Mammalian responses to LPS require the expression of Toll-like receptor 4 (TLR4), CD14, and MD-2. We expressed fluorescent TLR4 in cell lines and found that TLR4 densely localized to the surface and the Golgi. Similar distributions were observed in human monocytes. Confocal imaging revealed rapid recycling of TLR4-CD14-MD-2 complexes between the Golgi and the plasma membrane. Fluorescent LPS followed these trafficking pathways in CD14-positive cells. The TLR4- adapter protein, MyD88, translocated to the cell surface upon LPS exposure, and cross-linking of surface TLR4 with antibody induced signaling. Golgi-associated TLR4 expression was disrupted by brefeldin A, yet LPS signaling was preserved. We conclude that LPS signaling may be initiated by surface aggregation of TLR4 and is not dependent upon LPS trafficking to the Golgi. Mammalian responses to LPS require the expression of Toll-like receptor 4 (TLR4), CD14, and MD-2. We expressed fluorescent TLR4 in cell lines and found that TLR4 densely localized to the surface and the Golgi. Similar distributions were observed in human monocytes. Confocal imaging revealed rapid recycling of TLR4-CD14-MD-2 complexes between the Golgi and the plasma membrane. Fluorescent LPS followed these trafficking pathways in CD14-positive cells. The TLR4- adapter protein, MyD88, translocated to the cell surface upon LPS exposure, and cross-linking of surface TLR4 with antibody induced signaling. Golgi-associated TLR4 expression was disrupted by brefeldin A, yet LPS signaling was preserved. We conclude that LPS signaling may be initiated by surface aggregation of TLR4 and is not dependent upon LPS trafficking to the Golgi. The first line defense of multicellular organisms against bacterial pathogens relies upon germ line-encoded receptors that recognize a variety of conserved molecular structures on microorganisms (1Medzhitov R. Janeway Jr., C.A. Cell. 1997; 91: 295-298Google Scholar, 2Fearon D.T. Locksley R.M. Science. 1996; 272: 50-53Google Scholar, 3Aderem A. Ulevitch R.J. Nature. 2000; 406: 782-787Google Scholar). One family of such pattern recognition receptors is the type I transmembrane signaling receptors known as Toll-like receptors (TLRs). 1The abbreviations used are: TLR, Toll-like receptor; IL, interleukin; LPS, lipopolysaccharide; GFP, green fluorescent protein; CFP, cyan fluorescent protein; YFP, yellow fluorescent protein; HEK, human embryonic kidney; MALP-2, M. fermentans-derived membrane lipopeptide macrophage-activating lipopeptide of 2 kDa; mAb, monoclonal antibody; FRAP, fluorescent recovery after photobleaching; TNF, tumor necrosis factor; HRP, horseradish peroxidase; FACS, fluorescence-activated cell sorting; PBS, phosphate-buffered saline; TNFR, tumor necrosis factor receptor; ER, endoplasmic reticulum; PBMC, peripheral blood mononuclear cell; GM1, monosialo ganglioside GM1; APC, allophycocyanin; BODIPY, 4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-5-indacene; rLBP, recombinant lipopolysaccharide binding protein 1The abbreviations used are: TLR, Toll-like receptor; IL, interleukin; LPS, lipopolysaccharide; GFP, green fluorescent protein; CFP, cyan fluorescent protein; YFP, yellow fluorescent protein; HEK, human embryonic kidney; MALP-2, M. fermentans-derived membrane lipopeptide macrophage-activating lipopeptide of 2 kDa; mAb, monoclonal antibody; FRAP, fluorescent recovery after photobleaching; TNF, tumor necrosis factor; HRP, horseradish peroxidase; FACS, fluorescence-activated cell sorting; PBS, phosphate-buffered saline; TNFR, tumor necrosis factor receptor; ER, endoplasmic reticulum; PBMC, peripheral blood mononuclear cell; GM1, monosialo ganglioside GM1; APC, allophycocyanin; BODIPY, 4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-5-indacene; rLBP, recombinant