Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • Rights and Permissions
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • My alerts
  • Log in
  • Log out
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • Rights and Permissions
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

The Primate Lentiviral Receptor Bonzo/STRL33 Is Coordinately Regulated with CCR5 and Its Expression Pattern Is Conserved Between Human and Mouse

Derya Unutmaz, Wenkai Xiang, Mary Jean Sunshine, Jim Campbell, Eugene Butcher and Dan R. Littman
J Immunol September 15, 2000, 165 (6) 3284-3292; DOI: https://doi.org/10.4049/jimmunol.165.6.3284
Derya Unutmaz
Molecular Pathogenesis Program, Skirball Institute of Biomolecular Medicine, New York University Medical Center, and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wenkai Xiang
Molecular Pathogenesis Program, Skirball Institute of Biomolecular Medicine, New York University Medical Center, and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Mary Jean Sunshine
Molecular Pathogenesis Program, Skirball Institute of Biomolecular Medicine, New York University Medical Center, and Howard Hughes Medical Institute, New York, NY 10016; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jim Campbell
Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Eugene Butcher
Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Dan R. Littman
Molecular Pathogenesis Program, Skirball Institute of Biomolecular Medicine, New York University Medical Center, and Howard Hughes Medical Institute, New York, NY 10016; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Chemokines play necessary and important roles in regulating the trafficking of lymphocytes to intra- or interlymphoid tissues as well as to sites of inflammation. The complex migratory patterns of lymphoid lineage cells is governed by subset-specific expression of chemokine receptors and their access to specific ligands. Several chemokine receptors and chemokine receptor-like orphan receptors also serve, in conjunction with CD4, as coreceptors for infection by human and simian immunodeficiency viruses (HIV and SIV). Here we show that the expression pattern of Bonzo/STRL33, an orphan SIV/HIV coreceptor, is highly restricted to the memory subset of T cells and is up-regulated upon stimulation of these cells with IL-2 or IL-15. Both the pattern and the regulation of Bonzo expression closely paralleled that of CC family chemokine receptors CCR5 or CCR6 and inversely correlated with CXCR4 expression. However, in striking contrast to CCR5, Bonzo expression was not down-modulated by PMA or mitogen stimulation of T cells. Targeted replacement of the Bonzo gene with a gene encoding green fluorescent protein in mice revealed that the expression and cytokine regulation of mouse Bonzo are comparable to those of its human counterpart. The similar expression and regulation patterns of Bonzo and the HIV coreceptor CCR5 may have implications for understanding the role of HIV/SIV receptors in viral evolution and pathogenesis.

Chemokine receptors belong to a subset within the superfamily of seven-transmembrane domain, G protein-coupled receptors that generally function to direct the complex migratory or trafficking patterns of leukocytes (1, 2). Most chemokines are small secreted proteins that are grouped according to the highly conserved position of cysteine residues within their N-terminal region into C, CC, CXC, and CX3C families (3, 4, 5). Although chemokine receptors often exhibit multiple ligand specificities, this promiscuity is generally limited to the binding of chemokines within the same family (2). Because of this, chemokine receptors are classified based on the family of chemokines that they bind. Greater sequence homology also exists within each of the CC and CXC families of chemokine receptors than between the two families. Moreover, the CC chemokine receptors CCR1-CCR5, CCR8, CCR9/10, and CX3CR1 are closely linked on chromosome 3p21 (6, 7, 8, 9, 10).

Members of the chemokine receptor family also serve as coreceptors, in conjunction with the CD4 molecule, for entry of HIV and SIV into target cells (11, 12, 13, 14, 15, 16). CCR5 is the major coreceptor for R5 strains (previously referred as M-tropic) of HIV-1 and most SIV strains, while CXCR4 allows entry of X4 strains (previously referred as T-tropic) of HIV-1 (11, 12, 13, 14, 15, 16). Additionally, CCR2, CCR3, CCR8 and CX3CR1 have been reported to be used by some of the HIV/SIV isolates, albeit at lower efficiencies (17, 18, 19). Several chemokine-receptor-like orphan receptors have also been shown to function as coreceptors for HIV and SIV strains (20, 21, 22, 23). One of these seven-transmembrane domain orphan receptors, Bonzo (also named STRL33 or TYMSTR), was identified as a principal coreceptor for several strains of SIV (including SIVagm and SIVsm family viruses) as well as some HIV-2 and HIV-1 strains (20, 21, 22, 23). Bonzo is a putative chemokine receptor based on sequence homology with other chemokine receptor family members. However, its biologic function is not understood, and no natural ligand has yet been identified among the known human chemokines (21) (T. Schall, unpublished observation). Expression of Bonzo mRNA is restricted to lymphoid tissues, PBMC, and placenta (20, 21, 22, 23). Interestingly, the Bonzo gene maps to human chromosome 3 close to the region where genes encoding CC family chemokine receptors are clustered (21). Little is known about the cell subset-specific expression and regulation of Bonzo. The in vivo distribution of Bonzo-expressing cells and the modulation of this expression may be important in determining the role of Bonzo in SIV and HIV pathogenesis. To address this issue we have used a mAb specific for Bonzo to demonstrate that expression of this receptor is highly restricted to the memory subset of resting human T cells. We further show that, similar to CCR5, Bonzo expression is up-regulated by the cytokines IL-2 and IL-15. However, in contrast to CCR5, upon PMA treatment or TCR stimulation Bonzo was not down-modulated. A similar expression pattern was observed on mouse CD4+ T cells of mice in which a green fluorescent protein (GFP)4 gene was knocked into the endogenous Bonzo locus.

Materials and Methods

Generation of mAb against Bonzo

An mAb against Bonzo/STRL33 was raised at R&D Systems (Minneapolis, MN) by immunizing BALB/c mice with a syngeneic mouse myeloma (NSO) transfected with full-length human Bonzo/STRL-33 with a polyhistidine fused to the C-terminal end of the receptor sequence. The polyhis serves as an epitope tag that can be used to identify (via intracellular staining in FACS or via Western blot) transfected clones that appear to be expressing high levels of the gene of interest. An immunization protocol (24) for soluble protein was adapted for use with whole cells as the immunogen. The priming immunization was performed by mixing the cell suspension in PBS with an equal volume of emulsified MPL/TDM adjuvant (Ribi); subsequent boosts used cells in PBS alone. Following immunization, lymph node cells were used for polyethylene glycol-mediated fusion following conventional protocols. After 7 days of culture, supernatants were screened for Abs that could bind to paraformaldehyde-fixed NSO/STRL-33/polyHis cells used for immunization. Cultures that were positive in this primary screen were then tested for binding to NSO cells that had been transfected with an irrelevant gene (GDF-9) also expressed as a polyhis construct. One clone was chosen and subcloned based on strong binding of its supernatant to the unfixed relevant transfectants. This hybridoma secretes an IgG2b κ mAb that was purified and used in subsequent experiments. This Ab is designated MAB699.

Preparation of human PBMC and resting T cells

PBMC were separated from buffy coats of healthy donors (New York Blood Bank, New York, NY) through Ficoll-Hypaque (Pharmacia, Uppsala, Sweden). Purification of T cells was performed as previously described (25). Briefly, monocytes were first removed from PBMC by plastic adherence for 2 h at 37°C. Nonadherent cells were incubated with anti-CD4 or anti-CD8 conjugated with Dynabeads (Dynal, Oslo, Norway) at a 1:4 target/bead ratio. The bead-bound cells were recovered using a magnet (Dynal) washed at least four times to remove unbound cells. The CD4+ or CD8+ cells were detached from the beads using Detachabead according to the manufacturer’s instructions (Dynal). These cells were then incubated with anti-HLA-DR Ab followed by Dynabeads conjugated with goat anti-mouse IgG for magnetic removal of preactivated T cells and contaminating dendritic cells or macrophages.

Media, reagents, and T cell cultures

The culture medium used in all experiments was RPMI (Life Technologies, Grand Island, NY) supplemented with 10% FCS (HyClone, Logan, UT), penicillin (50 U/ml; Life Technologies), streptomycin (50 μg/ml), sodium pyruvate (1 mM; Life Technologies), and glutamine (2 mM; Life Technologies). T cell lines were prepared by activation of purified resting T cells with allogeneic PBMC and were treated with 50 μg/ml mitomycin C (Sigma) for 30 min at 37°C and 5 μg/ml PHA (Sigma). Cells were split 3 days postactivation, expanded, and maintained in culture medium supplemented with 200 U/ml recombinant IL-2 (Chiron). T cell lines were maintained by restimulating the cells every 2 wk with PHA and mitomycin C-treated allogeneic PBMCs. Culture of T cells with cytokines has been previously described (26, 27). Cytokines IFN-γ, IL-4, IL-7, IL-12, and IL-15 and chemokines RANTES, macrophage inflammatory protein-1α (MIP-1α), MIP-1β, and stromal cell-derived factor 1α (SDF-1α) were all obtained from R&D Systems.

Ab staining and FACS analyses

Cells were incubated with the relevant Ab on ice for 30 min in PBS buffer with 2% FCS and 0.1% sodium azide. For staining of Bonzo, cells were incubated with anti-Bonzo mAb at 3 μg/ml; after two washes, cells were incubated with goat anti-mouse IgG conjugated to PE or tricolor (TC) (Caltag, South San Francisco, CA). The cells were washed twice again and blocked with excess mouse IgG (100 μg/ml) followed by staining with directly conjugated Abs against the relevant cell surface molecules. The Abs used for staining were PE, FITC, TC, or peridin chlorophyll protein conjugates of anti-human CCR5 and CXCR4; anti-mouse CD3, CD4, CD8, CD44, CD45RB, and γδTCR (PharMingen, San Diego, CA); anti-human CD4 and CD45RO, and secondary Abs goat anti-mouse PE or FITC (Caltag); anti-human CD3, CD8, CD14, CD16, CD19, CD45RA, HLA-DR, and γδTCR (all from Becton Dickinson, Palo Alto, CA); or anti-human CCR6 (R&D Systems). Staining was analyzed on a FACScan using CellQuest software (Becton Dickinson). Live cells were gated based on forward and side scatter. Intracellular staining was performed using Cytofic/Cytoperm solution according to the manufacturer’s protocol (PharMingen). To perform FACS analyses on fresh human thymocytes, thymi were obtained from 7- to 9-mo-old pediatric heart surgery cases. Thymic tissue was disrupted by mincing and forcing through stainless steel mesh. Thymocytes were incubated twice at 37°C for 30 min in complete medium to remove adherent cells. Single-cell suspensions were then placed on ice and stained for FACS analysis as described above.

Gene targeting in embryonic stem cells and generation of mice

A 129/Sv mouse genomic DNA library was screened with human Bonzo cDNA and a 16-kb DNA fragment was isolated. Sequence analysis identified a 1.2-kb intronless open reading frame (ORF) homologous to that of the human Bonzo gene. To generate an EGFP (Clontech) knockin targeting vector, three DNA fragments were sequentially inserted into a pBS-KS+ plasmid: 1) a GFP expressing cassette followed by SV40 poly(A)n sequence, 2) a neomycin resistance cassette (neoR) flanked with loxP sites (28), and 3) a HSV thymidine kinase cassette (HSV-TK). Subsequently, an 8-kb ApaI-NotI genomic fragment downstream of the Bonzo ORF was inserted between the neoR and HSV-TK cassettes, whereas a 1.6-kb genomic fragment upstream of the Bonzo ORF and containing the 5′-untranslated region was inserted 5′ of the GFP ORF. The resulting targeting vector was linearized with ClaI and electroporated into 129/Sv-derived E14 embryonic stem (ES) cells. G418-resistant ES cell clones were then screened for homologous recombination. To eliminate possible interference from the neo gene, correctly targeted clones were electroporated with a Cre recombinase-expressing vector (pCMV-Cre) to delete the neoR cassette, which was confirmed by sensitivity to G418 and by PCR or Southern blotting for the absence of the neoR-coding region. The resulting clones were microinjected into C57BL/6 blastocysts. Chimeric mice were mated with wild-type C57BL/6 mice to produce heterozygous progeny. Six- to 8-wk-old littermates from the mating of heterozygous mice were then analyzed.

RT-PCR analysis

Total RNA was extracted from mouse lymph nodes and spleen using Trizol reagent (Life Technologies). The RNA was further treated with RNase-free DNase I (Roche, Indianapolis, IN). RNA (1 μg) was used in an Access RT-PCR system (Promega, Madison, WI) with 50 pmol of sense (GCT TGC TCA TTT GGG TG) and antisense (CGC CGC GTC GAC CTT CTC TAA GTG TGG CAA GGC) Bonzo primers. RT was performed for 45 min at 48°C, and cDNA amplification was conducted for 40 cycles at 94°C for 30 s, 58°C for 1 min, and 68°C for 2 min. To exclude possible contamination with genomic DNA, control reactions in which reverse transcriptase was omitted were performed in parallel.

Fluorescence microscopy

Mouse organs were fixed in 4% formaldehyde for at least 12 h and incubated in 15% sucrose in PBS for 1 h. The samples were embedded in HistoPrep (Fisher, Fairlawn, NJ) and later frozen with dry ice. Samples were cut into 15-μm sections with a cryostat. Fluorescence microscopy was used to visualize GFP expression.

