Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lionel B. Ivashkiv is active.

Publication


Featured researches published by Lionel B. Ivashkiv.


Nature Reviews Immunology | 2014

Regulation of type I interferon responses

Lionel B. Ivashkiv; Laura T. Donlin

Type I interferons (IFNs) activate intracellular antimicrobial programmes and influence the development of innate and adaptive immune responses. Canonical type I IFN signalling activates the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) pathway, leading to transcription of IFN-stimulated genes (ISGs). Host, pathogen and environmental factors regulate the responses of cells to this signalling pathway and thus calibrate host defences while limiting tissue damage and preventing autoimmunity. Here, we summarize the signalling and epigenetic mechanisms that regulate type I IFN-induced STAT activation and ISG transcription and translation. These regulatory mechanisms determine the biological outcomes of type I IFN responses and whether pathogens are cleared effectively or chronic infection or autoimmune disease ensues.


Immunity | 2009

Cross-regulation of Signaling Pathways by Interferon-γ: Implications for Immune Responses and Autoimmune Diseases

Xiaoyu Hu; Lionel B. Ivashkiv

Interferon-gamma (IFN-gamma) is an important mediator of immunity and inflammation that utilizes the JAK-STAT signaling pathway to activate the STAT1 transcription factor. Many functions of IFN-gamma have been ascribed to direct STAT1-mediated induction of immune effector genes, but recently it has become clear that key IFN-gamma functions are mediated by cross-regulation of cellular responses to other cytokines and inflammatory factors. Here, we review mechanisms by which IFN-gamma and STAT1 regulate signaling by Toll-like receptors, inflammatory factors, tissue-destructive cytokines, anti-inflammatory cytokines, and cytokines that activate opposing STATs. These signaling mechanisms reveal insights about how IFN-gamma regulates macrophage activation, inflammation, tissue remodeling, and helper and regulatory T cell differentiation and how Th1 and Th17 cell responses are integrated in autoimmune diseases.


Nature Immunology | 2012

Notch–RBP-J signaling regulates the transcription factor IRF8 to promote inflammatory macrophage polarization

Haixia Xu; Jimmy Zhu; Sinead Smith; Julia Foldi; Baohong Zhao; Allen Y. Chung; Hasina Outtz; Jan Kitajewski; Chao Shi; Silvio Weber; Paul Saftig; Yueming Li; Keiko Ozato; Carl P. Blobel; Lionel B. Ivashkiv; Xiaoyu Hu

Emerging concepts suggest that the functional phenotype of macrophages is regulated by transcription factors that define alternative activation states. We found that RBP-J, the main nuclear transducer of signaling via Notch receptors, augmented Toll-like receptor 4 (TLR4)-induced expression of key mediators of classically activated M1 macrophages and thus of innate immune responses to Listeria monocytogenes. Notch–RBP-J signaling controlled expression of the transcription factor IRF8 that induced downstream M1 macrophage–associated genes. RBP-J promoted the synthesis of IRF8 protein by selectively augmenting kinase IRAK2–dependent signaling via TLR4 to the kinase MNK1 and downstream translation-initiation control through eIF4E. Our results define a signaling network in which signaling via Notch–RBP-J and TLRs is integrated at the level of synthesis of IRF8 protein and identify a mechanism by which heterologous signaling pathways can regulate the TLR-induced inflammatory polarization of macrophages.


Immunological Reviews | 2008

Regulation of interferon and Toll-like receptor signaling during macrophage activation by opposing feedforward and feedback inhibition mechanisms.

Xiaoyu Hu; Soumya D. Chakravarty; Lionel B. Ivashkiv

Summary: Activated macrophages and their inflammatory products play a key role in innate immunity and in pathogenesis of autoimmune/inflammatory diseases. Macrophage activation needs to be tightly regulated to rapidly mount responses to infectious challenges but to avoid toxicity associated with excessive activation. Rapid and potent macrophage activation is driven by cytokine‐mediated feedforward loops, while excessive activation is prevented by feedback inhibition. Here we discuss feedforward mechanisms that augment macrophage responses to Toll‐like receptor (TLR) ligands and cytokines that are mediated by signal transducer and activator of transcription 1 (STAT1) and induced by interferon‐γ (IFN‐γ). IFN‐γ also drives full macrophage activation by inactivating feedback inhibitory mechanisms, such as those mediated by interleukin‐10 (IL‐10), and STAT3. Priming of macrophages with IFN‐γ reprograms cellular responses to other cytokines, such as type I IFNs and IL‐10, with a shift toward pro‐inflammatory STAT1‐dominated responses. Similar but partially distinct priming effects are induced by other cytokines that activate STAT1, including type I IFNs and IL‐27. We propose a model whereby opposing feedforward and feedback inhibition loops crossregulate each other to fine tune macrophage activation. In addition, we discuss how dysregulation of the balance between feedforward and feedback inhibitory mechanisms can contribute to the pathogenesis of autoimmune and inflammatory diseases, such as rheumatoid arthritis and systemic lupus erythematosus.


Journal of Leukocyte Biology | 2007

Crosstalk among Jak-STAT, Toll-like receptor, and ITAM-dependent pathways in macrophage activation

Xiaoyu Hu; Janice Chen; Lu Wang; Lionel B. Ivashkiv

Macrophage phenotype and activation are regulated by cytokines that use the Jak‐STAT signaling pathway, microbial recognition receptors that include TLRs, and immunoreceptors that signal via ITAM motifs. The amplitude and qualitative nature of macrophage activation are determined by crosstalk among these signaling pathways. Basal ITAM signaling restrains macrophage responses to TLRs and other activating ligands, whereas strong ITAM signals synergize with the same ligands to activate cells strongly. Similarly, basal ITAM signaling augments IFN signaling and function of receptor activator of NF‐κB, but extensive ITAM activation inhibits Jak‐STAT signaling. Thus, intensity and duration of ITAM signaling determine whether ITAM‐coupled receptors augment or attenuate TLR and Jak‐STAT responses. IFN‐γ synergizes with TLRs in part by suppressing TLR‐induced feedback inhibition, mediated by IL‐10 and Stat3, by a mechanism that depends on glycogen synthase kinase (GSK)3 regulation of AP‐1 and CREB. IFN‐γ suppresses TLR2 and TLR4 induction/activation of AP‐1 by overlapping mechanisms that include regulation of MAPKs, GSK3‐dependent suppression of DNA binding, and decreased Fos and Jun protein expression and stability. IFN‐γ suppression of TLR‐induced activation of AP‐1 and downstream target genes challenges current concepts about the inflammatory role of AP‐1 proteins in macrophage activation and is consistent with a role for AP‐1 in the generation of noninflammatory osteoclasts. Jak‐STAT, TLR, and ITAM pathways are basally active in macrophages and strongly induced during innate responses. Thus, signal transduction crosstalk is regulated in a dynamic manner, which differs under homeostatic and pathologic conditions, and dysregulation of signal transduction crosstalk may contribute to pathogenesis of chronic inflammatory diseases.


Nature Immunology | 2002

Sensitization of IFN-γ Jak-STAT signaling during macrophage activation

Xiaoyu Hu; Carmen Herrero; Wai-Ping Li; Taras T. Antoniv; Erik Falck-Pedersen; Alisa E. Koch; James M. Woods; G. Kenneth Haines; Lionel B. Ivashkiv

A general paradigm in signal transduction is ligand-induced feedback inhibition and the desensitization of signaling. We found that subthreshold concentrations of interferon-γ (IFN-γ), which did not activate macrophages, increased their sensitivity to subsequent IFN-γ stimulation; this resulted in increased signal transducer and activator of transcription 1 (STAT1) activation and increased IFN-γ–dependent gene activation. Sensitization of IFN-γ signaling was mediated by the induction of STAT1 expression by low doses of IFN-γ that did not effectively induce feedback inhibition. IFN-γ signaling was sensitized in vivo after IFN-γ injection, and STAT1 expression was increased after injection of lipopolysaccharide and in rheumatoid arthritis synovial cells. These results identify a mechanism that sensitizes macrophages to low concentrations of IFN-γ and regulates IFN-γ responses in acute and chronic inflammation.


Nature Immunology | 2009

Cross-regulation of signaling by ITAM-associated receptors

Lionel B. Ivashkiv

An important function of receptors that signal through immunoreceptor tyrosine-based activation motifs (ITAMs) is to regulate signaling by heterologous receptors. This review describes mechanisms by which ITAM-associated receptors modulate signaling by Toll-like receptors (TLRs), tumor necrosis factor receptor family members and cytokine receptors that use the Jak–STAT signaling pathway, and the biological importance of this signal transduction cross-talk. ITAM-mediated cross-regulation can either augment or dampen signaling by other receptors. Conversely, TLRs and cytokines modulate ITAM-mediated signaling, by means including activation of β2 integrins that are coupled to the ITAM-containing adaptors DAP12 and FcRγ. Integration of ITAM signaling into signaling networks through cross-talk with other signal transduction pathways results in tight regulation and fine tuning of cellular responses to various extracellular stimuli and contributes to induction of specific activation and differentiation pathways.


Journal of Biological Chemistry | 2006

Role of STAT3 in Type I Interferon Responses NEGATIVE REGULATION OF STAT1-DEPENDENT INFLAMMATORY GENE ACTIVATION

Hao H. Ho; Lionel B. Ivashkiv

Type I interferons (IFNα/β) induce antiviral responses and have immunomodulatory effects that can either promote or suppress immunity and inflammation. In myeloid cells IFNα/β activates signal transducers and activators of transcription STAT1, STAT2, and STAT3. STAT1 and STAT2 mediate the antiviral and inflammatory effects of IFNα/β, but the function of IFNα/β-activated STAT3 is not known. We investigated the role of STAT3 in type I IFN signaling in myeloid cells by modulating STAT3 expression and the intensity of STAT3 activation using overexpression and RNA interference and determining the effects on downstream signaling and gene expression. IFNα-activated STAT3 inhibited STAT1-dependent gene activation, thereby down-regulating IFNα-mediated induction of inflammatory mediators such as the chemokines CXCL9 (Mig) and CXCL10 (IP-10). At the same time, IFNα-activated STAT3 supported ISGF-3-dependent induction of antiviral genes. STAT3 did not suppress STAT1 tyrosine phosphorylation or nuclear translocation but instead sequestered STAT1 and suppressed the formation of DNA-binding STAT1 homodimers. These results identify a regulatory function for STAT3 in attenuating the inflammatory properties of type I IFNs and provide a mechanism of suppression of STAT1 function that differs from previously described suppression of tyrosine phosphorylation. The results suggest that changes in the relative expression and activation of STAT1 and STAT3 that occur during immune responses determine the nature of cellular responses to type I IFNs.


Journal of Experimental Medicine | 2006

Twist mediates suppression of inflammation by type I IFNs and Axl

M. Nusrat Sharif; Dražen Šošić; Carla V. Rothlin; Erin Kelly; Greg Lemke; Eric N. Olson; Lionel B. Ivashkiv

Type I interferons (IFNs) are pleiotropic cytokines with antiviral and immunomodulatory properties. The immunosuppressive actions of type I IFNs are poorly understood, but IFN-mediated suppression of TNFα production has been implicated in the regulation of inflammation and contributes to the effectiveness of type I IFNs in the treatment of certain autoimmune and inflammatory diseases. In this study, we investigated mechanisms by which type I IFNs suppress induction of TNFα production by immune complexes, Fc receptors, and Toll-like receptors. Suppression of TNFα production was mediated by induction and activation of the Axl receptor tyrosine kinase and downstream induction of Twist transcriptional repressors that bind to E box elements in the TNF promoter and suppress NF-κB–dependent transcription. Twist expression was activated by the Axl ligand Gas6 and by protein S and apoptotic cells. These results implicate Twist proteins in regulation of TNFα production by antiinflammatory factors and pathways, and provide a mechanism by which type I IFNs and Axl receptors suppress inflammatory cytokine production.


Immunity | 2008

Integrated Regulation of Toll-like Receptor Responses by Notch and Interferon-γ Pathways

Xiaoyu Hu; Allen Y. Chung; Indira Wu; Julia Foldi; Janice Chen; Jong Dae Ji; Tomoko Tateya; Young Jun Kang; Jiahuai Han; Manfred Gessler; Ryoichiro Kageyama; Lionel B. Ivashkiv

Toll-like receptor (TLR) responses are regulated to avoid toxicity and achieve coordinated responses appropriate for the cell environment. We found that Notch and TLR pathways cooperated to activate canonical Notch target genes, including transcriptional repressors Hes1 and Hey1, and to increase production of canonical TLR-induced cytokines TNF, IL-6, and IL-12. Cooperation by these pathways to increase target gene expression was mediated by the Notch-pathway component and transcription factor RBP-J, which also contributed to lethality after endotoxin injection. TLR- and Notch-induced Hes1 and Hey1 attenuated IL-6 and IL-12 production. This Hes1- and Hey1-mediated feedback inhibitory loop was abrogated by interferon-gamma (IFN-gamma), which blocked TLR-induced activation of canonical Notch target genes by inhibiting Notch2 signaling and downstream transcription. These findings identify new immune functions for RBP-J, Hes, and Hey proteins and provide insights into mechanisms by which Notch, TLR, and IFN-gamma signals are integrated to modulate specific effector functions in macrophages.

Collaboration


Dive into the Lionel B. Ivashkiv's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kyung-Hyun Park-Min

Hospital for Special Surgery

View shared research outputs
Top Co-Authors

Avatar

George D. Kalliolias

Hospital for Special Surgery

View shared research outputs
Top Co-Authors

Avatar

Yu Qiao

Hospital for Special Surgery

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Janice Chen

Hospital for Special Surgery

View shared research outputs
Top Co-Authors

Avatar

Sung Ho Park

Hospital for Special Surgery

View shared research outputs
Top Co-Authors

Avatar

Anna Yarilina

Hospital for Special Surgery

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Taras T. Antoniv

Hospital for Special Surgery

View shared research outputs
Researchain Logo
Decentralizing Knowledge