Anda di halaman 1dari 5

THE JOURNAL OF BIOLOGICAL CHEMISTRY

Minireview Vol. 275, No. 6, Issue of February 11, pp. 3729 3732, 2000
2000 by The American Society for Biochemistry and Molecular Biology, Inc.
Printed in U.S.A.

Cystic Fibrosis Transmembrane of CFTR in regulating the pH within intracellular organelles


(10).
Conductance Regulator
Structural Basis of Channel Function
CFTR is a member of the ATP-binding cassette (ABC) mem-
STRUCTURE AND FUNCTION OF AN EPITHELIAL
brane transporter gene superfamily that includes both eukary-
CHLORIDE CHANNEL*
otic and bacterial proteins, such as the multiple drug resistance
protein (MDR), the sulfonylurea receptor, the transporter for
Myles H. Akabas
antigen presentation, and the bacterial periplasmic permeases
From the Center for Molecular Recognition, (9, 11). Being an ion channel makes CFTR unique in this gene
Departments of Physiology & Cellular Biophysics superfamily in which most other members are ATP-driven
and Medicine, Columbia University, membrane transporters.
New York, New York 10032
CFTR contains 1480 amino acids and consists of two homol-
ogous halves (Fig. 1). Each half contains six membrane-span-
The cystic fibrosis transmembrane conductance regulator
ning segments and a nucleotide-binding domain (NBD). The
(CFTR)1 forms a Cl channel that is an essential component of
two halves of CFTR are linked by a cytoplasmic regulatory
epithelial Cl transport systems in many organs, including the domain (R-domain) that contains a number of consensus phos-
intestines, pancreas, lungs, sweat glands, and kidneys. In the phorylation sites (9). In the proposed transmembrane topology
Cl secretory intestinal epithelium, Cl enters the cells

Downloaded from http://www.jbc.org/ by guest on March 12, 2017


(Fig. 1), which is supported by experimental evidence (9, 12,
through a Na-K-2Cl cotransporter in the basolateral mem- 13), 77% of the protein is in the cytoplasm, 19% in membrane-
brane and exits through CFTR in the apical membrane; water spanning segments, and 4% in extracellular loops, which (ex-
follows osmotically (1). Absorptive epithelia use similar trans- cept for the M1M2 and M7M8 loops) are very short. The
porters and channels, but their polarized distribution between M7M8 loop contains two N-linked glycosylation sites that are
the apical and basolateral membranes is usually reversed. A used in vivo (12, 13). Covalent linkage of the two halves is not
major determinant of the transepithelial Cl transport rate is required for assembly and function. When expressed as sepa-
the level of activation of CFTR (2, 3), which depends on the rate proteins in the same cells the halves assembled into func-
extent to which it is phosphorylated. This is determined by the tional channels and could be co-immunoprecipitated. This as-
relative activities of kinases and phosphatases, the activities of sembly required interactions within the membrane-spanning
which are often hormonally regulated (1). domains (13, 14).
Defects in the gene encoding CFTR that reduce either its Cl CFTR was inferred to be a monomer because following solu-
transport capacity or its level of cell surface expression cause bilization, with either ionic or nonionic detergents, biochemi-
cystic fibrosis (CF) (4 6) as well as a form of male sterility due cally distinct forms of CFTR, expressed in the same cells, could
to congenital bilateral absence of the vas deferens (7). CF is the not be co-immunoprecipitated (15). In contrast, two studies
most common lethal genetic disease in Caucasians, with about concluded that CFTR is a homodimer in situ based on the size
30,000 CF patients in the United States. In contrast, in intes- of CFTR particles in freeze-fracture electron micrographs (16)
tinal epithelial cells overstimulation of CFTR because of the and on the functional effects of coexpressing mutants with
activation of protein kinases by bacterial enterotoxins causes distinct functional properties (17). In the homologous MDR
secretory diarrhea (1, 8). Secretory diarrhea is the second larg- transporter, however, dimers were not seen in cryoelectron
est cause of infant mortality in the developing world, causing 3 microscopic images (18).
million deaths per year of children under the age of 5. Thus, Channel-lining ResiduesThe channel lining is formed, at
although CFTR was named because of its association with CF, least in part, by residues from among the 12 membrane-span-
as a cause of disease, its relationship to secretory diarrhea is a ning segments. The major determinants of the channels func-
more widespread public health problem. tional properties are likely to be among the channel-lining
The cloning of CFTR in 1989 (9) has facilitated studies of its residues. Water-accessible residues inferred to line the channel
structure, function, regulation, biogenesis, and degradation, were identified in the M1, M3, and M6 segments using the
which will be reviewed in this article. Issues reviewed else- substituted cysteine accessibility method (19 21). These seg-
where and not discussed here include the mechanisms by ments secondary structure was inferred to be largely -helical
which mutations in CFTR cause CF (5, 6) and the possible role based on the patterns formed by the channel-lining residues
(19 21). Further support for an -helical secondary structure
of the membrane-spanning segments was provided by studies
* This minireview will be reprinted in the 2000 Minireview Compen-
dium, which will be available in December, 2000. This work was sup- of synthetic peptides, the sequences of which corresponded to
ported in part by National Institutes of Health Grant DK51794 and an those of the M1M6 segments. These peptides were largely
Established Scientist Award from the New York City Affiliate of the -helical in liposomes and in detergent micelles (22, 23).
American Heart Association. The minimum channel diameter was inferred to be 5.3
To whom correspondence should be addressed: Center for Molecular
Recognition, Depts. of Physiology & Cellular Biophysics and Medicine, based on the size of the largest permeant anion (24). Further
Columbia University, 630 W. 168th St., New York, NY 10032. Tel.: studies using patches with larger numbers of channels showed
212-305-3974; Fax: 212-305-5594; E-mail: ma14@columbia.edu. that anions as large as lactobionate (10 13 in diameter) were
1
The abbreviations used are: CFTR, cystic fibrosis transmembrane slightly permeable. Thus, at least transiently, the diameter of
conductance regulator; CF, cystic fibrosis; DIDS, 4,4-diisothiocyanos-
tilbene-2,2-disulfonic acid; MDR, multiple drug resistance protein;
the channel must be 10 13 (25). In the presence of cytoplas-
NBD, nucleotide-binding domain; PKA, cAMP-dependent protein ki- mic ATP the large anions were only permeable from the cyto-
nase; PKC, protein kinase C; ER, endoplasmic reticulum. plasmic side (25). The molecular basis for asymmetric perme-

This paper is available on line at http://www.jbc.org 3729


3730 Minireview: CFTR Channel Structure and Function

FIG. 1. Transmembrane topology of CFTR. Gray balloons on the


M7M8 loop indicate N-linked glycosylation sites. R, R-domain.

ation by large anions is unknown. There is no asymmetry in the


conduction of small anions; the channel has a linear current-
voltage relationship in Cl solutions (13). Although some stud-
ies suggested that ATP was permeable through CFTR (re-
viewed in Ref. 26), it has been convincingly demonstrated that
there is no measurable electrogenic ATP flux through CFTR
(2729). FIG. 2. Schematic illustrating the structure of CFTR and the

Downloaded from http://www.jbc.org/ by guest on March 12, 2017


Mechanism of Charge SelectivityThe ability to discrimi- relationships of its domains and associated proteins to the
nate between Cl and cations is essential for the role of CFTR channel. Membrane-spanning domains, red; the water-filled trans-
membrane channel, blue; NBD-1, yellow; NBD-2, orange; bound ATP,
in epithelial Cl transport. The extent of anion selectivity, as white ovals; R-domain, green; the N- and C termini are indicated by
measured by the Cl to Na permeability ratio (30), ranges black lines; the associated proteins syntaxin 1A (Syn 1A) and Na/H
between 10 and 300 (31, 32). We showed that the charge selec- exchanger regulatory factor/ezrin-binding protein 50 (NHERF/EBP-
tivity filter that discriminates between anions and cations is 50), purple. Note the position of Arg-352, a major determinant of charge
selectivity, at the narrow point in the channel. Cl ions probably do not
located at the cytoplasmic end of the channel and that both interact with the NBDs and R-domain as they pass through the chan-
anions and cations can enter the extracellular end of the chan- nel. The cytoplasmic vestibule has a binding site for large anions (see
nel (33). Arg-352, at the cytoplasmic end of M6, is a major channel-lining Residues). The interactions with PP2C and other chan-
determinant of charge selectivity. Removal of the positive nels and transporters are not shown (see Interactions with Other
Proteins).
charge at this position reduced the Cl to Na permeability
ratio approximately 10-fold (34). We hypothesized that the
positive charge of Arg-352 creates an electrostatic barrier to the vestibule because none of the CF-related loop mutations
cation permeation. Near Arg-352 the channel must be narrow affected ion conduction (40).
enough so that the electrostatic barrier extends across the The picture that is emerging is of a channel with a large
channel lumen (34). extracellular vestibule that extends into the plain of the mem-
Halide Selectivity and Multiple Ion OccupancyThe channel brane and is accessible to anions and cations from the extra-
displays modest selectivity between halides. The halide perme- cellular side (Fig. 2). The channel narrows toward the cytoplas-
ability sequence is I (1.8) Br (1.2) Cl (1.0) F (0.2), mic end where the charge selectivity filter is located. Finally,
indicative of weak interactions between permeating anions and there is a short cytoplasmic vestibule that contains an anion
channel binding sites (30, 32). Multiple anions can simulta- binding site. The channel lining is formed, in part, by residues
neously occupy the channel, thereby resulting in anomalous from the M1, M3, M6, and M12 segments. Little is known about
mole fraction effects in the single channel conductance in mix- whether the other eight membrane-spanning segments contrib-
tures of Cl and SCN. These effects were eliminated in the ute to the channel lining. The location of the gate that blocks
M6 mutant R347D, and Arg-347, therefore, was hypothesized ion conduction through the channel in the closed state is un-
to be at or near an anion binding site (35). Arg-347, however, known. The gate, however, is controlled by conformational
has been shown to form a salt bridge with Asp-924 (36). There- changes in the cytoplasmic domains.
fore, it is possible that rather than Arg-347 being a binding site
itself, structural perturbation caused by the loss of the salt Regulation of CFTR Channel Gating
bridge disrupted an anion binding site elsewhere (30). Two separate processes control the gating of CFTR: 1) phos-
Channel InhibitorsCFTR has few inhibitors and none are phorylation and 2) binding and hydrolysis of ATP. Phosphoryl-
of high affinity or specificity (13). The affinity of the channel ation is necessary for activation, but it is not sufficient. After
blocker diphenylamine 2-carboxylate was reduced by mutating phosphorylation, gating between the closed and open states is
two water-accessible residues in M6. Mutations of the aligned controlled by ATP hydrolysis (12, 13, 41).
M12 residues increased diphenylamine 2-carboxylate affinity Phosphorylation Is a Prerequisite for Channel Activation
suggesting that the channel is lined by residues from both The R-domain contains multiple consensus phosphorylation
halves of CFTR (37). sites for cAMP-dependent protein kinase (PKA), protein kinase
In contrast to their actions on other Cl channels and trans- C (PKC), and Type II cGMP-dependent protein kinase (9, 12,
porters, disulfonic stilbenes such as DIDS inhibit only when 13, 41, 42). The more extensive the phosphorylation, the
applied to the cytoplasmic side (38). Cytoplasmic application of greater the channel open probability (41, 43). There appears to
other large anions also caused flickery block of CFTR. This is be extensive redundancy in the regulatory PKA phosphoryla-
further evidence that the channel is asymmetric and contains tion sites. Five sites are phosphorylated in vivo, but removal of
an anion binding site in the cytoplasmic vestibule (39). The these sites or all 10 dibasic PKA sites reduced but did not
residues forming the cytoplasmic vestibule remain to be iden- eliminate PKA activation (44). CFTR is still phosphorylated
tified, but the cytoplasmic loops probably do not contribute to after removal of the 10 dibasic sites indicating that phospho-
Minireview: CFTR Channel Structure and Function 3731
rylation at non-dibasic PKA sites is sufficient for channel acti- rent hypothesis that ATP hydrolysis at NBD-1 opens the chan-
vation; one such site has been identified (12, 13, 41). PKC nel and ATP hydrolysis at NBD-2 closes the channel to termi-
phosphorylation may be a prerequisite for activation by PKA nate a burst is consistent with the functional effects of
(45). Finally, although genistein, a tyrosine kinase inhibitor, mutations in the NBDs (13, 41). Mutations in the NBD-2
activates CFTR this occurs through a direct interaction with Walker motifs that decrease its ATP hydrolysis rate slow chan-
CFTR and does not involve a tyrosine kinase (reviewed in Ref. nel closing (5558). Similar mutations in NBD-1 reduce chan-
41). nel opening. The NBD-1 mutations also altered channel closing
After phosphorylation, CFTR is deactivated by protein phos- rates, suggesting there are functional interactions between the
phatases (41). Dephosphorylation is largely mediated by pro- NBDs (5557). In both the crystal structure and in solution
tein phosphatases PP2C and PP2A (46, 47). PP2C is closely HisP forms a dimer, the interface of which is located along the
linked to CFTR in the membrane as it could be co-immunopre- edge of Arm I (54). Thus, direct contacts between the NBDs
cipitated with CFTR as well as cross-linked to CFTR by bifunc- may be the structural basis for the functional interaction. Mix-
tional cross-linkers, whereas PP1, PP2A, and PP2B could not tures of ATP and non-hydrolyzable analogs lock channels in the
(48). open state. Presumably ATP hydrolysis at NBD-1 opens the
Channel Gating Requires ATP HydrolysisFollowing phos- channels and binding of a non-hydrolyzable analog at NBD-2
phorylation, opening and closing of the channel is controlled by prevents closing (13, 41).
ATP binding and hydrolysis (12, 13, 41). Non-hydrolyzable ATP The gating behavior of CFTR has been described by several
analogs do not support channel opening (49). Purified, phos- kinetic models. Cyclic models with essentially irreversible
phorylated, reconstituted CFTR binds ATP with a Km of 0.3 mM steps corresponding to the ATP hydrolysis steps provide the
and hydrolyzes ATP at a rate of about 1 molecule/s (50), similar best descriptions of channel gating (reviewed in Refs. 13, 41,
to the rate of channel gating. ATP hydrolysis occurs in the and 59).
NBDs, which contain highly conserved Walker A and B motifs Interactions with Other Proteins

Downloaded from http://www.jbc.org/ by guest on March 12, 2017


found in other ATPases (9, 13, 41, 49). In addition to its function as a Cl channel, CFTR acts as a
A construct lacking the R-domain, CFTR-R, did not require regulator of other channels and transporters (26). CFTR medi-
phosphorylation to open and close in the presence of ATP (13). ates cAMP regulation of amiloride-sensitive Na channels (60),
Adding soluble, phosphorylated R-domain peptide to CFTR-R outward rectifying Cl channels (61, 62), Cl/HCO3 exchanger
enhanced its ATP sensitivity resulting in an increased channel (63), and the ROMK K channel (26). Whether these regulatory
open probability (51, 52). Thus, the R-domain interacts with functions result from direct interactions between CFTR and
the NBDs and regulates their ATP affinity. the channels and transporters or through indirect interactions
Structure of a Bacterial NBDIn the homologous bacterial via other proteins remains to be determined.
periplasmic permeases the NBDs and the transmembrane do- Direct interactions with CFTR have been shown for several
mains are usually separate gene products that assemble during proteins. Syntaxin 1A interacts with the cytoplasmic N-termi-
biosynthesis (11, 53). The histidine periplasmic permease con- nal domain of CFTR and inhibits channel activity (64). A family
tains two copies of its NBD protein, HisP; each copy has a of PDZ-binding domain proteins, including the Na/H ex-
single nucleotide binding site. The x-ray crystal structure of changer regulatory factor (65) and ezrin-binding protein 50
HisP was determined (54). The sequence conservation between (66), bind to the C terminus (D-(S/T)-X-L) (Fig. 2). These pro-
the bacterial and eukaryotic NBDs implies that their struc- teins presumably couple CFTR to the cytoskeleton and may be
tures are similar and that aligned residues perform similar involved in apical membrane localization. Finally, as men-
functions in HisP and CFTR. tioned above, CFTR is closely associated with protein phospha-
HisP has a novel structure (54). It contains only small re- tase 2C (48).
gions of structural similarity with RecA and the and F1-
Synthesis and Degradation
ATPase subunits. HisP is shaped like the letter L (Fig. 2). One
arm (Arm I) of the L contains the ATP binding site, including Attention has focused on the synthesis and degradation of
the Walker A and B motifs. Most interactions between the CFTR because many mutations that cause CF, such as F508,
Walker A residues and ATP are via hydrogen bonds between lead to the misfolding of CFTR in the endoplasmic reticulum
the backbone amides and the ATP phosphates. An aspartate at (ER) and subsequent degradation of CFTR (reviewed in Refs. 4
the C terminus of the Walker B motif, aligned with CFTR and 67). The synthesis of wild-type CFTR is inefficient. Only
residues Asp-572 and Asp-1370, in NBD-1 and -2, respectively, about 30% of newly synthesized CFTR develops endoglycosi-
dase H-resistant mature glycosylation suggestive of transloca-
was inferred to bind the Mg2 ion required for hydrolysis.
tion from the ER to the Golgi complex (68, 69). In contrast,
CFTR residues Gln-493 and Ser-573 in NBD-1 and Gln-1291
100% of the homologous MDR matures (70). CFTR molecules
and Glu-1371 in NBD-2 align with the catalytically important
that fail to mature are polyubiquitinated and degraded via a
residues in HisP that hydrogen bond to a water molecule that
cytoplasmic, proteasome-dependent pathway (71, 72). In the
in turn hydrogen bonds to the -phosphate. This water mole-
ER, CFTR transiently associates with several chaperones in-
cule is thought to attack ATP because its hydrogen bond lies
cluding the cytoplasmic Hsp70 and Hsp90 and the ER mem-
parallel to the P-O bond that is broken during ATP hydrol-
brane chaperone calnexin (67, 73). The molecular basis for the
ysis (54). The adenine base forms a hydrophobic interaction
inefficient processing of CFTR in the protein synthetic pathway
with a single aromatic residue, perhaps explaining the lack of
remains to be elucidated.
specificity among nucleotide triphosphates that can gate CFTR
(49). Conclusions
The other arm in the HisP structure, Arm II, contains a CFTR has an important role in epithelial Cl transport both
LSGGQ motif that forms the foundation around which Arm II as a Cl channel and as a regulator of other channels and
is built. Arm II was inferred to interact with the membrane- transporters. Studies of the channel have begun to provide a
spanning domains (54). The residue aligned with Phe-508 in picture of its structure and regulation. Understanding how
CFTR, the site of the most common CF-associated mutation, CFTR interacts with other channels and transporter will pro-
lies on an exposed surface of an -helix in Arm II. vide insights into the cellular regulation of epithelial transport
Channel Gating and the Sites of ATP HydrolysisThe cur- and the pathophysiology that results from defects in CFTR or
3732 Minireview: CFTR Channel Structure and Function
from its overstimulation by bacterial toxins. One hopes that 34. Guinamard, R., and Akabas, M. H. (1999) Biochemistry 38, 5528 5537
35. Tabcharani, J. A., Rommens, J. M., Hou, Y. X., Chang, X. B., Tsui, L. C.,
further studies will provide a basis for the design or identifi- Riordan, J. R., and Hanrahan, J. W. (1993) Nature 366, 79 82
cation of specific, high affinity CFTR inhibitors that would be 36. Cotten, J. F., and Welsh, M. J. (1999) J. Biol. Chem. 274, 5429 5435
useful for basic studies of the role of CFTR in epithelial Cl 37. McDonough, S., Davidson, N., Lester, H. A., and McCarty, N. A. (1994) Neuron
13, 623 634
transport and potentially as a therapy for secretory diarrhea, a 38. Linsdell, P., and Hanrahan, J. W. (1996) J. Physiol. (Lond.) 496, 687 693
major cause of morbidity and infant mortality in the developing 39. Linsdell, P., and Hanrahan, J. W. (1996) Am. J. Physiol. 271, C628 C634
40. Seibert, F. S., Jia, Y., Mathews, C. J., Hanrahan, J. W., Riordan, J. R., Loo,
world. T. W., and Clarke, D. M. (1997) Biochemistry 36, 11966 11974
41. Gadsby, D. C., and Nairn, A. C. (1999) Physiol. Rev. 79, S77S107
AcknowledgmentI thank Dr. Jonathan Javitch for comments on 42. Lohmann, S. M., Vaandrager, A. B., Smolenski, A., Walter, U., and De Jonge,
this manuscript. H. R. (1997) Trends Biochem. Sci. 22, 307312
43. Hwang, T. C., Horie, M., and Gadsby, D. C. (1993) J. Gen. Physiol. 101,
REFERENCES 629 650
1. Field, M., and Semrad, C. E. (1993) Annu. Rev. Physiol. 55, 631 655 44. Chang, X.-B., Tabcharani, J. A., Hou, Y.-X., Jensen, T. J., Kartner, N., Alon,
2. Gabriel, S. E., Brigman, K. N., Koller, B. H., Boucher, R. C., and Stutts, M. J. N., Hanrahan, J. W., and Riordan, J. R. (1993) J. Biol. Chem. 268,
(1994) Science 266, 107109 11304 11311
3. Cuthbert, A. W., Hickman, M. E., MacVinish, L. J., Evans, M. J., Colledge, 45. Jia, Y., Mathews, C. J., and Hanrahan, J. W. (1997) J. Biol. Chem. 272,
W. H., Ratcliff, R., Seale, P. W., and Humphrey, P. P. (1994) Br. J. Phar- 4978 4984
macol. 112, 3136 46. Berger, H. A., Travis, S. M., and Welsh, M. J. (1993) J. Biol. Chem. 268,
4. Welsh, M. J., and Smith, A. E. (1993) Cell 73, 12511254 20372047
5. Pilewski, J. M., and Frizzell, R. A. (1999) Physiol. Rev. 79, S215S255 47. Luo, J., Pato, M. D., Riordan, J. R., and Hanrahan, J. W. (1998) Am. J. Physiol.
6. Wine, J. J. (1999) J. Clin. Invest. 103, 309 312 274, C1397C1410
7. De Braekeleer, M., and Ferec, C. (1996) Mol. Hum. Reprod. 2, 669 677 48. Zhu, T., Dahan, D., Evagelidis, A., Zheng, S., Luo, J., and Hanrahan, J. W.
8. Hansen, M. B., and Skadhauge, E. (1995) Physiol. Res. 44, 6178 (1999) J. Biol. Chem. 274, 2910229107
9. Riordan, J. R., Rommens, J. M., Kerem, B. S., Alon, N., Rozmahel, R., 49. Anderson, M. P., Berger, H. A., Rich, D. P., Gregory, R. J., Smith, A. E., and
Grzelczak, Z., Zielenski, J., Lok, S., Plavsic, N., Chou, J. L., Drumm, M. T., Welsh, M. J. (1991) Cell 67, 775784
Iannuzzi, M. C., Collins, F. S., and Tsui, L. C. (1989) Science 254, 50. Li, C., Ramjeesingh, M., Wang, W., Garami, E., Hewryk, M., Lee, D.,
1066 1073 Rommens, J. M., Galley, K., and Bear, C. E. (1996) J. Biol. Chem. 271,
10. al-Awqati, Q. (1995) Curr. Opin. Cell Biol. 7, 504 508 2846328468

Downloaded from http://www.jbc.org/ by guest on March 12, 2017


11. Higgins, C. F. (1992) Annu. Rev. Cell Biol. 8, 67113 51. Ma, J., Zhao, J., Drumm, M. L., Xie, J., and Davis, P. B. (1997) J. Biol. Chem.
12. Riordan, J. R. (1993) Annu. Rev. Physiol. 55, 609 630 272, 2813328141
13. Sheppard, D. N., and Welsh, M. J. (1999) Physiol. Rev. 79, S23S45 52. Winter, M. C., and Welsh, M. J. (1997) Nature 389, 294 296
14. Ostedgaard, L. S., Rich, D. P., DeBerg, L. G., and Welsh, M. J. (1997) Bio- 53. Ames, G. F.-L., Mimura, C. S., Holbrook, S. R., and Shyamala, V. (1992) Adv.
chemistry 36, 12871294 Enzymol. Relat. Areas Mol. Biol. 65, 1 47
15. Marshall, J., Fang, S., Ostedgaard, L. S., ORiordan, C. R., Ferrara, D., Amara, 54. Hung, L. W., Wang, I. X., Nikaido, K., Liu, P. Q., Ames, G. F., and Kim, S. H.
J. F., Hoppe, H., IV, Scheule, R. K., Welsh, M. J., Smith, A. E., and Cheng, (1998) Nature 396, 703707
S. H. (1994) J. Biol. Chem. 269, 29872995 55. Carson, M. R., Travis, S. M., and Welsh, M. J. (1995) J. Biol. Chem. 270,
16. Eskandari, S., Wright, E. M., Kreman, M., Starace, D. M., and Zampighi, G. A. 17111717
(1998) Proc. Natl. Acad. Sci. U. S. A. 95, 1123511240 56. Gunderson, K. L., and Kopito, R. R. (1995) Cell 82, 231239
17. Zerhusen, B., Zhao, J., Xie, J., Davis, P. B., and Ma, J. (1999) J. Biol. Chem. 57. Wilkinson, D. J., Mansoura, M. K., Watson, P. Y., Smit, L. S., Collins, F. S., and
274, 76277630 Dawson, D. C. (1996) J. Gen. Physiol. 107, 103119
18. Rosenberg, M. F., Callaghan, R., Ford, R. C., and Higgins, C. F. (1997) J. Biol. 58. Zeltwanger, S., Wang, F., Wang, G. T., Gillis, K. D., and Hwang, T. C. (1999)
Chem. 272, 1068510694 J. Gen. Physiol. 113, 541554
19. Akabas, M. H., Kaufmann, C., Cook, T. A., and Archdeacon, P. (1994) J. Biol.
59. Csanady, L., and Gadsby, D. C. (1999) J. Gen. Physiol. 114, 49 53
Chem. 269, 1486514868
60. Stutts, M. J., Canessa, C. M., Olsen, J. C., Hamrick, M., Cohn, J. A., Rossier,
20. Cheung, M., and Akabas, M. H. (1996) Biophys. J. 70, 2688 2695
B. C., and Boucher, R. C. (1995) Science 269, 847 850
21. Akabas, M. H. (1998) Biochemistry 37, 1223312240
61. Egan, M., Flotte, T., Afione, S., Solow, R., Zeitlin, P. L., Carter, B. J., and
22. Peng, S., Liu, L. P., Emili, A. Q., and Deber, C. M. (1998) FEBS Lett. 431,
Guggino, W. B. (1992) Nature 358, 581584
29 33
62. Gabriel, S. E., Clarke, L. C., Boucher, R. C., and Stutts, M. J. (1993) Nature
23. Wigley, W. C., Vijayakumar, S., Jones, J. D., Slaughter, C., and Thomas, P. J.
(1998) Biochemistry 37, 844 853 363, 263266
24. Linsdell, P., Tabcharani, J. A., Rommens, J. M., Hou, Y. X., Chang, X. B., Tsui, 63. Lee, M. G., Choi, J. Y., Luo, X., Strickland, E., Thomas, P. J., and Muallem, S.
L. C., Riordan, J. R., and Hanrahan, J. W. (1997) J. Gen. Physiol. 110, (1999) J. Biol. Chem. 274, 14670 14677
355364 64. Naren, A. P., Nelson, D. J., Xie, W., Jovov, B., Pevsner, J., Bennett, M. K.,
25. Linsdell, P., and Hanrahan, J. W. (1998) J. Gen. Physiol. 111, 601 614 Benos, D. J., Quick, M. W., and Kirk, K. L. (1997) Nature 390, 302305
26. Schwiebert, E. M., Benos, D. J., Egan, M. E., Stutts, M. J., and Guggino, W. B. 65. Hall, R. A., Ostedgaard, L. S., Premont, R. T., Blitzer, J. T., Rahman, N.,
(1999) Physiol. Rev. 79, S145S166 Welsh, M. J., and Lefkowitz, R. J. (1998) Proc. Natl. Acad. Sci. U. S. A. 95,
27. Grygorczyk, R., Tabcharani, J. A., and Hanrahan, J. W. (1996) J. Membr. Biol. 8496 8501
151, 139 148 66. Short, D. B., Trotter, K. W., Reczek, D., Kreda, S. M., Bretscher, A., Boucher,
28. Li, C., Ramjeesingh, M., and Bear, C. E. (1996) J. Biol. Chem. 271, R. C., Stutts, M. J., and Milgram, S. L. (1998) J. Biol. Chem. 273,
1162311626 1979719801
29. Reddy, M. M., Quinton, P. M., Haws, C., Wine, J. J., Grygorczyk, R., 67. Kopito, R. R. (1999) Physiol. Rev. 79, S167S173
Tabcharani, J. A., Hanrahan, J. W., Gunderson, K. L., and Kopito, R. R. 68. Lukacs, G. L., Mohamed, A., Kartner, N., Chang, X. B., Riordan, J. R., and
(1996) Science 271, 1876 1879 Grinstein, S. (1994) EMBO J. 13, 6076 6086
30. Dawson, D. C., Smith, S. S., and Mansoura, M. K. (1999) Physiol. Rev. 79, 69. Ward, C. L., and Kopito, R. R. (1994) J. Biol. Chem. 269, 25710 25718
S47S75 70. Loo, T. W., and Clarke, D. M. (1997) J. Biol. Chem. 272, 709 712
31. Anderson, M. P., Gregory, R. J., Thompson, S., Souza, D. W., Paul, S., 71. Jensen, T. J., Loo, M. A., Pind, S., Williams, D. B., Goldberg, A. L., and
Mulligan, R. C., Smith, A. E., and Welsh, M. J. (1991) Science 253, 202205 Riordan, J. R. (1995) Cell 83, 129 135
32. Tabcharani, J. A., Linsdell, P., and Hanrahan, J. W. (1997) J. Gen. Physiol. 72. Ward, C. L., Omura, S., and Kopito, R. R. (1995) Cell 83, 121127
110, 341354 73. Loo, M. A., Jensen, T. J., Cui, L., Hou, Y., Chang, X. B., and Riordan, J. R.
33. Cheung, M., and Akabas, M. H. (1997) J. Gen. Physiol. 109, 289 300 (1998) EMBO J. 17, 6879 6887
Cystic Fibrosis Transmembrane Conductance Regulator: STRUCTURE AND
FUNCTION OF AN EPITHELIAL CHLORIDE CHANNEL
Myles H. Akabas
J. Biol. Chem. 2000, 275:3729-3732.
doi: 10.1074/jbc.275.6.3729

Access the most updated version of this article at http://www.jbc.org/content/275/6/3729

Alerts:
When this article is cited
When a correction for this article is posted

Click here to choose from all of JBC's e-mail alerts

Downloaded from http://www.jbc.org/ by guest on March 12, 2017


This article cites 73 references, 34 of which can be accessed free at
http://www.jbc.org/content/275/6/3729.full.html#ref-list-1

Anda mungkin juga menyukai