Elsevier

Matrix Biology

Volume 32, Issue 6, 8 August 2013, Pages 316-324
Matrix Biology

Thrombospondin-1 is a CD47-dependent endogenous inhibitor of hydrogen sulfide signaling in T cell activation

https://doi.org/10.1016/j.matbio.2013.02.009Get rights and content

Highlights

  • Endogenous and exogenous thrombospondin-1 limits the ability of H2S to enhance T cell receptor-mediated T cell activation.

  • CD47 is necessary for thrombospondin-1 to inhibit activation of T cells by H2S.

  • Thrombospondin-1 inhibits H2S -induced ERK1/2 phosphorylation in activated T cells.

  • Thrombospondin-1 limits activation-induced H2S production by inhibiting expression of cystathionine β-synthase and γ-lyase.

  • Thrombospondin-1 signaling through CD47 is the first identified endogenous inhibitor of H2S signaling.

Abstract

Thrombospondin-1 is a potent suppressor of T cell activation via its receptor CD47. However, the precise mechanism for this inhibition remains unclear. Because H2S is an endogenous potentiator of T cell activation and is necessary for full T cell activation, we hypothesized that thrombospondin-1 signaling through CD47 inhibits T cell activation by antagonizing H2S signaling. Primary T cells from thrombospondin-1 null mice were more sensitive to H2S-dependent activation assessed by proliferation and induction of interleukin-2 and CD69 mRNAs. Exogenous thrombospondin-1 inhibited H2S responses in wild type and thrombospondin-1 null T cells but enhanced the same responses in CD47 null T cells. Fibronectin, which shares integrin and glycosaminoglycan binding properties with thrombospondin-1 but not CD47 binding, did not inhibit H2S signaling. A CD47-binding peptide derived from thrombospondin-1 inhibited H2S-induced activation, whereas two other functional sequences from thrombospondin-1 enhanced H2S signaling. Therefore, engaging CD47 is necessary and sufficient for thrombospondin-1 to inhibit H2S-dependent T cell activation. H2S stimulated T cell activation by potentiating MEK-dependent ERK phosphorylation, and thrombospondin-1 inhibited this signaling in a CD47-dependent manner. Thrombospondin-1 also limited activation-dependent T cell expression of the H2S biosynthetic enzymes cystathionine β-synthase and cystathionine γ-lyase, thereby limiting the autocrine role of H2S in T cell activation. Thus, thrombospondin-1 signaling through CD47 is the first identified endogenous inhibitor of H2S signaling and constitutes a novel mechanism that negatively regulates T cell activation.

Introduction

Thrombospondin-1 (TSP1) is a large (450 kDa) matricellular glycoprotein that plays a pivotal role in regulating vascular homeostasis (Isenberg et al., 2009, Bauer et al., 2010), platelet activation (Isenberg et al., 2008), angiogenesis (Carlson et al., 2008, Miller et al., 2009, Roberts et al., 2012), and immunity (Lopez-Dee et al., 2011). TSP1 mediates these activities by binding to other extracellular matrix components and growth factors, mediating activation of latent TGF-β1 (Schultz-Cherry and Murphy-Ullrich, 1993, Sweetwyne and Murphy-Ullrich, 2012), and binding to at least 12 different cell surface receptors(Murphy-Ullrich and Iozzo, 2012). These receptors include five integrins (Lawler et al., 1988, Chandrasekaran et al., 2000, Calzada et al., 2003, Calzada et al., 2004a, Calzada et al., 2004b, Staniszewska et al., 2007), CD36 (Dawson et al., 1997), CD47 (Gao et al., 1996), CD148 (Takahashi et al., 2012), calreticulin/low density lipoprotein receptor-related protein-1 (LRP1) (Elzie and Murphy-Ullrich, 2004), proteoglycans (Feitsma et al., 2000), and sulfatides (Guo et al., 1992). Among these, TSP1 has the highest affinity for CD47, and this receptor is both necessary and sufficient for TSP1 to inhibit NO-cGMP signaling (Isenberg et al., 2006).

TSP1 regulates T cell activation and function in a domain specific manner. Although TSP1 enhances some T cell actions via its N-terminal domains, such as α4β1 integrin-dependent adhesion and chemotaxis (Li et al., 2002), the dominant effect of soluble TSP1 is the potent inhibition of TCR-mediated T cell activation (Li et al., 2001). This inhibition requires interaction of the C-terminal domain of TSP1 with a proteoglycan isoform of CD47 on the T cell surface (Li et al., 2002, Kaur et al., 2011). The inhibitory activity of TSP1 does not require β1 integrins (Li et al., 2002) and is independent of TGFβ, based on resistance to TGFβ-function blocking antibodies (Li et al., 2001) and the inhibitory activity of a recombinant signature domain of TSP1 that lacks the TGFβ binding and activation sequences in the type 1 repeats (Ramanathan et al., 2011). Further evidence that CD47 ligation is sufficient to inhibit T cell activation derives from the inhibitory activity of some CD47 antibodies and CD47-binding peptides such as 7N3 (FIRVVMYEGKK), but not the corresponding control peptide FIRGGMYEGKK (Li et al., 2001). Despite this evidence that CD47 ligation is necessary and sufficient for inhibiting TCR-dependent T cell activation, the lack of a substantial cytoplasmic domain in CD47 for docking of downstream signaling molecules suggests that lateral interactions with other membrane proteins such as growth factor receptors, integrins, PLIC-1, Fas receptor, and SIRPs are generally required for its signaling functions (reviewed in (Soto-Pantoja et al., 2013).

While the proximal intracellular targets of TSP1/CD47–mediated inhibition of T cell activation are not known, this inhibition occurs downstream of the TCR targeting linker for activated T cells (LAT) and Zap70, but upstream of NFAT activation (Li et al., 2001). TSP1 regulates the activation of soluble guanylate cyclase by NO in Jurkat T lymphoma cells in a calcium-dependent manner (Ramanathan et al., 2011), but this pathway cannot account for the broad effects of CD47 signaling on T cell activation as cGMP signaling is not reported to play a major role in T cell activation and is limited to T cell differentiation (Niedbala et al., 2006).

H2S is emerging as an important member of the gasotransmitter family that also includes NO and carbon monoxide (CO). At toxic environmental concentrations (> 200 ppm), H2S inhibits mitochondrial cytochrome c oxidase (Reiffenstein et al., 1992). Lower nontoxic concentrations have physiological functions in neuromodulation ((Abe and Kimura, 1996) and reviewed in (Tan et al., 2009)), metabolic hibernation (Blackstone et al., 2005, Blackstone and Roth, 2007), protection from ischemia/reperfusion injury (Sivarajah et al., 2006, Elrod et al., 2007, Fu et al., 2008, Jha et al., 2008, Tripatara et al., 2008), oxygen sensing (Olson and Whitfield, 2009), vasodilatation (Hosoki et al., 1997, Yang et al., 2008), and promotion of angiogenesis (Wang et al., 2010). Like its gasotransmitter cousins NO and CO, H2S has transitioned from being perceived exclusively as toxin to recognition that it is an important endogenous signaling molecule. In common with NO, H2S has been implicated as both a pro- (Bhatia et al., 2005, Collin et al., 2005, Zhang et al., 2007, Cunha et al., 2008) and anti-inflammatory molecule in innate immune cells (Zanardo et al., 2006, Li et al., 2007, Cunha et al., 2008, Sivarajah et al., 2009). Like NO, H2S relies on its distinctive chemistry for signal transduction, which includes modification of specific protein cysteine residues (termed sulfhydration) and ligation of ferric iron, zinc, or copper centers in metalloproteins (Fukuto et al., 2012).

Recently, we reported that H2S is a potentiator of T cell activation in primary murine and human T cells and T cell lines (Miller et al., 2012). Exogenously added H2S, at nanomolar physiological levels (Furne et al., 2008, Shen et al., 2012) enhances both polyclonal and antigen-specific T cell activation. Notably the capacity of T cells to endogenously make H2S via cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE) is turned on as a result of T cell activation. Suppression of CBS and CSE expression by siRNA inhibits T cell activation and T cell proliferation, which can be rescued by supplementation with exogenous H2S. Therefore, H2S signaling is a necessary component of T cell activation.