lipopolysaccharide binding protein These receptors are all characterized by an intracellular signaling domain that is homologous to that of the IL-1 receptor and an extracellular domain with leucine-rich repeats (4Rock F.L. Hardiman G. Timans J.C. Kastelein R.A. Bazan J.F. Proc. Natl. Acad. Sci. U. S. A. 1998; 95: 588-593Google Scholar, 5Du X. Poltorak A. Wei Y. Beutler B. Eur. Cytokine Netw. 2000; 11: 362-371Google Scholar). The most extensively studied microbial product known to engage TLRs is lipopolysaccharide (LPS; endotoxin), a complex glycolipid that comprises the major portion of the outer leaflet of the outer membrane of Gram-negative bacteria (6Raetz C.R. Annu. Rev. Biochem. 1990; 59: 129-170Google Scholar). A potent immune response is orchestrated upon the recognition of LPS by mammalian cells, including the production and release of cytokines, activation of complement, and various other effects that result in the killing and clearance of the pathogen. Uncontrolled hyperinflammatory host responses to LPS may lead to life-threatening complications such as septic shock, multiorgan failure, and death (7Bone R.C. Ann. Intern. Med. 1991; 115: 457-469Google Scholar). Toll-like receptor 4 is the signaling receptor for LPS and requires the small glycosylated protein MD-2 for optimal signaling (8Poltorak A. He X. Smirnova I. Liu M.Y. Huffel C.V. Du X. Birdwell D. Alejos E. Silva M. Galanos C. Freudenberg M. Ricciardi-Castagnoli P. Layton B. Beutler B. Science. 1998; 282: 2085-2088Google Scholar, 9Hoshino K. Takeuchi O. Kawai T. Sanjo H. Ogawa T. Takeda Y. Takeda K. Akira S. J. Immunol. 1999; 162: 3749-3752Google Scholar, 10Shimazu R. Akashi S. Ogata H. Nagai Y. Fukudome K. Miyake K. Kimoto M. J. Exp. Med. 1999; 189: 1777-1782Google Scholar, 11Schromm A.B. Lien E. Henneke P. Chow J.C. Yoshimura A. Heine H. Latz E. Monks B.G. Schwartz D.A. Miyake K. Golenbock D.T. J. Exp. Med. 2001; 194: 79-88Google Scholar, 12Visintin A. Mazzoni A. Spitzer J.A. Segal D.M. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 12156-12161Google Scholar, 13Nagai Y. Akashi S. Nagafuku M. Ogata M. Iwakura Y. Akira S. Kitamura T. Kosugi A. Kimoto M. Miyake K. Nat. Immunol. 2002; 3: 667-672Google Scholar). We have recently observed that a mutant form of MD-2 (C95Y) completely abrogated LPS responses and that wild-type MD-2 was able to confer LPS responsiveness in TLR4-positive cells lacking MD-2 expression (11Schromm A.B. Lien E. Henneke P. Chow J.C. Yoshimura A. Heine H. Latz E. Monks B.G. Schwartz D.A. Miyake K. Golenbock D.T. J. Exp. Med. 2001; 194: 79-88Google Scholar, 12Visintin A. Mazzoni A. Spitzer J.A. Segal D.M. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 12156-12161Google Scholar). Whereas there is widespread agreement that CD14, TLR4, and MD-2 expression are necessary for optimal responses to LPS, the mechanism of cellular activation remains in doubt. Wright and co-workers (14Thieblemont N. Thieringer R. Wright S.D. Immunity. 1998; 8: 771-777Google Scholar,25Thieblemont N. Wright S.D. J. Exp. Med. 1999; 190: 523-534Google Scholar) observed that LPS is internalized and trafficked to the Golgi apparatus. Indeed, they reported that in the absence of internalization and movement of LPS to the Golgi, LPS did not activate mammalian cells (14Thieblemont N. Thieringer R. Wright S.D. Immunity. 1998; 8: 771-777Google Scholar). Wright proposed these events to be the critical initiators of signal transduction. TLR4 has now been reported to be localized to the Golgi in certain epithelial cells (15Hornef M.W. Frisan T. Vandewalle A. Normark S. Richter-Dahlfors A. J. Exp. Med. 2002; 195: 559-570Google Scholar). Together, these data suggested that LPS stimulates innate immune responses by activating an internal receptor, TLR4, that normally resides in the Golgi apparatus. Since the discovery and development of green fluorescent protein (GFP), the subcellular localization, trafficking, and fate of proteins can be studied in living cells using the fluorescent protein as a fusion tag (16Tsien R.Y. Annu. Rev. Biochem. 1998; 67: 509-544Google Scholar, 17Lippincott-Schwartz J. Histochem. Cell Biol. 2001; 116: 97-107Google Scholar, 18Lippincott-Schwartz J. Snapp E. Kenworthy A. Nat. Rev. Mol. Cell. Biol. 2001; 2: 444-456Google Scholar). Moreover, spectral variants of GFP, such as yellow fluorescent protein (YFP) and cyan fluorescent protein (CFP), have been developed. These GFP variants can be exploited for simultaneous visualization of two or more different proteins. We have engineered a TLR-fluorescent protein chimera with CFP or YFP fused in frame to the C terminus of TLR2 and TLR4 and, as a control, fused to the TNF receptor p60. These cDNA constructs were stably expressed in human embryonic kidney 293 (HEK293) cells. HEK293 cells lack TLR2 or TLR4 expression, and expression of the fluorescent chimeric Toll receptors enabled these cells to respond to the appropriate TLR ligand. We defined the subcellular localization, trafficking, and reorganization of TLR4, CD14, MD-2, and LPS in living cells by employing time lapse confocal microscopy of monocolor- and dual and triple color-labeled cells as well as fluorescent photobleaching techniques. We confirmed that LPS traffics to the Golgi and found that TLR4 is expressed in the Golgi of transfected HEK cells as well as native monocytes. However, TLR4 is also surface-expressed. Furthermore, LPS continued to traffic between the cell surface and the Golgi under experimental conditions where signal transduction does not occur. Conversely, TLR4 expression in the Golgi was not necessary for cells to respond to LPS. In addition, antibody cross-linking of surface TLR4 was able to trigger strong signaling both in TLR4-transfected cells and human monocytes. These findings provide strong evidence that LPS signaling is initiated on the plasma membrane. Localization of TLR4 in an intact Golgi network and the movement of LPS to this internal pool of TLR4 is neither necessary nor sufficient for signaling to occur. Reagents were obtained from Sigma unless otherwise indicated. PBS, Dulbecco's modified Eagle's medium, G418, and trypsin/versene mixture were from BioWhittaker (Walkersville, MD). Low endotoxin fetal bovine serum was from Hyclone (Logan, UT). Ciprofloxacin was a gift from Miles Pharmaceuticals (West Haven, CT). LPS derived from Escherichia coli strain 0111:B4 was purchased from Sigma and re-extracted by phenol chloroform as described (19Hirschfeld M. Ma Y. Weis J.H. Vogel S.N. Weis J.J. J. Immunol. 2000; 165: 618-622Google Scholar). Human IL-1b and TNF-α were purchased from Genzyme Pharmaceuticals (Cambridge, MA). The Mycoplasma fermentans-derived membrane macrophage-activating lipopeptide of 2 kDa (MALP-2) was obtained from Dr. G. Rawadi (Institut Pasteur, Paris, France). The cDNA for human TLR4 was provided in the vector pcDNA3 by Drs. C. Janeway and R. Medzhitov (Yale University, New Haven, CT). The expression plasmid pRK7-TLR2 was obtained from Dr. C. Kirschning (Technical University of Munich). The vector pcDNA3 (Invitrogen) was previously modified to include either CFP or YFP as C-terminal epitope tags in frame with a cloning site; these vectors were provided by Drs. F. Chan and M. Lenardo (20Chan F.K. Chun H.J. Zheng L. Siegel R.M. Bui K.L. Lenardo M.J. Science. 2000; 288: 2351-2354Google Scholar). The same investigators also provided epitope-tagged p60 TNFR (20Chan F.K. Chun H.J. Zheng L. Siegel R.M. Bui K.L. Lenardo M.J. Science. 2000; 288: 2351-2354Google Scholar). Polymerase chain reaction of TLR2 and TLR4 was performed on pRK7-TLR2 and on pcDNA3-TLR4 in order to construct chimeric fluorescent cDNAs. The upper and lower primers for TLR2 were 5′-GAAGCAGGATCCATGCCACATACTTTGT-3′ and 5′-GGGCTCGAGGGACTTTATCGCAGCTCTCAGA-3′. The upper and lower primers for TLR4 were 5′-GATGATGGATCCATGATGTCTGCCTCGC-3′ and 5′-ATTTTTGGCTCGAGGATAGATGTTGCTTCC-3′. The PCR fragments were digested with BamHI and XhoI and cloned in frame into pcDNA3-CFP and pcDNA3-YFP. The hMD-2 mammalian expression plasmid pEFBOS containing C-terminal FLAG and His epitopes was a gift of Dr. K. Miyake (University of Tokyo). The Golgi subcelluluar localization vector consisting of the targeting sequence of human β-galactosyltransferase fused to CFP was purchased from CLONTECH. The fluorescent MyD88 constructs were made by PCR of MyD88 in pRK7 (provided by Dr. H. Wesche, Tularik, Inc., San Francisco, CA) using the following upper and lower primers for MyD88: 5′-CCACGGGGATCCATGGCTGCAGGAGGTC-3′ and 5′-GAACAGGTCGACGGGCAGGGACAAGGC-3′. The PCR fragments were trimmed withBamHI and SalI and cloned in frame into pcDNA3-CFP or pcDNA3-YFP, respectively. Stable cell lines of HEK293 cells expressing the fluorescent protein TLR constructs were engineered by calcium phosphate transfection (21Chen C. Okayama H. Mol. Cell. Biol. 1987; 7: 2745-2752Google Scholar), selection of bulk populations of cells in the neomycin analog G418 (1 mg of total drug/ml), and positive selection by fluorescence-activated cell sorting (BD Vantage, Becton Dickinson Immunocytometry). Clonal cell lines were obtained by limiting dilution. The fluorescent HEK293 cell lines were maintained in Dulbecco's modified Eagle's medium supplemented with 10% fetal calf serum and 0.5 mg/ml G418 in a 5% saturated CO2 atmosphere at 37 °C. A cell line stably expressing both TLR4 and MD-2 was generated by retroviral transduction of HEK-TLR4YFPcells as described with a retrovirus encoding human MD-2 (12Visintin A. Mazzoni A. Spitzer J.A. Segal D.M. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 12156-12161Google Scholar). Cells were analyzed by Western blot and immunoprecipitation as previously described (12Visintin A. Mazzoni A. Spitzer J.A. Segal D.M. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 12156-12161Google Scholar). Briefly, MD-2-transduced HEK-TLR4YFP or HEK-TLR2YFP cells were grown in 10-cm dishes and washed in ice-cold Hanks' balanced salt solution, and surface proteins were labeled at 4 °C for 30 min in 5 ml of a solution of sulfosuccinimidobiotin (1 mg/ml; Pierce) in Hanks' balanced salt solution. Cells were washed and then incubated in 1 mglycine, pH 9, for 5 min, washed again, and lysed in 1 ml of buffer (137 mm NaCl, 20 mm Tris·HCl, pH 7.4, 1 mm EDTA, 0.5% Triton X-100) containing 60 mm n-octylglucoside, 25 mm iodoacetamide, and a mixture of protease inhibitors (10 μg/ml leupeptin and aprotinin and 1 mm phenylmethylsulfonyl fluoride). Lysates were centrifuged, precleared for 1 h in 40 μl of packed protein A-Sepharose (Amersham Biosciences), and immunoprecipitated with 2 μg of a GFP polyclonal antibody (Molecular Probes, Inc., Eugene, OR) per ml and 20 μl of protein A-Sepharose for 16 h at 4 °C. Pellets were washed four times in lysis buffer, resolved by 4–15% SDS-PAGE under reducing conditions, and transferred to nitrocellulose membranes. These membranes were blocked in 5% powdered milk (Difco) and blotted with either the M2-HRP mAb (Sigma), anti-GFP mAb (CLONTECH), or Avidin-HRP (Bio-Rad). The blots were then incubated with HRP substrate (enhanced chemiluminescence substrate; Amersham Biosciences) and developed by exposure to film (Hyperfilm; Amersham Biosciences). HEK293 cells that stably express TLR4YFP, TLR2YFP, TNFRYFP, or empty vector (pcDNA) were seeded into 96-well tissue culture plates at a density of 2 × 104 cells/well. The following day, cells were transiently transfected with luciferase reporter genes using Genejuice (Novagen) per the manufacturer's recommendations. In order to assess NF-κB activation, an NF-κB-luciferase reporter gene consisting of an artificial promoter composed of a multimer of five NF-κB sites driving the firefly luciferase gene, was co-transfected with a constitutively active Renilla-luciferase reporter gene (Promega, Madison, WI). The following day, the cells were stimulated as indicated. When necessary, HEK-TLR4YFP cells were either co-transfected with MD-2, stimulated in the presence of soluble MD-2 in conditioned medium, or retrovirally transduced with the cDNA for MD-2 (12Visintin A. Mazzoni A. Spitzer J.A. Segal D.M. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 12156-12161Google Scholar). Note that we have established that all three of these methodologies for expressing MD-2 comparably enhance TLR4-mediated responses to LPS. 2E. Latz and A. Visintin, unpublished data. After 4–6 h of stimulation, the cells were lysed in passive lysis buffer (Promega), and reporter gene activity was measured using a plate reader luminometer (Victor2; PerkinElmer Life Sciences) using the Dual-Luciferase Assay Reporter System (Promega) and normalized for transfection efficiency. In all cases, the data shown represent one of three separate experiments and are presented as the mean values ± S.D. of triplicate samples. Confocal microscopy was performed with a Zeiss Axiovert 100-M inverted microscope equipped with an LSM 510 laser-scanning unit. A Zeiss 40× and a 63× 1.4 numerical aperture plan Apochromat oil immersion objective (Zeiss) was used. Cells were seeded on 35-mm glass bottom γ-irradiated tissue culture dishes (MatTek Corp., Ashland, MA). CFP-tagged proteins were visualized using the 453-nm argon laser line; for YFP and GFP, the 514- or 488-nm line of a 25-milliwatt argon laser was used. Red fluorophores were excited with a 1.0-milliwatt helium/neon laser emitting at 543 nm. Alexa 647 or Cy5-LPS were excited with a helium/neon laser emitting at 633 nm. Band pass or long pass filters were chosen to optimally separate the fluorescence emissions between the different photomultipliers using single-labeled samples of the probes as controls. When crossover of the fluorescence signal was measured to be more than 5%, two or more tracks were scanned alternately with only one laser active per scan and the respective detector channel active at each time. Fluorescent recovery after photobleaching (FRAP) experiments were performed by selecting an area of interest and rapidly applying 99 consecutive scans using the 514-nm line of a 25-milliwatt argon laser at full laser power. Fluorescence recovery was observed under low illumination over time as indicated in the figures. Additional FRAP experiments were done after pretreatment with 200 μg/ml cycloheximide (Sigma) in complete tissue culture medium for 2 h. Live cell images and confocal time lapse fluorescence imaging were performed at 37 °C using a warm stage apparatus (Zeiss). Indirect immunofluorescence staining for FACS was done using purified mouse monoclonal antibodies (TLR4, HTA125; TLR2, TL2.1; CD14, 3C10) and isotype-matched control antibodies (Sigma) as primary antibodies. Cells were counterstained with APC-conjugated goat anti-mouse secondary antibody (Caltag). Cells were analyzed by flow cytometry (Becton Dickinson LSR) using the argon laser at 488 nm for excitation of YFP fluorescence and the helium/neon laser emitting at 633 nm to excite APC. Under these conditions, no spectral overlap of the fluorophores was observed. Transient transfection of cells observed with confocal microscopy was done on cells plated in 35-mm glass bottom tissue culture dishes using Effectene™ Transfection Reagent (Qiagen) according to the manufacturer's recommendations. Golgi stain by BODIPY-TR-conjugated ceramide (Molecular Probes, Inc., Eugene, OR) was done per the manufacturer's recommendations. MD-2 was stained for confocal analysis by use of M2 (Sigma) as a primary antibody and Alexa 647-conjugated goat anti-mouse as the secondary antibody (Molecular Probes); mCD14 was stained by Tricolor-conjugated anti-CD14 antibody (Caltag). Monocytes were isolated by plastic adherence of PBMCs in confocal Petri dishes (MatTek) and cultivated in RPMI1640 with 5% human A+ serum (University Hospital, Trondheim, Norway) for 48 h before they were washed three times in Hanks' solution and fixed in 2% formaldehyde (Merck) in PBS for 15 min on ice. The cells were then washed twice in PBS with 1% A+ serum (PBS/A+) before they were treated with acetone (−20 °C) for 10 min followed by a careful wash with PBS/A+. Nonspecific binding was blocked by adding PBS with 20% A+ on ice for 20 min. The cells were then stained for 30 min at room temperature with 10 μg/ml HTA125 conjugated with Alexa 546 (Molecular Probes) or a control murine IgG (Caltag) also conjugated with Alexa 546. After three washes with PBS/A+, the cells were examined in the confocal microscope with a 543-nm excitation. Membrane expression of TLR4 and TLR2 in monocytes was measured by flow cytometry (BD LSR). Freshly isolated PBMCs were surface-stained with an APC-labeled anti-CD14 antibody (Caltag) together with biotinylated anti-TLR4, anti-TLR2, or isotype control antibodies (all from eBiosciences) after preincubation in 20% human serum for 20 min. Biotinylated antibodies were developed with fluorescein isothiocyanate-conjugated streptavidin (Sigma), and CD14-positive cells (gated according to their forward and sideward scatter characteristics) were analyzed for TLR expression. E. coli 0111:B4 LPS was subjected to a second phenol extraction to remove minor contaminants and labeled with the amine-reactive Cy5-fluorochrome (AmershamBiosciences) per the manufacturer's recommendations. Free dye was separated from labeled LPS by passage through a sizing column (PD-10 column; Amersham Biosciences). The bioreactivity of the labeled LPS preparations was comparable with unlabeled LPS. Labeled LPS was preincubated in PBS plus 5% human serum (Sigma), human rsCD14 (5 μg/ml), and human rLBP (1 μg/ml) for 10 min prior to use. Adherent HEK293 cells were cultured in 24-well plates and incubated with Cy5-conjugated LPS for the indicated times at 37 °C in 5% CO2. Cells were washed twice with ice-cold PBS, trypsinized, and suspended in cold growth medium, washed twice by centrifugation at 200 × g in ice-cold PBS, and analyzed by FACS at an excitation frequency of 633 nm. Antibodies were bound to sterile 96-well high protein binding plates (Costar) by overnight incubation at 4 °C in PBS. After extensive washing with PBS, 4 × 105 (HEK) or 7 × 105 cells (PBMCs) were plated in each well and incubated for 5–12 h. Supernatants were analyzed for IL-8 (DuoSet; R&D) or TNF-α (Diaclone) by enzyme-linked immunosorbent assay. Human embryonic kidney 293 cells are deficient in all of the known members of the LPS signaling receptor, including TLR4, MD-2, and CD14. Thus, HEK293 cells are normally unresponsive to LPS. Additionally, HEK293 cells do not express TLR2 or TLR9 and likewise are unresponsive to the respective stimulatory molecules known to activate these receptors. We and others have observed that HEK293 cells transfected with TLR4 gain LPS responsiveness if they are either co-transfected with MD-2 or stimulated in the presence of conditioned medium rich in recombinant soluble MD-2 (11Schromm A.B. Lien E. Henneke P. Chow J.C. Yoshimura A. Heine H. Latz E. Monks B.G. Schwartz D.A. Miyake K. Golenbock D.T. J. Exp. Med. 2001; 194: 79-88Google Scholar, 12Visintin A. Mazzoni A. Spitzer J.A. Segal D.M. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 12156-12161Google Scholar, 22Yang H. Young D.W. Gusovsky F. Chow J.C. J. Biol. Chem. 2000; 275: 20861-20866Google Scholar). HEK293 cells that stably express TLR4YFP, TLR2YFP, or control cDNA (TNFRYFP or empty vector-pcDNA) were established. The stable cell lines were transiently co-transfected with a reporter construct in that firefly luciferase is under the control of the transcription factor NF-κB and stimulated with the microbial cell wall components LPS and the Mycoplasma fermentans-derived membrane lipopeptide MALP-2. LPS conferred responsiveness to cells expressing TLR4YFP, whereas MALP-2 selectively activated TLR2YFP-expressing cells (Fig.1 A). The responses observed were indistinguishable from responses observed in transfected HEK293 cells using TLR constructs that do not include an epitope tag (data not shown). Proper cellular responses to TLR2 or TLR4 engaging stimuli by the chimeric constructs were further illustrated by a time-dependent decrease of total IκB-a and nuclear translocation of NF-κB as assessed by electrophoretic mobility shift assay or immunofluorescence and confocal imaging (not shown). All of the various stable cell lines were tested and were equally reactive to IL-1b, TNF-α, and phorbol 12-myristate 13-acetate (Fig. 1 Aand data not shown). The responsiveness was similar in each of several clones tested regardless of which C-terminal tag (CFP, GFP, or YFP) was used. Unlike prior observations with N-terminally FLAG-tagged constructs, expression of these chimeric proteins at the C terminus did not result in appreciable constitutive NF-κB activation (data not shown). Thus, fusion proteins of TLR4 and TLR2 with fluorescent tags at the C terminus are functional signaling receptors that confer both specific and sensitive recognition of their cognate ligands in HEK293 cells. The use of GFP or the spectral variants CFP and YFP as a protein tag allows protein function and dynamics to be investigated within the environment of the living cell. The tagged constructs were designed so that the subcellular localization of the TLR receptors remained directed by their native signal sequences rather than by a signal sequence contained in the vector, as is commonly done when employing FLAG-tagged proteins. Cells were grown on glass bottom tissue culture dishes and observed at 37 °C. TLR4YFP was primarily expressed in two different subcellular localizations. We observed plasma membrane expression and localization in a defined juxtanuclear area. Surface staining for TLR4 using a monoclonal antibody (HTA125) revealed a clear surface expression of the protein independently of the fluorescent protein tag used (Fig. 1 C). Similarly, human monocytes expressed significant levels of both TLR4 and TLR2. Surface biotinylation and immunoprecipitation with anti-GFP antibody, followed by Western blot with avidin-HRP, revealed that only the heavily glycosylated mature forms of TLR4 and MD-2 (Fig.2 B) are expressed on the cell surface. We tested whether TLR2 would bind to MD-2 or whether TLR4 would bind MD-1 under identical experimental conditions but failed to observe co-localization of these protein pairs (data not shown) (12Visintin A. Mazzoni A. Spitzer J.A. Segal D.M. Proc. Natl. Acad. Sci. U. S. A. 2001; 98: 12156-12161Google Scholar). These results suggest that both TLR4 and MD-2 follow the trans-Golgi secretory pathway and reside on the cell surface as a mature protein complex. We next sought to identify the intracellular compartment that is enriched in TLR4. A fluorescent subcellular localization marker for the Golgi complex consisting of the Golgi-targeting sequence of human β-galactosyltransferase fused to CFP co-localized with the juxtanuclear compartment positive for TLR4 (Fig. 2 A). Likewise, fluorescent ceramide, which is known to enrich in Golgi membranes (23Lipsky N.G. Pagano R.E. Science. 1985; 228: 745-747Google Scholar), also co-localized with juxtanuclear TLR4YFP(Fig. 2 B). These studies identify the juxtanuclear region enriched in TLR4 as the Golgi apparatus. To determine whether the localization pattern of TLR4 was an artifact of transfection and overexpression or reasonably reflected the distribution of TLR4 in native cells, we stained human monocytes with a monoclonal anti-TLR4 antibody. FACS analysis of monocytes revealed detectable TLR4 on the surface of these cells (Fig. 1 C). Intracellular staining of purified monocytes clearly showed that TLR4 is also expressed in a defined juxtanuclear region (Fig.2 C), consistent with the conclusion that TLR4 resides both on the cell surface and in the Golgi. The Golgi complex is an intracellular compartment that is specialized for secretory traffic. Newly synthesized proteins and lipids are received from the ER by the Golgi complex and are covalently modified in preparation for delivery to their final destination (such as plasma membrane, lysosomes, secretory granules) or to be recycled back to the ER. In addition to these sorting and filtering capabilities, the Golgi complex recycles plasma membrane components that are retrieved by endocytosis (17Lippincott-Schwartz J. Histochem. Cell Biol. 2001; 116: 97-107Google Scholar, 24Nichols B.J. Kenworthy A.K. Polishchuk R.S. Lodge R. Roberts T.H. Hirschberg K. Phair R.D. Lippincott-Schwartz J. J. Cell Biol. 2001; 153: 529-541Google Scholar). The physical properties of GFP allow the GFP chimera to be used in a technique known as FRAP. After GFP or its spectral variants are excited with very high illumination, these fluorophores are readily photobleached, a process that irreversibly extinguishes the fluorescence. When performing FRAP, a small area of interest in the cell is rapidly photobleached by applying scans with a high powered laser beam. Thereafter, the movement of nonbleached fluorophores into the photobleached area can be recorded at low illumination. Fluorescence recovery gives insights in the diffusional properties of the reporter chimera. We employed FRAP to investigate whether membrane-expressed TLR4YFP would cycle back to the Golgi complex after photobleaching of Golgi-associated TLR4YFP. Application of repetitive scans with full laser power to the Golgi almost completely extinguished the Golgi-localized fluorescence, whereas neither the surrounding areas nor the neighboring cells were affected (Fig.3 A). Observation of fluorescence recovery revealed a fast and complete recovery of Golgi fluorescence. Over 90% of the Golgi-associated fluorescence was recovered within 4 min after photobleaching, indicating rapid lateral movement of TLR4YFP between cell compartments. We next questioned whether the recovery of Golgi fluorescence after FRAP is primarily due to new protein synthesis or represents the recycling of TLR4YFP from the membrane pool to the Golgi apparatus. To address this question, cells were preincubated with the protein synthesis inhibitor cycloheximide for 2 h prior to FRAP. The recovery of Golgi fluorescence observed in the cycloheximide-treated cells was nearly identical to that seen in untreated cells, indicating that rapid transport of TLR4 from the plasma membrane replenished T