To the editor:

In an article in this issue, Pilch-Cooper et al report an artifact they observed when they tried to detect intracellular CCR5 molecules by flow cytometry.1  When they stained fixed and permeabilized cells with an anti-CCR5 antibody, they obtained a strong signal even in non–CCR5-expressing cells, with a significant off-target staining in the nucleus. They concluded that their procedure generated irrelevant antibody binding sites during the fixing and permeabilization resulting in a false-positive result and that T cells do not have large intracellular pools of CCR5. Whereas the first part of their statement is probably correct, the second one might not be.

With an alternative procedure of intracellular labeling we have previously published,2  we were able to detect CCR5 molecules in the peripheral blood CD4+ T cells from a wild-type (WT)/WT CCR5 donor but not from a delta32(Δ32)/Δ32 CCR5 donor, the epitope recognized by the anti-CCR5 monoclonal antibody (mAb) 2D7 we used being disrupted in Δ32 CCR5 molecules3,4  (Figure 1A). When we transduced the WT CCR5 gene into the Δ32/Δ32 CD4+ T cells, we detected a signal in permeabilized cells (Figure 1B). To distinguish between surface and intracellular labeling, we first exposed these transduced CD4+ T cells to a saturating concentration of the unlabeled anti-CCR5 mAb 2D7, permeabilized the cells or not, and thereafter stained them with the same mAb conjugated with a fluorescent dye. Under these conditions, the permeabilized cells were labeled, whereas the nonpermeabilized cells were not (Figure 1C). Fluorescence microscopy confirmed that the staining was in the cytoplasm (Figure 1D). Moreover, overnight treatment with brefeldin A, which inhibits transport of proteins from endoplasmic reticulum to Golgi, leading to protein accumulation inside the endoplasmic reticulum, increased the amount of intracellular CCR5 molecules that were stained (mean fluorescence intensity [MFI], 33 ± 2 and 61 ± 1, P < .001, without and with brefeldin A, respectively; n = 3, Figure 1E). Finally, when we exposed circulating CD4+ T cells to the CCR5 ligand MIP-1β, we observed a decrease in the intensity of the labeling obtained with nonpermeabilized cells (MFI, 38 ± 2 and 24 ± 1, P = .005, without and with MIP-1β, respectively; n = 3, Figure 1F), contrasting with an increase in the intensity of the labeling obtained with permeabilized cells pre-exposed to nonconjugated 2D7 (MFI, 121 ± 8 and 190 ± 3, P = .008, without and with MIP-1β, respectively; n = 3, Figure 1F). These data are compatible with the ligand-induced internalization of cell surface CCR5 molecules.

Figure 1

Immunofluorescence analyses of CCR5-stained fresh or permeabilized peripheral blood CD4+ T cells. (A) Specific detection of CCR5 molecules on and inside CD4+ T cells. CCR5 expression of nonpermeabilized (NP, top histograms) and permeabilized (P, bottom histograms) circulating CD4+ T cells from a WT/WT CCR5 (left histograms) and a Δ32/Δ32 (right histograms) donor were analyzed. For CCR5 extracellular detection, whole blood was stained with a phycoerythrin (PE)–conjugated anti-CD4 mAb and the anti-CCR5 mAb 2D7 conjugated to PE-cyanin-5 (PC5; open histogram) or with an isotype control (gray filled histogram). For CCR5 intracellular detection, peripheral blood mononuclear cells (PBMCs) were permeabilized using PBS containing 0.2% saponin as previously described.2  CCR5 expression was analyzed after gating for lymphocytes on the basis of forward and side scatter and then gating on CD4+ cells. The percentage of CD4+ T cells expressing CCR5 is indicated. (B) Detection of CCR5 molecules on and inside the Δ32/Δ32 CD4+ T cells transduced with CCR5. PBMC from a Δ32/Δ32 donor were transduced with an HIV-1–derived vector harboring the WT CCR5 gene (right histograms) or the LacZ gene (left histograms) as a negative control as previously described,7  permeabilized (P, bottom histograms) or not (NP, top histograms) 48 hours later, and processed as described in panel A. The percentage of CD4+ T cells expressing CCR5 is indicated. (C) Detection of intracellular CCR5 molecules by flow cytometry. PBMCs from a Δ32/Δ32 donor were transduced as described in panel B, incubated 48 hours later with 1 μg/mL 2D7, permeabilized (P, right histogram) or not (NP, left histogram), and processed as described in panel A. The percentage of CD4+ T cells expressing CCR5 is indicated. (D) Detection of intracellular CCR5 molecules by immunofluorescence microscopy. Wide-field immunofluorescence image of nonpermeabilized (NP) and permeabilized (P) PBMCs from a WT/WT CCR5 donor labeled using the 2D7 primary antibody (1 μg/mL) and a FITC-conjugated goat anti–mouse antibody. Cells were observed with a Leica DM6000 microscope. Scale bar, 3 μm. (E) Effect of brefeldin A on CCR5 staining of permeabilized (P) CD4+ T cells. PBMC from a WT/WT CCR5 donor were incubated (bottom histogram) or not (top histogram) for 24 hours with 10 μg/mL brefeldin A (BFA). Cells were then incubated with 1 μg/mL 2D7, permeabilized, and then processed as described in panel C. One representative experiment of 3 performed is shown. (F) Ligand-induced internalization of CCR5. PBMCs from a WT/WT CCR5 donor were incubated (bottom histograms) or not (top histograms) for 1 hour at 37°C with 500 ng/mL MIP-1β, and permeabilized (P, right histogram) or not (NP, left histogram). Nonpermeabilized cells were processed as described in panel A, and permeabilized cells as described in panel C. One representative experiment of 3 performed is shown.

Figure 1

Immunofluorescence analyses of CCR5-stained fresh or permeabilized peripheral blood CD4+ T cells. (A) Specific detection of CCR5 molecules on and inside CD4+ T cells. CCR5 expression of nonpermeabilized (NP, top histograms) and permeabilized (P, bottom histograms) circulating CD4+ T cells from a WT/WT CCR5 (left histograms) and a Δ32/Δ32 (right histograms) donor were analyzed. For CCR5 extracellular detection, whole blood was stained with a phycoerythrin (PE)–conjugated anti-CD4 mAb and the anti-CCR5 mAb 2D7 conjugated to PE-cyanin-5 (PC5; open histogram) or with an isotype control (gray filled histogram). For CCR5 intracellular detection, peripheral blood mononuclear cells (PBMCs) were permeabilized using PBS containing 0.2% saponin as previously described.2  CCR5 expression was analyzed after gating for lymphocytes on the basis of forward and side scatter and then gating on CD4+ cells. The percentage of CD4+ T cells expressing CCR5 is indicated. (B) Detection of CCR5 molecules on and inside the Δ32/Δ32 CD4+ T cells transduced with CCR5. PBMC from a Δ32/Δ32 donor were transduced with an HIV-1–derived vector harboring the WT CCR5 gene (right histograms) or the LacZ gene (left histograms) as a negative control as previously described,7  permeabilized (P, bottom histograms) or not (NP, top histograms) 48 hours later, and processed as described in panel A. The percentage of CD4+ T cells expressing CCR5 is indicated. (C) Detection of intracellular CCR5 molecules by flow cytometry. PBMCs from a Δ32/Δ32 donor were transduced as described in panel B, incubated 48 hours later with 1 μg/mL 2D7, permeabilized (P, right histogram) or not (NP, left histogram), and processed as described in panel A. The percentage of CD4+ T cells expressing CCR5 is indicated. (D) Detection of intracellular CCR5 molecules by immunofluorescence microscopy. Wide-field immunofluorescence image of nonpermeabilized (NP) and permeabilized (P) PBMCs from a WT/WT CCR5 donor labeled using the 2D7 primary antibody (1 μg/mL) and a FITC-conjugated goat anti–mouse antibody. Cells were observed with a Leica DM6000 microscope. Scale bar, 3 μm. (E) Effect of brefeldin A on CCR5 staining of permeabilized (P) CD4+ T cells. PBMC from a WT/WT CCR5 donor were incubated (bottom histogram) or not (top histogram) for 24 hours with 10 μg/mL brefeldin A (BFA). Cells were then incubated with 1 μg/mL 2D7, permeabilized, and then processed as described in panel C. One representative experiment of 3 performed is shown. (F) Ligand-induced internalization of CCR5. PBMCs from a WT/WT CCR5 donor were incubated (bottom histograms) or not (top histograms) for 1 hour at 37°C with 500 ng/mL MIP-1β, and permeabilized (P, right histogram) or not (NP, left histogram). Nonpermeabilized cells were processed as described in panel A, and permeabilized cells as described in panel C. One representative experiment of 3 performed is shown.

Close modal

Our data argue for the presence of intracellular CCR5 in peripheral blood CD4+ T cells. The reason we did not get the nonspecific binding reported by Pilch-Cooper et al may be because of differences in the method of permeabilization. Our duration of exposition to the detergent is shorter, and we used saponin instead of Triton X-100, which is known to affect proteins and to permeabilize nuclear membrane.5  Moreover, we did not fix the cells.

We have previously determined that about two-thirds of the CCR5 molecules are located in the cytoplasm of peripheral blood CD4+ T cells and one-third at their surface.2  In vivo, CCR5 ligands present in the plasma probably induce the permanent internalization of cell surface CCR5 molecules, causing part of their intracellular accumulation. Consequently, blocking the interaction between CCR5 and its ligands by the administration of a CCR5 antagonist to healthy volunteers results in an increase in CD4+ T-cell surface CCR5 density and a decrease in the intracellular pool of CCR5 molecules.6  Conversely, in case of overproduction of CCR5-binding chemokines by peripheral blood mononuclear cells, as in rheumatoid arthritis, the proportion of CCR5 molecules expressed at the surface of CD4+ T cells is decreased compared with the proportion of intracellular CCR5 molecules.2 

Conflict-of-interest disclosure: P.C. has received grants from Pfizer, Merch Sharp and Dohme-Chibret, Gilead, and GlaxSmithKline. The remaining authors declare no competing financial interests.

Correspondence: Pierre Corbeau, Laboratoire d'Immunologie, Hôpital Saint Eloi, 80 avenue A. Fliche, 34295, Montpellier cedex 5, France; e-mail: p-corbeau@chu-montpellier.fr.

1
Pilch-Cooper
 
HA
Sieg
 
SF
Hope
 
TJ
, et al. 
Circulating human CD4 and CD8 T cells do not have large intracellular pools of CCR5.
Blood
2011
, vol. 
118
 
4
(pg. 
1015
-
1019
)
2
Portalès
 
P
Fabre
 
S
Vincent
 
T
, et al. 
Peripheral blood T4 cell surface CCR5 density as a marker of activity in rheumatoid arthritis treated with anti-CD20 monoclonal antibody.
Immunology
2009
, vol. 
128
 
1 suppl
(pg. 
e738
-
e745
)
3
Khurana
 
S
Kennedy
 
M
King
 
LR
Golding
 
H
Identification of a linear peptide recognized by monoclonal antibody 2D7 capable of generating CCR5-specific antibodies with human immunodeficiency virus-neutralizing activity.
J Virol
2005
, vol. 
79
 
11
(pg. 
6791
-
6800
)
4
Liu
 
R
Paxton
 
WA
Choe
 
S
, et al. 
Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection.
Cell
1996
, vol. 
86
 
3
(pg. 
367
-
377
)
5
Willingham
 
MC
Fluorescence labeling of surface antigens of attached or suspended tissue-culture cells.
Methods Mol Biol
2010
, vol. 
588
 (pg. 
143
-
151
)
6
Lin
 
Y-L
Mettling
 
C
Portalès
 
P
, et al. 
The chemokine CCL5 regulates the in vivo cell surface expression of its receptor, CCR5.
AIDS
2008
, vol. 
22
 
3
(pg. 
430
-
432
)
7
Lin
 
Y-L
Mettling
 
C
Portalès
 
P
, et al. 
G-protein signaling triggered by R5 human immunodeficiency virus type 1 increases virus replication efficiency in primary T lymphocytes.
J Virol
2005
, vol. 
79
 
12
(pg. 
7938
-
7941
)
Sign in via your Institution