Abstract

Soluble leptin receptor (SLR) represents the major leptin binding activity in plasma. It is generated by alternative splicing of OB-R mRNA (OB-Re) and/or ectodomain shedding of membrane-spanning receptors. To determine the role of SLR in leptin activation of its long-form receptor, OB-Rb, we established in vitro assays using a cell line stably expressing OB-Rb. Human embryonic kidney 293 cell lines stably overexpressing OB-Rb show a dose-dependent leptin-induced signal transducer and activator of transcription 3 (STAT3) tyrosine phosphorylation and STAT3-responsive luciferase (STAT3-luc) activity. We demonstrate that when SLR is incubated with free leptin, binding of leptin to OB-Rb is reduced, with corresponding decrease of leptin-induced STAT3 tyrosine phosphorylation and STAT3-luc activity. However, by preparing leptin/SLR mixtures containing the same amount of free leptin but increasing amounts of leptin-SLR complex, we show that leptin-SLR complex does not inhibit OB-Rb activation by free leptin. These results suggest that in settings in which leptin and SLR coexist, SLR may sequester leptin from productive interactions with OB-Rb. Because plasma SLR levels are independently regulated by many different physiological and pathophysiological conditions, SLR may modulate the actions of leptin in tissues in which direct action of leptin has been demonstrated.

LEPTIN IS AN adipose tissue-derived hormone with potent effects on food intake, metabolism, and many other important physiological processes, ranging from fertility and angiogenesis to bone formation and immune function (1, 2). Leptin acts by binding to its receptor, OB-R, which is alternatively spliced into several isoforms. One of the splice variants, OB-Re, does not encode a transmembrane domain and is predicted to be secreted. Although OB-Re mRNA has been identified in rodents, this splice variant has not been found in any of the many human tissues examined (3, 4). However, soluble leptin receptor (SLR) does circulate in human plasma (510). In vitro and in vivo studies indicate that circulating leptin receptor may be generated by ectodomain shedding of membrane-spanning receptors, mediated by a metalloprotease (11, 12). The relative contribution of OB-Re mRNA-derived and ectodomain shedding-derived SLR in plasma is not known, because the sizes of circulating SLR generated by either mechanism are indistinguishable by Western blotting analysis (11). One exception was reported, however. It appears that pregnancy-induced production of SLR from OB-Re mRNA exhibits a different glycosylation pattern, although the mechanisms involved and consequences on leptin action are not known (13).

The generation of OB-Re mRNA and protein is regulated under both physiological and pathophysiological conditions. In mice, expression of OB-Re mRNA in the placenta is strongly induced at late stages of pregnancy, causing an up to 40-fold increase in circulating leptin receptor (14). In humans, levels of SLR are inversely related to adiposity (1519), increased in patients with advanced chronic heart failure (20), and higher in female insulin-dependent diabetic subjects (21). However, the effect of SLR on leptin action in target tissues has not been determined, especially at peripheral sites where both leptin and SLR are present. The role of SLR in leptin transport across the blood brain barrier also remains unresolved. Because the affinity of leptin toward membrane-spanning receptors and full-length SLR is indistinguishable (22, 23), SLR may not transport leptin to the hypothalamus, where it is required for leptin’s weight reducing and other effects (24). The blood-cerebrospinal fluid barrier at the choroid plexus and the blood-brain barrier at the cerebral endothelium are two major controlling sites for entry of circulating proteins into the brain, locations at which membrane-spanning leptin receptors are enriched and across which leptin is presumably transported (2527). Transcellular transport of intact leptin in vitro has been reported (28). No SLR has been detected in cerebrospinal fluid (29).

Previously, we and others reported that SLR is the major determinant of plasma leptin levels and acts to stabilize circulating leptin without increasing leptin mRNA transcription (30, 31). We also found that, similar to human SLR, mouse SLR may be generated by ectodomain shedding of membrane-spanning receptors both in vitro and in vivo (11). In this study, we report that when leptin is bound to its soluble receptor, the leptin-SLR complex is not capable of activating OB-Rb, nor does it inhibit the action of free leptin. When the ability of a fixed amount of leptin to activate OB-Rb with or without the presence of SLR is compared, leptin-stimulated OB-Rb signal transduction is reduced in the presence of SLR. These results suggest that SLR serves to sequester leptin from productive interactions with its signaling receptor, OB-Rb, and may play a role in leptin action in tissues in which both leptin and SLR coexist.

RESULTS

Determination of Leptin Signaling Using Stable Cell Lines

Leptin binding to its long-form receptor, OB-Rb, causes activation of the Jak2 kinase, which is constitutively associated with OB-Rb. Activation of Jak2 leads to the phosphorylation of tyrosine residues within the cytoplasmic domain of OB-Rb. Two tyrosines are phosphorylated after leptin stimulation and become docking sites for Src homology 2 (SH2) domain containing signaling proteins (32). In the mouse receptor, phosphorylation of Tyr 985 leads to the recruitment of SHP-2, a protein tyrosine phosphatase that serves to decrease Jak2 phosphorylation and as an adaptor protein to activate the ERK/MAPK pathway (32, 33); phosphorylation of Tyr 1138 leads to the recruitment of signal transducer and activator of transcription 3 (STAT3) in vivo (34) and other STATs in vitro. Activation of STAT3 can be determined by following STAT3 tyrosine phosphorylation or its ability to increase transcription of reporter genes containing multiple copies of STAT3 binding sites in the promoter region (STAT3-luc).

Efforts from many laboratories have failed to identify cell lines with high-level expression of endogenous OB-Rb. To overcome this difficulty in studying leptin signal transduction pathway, we and others have been performing transfection of OB-R expression vectors transiently or permanently to detect leptin signaling (32, 35). Using a Flp-In human embryonic kidney 293 (HEK293) cell line that allows the integration of one copy transfected cDNA per cell, based on Cre-Lox recombination strategy, we were able to generate stable cell lines that express recombinant proteins without the influence of insert copy number, therefore allowing expression of mRNA to similar levels among the cell lines generated (11).

We measured leptin response in stable HEK293 cells overexpressing either OB-Ra or OB-Rb. OB-Ra, which cannot signal by itself, is used as a negative control; OB-Rb, which encodes a long cytoplasmic domain containing both Tyr 985 and Tyr 1138, is a target of Jak2 phosphorylation. Cell surface expression of OB-R protein was quantified by incubation of cells with radiolabeled leptin with or without unlabeled, cold leptin as competitor (Fig. 1B). There is less leptin binding on the surface of OB-Rb-expressing cells, consistent with earlier studies that OB-Rb translocation to plasma membrane is less efficient compared with OB-Ra (36, 37). However, when cells are treated with leptin after incubation under serum-free conditions, leptin caused STAT3 activation only in OB-Rb-expressing cells, as determined by STAT3 tyrosine phosphorylation (Fig. 1C) and STAT3-luc activity (Fig. 1D), confirming the specificity of these cell lines as useful reagents in determining leptin receptor signal transduction in the absence or presence of SLR.

Diagram of OB-R Constructs (A) Used to Generate Stable HEK293 Cell Lines and Determination of Cell Surface OB-R Expression by [125I]Leptin Binding (B) Leptin activation of STAT3 tyrosine phosphorylation and STAT3-luc activity are shown in panels C and D. TMR, Transmembrane region of OB-R; Y1, Y2, and Y3 denote cytoplasmic tyrosines of OB-Rb; Y3, at position 1138 of murine OB-Rb, is the site of STAT3 binding after phosphorylation by Jak2. Shaded areas are regions at the extracellular region of OB-R that are homologous to each other and to the ligand binding domains of other cytokine receptors. The membrane proximal domain binds leptin. myc denotes the c-myc epitope fused in frame to the carboxyl terminus of OB-R coding region. In panel B, specificity of leptin binding to stable cell lines was verified by incubation with an excess amount of cold leptin. pSTAT3 indicates tyrosine-phosphorylated STAT3, detected by an antibody recognizing phosphorylated Tyr 705 of STAT3.
Fig. 1.

Diagram of OB-R Constructs (A) Used to Generate Stable HEK293 Cell Lines and Determination of Cell Surface OB-R Expression by [125I]Leptin Binding (B) Leptin activation of STAT3 tyrosine phosphorylation and STAT3-luc activity are shown in panels C and D. TMR, Transmembrane region of OB-R; Y1, Y2, and Y3 denote cytoplasmic tyrosines of OB-Rb; Y3, at position 1138 of murine OB-Rb, is the site of STAT3 binding after phosphorylation by Jak2. Shaded areas are regions at the extracellular region of OB-R that are homologous to each other and to the ligand binding domains of other cytokine receptors. The membrane proximal domain binds leptin. myc denotes the c-myc epitope fused in frame to the carboxyl terminus of OB-R coding region. In panel B, specificity of leptin binding to stable cell lines was verified by incubation with an excess amount of cold leptin. pSTAT3 indicates tyrosine-phosphorylated STAT3, detected by an antibody recognizing phosphorylated Tyr 705 of STAT3.

Dose-Dependent Activation of STAT3 in Stable Cell Lines

One purpose of generating stable cell lines overexpressing OB-Rb is to determine the effect of SLR on leptin signal transduction. We reasoned that SLR might either enhance or inhibit leptin action in this setting. To distinguish between these possibilities, our system must be able to respond to different levels of leptin stimulation with graded responses. To determine that our stable cell lines can indeed function in such a manner, we first generated a leptin dosage-responsive curve in the absence of exogenously added SLR. Figure 2 shows that when leptin concentrations range from 0.0016 nm to 1 nm, both STAT3-luc activity (Fig. 2A) and STAT3 tyrosine phosphorylation (Fig. 2B) in treated cells showed parallel increases. In the experiments described below, we have chosen free leptin concentrations of between 0.008 nm and 0.2 nm, a range that gives rise to STAT3 responses close to being linear.

Dose-Dependent Activation of STAT3-luc Activity (A) and STAT3 Tyrosine Phosphorylation (B) by Leptin in OB-Rb HEK293 Stable Cell Lines The concentrations of leptin added to cells are indicated, with luciferase activity shown above, and pSTAT3 and total STAT3 levels below the concentration of leptin used.
Fig. 2.

Dose-Dependent Activation of STAT3-luc Activity (A) and STAT3 Tyrosine Phosphorylation (B) by Leptin in OB-Rb HEK293 Stable Cell Lines The concentrations of leptin added to cells are indicated, with luciferase activity shown above, and pSTAT3 and total STAT3 levels below the concentration of leptin used.

Preparation of Leptin-SLR Complexes with Defined Concentrations of Free and Bound Leptin

To determine the effect of SLR on leptin receptor signal transduction, we also needed to prepare leptin-SLR complexes with defined absolute and relative concentrations of each component. To achieve this goal, we took advantage of an efficient adenoviral vector system encoding OB-Re (Ad-OB-Re), which expresses recombinant SLR, as reported earlier (31). We used commercial preparations of bacterially derived recombinant leptin and added to it conditioned serum-free media from cells infected with Ad-OB-Re or a control virus that encodes β-galactosidase, Ad-β-Gal. Figure 3A shows that SLR accumulation in media of Ad-OB-Re-infected cells is readily detected with a polyclonal antibody specifically recognizing the extracellular domain of all OB-R isoforms (11, 31).

Generation of SLR by Adenoviral Overexpression (A) and Determination of the Percentage of Free and Bound Leptin in Solution by FPLC (B and C) In panel A, Ad-β-Gal (β-Gal) or Ad-OB-Re (SLR) was used to infect HEK293 cells. Supernatant (10 μl) was loaded per lane and blotted with an OB-R-specific antibody (31 ). In panel B, a fixed amount of leptin was incubated with different volumes of conditioned media containing adenovirally generated SLR. With increasing amounts of SLR present, peak sizes of leptin-SLR complex increase (percentage size of each bound leptin peak indicated by numbers) whereas those of free leptin decrease. Panel C shows the absence of leptin binding activity in conditioned media of control virus-infected cells. An excess volume (500 μl) of conditioned media from cells infected with Ad-β-Gal was incubated with [125I]leptin and loaded onto the same gel filtration column. No additional leptin binding activity is observed.
Fig. 3.

Generation of SLR by Adenoviral Overexpression (A) and Determination of the Percentage of Free and Bound Leptin in Solution by FPLC (B and C) In panel A, Ad-β-Gal (β-Gal) or Ad-OB-Re (SLR) was used to infect HEK293 cells. Supernatant (10 μl) was loaded per lane and blotted with an OB-R-specific antibody (31 ). In panel B, a fixed amount of leptin was incubated with different volumes of conditioned media containing adenovirally generated SLR. With increasing amounts of SLR present, peak sizes of leptin-SLR complex increase (percentage size of each bound leptin peak indicated by numbers) whereas those of free leptin decrease. Panel C shows the absence of leptin binding activity in conditioned media of control virus-infected cells. An excess volume (500 μl) of conditioned media from cells infected with Ad-β-Gal was incubated with [125I]leptin and loaded onto the same gel filtration column. No additional leptin binding activity is observed.

To prepare SLR-leptin complexes with defined free leptin concentration, we took advantage of the differential migration properties of leptin in a gel filtration column in free form or bound to SLR (7). The distribution of peaks corresponding to bound and free leptin is determined by incubating SLR-containing media with [125I]leptin tracer, which can be quantitated by scintillation counting. The percentage of free and bound leptin is determined by integration of areas under the curve for each peak. Finally, the absolute concentrations of free and bound leptin are obtained by multiplying that of total leptin added to the incubation mixture with the percentage of each fraction. To validate this procedure, we used either a fixed amount of free leptin and varying amounts of SLR or a fixed amount of SLR but varying amounts of free leptin and performed gel filtration chromatography (Fig. 3B and data not shown). Other leptin binding activity is not present in supernatant of virus-infected cells as incubation of [125I]-leptin tracer with an excess volume of conditioned media of Ad-β-Gal-infected cells did not reveal any additional peaks that elute ahead of free leptin (Fig. 3C).

Addition of SLR to a fixed amount of leptin inhibits leptin binding to OB-Rb HEK293 cells as well as OB-Rb signal transduction.

With the establishment of stable cell lines that respond to the concentration of free leptin linearly at a defined range (Fig. 2) and our ability to prepare SLR-leptin complexes with accurate determination of the percentage of free and bound leptin (Fig. 3), we proceeded to determine whether SLR enhances or inhibits leptin action.

Based on the gel filtration data, we mixed 1.6 ng leptin in 1 ml of buffer, corresponding to 0.1 nm leptin, which by itself elicits an intermediate STAT3 tyrosine phosphorylation and luciferase response (Fig. 2), with varying volumes of SLR-containing serum-free media to obtain a final leptin preparation that is 20%, 43%, and 86% bound to SLR (Fig. 3B). OB-Rb HEK293 cells were then treated with these leptin samples, containing either free leptin alone or leptin bound to varying amounts of SLR. Because binding of leptin to SLR is maintained at equilibrium, it is impossible to obtain leptin samples that are 100% bound. As an added control, leptin was mixed with serum-free conditioned media from Ad-β-Gal-infected HEK293 cells.

We first assessed how leptin binding to cell surface OB-Rb is affected in the absence or presence of increasing amounts of SLR. [125I]leptin alone or added different volumes of SLR-containing supernatant were incubated with OB-Rb HEK293 cells, and specific leptin binding to cell surface OB-Rb was determined (Fig. 4A). Addition of SLR caused a dose-dependent decrease of cell surface [125I]leptin binding, suggesting that SLR sequesters leptin from interaction with membrane-bound receptors.

SLR Added to a Fixed Amount of Leptin Decreases Leptin Binding to Cell Surface OB-Rb (A) as Well as Decreases Signaling through OB-Rb (B) In panel A, [125I]leptin was mixed with PBS or different volumes of SLR (10 or 50 μl), or 50 μl of supernatant of HEK293 cells infected with Ad-β-Gal in 1 ml of binding buffer. Each mixture was added to OB-Rb HEK293 cells and specific [125I]leptin binding was determined. Experiment was performed in triplicate. In panel B, different volumes of SLR were added to tubes containing 0.1 nm of free leptin, and STAT3-luc activity was determined after addition to OB-Rb HEK293 cells. The concentration of free leptin in each tube, as measured by FPLC, is indicated in the table. To determine the nonspecific effects of conditioned media from Ad-β-Gal-infected cells, the same volumes of Ad-β-Gal conditioned media were added to separate tubes. Luciferase activity and levels of pSTAT3 and total STAT3 (B) were determined as in Fig. 2. Note that there is a trend toward a slight decrease of STAT3-luc activity in the presence of increasing volumes of media derived from Ad-β-Gal-infected cells. Addition of Ad-OB-Re-conditioned media caused a statistically significant decrease in leptin-activated STAT3-luc activity as well as STAT3 tyrosine phosphorylation (B). *, P < 0.05; **, P < 0.01; ***, P < 0.001.
Fig. 4.

SLR Added to a Fixed Amount of Leptin Decreases Leptin Binding to Cell Surface OB-Rb (A) as Well as Decreases Signaling through OB-Rb (B) In panel A, [125I]leptin was mixed with PBS or different volumes of SLR (10 or 50 μl), or 50 μl of supernatant of HEK293 cells infected with Ad-β-Gal in 1 ml of binding buffer. Each mixture was added to OB-Rb HEK293 cells and specific [125I]leptin binding was determined. Experiment was performed in triplicate. In panel B, different volumes of SLR were added to tubes containing 0.1 nm of free leptin, and STAT3-luc activity was determined after addition to OB-Rb HEK293 cells. The concentration of free leptin in each tube, as measured by FPLC, is indicated in the table. To determine the nonspecific effects of conditioned media from Ad-β-Gal-infected cells, the same volumes of Ad-β-Gal conditioned media were added to separate tubes. Luciferase activity and levels of pSTAT3 and total STAT3 (B) were determined as in Fig. 2. Note that there is a trend toward a slight decrease of STAT3-luc activity in the presence of increasing volumes of media derived from Ad-β-Gal-infected cells. Addition of Ad-OB-Re-conditioned media caused a statistically significant decrease in leptin-activated STAT3-luc activity as well as STAT3 tyrosine phosphorylation (B). *, P < 0.05; **, P < 0.01; ***, P < 0.001.

The effects of SLR on leptin activation of OB-Rb in HEK293 cells are shown in Fig. 4B. As opposed to when all leptin is in the free form, addition of SLR to a fixed amount of leptin caused a dose-dependent decrease in leptin-activated luciferase activity. In agreement with the luciferase response, STAT3 tyrosine phosphorylation is also similarly reduced by increasing the amounts of SLR. This decrease is not due to the nonspecific effects caused by the presence of conditioned media, because addition of supernatant from Ad-β-Gal-infected cells did not significantly decrease leptin-induced STAT3 tyrosine phosphorylation or STAT3-luc activity (Fig. 4B), although a trend toward a slight decrease in STAT3-luc activation was observed when leptin was incubated with increasing volumes of conditioned media from Ad-β-Gal-infected cells (Fig. 4B). In aggregate, these results demonstrate that addition of SLR to a fixed amount of leptin decreases the ability of leptin ability to activate OB-Rb, as measured by both STAT3 tyrosine phosphorylation and STAT3-luc reporter activity.

SLR-Leptin Complex Does Not Affect Activation of OB-Rb by Free Leptin

We then asked whether binding of leptin to its soluble receptor merely sequesters it from binding to OB-Rb, as shown above, or whether the leptin-SLR complex also inhibits OB-Rb activation by leptin in a dominant negative fashion. To address this question, leptin was mixed with conditioned media from Ad-β-Gal-infected cells to a final concentration of 0.08 nm, or mixed with different volumes of serum-free conditioned media from Ad-OB-Re-infected cells containing 0.08 nm free leptin but different amount of leptin-SLR complex. In these samples, 34% or 88% of leptin is complexed with SLR, verified by fast protein liquid chromatography (FPLC) (Fig. 5A). To take into consideration the potential influence of media components on leptin activation of OB-Rb, the difference in volume of SLR-containing media was corrected by adding that from Ad-β-Gal-infected HEK293 cells, so that total media volume in each sample is identical. OB-Rb HEK293 cells were transfected with STAT3-luc and treated with the leptin samples prepared above. Figure 5B shows that at identical concentrations of free leptin, the presence of leptin-SLR complex did not cause appreciable decrease in STAT3-luc activity, suggesting that SLRs simply sequester leptin away from being available for OB-Rb interaction without a dominant negative role. This result is consistent with earlier studies, which demonstrated that leptin receptors tend to form homodimers but do not form heterodimers (38). This result is also reminiscent of the role of the complex of GH/GH binding protein, which has been demonstrated to inhibit or enhance GH signaling under different conditions (39). The role of SLR on leptin action in vivo is also likely to be similarly complex.

SLR-Leptin Complex Does Not Interfere with OB-Rb Signaling by Free Leptin A, Gel filtration profile of leptin-SLR complexes used to activate OB-Rb HEK293 cells. There are 34% and 88% bound leptin in each sample, containing the same concentration of free leptin (0.08 nm) but different concentrations of leptin-SLR complex, so that total leptin concentration is 0.12 nm and 0.67 nm, respectively. B, Samples as shown in panel A containing the same concentration of free leptin (indicated in bold, final concentration 0.08 nm) but different amounts of leptin-SLR complex were added to OB-Rb HEK293 cells. Supernatant of cells infected with Ad-β-Gal was added to OB-Rb HEK293 cells alone or with the addition of 0.08 nm leptin as negative and positive controls. A different preparation of SLR was used to perform the experiment in Fig. 5 than that used in Fig. 4; thus the volume of SLR added varied between the two figures. STAT3-luc activity and levels of pSTAT3 and total STAT3 were determined as in Fig. 4.
Fig. 5.

SLR-Leptin Complex Does Not Interfere with OB-Rb Signaling by Free Leptin A, Gel filtration profile of leptin-SLR complexes used to activate OB-Rb HEK293 cells. There are 34% and 88% bound leptin in each sample, containing the same concentration of free leptin (0.08 nm) but different concentrations of leptin-SLR complex, so that total leptin concentration is 0.12 nm and 0.67 nm, respectively. B, Samples as shown in panel A containing the same concentration of free leptin (indicated in bold, final concentration 0.08 nm) but different amounts of leptin-SLR complex were added to OB-Rb HEK293 cells. Supernatant of cells infected with Ad-β-Gal was added to OB-Rb HEK293 cells alone or with the addition of 0.08 nm leptin as negative and positive controls. A different preparation of SLR was used to perform the experiment in Fig. 5 than that used in Fig. 4; thus the volume of SLR added varied between the two figures. STAT3-luc activity and levels of pSTAT3 and total STAT3 were determined as in Fig. 4.

DISCUSSION

The ability of leptin to activate its receptor and mediate signal transduction depends on the presence of the signaling form of its receptor, OB-Rb. Leptin activation of OB-Rb centrally causes reduced food intake and increased energy expenditure. These central effects are mediated by a complex neural circuit comprised of neuropeptide Y/agouti-related protein and proopiomelanocortin/cocaine- and amphetamine-regulated transcript neurons, which are modulated by leptin (4042). In peripheral tissues, direct leptin action has also been shown to mediate a wide range of physiologically important, tissue-specific processes, such as wound healing, angiogenesis, T helper cell function, and AMP-activated protein kinase activation (1, 43). We hypothesized that these central and peripheral actions of leptin might be enhanced or inhibited by leptin binding to SLR. To test this prediction, we used an in vitro luciferase reporter assay to determine how free vs. bound leptin affects leptin signal transduction.

Previously, we generated HEK293 cells stably expressing the signaling form of the leptin receptor, OB-Rb (11). Transfection of a luciferase reporter driven by STAT3 responsive elements into these cells allows leptin activation to be assayed by both STAT3 tyrosine phosphorylation and luciferase activity in the lysate (Fig. 1). We sought to determine how STAT3 phosphorylation and STAT3-luc reporter activity are differentially regulated when leptin is added to these cells either alone or in combination with different amounts of SLR. In this report, we demonstrate that using STAT3 tyrosine phosphorylation and STAT3-luc reporter assays, leptin bound to SLR is not capable of activating its long-form receptor, OB-Rb, nor does it inhibit activation of OB-Rb by free leptin. However, when leptin-free SLR is added to a fixed amount of leptin, OB-Rb activation is reduced, presumably by sequestering leptin from productive interactions with OB-Rb. Soluble receptors for a number of cytokines exist and, in many cases, also inhibit the action of these cytokines at their respective target sites (44, 45). These results, coupled with earlier studies showing that SLR is the main leptin-binding activity in plasma (30), have important implications for our understanding of the regulation of the biological activity of leptin in vivo.

Many earlier studies have demonstrated that leptin signals in a variety of peripheral tissues to mediate a variety of physiological functions (1). These tissues are also exposed to SLR. Acute SLR induction is thus likely to inhibit leptin signaling temporally at these sites. The neutralization of leptin action at these sites by SLR could be protective in times of famine. However, in a leptin resistance state, increased amounts of SLR might dampen leptin action when it is needed, potentially contributing to local leptin resistance and the pathogenesis of obesity.

The SLR may also affect leptin’s activity in the brain, in combination with other receptor isoforms (27, 46, 47). To gain access to the central nervous system, leptin needs to cross the blood brain barrier, a process that is saturable, yet not fully defined (25, 4650). SLR might play a role in this process. Further studies to determine the effect of SLR on leptin transport into the central nervous system should shed more light on this very important question.

Prior studies have demonstrated that levels of circulating leptin vary, such as between males and females (51). In humans, serum leptin per unit fat mass is significantly higher at 36 wk of gestation than at 3 and 6 months postpartum (52). In rodents, SLR levels are similarly increased by up to more than 40-fold during late stages of pregnancy (14). Mutation of leptin receptor in Zucker diabetic fatty rat also caused an increase of plasma SLR (31). Our present data suggest that SLR may play a significant role in determining the levels of total leptin and free leptin, as well as leptin’s biological activity. Determination of the levels of free and bound leptin in individuals with hyperleptinemia should be useful in the diagnosis and treatment of certain forms of obesity. Given that leptin acts both centrally, where SLR has not been detected, and peripherally, where both free and bound leptin circulate, the biology of SLR on leptin action is likely to be complex. Analysis of transgenic mice overexpressing SLR, currently underway, may provide additional interesting insights on the role of SLR on leptin action in vivo.

MATERIALS AND METHODS

Reagents

Recombinant leptin was from Sigma Chemical Co. (St. Louis, MO) or from Dr. A. F. Parlow, National Hormone & Peptide Program; murine recombinant 125I-leptin was from Perkin-Elmer Life Sciences (Boston, MA); DMEM, BSA, penicillin, and streptomycin were from Sigma. FUGENE 6 transfection reagent was from Roche (Indianapolis, IN); luciferase assay system was from Promega Corp. (Madison, WI). Enhanced chemiluminescence Western blotting detection reagents and gel filtration columns were from Amersham Biosciences (Piscataway, NJ). Antiphospho-STAT3 antibody was from Cell Signaling Technology, Inc. (Beverly, MA). Antibody against total STAT3 was from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). Bradford assay kit was from Bio-Rad Laboratories, Inc. (Hercules, CA).

Cell Culture

Flp-In HEK293 cell lines stably over-expressing OB-R were generated as described (11). Cells were maintained in DMEM containing 10% fetal bovine serum and 100 U/ml penicillin and 100 μg/ml streptomycin in a 5% CO2, humidified environment at 37 C. The two cell lines used in the current study, encoding OB-Ra and OB-Rb, are schematically diagrammed in Fig. 1A.

[125I]Leptin Binding to Stably Transfected Cells

Leptin binding to Flip-In OB-Rb HEK293 cell lines was performed in six-well plates as described (11). Briefly, cells stably transfected with OB-Ra or OB-Rb were grown to about 90% confluence and washed with cold PBS. Cells were incubated with approximately 60,000 cpm of [125I]leptin alone, or [125I]leptin supplemented with conditioned media containing SLR, in the presence or absence of an excess amount of cold leptin (2 μg/well) for 6 h at 4 C in a final volume of 1 ml binding buffer [PBS containing 1% (wt/vol) BSA (fraction V)]. At the end of incubation, unbound label was removed by two PBS washes; 1 ml of 1 n NaOH was then added, and radioactivity in lysate was measured using a COBRA II AUTO-GAMMA counter from Packard Instruments (Meriden, CT).

Western Blotting Analyses

Cells were lysed in RIPA buffer, containing 150 mm NaCl, 50 mm Tris-HCl, 1% Triton X-100, 2% Igepal CA-630, 1 mm phenylmethylsulfonyl fluoride, 1 mm Na3VO4, 1 mm NaF, 10 μg/ml leupeptin, and 10 μg/ml aprotinin. Lysate was incubated on ice for 20 min with gentle rocking and centrifuged at 13,000 × g for 15 min at 4 C. Protein concentration was determined by Bradford assay. Protein samples were boiled in 2× reducing sample buffer and subjected to SDS-PAGE, followed by electrophoretic transfer to nitrocellulose membrane. After incubation with antibodies, target protein was detected using enhanced chemiluminescence, following the manufacturer’s instructions.

STAT3-Driven Luciferase Assay

When cells were approximately 80% confluent, transfection was performed by using FUGENE 6 Transfection Reagent with 0.5 μg/well STAT3-luc DNA, p4Xm67, containing four copies of the STAT3 binding sequences fused to luciferase cDNA (11). Two days after transfection, media were aspirated from cells and replaced with DMEM containing 0.5% BSA. After overnight incubation under serum-free conditions, cells were stimulated with different concentrations of leptin. Cells were harvested 15 min after leptin stimulation to determine the level of STAT3 tyrosine phosphorylation or after 3 h to determine STAT3-luc activity.

Preparation of Conditioned Media Containing SLR

HEK293 cells were grown in 15-cm plates until 90% confluent. Cells were washed with PBS, and then 20 ml of DMEM without serum were added. Purified recombinant adenoviruses encoding OB-Re (Ad-OB-Re) or β-galactosidase (Ad-β-Gal) (20 μl), each with a physical titer of 4 × 1012 particles/ml, were added to each plate. Two days after virus infection, medium was collected and filtered through a 0.22-μm syringe filter. Expression of SLR was verified by Western blotting.

Titration of Leptin Binding to SLR and Determination of Free and Bound Leptin by FPLC

HEK293 cells were cultured in serum-free DMEM at 37 C, 5% CO2, and infected with Ad-OB-Re or Ad-β-Gal. After incubation for 3 d, conditioned medium was harvested and mixed with cold recombinant mouse leptin and tracer [125I]leptin. Murine recombinant [125I]leptin with a specific activity of 2200 Ci/mmol (81.4 TBq/mmol) was used as a tracer. In separate 1.5-ml Eppendorf tubes, 0, 8, 16, and 80 μl of conditioned medium from infected HEK293 cells were mixed with 1.6 ng cold leptin and 2 μl [125I]leptin and incubated at 4 C overnight in a total volume of 1 ml. Subsequently, the mixture was analyzed by FPLC, using an AKTA design chromatography system from Amersham Biosciences. Gel filtration was performed on a Superdex 200 HR (10/30) column, equilibrated and eluted with the same buffer (0.05 m NaPO4 and 0.15 m NaCl, pH 7.4) at 4 C. The 1-ml mixture (200 μl) was loaded onto the column, and 80 fractions were collected, each having a volume of 300 μl. The amount of radioactivity in each fraction was determined by the use of a COBRA II AUTO-GAMMA counter. The areas under the curve of each of the two peaks, corresponding to the elution position of bound and free leptin, were calculated with the software Prism (GraphPad Software, Inc., San Diego, CA). Based on the assumption that tracer distribution reflects that of cold leptin, we obtained the absolute amount of bound and free leptin in the mixture by multiplying the percentage of free and bound leptin with the amount of total leptin added in each incubation mixture. All data were analyzed with Prism 3 software.

Acknowledgments

We thank Drs. Roger Unger and Joyce Repa for many thoughtful comments and a critical review of this manuscript; and Drs. Jeff Friedman and Paul Cohen (Rockefeller University) for many discussions and sharing of unpublished data.

This work was supported by Grant 0255737Y from the American Heart Association, Texas Affiliate, and National Institutes of Health Grant DK 60137. C.L. is the recipient of a Career Development Award from the American Diabetes Association.

Abbreviations

     
  • FPLC

    Fast protein liquid chromatography;

  •  
  • HEK

    human embryonic kidney;

  •  
  • SLR

    soluble leptin receptor;

  •  
  • STAT

    signal transducer and activator of transcription.

1

Huang
L
,
Li
C

2000
Leptin: a multifunctional hormone.
Cell Res
10
:
81
92
2

Friedman
JM
,
Halaas
JL

1998
Leptin and the regulation of body weight in mammals.
Nature
395
:
763
770
3

Li
C
,
Ioffe
E
,
Fidahusein
N
,
Connolly
E
,
Friedman
JM

1998
Absence of soluble leptin receptor in plasma from dbPas/dbPas and other db/db mice.
J Biol Chem
273
:
10078
10082
4

Chua Jr
SC
,
Koutras
IK
,
Han
L
,
Liu
SM
,
Kay
J
,
Young
SJ
,
Chung
WK
,
Leibel
RL

1997
Fine structure of the murine leptin receptor gene: splice site suppression is required to form two alternatively spliced transcripts.
Genomics
45
:
264
270
5

Diamond
FB
,
Eichler
DC
,
Duckett
G
,
Jorgensen
EV
,
Shulman
D
,
Root
AW

1997
Demonstration of a leptin binding factor in human serum.
Biochem Biophys Res Commun
233
:
818
822
6

Houseknecht
KL
,
Mantzoros
CS
,
Kuliawat
R
,
Hadro
E
,
Flier
JS
,
Kahn
BB

1996
Evidence for leptin binding to proteins in serum of rodents and humans: modulation with obesity.
Diabetes
45
:
1638
1643
7

Sinha
MK
,
Opentanova
I
,
Ohannesian
JP
,
Kolaczynski
JW
,
Heiman
ML
,
Hale
J
,
Becker
GW
,
Bowsher
RR
,
Stephens
TW
,
Caro
JF

1996
Evidence of free and bound leptin in human circulation. Studies in lean and obese subjects and during short-term fasting.
J Clin Invest
98
:
1277
1282
8

Chan
JL
,
Bluher
S
,
Yiannakouris
N
,
Suchard
MA
,
Kratzsch
J
,
Mantzoros
CS

2002
Regulation of circulating soluble leptin receptor levels by gender, adiposity, sex steroids, and leptin: observational and interventional studies in humans.
Diabetes
51
:
2105
2112
9

Lahlou
N
,
Issad
T
,
Lebouc
Y
,
Carel
JC
,
Camoin
L
,
Roger
M
,
Girard
J

2002
Mutations in the human leptin and leptin receptor genes as models of serum leptin receptor regulation.
Diabetes
51
:
1980
1985
10

Lahlou
N
,
Clement
K
,
Carel
JC
,
Vaisse
C
,
Lotton
C
, Le
Bihan
Y
,
Basdevant
A
,
Lebouc
Y
,
Froguel
P
,
Roger
M
,
Guy-Grand
B

2000
Soluble leptin receptor in serum of subjects with complete resistance to leptin: relation to fat mass.
Diabetes
49
:
1347
1352
11

Ge
H
,
Huang
L
,
Pourbahrami
T
,
Li
C

2002
Generation of soluble leptin receptor by ectodomain shedding of membrane-spanning receptors in vitro and in vivo.
J Biol Chem
277
:
45898
45903
12

Maamra
M
,
Bidlingmaier
M
,
Postel-Vinay
MC
,
Wu
Z
,
Strasburger
CJ
,
Ross
RJ

2001
Generation of human soluble leptin receptor by proteolytic cleavage of membrane-anchored receptors.
Endocrinology
142
:
4389
4393
13

Lammert
A
,
Brockmann
G
,
Renne
U
,
Kiess
W
,
Bottner
A
,
Thiery
J
,
Kratzsch
J

2002
Different isoforms of the soluble leptin receptor in non-pregnant and pregnant mice.
Biochem Biophys Res Commun
298
:
798
804
14

Gavrilova
O
,
Barr
V
,
Marcus-Samuels
B
,
Reitman
M

1997
Hyperleptinemia of pregnancy associated with the appearance of a circulating form of the leptin receptor.
J Biol Chem
272
:
30546
30551
15

Kratzsch
J
,
Lammert
A
,
Bottner
A
,
Seidel
B
,
Mueller
G
,
Thiery
J
,
Hebebrand
J
,
Kiess
W

2002
Circulating soluble leptin receptor and free leptin index during childhood, puberty, and adolescence.
J Clin Endocrinol Metab
87
:
4587
4594
16

Monteleone
P
,
Fabrazzo
M
,
Tortorella
A
,
Fuschino
A
,
Maj
M

2002
Opposite modifications in circulating leptin and soluble leptin receptor across the eating disorder spectrum.
Mol Psychiatry
7
:
641
646
17

Monteleone
P
, Di
Lieto
A
,
Tortorella
A
,
Longobardi
N
,
Maj
M

2000
Circulating leptin in patients with anorexia nervosa, bulimia nervosa or binge-eating disorder: relationship to body weight, eating patterns, psychopathology and endocrine changes.
Psychiatry Res
94
:
121
129
18

Laimer
M
,
Ebenbichler
CF
,
Kaser
S
,
Sandhofer
A
,
Weiss
H
,
Nehoda
H
,
Aigner
F
,
Patsch
JR

2002
Weight loss increases soluble leptin receptor levels and the soluble receptor bound fraction of leptin.
Obes Res
10
:
597
601
19

van Dielen
FM
, van ’t
Veer
C
,
Buurman
WA
,
Greve
JW

2002
Leptin and soluble leptin receptor levels in obese and weight-losing individuals.
J Clin Endocrinol Metab
87
:
1708
1716
20

Schulze
PC
,
Kratzsch
J
,
Linke
A
,
Schoene
N
,
Adams
V
,
Gielen
S
,
Erbs
S
,
Moebius-Winkler
S
,
Schuler
G

2003
Elevated serum levels of leptin and soluble leptin receptor in patients with advanced chronic heart failure.
Eur J Heart Fail
5
:
33
40
21

Lewandowski
K
,
Horn
R
,
O’Callaghan
CJ
,
Dunlop
D
,
Medley
GF
,
O’Hare
P
,
Brabant
G

1999
Free leptin, bound leptin, and soluble leptin receptor in normal and diabetic pregnancies.
J Clin Endocrinol Metab
84
:
300
306
22

Liu
C
,
Liu
XJ
,
Barry
G
,
Ling
N
,
Maki
RA
, De
Souza
EB

1997
Expression and characterization of a putative high affinity human soluble leptin receptor.
Endocrinology
138
:
3548
3554
23

Fong
TM
,
Huang
RR
,
Tota
MR
,
Mao
C
,
Smith
T
,
Varnerin
J
,
Karpitskiy
VV
,
Krause
JE
, Van der
Ploeg
LH

1998
Localization of leptin binding domain in the leptin receptor.
Mol Pharmacol
53
:
234
240
24

Kowalski
TJ
,
Liu
SM
,
Leibel
RL
,
Chua
SC

2001
Transgenic complementation of leptin-receptor deficiency. I. Rescue of the obesity/diabetes phenotype of LEPR-null mice expressing a LEPR-B transgene.
Diabetes
50
:
425
435
25

Zlokovic
BV
,
Jovanovic
S
,
Miao
W
,
Samara
S
,
Verma
S
,
Farrell
CL

2000
Differential regulation of leptin transport by the choroid plexus and blood-brain barrier and high affinity transport systems for entry into hypothalamus and across the blood-cerebrospinal fluid barrier.
Endocrinology
141
:
1434
1441
26

Golden
PL
,
Maccagnan
TJ
,
Pardridge
WM

1997
Human blood-brain barrier leptin receptor. Binding and endocytosis in isolated human brain microvessels.
J Clin Invest
99
:
14
18
27

Hileman
SM
,
Pierroz
DD
,
Masuzaki
H
,
Bjorbaek
C
,
El-Haschimi
K
,
Banks
WA
,
Flier
JS

2002
Characterizaton of short isoforms of the leptin receptor in rat cerebral microvessels and of brain uptake of leptin in mouse models of obesity.
Endocrinology
143
:
775
783
28

Hileman
SM
,
Tornoe
J
,
Flier
JS
,
Bjorbaek
C

2000
Transcellular transport of leptin by the short leptin receptor isoform ObRa in Madin-Darby canine kidney cells.
Endocrinology
141
:
1955
1961
29

Landt
M
,
Parvin
CA
,
Wong
M

2000
Leptin in cerebrospinal fluid from children: correlation with plasma leptin, sexual dimorphism, and lack of protein binding.
Clin Chem
46
:
854
858
30

Lammert
A
,
Kiess
W
,
Bottner
A
,
Glasow
A
,
Kratzsch
J

2001
Soluble leptin receptor represents the main leptin binding activity in human blood.
Biochem Biophys Res Commun
283
:
982
988
31

Huang
L
,
Wang
Z
,
Li
C

2001
Modulation of circulating leptin levels by its soluble receptor.
J Biol Chem
276
:
6343
6349
32

Li
C
,
Friedman
JM

1999
Leptin receptor activation of SH2 domain containing protein tyrosine phosphatase 2 modulates Ob receptor signal transduction.
Proc Natl Acad Sci USA
96
:
9677
9682
33

Bjorbak
C
,
Buchholz
RM
,
Davis
SM
,
Bates
SH
,
Pierroz
DD
,
Gu
H
,
Neel
BG
, Myers
Jr
MG
,
Flier
JS

2000
The role of SHP-2 in MAPK activation by leptin receptors.
J Biol Chem
276
:
4747
4755
34

Vaisse
C
,
Halaas
JL
,
Horvath
CM
, Darnell
Jr
JE
,
Stoffel
M
,
Friedman
JM

1996
Leptin activation of Stat3 in the hypothalamus of wild-type and ob/ob mice but not db/db mice.
Nat Genet
14
:
95
97
35

Carpenter
LR
,
Farruggella
TJ
,
Symes
A
,
Karow
ML
,
Yancopoulos
GD
,
Stahl
N

1998
Enhancing leptin response by preventing SH2-containing phosphatase 2 interaction with Ob receptor.
Proc Natl Acad Sci USA
95
:
6061
6066
36

Baumann
H
,
Morella
KK
,
White
DW
,
Dembski
M
,
Bailon
PS
,
Kim
H
,
Lai
CF
,
Tartaglia
LA

1996
The full-length leptin receptor has signaling capabilities of interleukin 6-type cytokine receptors.
Proc Natl Acad Sci USA
93
:
8374
8378
37

Barr
VA
,
Lane
K
,
Taylor
SI

1999
Subcellular localization and internalization of the four human leptin receptor isoforms.
J Biol Chem
274
:
21416
21424
38

Devos
R
,
Guisez
Y
, Van der
Heyden
J
,
White
DW
,
Kalai
M
,
Fountoulakis
M
,
Plaetinck
G

1997
Ligand-independent dimerization of the extracellular domain of the leptin receptor and determination of the stoichiometry of leptin binding.
J Biol Chem
272
:
18304
18310
39

Baumann
G

2002
Growth hormone binding protein. The soluble growth hormone receptor.
Minerva Endocrinol
27
:
265
276
40

DeFalco
J
,
Tomishima
M
,
Liu
H
,
Zhao
C
,
Cai
X
,
Marth
JD
,
Enquist
L
,
Friedman
JM

2001
Virus-assisted mapping of neural inputs to a feeding center in the hypothalamus.
Science
291
:
2608
2613
41

Cohen
P
,
Zhao
C
,
Cai
X
,
Montez
JM
,
Rohani
SC
,
Feinstein
P
,
Mombaerts
P
,
Friedman
JM

2001
Selective deletion of leptin receptor in neurons leads to obesity.
J Clin Invest
108
:
1113
1121
42

Friedman
JM

2000
Obesity in the new millennium.
Nature
404
:
632
634
43

Minokoshi
Y
,
Kim
YB
,
Peroni
OD
,
Fryer
LG
,
Muller
C
,
Carling
D
,
Kahn
BB

2002
Leptin stimulates fatty-acid oxidation by activating AMP-activated protein kinase.
Nature
415
:
339
343
44

Kotenko
SV
,
Izotova
LS
,
Mirochnitchenko
OV
,
Esterova
E
,
Dickensheets
H
,
Donnelly
RP
,
Pestka
S

2001
Identification, cloning, and characterization of a novel soluble receptor that binds IL-22 and neutralizes its activity.
J Immunol
166
:
7096
7103
45

Muller-Newen
G
,
Kohne
C
,
Heinrich
PC

1996
Soluble receptors for cytokines and growth factors.
Int Arch Allergy Immunol
111
:
99
106
46

Banks
WA
,
Niehoff
ML
,
Martin
D
,
Farrell
CL

2002
Leptin transport across the blood-brain barrier of the Koletsky rat is not mediated by a product of the leptin receptor gene.
Brain Res
950
:
130
136
47

Kastin
AJ
,
Pan
W
,
Maness
LM
,
Koletsky
RJ
,
Ernsberger
P

1999
Decreased transport of leptin across the blood-brain barrier in rats lacking the short form of the leptin receptor.
Peptides
20
:
1449
1453
48

Banks
WA
,
Clever
CM
,
Farrell
CL

2000
Partial saturation and regional variation in the blood-to-brain transport of leptin in normal weight mice.
Am J Physiol
278
:
E1158
E1165
49

Banks
WA

2001
Leptin transport across the blood-brain barrier: implications for the cause and treatment of obesity.
Curr Pharm Des
7
:
125
33
50

Kastin
AJ
,
Akerstrom
V

2001
Glucose and insulin increase the transport of leptin through the blood-brain barrier in normal mice but not in streptozotocin-diabetic mice.
Neuroendocrinology
73
:
237
242
51

Comuzzie
AG
,
Hixson
JE
,
Almasy
L
,
Mitchell
BD
,
Mahaney
MC
,
Dyer
TD
,
Stern
MP
,
MacCluer
JW
,
Blangero
J

1997
A major quantitative trait locus determining serum leptin levels and fat mass is located on human chromosome 2.
Nat Genet
15
:
273
276
52

Butte
NF
,
Hopkinson
JM
,
Nicolson
MA

1997
Leptin in human reproduction: serum leptin levels in pregnant and lactating women.
J Clin Endocrinol Metab
82
:
585
589