Results

Bonzo expression pattern on PBMC

An mAb reacting against human Bonzo was obtained as described in Materials and Methods. This Ab binds specifically to HOS cells that stably express the Bonzo cDNA (Fig. 1⇓a) (20). Similar results were obtained using Bonzo-transduced NIH-3T3 cells or a mouse thymoma line (data not shown). In addition, 293T cells were transfected with a plasmid encoding Bonzo-GFP fusion and stained with anti-Bonzo Ab. Only Bonzo-GFP-expressing cells were positively stained with anti-Bonzo mAb, and this correlated with GFP expression. To exclude the cross-reactivity of the Ab with other chemokine receptors, we also stained HOS cell lines stably expressing CCR1, CCR3, CCR4, CCR7, CCR8, APJ, and V28. None of these cell lines was stained with anti-Bonzo mAb (data not shown). We then determined the expression pattern of Bonzo on PBMC of normal donors using multicolor FACS analysis. Expression of Bonzo was found to segregate primarily to CD3+ T cells (Fig. 2⇓a). Both CD4+ and CD8+ T cell subsets expressed Bonzo (Fig. 2⇓, b and c), although the percentage of Bonzo+ cells was always higher within the CD8+ subset in samples obtained from different donors (data not shown). T cells can also be subdivided into memory and naive subsets based on expression of CD45 isoforms, CD45RO+RA− and CD45RO−RA+, respectively (29). Bonzo was exclusively expressed on CD45RO+ T cells (Fig. 2⇓d). About 30–40% of γδ T cells also expressed Bonzo (Fig. 2⇓f). The few CD3-negative cells that expressed Bonzo were CD16+ NK cells (Fig. 2⇓e), and no expression was detected on B cells, monocytes, or dendritic cells (Fig. 2⇓, g, h, and i, respectively).

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Bonzo Ab specifically stains transfected cells. a, HOS cells stably expressing Bonzo were generated by retroviral transduction of the Bonzo or CCR5 cDNA using the pBabe vector (20 ). Cells were stained with the anti-Bonzo mAb followed by a PE-conjugated goat-anti-mouse Ab. b, 293T cells were transfected with a pcDNA3 plasmid encoding either GFP or Bonzo-GFP fusion protein. Cells were then stained, 48 h post-transfection, with Bonzo Ab as described above.

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Expression of Bonzo on PBMC. Freshly isolated PBMC were stained with anti-Bonzo mAb followed by the PE-conjugated secondary Ab and were subsequently stained with PE-conjugated Abs specific for other cell surface molecules. Three donors gave similar results, and data from one donor are shown. Numbers in quadrants are percent positives. Expression of Bonzo on a) T cells (CD3+); b and c) CD4+ and CD8+ T cell subsets; d) memory (CD4RO+) and naive (CD45RO−) T cells (gated for CD3+ cells); e) NK cells (CD16+); f) γδ T cells (gated on CD3+ cells); g) B cells (CD19+); h, monocyte expression (CD14+); and i) dendritic cells. The dendritic cells were identified as follows: PBMC were first stained with anti-Bonzo Ab as described above and then with FITC-conjugated anti-CD3, -CD14, and -CD19 and TC-conjugated anti-HLA-DR Abs. A gate was set to exclude FITC-positive cells (T cells, B cells, and monocytes). The remaining HLA-DR+ cells identify DC.

It has been reported that expression of some chemokine receptors, such as the major HIV coreceptor CCR5, is also restricted to the memory subset of T cells (30). As shown in Fig. 3⇓, the expression profile of CCR5 on resting T cells very closely parallels that of Bonzo. Indeed, costaining of PBMC with Abs specific for Bonzo and CCR5 showed that most Bonzo-positive cells were also CCR5+ (Fig. 4⇓). PBMC from a healthy donor with a mutation in the CCR5 gene that prevents its cell surface expression (31) exhibited normal expression of Bonzo (Fig. 3⇓b), demonstrating that absence of CCR5 from the cell surface does not negatively influence Bonzo expression. Most Bonzo-positive resting T cells also expressed CCR6 (Fig. 4⇓), whereas expression of Bonzo and CXCR4 was inversely correlated (Fig. 4⇓). This inverse expression pattern reflects lower levels of CXCR4 on memory T cells (30) (data not shown), which preferentially express Bonzo (Fig. 2⇑d).

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Bonzo and CCR5 are expressed on similar T cell subsets. PBMC were stained with mAbs against either Bonzo or CCR5 and then with secondary PE-conjugated Ab followed by CD3-TC and FITC-conjugated anti-CD8, -CD4, or -CD45RO mAbs. Gating was restricted to CD3+ cells. a, CCR5 expression on T cell subsets and comparative staining of Bonzo (bottom panel). b, Bonzo expression of T cell subsets from a CCR5-null individual.

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Coexpression of Bonzo with CCR5. PBMC were stained with Bonzo as previously described, followed by PE-conjugated anti-CCR5, -CCR6, or -CXCR4 and TC-conjugated anti-CD3 mAbs. Profiles are gated on CD3+ cells.

We also analyzed the expression of Bonzo within the thymus and secondary lymphoid organs, adenoids and tonsils. Bonzo, CCR5, and CCR6 expression was either undetectable or was present on <1% of thymocytes, whereas the majority of the thymocytes expressed CXCR4 (data not shown). Expression profiles of Bonzo, CCR5, and CCR6 on the lymphocytes isolated from tonsils or adenoids were similar to those observed on PBMC, although a greater proportion of T cells was positive, and most of these cells expressed CD45RO+ as well as the early activation marker CD69 (data not shown).

Regulation of Bonzo expression by cytokine stimulation of resting T cells

The concordant subset distribution of Bonzo and CCR5 suggested that the surface expression of these molecules may be regulated by similar mechanisms. It has been noted that IL-2 or IL-15 stimulation of human T cells can up-regulate some of the CC chemokine receptors (27, 32, 33, 34). We asked whether Bonzo is similarly regulated by cytokines on resting human T cells. CD4+ and CD8+ resting T cells were purified from PBMC and cultured for 8 days in the presence of various cytokines. Culture of CD4+ or CD8+ T cells with IL-2 or IL-15 resulted in up-regulated expression of Bonzo exclusively on CD45RO+ memory T cells (Fig. 5⇓), with expression on CD8+ T cells usually greater than that on CD4+ T cells. This result is consistent with the staining pattern observed on freshly isolated PBMC. Little expression was observed on CD4+ T cells cultured with IL-4 or IL-7 (Fig. 5⇓), and none was found with IL-6, IL-12, or IFN-γ (data not shown). However, significant up-regulation of Bonzo was observed on CD8+ T cells cultured with IL-7 (Fig. 5⇓, lower panel). IL-2 and IL-15 also up-regulated CCR5 and CCR6 expression on CD4+ T cells, and most Bonzo-expressing cells also coexpressed these CC chemokine receptors (Fig. 6⇓). Similar results were obtained with CD8+ T cells (data not shown). This result recapitulates the coordinate expression profile of Bonzo with CCR5 or CCR6 in freshly isolated PBMC (Fig. 4⇑). Conversely, Bonzo-positive cells had slightly lower CXCR4 expression than Bonzo-negative cells after stimulation with IL-2 or IL-15 (Fig. 6⇓). CXCR4 expression was significantly up-regulated in the presence of IL-4, and there was also a moderate increase in response to IL-2 or IL-15, in accordance with previous reports (35, 36, 37).

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

Cytokine-mediated regulation of Bonzo and CCR5 on resting CD4+ and CD8+ T cells. Purified CD4+ or CD8+ resting T cells were cultured for 8 days in the presence of the cytokines IL-2 (200 U/ml), IL-4 (20 ng/ml), IL-7 (10 ng/ml), and IL-15 (10 ng/ml). Cells were then stained with mAbs specific for Bonzo and for CD45RO and CD4 or CD8 (top and lower panels, respectively).

FIGURE 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 6.

Cytokine-mediated coregulation of Bonzo and chemokine receptors CCR5, CCR6, or CXCR4 on resting T cells. Purified CD4+ T cells were cultured with cytokines as described in Fig. 5⇑. Cells were stained for Bonzo, CCR5, CCR6, or CXCR4 expression as described in Fig. 4⇑.

Post-transcriptional regulation of Bonzo cell surface expression

Most chemokine receptors contain a DRY sequence motif that appears to be required for coupling to G proteins (38). In contrast to other CC and CXC-family chemokine receptors, Bonzo possesses a noncanonic DRY box sequence at the second intracellular loop. This raised the possibility that in response to its physiologic ligand Bonzo may not signal in a similar fashion to the other chemokine receptors. Recently, it has been reported that chemokine receptors are down-regulated through ligand-mediated endocytosis (39, 40, 41, 42). Therefore, we reasoned that if any of the CCR5 ligands also bound to Bonzo, we may be able to detect this through down-regulation of Bonzo in the absence of detectable signaling. Fig. 7⇓a shows that none of the known CCR5 ligands (MIP-1α, MIP-1β, and RANTES) had any effect on Bonzo expression, whereas they completely down-modulated CCR5 at the same concentrations. As expected, CXCR4 was down-modulated by its ligand SDF-1α, but not by CCR5 ligands (Fig. 7⇓a). Interestingly, stimulation of cells with phorbol ester (PMA) also did not have any effect on Bonzo expression, although CCR5 and CXCR4 expression was completely down-modulated (Fig. 7⇓b). Similarly, upon restimulation of T cell lines with mitogen, CCR5 expression was down-modulated, but that of Bonzo was unaffected (Fig. 7⇓c). To determine whether Bonzo is premade, stored in intracellular compartments, and released to cell surface upon signaling, we performed intracellular staining on resting or cytokine-stimulated primary T cells with anti-Bonzo Ab. No difference was observed between cell surface staining alone or in conjunction with intracellular staining (data not shown).

FIGURE 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 7.

Effects of chemokines, PMA, and mitogen stimulation on Bonzo, CCR5, and CXCR4 cell surface expression. To maximize expression levels, CD4+ cells cultured with IL-15 for 12 days were used in the Bonzo and CCR5 down-regulation experiments, and IL-4-cultured cells were used for examining CXCR4 down-regulation. a, Cells were cultured for 24 h in the presence of the chemokines RANTES, MIP-1α, MIP-1β, or SDF-1α (1 μg/ml each) before staining. b, Cells were stimulated with 15 ng/ml PMA for 12 h and stained for chemokine receptor expression. c, Activated CD4+ human T cell lines maintained in IL-2 were stained for chemokine receptor expression 2 wk poststimulation (upper panel) or 3days after restimulation through TCR (lower panel).

Targeted disruption and knockin of EGFP into the mouse Bonzo gene locus

To gain insight into the expression pattern and function of Bonzo in mice, we used a gene-targeting strategy that replaced the Bonzo-coding sequence with that of EGFP (Fig. 8⇓A) Germline transmission was obtained with two independent ES cell clones. Deletion of the Bonzo gene was verified by Southern blot analysis (Fig. 8⇓B). In an RT-PCR assay, a 3′ portion of Bonzo mRNA was amplified from total RNA derived from lymph node and spleen of both Bonzo+/− (Bz+/−) and Bonzo−/− (Bz−/−) mice. No amplification was observed from Bz−/− mice (Fig. 8⇓C). Mating of heterozygous mice gave rise to Bz−/− mice in Mendelian proportions. Bz−/− mice were phenotypically indistinguishable from Bz+/+ and Bz+/− littermates in a specific pathogen-free environment. Histologic analysis of organs displayed no morphologic difference among the three genotypes. Flow cytometric analysis of cells from lymphoid organs showed no alteration in various cell populations (data not shown). Proliferation of T cells from Bz+/− or Bz−/− mice was assessed in response to different concentrations of anti-CD3 Ab in the presence or the absence of anti-CD28 Ab. No difference was observed between Bz−/+ and Bz−/− cells (data not shown). Nucleoprotein-keyhole limpet hemocyanin-immunized Bz+/− and Bz−/− mice also displayed similar levels of nucleoprotein-specific IgM and IgG1 production (data not shown), suggesting that T cell help and B cell function were normal in Bonzo-deficient mice.

FIGURE 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 8.

Targeted knockin of EGFP in the mouse Bonzo gene. A, From the top, the wild-type Bonzo genomic structure, the targeting construct, and the targeted Bonzo/EGFP knockin structure, respectively. The arrows above each gene depict the directions of transcription. ▵, loxP sites. H, HindIII; A, ApaI; RV, EcoRI; C, ClaI. B, Southern blot analysis of progeny of heterozygous intercrossing. The probe used is depicted by the solid bar beneath the targeted structure in A. The wild-type allele yields a 7-kb EcoRV genomic fragment, whereas the targeted allele yields a 6.6-kb fragment. C, RT-PCR analysis of expression of Bonzo mRNA in mutant mice. The primers used are described in Materials and Methods. In a TAE gel, a 590-bp DNA fragment is amplified from wide-type (top panel, lane 2) and heterozygous (lane 3) mice, but is absent in Bz−/− mice (lane 4). A similar RT-PCR assay was conducted for GADPH mRNA, as a loading control (lower panel).

Tracing mouse Bonzo expression in EGFP knockin mice

The in-frame substitution of EGFP for Bonzo yields a sensitive tool to trace Bonzo expression and regulation in vivo. Histologic analysis did not show any GFP expression in nonlymphoid tissues. Strong green fluorescence was detected in spleen and lymphoid nodes. In spleen, GFP-positive cells were clustered in the periarteriolar lymphoid sheath area in the white pulp, where T cells reside (Fig. 9⇓). Notably, there was no difference in expression pattern between Bz+/− and Bz−/− mice.

FIGURE 9.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 9.

Expression of GFP in spleen sections of Bonzo knockin mice. The section shows GFP fluorescence in T cell-rich periarteriolar lymphoid sheath area from Bonzo knockin mice.

We next performed FACS analysis of GFP expression in different lymphoid subsets. GFP expression was undetectable in B cells, granulocytes, monocytes or macrophage, and dendritic cells (data not shown). In thymus, no expression was observed in CD4+CD8+ cells or double-negative thymocytes. Only a small subset of single-positive cells showed strong expression (data not shown).

Within the lymph nodes and spleen almost all GFP+ cells were CD3+ T cells (Fig. 10⇓a). The mean fluorescence of GFP-expressing Bz−/− cells was slightly higher than that of Bz+/− cells, most likely due to GFP expression from both targeted alleles (data not shown). Few CD4+ T cells expressed GFP, whereas the majority of CD8+ T cells were positive, with a broad distribution of intensity of GFP expression. We next analyzed the distribution of GFP expression on naive vs memory T cells in Bonzo knockin mice. CD8+ cells were all CD44+, and it was difficult to discriminate naive vs memory cells (Fig. 10⇓b). γδ T cells from lymphoid organs or resident within the skin, reproductive tract, or gut also expressed GFP (Fig. 10⇓c). Interestingly, GFP expression on CD4+ murine T cells was restricted to the CD44+ and CD45RBlow subset (Fig. 10⇓d), suggesting that, as in human CD4+ T cells, Bonzo is primarily expressed in the memory subset of mouse CD4+ T cells.

FIGURE 10.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 10.

GFP expression on lymphocyte subsets from Bonzo knockin mice. FACS analysis of T cell subsets from lymph nodes of Bonzo-GFP knockin mice was performed. a, GFP expression on CD3+ T cells; b, cells stained with anti-mouse CD8-TC and CD44-PE, gated on CD8+ T cells; c, IEL stained with PE-conjugated anti-TCRγδ Ab; d, GFP expression on naive and memory mouse CD4 T cells. Lymph node cells stained with CD4-TC and with either CD44-PE or CD45RB-PE and analyzed after gating was set on CD4+ cells.

Finally we determined whether, similar to human Bonzo, expression of mouse Bonzo can be modulated by the presence of cytokines. Indeed, we observed dramatic up-regulation of GFP expression after 10 days of culture with IL-2, but less with IL-7 and none with IL-4. These results suggest that cytokine-mediated regulation of murine Bonzo follows the pattern of its human counterpart (Fig. 11⇓).

FIGURE 11.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 11.

Regulation of mouse Bonzo expression by cytokines. CD4+ T cells were purified from lymph nodes of Bz+/− and Bz−/− mice. Cells (4 × 105) were treated with cytokines (IL-2, IL-7, and IL-4) or activated by plate-bound CD3 Ab and assayed for GFP expression 3 and 10 days later. Most GFP-expressing cells were CD44high. The result is plotted as the percentage of CD4+ T cells that are GFP positive.

Discussion

The discovery that HIV and SIV use chemokine receptors to enter target cells raised hopes that chemokines and chemokine receptors might hold clues to understanding the progression of HIV-mediated pathogenesis. These discoveries also bolstered the general interest in the regulation of lymphocyte migration by chemokine receptors. The recent development of Abs to chemokine receptors has revealed the complex subset-specific expression of these receptors on T lymphocytes (43). In this report we demonstrate that the expression pattern and regulation of the SIV/HIV coreceptor and putative orphan chemokine receptor Bonzo strongly parallels those of CC family chemokine receptors.

It has been shown that CC family chemokine receptors CCR2, CCR5, and CCR6 are expressed primarily on the CD45RO+ or CD26+ memory subset of T cells (30, 44, 45, 46). The expression pattern of Bonzo was similarly restricted to memory T cells. In this regard we found that most Bonzo-positive cells also coexpressed CCR5 or CCR6 on peripheral blood T cells. In contrast, Bonzo was not coexpressed with high levels of CXCR4, reflecting lower CXCR4 expression on memory vs naive T cells (30). Although Bonzo was expressed on CD4+, CD8+ and γδ subsets of T cells, expression levels were usually higher on CD8+ and TCRγδ+ T cells. Few NK cells were found to express Bonzo, and those that did had relatively low levels of CD16 expression (Fig. 2⇑e), possibly representing recently activated NK cells.

The coexpression of Bonzo and CCR5 on memory T cells is notable from the perspective of HIV coreceptor usage, because CCR5 is the major receptor used by most strains of HIV and almost all SIVs. Expression of CCR5 on the cell surface has been shown to be critical in transmission of HIV-1 infection, because a homozygous 32-bp deletion in the CCR5 gene prevents CCR5 expression and confers resistance against HIV-1 infection (31, 47, 48). We showed that T cells isolated from individuals who are homozygous for the CCR5Δ32 mutation expressed normal levels of Bonzo. Although Bonzo is a minor HIV-1 coreceptor, its role during transmission of infection and pathogenesis is unclear. However, Bonzo is a major SIV receptor and is also used by many HIV-2 strains in vitro (18, 20). It is interesting to note that different species of nonhuman primates vary widely in their responses to SIV infection, and people infected with HIV-2 usually have a more delayed progression to AIDS. It is possible that Bonzo may substitute for CCR5 usage during infection with SIV and HIV-2 isolates and perhaps influences the course of infection and pathogenesis. In contrast to CCR5, Bonzo was not expressed by macrophages or dendritic cells. This expression pattern may preclude its involvement during the initial phase of infection, where macrophage or dendritic cell infection is thought to be necessary for the virus to gain a foothold in the body (49).

Cytokine stimulation of T cells differentially regulates the surface expression of chemokine receptors. IL-2 has been described to potently up-regulate CCR1, CCR2, CCR5, CCR6, and CXCR3 (27, 30, 32, 50, 51). More recently, IL-15 has been shown to have similar effects on some of the CC chemokine receptors (27, 33). In contrast, CXCR4 expression is dramatically up-regulated in the presence of IL-4 (35, 36, 37). We have shown that Bonzo, similar to CC chemokine receptors, is induced through stimulation of highly purified resting T cells with IL-2 or IL-15 and is coexpressed with CCR5 or CCR6. The coordinated regulation of Bonzo and CCR5 on resting T cells by IL-2 or IL-15 recapitulates the ex vivo expression pattern.

Notably, the gene for Bonzo maps to chromosome 3, where most of the chemokine receptors, especially those of the CC family, are localized (21). Additionally, the amino acid sequence of Bonzo is similar to that of other members of the chemokine receptor family. Taken together with our results on its expression pattern and cytokine-mediated regulation, these findings suggest that Bonzo is also a chemokine receptor. However, Bonzo stands apart from other chemokine receptors, because its expression is resistant to PMA-induced down-modulation. This result contrasts with PMA-induced down-modulation of CXCR4 (52, 53), CCR5 (Fig. 7⇑b), or CCR6 (data not shown). However, Bonzo may still be down-modulated by its ligand(s), because the mechanisms of ligand- and PMA-induced down-modulation appear to be different (42, 52, 53). We did not detect any ligand-induced down-modulation of Bonzo in the presence of CCR5 ligands, but we cannot rule out that Bonzo is resistant to ligand-induced endocytosis and thus may still be able to bind to these chemokines.

Because there is no Ab yet available against mouse Bonzo, we used the targeted knockin of GFP in place of the Bonzo gene as an indicator of Bonzo expression. We found that murine Bonzo is expressed in cell subsets similar to human Bonzo, namely in T cells, particularly those displaying markers of effector/memory cells and those stimulated by IL-2 (data not shown). However, in both mouse and human only a subset of memory cells expressed Bonzo, similar to CCR5 expression. Chemokines regulate both the inter- and intraorgan migration patterns of human T cell subsets. Recently, human memory T cells were subdivided into two functionally distinct subsets based on CCR7 expression (54). CCR7− memory cells express receptors for migration to inflamed tissues and display immediate effector function. In contrast, CCR7+ memory cells express lymph node-homing receptors and lack immediate effector function. Interestingly, the CCR7− cells were enriched for expression of CCR5, CCR6, and CCR1 (54). Based on the similar expression patterns, we would predict that the majority of Bonzo+ T cells will be compartmentalized to the CCR7− memory subset, suggesting a role for Bonzo in recruiting memory T cells to sites of inflammation.

In homozygous Bonzo knockout mice we have not yet detected any functional defect. This may be due to redundancy in the chemokine receptor system. However, it will be important to perform more detailed analyses of migration patterns of T cells in response to a wide variety of pathogens and in particular to study T cell memory responses to the pathogens. Mutations in other chemokine receptor family members, such as CCR5 and CCR2, resulted in phenotypes revealed only after studies in responses to pathogen or in disease models (55, 56, 57, 58). It will also be interesting to establish multiple knockouts between the Bonzo mutant mice and mutants for other chemokine receptors expressed in memory T cells to identify potentially overlapping functions of these molecules.

Bonzo is unique in that its sequence bears a noncanonic DRY box motif that is thought to couple chemokine receptors to G proteins (18, 20). It is currently not known whether Bonzo elicits signals that are similar to those of other chemokine receptors. It will be critical to identify the natural ligand(s) of Bonzo to understand the receptor’s signaling function. In addition, mice in which the Bonzo gene has been replaced with EGFP may be of considerable utility for tracking the migration patterns of memory T cell in responses to inflammatory stimuli. New insight into the expression pattern and function of Bonzo may also help elucidate its role during SIV/HIV infection.

Acknowledgments

We thank Drs. Chris Arendt and Monica Tsang for their critical reading of the manuscript and helpful suggestions, Dr. Michael Black for providing the human thymus, Christie Doxsee for excellent technical help, and Dr. Vineet N. Kewal-Ramani for many helpful suggestions, critique, and reagents.

Footnotes

  • ↵1 This work was supported by National Institutes of Health Grants AI33856 and AI36606 to D.R.L.

  • ↵2 Address correspondence and reprint requests to Dr. Derya Unutmaz, Department of Microbiology and Immunology, Vanderbilt University Medical School, AA-5216 Medical Center North, Nashville, TN 37215. E-mail address: derya.unutmaz{at}mcmail.vanderbilt.edu

  • ↵3 D.R.L. is an Investigator with the Howard Hughes Medical Institute.

  • ↵4 Abbreviations used in this paper: GFP, green fluorescent protein; MIP, macrophage inflammatory protein; SDF-1α, stromal cell-derived factor; TC, tricolor; ORF, open reading frame; ES, embryonic stem.

  • Received June 16, 2000.
  • Accepted June 28, 2000.
  • Copyright © 2000 by The American Association of Immunologists

References

  1. ↵
    Murphy, P. M.. 1996. Chemokine receptors: structure, function and role in microbial pathogenesis. Cytokine Growth Factor Rev. 7: 47
    OpenUrlCrossRefPubMed
  2. ↵
    Ward, S. G., K. Bacon, J. Westwick. 1998. Chemokines and T lymphocytes: more than an attraction. Immunity 9: 1
    OpenUrlCrossRefPubMed
  3. ↵
    Schall, T. J., K. B. Bacon. 1994. Chemokines, leukocyte trafficking, and inflammation. Curr. Opin. Immunol. 6: 865
    OpenUrlCrossRefPubMed
  4. ↵
    Baggiolini, M., B. Dewald, B. Moser. 1997. Human chemokines: an update. Annu. Rev. Immunol. 15: 675
    OpenUrlCrossRefPubMed
  5. ↵
    Baggiolini, M.. 1998. Chemokines and leukocyte traffic. Nature 392: 565
    OpenUrlCrossRefPubMed
  6. ↵
    Raport, C. J., V. L. Schweickart, R. J. Eddy, T. B. Shows, P. W. Gray. 1995. The orphan G-protein-coupled receptor-encoding gene V28 is closely related to genes for chemokine receptors and is expressed in lymphoid and neural tissues. Gene 163: 295
    OpenUrlCrossRefPubMed
  7. ↵
    Samson, M., P. Soularue, G. Vassart, M. Parmentier. 1996. The genes encoding the human CC-chemokine receptors CC-CKR1 to CC-CKR5 (CMKBR1-CMKBR5) are clustered in the p21.3-p24 region of chromosome 3. Genomics 36: 522
    OpenUrlCrossRefPubMed
  8. ↵
    Daugherty, B. L., M. S. Springer. 1997. The β-chemokine receptor genes CCR1 (CMKBR1), CCR2 (CMKBR2), and CCR3 (CMKBR3) cluster within 285 kb on human chromosome 3p21. Genomics 41: 294
    OpenUrlCrossRefPubMed
  9. ↵
    Bonini, J. A., S. K. Martin, F. Dralyuk, M. W. Roe, L. H. Philipson, D. F. Steiner. 1997. Cloning, expression, and chromosomal mapping of a novel human CC-chemokine receptor (CCR10) that displays high-affinity binding for MCP-1 and MCP-3. DNA Cell Biol. 16: 1249
    OpenUrlCrossRefPubMed
  10. ↵
    Roos, R. S., M. Loetscher, D. F. Legler, I. Clark-Lewis, M. Baggiolini, B. Moser. 1997. Identification of CCR8, the receptor for the human CC chemokine I-309. J. Biol. Chem. 272: 17251
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Feng, Y., C. C. Broder, P. E. Kennedy, E. A. Berger. 1996. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science 272: 872
    OpenUrlAbstract
  12. ↵
    Alkhatib, G., C. Combadiere, C. C. Broder, Y. Feng, P. E. Kennedy, P. M. Murphy, E. A. Berger. 1996. CC CKR5: a RANTES, MIP-1α, MIP-1β receptor as a fusion cofactor for macrophage-tropic HIV-1. Science 272: 1955
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Deng, H., R. Liu, W. Ellmeier, S. Choe, D. Unutmaz, M. Burkhart, P. Di Marzio, S. Marmon, R. E. Sutton, C. M. Hill, et al 1996. Identification of a major coreceptor for primary isolates of HIV-1. Nature 381: 661
    OpenUrlCrossRefPubMed
  14. ↵
    Dragic, T., V. Litwin, G. P. Allaway, S. R. Martin, Y. Huang, K. A. Nagashima, C. Cayanan, P. J. Maddon, R. A. Koup, J. P. Moore, et al 1996. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 381: 667
    OpenUrlCrossRefPubMed
  15. ↵
    Doranz, B. J., J. Rucker, Y. Yi, R. J. Smyth, M. Samson, S. C. Peiper, M. Parmentier, R. G. Collman, R. W. Doms. 1996. A dual-tropic primary HIV-1 isolate that uses fusin and the β-chemokine receptors CKR-5, CKR-3, and CKR-2b as fusion cofactors. Cell 85: 1149
    OpenUrlCrossRefPubMed
  16. ↵
    Choe, H., M. Farzan, Y. Sun, N. Sullivan, B. Rollins, P. D. Ponath, L. Wu, C. R. Mackay, G. LaRosa, W. Newman, et al 1996. The β-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 85: 1135
    OpenUrlCrossRefPubMed
  17. ↵
    Rucker, J., A. L. Edinger, M. Sharron, M. Samson, B. Lee, J. F. Berson, Y. Yi, B. Margulies, R. G. Collman, B. J. Doranz, et al 1997. Utilization of chemokine receptors, orphan receptors, and herpesvirus-encoded receptors by diverse human and simian immunodeficiency viruses. J. Virol. 71: 8999
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Unutmaz, D., V. N. KewalRamani, D. R. Littman. 1998. G protein-coupled receptors in HIV and SIV entry: new perspectives on lentivirus-host interactions and on the utility of animal models. Semin. Immunol. 10: 225
    OpenUrlCrossRefPubMed
  19. ↵
    Choe, H., K. A. Martin, M. Farzan, J. Sodroski, N. P. Gerard, C. Gerard. 1998. Structural interactions between chemokine receptors, gp120 Env and CD4. Semin. Immunol. 10: 249
    OpenUrlCrossRefPubMed
  20. ↵
    Deng, H. K., D. Unutmaz, V. N. KewalRamani, D. R. Littman. 1997. Expression cloning of new receptors used by simian and human immunodeficiency viruses. Nature 388: 296
    OpenUrlCrossRefPubMed
  21. ↵
    Loetscher, M., A. Amara, E. Oberlin, N. Brass, D. Legler, P. Loetscher, M. D’Apuzzo, E. Meese, D. Rousset, J. L. Virelizier, et al 1997. TYMSTR, a putative chemokine receptor selectively expressed in activated T cells, exhibits HIV-1 coreceptor function. Curr. Biol. 7: 652
    OpenUrlCrossRefPubMed
  22. ↵
    Liao, F., G. Alkhatib, K. W. Peden, G. Sharma, E. A. Berger, J. M. Farber. 1997. STRL33, a novel chemokine receptor-like protein, functions as a fusion cofactor for both macrophage-tropic and T cell line-tropic HIV-1. J. Exp. Med. 185: 2015
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Alkhatib, G., F. Liao, E. A. Berger, J. M. Farber, K. W. Peden. 1997. A new SIV coreceptor, STRL33. Nature 388: 238
    OpenUrlCrossRefPubMed
  24. ↵
    Lucas, C., L. N. Bald, B. M. Fendly, M. Mora-Worms, I. S. Figari, E. J. Patzer, M. A. Palladino. 1990. The autocrine production of transforming growth factor-β1 during lymphocyte activation: a study with a monoclonal antibody-based ELISA. J. Immunol. 145: 1415
    OpenUrlAbstract
  25. ↵
    Unutmaz, D., V. N. KewalRamani, S. Marmon, D. R. Littman. 1999. Cytokine signals are sufficient for HIV-1 infection of resting human T lymphocytes. J. Exp. Med. 189: 1735
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Unutmaz, D., P. Pileri, S. Abrignani. 1994. Antigen-independent activation of naive and memory resting T cells by a cytokine combination. J. Exp. Med. 180: 1159
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Unutmaz, D., V. N. KewalRamani, S. Marmon, D. R. Littman. 1999. Cytokine signals are sufficient for HIV-1 infection of resting human T lymphocytes. J. Exp. Med. 189: 1735
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Gu, H., Y. R. Zou, K. Rajewsky. 1993. Independent control of immunoglobulin switch recombination at individual switch regions evidenced through Cre-lox P mediated gene targeting. Cell 73: 1155
    OpenUrlCrossRefPubMed
  29. ↵
    Beverley, P. C.. 1992. Functional analysis of human T cell subsets defined by CD45 isoform expression. Semin. Immunol. 4: 35
    OpenUrlPubMed
  30. ↵
    Bleul, C. C., L. Wu, J. A. Hoxie, T. A. Springer, C. R. Mackay. 1997. The HIV coreceptors CXCR4 and CCR5 are differentially expressed and regulated on human T lymphocytes. Proc. Natl. Acad. Sci. USA 94: 1925
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Liu, R., W. A. Paxton, S. Choe, D. Ceradini, S. R. Martin, R. Horuk, M. E. MacDonald, H. Stuhlmann, R. A. Koup, N. R. Landau. 1996. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 86: 367
    OpenUrlCrossRefPubMed
  32. ↵
    Loetscher, P., M. Seitz, M. Baggiolini, B. Moser. 1996. Interleukin-2 regulates CC chemokine receptor expression and chemotactic responsiveness in T lymphocytes. J. Exp. Med. 184: 569
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Perera, L. P., C. K. Goldman, T. A. Waldmann. 1999. IL-15 induces the expression of chemokines and their receptors in T lymphocytes. J. Immunol. 162: 2606
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Jinquan, T., S. Quan, G. Feili, C. G. Larsen, K. Thestrup-Pedersen. 1999. Eotaxin activates T cells to chemotaxis and adhesion only if induced to express CCR3 by IL-2 together with IL-4. J. Immunol. 162: 4285
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Jourdan, P., C. Abbal, N. Nora, T. Hori, T. Uchiyama, J. P. Vendrell, J. Bousquet, N. Taylor, J. Pene, H. Yssel. 1998. IL-4 induces functional cell-surface expression of CXCR4 on human T cells. J. Immunol. 160: 4153
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Wang, J., A. Harada, S. Matsushita, S. Matsumi, Y. Zhang, T. Shioda, Y. Nagai, K. Matsushima. 1998. IL-4 and a glucocorticoid up-regulate CXCR4 expression on human CD4+ T lymphocytes and enhance HIV-1 replication. J. Leukocyte Biol. 64: 642
    OpenUrlAbstract
  37. ↵
    Valentin, A., W. Lu, M. Rosati, R. Schneider, J. Albert, A. Karlsson, G. N. Pavlakis. 1998. Dual effect of interleukin 4 on HIV-1 expression: implications for viral phenotypic switch and disease progression. Proc. Natl. Acad. Sci. USA 95: 8886
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Gosling, J., F. S. Monteclaro, R. E. Atchison, H. Arai, C. L. Tsou, M. A. Goldsmith, I. F. Charo. 1997. Molecular uncoupling of C-C chemokine receptor 5-induced chemotaxis and signal transduction from HIV-1 coreceptor activity. Proc. Natl. Acad. Sci. USA 94: 5061
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Solari, R., R. E. Offord, S. Remy, J. P. Aubry, T. N. Wells, E. Whitehorn, T. Oung, A. E. Proudfoot. 1997. Receptor-mediated endocytosis of CC-chemokines. J. Biol. Chem. 272: 9617
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Amara, A., S. L. Gall, O. Schwartz, J. Salamero, M. Montes, P. Loetscher, M. Baggiolini, J. L. Virelizier, F. Arenzana-Seisdedos. 1997. HIV coreceptor downregulation as antiviral principle: SDF-1α-dependent internalization of the chemokine receptor CXCR4 contributes to inhibition of HIV replication. J. Exp. Med. 186: 139
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Alkhatib, G., M. Locati, P. E. Kennedy, P. M. Murphy, E. A. Berger. 1997. HIV-1 coreceptor activity of CCR5 and its inhibition by chemokines: independence from G protein signaling and importance of coreceptor downmodulation. Virology 234: 340
    OpenUrlCrossRefPubMed
  42. ↵
    Mack, M., B. Luckow, P. J. Nelson, J. Cihak, G. Simmons, P. R. Clapham, N. Signoret, M. Marsh, M. Stangassinger, F. Borlat, et al 1998. Aminooxypentane-RANTES induces CCR5 internalization but inhibits recycling: a novel inhibitory mechanism of HIV infectivity. J. Exp. Med. 187: 1215
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Sallusto, F., A. Lanzavecchia, C. R. Mackay. 1998. Chemokines and chemokine receptors in T-cell priming and Th1/Th2-mediated responses. Immunol. Today 19: 568
    OpenUrlCrossRefPubMed
  44. ↵
    Oin, S., G. LaRosa, J. J. Campbell, H. Smith-Heath, N. Kassam, X. Shi, L. Zeng, E. C. Buthcher, C. R. Mackay. 1996. Expression of monocyte chemoattractant protein-1 and interleukin-8 receptors on subsets of T cells: correlation with transendothelial chemotactic potential. Eur. J. Immunol. 26: 640
    OpenUrlCrossRefPubMed
  45. ↵
    Rabin, R. L., M. K. Park, F. Liao, R. Swofford, D. Stephany, J. M. Farber. 1999. Chemokine receptor responses on T cells are achieved through regulation of both receptor expression and signaling. J. Immunol. 162: 3840
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Liao, F., R. L. Rabin, C. S. Smith, G. Sharma, T. B. Nutman, J. M. Farber. 1999. CC-chemokine receptor 6 is expressed on diverse memory subsets of T cells and determines responsiveness to macrophage inflammatory protein 3α. J. Immunol. 162: 186
    OpenUrlAbstract/FREE Full Text
  47. ↵
    Huang, Y., W. A. Paxton, S. M. Wolinsky, A. U. Neumann, L. Zhang, T. He, S. Kang, D. Ceradini, Z. Jin, K. Yazdanbakhsh, et al 1996. The role of a mutant CCR5 allele in HIV-1 transmission and disease progression. Nat. Med. 2: 1240
    OpenUrlCrossRefPubMed
  48. ↵
    Dean, M., M. Carrington, C. Winkler, G. A. Huttley, M. W. Smith, R. Allikmets, J. J. Goedert, S. P. Buchbinder, E. Vittinghoff, E. Gomperts, et al 1996. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene: Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science 273: 1856
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Unutmaz, D., D. R. Littman. 1997. Expression pattern of HIV-1 coreceptors on T cells: implications for viral transmission and lymphocyte homing. Proc. Natl. Acad. Sci. USA 94: 1615
    OpenUrlFREE Full Text
  50. ↵
    Baba, M., T. Imai, M. Nishimura, M. Kakizaki, S. Takagi, K. Hieshima, H. Nomiyama, O. Yoshie. 1997. Identification of CCR6, the specific receptor for a novel lymphocyte-directed CC chemokine LARC. J. Biol. Chem. 272: 14893
    OpenUrlAbstract/FREE Full Text
  51. ↵
    Loetscher, M., P. Loetscher, N. Brass, E. Meese, B. Moser. 1998. Lymphocyte-specific chemokine receptor CXCR3: regulation, chemokine binding and gene localization. Eur. J. Immunol. 28: 3696
    OpenUrlCrossRefPubMed
  52. ↵
    Signoret, N., J. Oldridge, A. Pelchen-Matthews, P. J. Klasse, T. Tran, L. F. Brass, M. M. Rosenkilde, T. W. Schwartz, W. Holmes, W. Dallas, et al 1997. Phorbol esters and SDF-1 induce rapid endocytosis and down modulation of the chemokine receptor CXCR4. J. Cell Biol. 139: 651
    OpenUrlAbstract/FREE Full Text
  53. ↵
    Signoret, N., M. M. Rosenkilde, P. J. Klasse, T. W. Schwartz, M. H. Malim, J. A. Hoxie, M. Marsh. 1998. Differential regulation of CXCR4 and CCR5 endocytosis. J. Cell Sci 111: 2819
    OpenUrlAbstract/FREE Full Text
  54. ↵
    Sallusto, F., D. Lenig, R. Forster, M. Lipp, A. Lanzavecchia. 1999. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401: 708
    OpenUrlCrossRefPubMed
  55. ↵
    Kurihara, T., G. Warr, J. Loy, R. Bravo. 1997. Defects in macrophage recruitment and host defense in mice lacking the CCR2 chemokine receptor. J. Exp. Med. 186: 1757
    OpenUrlAbstract/FREE Full Text
  56. ↵
    Zhou, Y., T. Kurihara, R. P. Ryseck, Y. Yang, C. Ryan, J. Loy, G. Warr, R. Bravo. 1998. Impaired macrophage function and enhanced T cell-dependent immune response in mice lacking CCR5, the mouse homologue of the major HIV-1 coreceptor. J. Immunol. 160: 4018
    OpenUrlAbstract/FREE Full Text
  57. ↵
    Huffnagle, G. B., L. K. McNeil, R. A. McDonald, J. W. Murphy, G. B. Toews, N. Maeda, W. A. Kuziel. 1999. Cutting edge: Role of C-C chemokine receptor 5 in organ-specific and innate immunity to Cryptococcus neoformans. J. Immunol. 163: 4642
    OpenUrlAbstract/FREE Full Text
  58. ↵
    Traynor, T. R., W. A. Kuziel, G. B. Toews, G. B. Huffnagle. 2000. CCR2 expression determines T1 versus T2 polarization during pulmonary Cryptococcus neoformans infection. J. Immunol. 164: 2021
    OpenUrlAbstract/FREE Full Text
View Abstract
PreviousNext
Back to top

In this issue

The Journal of Immunology: 165 (6)
The Journal of Immunology
Vol. 165, Issue 6
15 Sep 2000
  • Table of Contents
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
The Primate Lentiviral Receptor Bonzo/STRL33 Is Coordinately Regulated with CCR5 and Its Expression Pattern Is Conserved Between Human and Mouse
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
Citation Tools
The Primate Lentiviral Receptor Bonzo/STRL33 Is Coordinately Regulated with CCR5 and Its Expression Pattern Is Conserved Between Human and Mouse
Derya Unutmaz, Wenkai Xiang, Mary Jean Sunshine, Jim Campbell, Eugene Butcher, Dan R. Littman
The Journal of Immunology September 15, 2000, 165 (6) 3284-3292; DOI: 10.4049/jimmunol.165.6.3284

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
The Primate Lentiviral Receptor Bonzo/STRL33 Is Coordinately Regulated with CCR5 and Its Expression Pattern Is Conserved Between Human and Mouse
Derya Unutmaz, Wenkai Xiang, Mary Jean Sunshine, Jim Campbell, Eugene Butcher, Dan R. Littman
The Journal of Immunology September 15, 2000, 165 (6) 3284-3292; DOI: 10.4049/jimmunol.165.6.3284
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Early Self-Regulatory Mechanisms Control the Magnitude of CD8+ T Cell Responses Against Liver Stages of Murine Malaria
  • Sublethal Hyperoxia Impairs Pulmonary Innate Immunity
  • Dependence of IL-4, IL-13, and Nematode-Induced Alterations in Murine Small Intestinal Smooth Muscle Contractility on Stat6 and Enteric Nerves
Show more Host Defense

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • Public Access
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2019 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606