Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2009 Aug 11;106(32):13445-50.
doi: 10.1073/pnas.0901944106. Epub 2009 Jul 27.

GARP (LRRC32) is essential for the surface expression of latent TGF-beta on platelets and activated FOXP3+ regulatory T cells

Affiliations

GARP (LRRC32) is essential for the surface expression of latent TGF-beta on platelets and activated FOXP3+ regulatory T cells

Dat Q Tran et al. Proc Natl Acad Sci U S A. .

Abstract

TGF-beta family members are highly pleiotropic cytokines with diverse regulatory functions. TGF-beta is normally found in the latent form associated with latency-associated peptide (LAP). This latent complex can associate with latent TGFbeta-binding protein (LTBP) to produce a large latent form. Latent TGF-beta is also found on the surface of activated FOXP3(+) regulatory T cells (Tregs), but it is unclear how it is anchored to the cell membrane. We show that GARP or LRRC32, a leucine-rich repeat molecule of unknown function, is critical for tethering TGF-beta to the cell surface. We demonstrate that platelets and activated Tregs co-express latent TGF-beta and GARP on their membranes. The knockdown of GARP mRNA with siRNA prevented surface latent TGF-beta expression on activated Tregs and recombinant latent TGF-beta1 is able to bind directly with GARP. Confocal microscopy and immunoprecipitation strongly support their interactions. The role of TGF-beta on Tregs appears to have dual functions, both for Treg-mediated suppression and infectious tolerance mechanism.

PubMed Disclaimer

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Fig. 1.
Fig. 1.
GARP and LAP are selectively expressed on activated FOXP3+ Tregs and platelets. (A) Level of FOXP3 and GARP mRNA on fresh (0 h) and activated CD4+CD25 T cells and Tregs (CD25hi) at 12, 24, 120 h, or 120 h with TGF-β1. Tregs were rested until day 14 (0 h) and restimulated for 18 h. (B) Flow cytometric analysis of surface LAP, GARP and intracellular FOXP3 on fresh (0 h) and activated Tregs (CD25hi) and CD4+CD25int T cells. (C) Surface staining of LAP and GARP on platelets based on FSC/SSC and CD61 expression. (D) LAP and GARP surface staining of plasmacytoid and myeloid DCs from PBMCs by gating on CD303+Lin-1 and CD1c+, respectively. (E) Surface LAP and GARP expression on Tregs after 12 h activation in the absence (none) or presence of monensin or brefeldin A for the last 8 h. Data are representative of 3 independent experiments. Numbers indicate percentage in each quadrant for B–E.
Fig. 2.
Fig. 2.
Surface expression of latent TGF-β1 requires GARP association. (A) Tregs were transfected with nonspecific (siNS), TGF-β1 (siTGFβ1), or GARP (siGARP) siRNA and rested in IL-2 culture medium for 24 h before stimulation for 48 h to assess surface expression of GARP and LAP. (B) Tregs transfected with TGF-β1 or GARP siRNA were activated for 48 h then incubated for 30 min at 37 °C with 5 μg/mL mature rhTGF-β1, LAP, LAP + TGF-β1, or latent TGF-β1 and then surface stained for LAP. (C) GARP transfected Jurkat cells were incubated for 30 min at 37 °C without (NONE) or with 5 μg/mL LAP, TGF-β1, TGF-β2, or TGF-β3 alone or mixed with LAP and then stained for surface LAP. Jurkat cells transfected with the RFP vector were used as a control. (D) GARP-transfected Jurkat cells were incubated with 5 μg/mL latent TGF-β1 (LTGFβ1) or preincubated for 20 min at 37 °C with a 10-fold excess of TSP1, RGD, or RGDS peptides before latent TGF-β1 incubation and then stained for surface LAP. (E) Tregs were expanded in vitro for 14 days and then restimulated for 48 h before immunoprecipitation with anti-LAP (left lane) or isotype (right lane) followed by immunoblot with anti-GARP. (F) Dynabeads conjugated to αGARP (Left) or αLAP (Right) were incubated with a mixture of GARP-Fc + TGF-β1 (shaded histogram), GARP-Fc + LAP (dashed histogram), or GARP-Fc + latent TGF-β1 (solid histogram) followed by flow cytometric analysis with PE-labeled anti-hLAP (Left) or anti-hFc (Right). Data are representative of 3 independent experiments for A, B, C, D, and F and 2 for E. Numbers indicate percentage in each quadrant for A–D.
Fig. 3.
Fig. 3.
Colocalization of GARP and latent TGF-β1 on the surface of activated Tregs. Tregs activated for 48 h from 3 different donors (D1, D2, D3) were surface-stained with anti-GARP and anti-LAP or isotype controls. The cells were imaged with a Leica SP2-AOBS confocal microscope.
Fig. 4.
Fig. 4.
GARP is required for the surface expression of latent TGF-β1. (A) FACS-sorted Tregs (CD25hi), naïve (CD45RA+), and memory (CD45RO+) T cells were activated for 5 days in the absence or presence of TGF-β1 then rested for 7 days and restimulated for 48 h without TGF-β1 before analysis of FOXP3 with surface GARP and LAP expressions. (B) Tregs were transfected with nonspecific (siNS) or FOXP3 (siFOXP3) siRNA and cultured for 5 days before activation for 48 h and analysis of FOXP3 with surface GARP and LAP expressions. (C) Naïve T cells were transduced with control (vector) or GARP encoded RFP-expressing lentiviral vectors and expanded for 7 days with anti-CD3/CD28 Dynabeads before restimulation for 48 h to evaluate for surface GARP and LAP (Left) and intracellular FOXP3 expressions among RFP+ and RFP cells (Right). Data are representative of 3 independent experiments. Numbers indicate percentage in each quadrant.
Fig. 5.
Fig. 5.
Dual functions of TGF-β in Treg-mediated suppression and infectious tolerance mechanism. (A) Tregs were transfected with siNS, siGARP, or siTGFβ1 and preactivated for 24 h before assessing their suppression of the proliferation of CD4+CD25 T cells stimulated with HLA-DR+ APCs and soluble anti-CD3 for 3 days and pulsed with 3H-TdR. The top panel is the FOXP3 expression, and the middle panel is the surface GARP and LAP expression of the 3 Treg populations at the end of the 3-day suppression assay. *, P < 0.05 between the siNS and siGARP or siTGFβ1 Treg suppression. (B) Similarly, CFSE-labeled CD4+CD25 responders were activated with HLA-DR+ APCs and soluble anti-CD3 for 3 days alone (top row) or in the presence of Tregs transfected with siNS, siGARP, or siTGFβ1 at a ratio of 2:1, 4:1, and 8:1 responder:Treg (bottom rows). (C) CFSE-labeled CD4+CD25CD127+CD45RA+ T cells were stimulated with anti-CD3/CD28 Dynabeads for 5 days alone (top panel) or with 24-h preactivated Tregs transfected with siNS, siGARP, or siTGFβ1 and analyzed for FOXP3 induction in the CFSE-labeled cells. Data are representative of 3 independent experiments. Numbers indicate percentage in each quadrant.

Similar articles

Cited by

References

    1. Wan YY, Flavell RA. TGF-beta and regulatory T cell in immunity and autoimmunity. J Clin Immunol. 2008;28:647–659. - PMC - PubMed
    1. Gleizes PE, et al. TGF-beta latency: Biological significance and mechanisms of activation. Stem Cells. 1997;15:190–197. - PubMed
    1. Lawrence DA. Latent-TGF-beta: An overview. Mol Cell Biochem. 2001;219:163–170. - PubMed
    1. Grainger DJ, Wakefield L, Bethell HW, Farndale RW, Metcalfe JC. Release and activation of platelet latent TGF-beta in blood clots during dissolution with plasmin. Nat Med. 1995;1:932–937. - PubMed
    1. Gandhi R, Anderson DE, Weiner HL. Cutting edge: Immature human dendritic cells express latency-associated peptide and inhibit T cell activation in a TGF-beta-dependent manner. J Immunol. 2007;178:4017–4021. - PubMed

Publication types

MeSH terms

-