Departments of 1 Pediatrics and 2 Physiology and Biophysics, Case Western Reserve University School of Medicine, and Rainbow Center for Childhood PKD, Rainbow Babies and Children's Hospital, University Hospitals of Cleveland, Research Institute, Cleveland, Ohio 44106-6003
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Developmental control of cell
proliferation is crucial, and abnormal principal cell proliferation may
contribute to cystogenesis in polycystic kidney disease. This study
investigates roles of cAMP and its primary effector, cAMP-dependent
protein kinase (protein kinase A; PKA), in control of cell
proliferation in filter-grown noncystic (NC) and cystic (CY)-derived
principal cell cultures. These cultures had similar cAMP pathway
characteristics upstream of PKA subunit distribution but differed in
predicted PKA subtype distribution. Functionally, cultures were
proliferative before polarization, with constitutively higher
proliferation in CY cultures. NC cultures achieved levels similar to
those of CY cultures on pharmacological manipulation of cAMP production
or PKA activation or inhibition of PKA subtype I activity. Inhibition
of overall PKA activity, or of PKA subtype II anchoring, diminished
cAMP/PKA-mediated proliferation in NC cultures but had no effect on CY
cultures. Polarized CY monolayers remained proliferative, but NC
monolayers lost responsiveness. No large proliferation changes resulted
from treatments of polarized cultures; however, polarized NC and CY cultures differed in poststimulation handling of PKA catalytic and type
II regulatory subunits. Our results support PKA subtype regulation
of prepolarization proliferation in NC principal cells and altered
regulation of PKA in CY cells and suggest that differences at or
downstream of PKA can contribute to altered proliferation in a
developmental renal disease.
epidermal growth factor receptor; polycystic kidney disease; protein kinase subtypes; immunolocalization
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
CELL PROLIFERATION is a normal physiological component of development. In the kidney increased proliferation can result in cancers (reviewed in Ref. 40) or cysts (reviewed in Ref. 39), whereas decreased proliferation can result in loss or diminished size of renal structures (2). Study of disease models with abnormal cell proliferation can help to define specific regulatory mechanisms by identifying differences from normal. The intracellular signaling molecule cAMP has been implicated in regulation of proliferation and differentiation in many cell types (reviewed in Ref. 64). Effects are either mediated by cAMP-dependent protein kinase (protein kinase A, PKA; Refs. 4, 12, 15, 26, 44) or are independent of PKA and involve small guanine nucleotide binding (G) proteins (46, 56, 67). Both positive and negative modulation has been reported, which appeared to depend on cell type, culture conditions, or the presence of other stimuli.
In addition to roles in regulating renal fluid and ion transport, cAMP and/or PKA modulate proliferation in cell cultures derived from throughout the kidney: human proximal tubular cells (24, 28); proximal tubular epithelial cell lines MCT (71), LLC-PK1 (5, 71), and PKSV-PCT (68); glomerular mesangial cells (7, 33); Madin-Darby canine kidney (MDCK) cell lines (21, 24, 72, 73); immortalized human kidney epithelial cell line IHKE-1 (3); human embryonic kidney cell line HEK293 (56); and renal cancer cells (8). Because cAMP can induce proliferation and formation of renal epithelial cysts in vitro (21), and abnormally increased principal cell proliferation is a factor in collecting duct cyst formation and expansion in a developmental renal disease, autosomal recessive polycystic kidney disease (ARPKD) (39), cAMP could play a role in cell proliferation in normal developing principal cell epithelium. Earlier ureteric bud and collecting duct proliferation is regulated by bone morphogenetic protein (BMP)7 (47), but a later developmental switch to collecting duct cyst formation and progressive expansion occurs in ARPKD (39).
The inherited polycystic kidney diseases (PKDs) autosomal dominant PKD (ADPKD) and ARPKD make up the most common life-threatening genetic disease family (reviewed in Ref. 49). The family is characterized by progressive fluid accumulation in renal tubules to form cysts that eventually lead to renal failure. Genes responsible for ADPKD1 and ADPKD2 subtypes, and a gene associated with murine ARPKD in orpk mice, were identified (reviewed in Refs. 35, 36, and 69). Their gene products (polycystin-1, polycystin-2, and polaris) may form a membrane-associated signaling complex involved in nephrogenesis, and mutations may contribute to PKD by altering the complex's function (reviewed in Refs. 18 and 35). Positional cloning studies identified chromosomal loci linked to ARPKD and several murine ARPKD models (reviewed in Refs. 39 and 40), but specific genes for these PKDs have not been found. All known PKD genes and genetic loci are on distinct chromosomes, and cellular mechanisms for initiation and progression of cystic disease are still unclear.
All PKDs have a consistent molecular phenotype of abnormal overexpression of epidermal growth factor (EGF) receptor (EGFR) on apical surfaces of cystic collecting tubular (CT) principal cell epithelium, whereas noncystic receptor expression is basolateral (reviewed in Ref. 61). EGFR activation promotes increased CT mitogenesis, cyst formation, and progressive cystic enlargement that is inhibited by blocking EGF-activated EGFR tyrosine kinase activity (48, 53, 61, 63). In addition to characteristic altered fluid transport and abnormal extracellular matrix, EGFR-mediated cell proliferation is very likely a mechanism for cyst formation and expansion in PKD (reviewed in Refs. 39 and 60). However, because the EGFR phenotype occurs downstream from several different primary gene mutations, other signal transduction systems present in principal cells could also modulate abnormal cell proliferation.
In classic cAMP signaling, a receptor family exerts its effects by activating an intracellular signaling cascade of heterotrimeric G proteins, adenylyl cyclase (AC), cAMP synthesis, and cAMP-dependent protein kinase (PKA) activation. The PKA is a tetrameric holoenzyme of two cAMP-binding regulatory (R) and two catalytic (C) subunits. Holoenzymes with different pairs of R subunit isoforms (RI or RII) are classified as type I and type II PKA subtypes, respectively (reviewed in Ref. 19). Activation is accomplished when cAMP binding to R occurs (reviewed in Refs. 19 and 23). Activated PKA is believed to move throughout the cell as free C subunits, although holoenzyme may be capable of activation without dissociation (76). Activation mediates serine or threonine phosphorylation, an effect reversible by specific protein phosphatases. Phosphodiesterases (PDEs) degrade cAMP (14) and promote the return of PKA to its inactive, tetrameric state. Any C subunits sent to the nucleus on activation are inhibited and shuttled out by the endogenous PKA-specific protein kinase inhibitor (PKI) (reviewed in Ref. 19). Subcellular distribution of PKA holoenzyme is mediated partly by A kinase anchoring proteins (AKAPs) (Refs. 6 and 75; reviewed in Ref. 11) that bind R subunits and allow assembly of signaling complexes (57, 70). Therefore, R binding to AKAPs may regulate subtype activity distribution. Isoforms of each cAMP pathway component have been identified, and their diverse expression patterns probably contribute to the range of cellular functions credited to cAMP (reviewed in Refs. 19 and 23).