As there are no reported endogenous inhibitors of H2S signaling, we sought to examine the effect of inhibitory TSP1 signaling through CD47 on H2S-mediated T cell activation. TSP1 is a logical candidate for regulation of this pathway given its potent and broad T cell inhibitory effects and its potent regulation of NO gasotransmitter signaling.

Section snippets

TSP1 null T cells are more sensitive to H2S-potentiated activation

In order to examine the role of TSP1 in H2S-dependent T cell activation, we compared the activation of WT and TSP1 null CD3+ murine T cells via plate-bound anti-CD3 and anti-CD28 antibodies in the presence of H2S. Using IL-2 gene expression as a marker of T cell activation, we observed, as previously, that H2S dose-dependently enhanced IL-2 expression in WT T cells by up to 4-fold over control activated cells not treated with H2S (Fig. 1A). H2S enhancement of IL-2 expression was greater at all H

Discussion

Recently we reported that physiological levels of the gasotransmitter H2S in the nanomolar range function as an endogenous potentiator of T cell activation (Miller et al., 2012). The present work identifies an extracellular matrix signaling pathway that limits this H2S function in T cells (summarized in Fig. 7). We demonstrate that the previously reported potent inhibition of T cell activation by TSP1 (Li et al., 2001) is mediated at least in part through inhibiting T cell responses to H2S and

Cells and reagents

H2S refers to any of its various protonation states (H2S  HS + H+  S2  + H+) with HS being the predominant form at physiological pH (pKa = 6.8). Na2S and NaHS, the corresponding sodium salts of these anionic forms of H2S, are considered H2S donors at physiological pH and are used as sources of H2S for this study. C57Bl/6 mice were anesthetized and sacrificed by cervical dislocation, and their spleens were harvested for T cell culture. The spleens were gently ruptured in a 40 micron cell strainer (BD

Acknowledgments

This work was supported by the Intramural Research Program of the National Institutes of Health, National Cancer Institute, Center for Cancer Research.

References (85)

  • J.S. Isenberg et al.

    Thrombospondin-1 stimulates platelet aggregation by blocking the antithrombotic activity of nitric oxide/cGMP signaling

    Blood

    (2008)
  • J.S. Isenberg et al.

    Thrombospondin-1 and CD47 regulate blood pressure and cardiac responses to vasoactive stress

    Matrix Biol.

    (2009)
  • S. Kaur et al.

    Identification of CD47 and amyloid precursor-like protein-2 as the major heparan sulfate proteoglycans on T lymphocytes and this isoform of CD47 as the signaling receptor for thrombospondin-1

    J. Biol. Chem.

    (2011)
  • J. Kwon et al.

    T cell receptor-stimulated generation of hydrogen peroxide inhibits MEK–ERK activation and lck serine phosphorylation

    Free Radic. Biol. Med.

    (2003)
  • S.S. Li et al.

    Endogenous thrombospondin-1 is a cell-surface ligand for regulation of integrin-dependent T-lymphocyte adhesion

    Blood

    (2006)
  • L. Li et al.

    Anti-inflammatory and gastrointestinal effects of a novel diclofenac derivative

    Free Radic. Biol. Med.

    (2007)
  • K.N. Maclean et al.

    The dominant role of Sp1 in regulating the cystathionine beta-synthase -1a and -1b promoters facilitates potential tissue-specific regulation by Kruppel-like factors

    J. Biol. Chem.

    (2004)
  • T.W. Miller et al.

    Hydrogen sulfide is an endogenous potentiator of T cell activation

    J. Biol. Chem.

    (2012)
  • J.E. Murphy-Ullrich et al.

    Thrombospondins in physiology and disease: new tricks for old dogs

    Matrix Biol.

    (2012)
  • E. Negre et al.

    The collagen binding domain of fibronectin contains a high affinity binding site for Candida albicans

    J. Biol. Chem.

    (1994)
  • M. Nejmeddine et al.

    HTLV-1-Tax and ICAM-1 act on T-cell signal pathways to polarize the microtubule-organizing center at the virological synapse

    Blood

    (2009)
  • D.D. Roberts et al.

    The matricellular protein thrombospondin-1 globally regulates cardiovascular function and responses to stress via CD47

    Matrix Biol.

    (2012)
  • X. Shen et al.

    Analytical measurement of discrete hydrogen sulfide pools in biological specimens

    Free Radic. Biol. Med.

    (2012)
  • M.T. Sweetwyne et al.

    Thrombospondin1 in tissue repair and fibrosis: TGF-beta-dependent and independent mechanisms

    Matrix Biol.

    (2012)
  • P. Tripatara et al.

    Generation of endogenous hydrogen sulfide by cystathionine gamma-lyase limits renal ischemia/reperfusion injury and dysfunction

    Lab. Invest.

    (2008)
  • D. Tulasne et al.

    C-terminal peptide of thrombospondin-1 induces platelet aggregation through the Fc receptor gamma-chain-associated signaling pathway and by agglutination

    Blood

    (2001)
  • G. Yang et al.

    Specificity protein-1 as a critical regulator of human cystathionine gamma-lyase in smooth muscle cells

    J. Biol. Chem.

    (2011)
  • K. Abe et al.

    The possible role of hydrogen sulfide as an endogenous neuromodulator

    J. Neurosci.

    (1996)
  • E.M. Bauer et al.

    Thrombospondin-1 supports blood pressure by limiting eNOS activation and endothelial-dependent vasorelaxation

    Cardiovasc. Res.

    (2010)
  • M. Bhatia et al.

    Hydrogen sulphide is a mediator of carrageenan-induced hindpaw oedema in the rat

    Br. J. Pharmacol.

    (2005)
  • E. Blackstone et al.

    Suspended animation-like state protects mice from lethal hypoxia

    Shock

    (2007)
  • E. Blackstone et al.

    H2S induces a suspended animation-like state in mice

    Science

    (2005)
  • S. Bouguermouh et al.

    CD47 expression on T cell is a self-control negative regulator of type 1 immune response

    J. Immunol.

    (2008)
  • B.J. Burbach et al.

    T-cell receptor signaling to integrins

    Immunol. Rev.

    (2007)
  • M.J. Calzada et al.

    α4β1 integrin mediates selective endothelial cell responses to thrombospondins in vitro and modulates angiogenesis in vivo

    Circ. Res.

    (2004)
  • C.B. Carlson et al.

    Structures of thrombospondins

    Cell. Mol. Life Sci.

    (2008)
  • L. Chandrasekaran et al.

    Cell contact-dependent activation of α3β1 integrin modulates endothelial cell responses to thrombospondin-1

    Mol. Biol. Cell

    (2000)
  • X. Chen et al.

    Many NK cell receptors activate ERK2 and JNK1 to trigger microtubule organizing center and granule polarization and cytotoxicity

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • M. Collin et al.

    Inhibition of endogenous hydrogen sulfide formation reduces the organ injury caused by endotoxemia

    Br. J. Pharmacol.

    (2005)
  • D.W. Dawson et al.

    CD36 mediates the In vitro inhibitory effects of thrombospondin-1 on endothelial cells

    J. Cell Biol.

    (1997)
  • J.B. de Kok et al.

    Normalization of gene expression measurements in tumor tissues: comparison of 13 endogenous control genes

    Lab. Invest.

    (2004)
  • S. Devadas et al.

    Discrete generation of superoxide and hydrogen peroxide by T cell receptor stimulation: selective regulation of mitogen-activated protein kinase activation and fas ligand expression

    J. Exp. Med.

    (2002)
  • Cited by (66)

    • Thrombospondin-1 in drug activity and tumor response to therapies

      2024, Seminars in Cell and Developmental Biology
    • Emerging functions of thrombospondin-1 in immunity

      2024, Seminars in Cell and Developmental Biology
    • Thrombospondins in the tumor microenvironment

      2024, Seminars in Cell and Developmental Biology
    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text