GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Elsevier BV ; 2010
    In:  Journal of Biological Chemistry Vol. 285, No. 10 ( 2010-03), p. 7271-7280
    In: Journal of Biological Chemistry, Elsevier BV, Vol. 285, No. 10 ( 2010-03), p. 7271-7280
    Type of Medium: Online Resource
    ISSN: 0021-9258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2010
    detail.hit.zdb_id: 2141744-1
    detail.hit.zdb_id: 1474604-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 7, No. 1 ( 2017-03-21)
    Abstract: C1q collagen-like region (CLR) engaging and activating the LAIR-1 inhibitory immunoreceptor represents a non-complement mechanism for maintaining immune quiescence. Given the binding promiscuity of C1q’s globular region (gC1q), we hypothesized that C1q concurrently associates with distinct inhibitory immunoreceptors to produce C1q-mediated modulatory networking. Like LAIR-1, CD33 inhibitory immunoreceptors are highly expressed on monocytes. Binding CD33 restricts cell activation/differentiation; however, natural ligands for CD33 remain elusive. CD33 has IgC2-like domains potentially recognized by gC1q. Thus, we asked whether C1q binds to CD33 and if C1q mediates CD33/LAIR-1 crosslinking. Our findings demonstrate that C1q and gC1q interact with CD33 to activate its inhibitory motifs, while CLR does not. Whole C1q is required to crosslink CD33 and LAIR-1 and concurrently activate CD33/LAIR-1 inhibitory motifs. While C1q binds CD33C2 domains, decreased C1q-CD33 interactions resulting from sialic acid masking of CD33C2 domains suggests a process for regulating C1q-CD33 activity. Consistent with defective self-tolerance, CD33/LAIR-1 expression is reduced in systemic lupus erythematosus (SLE) myelomonocytes. The anti-inflammatory cytokine M-CSF, but not DC growth factors, sustains CD33/LAIR-1 expression on both healthy and SLE cells suggesting further biological control of C1q-CD33/LAIR-1 processes.
    Type of Medium: Online Resource
    ISSN: 2045-2322
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2017
    detail.hit.zdb_id: 2615211-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Wiley ; 2016
    In:  Immunological Reviews Vol. 269, No. 1 ( 2016-01), p. 100-117
    In: Immunological Reviews, Wiley, Vol. 269, No. 1 ( 2016-01), p. 100-117
    Abstract: Numerous risk alleles for systemic lupus erythematosus ( SLE ) have now been identified. Analysis of the expression of genes with risk alleles in cells of hematopoietic origin demonstrates them to be most abundantly expressed in B cells and dendritic cells ( DC s), suggesting that these cell types may be the drivers of the inflammatory changes seen in SLE . DC s are of particular interest as they act to connect the innate and the adaptive immune response. Thus, DC s can transform inflammation into autoimmunity, and autoantibodies are the hallmark of SLE . In this review, we focus on mechanisms of tolerance that maintain DC s in a non‐activated, non‐immunogenic state. We demonstrate, using examples from our own studies, how alterations in DC function stemming from either DC ‐intrinsic abnormalities or DC ‐extrinsic regulators of function can predispose to autoimmunity.
    Type of Medium: Online Resource
    ISSN: 0105-2896 , 1600-065X
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2016
    detail.hit.zdb_id: 2038276-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    MDPI AG ; 2021
    In:  Cells Vol. 10, No. 3 ( 2021-03-05), p. 564-
    In: Cells, MDPI AG, Vol. 10, No. 3 ( 2021-03-05), p. 564-
    Abstract: The disruption of the immune tolerance induces autoimmunity such as systemic lupus erythematosus and vasculitis. A chromatin-binding non-histone protein, high mobility group box 1 (HMGB1), is released from the nucleus to the extracellular milieu in particular environments such as autoimmunity, sepsis and hypoxia. Extracellular HMGB1 engages pattern recognition receptors, including Toll-like receptors (TLRs) and the receptor for advanced glycation endproducts (RAGE). While the HMGB1-RAGE axis drives inflammation in various diseases, recent studies also focus on the anti-inflammatory effects of HMGB1 and RAGE. This review discusses current perspectives on HMGB1 and RAGE’s roles in controlling inflammation and immune tolerance. We also suggest how RAGE heterodimers responding microenvironments functions in immune responses.
    Type of Medium: Online Resource
    ISSN: 2073-4409
    Language: English
    Publisher: MDPI AG
    Publication Date: 2021
    detail.hit.zdb_id: 2661518-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Proceedings of the National Academy of Sciences ; 2021
    In:  Proceedings of the National Academy of Sciences Vol. 118, No. 26 ( 2021-06-29)
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 118, No. 26 ( 2021-06-29)
    Abstract: Patients with severe COVID-19 infection exhibit a low level of oxygen in affected tissue and blood. To understand the pathophysiology of COVID-19 infection, it is therefore necessary to understand cell function during hypoxia. We investigated aspects of human monocyte activation under hypoxic conditions. HMGB1 is an alarmin released by stressed cells. Under normoxic conditions, HMGB1 activates interferon regulatory factor (IRF)5 and nuclear factor-κB in monocytes, leading to expression of type I interferon (IFN) and inflammatory cytokines including tumor necrosis factor α, and interleukin 1β, respectively. When hypoxic monocytes are activated by HMGB1, they produce proinflammatory cytokines but fail to produce type I IFN. Hypoxia-inducible factor-1α, induced by hypoxia, functions as a direct transcriptional repressor of IRF5 and IRF3. As hypoxia is a stressor that induces secretion of HMGB1 by epithelial cells, hypoxia establishes a microenvironment that favors monocyte production of inflammatory cytokines but not IFN. These findings have implications for the pathogenesis of COVID-19.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2021
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Elsevier BV ; 2013
    In:  Journal of Autoimmunity Vol. 44 ( 2013-08), p. 34-39
    In: Journal of Autoimmunity, Elsevier BV, Vol. 44 ( 2013-08), p. 34-39
    Type of Medium: Online Resource
    ISSN: 0896-8411
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2013
    detail.hit.zdb_id: 1468989-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Clinical Immunology, Elsevier BV, Vol. 250 ( 2023-05), p. 109317-
    Type of Medium: Online Resource
    ISSN: 1521-6616
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 1462862-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2016
    In:  The Journal of Immunology Vol. 196, No. 1_Supplement ( 2016-05-01), p. 196.18-196.18
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 196, No. 1_Supplement ( 2016-05-01), p. 196.18-196.18
    Abstract: Cross-linking of the inhibitory immunoreceptor LAIR-1 (CD305) on the cell surface phosphorylates LAIR-1 inhibitory motifs (ITIM) and restricts the growth of monocyte-derived dendritic cells (mono-DCs). Despite the suggestion that LAIR-1’s ability to prevent unwarranted immunity is compromised in autoimmunity, much remains to be learned about the control of LAIR-1 expression. Here, we compared the ability of various myeloid cytokines to control LAIR-1 expression on the surface of monocytes/macrophages (mϕs) and mono-DCs. Treatment of freshly isolated blood monocytes from healthy donors with mono-DC growth factors (GM-CSF+IL-4/GM-CSF+IL-13) led to reduced levels of LAIR-1 at the critical monocyte-to-mono-DC juncture. In contrast, M-CSF (a steady state/M2 cytokine) sustained LAIR-1+CD14+CD33m/M+CD16+ mono-mϕs. M-CSF effects on LAIR-1 occurred under serum-supplemented and serum-free conditions. Addition of M-CSF to mono-DCs previously cultured in GM-CSF+IL-4 promptly reversed the down-regulatory effects of GM-CSF+IL-4 producing increases in LAIR-1+CD14+ cells. Conversely, addition of GM-CSF+IL-4 to M-CSF exposed cultures lowered levels of LAIR-1 and CD14. M-CSF treatment of SLE blood myeloid cells (featuring activated monocytes/mono-DCs lacking LAIR-1, CD14) produced high levels of LAIR-1 and CD14 vs GM-CSF/IL-4. Thus: 1. The expression of LAIR-1 on monocytes/mϕs and mono-DCs is distinctly and reversibly controlled by myeloid cytokines. 2. LAIR-1 expression on circulating myeloid cells in SLE may be regulated by M-CSF.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2016
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2012
    In:  The Journal of Immunology Vol. 188, No. 1_Supplement ( 2012-05-01), p. 167.1-167.1
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 188, No. 1_Supplement ( 2012-05-01), p. 167.1-167.1
    Abstract: Leukocyte-associated inhibitory receptor (LAIR)-1 is a collagen receptor. Cross-linking of LAIR-1 suppresses the differentiation of dendritic cells (DCs). We found that C1q, which is known to have an inhibitory role in the differentiation of DCs, maintained the expression of LAIR-1 in cells transitioning between monocytes and DCs. Since C1q contains a collagen-like tail, we hypothesized that a C1q/LAIR-1 interaction leads to DC lineage arrest. In order to assess interactions between C1q and LAIR-1, we explored three models. First, HEK293T cells were transfected with a plasmid encoding LAIR-1. Labeled C1q associated with LAIR-1 expressing cells in a dose specific manner; this binding was inhibited by soluble LAIR-2, which exhibits the same collagen binding capacity as LAIR-1. Second, THP-1 cells, a LAIR-1hi human monocytic leukemia cell line, exhibited a high degree of binding to C1q which was also blocked by sLAIR-2. Third, in primary human monocytes, sLAIR-2 blocked the effects of C1q on monocyte-derived DC differentiation. We confirmed a direct interaction between C1q and LAIR-1 by dot blot analysis. Moreover, LAIR-1 was phosphorylated in a C1q-dependent manner in primary monocytes, demonstrating that C1q is an inducer of LAIR-1/ITIM-specific phosphorylation. Collectively, these data indicate that C1q functions as a natural ligand for LAIR-1. These findings provide novel insights into how C1q may prevent unwarranted immune responses.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2012
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2012
    In:  The Journal of Immunology Vol. 188, No. 1_Supplement ( 2012-05-01), p. 159.26-159.26
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 188, No. 1_Supplement ( 2012-05-01), p. 159.26-159.26
    Abstract: Systemic Lupus Erythematosus (SLE) is an autoimmune disease characterized by the loss of tolerance to autoantigens, and to DNA in particular. Although Immune complexes (ICs) of IgG anti-DNA antibodies are pathogenic, IgM anti-DNA antibodies in SLE are protective. The mechanism responsible for the protective effect of IgM anti-DNA antibody is unclear. We found that a monoclonal IgM anti-DNA antibody cooperated with C1q to suppress interferon (IFN)-α production by plasmacytoid DCs (pDCs). We tested the effects of the antibody on bone marrow-derived pDCs from wild type (WT) and C1q knockout (KO) mice. Interestingly, the absence of C1q significantly enhanced the production of IFN-α induced by IgM anti-DNA antibody. The inhibitory effect of C1q was confirmed using in vivo generated pDCs or in vitro generated pDCs under serum-free conditions. Exogenous C1q suppressed IFN-α transcription in both WT and C1q KO pDCs. Moreover, we identified leukocyte-associated inhibitory receptor (LAIR)-1 as a potential receptor for C1q. Fluorescence labeled C1q strongly bound to pDCs which were highly expressing LAIR-1. LAIR-1 is a collagen receptor containing two ITIMs; its cross-linking with antibody inhibits IFN-a production by pDCs stimulated with DNA containing ICs. In summary, protective IgM anti-DNA antibody/C1q complexes inhibit unwanted IFN-α production through binding to LAIR-1.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2012
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...