Exactly how these isoforms act to regulate cellular function is a
central question in cAMP research. A physiological model for
investigating the role of PKA subtypes in cell proliferation can be
found in the abnormal cell proliferation of cancer. Predominant type II
activity is found in normal, nonproliferating tissues and in
growth-arrested cells, whereas type I activity predominates in tumor
cells (reviewed in Ref. 9). Human mammary epithelial cells
grow in serum-free medium when they overexpress transfected RI
(66). Specific inhibition of type I activity by cAMP
analogs 8-chloro-cAMP (8-ClcAMP) (38, 54) and the Rp
diastereomer of adenosine 3',5'-cyclic monophosphothioate (Rp-cAMPS;
Ref. 20) inhibits growth in human cancer cell lines, and
EGFR and type I PKA inhibition cooperatively limits proliferation
(reviewed in Ref. 9). Therefore, coordinate cAMP/EGFR
regulation of cancer cell proliferation may occur. PKD is another
physiological model in which coordinate cAMP/EGFR regulation of
abnormal cell proliferation may occur. Mechanistic similarities between
cyst formation and the development of cancerous tumors could exist,
because increased CT cell proliferation and altered extracellular
matrix properties might be expected to result in tumors in the absence
of the altered fluid transport required for cyst formation.
cAMP pathway regulation of epithelial cyst formation was demonstrated
in three-dimensional collagen gel culture, where MDCK cells form
spherical cysts and enlarge after stimulation with cAMP analogs
(21, 30, 31, 34). MDCK cell clones with comparable capacities for cAMP production despite differences in responsiveness to
cAMP agonists have unique distributions of RI and RII
subunits that probably reflect PKA anchoring differences. This is likely to
allow PKA subtype regulation of the clones' distinct tubule and cyst
morphologies (41, 42).
Does cAMP regulate proliferation in normal and ARPKD principal cells? Because the cellular balance of PKA type I and type II activities, their subcellular locations, their preferred targets, and their ultimate functional effects may vary depending on the cell's unique complement of R and C subunits, the present study was undertaken to characterize cAMP signaling and PKA expression in normal murine principal cells and in cells derived from a murine model of ARPKD.
![]() |
EXPERIMENTAL PROCEDURES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Cell isolation and culture. An animal model of ARPKD, the C57BL/6J(cpk/cpk) (cpk) mouse (55), arose from a spontaneous mutation in the C57BL/6J(C57+/C57+) (C57) strain. The murine disease phenotype and its progression have been described extensively, and it has been shown to mimic the stages and tubular profile of the human disease ARPKD (reviewed in Refs. 1 and 60). As previously described (43), cystic cpk mice were killed to obtain kidney tissue at postnatal days 8-14. Collecting tubular epithelial cells were isolated from cystic kidneys with a selective enrichment method that resulted in an isolated cell population comprised predominantly of principal cells (60). The enriched population of principal cells was cultured and passaged six times without loss of the apical EGFR phenotype. Control, noncystic cells were obtained in the same way from C57 kidneys at corresponding ages.
Cell cultures were maintained as previously described (60). Stock cultures were seeded on 10-cm plastic culture plates (Corning Costar, Cambridge, MA) at ~1.2 × 104 cells/cm2 and were passaged every 7 days. For experiments, cells were seeded at 1.2 × 104 cells/cm2 on 0.4-µm pore Transwell-Clear filter inserts (no. 3460, Corning Costar) unless otherwise noted. In defined growth medium (60), cells formed confluent monolayers by 1-2 wk. Conditions for preparing subconfluent and confluent filter-grown cultures were standardized by consistent seeding and growth conditions, followed by visual inspection with a standard phase-contrast microscope and correlation with ZO-1 immunostaining to determine when polarized monolayers were established (see Fig. 1). The physiological tightness of the confluent monolayers under these growth conditions was established previously by measuring 3H-labeled inulin leakage (60). Medium changes for experiments are described individually.
|
Immunological reagents.
Primary antibodies used were anti-PKA-catalytic subunit- (sc-903), a
rabbit IgG against human C
(Santa Cruz Biotechnology, Santa Cruz,
CA); anti-PKA-regulatory subunit type I (P19920), a mouse IgG against
mouse RI (Transduction Laboratories, Lexington, KY);
anti-PKA-regulatory subunit type II
(sc-909), a rabbit IgG against
mouse RII
(Santa Cruz Biotechnology); anti-EGFR (sc-003), a rabbit
IgG against human EGFR (Santa Cruz Biotechnology); and anti-ZO-1
(61-7300), a rabbit IgG against human ZO-1 (Zymed Laboratories, South San Francisco, CA). The fluorescence-tagged secondary antibodies used were Oregon green 488 goat anti-mouse IgG conjugate (O-6380, Molecular Probes, Eugene, OR), Oregon green 488 goat anti-rabbit IgG
conjugate (O-6381, Molecular Probes), Texas red-X goat anti-mouse IgG
conjugate (T-6390, Molecular Probes), and Texas red-X goat anti-rabbit
IgG conjugate (T-6391, Molecular Probes). Peroxidase-linked secondary
antibodies used were goat IgG against rabbit IgG (sc-2004, Santa Cruz
Biotechnology), goat IgG against mouse IgG (sc-2005, Santa Cruz
Biotechnology), and mouse IgG against biotin (200-032-096, Jackson
ImmunoResearch Laboratories, West Grove, PA).
Western blot analysis of protein expression.
To characterize protein expression in cultured cells, samples grown on
filter inserts were harvested and 10 µg of protein (unless otherwise
noted in individual figures) from each sample was subjected to
SDS-PAGE, immunoblotting, and signal detection as previously described
(41). Biotinylated ECL protein molecular weight markers
(RPN2107; Amersham Pharmacia Biotech, Piscataway, NJ) were included in
each gel and were detected by immunoblotting with anti-biotin
200-032-096 at 1:40,000. Primary antibodies used were anti-C sc-903
at 1:2000, anti-RI P19920 at 1:25, anti-RII
sc-909 at
1:500, and anti-EGFR sc-003 at 1:500. Peroxidase-linked secondary
antibodies were used at 1:500-2,000 dilutions. Specific primary
and secondary antibody combinations are indicated in Figs. 1, 4, 8, and
9.
cAMP-dependent protein kinase assays. Cells were seeded onto 24-mm diameter Transwell-Clear filter inserts (Corning Costar) and maintained in culture as described in Cell isolation and culture. Samples were prepared (10) for assay, and total cAMP-dependent protein kinase activity toward the specific substrate kemptide was measured as previously described (41). Sample activity was compared with a standard curve generated with 0.0063-0.1 units of purified bovine heart PKA C subunit (no. 29538; Pierce, Rockford, IL). A unit is equivalent to 1 picomole of phosphate transferred to substrate per minute. Values were normalized per milligram of protein assayed. In the present study, activity in the presence of cAMP plus the specific PKA inhibitor peptide PKA inhibitor 6-22 amide (PKI 6-22, 100 nM; no. 539684, Calbiochem-Novabiochem, La Jolla, CA), or in the absence of cAMP, was similar (data not shown).
Localization studies. Confocal laser scanning microscopy was used to localize specific proteins identified by immunofluorescence. Cells were seeded onto 12-mm diameter Transwell-Clear filter inserts and maintained in culture as described in Cell isolation and culture. For monolayers treated before fixation, growth medium was removed, the confluent cell monolayers were washed three times with phosphate-buffered saline (PBS; 10 mM NaPO4, 150 mM NaCl, pH 6.9) and cells were incubated at 37°C in defined medium containing additions indicated in Fig. 6. All subsequent steps were performed as previously described (41). Specific primary and secondary antibody combinations used are listed in Figs. 1, 5, and 6. The specificity of antibody immunoreactivity was demonstrated with controls lacking primary antibodies, which in all cases resulted in no detectable fluorescent signal (data not shown). Actin labeling was performed after immunofluorescent labeling was completed with an Oregon green 514-phalloidin conjugate (O-7465; Molecular Probes) according to the manufacturer's protocol. All samples were examined with a scanning laser confocal microscope, by using a 100× objective and software to optically section the cell monolayer every 0.5 µm, as previously described (41).
Intracellular cAMP determinations.
Cells were seeded onto 12-mm-diameter Transwell-Clear filter inserts
(Corning Costar) and maintained in culture as described in Cell
isolation and culture. Growth medium was removed, and confluent
cell monolayers were washed three times with PBS. Defined medium with
additions indicated in Fig. 2 was added, and cultures were replaced in
the tissue culture incubator for 1 h. Monolayers were then
assayed for cAMP as previously described (41). Values were
normalized for protein content. Fold changes in accumulation were
calculated as follows: [(stimulated) (control)]/ [(IBMX + rolipram)
(control)].
|
Cell proliferation assays.
The proportion of proliferating cells in cell cultures was determined
by seeding cells onto Transwell-Clear filter inserts, as described in
Cell isolation and culture. Cultures were expanded to the desired confluence (subconfluent cultures at 20,000
cells/insert, 100% confluence at
100,000 cells/insert) and then were
treated with agents added to standard growth medium for an additional 16 h as described in Figs. 7-9. Proliferation was
assayed using the 5-bromo-2'-deoxyuridine (BrdU) labeling and detection
kit (no. 1-296-736; Roche Molecular Biochemicals) according
to the manufacturer's protocol. Briefly, immunofluorescent fluorescein labeling was used to detect BrdU incorporated into cellular DNA. All
samples were then stained with 500 nM propidium iodide (Molecular Probes) in PBS for 5 min to detect nuclei according to the
manufacturer's protocol. Filter inserts were mounted on standard
microscope slides as described in Localization studies.
Samples were viewed with an Optiphot-2 fluorescence microscope (Nikon,
Japan) with FITC HQ and Texas red HQ filter sets (Chroma Technologies,
Brattleboro, VT), and three rectangular fields (270 × 210 µm)
in each sample were photographed with a SPOT digital color camera
(Diagnostic Instruments, Sterling Heights, MI). Semitransparent layered
images in Adobe Photoshop 5.5 software (Adobe Systems, San Jose, CA) were scored for the number of nuclei showing BrdU immunostaining and
the total number of nuclei observed (propidium iodide staining). The
ratio of these values was used to reflect BrdU incorporation into
cells. Higher values reflected increased levels of BrdU uptake and
incorporation into cellular DNA, an indication of relatively greater
levels of cell proliferation. In a tightly packed confluent monolayer,
the number (mean ± SE; n = 4) of propidium
iodide-stained nuclei per well was equal to the number of trypan
blue-excluding live cells removed by trypsinization (567,000 ± 42,438 vs. 583,875 ± 48,549, respectively). If cultures were
allowed to overgrow, more cells were removed by trypsinization than
were scored visually. However, because cysts enlarge progressively,
producing and maintaining an epithelial monolayer without overgrowth,
cultures were used and scored only if cells or monolayers were present
without overgrowth.
Other information. Statistical determinations were calculated with InStat3 v3.05 software (GraphPad Software, San Diego, CA). Protein was quantitated with either Bio-Rad (Bio-Rad Laboratories, Hercules, CA) or bicinchoninic acid (Pierce, Rockford, IL) protein assay reagents, according to the manufacturers' protocols. The EGFR tyrosine kinase inhibitor EKI-785 (13, 61) was obtained from Dr. Philip Frost (Wyeth-Ayerst Research, Pearl River, NY) for the Rainbow Center for Childhood PKD. The AKAP binding inhibitor InCELLect AKAP St-Ht31 inhibitor peptide was obtained from Promega (Madison, WI). Reagents not specifically listed were either cell culture or molecular biology grade and were obtained from Sigma (St. Louis, MO), Life Technologies, and Calbiochem-Novachem.
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
cAMP accumulation was normal in cultured cells that expressed the cystic phenotype. Selectively enriched populations of isolated CT principal cells used in this study maintained characteristic phenotypes associated with either PKD cysts or unaffected collecting tubules (43, 60). Apical and basolateral EGFR immunostaining was observed in filter-grown cystic cpk cell cultures (Fig. 1). Only lateral EGFR immunostaining was visible in noncystic C57 cells (Fig. 1). Unlike the overexpression observed in tissue samples harvested from murine cystic kidneys (43), EGFR protein expression was similar in cell cultures grown in the absence of EGF (Fig. 1). Immunostaining for the tight junction protein ZO-1 was identical for both culture types (Fig. 1). Therefore, both isolated cystic and noncystic cells formed epithelial layers in filter culture, and phenotypic differences were maintained.
To determine whether the cystic phenotype included alterations in the initial cAMP signaling step, intracellular cAMP levels in cystic cpk and noncystic C57 CT cells were measured. Basal levels of intracellular cAMP were similar in the two cell types (Fig. 2). Similar low levels of cAMP accumulated in the two cell types in response to the PDE inhibitors IBMX and rolipram. The two inhibitors likely inhibit the majority of PDE isoform activity present in these cells (59). To maximize the likelihood that cAMP-mediated responses would be detected and to allow direct comparisons between treatments, PDE inhibitors were included for all subsequent treatment conditions and experiments unless specifically noted. In response to EGF (in the presence of PDE inhibitors, as noted), no intracellular cAMP accumulation occurred in noncystic cells and no consistent accumulation was observed in cystic cell cultures. Therefore, EGF, which was required to maintain cysts in murine metanephric organ culture (48, 61), did not promote increased cAMP production. Forskolin, a direct activator of AC, induced similar large increases in cAMP accumulation in both C57 and cpk cells, although the absolute amount of cAMP present in cpk cells was smaller. Therefore, the cell types had similar, normal capabilities for cAMP production.Subcellular distribution of PKA subunits differed between noncystic
and cystic cell cultures.
Because the primary effector of cAMP action is PKA, protein kinase
activity was measured in confluent cultures of each cell type to
determine whether both cystic and noncystic cell cultures expressed
similar overall levels of PKA activity. Total cAMP-dependent protein
kinase activity levels were similar in both cell types (Fig.
3), but this assay does not distinguish
between type I and type II subtypes. cAMP-independent basal activity
and activity in the presence of cAMP plus PKI 6-22 were similar
(data not shown).
|
|
|
|
Regulation of proliferation differed between noncystic and cystic cell cultures. Progressive enlargement of CT cysts during the disease course is a hallmark of ARPKD and murine models for it and is postulated to be due partly to increased cell proliferation, although expanding cysts maintain a single layer lining of cells (60). Proliferation rates are low in normal collecting tubules, so why proliferation persists in cystic collecting tubules is a central question (37). In organ culture, EGF stimulated cystic kidney cell proliferation and was required for maintenance of cysts (48, 60, 61), but the organ culture model includes many cell types other than the principal cells that express the characteristic cystic phenotype of apical EGFR expression. The mechanism responsible for apical targeting of the EGFR in PKD is unknown. Because it is unlikely that new principal cells appear fully epithelialized, any new cells produced in the normal tubule or in a cyst as the result of proliferation are likely to transition from unpolarized to polarized and to acquire the epithelial phenotype as they develop. Therefore, subconfluent cultures were postulated to model regions of growing collecting tubule before establishment of a polarized epithelium, whereas confluent cultures were shown to express a polarized phenotype (Fig. 1).
To determine whether proliferation in the cell cultures reflected proliferation observed in the developing PKD kidney (48), cell proliferation was measured in both subconfluent and confluent cultures of CT cells. Control, or basal, levels of cell proliferation were higher in subconfluent cpk cell cultures than in C57 cell cultures (Table 1; Fig. 7A). Relative proliferation leveled off to minimum detectable values as C57 cultures reached confluence. Cystic cultures maintained high relative proliferation as confluence increased, with control proliferation levels similar to those for subconfluent cultures. To determine whether cAMP and PKA activity regulated proliferation in principal cells, effects of the cAMP analog 8-bromo-cAMP (8-BrcAMP) in the absence or presence of the PKA activity inhibitor H-89 were tested in the proliferation assay. As shown, 8-BrcAMP stimulated proliferation in subconfluent C57 cultures and H-89 blocked 84% of the 8-BrcAMP-induced increase (Table 1). This overall PKA activity inhibitor had no significant effect on control levels of proliferation in any culture condition.
|
|
Pharmacological stimulation of cystic and noncystic cultures
resulted in differential protein expression changes.
The proliferation assay results (Fig. 7, B and C)
suggest that PKA subtype differences exist between cell types and
confluence states, but there were no major differences in expression of
any PKA subunits when subunit expression was compared in subconfluent and confluent (Fig. 4) cultures and overall PKA activity was the same
(Fig. 3). However, when confluent cultures were treated to determine
whether stimulation affected expression of specific subunits, an
expression difference was found between cystic and noncystic cells
(Fig. 8). Stimuli that elevated
intracellular cAMP levels (PDE inhibitors, with and without forskolin)
or were expected to activate EGFR (EGF in the presence of PDE
inhibitors) resulted in either no changes or very modest changes in
C or RI expression. In C57 cells, a shift in RII
expression from
an apparent molecular weight of 51,000 to 54,000 was observed only in
the presence of forskolin. The same stimuli resulted in the appearance of an immunoreactive RII
doublet in cpk
cultures whenever PDE inhibitors were present. Therefore, only a large
increase in intracellular cAMP (Fig. 2) produced an RII
shift in C57
cells, whereas any increase over control (Fig. 2) resulted in the
appearance of the doublet pattern in cpk cells. Unlike RI,
which contains a pseudosubstrate motif lacking a PKA-phosphorylatable
serine residue, RII may be autophosphorylated in its pseudosubstrate region. The apparent molecular weight shift observed (Fig. 8) suggests
endogenous autophosphorylation of the RII
subunit by C (reviewed in
Ref. 25).
|
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
The results presented in this study suggest that an appropriate
balance of PKA distribution and activity is necessary for normal
proliferation in principal cells. The results demonstrate that PKA type
I activity limits proliferation to low, basal levels in normal
principal cells before polarization. The results also demonstrate that
cAMP-stimulated proliferation, observed only in noncystic, expanding
cell cultures, is mediated by PKA activity and AKAP anchoring. On
reaching confluence, normal cells proliferate only at very low levels
and proliferation is unresponsive to agonists or inhibitors. In
contrast, both unpolarized and polarized cystic principal cells are
highly proliferative and also mostly unresponsive to agonists and
inhibitors, despite normal cAMP accumulation, PKA activity, and subunit
expression. Striking differences are observed in the subcellular
distribution of both RI and RII PKA subunits and in the distinctive
redistribution of catalytic subunit and altered expression of RII
subunit in response to cAMP-elevating agents. In summary, these studies
suggest that differences in the predicted location of both PKA type I
and type II activities result in loss of normal PKA-regulated control
of proliferation so that nearly constitutive proliferation is
observed in cystic principal cells (Fig.
10).
|
Proliferative activity of cyst epithelium in human cystic diseases was evaluated in a previous report. Approximately 32- and 8-fold greater proliferative indexes were measured in ARPKD and ADPKD epithelia, respectively, compared with normal kidney (37). The results of the present study suggest that cultured cpk principal cells maintain a proliferative phenotype similar to that observed in the cystic epithelium of human ARPKD. The double enrichment method used is selective for principal cells and was used with kidneys from animals at early stage to midstage of the disease, when CT cysts are forming and expanding rapidly. These results are in contrast to the low proliferation levels reported for Dolichos biflorus lectin-expressing collecting duct cells derived from the same animal model (52). However, the two culture systems were dissimilar because both collecting duct principal and intercalated cells express the lectin and lectin-expressing cells were obtained from 21-day-old animals at the end stage of the disease, near the end of the animals' life span.
There are few data in the literature about PKA subtype activation in PKD. In the pcy mouse model of ARPKD, kidney cAMP content increased over the disease time course whereas cAMP content in normal murine kidney remained constant, and a lipid found in pcy cyst fluid stimulated cellular cAMP production, fluid transport, and cell proliferation in renal cell lines (73). In two previous reports, activation of the cAMP pathway regulated cell proliferation in ADPKD cyst cells (cultured on plastic or on collagen) but not in human kidney cortex cells (22, 74). In the former report, cAMP-mediated proliferation occurred by activation of the extracellular signal-regulated kinase (ERK)1/2 kinase pathway at a step distinct from that stimulated by EGFR activation (74). The type I PKA inhibitor used in that study was as inhibitory as the general PKA inhibitor H-89, suggesting that cAMP-induced proliferation through the ERK pathway required type I PKA activity. The latter report suggested that effects of EGF and cAMP on ADPKD cell proliferation were independent (22). In contrast to the ADPKD studies, the results of the present study demonstrate a role for PKA-mediated proliferation in normal murine principal cells and suggest that high levels of cell proliferation in murine ARPKD cells assayed on permeable filter supports could result from a shift in the balance and distribution of PKA subtype activities, that the shift may contribute to altered EGFR activity, and that differences exist even before a polarized epithelium is firmly established. The ability of H-89 to inhibit all but 16% of the cAMP-stimulated effect on proliferation in noncystic cultures suggests that normal principal cells have a small PKA-independent mechanism regulating cAMP effects on proliferation. Perhaps this becomes a major factor in murine ARPKD cells in the absence of normal PKA distribution.
Although individual subtype activities were not measured in the present
study, the functional effects of the two subtypes were addressed
pharmacologically. Type I PKA was responsible for maintaining basal
proliferation at low levels in expanding cultures of noncystic cells
and was diffusely distributed within the cell in polarized cultures. In
contrast, perhaps the distinct apical distribution of type I in cystic
cells prevented its inhibition and contributed to the cell's
constitutive proliferation levels. Binding of RI to the Grb2 protein
recruited to autophosphorylated EGFR in cancer cells has been reported
(65), and inhibition of EGFR and PKA cooperatively
inhibits cancer cell growth (reviewed in Ref. 9). If a
similar interaction occurred between apical EGFR and apical RI
in
PKD cells, this might contribute to the persistent proliferation
observed, although this is pure speculation.
Type II PKA anchoring to AKAPs was required for forskolin-mediated
proliferation in subconfluent, noncystic cells. In contrast, in
expanding and confluent cystic cell cultures, normal anchoring appeared
to be absent or dysfunctional. With the RII distribution differences
in unstimulated confluent cells, and the prolonged redistribution of
C
in stimulated cpk cells, these results support the
hypothesis that cystic cells have a defect in PKA anchoring via AKAPs
or redistribution mechanisms via PKI or slightly elevated RII
and
C
expression. Some AKAPs are associated with the cytoskeleton via
actin, and phalloidin labeling demonstrated that although both C57 and
cpk cells had similar patterns of lateral actin staining, only C57 cells showed basal stress fiber staining (Fig. 1). Basal actin
in the cpk cells appeared to be disorganized (Fig. 1),
supporting the likelihood of anchoring defects. However, use of the
anchoring inhibitor does not address possible type II activity
differences. Evidence that type II activity is altered in
cpk is provided by the observation that immunoreactive
RII
bands of higher apparent molecular weight appeared in a distinct
pattern on cAMP stimulation of the two cell types. Such shifts have
been accounted for by RII
"autophosphorylation" by C, a
modification that can result in the appearance of additional bands of
higher apparent molecular weight in polyacrylamide gels, and have been
linked to cell cycle differences, cell proliferation, apical endocytic
recycling, and altered subcellular location of RII
(Refs.
17, 32, and 58; reviewed in Ref.
25). Autophosphorylation in vitro shifted RII
from
51,000 to 54,000, decreased the affinity between RII
and C
(51), and promoted a more efficient association with AKAPs (77, 78). If the presence of apparent molecular weight
differences in CT cell RII
observed in the present study reflects
RII
autophosphorylation, then these data suggest that Type II PKA
activity, or phosphorylation/dephosphorylation mechanisms, differed
between confluent C57 and cpk cultures.
Because EGF and EGFR were previously observed to play roles in ARPKD proliferation and cystogenesis (reviewed in Refs. 39 and 60), and because EGFR and PKA type I appear to regulate proliferation coordinately in cancer (reviewed in Ref. 9), EGF was included in the present study. In contrast to tissue and organ culture models, cultured cystic principal cells were constitutively proliferative rather than hyperresponsive to EGF. The cell culture system lacks normal organotypic architecture and other cell types that may contribute endogenous growth controls. However, the characteristic EGFR phenotype was maintained. Principal cells are the affected cells that line cysts, so regardless of the mechanism, EGF may activate apical EGFR and provide activated receptors access to cellular molecules with which they might not normally communicate. This concept is supported by the clear difference seen in ligand-induced loss of EGFR protein expression in cystic compared with noncystic cells. Although membrane-associated proteins other than the EGFR were not found to be clearly mislocalized in PKD (reviewed in Ref. 61), the observation that basal actin appeared disorganized in the cystic cpk cells suggests that cytoskeletal anchoring or signal transduction mechanisms may differ in cystic compared with noncystic cells.
The EGFR autophosphorylation/tyrosine kinase activity inhibitor was included because an effect on EGF-mediated proliferation was expected, the inhibitor blocked cyst progression in vivo, and it has been considered as a potential therapeutic agent (49, 61). Unexpectedly, the experimental results suggested a role for PKA regulation of the EGFR. The observation that the inhibitor blocked the proliferative effects of both EGF and forskolin in noncystic cells suggested that cAMP/PKA regulated EGFR expression or activity to regulate proliferation and that this was a normal CT cell process. However, elevation of intracellular cAMP did not increase equilibrium EGFR protein expression, so EKI-785 was unlikely to be acting on cAMP-induced EGFRs. Possible PDE inhibitor, or PKA, stimulation of EGFR activity was not measured in the present study. Because the inhibitor has some effects on c-erbB-2 tyrosine autophosphorylation (13), questions remain about the mechanism of action of EKI-785. The inhibitor was reported to have greater efficacy at early stages of cystic disease (62, 63), perhaps corresponding to its effect on the subconfluent cystic cultures, as a potential model for the growing collecting duct, and supporting the idea that there is a therapeutic "window of opportunity," because confluent cultures were unaffected. If cellular PKA inhibition by EKI-785 did occur, this result would confirm PKA regulation of proliferation in these cells. Perhaps EKI-785 was effective at inhibiting PKD cystogenesis because of its combined effects on activated EGFR and PKA.
The ability of normal principal cells to cease proliferation appropriately after reaching confluence although cystic cells that express and handle PKA differently do not suggests that appropriate PKA action is crucial for normal principal cell proliferation. Type I PKA may maintain low basal proliferation levels, whereas AKAP anchoring may mediate proliferation in response to elevated cAMP. In cystic principal cells, perhaps unusual coupling of type I activity could mediate altered proliferation and variable type II anchoring or activation could mediate altered endocytic recycling or activation of the EGFR, making PKA abnormalities an upstream step in the process of PKD pathogenesis. Preferential access of PKA subtypes to polarized signaling complexes or substrates could regulate specific functions or phenotypes. For example, polycystin-1, the transmembrane protein product of the PKD1 gene, was a PKA substrate in vitro (29, 45), although the functional significance of this finding is unknown. If a PKD protein complex regulates normal nephrogenesis, then mutations that alter its regulation by PKA subtypes could contribute to cyst formation. Murine ARPKD may be an example of a disease in which cAMP pathway variations have serious physiological consequences. This study is the first demonstration of PKA subunit localization in PKD. Further study is required to identify and pinpoint direct subtype links to PKA substrates and to demonstrate phosphorylation-specific regulation of proliferation in normal cells as well as proliferation and cystogenesis in cystic disease.
![]() |
ACKNOWLEDGEMENTS |
---|
This work was supported by National Institute of Diabetes and Digestive and Kidney Diseases Grants DK-50707 and DK-57306 (in part), by the PKD Foundation, and by the Department of Pediatrics of Rainbow Babies and Children's Hospital of the University Hospitals of Cleveland.
![]() |
FOOTNOTES |
---|
Present address of C. Marfella-Scivittaro: Dept. of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106.
Address for reprint requests and other correspondence: S. A. Orellana, 11100 Euclid Ave., Mail Stop 6003, Cleveland, OH 44106-6003 (E-mail: sao3{at}po.cwru.edu).
The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
10.1152/ajpcell.00122.2001
Received 8 March 2001; accepted in final form 6 November 2001.
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
1.
Avner, ED.
Renal developmental diseases.
Semin Nephrol
13:
427-435,
1993[ISI][Medline].
2.
Bates, CM,
Kharzai S,
Erwin T,
Rossant J,
and
Parada LF.
Role of N-myc in the developing mouse kidney.
Dev Biol
222:
317-325,
2000[ISI][Medline].
3.
Benesic, A,
Mildenberger S,
and
Gekle M.
Nephritogenic ochratoxin A interferes with hormonal signalling in immortalized human kidney epithelial cells.
Pflügers Arch
439:
278-287,
2000[ISI][Medline].
4.
Braun, CM,
Huang SK,
Kagey-Sobotka A,
Lichtenstein LM,
and
Essayan DM.
Co-regulation of antigen-specific T lymphocyte responses by type I and type II cyclic AMP-dependent protein kinases (cAK).
Biochem Pharmacol
56:
871-879,
1998[ISI][Medline].
5.
Bringhurst, FR,
Juppner H,
Guo J,
Urena P,
Potts JT, Jr,
Kronenberg HM,
Abou-Samra AB,
and
Segre GV.
Cloned, stably expressed parathyroid hormone (PTH)/PTH-related peptide receptors activate multiple messenger signals and biological responses in LLC-PK1 kidney cells.
Endocrinology
132:
2090-2098,
1993[Abstract].
6.
Carr, DW,
Cutler RE,
Cottom JE,
Salvador LM,
Fraser ID,
Scott JD,
and
Hunzicker-Dunn M.
Identification of cAMP-dependent protein kinase holoenzymes in preantral- and preovulatory-follicle-enriched ovaries, and their association with A-kinase-anchoring proteins.
Biochem J
344:
613-623,
1999[ISI][Medline].
7.
Chini, CC,
Grande JP,
Chini EN,
and
Dousa TP.
Compartmentalization of cAMP signaling in mesangial cells by phosphodiesterase isozymes PDE3 and PDE4. Regulation of superoxidation and mitogenesis.
J Biol Chem
272:
9854-9859,
1997
8.
Ciardiello, F,
Caputo R,
Bianco R,
Damiano V,
Pomatico G,
Pepe S,
Bianco AR,
Agrawal S,
Mendelsohn J,
and
Tortora G.
Cooperative inhibition of renal cancer growth by anti-epidermal growth factor receptor antibody and protein kinase A antisense oligonucleotide.
J Natl Cancer Inst
90:
1087-1094,
1998
9.
Ciardiello, F,
and
Tortora G.
Interactions between the epidermal growth factor receptor and type I protein kinase A: biological significance and therapeutic applications.
Clin Cancer Res
4:
821-828,
1998[Abstract].
10.
Clegg, HC,
Correll LA,
Cadd GG,
and
McKnight GS.
Inhibition of intracellular cAMP-dependent protein kinase using mutant genes of the regulatory type I subunit.
J Biol Chem
262:
13111-13119,
1987
11.
Colledge, M,
and
Scott JD.
AKAPs: from structure to function.
Trends Cell Biol
9:
216-221,
1999[ISI][Medline].
12.
Della Fazia, MA,
Servillo G,
and
Sassone-Corsi P.
Cyclic AMP signalling and cellular proliferation: regulation of CREB and CREM.
FEBS Lett
410:
22-24,
1997[ISI][Medline].
13.
Discafani, CM,
Carroll ML,
Floyd MB, Jr,
Hollander IJ,
Husain Z,
Johnson BD,
Kitchen D,
May MK,
Malo MS,
Minnick AA, Jr,
Nilakantan R,
Shen R,
Wang YF,
Wissner A,
and
Greenberger LM.
CL-387,785: an irreversible inhibitor of epidermal growth factor receptor tyrosine kinase with in vivo activity.
Biochem Pharmacol
57:
917-925,
1999[ISI][Medline].
14.
Dousa, TP.
Cyclic-3',5'-nucleotide phosphodiesterase isozymes in cell biology and pathophysiology of the kidney.
Kidney Int
55:
29-62,
1999[ISI][Medline].
15.
Dremier, S,
Pohl V,
Poteet-Smith C,
Roger PP,
Corbin J,
Doskeland SO,
Dumont JE,
and
Maenhaut C.
Activation of cyclic AMP-dependent kinase is required but may not be sufficient to mimic cyclic AMP-dependent DNA synthesis and thyroglobulin expression in dog thyroid cells.
Mol Cell Biol
17:
6717-6726,
1997[Abstract].
16.
Edwards, AS,
and
Scott JD.
A-kinase anchoring proteins: protein kinase A and beyond.
Curr Opin Cell Biol
12:
217-221,
2000[ISI][Medline].
17.
Eide, T,
Coghlan V,
Orstavik S,
Holsve C,
Solberg R,
Skalhegg BS,
Lamb NJ,
Langeberg L,
Fernandez A,
Scott JD,
Jahnsen T,
and
Tasken K.
Molecular cloning, chromosomal localization, and cell cycle-dependent subcellular distribution of the A-kinase anchoring protein, AKAP95.
Exp Cell Res
238:
305-316,
1998[ISI][Medline].
18.
Emmons, SW,
and
Somlo S.
Mating, channels, and kidney cysts.
Nature
401:
339-340,
1999[ISI][Medline].
19.
Francis, SH,
and
Corbin JD.
Cyclic nucleotide-dependent protein kinases: intracellular receptors for cAMP and cGMP action.
Crit Rev Clin Lab Sci
36:
275-328,
1999[ISI][Medline].
20.
Gjertsen, BT,
Mellgren G,
Otten A,
Maronde E,
Genieser HG,
Jastorff B,
Vintermyr OK,
McKnight GS,
and
Doskeland SO.
Novel (Rp)-cAMPS analogs as tools for inhibition of cAMP-kinase in cell culture. Basal cAMP-kinase activity modulates interleukin-1 beta action.
J Biol Chem
270:
20599-20607,
1995
21.
Grantham, JJ,
Uchic M,
Cragoe EJ, Jr,
Kornhaus J,
Grantham JA,
Donoso V,
Mangoo-Karim R,
Evan A,
and
McAteer J.
Chemical modification of cell proliferation and fluid secretion in renal cysts.
Kidney Int
35:
1379-1389,
1989[ISI][Medline].
22.
Hanaoka, K,
and
Guggino WB.
cAMP regulates cell proliferation and cyst formation in autosomal polycystic kidney disease cells.
J Am Soc Nephrol
11:
1179-1187,
2000
23.
Hansson, V,
Skalhegg BS,
and
Tasken K.
Cyclic-AMP-dependent protein kinase (PKA) in testicular cells. Cell specific expression, differential regulation and targeting of subunits of PKA.
J Steroid Biochem Mol Biol
69:
367-378,
1999[ISI][Medline].
24.
Izevbigie, EB,
Gutkind JS,
and
Ray PE.
Isoproterenol inhibits fibroblast growth factor-2-induced growth of renal epithelial cells.
Pediatr Nephrol
14:
726-734,
2000[ISI][Medline].
25.
Keryer, G,
Yassenko M,
Labbe JC,
Castro A,
Lohmann SM,
Evain-Brion D,
and
Tasken K.
Mitosis-specific phosphorylation and subcellular redistribution of the RIIalpha regulatory subunit of cAMP-dependent protein kinase.
J Biol Chem
273:
34594-34602,
1998
26.
Kimura, M,
and
Ogihara M.
Proliferation of adult rat hepatocytes by hepatocyte growth factor is potentiated by both phenylephrine and metaproterenol.
J Pharmacol Exp Ther
282:
1146-1154,
1997
27.
Klussmann, E,
Maric K,
Wiesner B,
Beyermann M,
and
Rosenthal W.
Protein kinase A anchoring proteins are required for vasopressin-mediated translocation of aquaporin-2 into cell membranes of renal principal cells.
J Biol Chem
274:
4934-4938,
1999
28.
Kumagai, N,
Inoue CN,
Kondo Y,
and
Iinuma K.
Mitogenic action of lysophosphatidic acid in proximal tubular epithelial cells obtained from voided human urine.
Clin Sci (Colch)
99:
561-567,
2000[ISI][Medline].
29.
Li, HP,
Geng L,
Burrow CR,
and
Wilson PD.
Identification of phosphorylation sites in the PKD1-encoded protein C-terminal domain.
Biochem Biophys Res Commun
259:
356-363,
1999[ISI][Medline].
30.
Mangoo-Karim, R,
Uchic M,
Lechene C,
and
Grantham JJ.
Renal epithelial cyst formation and enlargement in vitro: dependence on cAMP.
Proc Natl Acad Sci USA
86:
6007-6011,
1989[Abstract].
31.
Mangoo-Karim, R,
Uchic ME,
Grant M,
Shumate WA,
Calvet JP,
Park CH,
and
Grantham JJ.
Renal epithelial fluid secretion and cyst growth: the role of cyclic AMP.
FASEB J
3:
2629-2632,
1989
32.
Martin, ME,
Hidalgo J,
Vega FM,
and
Velasco A.
Trimeric G proteins modulate the dynamic interaction of PKAII with the Golgi complex.
J Cell Sci
112:
3869-3878,
1999
33.
Matousovic, K,
Tsuboi Y,
Walker H,
Grande JP,
and
Dousa TP.
Inhibitors of cyclic nucleotide phosphodiesterase isozymes block renal tubular cell proliferation induced by folic acid.
J Lab Clin Med
130:
487-495,
1997[ISI][Medline].
34.
McAteer, JA,
Evan AP,
and
Gardner KD.
Morphogenetic clonal growth of kidney epithelial cell line MDCK.
Anat Rec
217:
229-239,
1987[ISI][Medline].
35.
Murcia, NS,
Sweeney WE, Jr,
and
Avner ED.
New insights into the molecular pathophysiology of polycystic kidney disease.
Kidney Int
55:
1187-1197,
1999[ISI][Medline].
36.
Murcia, NS,
Woychik RP,
and
Avner ED.
The molecular biology of polycystic kidney disease.
Pediatr Nephrol
12:
721-726,
1998[ISI][Medline].
37.
Nadasdy, T,
Laszik Z,
Lajoie G,
Blick KE,
Wheeler DE,
and
Silva FG.
Proliferative activity of cyst epithelium in human renal cystic diseases.
J Am Soc Nephrol
5:
1462-1468,
1995[Abstract].
38.
Noguchi, K,
Murata T,
and
Cho-Chung YS.
8-Chloroadenosine 3',5'-monophosphate (8-Cl-cAMP) selectively eliminates protein kinase A type I to induce growth inhibition in c-ras-transformed fibroblasts.
Eur J Cancer
34:
1260-1267,
1998[ISI][Medline].
39.
Orellana, SA,
and
Avner ED.
Cystic maldevelopment of the kidney.
Semin Nephrol
15:
341-352,
1995[ISI][Medline].
40.
Orellana, SA,
and
Avner ED.
Cell and molecular biology of kidney development.
Semin Nephrol
18:
233-243,
1998[ISI][Medline].
41.
Orellana, SA,
and
Marfella-Scivittaro C.
Distinctive cyclic AMP-dependent protein kinase subunit localization is associated with cyst formation and loss of tubulogenic capacity in Madin Darby canine kidney cell clones.
J Biol Chem
275:
21233-21240,
2000
42.
Orellana, SA,
Neff CD,
Sweeney WE,
and
Avner ED.
Novel Madin Darby canine kidney cell clones exhibit unique phenotypes in response to morphogens.
In Vitro Cell Dev Biol
32:
329-339,
1996[ISI].
43.
Orellana, SA,
Sweeney WE,
Neff CD,
and
Avner ED.
Epidermal growth factor receptor expression is abnormal in murine polycystic kidney.
Kidney Int
47:
490-499,
1995[ISI][Medline].
44.
Osinski, MT,
and
Schror K.
Inhibition of platelet-derived growth factor-induced mitogenesis by phosphodiesterase 3 inhibitors: role of protein kinase A in vascular smooth muscle cell mitogenesis.
Biochem Pharmacol
60:
381-387,
2000[ISI][Medline].
45.
Parnell, SC,
Magenheimer BS,
Maser RL,
and
Calvet JP.
Identification of the major site of in vitro PKA phosphorylation in the polycystin-1 C-terminal cytosolic domain.
Biochem Biophys Res Commun
259:
539-543,
1999[ISI][Medline].
46.
Pham, N,
Cheglakov I,
Koch CA,
de Hoog CL,
Moran MF,
and
Rotin D.
The guanine nucleotide exchange factor CNrasGEF activates ras in response to cAMP and cGMP.
Curr Biol
10:
555-558,
2000[ISI][Medline].
47.
Piscione, TD,
Phan T,
and
Rosenblum ND.
BMP7 controls collecting tubule cell proliferation and apoptosis via Smad1-dependent and -independent pathways.
Am J Physiol Renal Physiol
280:
F19-F33,
2001
48.
Pugh, JL,
Sweeney WE, Jr,
and
Avner ED.
Tyrosine kinase activity of the EGF receptor in murine metanephric organ culture.
Kidney Int
47:
774-781,
1995[ISI][Medline].
49.
Qian, Q,
Harris PC,
and
Torres VE.
Treatment prospects for autosomal-dominant polycystic kidney disease.
Kidney Int
59:
2005-2022,
2001[ISI][Medline].
50.
Quigley, R,
and
Baum M.
Effects of epidermal growth factor and transforming growth factor- on rabbit proximal tubule solute transport.
Am J Physiol Renal Fluid Electrolyte Physiol
266:
F459-F465,
1994
51.
Rangel-Aldao, R,
and
Rosen OM.
Dissociation and reassociation of the phosphorylated and nonphosphorylated forms of adenosine 3':5'-monophosphate-dependent protein kinase from bovine cardiac muscle.
J Biol Chem
251:
3375-3380,
1976[Abstract].
52.
Rankin, CA,
Ziemer DM,
Maser RL,
Foo I,
and
Calvet JP.
Growth characteristics of cells cultured from two murine models of polycystic kidney disease.
In Vitro Cell Dev Biol Anim
32:
100-106,
1996[ISI][Medline].
53.
Richards, WG,
Sweeney WE,
Yoder BK,
Wilkinson JE,
Woychik RP,
and
Avner ED.
Epidermal growth factor receptor activity mediates renal cyst formation in polycystic kidney disease.
J Clin Invest
101:
935-939,
1998
54.
Rohlff, C,
Clair T,
and
Cho-Chung YS.
8-Cl-cAMP induces truncation and down-regulation of the RI alpha subunit and up-regulation of the RII beta subunit of cAMP-dependent protein kinase leading to type II holoenzyme-dependent growth inhibition and differentiation of HL-60 leukemia cells.
J Biol Chem
268:
5774-5782,
1993
55.
Russell, ES,
and
McFarland EC.
Cystic kidneys.
Mouse Newsl
56:
40-43,
1977.
56.
Schmitt, JM,
and
Stork PJ.
Beta 2-adrenergic receptor activates extracellular signal-regulated kinases (ERKs) via the small G protein rap1 and the serine/threonine kinase B-Raf.
J Biol Chem
275:
25342-25350,
2000
57.
Shih, M,
Lin F,
Scott JD,
Wang HY,
and
Malbon CC.
Dynamic complexes of beta2-adrenergic receptors with protein kinases and phosphatases and the role of gravin.
J Biol Chem
274:
1588-1595,
1999
58.
Singh, AK,
Tasken K,
Walker W,
Frizzell RA,
Watkins SC,
Bridges RJ,
and
Bradbury NA.
Characterization of PKA isoforms and kinase-dependent activation of chloride secretion in T84 cells.
Am J Physiol Cell Physiol
275:
C562-C570,
1998
59.
Soderling, SH,
Bayuga SJ,
and
Beavo JA.
Cloning and characterization of a cAMP-specific cyclic nucleotide phosphodiesterase.
Proc Natl Acad Sci USA
95:
8991-8996,
1998
60.
Sweeney, WE,
and
Avner ED.
Functional activity of epidermal growth factor receptors in autosomal recessive polycystic kidney disease.
Am J Physiol Renal Physiol
275:
F387-F394,
1998
61.
Sweeney, WE,
Chen Y,
Nakanishi K,
Frost P,
and
Avner ED.
Treatment of polycystic kidney disease with a novel tyrosine kinase inhibitor 1.
Kidney Int
57:
33-40,
2000[ISI][Medline].
62.
Sweeney, WE,
Futey L,
Frost P,
and
Avner ED.
In vitro modulation of cyst formation by a novel tyrosine kinase inhibitor.
Kidney Int
56:
406-413,
1999[ISI][Medline].
63.
Sweeney, WE, Jr,
Futey L,
Frost P,
and
Avner ED.
In vitro modulation of cyst formation by a novel tyrosine kinase inhibitor.
Kidney Int
56:
406-413,
1999[ISI][Medline].
64.
Tasken, K,
Skalhegg BS,
Tasken KA,
Solberg R,
Knutsen HK,
Levy FO,
Sandberg M,
Orstavik S,
Larsen T,
Johansen AK,
Vang T,
Schrader HP,
Reinton NT,
Torgersen KM,
Hansson V,
and
Jahnsen T.
Structure, function, and regulation of human cAMP-dependent protein kinases.
Adv Second Messenger Phosphoprotein Res
31:
191-204,
1997[ISI][Medline].
65.
Tortora, G,
Damiano V,
Bianco C,
Baldassare G,
Bianco AR,
Lanfrancone L,
Pelicci PG,
and
Ciardiello F.
The RI subunit of protein kinase A (PKA) binds to Grb2 and allows PKA interaction with the activated EGF-receptor.
Oncogene
14:
923-928,
1997[ISI][Medline].
66.
Tortora, G,
Pepe S,
Bianco C,
Baldassare G,
Budillon A,
Clair T,
Cho-Chung YS,
Bianco AR,
and
Ciardiello F.
The RI subunit of protein kinase A controls serum dependency and entry into cell cycle of human mammary epithelial cells.
Oncogene
9:
3233-3240,
1994[ISI][Medline].
67.
Tsygankova, OM,
Kupperman E,
Wen W,
and
Meinkoth JL.
Cyclic AMP activates Ras.
Oncogene
19:
3609-3615,
2000[ISI][Medline].
68.
Voisin, T,
Bens M,
Cluzeaud F,
Vandewalle A,
and
Laburthe M.
Peptide YY receptors in the proximal tubule PKSV-PCT cell line derived from transgenic mice. Relation with cell growth.
J Biol Chem
268:
20547-20554,
1993
69.
Watnick, T,
and
Germino GG.
Molecular basis of autosomal dominant polycystic kidney disease.
Semin Nephrol
19:
327-343,
1999[ISI][Medline].
70.
Westphal, RS,
Tavalin SJ,
Lin JW,
Alto NM,
Fraser IDC,
Langeberg LK,
Sheng M,
and
Scott JD.
Regulation of NMDA receptors by an associated phosphatase-kinase signaling complex.
Science
285:
93-96,
1999
71.
Wolf, G,
and
Neilson EG.
Isoproterenol induces mitogenesis in MCT and LLC-PK1 tubular cells.
J Am Soc Nephrol
4:
1995-2002,
1994[Abstract].
72.
Yamada, T,
Terada Y,
Homma MK,
Nonoguchi H,
Sasaki S,
Yuasa Y,
Tomita K,
and
Marumo F.
AVP inhibits EGF-stimulated MAP kinase cascade in Madin-Darby canine kidney cells.
Kidney Int
48:
745-752,
1995[ISI][Medline].
73.
Yamaguchi, T,
Nagao S,
Kasahara M,
Takahashi H,
and
Grantham JJ.
Renal accumulation and excretion of cyclic adenosine monophosphate in a murine model of slowly progressive polycystic kidney disease.
Am J Kidney Dis
30:
703-709,
1997[ISI][Medline].
74.
Yamaguchi, T,
Pelling JC,
Ramaswamy NT,
Eppler JW,
Wallace DP,
Nagao S,
Rome LA,
Sullivan LP,
and
Grantham JJ.
cAMP stimulates the in vitro proliferation of renal cyst epithelial cells by activating the extracellular signal-regulated kinase pathway.
Kidney Int
57:
1460-1471,
2000[ISI][Medline].
75.
Yang, J,
Drazba JA,
Ferguson DG,
and
Bond M.
A-kinase anchoring protein 100 (AKAP100) is localized in multiple subcellular compartments in the adult rat heart.
J Cell Biol
142:
511-522,
1998
76.
Yang, S,
Fletcher WH,
and
Johnson DA.
Regulation of cAMP-dependent protein kinase: enzyme activation without dissociation.
Biochemistry
34:
6267-71,
1995[ISI][Medline].
77.
Zakhary, DR,
Fink MA,
Ruehr ML,
and
Bond M.
Selectivity and regulation of A-kinase anchoring proteins in the heart. The role of autophosphorylation of the type II regulatory subunit of cAMP-dependent protein kinase.
J Biol Chem
275:
41389-41395,
2000
78.
Zakhary, DR,
Moravec CS,
and
Bond M.
Regulation of PKA binding to AKAPs in the heart: alterations in human heart failure.
Circulation
101:
1459-1464,
2000