induced by Loa22-LPGN complexes. In summary, this study identified LPGN in leptospira mediates interactions between Loa22 and TLR2 and induces downstream signals to trigger inflammatory responses. Interactions between Loa22-LPGN-TLR2 reveal a novel binding mechanism for the innate immune system and infection induced by leptospira.Author summaryLeptospirosis is one of the most overlooked zoonotic diseases caused by pathogenic Leptospira in warm and humid regions worldwide. With the infection by Leptospira, many organs are invaded and can result in multiple-organ failure (Weil’s syndrome). Kidney is the major organ infected by pathogenic Leptospira, which would manifest as tubulointerstitial nephritis. In this study, we focused on the outer membrane lipoprotein Loa22 (Leptospiral OmpA-like domain 22) from pathogenic Leptospira which triggers inflammatory responses on renal tubular cell. Protein domain prediction indicated that Loa22 contains an important domain termed OmpA-like domain and the function of this domain is peptidoglycan (PGN) binding. From sequence alignments of Loa22 with other OmpA proteins, two important amino acids, Asp122 and Arg143, were found to be highly conserved. The role of the two conserved residues in AbOmpA (OmpA protein in A. baumannii) and Pal (peptidoglycan-associated lipoprotein in E. coli) proteins are important for PGN binding. These two residues in Loa22 were altered by site-directed mutagenesis to obtain D122A and R143A variants. In pull-down and AFM analysis, the binding capacities of Loa22 variants to Leptospira PGN (LPGN) were significantly decreased as compared to rLoa22WT, indicating that the two residues are involved in LPGN binding. Furthermore, recombinant Loa22 and its variants in the absence or presence of LPGN, were incubated with HEK293-TLR2 cells, to confirm the role of LPGN in triggering inflammatory responses involving CXCL8/IL8, hCCL2/MCP-1, and hTNF-α. These factors are involved in downstream signaling of inflammatory responses through Toll-like receptor 2 (TLR2). In addition, confocal microscopy was employed to observe the co-localization of Loa22-LPGN complexes and TLR2 receptors on HEK293-TLR2 cell surfaces. Finally, the interaction forces between rTLR2 and rLoa22-LPGN complexes were measured by AFM and ELISA to conclude the necessary role of LPGN in rLoa22-TLR2 complex formation. In summary, these results demonstrate that the interaction of Loa22 protein with the important cell wall component, PGN, concomitantly triggered inflammatory responses of host cells through interaction with TLR2.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ying Tang ◽  
Mengchun Zhou ◽  
Rongrong Huang ◽  
Ling Shen ◽  
Li Yang ◽  
...  

Abstract Background Astrocytes participate in innate inflammatory responses within the mammalian central nervous system (CNS). HECT domain E3 ubiquitin protein ligase 1 (HECTD1) functions during microglial activation, suggesting a connection with neuroinflammation. However, the potential role of HECTD1 in astrocytes remains largely unknown. Results Here, we demonstrated that HECTD1 was upregulated in primary mouse astrocytes after 100 ng/ml lipopolysaccharide (LPS) treatment. Genetic knockdown of HECTD1 in vitro or astrocyte-specific knockdown of HECTD1 in vivo suppressed LPS-induced astrocyte activation, whereas overexpression of HECTD1 in vitro facilitated LPS-induced astrocyte activation. Mechanistically, we established that LPS activated σ-1R-JNK/p38 pathway, and σ-1R antagonist BD1047, JNK inhibitor SP600125, or p38 inhibitor SB203580 reversed LPS-induced expression of HECTD1, thus restored LPS-induced astrocyte activation. In addition, FOXJ2 functioned as a transcription factor of HECTD1, and pretreatment of primary mouse astrocytes with BD1047, SB203580, and SP600125 significantly inhibited LPS-mediated translocation of FOXJ2 into the nucleus. Conclusions Overall, our present findings suggest that HECTD1 participates in LPS-induced astrocyte activation by activation of σ-1R-JNK/p38-FOXJ2 pathway and provide a potential therapeutic strategy for neuroinflammation induced by LPS or any other neuroinflammatory disorders.


Vaccines ◽  
2021 ◽  
Vol 9 (7) ◽  
pp. 692
Author(s):  
Giulia Franzoni ◽  
Antonio Anfossi ◽  
Chiara Grazia De Ciucis ◽  
Samanta Mecocci ◽  
Tania Carta ◽  
...  

Toll-like receptor 2 (TLR2) ligands are attracting increasing attention as prophylactic and immunotherapeutic agents against pathogens and tumors. We previously observed that a synthetic diacylated lipopeptide based on a surface protein of Mycoplasma agalactiae (Mag-Pam2Cys) strongly activated innate immune cells, including porcine monocyte-derived macrophages (moMΦ). In this study, we utilized confocal microscopy, flow cytometry, multiplex cytokine ELISA, and RT-qPCR to conduct a comprehensive analysis of the effects of scalar doses of Mag-Pam2Cys on porcine moMΦ. We observed enhanced expression of activation markers (MHC class I, MHC class II DR, CD25), increased phagocytotic activity, and release of IL-12 and proinflammatory cytokines. Mag-Pam2Cys also upregulated the gene expression of several IFN-α subtypes, p65, NOS2, and molecules with antimicrobial activities (CD14, beta defensin 1). Overall, our data showed that Mag-Pam2Cys polarized porcine macrophages towards a proinflammatory antimicrobial phenotype. However, Mag-Pam2Cys downregulated the expression of IFN-α3, six TLRs (TLR3, -4, -5, -7, -8, -9), and did not interfere with macrophage polarization induced by the immunosuppressive IL-10, suggesting that the inflammatory activity evoked by Mag-Pam2Cys could be regulated to avoid potentially harmful consequences. We hope that our in vitro results will lay the foundation for the further evaluation of this diacylated lipopeptide as an immunopotentiator in vivo.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Shinjini Chakraborty ◽  
Veronika Eva Winkelmann ◽  
Sonja Braumüller ◽  
Annette Palmer ◽  
Anke Schultze ◽  
...  

AbstractSingular blockade of C5a in experimental models of sepsis is known to confer protection by rescuing lethality and decreasing pro-inflammatory responses. However, the role of inhibiting C5a has not been evaluated in the context of sterile systemic inflammatory responses, like polytrauma and hemorrhagic shock (PT + HS). In our presented study, a novel and highly specific C5a L-aptamer, NoxD21, was used to block C5a activity in an experimental murine model of PT + HS. The aim of the study was to assess early modulation of inflammatory responses and lung damage 4 h after PT + HS induction. NoxD21-treated PT + HS mice displayed greater polymorphonuclear cell recruitment in the lung, increased pro-inflammatory cytokine levels in the bronchoalveolar lavage fluids (BALF) and reduced myeloperoxidase levels within the lung tissue. An in vitro model of the alveolar-capillary barrier was established to confirm these in vivo observations. Treatment with a polytrauma cocktail induced barrier damage only after 16 h, and NoxD21 treatment in vitro did not rescue this effect. Furthermore, to test the exact role of both the cognate receptors of C5a (C5aR1 and C5aR2), experimental PT + HS was induced in C5aR1 knockout (C5aR1 KO) and C5aR2 KO mice. Following 4 h of PT + HS, C5aR2 KO mice had significantly reduced IL-6 and IL-17 levels in the BALF without significant lung damage, and both, C5aR1 KO and C5aR2 KO PT + HS animals displayed reduced MPO levels within the lungs. In conclusion, the C5aR2 could be a putative driver of early local inflammatory responses in the lung after PT + HS.


Sign in / Sign up

Export Citation Format

Share Document