Academic literature on the topic 'WASp, pDCs, Type-I-IFN, autoimmunity'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'WASp, pDCs, Type-I-IFN, autoimmunity.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "WASp, pDCs, Type-I-IFN, autoimmunity"

1

Prete, Francesca, Marco Catucci, Mayrel Labrada, Stefania Gobessi, Maria Carmina Castiello, Elisa Bonomi, Alessandro Aiuti, et al. "Wiskott-Aldrich syndrome protein–mediated actin dynamics control type-I interferon production in plasmacytoid dendritic cells." Journal of Experimental Medicine 210, no. 2 (January 21, 2013): 355–74. http://dx.doi.org/10.1084/jem.20120363.

Full text
Abstract:
Mutations in Wiskott-Aldrich syndrome (WAS) protein (WASp), a regulator of actin dynamics in hematopoietic cells, cause WAS, an X-linked primary immunodeficiency characterized by recurrent infections and a marked predisposition to develop autoimmune disorders. The mechanisms that link actin alterations to the autoimmune phenotype are still poorly understood. We show that chronic activation of plasmacytoid dendritic cells (pDCs) and elevated type-I interferon (IFN) levels play a role in WAS autoimmunity. WAS patients display increased expression of type-I IFN genes and their inducible targets, alteration in pDCs numbers, and hyperresponsiveness to TLR9. Importantly, ablating IFN-I signaling in WASp null mice rescued chronic activation of conventional DCs, splenomegaly, and colitis. Using WASp-deficient mice, we demonstrated that WASp null pDCs are intrinsically more responsive to multimeric agonist of TLR9 and constitutively secrete type-I IFN but become progressively tolerant to further stimulation. By acute silencing of WASp and actin inhibitors, we show that WASp-mediated actin polymerization controls intracellular trafficking and compartmentalization of TLR9 ligands in pDCs restraining exaggerated activation of the TLR9–IFN-α pathway. Together, these data highlight the role of actin dynamics in pDC innate functions and imply the pDC–IFN-α axis as a player in the onset of autoimmune phenomena in WAS disease.
APA, Harvard, Vancouver, ISO, and other styles
2

Schmitt, Heike, Sabrina Sell, Julia Koch, Martina Seefried, Sophia Sonnewald, Christoph Daniel, Thomas H. Winkler, and Lars Nitschke. "Siglec-H protects from virus-triggered severe systemic autoimmunity." Journal of Experimental Medicine 213, no. 8 (July 4, 2016): 1627–44. http://dx.doi.org/10.1084/jem.20160189.

Full text
Abstract:
It is controversial whether virus infections can contribute to the development of autoimmune diseases. Type I interferons (IFNs) are critical antiviral cytokines during virus infections and have also been implicated in the pathogenesis of systemic lupus erythematosus. Type I IFN is mainly produced by plasmacytoid dendritic cells (pDCs). The secretion of type I IFN of pDCs is modulated by Siglec-H, a DAP12-associated receptor on pDCs. In this study, we show that Siglec-H–deficient pDCs produce more of the type I IFN, IFN-α, in vitro and that Siglec-H knockout (KO) mice produce more IFN-α after murine cytomegalovirus (mCMV) infection in vivo. This did not impact control of viral replication. Remarkably, several weeks after a single mCMV infection, Siglec-H KO mice developed a severe form of systemic lupus–like autoimmune disease with strong kidney nephritis. In contrast, uninfected aging Siglec-H KO mice developed a mild form of systemic autoimmunity. The induction of systemic autoimmune disease after virus infection in Siglec-H KO mice was accompanied by a type I IFN signature and fully dependent on type I IFN signaling. These results show that Siglec-H normally serves as a modulator of type I IFN responses after infection with a persistent virus and thereby prevents induction of autoimmune disease.
APA, Harvard, Vancouver, ISO, and other styles
3

Karnell, Jodi L., Yanping Wu, Nanette Mittereder, Michael A. Smith, Michele Gunsior, Li Yan, Kerry A. Casey, et al. "Depleting plasmacytoid dendritic cells reduces local type I interferon responses and disease activity in patients with cutaneous lupus." Science Translational Medicine 13, no. 595 (May 26, 2021): eabf8442. http://dx.doi.org/10.1126/scitranslmed.abf8442.

Full text
Abstract:
Plasmacytoid dendritic cells (pDCs) not only are specialized in their capacity to secrete large amounts of type I interferon (IFN) but also serve to enable both innate and adaptive immune responses through expression of additional proinflammatory cytokines, chemokines, and costimulatory molecules. Persistent activation of pDCs has been demonstrated in a number of autoimmune diseases. To evaluate the potential benefit of depleting pDCs in autoimmunity, a monoclonal antibody targeting the pDC-specific marker immunoglobulin-like transcript 7 was generated. This antibody, known as VIB7734, which was engineered for enhanced effector function, mediated rapid and potent depletion of pDCs through antibody-dependent cellular cytotoxicity. In cynomolgus monkeys, treatment with VIB7734 reduced pDCs in blood below the lower limit of normal by day 1 after the first dose. In two phase 1 studies in patients with autoimmune diseases, VIB7734 demonstrated an acceptable safety profile, comparable to that of placebo. In individuals with cutaneous lupus, VIB7734 profoundly reduced both circulating and tissue-resident pDCs, with a 97.6% median reduction in skin pDCs at study day 85 in VIB7734-treated participants. Reductions in pDCs in the skin correlated with a decrease in local type I IFN activity as well as improvements in clinical disease activity. Biomarker analysis suggests that responsiveness to pDC depletion therapy may be greater among individuals with high baseline type I IFN activity, supporting a central role for pDCs in type I IFN production in autoimmunity and further development of VIB7734 in IFN-associated diseases.
APA, Harvard, Vancouver, ISO, and other styles
4

Cao, Wei, Jeremy Di Domizio, Stephanie Dorta-Estramera, Mihai Gagea, Ping Li, Liqi Bi, and Michel Gilliet. "Nucleic acid-containing amyloid fibrils potently induce Type I interferon and stimulate systemic autoimmunity (P4075)." Journal of Immunology 190, no. 1_Supplement (May 1, 2013): 127.9. http://dx.doi.org/10.4049/jimmunol.190.supp.127.9.

Full text
Abstract:
Abstract The immunopathophysiologic development of systemic autoimmunity involves numerous factors through complex mechanisms. In systemic lupus erythematosus, type I IFN (IFNI) produced by plasmacytoid dendritic cells (pDCs) critically promotes the autoimmunity through its pleiotropic effects on immune cells. However, the host-derived factors that enable abnormal IFN-I production and initial immune tolerance breakdown are largely unknown. Amyloid precursor proteins can form amyloid fibrils in the presence of nucleic acids. Here we report that nucleic acid-containing amyloid fibrils can potently activate pDCs and enable IFN-I production in response to self-DNA, self-RNA, and dead cell debris. pDCs can take up DNA-containing amyloid fibrils, which are retained in the early endosomes to activate TLR9, leading to high IFNα/β production. In mice treated with DNA-containing amyloid fibrils, a rapid IFN response correlated with pDC infiltration and activation. Immunization of nonautoimmune mice with DNA-containing amyloid fibrils induced antinuclear serology against a panel of selfantigens. The mice exhibited positive proteinuria and deposited antibodies in their kidneys. Intriguingly, pDC depletion obstructed IFN-I response and selectively abolished autoantibody generation. Our study reveals an innate immune function of nucleic acid-containing amyloid fibrils and provides a potential link between compromised protein homeostasis and autoimmunity via a pDC-IFN axis.
APA, Harvard, Vancouver, ISO, and other styles
5

Barrat, Franck J., and Lishan Su. "A pathogenic role of plasmacytoid dendritic cells in autoimmunity and chronic viral infection." Journal of Experimental Medicine 216, no. 9 (August 16, 2019): 1974–85. http://dx.doi.org/10.1084/jem.20181359.

Full text
Abstract:
Following the discovery of plasmacytoid dendritic cells (pDCs) and of their extraordinary ability to produce type I IFNs (IFN-I) in response to TLR7 and TLR9 stimulation, it is assumed that their main function is to participate in the antiviral response. There is increasing evidence suggesting that pDCs and/or IFN-I can also have a detrimental role in a number of inflammatory and autoimmune diseases, in the context of chronic viral infections and in cancers. Whether these cells should be targeted in patients and how much of their biology is connected to IFN-I production remains unclear and is discussed here.
APA, Harvard, Vancouver, ISO, and other styles
6

Lorant, Alina K., and Adam Lacy-Hulbert. "Integrin αvβ3 limits TLR signaling in plasmacytoid dendritic cells to prevent autoimmunity." Journal of Immunology 206, no. 1_Supplement (May 1, 2021): 60.09. http://dx.doi.org/10.4049/jimmunol.206.supp.60.09.

Full text
Abstract:
Abstract SLE is a chronic, systemic autoimmune disease affecting millions of people worldwide. SLE is caused by a breakdown of tolerance to self-derived nucleic acids (NA) and characterized by the presence of autoantibodies to DNA/RNA. Plasmacytoid dendritic cells (pDCs) are highly tuned to respond to NA and express a range of receptors for RNA and DNA, including TLRs 7 and 9. Following TLR engagement, pDCs produce high levels of type I interferon (IFN-I), which promotes loss of tolerance, implicating pDCs in development of autoimmunity. However, relatively little is known about how innate immune signaling is regulated in pDCs, and particularly how they distinguish self-derived from foreign NA. TLRs 7 and 9 are both localized in endosomal compartments, and their NA ligands must be internalized and processed before they can bind. One source of these NA is dying cells and loss of their receptors is associated with SLE. We studied the role of an apoptotic cell receptor, Integrin αvβ3, in TLR signaling of pDCs and found that αvβ3 limits IFN-I production. αvβ3-knockout pDCs show increased TLR signaling and produce more IFN-I and inflammatory cytokines when stimulated in culture. In mice without αvβ3, there is a systemic increase in circulating ISGs and IFN-I after TLR7 stimulation. This corresponds with an increase in pDC activation and a decrease in marginal zone B cells, indicating elevated IFN-I production. These data indicate αvβ3 has a pDC-intrinsic role in limiting TLR signaling, which we propose is through activation of non-canonical autophagy. Based on these data, we hypothesize that co-receptors involved in capture and internalization of nucleic acids provide contextual ‘cues’ to regulate TLR signaling and prevent autoimmunity.
APA, Harvard, Vancouver, ISO, and other styles
7

Bencze, Dóra, Tünde Fekete, and Kitti Pázmándi. "Type I Interferon Production of Plasmacytoid Dendritic Cells under Control." International Journal of Molecular Sciences 22, no. 8 (April 18, 2021): 4190. http://dx.doi.org/10.3390/ijms22084190.

Full text
Abstract:
One of the most powerful and multifaceted cytokines produced by immune cells are type I interferons (IFNs), the basal secretion of which contributes to the maintenance of immune homeostasis, while their activation-induced production is essential to effective immune responses. Although, each cell is capable of producing type I IFNs, plasmacytoid dendritic cells (pDCs) possess a unique ability to rapidly produce large amounts of them. Importantly, type I IFNs have a prominent role in the pathomechanism of various pDC-associated diseases. Deficiency in type I IFN production increases the risk of more severe viral infections and the development of certain allergic reactions, and supports tumor resistance; nevertheless, its overproduction promotes autoimmune reactions. Therefore, the tight regulation of type I IFN responses of pDCs is essential to maintain an adequate level of immune response without causing adverse effects. Here, our goal was to summarize those endogenous factors that can influence the type I IFN responses of pDCs, and thus might serve as possible therapeutic targets in pDC-associated diseases. Furthermore, we briefly discuss the current therapeutic approaches targeting the pDC-type I IFN axis in viral infections, cancer, autoimmunity, and allergy, together with their limitations defined by the Janus-faced nature of pDC-derived type I IFNs.
APA, Harvard, Vancouver, ISO, and other styles
8

Lorant, Alina K., and Adam Lacy-Hulbert. "Integrin αv limits TLR signaling in plasmacytoid dendritic cells." Journal of Immunology 204, no. 1_Supplement (May 1, 2020): 226.3. http://dx.doi.org/10.4049/jimmunol.204.supp.226.3.

Full text
Abstract:
Abstract SLE is a chronic, systemic autoimmune disease affecting millions of people worldwide. SLE is caused by a breakdown of tolerance to self-derived nucleic acids (NA) and characterized by the presence of autoantibodies to DNA/RNA. Plasmacytoid dendritic cells (pDCs) are highly tuned to respond to NA and express a range of receptors for RNA and DNA, including TLRs 7 and 9. Following TLR engagement, pDCs produce high levels of type I interferon (IFN-I), which promotes loss of tolerance, implicating pDCs in development of autoimmunity. However, relatively little is known about how innate immune signaling is regulated in pDCs, and particularly how they distinguish self-derived from foreign NA. TLRs 7 and 9 are both localized in endosomal compartments, and their NA ligands must be internalized and processed before they can bind. One source of these NA is dying cells and loss of their receptors is associated with SLE. We studied the role of an apoptotic cell receptor, Integrin αv, in TLR signaling of pDCs and found that αv limits IFN-I production. In mice without αv, there is a systemic increase in IFN-I and inflammatory cytokines after TLR7 stimulation. This corresponds with an increase in pDC activation and the number of IFN-I-producing pDCs in the spleen. αv-knockout pDCs show increased TLR signaling and produce more IFN-I and inflammatory cytokines when stimulated in culture. These data indicate αv has a pDC-intrinsic role in limiting TLR signaling, which we propose is through activation of non-canonical autophagy. Based on these data, we hypothesize that co-receptors involved in capture and internalization of nucleic acids provide contextual ‘cues’ to regulate TLR signaling and prevent autoimmunity.
APA, Harvard, Vancouver, ISO, and other styles
9

Lande, Roberto, Anna Mennella, Raffaella Palazzo, Immacolata Pietraforte, Katia Stefanantoni, Nicoletta Iannace, Alessia Butera, et al. "Anti-CXCL4 Antibody Reactivity Is Present in Systemic Sclerosis (SSc) and Correlates with the SSc Type I Interferon Signature." International Journal of Molecular Sciences 21, no. 14 (July 19, 2020): 5102. http://dx.doi.org/10.3390/ijms21145102.

Full text
Abstract:
Systemic sclerosis (SSc) is characterized by skin/internal organ fibrosis, vasculopathy and autoimmunity. Chemokine (C-X-C motif) ligand 4 (CXCL4) is an SSc biomarker, predicting unfavorable prognosis and lung fibrosis. CXCL4 binds DNA/RNA and favors interferon (IFN)-α production by plasmacytoid dendritic cells (pDCs), contributing to the type I IFN (IFN-I) signature in SSc patients. However, whether CXCL4 is an autoantigen in SSc is unknown. Here, we show that at least half of SSc patients show consistent antibody reactivity to CXCL4. T-cell proliferation to CXCL4, tested in a limited number of patients, correlates with anti-CXCL4 antibody reactivity. Antibodies to CXCL4 mostly correlate with circulating IFN-α levels and are significantly higher in patients with lung fibrosis in two independent SSc cohorts. Antibodies to CXCL4 implement the CXCL4–DNA complex’s effect on IFN-α production by pDCs; CXCL4–DNA/RNA complexes stimulate purified human B-cells to become antibody-secreting plasma cells in vitro. These data indicate that CXCL4 is indeed an autoantigen in SSc and suggest that CXCL4, and CXCL4-specific autoantibodies, can fuel a harmful loop: CXCL4–DNA/RNA complexes induce IFN-α in pDCs and direct B-cell stimulation, including the secretion of anti-CXCL4 antibodies. Anti-CXCL4 antibodies may further increase pDC stimulation and IFN-α release in vivo, creating a vicious cycle which sustains the SSc IFN-I signature and general inflammation.
APA, Harvard, Vancouver, ISO, and other styles
10

Babcock, Rachel Lauren, Haiyan S. Li, Yifan Zhou, Taylor T. Chrisikos, and Stephanie S. Watowich. "Regulation of Id2 gene expression in plasmacytoid dendritic cells." Journal of Immunology 204, no. 1_Supplement (May 1, 2020): 229.3. http://dx.doi.org/10.4049/jimmunol.204.supp.229.3.

Full text
Abstract:
Abstract Plasmacytoid dendritic cells (pDCs) are specialized type I interferon (IFN-I) producing cells that mediate anti-viral responses, anti-tumor immunity, and autoimmunity. When exposed to Toll-like receptor 7 (TLR7)- and TLR9-ligands, pDCs mature and produce IFN-Is; pDCs also acquire conventional DC (cDC)-like morphology and features, including the cell surface expression of antigen presentation and co-stimulatory molecules, secretion of additional cytokines, and ability to activate adaptive T cell responses. Yet, the transcriptional pathways regulating TLR-induced pDC maturation are poorly characterized. We found the mRNA and protein expression of the inhibitor of DNA binding protein 2 (Id2) was induced in TLR7/9-stimulated pDCs, and Id2 expression correlated with greater cDC-like features. Id2 normally blocks pDC development by antagonizing the pDC-master regulator, E2-2, while E2-2 suppresses Id2 transcription. Thus, we hypothesize TLR-activated signaling pathways in pDCs control the abundance of transcriptional regulators (e.g., E2-2) at the Id2 promoter as well as the promoter chromatin state, leading to induction of Id2 transcription during pDC maturation. Using genetic knockout mouse models, we found Id2 induction was independent of interferon alpha receptor and nuclear factor kappa beta signaling pathways. Chromatin-immunoprecipitation assays suggested E2-2 abundance at the Id2 promoter decreased following TLR stimulation, while activating histone modifications increased. These data are consistent with transcriptional induction of Id2 upon TLR stimulation. Future work will investigate the effect of additional regulators and TLR-regulated pathways on Id2 induction in TLR7/9-stimulated pDCs.
APA, Harvard, Vancouver, ISO, and other styles

Dissertations / Theses on the topic "WASp, pDCs, Type-I-IFN, autoimmunity"

1

Prete, Francesca. "The role of Wiskott-Aldrich Syndrome protein-mediated actin dynamics in controlling type-I IFN production in plasmacytoid dendritic cells." Doctoral thesis, Università degli studi di Trieste, 2012. http://hdl.handle.net/10077/7867.

Full text
Abstract:
2010/2011
Wiskott-Aldrich syndrome (WAS) is an X-linked primary immunodeficiency characterized by recurrent infections, and a marked predisposition to develop autoimmune phenomena. The disease is caused by mutations in WASp, a key regulator of actin polymerization expressed only in hematopoietic cells. A general impairment of hematopoietic cell functions contributes to the pathogenesis of the disease. Neutrophils, B cells, T cells and DCs deficient for WASp were all shown to have impaired homing ability, a cellular function that strictly depends on spatio-temporal regulation of actin polymerization. WASp null T cells present severe impairment in coupling TCR stimulation to proliferation and fail to organize signaling molecules within the immunological synapse (Badour et al., 2004; Dupre et al., 2002; Sims et al., 2007). B cells intrinsic defects include altered B cell receptor clustering, defective homeostasis of mature B cells and a specific reduction in the expression of the complement receptor (Park et al., 2005; Simon et al., 1992; Westerberg et al., 2001). WASp null DCs fail to assemble podosomes and display late migration from the periphery to lymph nodes (de Noronha et al., 2005). Moreover, WASp expression in DCs is critical to organize the dynamic cytoskeletal changes that facilitate DC-T cell interaction during antigen presentation (Pulecio et al., 2008; Bouma et al., 2011). Together, these cellular alterations provided clues to understand the reduced response to pathogens and the immunodeficiency of WAS patients. However, the mechanisms by which perturbation of actin dynamics promote autoimmune phenomena are less clear. Autoimmune complications occur in 40-72% of children with severe WAS phenotype. The most common autoimmune features that develop in WAS patients include hemolytic anemia, vasculitis, renal disease, and arthritis (Dupuis-Girod et al., 2003; Sullivan et al., 1994; Humblet-baron et al., 2007). Impairment of T and B cell tolerance have been reported in WAS patients and in Was-/- mice, but the exact cellular mechanisms that link loss of WASp function to autoimmunity have not been fully elucidated yet (Becker-Herman et al., 2011; Recher et al., 2012; Marangoni et al., 2007; Maillard et al., 2007; Humblet-Baron et al., 2007). It is increasingly recognized that excessive activation of pDCs and elevated type-I interferon (IFN) levels are pathogenic in several human autoimmune diseases such as SLE, psoriasis, Sjogren’s syndrome. Since WAS autoimmune manifestations partially overlap with those of type-I IFN diseases, we hypothesized that pDCs/IFN-α axis may have a role in WAS-associated autoimmunity. In the first part of results we present the analysis of the pDC compartment in a mouse model of the disease, a mouse knock-out for WASp (WKO mice). We show that pDCs from WKO animals are chronically activated, secrete type-I IFN constitutively and become refractory to further stimulation in vivo. By depleting WASp expression or by interfering with actin dynamics in pDCs we prove that WASp-mediated actin dynamics control the activation of the TLR9/IFN-α pathway in a cell autonomous fashion. Based on the results of previous section, we speculated that WASp may regulate IFN-α production by controlling the correct organization of the endocytic pathway. It is well known, in fact, that type-I IFN production relies on spatio-temporal regulation of TLR9 signaling in the endocytic pathway, at the same time, it is largely established that many steps of the endocytosis are regulated by actin regulatory proteins of the WASp family. Therefore, we proceed presenting a series of experiments exploring the role of WASp in mediating signaling downstream TLR9 activation in pDCs. Tracking of TLR9 agonist in pDCs show that WASp controls cellular architecture and early endosomes size leading to accumulation of large aggregates of TLR9 agonist in WASp-deficient cells. To demonstrate a link between the alteration in the pDCs/IFN-α axis found in WKO mice and the pathogenesis of WAS autoimmunity, we first analyzed the presence in patients of clinical aspects known to be associated with type-I IFN diseases. We report that WAS patients display a moderate type-I interferon signature, indicating an exposure to elevated levels of this cytokine. Finally, we moved back to the mouse model of WAS to start investigating the presence of alterations of innate/adaptive immunity classically attributed to unrestrained type-I IFN production. In line with the well known inducing effects of systemic type-I IFNs, our preliminary data illustrate a generally more activated phenotype of immunocytes in WKO mice. In sum, our work provide 1) the first demonstration of an altered pDCs/IFN-α axis in WAS, 2) the existence of a cell intrinsic mechanism of increased pDCs activation in WASp-null pDC, and 3) a new role for actin in restraining excessive activation of TLR9 in pDCs. These observations add a new layer of complexity to our understanding of the pathophysiology of WAS, and raise important considerations about the treatment of patients with autoimmune phenomena.
XXIV Ciclo
1983
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography