WO2000050591A1 - Enhancers of cftr chloride channel function - Google Patents

Enhancers of cftr chloride channel function Download PDF

Info

Publication number
WO2000050591A1
WO2000050591A1 PCT/US2000/004642 US0004642W WO0050591A1 WO 2000050591 A1 WO2000050591 A1 WO 2000050591A1 US 0004642 W US0004642 W US 0004642W WO 0050591 A1 WO0050591 A1 WO 0050591A1
Authority
WO
WIPO (PCT)
Prior art keywords
cftr
polypeptide
protein
channel
open probability
Prior art date
Application number
PCT/US2000/004642
Other languages
French (fr)
Inventor
Lynn M. Adams
Pamela B. Davis
Jianjie Ma
Original Assignee
Case Western Reserve University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Case Western Reserve University filed Critical Case Western Reserve University
Priority to AU32419/00A priority Critical patent/AU3241900A/en
Publication of WO2000050591A1 publication Critical patent/WO2000050591A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4712Cystic fibrosis

Definitions

  • This invention is related to the field of cystic fibrosis. More particularly, it is related to the area of therapeutic treatments and drug discovery for treating cystic fibrosis.
  • CFTR a chloride channel located in the apical membrane of epithelial cells
  • cystic fibrosis a 1480 amino acid protein that is a member of the ATP binding cassette (ABC) transporter family (Riordan et al, 1989, Higgins, 1992).
  • ABSC ATP binding cassette
  • Each half of CFTR contains a transmembrane domain and a nucleotide binding fold (NBF), and the two halves are connected by a regulatory, or R domain.
  • the R domain is unique to CFTR and contains several consensus PKA phosphorylation sites (Cheng et al, 1991, Picciotto et al., 1992).
  • Opening of the CFTR channel is controlled by PKA phosphorylation of serine residues in the R domain (Tabcharani et al., 1991, Bear et al., 1992) and ATP binding and hydrolysis at the NBFs (Anderson et al., 1991 , Gunderson and Kopito, 1995).
  • Phosphorylation adds negative charges to the R domain, and introduces global conformational changes reflected by the reduction in the ⁇ -helical content of the R domain protein (Dulhanty and Riordan, 1994).
  • electrostatic and/or allosteric changes mediated by phosphorylation are likely to be responsible for interactions between the R domain and other CFTR domains that regulate channel function (Rich et al, 1993, Gadsby and Nairn, 1994).
  • polypeptide comprises a portion of CFTR (cystic fibrosis transmembrane conductance regulator) protein of between 10 and 100 amino acids, said portion comprising 18 amino acids as shown in SEQ ID NO: 1.
  • a method for activating a CFTR protein is provided.
  • a polypeptide is applied to a CFTR protein which forms a cAMP regulated chloride channel.
  • the polypeptide consists of a portion of CFTR protein which comprises 18 amino acids as shown in SEQ ID NO: 1, whereby the open probability of the channel formed by the CFTR increases by at least 25%.
  • a method for activating a CFTR protein is provided.
  • a polypeptide is applied to a CFTR protein which forms a cAMP regulated chloride channel.
  • the polypeptide consists of a portion of CFTR protein which comprises 22 amino acids as shown in SEQ ID NO: 2, whereby the open probability of the channel formed by the CFTR increases by at least 25%.
  • the present invention thus provides the art with reagents and tools for enhancing function of channels which are defective in cystic fibrosis patients.
  • FIG. 1A Sequences of NEGl and NEG2 within the R domain. Residues where mutations have been identified in the CFTR cDNA are underlined (E822K, E826K, D836Y).
  • Figure IB NEG2 is predicted to form an amphipathic ⁇ -helix as determined by secondary structure determination (Geourjon and Deleage, 1995, Rost and Sander, 1993, Rost and Sander, 1994) and illustrated in this space filling model. Negatively charged residues are colored pink, and the positively charged lysine is colored green.
  • FIG. 2A Single channel currents of wt, ⁇ NEG1- and ⁇ NEG2-CFTR incorporated into the lipid bilayer. While activities of wt-and ⁇ NEG1-CFTR absolutely require the presence of PKA in the c.-s'-intracellular solution, the ⁇ NEG2-CFTR channel opens without PKA phosphorylation.
  • Figure 2B Diary plot of ⁇ NEG2-CFTR channel open probability versus time shows that addition of 200 units/ml of PKA, a maximally stimulating concentration, does not affect channel activity. The dashed line indicates the average open probability for each segment of the experiment. Channels were recorded at -100 mV.
  • Figure 3 The Synthetic NEG2 Peptide both Stimulates and Inhibits CFTR (Figure 3A) Diary plot (open probability versus time) of a wt-CFTR channel illustrating the effect of the NEG2 peptide on the open probability of the channel in the planar lipid bilayer. The concentration of synthetic NEG2 in the cw-intracellular solution is indicated above the plot.
  • Rate of the Channel Histograms of open and closed events of the wt-CFTR channel at -80 mV were generated without peptide (control, left panel) and with 4.4 ⁇ M NEG2 peptide in the c/s-solution (right panel).
  • P 2 probability of the intermediate closed component
  • P 3 probability of the long closed component.
  • the isolated polypeptide according to the invention consists of a portion of CFTR (cystic fibrosis transmembrane conductance regulator) protein.
  • the portion preferably contains at least 18 ammo acids as shown m SEQ ID NO: 1. However, fewer amino acid residues of the sequence may be used if they retain the channel enhancing function desc ⁇ bed herein for the 18 and 22 residue polypeptides. See also SEQ ID NO. 2. Thus the polypeptide may be from about 10 or 15 ammo acid residues up to about 30 or even 100 amino acid residues
  • An isolated polypeptide may be synthetic or made in a recombinant organism. It may be a proteolytic cleavage product of a larger primary expression product, including full-length, wild-type CFTR.
  • the polypeptide will be free of full-length CFTR.
  • the polypeptide will preferably be free of other proteins and polypeptides as well.
  • fusion to another protein such as keyhole limpet hemocyanin would be used to increase immunogenicity.
  • Another desirable fusion partner is a membrane- penetrating peptide.
  • Such peptides include VP-22 (SEQ ID NO: 3), as well as the peptides shown in SEQ ID NO: 4 and SEQ ID NO: 5.
  • Such peptides can be used to facilitate the uptake by target cells of the polypeptide.
  • polypeptides of the present invention can be used to enhance the function of wild-type or minimally active mutant CFTR proteins.
  • the polypeptide functions to decrease the closed time of the channels formed by CFTR.
  • a polypeptide can be applied to the CFTR protein in any context. It can be applied in vitro or in vivo. If in vitro it can be to CFTR in cultured cells or to planar bilayer membranes containing
  • the polypeptide can be applied directly to airway epithelial cells. Such application can be by any means known in the art, including but not limited to using a gargle or a nebulizer to deliver aerosolized polypeptide to the target cells.
  • the peptide can be delivered in an indirect mode, by delivering a gene construct to the airway epithelial cells, which when taken up by the cells causes them to express the polypeptide.
  • the delivery of the polypeptide to the CFTR preferably increases the open probability of the channel formed by the CFTR by at least 25%. More preferably it increases the open probability by at least 50%, at least 75%, at least 100%), at least 125%, at least 150%, or at least 200%.
  • a CFTR construct comprises a nucleic acid sequence encoding the amino acid sequence shown in SEQ ID NO 1.
  • a suitable promoter for expression in lung epitheha is also desirable. Many such promoters are known in the art, and any can be used as appropriate for a particular application. It is believed that the administration of the polypeptide of the present invention will be the most useful m treatment of a class of mutants which produce CFTR proteins which are properly delivered to the plasma membrane but which are only residually or minimally active. Known mutants of CFTR are listed at http://www.genet.sickkids.on.ca/cftr-cgi-bin/fulltable. One can determine that a particular CFTR mutant is fully processed and reaches the plasma membrane m a
  • Example 1 Deletion of a negatively charged region (a.a. 817-838) from the R domain of CFTR alters PKA-dependent regulation of the CFTR channel
  • CFTR contains a large intracellular regulatory (R) domain where multiple PKA phosphorylation sites are located. There are two regions withm the R domain that contain a high proportion of negatively charged amino acids, a.a. 725-733 (NEGl) and a.a. 817-838 (NEG2). It is possible that these two regions could have alloste ⁇ c or electrostatic interactions with other regions of CFTR and thus affect its chloride channel function.
  • NEGl and NEG2 two deletion mutants, NEGl -CFTR and NEG2-CFTR, were created. The CFTR mutants were transiently expressed in HEK 293 cells, and their single channel functions were studied using the bilayer reconstitution system.
  • Example 2 ⁇ NEGl- and ⁇ NEG2-CFTR are glycosylated.
  • the R domain of CFTR contains two negatively charged regions, amino acids 725-733 (NEGl) and amino acids 817-838 (NEG2), that reside in close proximity to two PKA phosphorylation sites, S737 and S813, used in vivo ( Figure 1A) (Cheng, et al. 1991). NEG2 is predicted to form an amphipathic (-helical structure with a negatively charged face ( Figure IB) (Geourjon and Deleage, 1995, Rost and Sander, 1993, Rost and Sander, 1994). Three mutations (E822K, E826K, D836Y), two of which were clearly obtained from patients with CF (E822K and D836Y).
  • the E822K CFTR channel has a low open probability relative to wt-CFTR (wild type-CFTR), but the E826K CFTR channel has single channel properties similar to wt-CFTR (Vankeerberghen et al., 1998). The presence of these disease-causing mutations suggests the potential importance of the NEG2 region.
  • these regions were deleted from CFTR using mutagenesis and subclonmg.
  • the ⁇ NEG1- and ⁇ NEG2-CFTR proteins were transiently expressed in human embryonic kidney 293 cells. Membrane vesicles containing the CFTR proteins were isolated and subjected to
  • NEG2 may represent an inhibitory region, removal of these amino acids does not result in a fully activated channel.
  • the failure of the ⁇ NEG2-CFTR channel to respond to PKA does not result from inability of the channel to be phosphorylated, for an in vitro assay using (- 32 P-ATP showed comparable phosphorylation of wt-CFTR and ⁇ NEG2-CFTR ( Figure 1C).
  • removal of NEG2 from CFTR completely eliminates the PKA dependence of the chloride channel, although the ⁇ NEG2-CFTR channel still contains all ten PKA sites and can be phosphorylated.
  • NEG2 polypeptide stimulates both wild-type and ⁇ NEG2 CFTR proteins at concentrations greater than 0.44 ⁇ M.
  • synthetic NEG2 a 22 amino acid peptide
  • the diary plot of open probability as a function of time shows the activity of a single wt-CFTR channel during the course of the experiment ( Figure 3A). After peptide addition, there are periods of intense stimulation that last 4 to 8 minutes.
  • Example 5 The NEG2 peptide decreases the closed time of the wild-type CFTR protein.
  • the open time distributions of the wt-CFTR did not change during peptide stimulation, as both control (without NEG2 peptide) and peptide-stimulated channels had an open lifetime of approximately 120 ms (Figure 4A).
  • the increase in the open probability is not due to a change in the closing rate of the channel.
  • the closed time distribution for the stimulated channel is clearly shifted to the left compared to the control channel ( Figure 4B).
  • ⁇ cl fast ( ⁇ cl )
  • ⁇ c2 intermediate ( ⁇ c2 )
  • ⁇ c3 long ( ⁇ c3 ) closed component.
  • NEM NAM results in irreversible stimulation of PKA-phosphorylated CFTR chloride channel activity (Cotten and Welsh, 1997), further emphasizing the importance of NEG2 in CFTR regulation.
  • the wt, ⁇ NEG1-, and ⁇ NEG2-CFTR cDNAs were subcloned into an Epstein-Barr virus-based episomal eukaryotic expression vector, pCEP4 (Invitrogen, San Diego, CA), between the Nhel and Xhol restriction sites.
  • pCEP4 Epstein-Barr virus-based episomal eukaryotic expression vector
  • ⁇ NEG2 deletion mutants were created using the pALTER mutagenesis system and shuttled from pALTER into pCEP4 by substituting the corresponding fragment in pCEP4 wt-CFTR with the mutant fragment between the Xhol and BstZ171 restriction sites.
  • the ⁇ NEG1-CFTR cDNA has 27 bases deleted (amino acids 725-733).
  • the ⁇ NEG2-CFTR cDNA has 66 bases deleted (amino acids 817-838).
  • a human embryonic kidney cell line (293-EBNA HEK; Invitrogen) was used for transfection and expression of the CFTR proteins (Ma et al., 1997, Ma et al, 1996, Xie et al, 1995).
  • the HEK-293 cell line contains a pCMV-EBNA vector, which constitutively expresses the Epstein-Barr virus nuclear antigen- 1 (EBNA-1) gene product and increases the transfection efficiency of Epstein-Barr virus-based vectors.
  • the cells were maintained in Dulbecco's Modified Eagle Medium with 10% FBS and 1% L-glutamine.
  • Vesicle preparation from transfected HEK 293 cells HEK-293 cells transfected with pCEP4(CFTR) were harvested and homogenized using a combination of hypotonic lysis and Dounce homogenization in the presence of protease inhibitors (Ma et al, 1997, Ma et al, 1996, Xie et al, 1995). Microsomes were collected by centrifugation of postnuclear supernatant (4500 x g, 15 min) at 100,000 x g for 20 min and resuspended in a buffer containing 250 mM sucrose, 10 mM HEPES, pH 7.2. The membrane vesicles were stored at -75°C until use.
  • CFTR proteins isolated in membrane vesicles were bound to protein G agarose using a mouse monoclonal anti-human CFTR antibody (Genzyme).
  • the protein G agarose was washed, and (- 32 P-ATP (10 (Ci) and protein kinase A (-10 units/50(1) was added. Samples were incubated at 30(C for one hour during phosphorylation. Excess
  • the 22 amino acid peptide corresponding to NEG2 was custom made by Quality Controlled Biochemicals, Inc.
  • the peptide was resuspended in water to a concentration of 1 mg/ml and pH was adjusted to a physiological range (7.2-7.4) using KOH and HC1.
  • the space filling model of the NEG2 peptide was generated, based on secondary structure predictions (Geourjon and Deleage, 1995, Rost and Sander, 1993, Rost and Sander, 1994), using the Insight II program from Molecular Simulations Incorporated.
  • Lipid bilayer membranes were formed across an aperture of -200 (m diameter with a mixture of phosphatidylethanolamine:phosphatidylserine:cholesterol in a ratio of 5:5: 1.
  • the lipids were dissolved in decane at a concentration of 33 mg/ml.
  • the recording solutions contained: cis (intracellular), 200 mM CsCl, 1 mM MgCl 2 , 2 mM
  • HEPES-Tris (pH 7.4). Vesicles (1-4 (1) containing either wild-type, ⁇ NEG1-, or ⁇ NEG2-CFTR were added to the cis solution. The PKA catalytic subunit was present at a concentration of 50 units/ml in the cis solution unless noted otherwise. Single channel currents were recorded with an Axopatch 200A patch clamp unit (Axon
  • SOPMA significant improvements in protein secondary structure prediction by consensus prediction from multiple alignments.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pulmonology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Phosphorylation of the cystic fibrosis transmembrane conductance regulator (CFTR) by cyclic AMP-dependent protein kinase (PKA) is essential for opening the CFTR chloride channel. A short segment containing many negatively charged amino acids (817-838, NEG 2) within the regulatory (R) domain of CFTR is a critical regulator of the chloride channel activity. Deletion of NEG2 from CFTR completely eliminates the PKA dependence of the chloride channel. Exogenous NEG2 peptide interacts with the CFTR molecule and exhibits stimulatory effects on CFTR function. Our data suggest that NEG2 interact with other cytosolic domains of CFTR to control the opening transitions of the chloride channel.

Description

ENHANCERS OF CFTR CHLORIDE CHANNEL FUNCTION
This invention was made with government support under RO1 HL/DK 49003, P30 DK27651 and RO1 DK51770 awarded by the National Institute of Health. The government has certain rights in the invention.
TECHNICAL FIELD OF THE INNENTΪON
This invention is related to the field of cystic fibrosis. More particularly, it is related to the area of therapeutic treatments and drug discovery for treating cystic fibrosis.
BACKGROUND OF THE INVENTION Defects in CFTR, a chloride channel located in the apical membrane of epithelial cells, are associated with the common genetic disease, cystic fibrosis (Quinton, 1986, Welsh and Smith, 1993, Zielenski and Tsui, 1995). CFTR is a 1480 amino acid protein that is a member of the ATP binding cassette (ABC) transporter family (Riordan et al, 1989, Higgins, 1992). Each half of CFTR contains a transmembrane domain and a nucleotide binding fold (NBF), and the two halves are connected by a regulatory, or R domain. The R domain is unique to CFTR and contains several consensus PKA phosphorylation sites (Cheng et al, 1991, Picciotto et al., 1992).
Opening of the CFTR channel is controlled by PKA phosphorylation of serine residues in the R domain (Tabcharani et al., 1991, Bear et al., 1992) and ATP binding and hydrolysis at the NBFs (Anderson et al., 1991 , Gunderson and Kopito, 1995). Phosphorylation adds negative charges to the R domain, and introduces global conformational changes reflected by the reduction in the α-helical content of the R domain protein (Dulhanty and Riordan, 1994). Thus, electrostatic and/or allosteric changes mediated by phosphorylation are likely to be responsible for interactions between the R domain and other CFTR domains that regulate channel function (Rich et al, 1993, Gadsby and Nairn, 1994).
Rich et al, 1991 showed that deletion of amino acids 708-835 from the R domain (ΔR-CFTR), which removes most of the PKA consensus sites, renders the
CFTR channel PKA independent, but the open probability of ΔR-CFTR is one-third that of the wild type channel and does not increase upon PKA phosphorylation (Ma et al., 1997, Winter and Welsh, 1997). Thus, it is possible that deletion of the R domain removes both inhibitory and stimulatory effects conferred by the R domain on CFTR chloride channel function. This conclusion is supported by studies that show that addition of exogenous unphosphorylated R domain protein (amino acids 588-858) to wt-CFTR blocks the chloride channel (Ma et al., 1996), suggesting that the unphosphorylated R domain is inhibitory. Conversely, exogenous phosphorylated R domain protein (amino acids 588-855 or 645-834) stimulated the ΔR-CFTR channel, suggesting that the phosphorylated R domain is stimulatory (Ma et al, 1997, Winter and Welsh, 1997). Therefore, it appears that the manifest activity (stimulatory or inhibitory) depends on the phosphorylation state of the R domain.
About 25% of the known 700 mutations in CFTR produce a mutant CFTR protein which is properly transported to the apical membrane of epithelial cells but have only low level, residual channel activity. There is a need in the art for agents which can boost the level of channel activity in those mutants having low level activity. SUMMARY OF THE INVENTION
It is an object of the present invention to provide an isolated polypeptide useful for enhancing the open probability of CFTR chloride channels.
It is another object of the present invention to provide a method of activating a CFTR protein to enhance its open probability. These and other objects of the invention are achieved by providing one or more of the embodiments described below. In one embodiment of the invention an isolated polypeptide is provided. The polypeptide comprises a portion of CFTR (cystic fibrosis transmembrane conductance regulator) protein of between 10 and 100 amino acids, said portion comprising 18 amino acids as shown in SEQ ID NO: 1.
In another embodiment of the invention a method is provided for activating a CFTR protein. A polypeptide is applied to a CFTR protein which forms a cAMP regulated chloride channel. The polypeptide consists of a portion of CFTR protein which comprises 18 amino acids as shown in SEQ ID NO: 1, whereby the open probability of the channel formed by the CFTR increases by at least 25%.
According to another aspect of the invention a method is provided for activating a CFTR protein. A polypeptide is applied to a CFTR protein which forms a cAMP regulated chloride channel. The polypeptide consists of a portion of CFTR protein which comprises 22 amino acids as shown in SEQ ID NO: 2, whereby the open probability of the channel formed by the CFTR increases by at least 25%.
The present invention thus provides the art with reagents and tools for enhancing function of channels which are defective in cystic fibrosis patients. DETAILED DESCRIPTION OF THE DRAWINGS Figure 1. Deletion of Negatively Charged Regions from the R Domain Results in
Expression of Mature Glycosylated, Phosphorylatable CFTR Proteins (Figure 1A) Sequences of NEGl and NEG2 within the R domain. Residues where mutations have been identified in the CFTR cDNA are underlined (E822K, E826K, D836Y). (Figure IB) NEG2 is predicted to form an amphipathic α-helix as determined by secondary structure determination (Geourjon and Deleage, 1995, Rost and Sander, 1993, Rost and Sander, 1994) and illustrated in this space filling model. Negatively charged residues are colored pink, and the positively charged lysine is colored green.
(Figure 1C) In vitro phosphorylation of wt-(lane 1), ΔNEG1- (lane 2) and ΔNEG2-CFTR (lane 3) by PKA in the presence of γ-32P-ATP. Both the core (band B) and fully glycosylated (band C) forms of all three CFTR molecules are phosphorylated.
Figure 2. ΔNEG2-CFTR Forms a Chloride Channel that is Unregulated by PKA
(Figure 2A) Single channel currents of wt, ΔNEG1- and ΔNEG2-CFTR incorporated into the lipid bilayer. While activities of wt-and ΔNEG1-CFTR absolutely require the presence of PKA in the c.-s'-intracellular solution, the ΔNEG2-CFTR channel opens without PKA phosphorylation. (Figure 2B) Diary plot of ΔNEG2-CFTR channel open probability versus time shows that addition of 200 units/ml of PKA, a maximally stimulating concentration, does not affect channel activity. The dashed line indicates the average open probability for each segment of the experiment. Channels were recorded at -100 mV.
Figure 3. The Synthetic NEG2 Peptide both Stimulates and Inhibits CFTR (Figure 3A) Diary plot (open probability versus time) of a wt-CFTR channel illustrating the effect of the NEG2 peptide on the open probability of the channel in the planar lipid bilayer. The concentration of synthetic NEG2 in the cw-intracellular solution is indicated above the plot.
(Figure 3B) Single channel currents from the wt-CFTR channel were acquired at -80 mV at the points indicated in A. The cw-intracellular solution contained 2 mM ATP and 50 units PKA/ml.
(Figure 3C) Single channel trace from ΔNEG2-CFTR incorporated into the lipid bilayer membrane. Traces were acquired at -80 mV. The c/s-solution contained 2 mM
ATP and no PKA. The top two traces were acquired before synthetic NEG2 peptide addition, with the second trace being an expansion of the first. In the bottom two traces, 0.44 μM of the NEG2 peptide has been added and stimulation is observed. The closed time visibly decreases after peptide addition.
Figure 4. NEG2 Enhances CFTR Channel Activity by Increasing the Opening
Rate of the Channel Histograms of open and closed events of the wt-CFTR channel at -80 mV were generated without peptide (control, left panel) and with 4.4 μM NEG2 peptide in the c/s-solution (right panel).
(Figure 4A) The open time histograms contain a single exponential component with a time constant of 124 ms (control) and 105 ms (pep tide-stimulated). (Figure 4B) The closed time histograms contain a fast component and multiple slow components
(Figure 4C) The closed-burst duration histograms were constructed using a delimiter of 40 ms (represented by the arrow in B). The solid lines in C represent the fit according to the double exponential equation y = P2*exp[t-α -expαt-α )] 2+
P3*exp[t-α3-exp(t-α3)] where a 2 = log τ Λ a 3= log τ t3P 2= probability of the intermediate closed component, and P3 = probability of the long closed component. The best fit parameters are P2 = 0.811, τc2 = 459 ms, P3 = 0.189, τc3 = 2494 ms (control); P2 = 0.957, τc2 = 105 ms, P3 = 0.043, τc3 = 1652 ms (peptide-stimulated).
DETAILED DESCRIPTION OF THE INVENTION
It is a discovery of the present inventors that negatively charged amino acids at the carboxyl terminal of the R domain (817-838, NEG2) is involved in both the stimulatory and inhibitory functions of the R domain on the chloπde channel. Moreover, a polypeptide which contains this portion of the CFTR ammo acid sequence can be used to enhance the open probability of both wild-type and minimally active mutant CFTR protein.
The isolated polypeptide according to the invention consists of a portion of CFTR (cystic fibrosis transmembrane conductance regulator) protein. The portion preferably contains at least 18 ammo acids as shown m SEQ ID NO: 1. However, fewer amino acid residues of the sequence may be used if they retain the channel enhancing function descπbed herein for the 18 and 22 residue polypeptides. See also SEQ ID NO. 2. Thus the polypeptide may be from about 10 or 15 ammo acid residues up to about 30 or even 100 amino acid residues An isolated polypeptide may be synthetic or made in a recombinant organism. It may be a proteolytic cleavage product of a larger primary expression product, including full-length, wild-type CFTR. Preferably the polypeptide will be free of full-length CFTR. The polypeptide will preferably be free of other proteins and polypeptides as well. However, it may be desirable that the polypeptide be fused to another polypeptide to provide additional functional propeπies. For example, fusion to another protein such as keyhole limpet hemocyanin would be used to increase immunogenicity. Another desirable fusion partner is a membrane- penetrating peptide. Such peptides include VP-22 (SEQ ID NO: 3), as well as the peptides shown in SEQ ID NO: 4 and SEQ ID NO: 5. Such peptides can be used to facilitate the uptake by target cells of the polypeptide.
The polypeptides of the present invention can be used to enhance the function of wild-type or minimally active mutant CFTR proteins. The polypeptide functions to decrease the closed time of the channels formed by CFTR. A polypeptide can be applied to the CFTR protein in any context. It can be applied in vitro or in vivo. If in vitro it can be to CFTR in cultured cells or to planar bilayer membranes containing
CFTR. If in vivo, the polypeptide can be applied directly to airway epithelial cells. Such application can be by any means known in the art, including but not limited to using a gargle or a nebulizer to deliver aerosolized polypeptide to the target cells. In addition, the peptide can be delivered in an indirect mode, by delivering a gene construct to the airway epithelial cells, which when taken up by the cells causes them to express the polypeptide. The delivery of the polypeptide to the CFTR preferably increases the open probability of the channel formed by the CFTR by at least 25%. More preferably it increases the open probability by at least 50%, at least 75%, at least 100%), at least 125%, at least 150%, or at least 200%. A CFTR construct comprises a nucleic acid sequence encoding the amino acid sequence shown in SEQ ID NO 1. A suitable promoter for expression in lung epitheha is also desirable. Many such promoters are known in the art, and any can be used as appropriate for a particular application. It is believed that the administration of the polypeptide of the present invention will be the most useful m treatment of a class of mutants which produce CFTR proteins which are properly delivered to the plasma membrane but which are only residually or minimally active. Known mutants of CFTR are listed at http://www.genet.sickkids.on.ca/cftr-cgi-bin/fulltable. One can determine that a particular CFTR mutant is fully processed and reaches the plasma membrane m a
Western blot assay using antibody against CFTR. Fully processed mutants achieve mature glycosylation status and appear on the gel as "band C and band B" whereas mutants that are retained in the endoplasmic reticulum are not fully glycosylated and show only "band B". See Example 2, below and Figure lC. The above disclosure generally descπbes the present mvention. A more complete understanding can be obtained by reference to the following specific examples which are provided herein for purposes of illustration only, and are not intended to limit the scope of the invention.
EXAMPLES
Example 1: Deletion of a negatively charged region (a.a. 817-838) from the R domain of CFTR alters PKA-dependent regulation of the CFTR channel
CFTR contains a large intracellular regulatory (R) domain where multiple PKA phosphorylation sites are located. There are two regions withm the R domain that contain a high proportion of negatively charged amino acids, a.a. 725-733 (NEGl) and a.a. 817-838 (NEG2). It is possible that these two regions could have allosteπc or electrostatic interactions with other regions of CFTR and thus affect its chloride channel function. To test the role of NEGl and NEG2, two deletion mutants, NEGl -CFTR and NEG2-CFTR, were created. The CFTR mutants were transiently expressed in HEK 293 cells, and their single channel functions were studied using the bilayer reconstitution system. Western blots indicate that both NEGl -CFTR and NEG2-CFTR process normally and traffic to the plasma membrane of HEK 293 cells. Both mutants form functional chloride channels in the bilayer membrane, with single channel conductances similar to the wt-CFTR channel. Like wt-CFTR, opening of NEG1-CFTR requires absolutely PKA phosphorylation and ATP binding/hydrolysis. In contrast to wt-CFTR, opening of NEG2-CFTR does not require PKA phosphorylation. Thus, deletion of NEG2, but not NEGl, alters PKA-dependent regulation of the CFTR chloride channel. Our data suggest that NEG2 could form a 'putative gating particle' of the CFTR channel possibly through electrostatic and/or allosteric interactions with other domains of CFTR.
Example 2: Δ NEGl- and Δ NEG2-CFTR are glycosylated.
The R domain of CFTR contains two negatively charged regions, amino acids 725-733 (NEGl) and amino acids 817-838 (NEG2), that reside in close proximity to two PKA phosphorylation sites, S737 and S813, used in vivo (Figure 1A) (Cheng, et al. 1991). NEG2 is predicted to form an amphipathic (-helical structure with a negatively charged face (Figure IB) (Geourjon and Deleage, 1995, Rost and Sander, 1993, Rost and Sander, 1994). Three mutations (E822K, E826K, D836Y), two of which were clearly obtained from patients with CF (E822K and D836Y). have been identified withm the NEG2 region that result in the removal of negative charges (www.genet.sickkids.on.ca). The E822K CFTR channel has a low open probability relative to wt-CFTR (wild type-CFTR), but the E826K CFTR channel has single channel properties similar to wt-CFTR (Vankeerberghen et al., 1998). The presence of these disease-causing mutations suggests the potential importance of the NEG2 region. To investigate the roles of NEGl and NEG2 in CFTR function, these regions were deleted from CFTR using mutagenesis and subclonmg. The ΔNEG1- and ΔNEG2-CFTR proteins were transiently expressed in human embryonic kidney 293 cells. Membrane vesicles containing the CFTR proteins were isolated and subjected to
SDS-PAGE. Like wt-CFTR, both ΔNEG1- and ΔNEG2-CFTR are present both in the core glycosylated (band B) and the fully glycosylated form (band C) (Figure 1C). Example 3: The open probability of the ΔNEG2-CFTR chloride channel is much less than that of wild type but is independent PKA, although it contains all PKA phosphorylation sites.
Single channel measurements indicate that the ΔNEG1-CFTR channel is similar to wt-CFTR in its PKA dependence. No chloride channels are observed in the absence of PKA (Figure 2A) and the open probability in the presence of PKA and ATP is similar to wt-CFTR. In contrast, the ΔNEG2-CFTR channel opens without PKA (Figure 2A). The constitutive activity of the ΔNEG2-CFTR channel is unlikely to be due to the endogenous phosphorylation of the ΔNEG2-CFTR protein, since protein phosphatase 2A, which decreases activity of the wt-CFTR opened by PKA and ATP (Ma et al., 1997), has no effect on the ΔNEG2-CFTR channel (n=4) Moreover, addition of PKA up to 200 units/ml, four times the concentration required to fully activate wt-CFTR (Ma et al., 1997), does not increase the open probability of the channel (Figure 2B). ΔNEG2-CFTR has conductance properties similar to wild type (Tao et al., 1996). However, the open probability of the ΔNEG2-CFTR chloride channel is much less than that of wild type and cannot be increased by PKA (P0= 0.035 ( 0.012 and P0= 0.026 ( 0.013 without and with PKA respectively, n=5). While NEG2 may represent an inhibitory region, removal of these amino acids does not result in a fully activated channel. The failure of the ΔNEG2-CFTR channel to respond to PKA does not result from inability of the channel to be phosphorylated, for an in vitro assay using (-32P-ATP showed comparable phosphorylation of wt-CFTR and ΔNEG2-CFTR (Figure 1C). Thus, it appears that removal of NEG2 from CFTR completely eliminates the PKA dependence of the chloride channel, although the ΔNEG2-CFTR channel still contains all ten PKA sites and can be phosphorylated.
Example 4: NEG2 polypeptide stimulates both wild-type and Δ NEG2 CFTR proteins at concentrations greater than 0.44 μM. To test whether the NEG2 region is responsible for both stimulatory and inhibitory interactions between the R domain and other domains, synthetic NEG2, a 22 amino acid peptide, was added to the cis-intracellular side of single CFTR channels captured in the planar lipid bilayer (Figure 3). The diary plot of open probability as a function of time shows the activity of a single wt-CFTR channel during the course of the experiment (Figure 3A). After peptide addition, there are periods of intense stimulation that last 4 to 8 minutes. These stimulatory periods are followed by either a return to the basal level of activity before peptide addition, or by an almost complete inhibition of the channel, where only a flickery 3 pS conductance is observed. During stimulation, the open probability more than doubles and more transitions are observed between the open and closed states (Figure 3B). The stimulatory response was observed in 6 of 7 experiments at concentrations >0.44 μM (the remaining channel was inhibited upon peptide addition (4.4 μM) and no stimulation was seen). Profound inhibition was observed in three channels at concentrations > 4.4 μM. When the NEG2 peptide was added to the intracellular side of the ΔNEG2-CFTR channel, which lacks its own endogenous NEG2 sequence, a similar stimulatory response was observed (Figure 3C).
Example 5: The NEG2 peptide decreases the closed time of the wild-type CFTR protein. In order to understand the mechanism responsible for the increase in open probability, the gating kinetics of wt-CFTR without peptide and during stimulation by synthetic NEG2 were analyzed. The open time distributions of the wt-CFTR did not change during peptide stimulation, as both control (without NEG2 peptide) and peptide-stimulated channels had an open lifetime of approximately 120 ms (Figure 4A). Thus, the increase in the open probability is not due to a change in the closing rate of the channel. However, the closed time distribution for the stimulated channel is clearly shifted to the left compared to the control channel (Figure 4B). There are three components to the closed state, a fast (τcl), an intermediate (τc2), and a long (τc3) closed component. The fast closed component is probably due to closings within a burst (Carson et al, 1995). Therefore, to identify better the closed times between bursts, a delimiter of τc = 40 ms was set at the nadir between the fast and intermediate closed times (illustrated by the arrow in Figure 4B) to generate the closed-burst duration histograms. As shown in Figure 4C, following peptide stimulation, the intermediate closed time was reduced from 459 ms to 105 ms, whereas the long closed time remained relatively unchanged Thus, the interaction of NEG2 with CFTR increased the intermediate-opening rate of the channel This increase in opening rate is similar to that observed when the phosphorylated R domain protein (amino acids 645-834) was added to CFTR- ΔR/S660A in excised, inside-out patches (Winter and Welsh, 1997) Additionally, modification of C832, which resides within NEG2, by N-ethylmaleimide
(NEM) results in irreversible stimulation of PKA-phosphorylated CFTR chloride channel activity (Cotten and Welsh, 1997), further emphasizing the importance of NEG2 in CFTR regulation.
These data, taken together, show that the NEG2 region confers both stimulatory and inhibitory functions of the R domain on the CFTR channel. When this region is deleted from CFTR, the resultant channel opens without PKA (loss of inhibitory function), but it never achieves open probability comparable to wild type even when phosphorylated with PKA (loss of stimulatory function) This same sequence, expressed as a peptide, results in stimulation of channel openings at lower concentrations and profound inhibition of charmel activity at higher concentrations, when added to the intracellular side of CFTR channels. It seems likely that this sequence interacts with CFTR at different sites on the nucleotide binding domains to either stimulate or inhibit channel openings. Phosphorylation of the R domain, in this model, changes its conformation and thus presents the NEG2 sequence better to the stimulatory than the inhibitory site. A current model for charmel opening is that phosphorylated channels open in response to ATP binding and hydrolysis at the first nucleotide binding fold (NBF1) (Gadsby and Nairn, 1994, Ma and Davis, 1998). Since stimulation by NEG2 occurs by increasing channel openings, a likely site of stimulation is NBF1, though other models are possible. METHODS USED IN EXAMPLES 1-5 Subcloning of CFTR gene
The wt, ΔNEG1-, and ΔNEG2-CFTR cDNAs were subcloned into an Epstein-Barr virus-based episomal eukaryotic expression vector, pCEP4 (Invitrogen, San Diego, CA), between the Nhel and Xhol restriction sites. The ΔNEG1 and
ΔNEG2 deletion mutants were created using the pALTER mutagenesis system and shuttled from pALTER into pCEP4 by substituting the corresponding fragment in pCEP4 wt-CFTR with the mutant fragment between the Xhol and BstZ171 restriction sites. The ΔNEG1-CFTR cDNA has 27 bases deleted (amino acids 725-733). The ΔNEG2-CFTR cDNA has 66 bases deleted (amino acids 817-838).
Expression of CFTR in HEK 293 cells
A human embryonic kidney cell line (293-EBNA HEK; Invitrogen) was used for transfection and expression of the CFTR proteins (Ma et al., 1997, Ma et al, 1996, Xie et al, 1995). The HEK-293 cell line contains a pCMV-EBNA vector, which constitutively expresses the Epstein-Barr virus nuclear antigen- 1 (EBNA-1) gene product and increases the transfection efficiency of Epstein-Barr virus-based vectors. The cells were maintained in Dulbecco's Modified Eagle Medium with 10% FBS and 1% L-glutamine. Geneticin (G418, 250 (g/ml) was added to the cell culture medium to maintain selection of the cells containing the pCMV-EBNA vector. Lipofectamine reagent (Life Technologies, Inc) in Optimem media (serum-free) was used to transfect the HEK-293 cells with pCEP4(wt), pCEP4(ΔNEGl), or pCEP4(ΔNEG2). After 5 hours, serum was added to the media (10% final serum concentration). Twenty-four hours after transfection, the transfection media was replaced with fresh media. The cells were harvested two days after transfection and microsomal membrane vesicles were prepared for single channel measurements in the lipid bilayer reconstitution system.
Vesicle preparation from transfected HEK 293 cells HEK-293 cells transfected with pCEP4(CFTR) were harvested and homogenized using a combination of hypotonic lysis and Dounce homogenization in the presence of protease inhibitors (Ma et al, 1997, Ma et al, 1996, Xie et al, 1995). Microsomes were collected by centrifugation of postnuclear supernatant (4500 x g, 15 min) at 100,000 x g for 20 min and resuspended in a buffer containing 250 mM sucrose, 10 mM HEPES, pH 7.2. The membrane vesicles were stored at -75°C until use.
In vitro phosphorylation of CFTR proteins
CFTR proteins isolated in membrane vesicles were bound to protein G agarose using a mouse monoclonal anti-human CFTR antibody (Genzyme). The protein G agarose was washed, and (-32P-ATP (10 (Ci) and protein kinase A (-10 units/50(1) was added. Samples were incubated at 30(C for one hour during phosphorylation. Excess
(γ-32P-ATP was removed, and SDS-PAGE sample buffer (200 mM Tris-Cl, pH 6.7, 9% SDS, 6% beta-mercaptoethanol, 15% glycerol, and 0.01 % bromophenol blue) was added to denature CFTR and release it from the protein G agarose. The samples were subjected to electrophoresis on a 5% SDS-polyacrylamide gel, transferred to a polyvinylidene difluoride membrane, and exposed to film. Preparation ofNEG2 peptides
The 22 amino acid peptide corresponding to NEG2 was custom made by Quality Controlled Biochemicals, Inc. The peptide was resuspended in water to a concentration of 1 mg/ml and pH was adjusted to a physiological range (7.2-7.4) using KOH and HC1. The space filling model of the NEG2 peptide was generated, based on secondary structure predictions (Geourjon and Deleage, 1995, Rost and Sander, 1993, Rost and Sander, 1994), using the Insight II program from Molecular Simulations Incorporated.
Reconstitution of CFTR channels in lipid bilayer membranes
Lipid bilayer membranes were formed across an aperture of -200 (m diameter with a mixture of phosphatidylethanolamine:phosphatidylserine:cholesterol in a ratio of 5:5: 1. The lipids were dissolved in decane at a concentration of 33 mg/ml. The recording solutions contained: cis (intracellular), 200 mM CsCl, 1 mM MgCl2, 2 mM
ATP, and 10 mM HEPES-Tris (pH 7.4); trans (extracellular), 50 mM CsCl, 10 mM
HEPES-Tris (pH 7.4). Vesicles (1-4 (1) containing either wild-type, ΔNEG1-, or ΔNEG2-CFTR were added to the cis solution. The PKA catalytic subunit was present at a concentration of 50 units/ml in the cis solution unless noted otherwise. Single channel currents were recorded with an Axopatch 200A patch clamp unit (Axon
Instruments). The currents were sampled at 1-2.5 ms/point. Single charmel data analyses were performed with pClamp and TIPS softwares. References
Anderson, M.P., Berger, H.A., Rich, D.P., Gregory, R.J., Smith, A.E., and Welsh, M.J. (1991). Nucleoside triphosphates are required to open the CFTR chloride channel. Cell 67, 775-784.
Bear, C.E., Li, C, Kartner, N., Bridges, R.J., Jensen, T.J., Ramjeesingh, M., and
Riordan, J.R. (1992). Purification and functional reconstitution of the cystic fibrosis transmembrane conductance regulator (CFTR). Cell 68, 809-818. Carson, M.R., Travis, S.M., and Welsh, M.J. (1995). The two nucleotide-binding domains of cystic fibrosis transmembrane conductance regulator (CFTR) have distinct functions in controlling channel activity. J. Biol. Chem. 270, 1711-1717.
Cheng, S.H., Rich, D.P., Marshall, J., Gregory, R.J., Welsh, M.J., and Smith, A.E.
(1991). Phosphorylation of the R domain by cAMP-dependent protein kinase regulates the CFTR chloride channel. Cell 66, 1027-1036.
Cotten, J.F. and Welsh, M.J. (1997). Covalent modification of the regulatory domain irreversibly stimulates cystic fibrosis transmembrane conductance regulator. J. Biol.
Chem. 272, 25617-25622.
Dulhanty, A.M. and Riordan, J.R. (1994). Phosphorylation by cAMP-dependent protein kinase causes a conformational change in the R domain of the cystic fibrosis transmembrane conductance regulator. Biochemistry 22, 4072-4079.
Gadsby, D.C. and Nairn, A.C. (1994). Regulation of CFTR channel gating. Trends
Biochem. Sci. 19, 513-518.
Geourjon, C. and Deleage, G. (1995). SOPMA: significant improvements in protein secondary structure prediction by consensus prediction from multiple alignments.
CABIOS 11, 681-684.
Gunderson, K.L. and Kopito, R.R. (1995). Conformational states of CFTR associated with channel gating: the role of ATP binding and hydrolysis. Cell 82, 231-239.
Higgens, C.F. (1992). ABC transporters: from microorganisms to man. Annu. Rev. Cell Biol. 8, 67-113.
Ma, J. and Davis, P.B. (1998). What we know and what we do not know about cystic fibrosis transmembrane conductance regulator. Clinics in Chest Medicine 19, 459-471.
Ma, J., Tasch, J.E., Tao, T., Zhao, J., Xie, J., Drumm, M ., and Davis, P.B. (1996). Phosphorylation-dependent block of cystic fibrosis transmembrane conductance regulator chloride channel by exogenous R domain protein. J. Biol. Chem. 271,
7351-7356.
Ma, J., Zhao, J., Drumm, M ., Xie, J., and Davis, P.B. (1997). Function of the R domain in the cystic fibrosis transmembrane conductance regulator chloride channel. J. Biol. Chem. 272, 28133-28141.
Picciotto, M.R., Conn, J.A., Bertuzzi, G., Greengard, P., and Nairn, A.C. (1992).
Phosphorylation of the cystic fibrosis transmembrane conductance regulator. J. Biol.
Chem. 267, 12742-12752.
Quinton, P.M. (1986). Missing Cl" conductance in cystic fibrosis. Am. J. Physiol. 251, C649-C652.
Rich, D.P., Berger, H.A., Cheng, S.H., Travis, S.M., Saxena, M., Smith, A.E., and
Welsh, M.J. (1993). Regulation of the cystic fibrosis transmembrane conductance regulator Cf channel by negative charge in the R domain. J. Biol. Chem. 268,
20259-20267. Rich, D.P., Gregory, R.J., Anderson, M.P., Manavalan, P., Smith, A.E., and Welsh,
M.J. (1991). Effect of deleting the R domain on CFTR-generated chloride channels.
Science 253, 205-207.
Riordan, J., Rommens, J., Kerem, B.-S., Noa, A., Rozmahel, R., Grzelczak, Z.,
Zielenski, J., Lok, S., Plavsic, N., Chou, J.-L., Drumm, M., Iannuzzi, M., Collins, F., and Tsui, L.-C. (1989). Identification of the cystic fibrosis gene: cloning and characterization of complementary DNA. Science 245, 1066-1073.
Rost, B. and Sander, C. (1993). Prediction of protein structure at better than 70% accuracy. J. Mol. Biol. 232, 584-599. Rost, B. and Sander, C. (1994). Combining evolutionary information and neural networks to predict protein secondary structure. Proteins 19, 55-72.
Tabcharani, J.A., Chang, X.-B., Riordan, J.R. and Hanrahan, J.W. (1991).
Phosphorylation-regulated Cl- channel in CHO cells stably expressing the cystic fibrosis gene. Nature 352, 628-631. Tao, T., Xie, J., Drumm, M ., Zhao, J., Davis, P.B., and Ma, J. (1996). Slow conversions among subconductance states of cystic fibrosis transmembrane conductance regulator chloride channel. Biophys. J. 70, 743-753.
Vankeerberghen, A., Wei, L., Jaspers, M., Cassiman, J.-J., Nilius, B., and Cuppens, H.
(1998). Characterization of 19 disease-associated missense mutations in the regulatory domain of the cystic fibrosis transmembrane conductance regulator. Hum. Mol. Genet.
7, 1761-1769.
Welsh, M.J. and Smith, A.E. (1993). Molecular mechanisms of CFTR chloride channel dysfunction in cystic fibrosis. Cell 73, 1251-1254.
Winter, M.C. and Welsh, M.J. (1997). Stimulation of CFTR activity by its phosphorylated R domain. Nature 389, 294-296.
Xie, J., Drumm, M ., Ma, J., and Davis, P.B. (1995). Intracellular loop between transmembrane segments IV and V of cystic fibrosis transmembrane conductance regulator is involved in regulation of chloride channel conductance state. J. Biol. Chem.
270, 28084-28091. Zielenski, J. and Tsui, L.C. (1995). Cystic fibrosis: genotypic and phenotypic variations. Annu. Rev. Genetics 29, 777-807.

Claims

CLAIMS:
1. An isolated polypeptide comprising a portion of CFTR (cystic fibrosis transmembrane conductance regulator) protein of between 10 and 100 amino acids, said portion comprising 18 amino acids as shown in SEQ ID NO: 1.
2. The polypeptide of claim 1 which comprises 22 amino acids as shown in SEQ
ID NO: 2.
3. The polypeptide of claim 1 wherein the polypeptide is fused to a membrane- penetrating peptide.
4. The polypeptide of claim 2 wherein the polypeptide is fused to a membrane- penetrating peptide.
5. The polypeptide of claim 3 wherein the membrane-penetrating peptide is selected from the group consisting of: VP-22 (SEQ ID NO: 3), (SEQ ID NO: 4), and (SEQ ID NO: 5).
6. The polypeptide of claim 4 wherein the membrane-penetrating peptide is selected from the group consisting of: VP-22 (SEQ ID NO: 3), (SEQ ID NO:
4), and (SEQ ID NO: 5).
7. The polypeptide of claim 1 which is free of phosphorylation.
8. A method of activating a CFTR protein comprising: applying a polypeptide to a CFTR protein which forms a cAMP regulated chloride channel, said polypeptide comprising a portion of
CFTR protein of between about 10 and 100 amino acids, said portion comprising 18 amino acids as shown in SEQ ID NO: 1, whereby the open probability of the channel formed by the CFTR increases by at least 25%.
9. The method of claim 8 wherein the open probability of the channel formed by the CFTR increases by at least 50%.
10. The method of claim 8 wherein the open probability of the channel formed by the CFTR increases by at least 75%.
1 1. The method of claim 8 wherein the open probability of the channel formed by the CFTR increases by at least 100%.
12. The method of claim 8 wherein the open probability of the channel formed by the CFTR increases by at least 125%.
13. The method of claim 8 wherein the open probability of the channel formed by the CFTR increases by at least 150%.
14. The method of claim 8 wherein the open probability of the channel formed by the CFTR increases by at least 200%.
15. The method of claim 8 wherein the CFTR protein is a mutant which reaches a cell's plasma membrane but fails to undergo full activation.
16. The method of claim 15 wherein the CFTR protein is listed at http://www.genet.sickkids.on.ca/cftr-cgi-bin/fulltable.
17. The method of claim 8 wherein the step of applying is performed by administering an aerosolized polypeptide to a patient with a mutant CFTR protein.
18. The method of claim 8 wherein the CFTR protein is in a lipid bilayer and a change in conductance is measured upon applying the polypeptide.
19. The method of claim 8 wherein the step of applying the polypeptide is accomplished by administering a nucleic acid encoding the polypeptide to a patient who expresses the CFTR protein, whereby the polypeptide is expressed.
20. The method of claim 19 wherein the nucleic acid is administered as an aerosol to the patient's airways.
21. A method of activating a CFTR protein comprising: applying a polypeptide to a CFTR protein which forms a cAMP regulated chloride channel, said polypeptide comprising a portion of CFTR protein of between 10 and 100 amino acids, said portion comprising 22 amino acids as shown in SEQ ID NO: 1, whereby the open probability of the channel formed by the CFTR increases by at least 25%.
22. The method of claim 21 wherein the open probability of the channel formed by the CFTR increases by at least 50%..
23. The method of claim 21 wherein the open probability of the channel formed by the CFTR increases by at least 75%.
24. The method of claim 21 wherein the open probability of the charmel formed by the CFTR increases by at least 100%.
25. The method of claim 21 wherein the open probability of the channel formed by the CFTR increases by at least 125%.
26. The method of claim 21 wherein the open probability of the charmel formed by the CFTR increases by at least 150%.
27. The method of claim 21 wherein the open probability of the channel formed by the CFTR increases by at least 200%.
28. The method of claim 21 wherein the CFTR protein is a mutant which reaches a cell's plasma membrane but fails to undergo full activation.
29. The method of claim 28 wherein the CFTR protein is listed at http://www.genet.sickkids.on.ca/cftr-cgi-bin/fulltable.
30. The method of claim 21 wherein the step of applying is performed by administering an aerosolized polypeptide to a patient with a mutant CFTR protein.
31. The method of claim 21 wherein the CFTR protein is in a lipid bilayer and a change in conductance is measured upon applying the polypeptide.
32. The method of claim 21 wherein the step of applying the polypeptide is accomplished by administering a nucleic acid encoding the polypeptide to a patient who expresses the CFTR protein, whereby the polypeptide is expressed.
33. The method of claim 32 wherein the nucleic acid is administered as an aerosol to the patient's airways.
34. The method of claim 8 or 21 wherein the polypeptide is free of phosphorylation.
PCT/US2000/004642 1999-02-24 2000-02-24 Enhancers of cftr chloride channel function WO2000050591A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU32419/00A AU3241900A (en) 1999-02-24 2000-02-24 Enhancers of cftr chloride channel function

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US12149599P 1999-02-24 1999-02-24
US60/121,495 1999-02-24

Publications (1)

Publication Number Publication Date
WO2000050591A1 true WO2000050591A1 (en) 2000-08-31

Family

ID=22397082

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/004642 WO2000050591A1 (en) 1999-02-24 2000-02-24 Enhancers of cftr chloride channel function

Country Status (2)

Country Link
AU (1) AU3241900A (en)
WO (1) WO2000050591A1 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995025796A1 (en) * 1994-03-23 1995-09-28 University Of Iowa Research Foundation C-terminally truncated cftr genes and proteins

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995025796A1 (en) * 1994-03-23 1995-09-28 University Of Iowa Research Foundation C-terminally truncated cftr genes and proteins

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
ADAMS L M ET AL: "Deletion of a negatively charged region (a.a 817-838) from the R domain of CFTR alters PKA-dependent regulation of the CFTR channel.", BIOPHYSICAL JOURNAL, vol. 74, no. 2 PART 2, February 1998 (1998-02-01), Forty-second Annual Meeting of the Biophysical Society;Kansas City, Missouri, USA; February 22-26, 1998, pages A344, XP000923128, ISSN: 0006-3495 *
COTTEN JOSEPH F ET AL: "Covalent modification of the regulatory domain irreversibly stimulates cystic fibrosis transmembrane conductance regulator.", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 272, no. 41, 1997, pages 25617 - 25622, XP002142118, ISSN: 0021-9258 *
MA JIANJIE ET AL: "Function of the R domain in the cystic fibrosis transmembrane conductance regulator chloride channel.", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 272, no. 44, 31 October 1997 (1997-10-31), pages 28133 - 28141, XP002142117, ISSN: 0021-9258 *
MA JIANJIE ET AL: "Phosphorylation-dependent block of cystic fibrosis transmembrane conductance regulator chloride channel by exogenous R domain protein.", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 271, no. 13, 1996, pages 7351 - 7356, XP002142116, ISSN: 0021-9258 *
RICH DEVRA P ET AL: "Regulation of the cystic fibrosis transmembrane conductance regulator chloride channel by negative charge in the R domain.", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 268, no. 27, 1993, pages 20259 - 20267, XP002142119, ISSN: 0021-9258 *
TASCH JASON E ET AL: "Functional dissection of the R domain of cystic fibrosis transmembrane conductance regulator.", FEBS LETTERS, vol. 445, no. 1, 19 February 1999 (1999-02-19), pages 63 - 68, XP002142114, ISSN: 0014-5793 *
WINTER MICHAEL C ET AL: "Stimulation of CFTR activity by its phosphorylated R domain.", NATURE (LONDON), vol. 389, no. 6648, 1997, pages 294 - 296, XP002142115, ISSN: 0028-0836 *

Also Published As

Publication number Publication date
AU3241900A (en) 2000-09-14

Similar Documents

Publication Publication Date Title
Girard et al. p11, an annexin II subunit, an auxiliary protein associated with the background K+ channel, TASK-1
Paschal et al. Identification of NTF2, a cytosolic factor for nuclear import that interacts with nuclear pore complex protein p62.
Teem et al. Identification of revertants for the cystic fibrosis ΔF508 mutation using STE6-CFTR chimeras in yeast
US5240846A (en) Gene therapy vector for cystic fibrosis
WO2013039996A1 (en) Compositions and methods for brown fat induction and activity using fndc5
WO2002064790A2 (en) Jfy1 protein induces rapid apoptosis
MX2007002379A (en) Systems and methods for protein production.
US20030100501A1 (en) Q4N2NEG2 enhances CFTR activity
AU2017212629A1 (en) Wnt compositions and methods for serum-free synthesis
US6770739B1 (en) Enhancers of CFTR chloride channel function
EP1243595B1 (en) Bh4-fused polypeptides
WO2000050591A1 (en) Enhancers of cftr chloride channel function
US6468793B1 (en) CFTR genes and proteins for cystic fibrosis gene therapy
Johnston et al. Harp (harmonin‐interacting, ankyrin repeat‐containing protein), a novel protein that interacts with harmonin in epithelial tissues
JP2003523723A (en) Hermansky-Padrack syndrome protein-interacting proteins and methods of use
CA2407945A1 (en) Molecules that modulate ubiquitin-dependent proteolysis and methods for identifying same
JP2004502416A (en) Inhibitory PAS domain protein (IPAS) associated with angiogenesis and tumor progression and screening method
AU750395B2 (en) A method for selectively controlling membrane protein display and protein secretion in eukaryotic cells
JP2000135090A (en) HUMAN H37 PROTEIN, AND cDAN CODING FOR THE SAME
WO2002050104A9 (en) Methods and cells for detecting modulators of rgs proteins
US6790949B1 (en) Protein Rim2
JP2001161363A (en) Novel endoplasmic localization protein, agent for curing abnormal glycoprotein disease containing the same, dna coding for the protein and gene curing agent for curing abnormal glycoprotein disease containing the same
WO2007041360A2 (en) Peptides and calcium regulation in mammalian cells
JP2001054391A (en) Endoplasmic reticulum stress transcription factor
DeGiosio et al. Differential regulation of MAP2 by phosphorylation events in proline‐rich versus C‐terminal domains

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 09914213

Country of ref document: US

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase