当前位置: > 正文
编号:11370640
A foldable CFTRF508 biogenic intermediate accumulates upon inhibition
http://www.100md.com 《细胞学杂志》
     1 Department of Cell and Developmental Biology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599

    2 The Cystic Fibrosis Center, School of Medicine, University of North Carolina, Chapel Hill, NC 27599

    Correspondence to D.M. Cyr: dmcyr@med.unc.edu

    Abstract

    CFTRF508 exhibits a correctable protein-folding defect that leads to its misfolding and premature degradation, which is the cause of cystic fibrosis (CF). Herein we report on the characterization of the CFTRF508 biogenic intermediate that is selected for proteasomal degradation and identification of cellular components that polyubiquitinate CFTRF508. Nonubiquitinated CFTRF508 accumulates in a kinetically trapped, but folding competent conformation, that is maintained in a soluble state by cytosolic Hsc70. Ubiquitination of Hsc70-bound CFTRF508 requires CHIP, a U box containing cytosolic cochaperone. CHIP is demonstrated to function as a scaffold that nucleates the formation of a multisubunit E3 ubiquitin ligase whose reconstituted activity toward CFTR is dependent upon Hdj2, Hsc70, and the E2 UbcH5a. Inactivation of the Hsc70–CHIP E3 leads CFTRF508 to accumulate in a nonaggregated state, which upon lowering of cell growth temperatures, can fold and reach the cell surface. Inhibition of CFTRF508 ubiquitination can increase its cell surface expression and may provide an approach to treat CF.

    Abbreviations used in this paper: ApoB48, apolipoprotein B48; BFA, brefeldin A; CF, cystic fibrosis; ERQC, endoplasmic reticulum quality control system; NBD, nucleotide binding domain; TCR, T cell receptor subunit; TPR, tetratricopeptide repeat motifs.

    Introduction

    CFTR is a Cl– ion channel that is localized to the apical surface of epithelial cells that line lung airways and glands (Riordan et al., 1989). Cystic fibrosis (CF) is an autosomal recessive disease and most CF patients inherit at least one CFTRF508 mutant allele. CFTRF508 lacks F508 in nucleotide binding domain (NBD) I and has a temperature-sensitive folding defect, which causes its premature degradation by the ubiquitin proteasome system (Denning et al., 1992; Jensen et al., 1995a; Ward et al., 1995). Loss of CFTR function at the cell surface leads to mortality in CF patients because of altered hydration of airway epithelia and persistent lung infections (Welsh and Smith, 1993).

    An interesting feature of CFTRF508 is that when growth conditions are altered, it can fold, escape the endoplasmic reticulum quality control system (ERQC), and function at the cell surface (Denning et al., 1992; Brown et al., 1996). Thus, the development of agents that promote the folding or block the degradation of nascent CFTRF508 has the potential to provide a therapeutic avenue for the treatment of CF. Rational design of such therapeutics requires a basic understanding of the mechanism for CFTRF508 misfolding and the identification of the ERQC machinery that selects CFTRF508 for degradation.

    CFTR is a 1,480 residue glycomembrane protein whose proper function requires the formation of intramolecular contacts between its two transmembrane domains, two cytoplasmic NBDs, and a regulatory domain (R-domain; Xiong et al., 1997). CFTR and CFTRF508 biogenesis is inefficient with 60–75% of CFTR and nearly 99% of CFTRF508 being degraded before reaching the cell surface (Ward and Kopito, 1994). Thus, the kinetics of CFTR and CFTRF508 folding are slow and nonnative biogenic intermediates of each appear to be actively selected for degradation by ERQC systems. The nature of the CFTRF508 biogenic intermediate that is selected for degradation is unknown, but CFTR and CFTRF508 appear to assume similar conformations at early stages of assembly (Zhang et al., 1998). Because F508 is located on the surface of NBD1, CFTRF508 assembly is proposed to go off pathway at a late stage (Lewis et al., 2004). The inability of CFTRF508 to fold properly makes it prone to aggregation (Qu and Thomas, 1996) and causes some of its degradation intermediates to accumulate in detergent-insoluble aggregates (Ward et al., 1995).

    The mechanism by which the cell monitors the conformational state of CFTRF508 and makes protein triage decisions that determine its fate are unclear. Current models suggest that the folded state of CFTR and CFTRF508 is surveyed by the cytosolic chaperone Hsc70 and Hsp90 (Yang et al., 1993; Loo et al., 1998; Zhang et al., 2001). The ER luminal lectin-binding chaperone calnexin can form a complex with the immaturely glycosylated B form of CFTRF508, but a direct role for calnexin in CFTR folding and/or degradation has not been demonstrated (Pind et al., 1994; Okiyoneda et al., 2004).

    Cytosolic Hsc70 functions in complexes with either folding or degradatory cochaperones to mediate steps in CFTR folding and degradation (Cyr et al., 2002). The type I Hsp40 cochaperone Hdj-2 is farnesylated and localized to the cytoplasmic face of the ER where it recruits Hsc70 to bind ribosome associated CFTR to promote early stages of its assembly (Meacham et al., 1999). Hsc70 can also interact with the degradatory cochaperone CHIP to facilitate the proteasomal degradation of ER forms of CFTR and CFTRF508 (Meacham et al., 2001). Thus, cochaperones of Hsc70 play a central role in determining the fate of nascent CFTR.

    Mechanistic insight into how CHIP functions as a degradatory cochaperone is now required to understand how the cell makes protein triage decisions for CFTRF508. CHIP contains three NH2-terminal tetratricopeptide repeat motifs (TPR) that bind the COOH terminus of Hsc70 (Scheufler et al., 2000) and a noncanonical RING domain, termed the U box, which promotes interactions with E2 enzymes (Ballinger et al., 1999). The TPR repeat motifs and U box are essential for CHIP to mediate CFTR and CFTRF508 degradation (Meacham et al., 2001). Thus, CHIP is proposed to interact with Hsc70 to form an E3 ubiquitin ligase that targets CFTRF508 for proteasomal degradation. However, CHIP can function in a U box independent manner to alter the Hsc70 polypeptide binding and release cycle and negatively influence the folding of some client proteins (Ballinger et al., 1999; Dai et al., 2003). Thus, it is plausible that CHIP targets CFTR and CFTRF508 for degradation by acting as an antifolding factor.

    Herein, we reconstituted CFTR ubiquitination with purified components and demonstrated that Hsc70 and CHIP functionally interact with the E2 UbcH5a to form a multisubunit E3 ubiquitin ligase that polyubiquitinates the cytosolic subdomains of CFTR. Inactivation of the Hsc70–CHIP E3 in cultured cells drove the accumulation of a nonaggregated and foldable CFTRF508 degradation intermediate. These data support a model for quality control in which aggregation of CFTRF508 is suppressed by Hsc70 and Hdj-2. However, when CFTRF508 folding intermediates become kinetically trapped in a nonnative state, CHIP attracts UbcH5a to Hsc70–CFTRF508 complexes and thereby facilitates CFTRF508 ubiquitination and degradation.

    Results

    Purified CHIP functions as an Hdj-2–, Hsc70-, and UbcH5a-dependent E3 ubiquitin ligase to polyubiquitinate CFTR

    To investigate whether CHIP targets CFTR for degradation by acting as a U box–dependent E3 ubiquitin ligase we sought to reconstitute CFTR ubiquitination. To accomplish this goal CHIP, Hsc70, Hdj-2, and the E2 UbcH5a were overexpressed and purified from Escherichia coli. UbcH5a was chosen as the E2 for these experiments because it cooperates with CHIP to promote polyubiquitin chain assembly (Jiang et al., 2001). The CFTR substrate used for this ubiquitination reaction was gst–NBD1–R (Naren et al., 1999), which contains binding sites for Hdj-2 and Hsc70 (Meacham et al., 1999).

    Gst–NBD1–R was incubated with different combinations of chaperones and ubiquitination enzymes and its ubiquitination was monitored by analyzing the retardation of its mobility on SDS-PAGE gels (Fig. 1 A). E1 and UbcH5a were unable to facilitate the ubiquitination of gst–NBD1–R and the addition of CHIP lead to the formation of a small quantity of ubiquitinated gst–NBD1–R. The presence of Hsc70 or Hdj-2 in combination with CHIP further stimulated the ubiquitination by UbcH5a, but the efficiency of this reaction remained low with <3% of gst–NBD1–R being detected as a mono-, di-, or triubiquitinated species. However, >85% of gst–NBD1–R was polyubiquitinated when E1, UbcH5a, Hsc70, Hdj-2, and CHIP were jointly present. Mutation of conserved residues in CHIPs U box, H260A and P269A, greatly reduced gst–NBD1–R polyubiquitination. In addition, mutation of K30A in the TPR repeat domain reduced CHIPs ubiquitination activity to levels observed when CHIP and Hdj-2 were present, but Hsc70 was omitted (Fig. 1, A and C). CHIP cooperates with UbcH5a, Hsc70, and Hdj-2 in a TPR repeat and U box–dependent manner to facilitate CFTR polyubiquitination.

    Figure 1. Reconstitution of CFTR ubiquitination. (A) Ubiquitination of gst–NBD1–R. (B) Ubc6 and Ubc7 cannot cooperate with Hsc70 and CHIP to ubiquitinate gst–NBD1–R. (C) CHIP K30A is defective in polyubiquitination. (D) UbcH5a C85A cannot ubiquitinate Gst–NBD–R. Ubiquitination of Gst–NBD–R (1 μM) was performed at 37°C and where indicated the following components were present: UbcH5a (4 μM), UbcH5a C85A (4 μM), Ubc6 (4 μM), Ubc7 (4 μM), CHIP (3 μM), CHIP K30A (3 μM), Hsc70 (2 μM), and Hdj2 (4 μM). In 1B-D Hsc70 and Hdj-2 were in all reactions except for lane 1. Incubations in A, C, and D were for 2 h. Ubiquitination of Gst–NBD–R was determined by Western blot with -R domain antibody. In the x-ray films shown in A–D the band that corresponds to Gst–NBD–R is overexposed to allow for the visualization of its ubiquitinated forms. Quantitation of shorter exposures of these x-ray films indicates that >85% of Gst–NBD–R is polyubiquitinated by the joint action of UbcH5a, Hsc70, Hdj-2, and CHIP (A, lane 5). When Hsc70 or Hdj-2 were absent (A, lanes 2 and 3), UbcH5a and CHIP ubiquitinated <3% of Gst–NBD–R.

    U box proteins function as E3 enzymes to stimulate E2 dependent polyubiquitin chain assembly and in some cases act as E4 enzymes to elongate ubiquitin chains on monoubiquitinated proteins (Koegl et al., 1999; Cyr et al., 2002). The data presented indicates that CHIP acts in concert with Hsc70 and Hdj-2 as an E3 to stimulate UbcH5a ubiquitination activity. This conclusion is supported by the observation that UbcH5a does not monoubiquitinate, and CHIP cooperates with Hsc70 and Hdj-2 to stimulate the rate at which UbcH5a converts gst–NBD1–R into a polyubiquitinated species (Fig. S1).

    To evaluate the specificity of CHIPs ubiquitination activity, its ability to cooperate with the mammalian E2s Ubc6 and Ubc7 (Tiwari and Weissman, 2001; Lenk et al., 2002) to facilitate gst–NBD1–R ubiquitination was examined (Fig. 1 B). Ubc6 and Ubc7 were used because these E2s mediate the ubiquitination of ERAD substrates in cultured cells and their purified forms function in vitro to facilitate polyubiquitin chain assembly (Tiwari and Weissman, 2001). In reaction cocktails that contained E1, UbcH5a, Hsc70, Hdj2, and CHIP, gst–NBD1–R was converted to a polyubiquitinated species in time-dependent fashion (Fig. 1 B). However, after 180 min of incubation a polyubiquitinated form of gst–NBD1–R was not detected when the purified E2 domain of Ubc6 or Ubc7 was substituted for UbcH5a in otherwise identical reaction cocktails. Thus, CHIP appears to specifically recognize UbcH5a.

    Next, the ability of UbcH5a C85A to facilitate the polyubiquitination of gst–NBD1–R was determined (Fig. 1 D). UbcH5a C85A is a form of UbcH5a in which its active site cysteine that accepts a charged ubiquitin from E1 has been mutated. UbcH5a C85A is predicted to interact with the U box on CHIP, but should not facilitate polyubiquitin chain assembly because it cannot be conjugated to ubiquitin. This supposition was found to be true as UbcH5a C85A was unable to cooperate with CHIP to promote the polyubiquitination of gst–NBD1–R. Hence, the Hsc70- and Hdj-2–dependent reconstitution of CFTR polyubiquitination requires the action of the CHIP TPR and U box domains and the active site cysteine of UbcH5a.

    Overexpression of UbcH5a C85A in cultured cells inhibits CFTR and CFTRF508 degradation

    To access whether or not UbcH5a is an in vivo component of Hsc70–CHIP E3, the effect that UbcH5a and UbcH5a C85A overexpression in HEK293 cells had on CFTR biogenesis was determined (Fig. 2 A). In addition, we compared the effect of UbcH5a or UbcH5a C85A overexpression on CFTR biogenesis to that of wild-type and dominant negative mutant forms of the human E2s Ubc6 and Ubc7 (Fig. 2 A). Elevation of UbcH5a levels caused a decrease in the accumulation of the immaturely glycosylated ER localized B form and the maturely glycosylated plasma membrane localized C form of CFTR. In contrast, overexpression of UbcH5a C85A led to a severalfold increase in the steady-state level of the B and C form of CFTR. Ubc6 and Ubc6 C91S overexpression were also observed to influence CFTR expression levels, but the effect that Ubc6 C91S had on the accumulation of the B form of CFTR was modest when compared with results obtained with UbcH5a C85A. On the other hand, Ubc7 and Ubc7 C89S overexpression did not cause a detectable change in the steady-state level of CFTR and CFTRF508. Because Ubc6 does not appear to interact with CHIP (Fig. 1), the effect that its overexpression has on CFTR biogenesis appears to result from its ability to function with additional quality control factors that monitor the folded state of CFTR (Gnann et al., 2004).

    Figure 2. Degradation of CFTR and CFTRF508 is inhibited by overexpression of UbcH5A C85A. (A) Western blot analysis of CFTR levels upon overexpression of the E2s UbcH5A, Ubc6, and Ubc7. HEK293 cells were transfected transiently with expression plasmids that encode the indicated proteins. Total cell extracts were prepared 24 h after transfection in SDS-PAGE sample buffer. Nitrocellulose was probed with -CFTR and developed. The immaturely glycosylated ER localized B form and maturely glycosylated plasma membrane associated C form of CFTR are denoted as B and C, respectively. (B and C) UbcH5A C85A overexpression slows the rate of CFTR and CFTRF508 degradation. Cells were labeled for 20 min with 35S-translabel and a chase period was initiated by the addition of cycloheximide. At the indicated times, 35S-CFTR or 35S-CFTRF508 was immunoprecipitated from cell extracts. CFTR or CFTRF508 isolated in this manner was detected by SDS-PAGE and fluorography. (D and E) Graphs illustrate the processing efficiency and half-life of CFTR and CFTRF508. Relative CFTR and CFTRF508 levels were quantitated by laser densitometry of the x-ray films shown in B and C, respectively. Values were normalized to the quantity of the B form of CFTR and CFTRF508 present at t = 0 under the indicated experimental condition levels. t = 0 values for the B form of CFTR were 1.1 and 1.8 OD, in the absence and presence of UbcH5a C85A, respectively. t = 0 values for the B form of CFTRF508 were 2.3 and 7.0 OD, in the absence and presence of UbcH5a C85A, respectively. (F) CHIP and UbcH5a jointly reduce levels of CFTRF508 in Western blots.

    To explore the reason why overexpression of UbcH5a C85A drove the B form of CFTR to accumulate, its effect on the kinetics of CFTR and CFTRF508 degradation was determined (Fig. 2, B and C). In pulse-chase experiments, UbcH5a C85A overexpression increased the quantity of the B form of CFTR and CFTRF508 present at the beginning of the chase period 1.5–3-fold (Fig. 2, legend). The UbcH5a C85A induced increase in CFTR and CFTRF508 levels appeared to result from the impaired degradation of nascent CFTR and CFTR508 because the half-life of the B form of each was increased two to threefold (Fig. 2, D and E). In addition, the maturation efficiency of CFTR in the presence or absence of UbcH5a C85A was around 25% (Fig. 2 D). Thus, inhibition of the Hsc70–CHIP E3 complex via UbcH5a C85A overexpression inhibits CFTR degradation, but does not interfere with CFTR folding efficiency.

    To gain additional support for the interpretation that CHIP and UbcH5a interact with each other to select nascent CFTR and CFTRF508 for degradation we demonstrated that the coexpression of CHIP and UbcH5a enhanced the effect that individual forms of each had on CFTRF508 degradation (Fig. 2 D). When CHIP or UbcH5a was overexpressed alone, steady-state levels of CFTRF508 were reduced to 30 and 41% of control levels, respectively. Yet, when CHIP and UbcH5a were coexpressed, CFTRF508 accumulation was reduced by >98%.

    Overexpression of Ubch5a C85A does not generally inhibit ERAD

    To ascertain whether or not UbcH5a C85A overexpression generally inhibited ERAD or specifically blocked CFTR degradation, its effect on the degradation of the T cell receptor subunit (TCR) was examined (Fig. 3 A). TCR is a transmembrane protein that exposes a large extracellular domain in the ER lumen whose unassembled form is degraded via an ERAD pathway that uses the E2s Ubc6 and Ubc7 (Tiwari and Weissman, 2001; Lenk et al., 2002). Pulse chase analysis revealed that TCR had a half-life of around 1 h in absence or presence of UbcH5a C85A (Fig. 3, A and C). In addition, the overexpression of CHIP was not observed to influence the rate of TCR turnover (unpublished data). In contrast, overexpression of either Ubc6 C91S or Ubc7 C89S led to an increase in the half-life of TCR from 1 h to 1.5 and 2 h, respectively. Thus, UbcH5a C85A overexpression does not detectably hinder the turnover of a transmembrane ERAD substrate whose degradation can be blocked by interference with the action of Ubc6 and Ubc7.

    Figure 3. Insensitivity of TCR and ApoB48 turnover to inhibition of the Hsc70–CHIP E3. (A) TCR degradation. (B) ApoB48 turnover. HEK293 cells were transiently transfected with expression plasmids for the indicated proteins. 24 h after transfection, cells were labeled for 20 min with 35S-translabel. A chase period was initiated by the addition of cycloheximide and, at the indicated times, cells were harvested and lysed. TCR or ApoB48 were immunoprecipitated from cell extracts and detected by SDS-PAGE and autoradiography. (C) Quantitation of TCR and Apob48 levels, the relative amount of each protein present at t = 0 is expressed as 100% of control.

    Because the membrane topology of TCR differs from that of CFTR, we also examined the sensitivity of apolipoprotein B48 (ApoB48) degradation to overexpression of UbcH5a C85A. ApoB48 is a 2,15–amino acid residue secretory protein whose nascent form has features that are similar to CFTR because it exposes surfaces in the ER lumen and cytosol, and is a substrate of cytosolic Hsp70 and Hsp90 (Gusarova et al., 2001). ApoB48 folding and exit from the ER requires its assembly into complexes with lipids, and unassembled forms are degraded via a pathway that involves the transmembrane E3 Gp78, which can interact with Ubc7 (Cyr et al., 2002; Liang et al., 2003). The overexpression of CHIP does not accelerate Apob48 degradation (Meacham et al., 2001). Likewise, the overexpression of UbcH5a C85A does not have a detectable effect on the rate of ApoB48 degradation (Fig. 3, B and C). Thus, the overexpression of UbcH5 C85A does not generally inhibit the function the cellular quality control machinery.

    A detergent-soluble CFTRF508 degradation intermediate accumulates upon inactivation of the Hsc70–CHIP E3 complex

    CFTR and CFTRF508 biogenic intermediates appear to be aggregation prone and therefore are selected for ubiquitination and proteasomal degradation. When the proteasome is inhibited, polyubiquitinated CFTRF508 accumulates in detergent-insoluble aggregates (Ward et al., 1995), but nonubiquitinated CFTRF508 biogenic intermediates are not well characterized. To investigate the aggregation state of nonubiquitinated CFTRF508, we modulated the activity of the Hsc70–CHIP E3 and determined the detergent solubility of the CFTR and CFTRF508 biogenic intermediates that accumulated (Fig. 4). Overexpression of UbcH5a and CHIP caused an overall decrease in the pool of CFTR and CFTRF508 and none of the B form was detected in the Triton X-100–insoluble fraction. When UbcH5a C85A was overexpressed, severalfold more of the B and C form of CFTR accumulated in the Triton X-100–soluble fraction. To our surprise, we also observed UbcH5a C85A overexpression to drive a severalfold increase in the quantity of the B form of CFTRF508 that accumulated in a Triton X-100–soluble state. Under these experimental conditions, we also observed a small quantity of the C form of CFTR in the detergent-insoluble fraction. Because the C form of CFTR is typically soluble in Triton X-100, this material appears to represent a minor contamination of the detergent-insoluble fraction with detergent soluble material.

    Figure 4. Triton X-100–soluble CFTR and CFTRF508 degradation intermediates accumulate in response to overexpression of UbcH5a C85A. (A and B) Analysis of the solubility of CFTR or CFTRF508. HEK293 were transiently tranfected with vectors that express the indicated proteins. As indicated, the proteasome inhibitor ALLN (200 μM) was added to the growth medium 4 h preceding cell lysis. (C) Coexpression of CHIP and UbcH5a C85A blocks CFTR folding and causes its biogenic intermediates to aggregate. CFTR and CFTRF508 were detected by Western blot. The fractionation of cell extracts into Triton X-100–soluble and –insoluble material was performed as described in the Materials and methods. The upper panel represents Triton X-100–soluble material, where as the lower part represents Triton X-100–insoluble material. The immaturely glycosylated B form of CFTR and CFTRF508, and maturely glycosylated C form of CFTR are denoted.

    Paradoxically, we observed markedly different results when the action of the Hsc70–CHIP E3 was blocked via overexpression of CHIP P269A. CHIP P269A inhibited degradation of the B form of CFTR and CFTRF508, but the degradation intermediate that accumulated was insoluble in Triton X-100 (Fig. 4, A and B). In addition, CHIP P269A blocked the glycolytic maturation of CFTR from the B to C form. The effect that CHIP and CHIP P269A had on CFTR and CFTRF508 biogenesis was resultant from interactions with Hsc70 because the TPR mutant CHIP K30A had no effect on CFTR biogenesis (Fig. 4).

    How do we explain the observation that the overexpression of UbcH5a C85A and CHIP P269A block CFTR degradation, but yet cause the degradation intermediates that accumulate to exhibit differential detergent solubility? CHIP P269A blocks the glycolytic maturation of CFTR, and thus retains the ability of CHIP to interact with Hsc70 to arrest CFTR folding (Meacham et al., 2001). Therefore, because CHIP P269A can hinder Hsc70s protein folding function, but can't promote CFTR degradation, it causes nonnative CFTR to aggregate. In HEK293 cells CHIP levels are normally 10-fold lower than those for Hsc70 (Meacham et al., 2001). Hence, when UbcH5a C85A is overexpressed the levels of CHIP–UbcH5a C85A complexes that form should not be more than one-tenth the level of Hsc70. Ergo, the overexpression of UbcH5a C85A can block CFTR degradation, but the levels of CHIP–UbcH5a C85A complexes are not high enough to interfere with Hsc70s ability to suppress CFTR aggregation. Therefore, UbcH5a C85A drives the accumulation of a CFTRF508 degradation intermediate that is stabilized in a nonaggregated state.

    If the aforementioned interpretations are correct and UbcH5a C85A specifically inactivates the U box of CHIP to block CFTR degradation, then the coexpression of UbcH5a C85A and CHIP, should have the same effect on CFTR biogenesis as CHIP P269A. Indeed, we observed that the simultaneous overexpression of CHIP and UbcH5a C85A blocked the glycolytic maturation and degradation of CFTR and drove the B form of CFTR to accumulate as a detergent-insoluble aggregate (Fig. 4 D, lane 1 vs 5).

    The data presented in Fig. 4 are important for the following reasons. First, these data indicate that interference with Hsc70–CHIP E3 activity drives the accumulation of a novel nonaggregated CFTRF508 biogenic intermediate. Second, these data demonstrate that proper chaperone function of Hsc70 is critically important for maintenance of nonnative CFTR and CFTRF508 in a detergent soluble state. Third, the observation that UbcH5a C85A and CHIP coexpression makes CHIP behave like CHIP P269A supports the interpretation that CHIP and UbcH5a functionally interact in vivo to ubiquitinate CFTR.

    Characterization of the nonaggregated CFTRF508 degradation intermediate

    To investigate the nature of the detergent-soluble CFTRF508 degradation intermediate that accumulated when UbcH5a C85A was overexpressed, we compared its ubiquitination state to that of degradation intermediates that accumulate in response to proteasome inhibition by ALLN, or overexpression of a dominant negative form of p97 (p97 QQ; Ye et al., 2003). P97 is a cytosolic chaperone that extracts polyubiquitinated proteins from the ER and participates in CFTRF508 degradation (Ye et al., 2003; Dalal et al., 2004). Nonubiquitinated CFTRF508 migrates on SDS-PAGE gels with the same mobility as its B form, where as polyubiquitinated CFTRF508, which accumulated when its degradation was blocked by ALLN or p97 QQ, migrates on SDS-PAGE gels as a high molecular weight smear (Fig. 5 A). CFTRF508 that accumulated in response to UbcH5a C85A overexpression did not migrate as a high molecular smear and, therefore, represents a nonubiquitinated species. This result is consistent with the notion that UbcH5a C85A inhibits CFTRF508 ubiquitination and thereby blocks its degradation.

    Figure 5. Characterization of the CFTRF508 biogenic intermediate that accumulates when UbcH5a C85A is overexpressed. (A) Analysis of CFTR508 levels when its proteasomal degradation is inhibited by different methods. HEK293 cells were transfected with the indicated expression vectors and harvested in SDS sample buffer 24 h later. ALLN (200 μM) was added to culture media 4 h before cell harvest. CFTRF508 levels were determined by Western blot. Ubiquitinated CFTRF508 runs as a high molecular smear on SDS-PAGE gels and is denoted. (B) Complex formation between Hsc70 and CFTRF508. Cells were transfected with the indicated expression plasmids and 24 h after incubation they were radiolabeled for 30 min with 35S-translabel and harvested. Half of each cell pellet was lysed under denaturing or native buffer conditions and immunoprecipitations were performed and analyzed by fluorography. (C) CFTRF508 that accumulates in the presence of UbcH5a C85A is glycosylated. Cells were harvested and half of each respective lysate was treated with endoglycosidase H whereas the other half served as the control. A and B denote the mobility of nonglycosylated and immaturely glycosylated forms of CFTRF508, respectively.

    Next, we examined the effect that modulating Hsc70–CHIP E3 action had on complex formation between Hsc70 and CFTRF508 degradation intermediates (Fig. 5 B). This was accomplished by coimmunoprecipitating Hsc70–CFTRF508 complexes with -Hsc70 antibody from radiolabeled cells that were transfected with the indicated form of UbcH5a or CHIP. Overexpression of UbcH5a or CHIP reduced the levels of immunoprecipitable CFTRF508, whereas UbcH5a C85A overexpression caused CFTRF508 to accumulate several fold. On the other hand, CHIP P269A, which inhibits CFTR degradation and caused CFTRF508 to aggregate (Fig. 4), reduced the total amount of CFTR508 that could be immunoprecipitated from cell extracts. The newly synthesized pool of Hsc70 detected by immunoprecipitation was not significantly changed when the levels of the components of the Hsc70–CHIP–UbcH5a E3 complex were altered. Yet, we did observe that the levels of Hsc70–CFTRF508 complexes were lower when UbcH5a and CHIP were overexpressed, and were elevated when UbcH5a C85A was overexpressed. Nonetheless, under all of the aforementioned experimental conditions tested, the changes in the levels of Hsc70–CFTRF508 complexes appeared proportional to changes in the total amount of immunoprecipitable CFTRF508 present in the cell extracts. The major conclusion drawn from these results is that the detergent soluble CFTRF508 degradation intermediate that accumulates upon UbcH5a C85A overexpression is associated with Hsc70.

    Data presented thus far suggest that inhibition of Hsc70–CHIP E3 activity drives the accumulation of an ER membrane inserted CFTRF508 biogenic intermediate that is arrested at a biogenic stage where it has the potential to either fold or be degraded. If this is the case then the CFTRF508 that accumulates in response to UbcH5a C85A overexpression should be glycosylated. Indeed, we observed that the gel mobility of the CFTRF508 that accumulates in response to UbcH5a C85A overexpression was increased when total cell extracts were treated with endoglycosidase H, which removes ASN-linked glycans from glycoproteins (Fig. 5 C). Thus, inhibition of the Hsc70–CHIP E3 activity promotes the accumulation of an immaturely glycosylated and detergent soluble form of CFTRF508 that is bound to Hsc70.

    CFTRF508 that accumulates in the ER when Hsc70–CHIP action is blocked can fold to the native state

    To ascertain whether or not the CFTRF508 biogenic intermediate that accumulates in response to inhibition of Hsc70–CHIP E3 function is capable of folding we determined if it could be chased to its maturely glycosylated C form (Fig. 6 A). Transiently transfected HEK293 cells were grown for 24 h after transfection and then treated with cycloheximide to inhibit new protein synthesis. The fate of the accumulated CFTRF508 was then monitored by Western blot after the indicated chase incubation at 37° or 26°C (Fig. 6 A). The low-temperature chase incubation was incorporated into the design of this experiment because nascent CFTRF508 exhibits a temperature-sensitive folding defect and can fold to the native state and accumulate in its maturely glycosylated C form when cells are cultured at 26°C (Denning et al., 1992).

    Figure 6. Glycolytic maturation of CFTRF508 degradation intermediates that accumulate in response to UbcH5a C85A overexpression. (A) The CFTRF508 that accumulates when UbcH5a C85A is overexpressed becomes maturely glycosylated when cell growth temperatures are reduced to 26°C. HEK293 cells were transiently transfected with the indicated plasmids and were treated with cycloheximide (25 μg/ml) 24 h later. Cells were then either shifted to 26°C or maintained at 37°C for the indicated time.(B) Glycolytic processing of CFTRF508 in cells incubated at 26°C for 16 h is blocked by BFA and enhanced by the chemical chaperone TMAO (75 mM). (C) COS-7 cells are capable of maintaining CFTRF508 degradation intermediates in a folding competent state. This experiment was performed as described for panel A except that the chase time was for 24 h at 26°C. The immaturely glycosylated and maturely glycosylated forms of CFTRF508 are denoted as B and C, respectively.

    When control cells were allowed to synthesize CFTRF508 at 37°C and were then incubated at either 37° or 26°C, >90% of the total protein present at t = 0 was degraded during the 8-h chase period. When UbcH5a C85A was coexpressed with CFTRF508 and a chase incubation was performed at 37°C, 40 to 50% of the total CFTRF508 present at t = 0 remained in the cell for up to 24 h. However, even though UbcH5a C85A stabilized the B form of CFTRF508, its conversion to the C form was not detected during the chase reaction at 37°C. In contrast, when chase incubations were performed at 26°C, a significant portion of the CFTRF508 that was stabilized in the B form by UbcH5a C85A, was converted to the maturely glycosylated C form. The formation of the maturely glycosylated C form of CFTRF508 was proportional to the loss of B form and could be detected in cells after 8 h of chase time and appeared to be complete after 16 h. The glycolytic maturation of CFTRF508 observed in the presence of UbcH5a C85A during the chase incubation at 26°C was inhibited by brefeldin A (BFA) and appears to result from trafficking of CFTRF508 out of the ER (Fig. 6 B). To determine if we could further increase the folding efficiency of the CFTRF508 that accumulated in the presence of UbcH5a C85A, cells were treated with the chemical chaperone TMAO just before the initiation of the chase reaction (Brown et al., 1996). TMAO treatment of cells increased the quantity of UbcH5a C85A stabilized CFTRF508 that could be processed to a maturely glycosylated C form around twofold. Thus, a portion of CFTRF508 that accumulated in response to inhibition of its ubiquitination remains in a foldable state that can be brought back on pathway by alteration of cell growth temperatures or chemical chaperones.

    To probe whether inhibition of the Hsc70–CHIP complex stabilizes CFTRF508 in a folding competent conformation in more than one cell type, we examined the effect that UbcH5a C85A overexpression had on CFTRF508 expression and folding in COS7 cells (Fig. 6 C). UbcH5a C85A overexpression was again observed to drive the accumulation of the B form of CFTRF508. In addition, a significant portion of the B form that accumulated in the presence of Ubc5Ha C85A could be chased at 26°C, in a BFA-sensitive manner, to the maturely glycosylated C form. Hence, the Hsc70–CHIP complex can regulate biogenesis of CFTRF508 in more than one cell type.

    To demonstrate that the CFTRF508 that was converted to the C form during the 26°C chase incubation was trafficked to the cell surface the localization of GFP–CFTRF508 was examined under these experimental conditions and compared with that of GFP–CFTR (Fig. 7). At 37°C, GFP–CFTR was detected both at the cell surface and in a perinuclear location that corresponds to the ER (Moyer et al., 1998). At 37°C, in the presence or absence of UbcH5a C85A, GFP–CFTRF508 was only detected in its soluble ER form. However, when UbcH5a C85A-transfected cells were cultured for 24 h at 37°C, treated with cycloheximide, and then incubated at 26°C for 4 h, a pool of GFP–CFTRF508 accumulated, in a BFA-sensitive fashion, at the cell surface.

    Figure 7. Localization of GFP–CFTRF508 in HEK293 cells. Cells grown on glass coverslips were transiently transfected with GFP–CFTR or GFP–CFTRF508 with or without UbcH5a C85A and cultured for 24 h at 37°C. Where indicated 25 μg/ml cyclohexamide and/or brefeldin A (10 μM) was added to culture media. After 4 h of culture at 26°C these cells were fixed and the coverslips were mounted on glass slides. Images were collected and processed as described in the Materials and methods section.

    The collective data presented in Figs. 5–7 demonstrate that when the activity of the Hsc70–CHIP ubiquitin ligase is reduced, CFTRF508 accumulates as an immaturely glycosylated species that is not a dead-end folding intermediate. Instead, the cell can maintain a pool of kinetically trapped CFTRF508 folding intermediates in a detergent soluble and foldable state.

    Discussion

    Herein we provide new insights into how CFTR and CFTRF508 biogenic intermediates are partitioned between folding and degradation pathways. The data presented suggest a model for quality control in which newly synthesized CFTR and CFTRF508 initiate folding, but intermediates of each accumulate in a kinetically trapped conformation that is maintained in a soluble state by Hsc70. CHIP then interacts with Hsc70 and functions via a two-step mechanism to attract UbcH5a into Hsc70–CFTR complexes. Then the Hsc70–CHIP–UbcH5a E3 formed acts to polyubiquitinate CFTR.

    Interestingly, inhibition of Hsc70–CHIP E3 activity caused the accumulation of a nonaggregated and ER localized CFTRF508 biogenic intermediate that was bound by Hsc70. Addition of chemical chaperones to growth media and reduction of cell growth temperatures permitted this nonubiquitinated CFTRF508 degradation intermediate to fold, exit the ER, and accumulate on the cell surface. These are the first data that describe a nonubiquitinated CFTRF508 biogenic intermediate and they demonstrate that it can be maintained in a nonaggregated and foldable state. This new information suggests that the development of drug cocktails that contain ubiquitination blockers and chemical chaperones could increase the cell surface expression of CFTRF508 and provide a treatment for CF.

    The nature of the folding defect that arrests the progression of CFTRF508 through its folding cascade and what causes it to be selected for proteasomal degradation is not entirely clear. One school of thought is that CFTRF508 is highly prone to misfolding and aggregation and is therefore selected for ERAD. Such a notion is supported by the observation that inhibition of the proteasome blocks CFTRF508 degradation and drives the accumulation of ubiquitinated forms of CFTRF508 in Triton X-100–insoluble aggregates (Ward and Kopito, 1998). However, because the inactivation of the Hsc70–CHIP E3 ligase leads to the accumulation of a soluble ER localized CFTRF508 biogenic intermediate, the data we present support a different view. It appears that inhibition of the proteasome leads polyubiquitinated CFTRF508 to aggregate because it can be extracted from the ER membrane by the p97–UFD1–NPL4 complex (Ye et al., 2003), and because it cannot be degraded, polyubiquitinated CFTRF508 accumulates in aggresomes (Ward and Kopito, 1998). On the other hand, the nonubiquitinated CFTRF508 that accumulates in response to inhibition of the Hsc70–CHIP E3 does not aggregate because it is inserted into the ER membrane and is bound by cytosolic Hsc70. Thus, while CFTRF508 has a folding defect that prevents it from passing quality control and escaping the ER, it does not appear to be overly aggregation prone and cellular chaperones can maintain it in a foldable state.

    Because the cellular activity of CHIP and UbcH5a influence the partitioning of CFTR biogenic intermediates between folding and degradation pathways, we were interested in investigating whether inhibition of the Hsc70–CHIP E3 would influence the processing efficiency of CFTR and CFTRF508. In pulse-chase experiments UbcH5a C85A overexpression increased the half-life of the B form of CFTR and CFTRF508 from two- to threefold. Therefore, UbcH5a C85A overexpression increased the steady-state levels of CFTR and CFTRF508 severalfold. However, the processing efficiency of CFTR from its B form to its C form remained at around 25% whether or not the Hscp70–CHIP E3 complex was active. Thus, while the elevation of cellular Hsc70–CHIP E3 activity can divert the B form of CFTR away from its folding pathway, the ability of full-length CFTR to stay on pathway and collapse to the native state appears to be limited by its intrinsic folding pathway and/or additional quality control factors.

    We conclude that the E2 UbcH5a is a cytosolic factor that functions with Hsc70 and CHIP to mediate CFTR ubiquitination. This conclusion is supported by three lines of experimental evidence. First, purified CHIP and UbcH5a cooperated to facilitate the polyubiquitination of CFTR. Second, when CHIP and UbcH5a were coexpressed together they appeared to act synergistically to reduce the steady-state levels of CFTRF508. Third, the coexpression of UbcH5a C85A with CHIP, blocked CHIPs ability to degrade CFTR and converted it into a protein that behaved like the CHIP U box mutant P269A.

    UbcH5a is a member of a family of conserved E2 proteins that include UbcH5b and UbcH5c that are nearly 90% identical to each other (Scheffner et al., 1994; Jensen et al., 1995b). In addition to UbcH5a, purified CHIP can interact with UbcH5b and UbcH5c, and mRNAs for each of these E2 proteins is present in all tissues tested (Jiang et al., 2001; Jensen et al., 1995b). Thus, we propose that CHIP functions with an UbcH5 E2 family member to ubiquitinate CFTR and other Hsc70 substrates, but we are not able to state whether it prefers one family member to the other. At this point, it is interesting to note that UbcH5 proteins are related to the yeast Ubc4/5 proteins that function to target misfolded proteins for degradation and protect cells from protein denaturing physiological stress (Seufert and Jentsch, 1990). In fact, one member of the Ubc4/5 family, Ubc1, has been shown to function on the ER surface to ubiquitinate ERAD substrates (Bays et al., 2001). Thus, it is logical that UbcH5 is a component of an E3 complex, which contains molecular chaperones, that serves to prevent the accumulation of toxic protein aggregates.

    A potential caveat to the interpretation that Hsc70 and CHIP interact with a UbcH5 family member to select CFTR for degradation is that the overexpression of UbcH5a C85A may nonspecifically inhibit the action of other cytosolic quality control factors that function on the ER surface to mediate ERAD. Though possible, data from the control studies with the ERAD substrates TCR and ApoB48, whose degradation relies on cytsolic E2s, demonstrate that their degradation was not delayed by overexpression of UbcH5a C85A. Thus, it appears that the reduced rates of CFTR degradation caused by UbcH5a C85A overexpression are due to specific inactivation of the Hsc70–CHIP–UbcH5 E3 complex.

    The E2s Ubc6 and Ubc7 function with E3s such as gp78 and Doa10 on the cytoplasmic face of the ER to ubiquitinate a variety of substrates (Cyr et al., 2002). Hence, it is plausible that E2–E3 complexes that contain Ubc6 and/or Ubc7 function to select CFTR and CFTRF508 for degradation. Sommer and colleagues have explored this concept and found that overexpression of Ubc6, but not Ubc7, modulates the rate of CFTRF508 degradation (Lenk et al., 2002). When we compared the effect that dominant negative forms of Ubc6, Ubc7, and UbcH5a had on CFTR and CFTRF508 expression, UbcH5a C85A and Ubc6 C91S drove the accumulation of the B form of CFTR, whereas Ubc7 had no apparent effect. The influence that UbcH5a C85A had on the accumulation of the B form of CFTR and CFTRF508, was markedly more dramatic than that of Ubc6 C91S, yet Ubc6 clearly plays a role in CFTR quality control. Studies in yeast demonstrate that Ubc6 cooperates with the transmembrane E3 Doa10 to degrade membrane and cytosolic proteins (Swanson et al., 2001) and Doa10 is required for efficient CFTR turnover in yeast (Gnann et al., 2004). Thus, the Doa10–Ubc6 E3 may function alongside the Hsc70–CHIP–UbcH5 E3 to mediate quality control of CFTR. The Hsc70–CHIP–UbcH5 E3 recognizes cytosolic regions of CFTR, whereas the Doa10/Ubc6 E3 may recognize unassembled transmembrane regions. This scenario would explain why turnover of the B form of CFTR and CFTRF508 is delayed, but not completely blocked, by the inactivation of the Hsc70–CHIP E3 complex. A critical question pertaining to the function of CHIP as a quality control factor is related to the mechanism by which it regulates Hsc70 polypeptide binding and protein folding activity. The data presented suggest that CHIP functions via a two-step mechanism to determine the fate of Hsc70 clients such as CFTR. The first step involves the binding of CHIP to the COOH-terminal EEVD motif in the lid domain of Hsc70 (Ballinger et al., 1999; Scheufler et al., 2000). This event alters the Hsc70 polypeptide binding and release cycle to arrest CFTR folding and may involve the transient stabilization of Hsc70–CFTR complexes. This putative event would give the U box on CHIP the time required to attract UbcH5 to Hsc70–CFTR complexes and facilitate CFTR ubiquitination.

    Interestingly, the ability of CHIP to ubiquitinate Hsc70 clients can be modified by other cochaperones. Data presented herein demonstrate that Hdj-2 cooperates with Hsc70 and CHIP to mediate CFTR ubiquitination. However, the cochaperone HspBP1, which is a member of a family of nucleotide exchange factors that promote substrate release from Hsc70, blocks the ability of CHIP to ubiquitinate CFTR (Alberti et al., 2004). Thus, the fate of proteins that are bound to Hsc70 is regulated by its interactions with multiple cochaperones. To understand this process, the temporal relationship and driving force for interactions between Hsc70 and its folding or degradatory cochaperones needs to be determined.

    Materials and methods

    Plasmids and antibodies

    The plasmids used for cell transfection were: pCDNA3.1CFTR and pCDNA3.1CFTRF508 (Meacham et al., 2001); pEGFPC2-CFTR and pEGFPC2-CFTRF508 (Moyer et al., 1998); pCDM8 2B4 TCR (Bonifacino et al., 1989); pCDNA-ApoB48 (H. Ginsberg, Columbia Presbyterian Medical Center, New York, NY); pCAGGS His6UBCH5A C85A and pCAGGS His6UBCH5A (Jiang et al., 2001); pCMVPLDmyc-UBC6 and pCMVPLDmyc-UBC6 C91S (Lenk et al., 2002); pCDNA myc-UBC7 and pCDNA myc-UBC7 C89S (Tiwari and Weissman, 2001); pCDNA3.1CHIP, pCDNA3.1CHIP K30A, and pCDNA3.1CHIP P269A (Jiang et al., 2001; Meacham et al., 2001). pGEXCFTR 371–855 was termed pGSTNBD1-R (Naren et al., 1999).

    The following plasmids were used for overexpression of the indicated proteins in E. coli; pET9d Hdj2 and pET11a Hsc70 (Meacham et al., 1999). Plasmids prepared for this study were pET11a His6UbcH5a, pET11a-His6UbcH5a C85A, pET11d His6Ubc7, pET11a His6Ubc6 1–243, pET30His6CHIP, pET30His6CHIPK30A, pET30His6CHIPH260A, and pET30 His6CHIPP269A. E1 was purchased from Calbiochem.

    The antibodies used for Western blots and/or immunoprecipitations were CFTR clone MM13-4 from Upstate Biotechnology and CFTR R-domain antibody from R&D Systems. TCR was from BD Biosciences and Hsc70 was from Stressgen Biotechnologies.

    Protein purification

    The proteins used in in vitro ubiquitination assays were purified after overexpression in E. coli. Hsc70 was purified by a combination of ATP–agarose and anion exchange chromatography (Cyr et al., 1992). Hdj-2 was purified by anion exchange and hydroxyapatite chromatography (Meacham et al., 1999). His6-tagged proteins were purified by metal chelate chromatography (Lu and Cyr, 1998).

    GstNBD1-R was expressed in E. coli strain BL21 (DE3) and cells from a 600-ml culture were harvested after a 16-h induction at 30°C with 0.2 mM IPTG. Cell pellets were resuspended and incubated for 30 min on ice in 10 mM Tris-HCl, pH 8.0, 1 mM EDTA, 150 mM NaCl, 1 mM phenylmethyl sulfonyl fluoride, 1 mM DTT, and lysozyme 0.1 μg/ml. The extract was supplemented with 1% sarkosyl and sonicated. Gst–NBD1–R was then purified with glutathione–agarose beads and had a final concentration near 2 mg/ml.

    Reconstitution of CFTR ubiquitination

    The experimental conditions for the reconstitution of gst–NBD1–R ubiquitination were described previously (Koegl et al., 1999). Ubiquitination assays were performed in a reaction buffer composed of 20 mM Hepes, pH 7.4, 50 mM NaCl, 5 mM MgCl2, 2.5 mM ATP, 2 mM DTT, 10 μM bovine ubiquitin, 0.1 μM rabbit E1 and 1 μM gst–NBD1–R. The indicated E2 protein was added at 4 μM and the other factors were included at the concentration indicated in Fig. 1. Incubations were performed at 37°C for the indicated time and terminated by the addition of 20 μl of SDS sample buffer to 25 μl reaction cocktails. Proteins were resolved on 7% SDS-PAGE gels and then transferred to nitrocellulose membranes that were decorated with R domain antibody and developed.

    Assays for CFTR biogenesis

    HEK293 and Cos-7 cells were maintained in DMEM supplemented with 10% fetal bovine serum and a mixture of 1% penicillin and streptomycin at 37°C and transfected with CFTR and CFTRF508 expression plasmids. Steady-state levels of CFTR and CFTRF508 levels were determined by Western blot and CFTR processing efficiency was measured by pulse chase analysis (Meacham et al., 2001). Details of the protocols for direct immunoprecipitations or coimmunoprecipitations were as described previously (Meacham et al., 2001). To verify the identity of CFTR isolated from chaperone complexes CFTR was reimmunoprecipitated from coimmunoprecipitates with -CFTR (Meacham et al., 1999). Treatment of cell extracts with endoglycosidase H was conducted as described previously (Meacham et al., 1999).

    Fluorescence microscopy

    HEK293 cells were cultured on glass coverslips and transfected transiently with 1μg pEGFPC2–CFTR or pEGFPC2–CFTRF508 either alone or in combination with 3 μg pCAGGS His6UBCH5A C85A. 24 h later, cells were washed twice for 5 min with 2 mls of PBS and fixed with 4% paraformaldehyde at room temperature. As indicated, cells were grown for 24 h after transfection and then treated with 25 μg/ml cyclohexamide and incubated for an additional 4 h at 26°C. Coverslips were mounted on glass slides with the preservative Fluoromount-G. Images were collected using a Nikon E600 microscope and a Princeton Instruments CCD camera. Images were processed with Metamorph (Universal Imaging Corp.) and Adobe Photoshop software.

    Online supplemental material

    Data presented in Fig. S1 demonstrate that the joint presence of Hsc70, Hdj-2, and CHIP stimulates dramatically the rate at which UbcH5a conjugates gst–NBD1–R with ubiquitin. When Hsc70 or Hdj-2 were omitted from reactions mono-, di-, and triubiquitination of gst–NBD1–R was observed, but this reaction was slow and inefficient. Thus, Hsc70, Hdj-2, and CHIP act jointly to stimulate the ubiquitination activity of UbcH5a and therefore exhibit an activity that fits the definition of a multisubunit E3 ubiquitin ligase. Fig. S1 is available at http://www.jcb.org/cgi/content/full/jcb.200410065/DC1.

    Acknowledgments

    The authors thank Drs. Bonificano, Ginsberg, Kirk, Sommer, Stanton, and Weissman for providing plasmids.

    C. Patterson is funded by the National Institutes of Health (NIH). D.M. Cyr is funded by the NIH and the Cystic Fibrosis Foundation. A predoctoral fellowship from the American Heart Association supports J.M. Younger.

    References

    Alberti, S., K. Bohse, V. Arndt, A. Schmitz, and J. Hohfeld. 2004. The cochaperone HspBP1 inhibits the CHIP ubiquitin ligase and stimulates the maturation of the cystic fibrosis transmembrane conductance regulator. Mol. Biol. Cell. 15: 4003–4010.

    Ballinger, C.A., P. Connell, Y. Wu, Z. Hu, L.J. Thompson, L.Y. Yin, and C. Patterson. 1999. Identification of CHIP, a novel tetratricopeptide repeat-containing protein that interacts with heat shock proteins and negatively regulates chaperone functions. Mol. Cell. Biol. 19:4535–4545.

    Bays, N.W., R.G. Gardner, L.P. Seelig, C.A. Joazeiro, and R.Y. Hampton. 2001. Hrd1p/Der3p is a membrane-anchored ubiquitin ligase required for ER-associated degradation. Nat. Cell Biol. 3:24–29.

    Bonifacino, J.S., C.K. Suzuki, J. Lippincott-Schwartz, A.M. Weissman, and R.D. Klausner. 1989. Pre-Golgi degradation of newly synthesized T-cell antigen receptor chains: intrinsic sensitivity and the role of subunit assembly. J. Cell Biol. 109:73–83.

    Brown, C.R., L.Q. Hong-Brown, J. Biwersi, A.S. Verkman, and W.J. Welch. 1996. Chemical chaperones correct the mutant phenotype of the delta F508 cystic fibrosis transmembrane conductance regulator protein. Cell Stress Chaperones. 1:117–125.

    Cyr, D.M., X. Lu, and M.G. Douglas. 1992. Regulation of Hsp70 function by a eukaryotic DnaJ homolog. J. Biol. Chem. 267:20927–20931.

    Cyr, D.M., J. Hohfeld, and C. Patterson. 2002. Protein quality control: U-box-containing E3 ubiquitin ligases join the fold. Trends Biochem. Sci. 27:368–375.

    Dai, Q., C. Zhang, Y. Wu, H. McDonough, R.A. Whaley, V. Godfrey, H.H. Li, N. Madamanchi, W. Xu, L. Neckers, et al. 2003. CHIP activates HSF1 and confers protection against apoptosis and cellular stress. EMBO J. 22:5446–5458.

    Dalal, S., M.F. Rosser, D.M. Cyr, and P.I. Hanson. 2004. Distinct roles for the AAA ATPases NSF and p97 in the secretory pathway. Mol. Biol. Cell. 15:637–648.

    Denning, G.M., M.P. Anderson, J.F. Amara, J. Marshall, A.E. Smith, and M.J. Welsh. 1992. Processing of mutant cystic fibrosis transmembrane conductance regulator is temperature-sensitive. Nature. 358:761–764.

    Gnann, A., J.R. Riordan, and D.H. Wolf. 2004. Cystic fibrosis transmembrane conductance regulator degradation depends on the lectins Htm1p/EDEM and the Cdc48 protein complex in yeast. Mol. Biol. Cell. 15:4125–4135.

    Gusarova, V., A.J. Caplan, J.L. Brodsky, and E.A. Fisher. 2001. Apoprotein B degradation is promoted by the molecular chaperones hsp90 and hsp70. J. Biol. Chem. 276:24891–24900.

    Jensen, T.J., M.A. Loo, S. Pind, D.B. Williams, A.L. Goldberg, and J.R. Riordan. 1995a. Multiple proteolytic systems, including the proteasome, contribute to CFTR processing. Cell. 83:129–135.

    Jensen, J.P., P.W. Bates, M. Yang, R.D. Vierstra, and A.M. Weissman. 1995b. Identification of a family of closely related human ubiquitin conjugating enzymes. J. Biol. Chem. 270:30408–30414.

    Jiang, J., C.A. Ballinger, Y. Wu, Q. Dai, D.M. Cyr, J. Hohfeld, and C. Patterson. 2001. CHIP is a U-box-dependent E3 ubiquitin ligase: identification of Hsc70 as a target for ubiquitylation. J. Biol. Chem. 276:42938–42944.

    Koegl, M., T. Hoppe, S. Schlenker, H.D. Ulrich, T.U. Mayer, and S. Jentsch. 1999. A novel ubiquitination factor, E4, is involved in multiubiquitin chain assembly. Cell. 96:635–644.

    Lenk, U., H. Yu, J. Walter, M.S. Gelman, E. Hartmann, R.R. Kopito, and T. Sommer. 2002. A role for mammalian Ubc6 homologues in ER-associated protein degradation. J. Cell Sci. 115:3007–3014.

    Lewis, H.A., S.G. Buchanan, S.K. Burley, K. Conners, M. Dickey, M. Dorwart, R. Fowler, X. Gao, W.B. Guggino, W.A. Hendrickson, et al. 2004. Structure of nucleotide-binding domain 1 of the cystic fibrosis transmembrane conductance regulator. EMBO J. 23:282–293.

    Liang, J.-S., T. Kim, S. Fang, J. Yamaguchi, A.M. Weissman, E.A. Fisher, and H.N. Ginsberg. 2003. Overexpression of the tumor autocrine motility factor receptor Gp78, a ubiquitin protein ligase, results in increased ubiquitinylation and decreased secretion of apolipoprotein B100 in HepG2 cells. J. Biol. Chem. 278:23984–23988.

    Loo, M.A., T.J. Jensen, L. Cui, Y. Hou, X.B. Chang, and J.R. Riordan. 1998. Perturbation of Hsp90 interaction with nascent CFTR prevents its maturation and accelerates its degradation by the proteasome. EMBO J. 17:6879–6887.

    Lu, Z., and D.M. Cyr. 1998. Protein folding activity of Hsp70 is modified differentially by the hsp40 co-chaperones Sis1 and Ydj1. J Biol Chem. 273:27824–27830.

    Meacham, G.C., Z. Lu, S. King, E. Sorscher, A. Tousson, and D.M. Cyr. 1999. The Hdj-2/Hsc70 chaperone pair facilitates early steps in CFTR biogenesis. EMBO J. 18:1492–1505.

    Meacham, G.C., C. Patterson, W. Zhang, J.M. Younger, and D.M. Cyr. 2001. The Hsc70 co-chaperone CHIP targets immature CFTR for proteasomal degradation. Nat. Cell Biol. 3:100–105.

    Moyer, B.D., J. Loffing, E.M. Schwiebert, D. Loffing-Cueni, P.A. Halpin, K.H. Karlson, I.I. Ismailov, W.B. Guggino, G.M. Langford, and B.A. Stanton. 1998. Membrane trafficking of the cystic fibrosis gene product, cystic fibrosis transmembrane conductance regulator, tagged with green fluorescent protein in Madin-Darby canine kidney cells. J. Biol. Chem. 273:21759–21768.

    Naren, A.P., E. Cormet-Boyaka, J. Fu, M. Villain, J.E. Blalock, M.W. Quick, and K.L. Kirk. 1999. CFTR chloride channel regulation by an interdomain interaction. Science. 286:544–548.

    Okiyoneda, T., K. Harada, M. Takeya, K. Yamahira, I. Wada, T. Shuto, M.A. Suico, Y. Hashimoto, and H. Kai. 2004. Delta F508 CFTR pool in the endoplasmic reticulum is increased by calnexin overexpression. Mol. Biol. Cell. 15:563–574.

    Pind, S., J.R. Riordan, and D.B. Williams. 1994. Participation of the endoplasmic reticulum chaperone calnexin (p88, IP90) in the biogenesis of the cystic fibrosis transmembrane conductance regulator. J. Biol. Chem. 269:12784–12788.

    Qu, B.H., and P.J. Thomas. 1996. Alteration of the cystic fibrosis transmembrane conductance regulator folding pathway. J Biol Chem. 271:7261–7264.

    Riordan, J.R., J.M. Rommens, B. Kerem, N. Alon, R. Rozmahel, Z. Grzelczak, J. Zielenski, S. Lok, N. Plavsic, J.L. Chou, and et al. 1989. Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science. 245:1066–1073.

    Scheffner, M., J.M. Huibregtse, and P.M. Howley. 1994. Identification of a human ubiquitin-conjugating enzyme that mediates the E6-AP-dependent ubiquitination of p53. Proc. Natl. Acad. Sci. USA. 91:8797–8801.

    Scheufler, C., A. Brinker, G. Bourenkov, S. Pegoraro, L. Moroder, H. Bartunik, F.U. Hartl, and I. Moarefi. 2000. Structure of TPR domain-peptide complexes: critical elements in the assembly of the Hsp70-Hsp90 multichaperone machine. Cell. 101:199–210.

    Seufert, W., and S. Jentsch. 1990. Ubiquitin-conjugating enzymes UBC4 and UBC5 mediate selective degradation of short-lived and abnormal proteins. EMBO J. 9:543–550.

    Swanson, R., M. Locher, and M. Hochstrasser. 2001. A conserved ubiquitin ligase of the nuclear envelope/endoplasmic reticulum that functions in both ER-associated and Matalpha2 repressor degradation. Genes Dev. 15:2660–2674.

    Tiwari, S., and A.M. Weissman. 2001. Endoplasmic reticulum (ER)-associated degradation of T cell receptor subunits. Involvement of ER-associated ubiquitin-conjugating enzymes (E2s). J. Biol. Chem. 276:16193–16200.

    Ward, C.L., and R.R. Kopito. 1994. Intracellular turnover of cystic fibrosis transmembrane conductance regulator. Inefficient processing and rapid degradation of wild-type and mutant proteins. J. Biol. Chem. 269:25710–25718.

    Ward, C.L., and R.R. Kopito. 1998. Aggresomes: a cellular response to misfolded proteins. J. Cell Biol. 143:1883–1898.

    Ward, C.L., S. Omura, and R.R. Kopito. 1995. Degradation of CFTR by the ubiquitin-proteasome pathway. Cell. 83:121–127.

    Welsh, M.J., and A.E. Smith. 1993. Molecular mechanisms of CFTR chloride channel dysfunction in cystic fibrosis. Cell. 73:1251–1254.

    Xiong, X., A. Bragin, J.H. Widdicombe, J. Cohn, and W.R. Skach. 1997. Structural cues involved in endoplasmic reticulum degradation of G85E and G91R mutant cystic fibrosis transmembrane conductance regulator. J. Clin. Invest. 100:1079–1088.

    Yang, Y., S. Janich, J.A. Cohn, and J.M. Wilson. 1993. The common variant of cystic fibrosis transmembrane conductance regulator is recognized by hsp70 and degraded in a pre-Golgi nonlysosomal compartment. Proc. Natl. Acad. Sci. USA. 90:9480–9484.

    Ye, Y., H.H. Meyer, and T.A. Rapoport. 2003. Function of the p97-Ufd1-Npl4 complex in retrotranslocation from the ER to the cytosol: dual recognition of nonubiquitinated polypeptide segments and polyubiquitin chains. J. Cell Biol. 162:71–84.

    Zhang, F., N. Kartner, and G.L. Lukacs. 1998. Limited proteolysis as a probe for arrested conformational maturation of delta F508 CFTR. Nat. Struct. Biol. 5:180–183.

    Zhang, Y., G. Nijbroek, M.L. Sullivan, A.A. McCracken, S.C. Watkins, S. Michaelis, and J.L. Brodsky. 2001. Hsp70 molecular chaperone facilitates endoplasmic reticulum-associated protein degradation of cystic fibrosis transmembrane conductance regulator in yeast. Mol. Biol. Cell. 12:1303–1314.(J. Michael Younger1,2, Hong-Yu Ren1,2, L)