Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
1.
Kidney Int ; 73(1): 108-16, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17960141

ABSTRACT

The Consortium of Radiologic Imaging Studies of Polycystic Kidney Disease (CRISP) recently showed that renal enlargement in autosomal-dominant polycystic kidney disease mimicked exponential growth. We determined the effects of cyst initiation rate, total number, and growth rate on the time-dependent change of total cyst volume (TCV). Mathematical models with equations integrating cyst surface area, volume, and an invariant growth rate constant were used to compute the time-dependent change in volume of solitary and multiple cysts. Multiple expanding cysts increased TCV in an exponential-like pattern even when individual cysts formed at different rates or exhibited different but constant growth rates. TCV depended on the rate of cyst initiation and on the total number of cysts; however, the compounding effect of exponential-like growth was the most powerful determinant of long-term cyst expansion. Extrapolation of TCV data plots for individual subjects back to an age of 18 predicted TCV values within an established range. We conclude that cysts started early in life were the main contributor to eventual TCV while their growth rate primarily determined renal size; although the rate of formation and the ultimate number of cysts also contributed. The good fit between the exponential models and the extrapolated CRISP data indicates that the TCV growth rate is a defining trait for individual patients and may be used as a prognostic marker.


Subject(s)
Cysts/pathology , Kidney/pathology , Models, Biological , Polycystic Kidney, Autosomal Dominant/pathology , Humans , Organ Size
2.
Kidney Int ; 60(2): 484-94, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11473631

ABSTRACT

BACKGROUND: Polycystic kidney disease (PKD) is characterized by the abnormal proliferation of tubular epithelial cells. It was recently shown that the growth of PKD cyst-lining cells is stimulated by cyclic adenosine monophosphate (cAMP), whereas the growth of normal human kidney cortex cells is inhibited. METHODS: We have examined the effects of overexpressing the C-terminal cytosolic tail of mouse polycystin-1, as a membrane-targeted fusion protein, on cAMP-responsive cell proliferation in stably transfected M-1 cortical collecting duct cells. Two cell lines that express high levels of the polycystin-1 fusion protein and two control cell lines that do not express the fusion protein were tested. RESULTS: Growth of parental M-1 cells and the control cell lines was inhibited by 8-Br-cAMP and by a variety of cAMP agonists. In contrast, growth of the polycystin-1-expressing clones was stimulated by cAMP. Consistent with this, the protein kinase A (PKA) inhibitor H-89 caused either a positive or a negative growth effect depending on the primary response to cAMP. PD98059 blocked the cAMP stimulation of cell proliferation, indicating that the pathway is MEK1 dependent. CONCLUSIONS: Expression of the polycystin-1 C-terminal tail disrupts normal cellular signaling and transforms the stably transfected M-1 cells to an abnormal PKD cell proliferation phenotype.


Subject(s)
Cyclic AMP/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Proteins/genetics , Proteins/metabolism , Sulfonamides , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Line , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression/physiology , Isoquinolines/pharmacology , Kidney Tubules, Collecting/cytology , Mice , Phenotype , RNA, Messenger/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , TRPP Cation Channels , Transfection
3.
Curr Opin Nephrol Hypertens ; 10(4): 533-42, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11458035

ABSTRACT

Collated in this highly personal commentary are the most important research findings of the past 10 years that deal primarily with the renal manifestations of inherited polycystic kidney diseases. Progress in understanding these complex disorders has followed two major concurrent and convergent lines of investigation: genes and genetic mechanisms, and pathogenesis and progression. The field has moved from descriptive pathobiology to the elucidation of molecular mechanisms consequent to genetic and epigenetic events. Doubtless, the favorite works of some who have labored diligently in this field have not been fully exalted, and for this I apologize. Were I the editor, this entire celebratory volume would be used to extol the thrilling growth of knowledge during the tenure of this polycystic kidney disease watcher.


Subject(s)
Polycystic Kidney Diseases/history , Animals , Disease Models, Animal , History, 20th Century , Humans , Mice , Mutation , Polycystic Kidney Diseases/etiology , Polycystic Kidney Diseases/genetics , Polycystic Kidney, Autosomal Dominant/etiology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/history , United States
4.
Am J Physiol Renal Physiol ; 280(6): F1019-29, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11352842

ABSTRACT

We used an unambiguous in vitro method to determine if inner medullary collecting ducts (IMCD) have intrinsic capacities to absorb and secrete solutes and fluid in an isotonic medium. IMCD(1), IMCD(2), and IMCD(3) were dissected from kidneys of young Sprague-Dawley rats. 8-Bromo-3',5'-cyclic monophosphate (8-BrcAMP) stimulated lumen formation and progressive dilation in all IMCD subsegments; lumen formation was greatest in IMCD(1.) Benzamil potentiated the rate of lumen expansion in response to 8-BrcAMP. Fluid entered tubule lumens by transcellular secretion rather than simple translocation of intracellular fluid. Secreted lumen solutes were osmometrically active. Inhibition of protein kinase A with H-89 and Rp diastereomer of adenosine 3',5'-cyclic monophosphorothioate blocked fluid secretion. The rate of lumen expansion was reduced by the selective addition of ouabain, barium, diphenyl-2-carboxylate, bumetanide, glybenclamide, or DIDS, or reduction of extracellular Cl(-). We conclude that IMCD absorb and secrete electrolytes and fluid in vitro and that secretion is accelerated by cAMP. We suggest that salt and fluid secretion by the terminal portions of the renal collecting system may have a role in modulating the composition and volume of the final urine.


Subject(s)
Cyclic AMP/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Collecting/metabolism , Sulfonamides , Water-Electrolyte Balance/physiology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Barium/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Blood Proteins/pharmacology , Calcium Channel Blockers/pharmacology , Chlorides/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Dissection , Enzyme Inhibitors/pharmacology , Glyburide/pharmacology , Hypoglycemic Agents/pharmacology , Isoquinolines/pharmacology , Male , Ouabain/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Chloride/pharmacokinetics , Vanadates/pharmacology , Water/metabolism , ortho-Aminobenzoates/pharmacology
5.
Semin Nephrol ; 21(2): 107-23, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11245774

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a major, inherited disorder that is characterized by the growth of large, fluid-filled cysts from the tubules and collecting ducts of affected kidneys, and by a number of extrarenal manifestations including liver and pancreatic cysts, hypertension, heart valve defects, and cerebral and aortic aneurysms. Mutations in either of 2 different genes (PKD1 or PKD2) give rise to ADPKD. Most mutations identified in affected families appear to inactivate the PKD genes, and accumulating evidence suggests that a 2-hit mechanism, in which the normal PKD1 or PKD2 allele is also mutated, may be required for cyst growth. The protein products of the PKD genes (polycystin-1 and polycystin-2) are thought to function together as part of a multiprotein membrane-spanning complex involved in cell-cell or cell-matrix interactions. Polycystin-1 and polycystin-2 can initiate signal transduction, leading to the activation of a number of downstream effectors, including heterotrimeric G-proteins, protein kinase C, mitogen-activated protein kinases, beta-catenin, and the AP-1 transcription factor. In addition, polycystin-2 may function in mediating calcium flux. The pathogenesis of cyst formation is currently thought to involve increased cell proliferation, fluid accumulation, and basement membrane remodeling. It now appears that cyclic adenosine monophosphate (cAMP) metabolism is a central component of cyst formation, stimulating apical chloride secretion and driving the accumulation of cyst fluid. Recent evidence has shown that ADPKD cells also have an altered responsiveness to cyclic AMP. In contrast to normal kidney cells whose cell proliferation is inhibited by cyclic AMP, ADPKD cells are stimulated to proliferate. Thus, it is likely that an alteration in polycystin function transforms the normal cellular phenotype to one that responds to elevated cyclic AMP by an increased rate of cell proliferation and that the enlarging cyst expands by an increased rate of cyclic AMP-driven fluid secretion. Cyclic AMP and growth factors, including epidermal growth factor, have complementary effects to accelerate the enlargement of ADPKD cysts, and thereby to contribute to the progression of the disease. This knowledge should facilitate the discovery of inhibitors of signal transduction cascades that can be used in the treatment of ADPKD.


Subject(s)
Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/physiopathology , Cyclic AMP/physiology , Humans , Membrane Proteins/physiology , Mutation , Proteins/physiology , Signal Transduction , TRPP Cation Channels
7.
Kidney Int ; 58(6): 2492-501, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11115083

ABSTRACT

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is characterized by progressive renal enlargement and renal failure. We evaluated sequential radiocontrast-enhanced computed tomography (CT) scans to determine the rate of kidney enlargement in patients with ADPKD. METHODS: Ten adult patients with ADPKD (4 men and 6 women) with initial serum creatinine levels 0.05), and 185 +/- 52 mL (P < 0.01), respectively. In 19 individual spherical cysts selected in six patients, the mean initial volume was 15.0 +/- 7.2 mL (range 1.1 to 137 mL), and the average rate of volume increase was 0.52 +/- 0.21 mL/month (P < 0.025, range 0.02 to 4.15 mL/month). In five patients who eventually required dialysis, 11.2 years after the initial CT scan, the initial cyst/kidney volume ratio (combined for both kidneys) exceeded 0.43; four patients with lower cyst/kidney volume ratios had serum creatinine levels <1.5 mg/dL, 8.7 years after the initial CT scan. CONCLUSIONS: On the basis of this preliminary survey of archival material, we conclude that conventional contrast-enhanced CT scans can be used to quantitate volume components of progression in ADPKD. The rates of individual kidney and cyst enlargement are highly variable within and between patients, but overall, the values increase over time. The volume fraction of kidneys comprised of cysts may be a useful indicator of ADPKD progression.


Subject(s)
Polycystic Kidney, Autosomal Dominant/diagnostic imaging , Polycystic Kidney, Autosomal Dominant/pathology , Tomography, X-Ray Computed/methods , Adult , Age Factors , Body Fluids/metabolism , Chlorides/metabolism , Disease Progression , Female , Humans , Kidney/pathology , Kidney Failure, Chronic/diagnostic imaging , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Male , Middle Aged , Polycystic Kidney, Autosomal Dominant/metabolism , Predictive Value of Tests , Prognosis
8.
Kidney Int ; 57(4): 1460-71, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10760082

ABSTRACT

BACKGROUND: : Cellular proliferation is a key factor in the enlargement of renal cysts in autosomal dominant polycystic kidney disease (ADPKD). We determined the extent to which adenosine 3':5'-cyclic monophosphate (cAMP) may regulate the in vitro proliferation of cyst epithelial cells derived from human ADPKD cysts. METHODS: : Epithelial cells from cysts of individuals with ADPKD and from normal human kidney cortex (HKC) of individuals without ADPKD were cultured. The effects of agonists and inhibitors on the rate of cellular proliferation and the activation of extracellular signal-regulated kinase (ERK1/2) were determined. RESULTS: : 8-Br-cAMP (100 micromol/L) stimulated the proliferation of cells from eight different ADPKD subjects to 99.0% above baseline; proliferation was inhibited by protein kinase A (PKA) antagonists H-89 (97%) and Rp-cAMP (90%). Forskolin (10 micromol/L), which activates adenylyl cyclase, increased proliferation 124%, and receptor-mediated agonists arginine vasopressin, desmopressin, secretin, vasoactive intestinal polypeptide, and prostaglandin E2 stimulated proliferation 54.2, 56.3, 46.7, 37.1, and 48.3%, respectively. The mitogen extracellular kinase (MEK) inhibitor PD98059 completely inhibited ADPKD cell proliferation in response to cAMP agonists, but genistein, a receptor tyrosine kinase inhibitor, did not block cAMP-dependent proliferation. cAMP agonists increased the activity of ERK above control levels within five minutes. In contrast to ADPKD, proliferation and ERK activity of cells derived from normal HKC were not stimulated by cAMP agonists, although electrogenic Cl- secretion was increased by these agonists in both ADPKD and HKC cell monolayers. CONCLUSIONS: : We conclude that cAMP agonists stimulate the proliferation of ADPKD but not HKC epithelial cells through PKA activation of the ERK pathway at a locus distal to receptor tyrosine kinase. We suggest that the adenylyl cyclase signaling pathway may have a unique role in determining the rate of cyst enlargement in ADPKD through its actions to stimulate cellular proliferation and transepithelial solute and fluid secretion.


Subject(s)
Cyclic AMP/pharmacology , Kidney/pathology , Mitogen-Activated Protein Kinases/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Cell Division/drug effects , Cells, Cultured , Electric Impedance , Enzyme Activation/physiology , Epithelial Cells/enzymology , Epithelial Cells/pathology , Epithelial Cells/physiology , Humans , Kidney/enzymology , Kidney/physiopathology , Kidney Cortex/cytology , Kidney Cortex/physiology , Polycystic Kidney, Autosomal Dominant/enzymology , Polycystic Kidney, Autosomal Dominant/physiopathology
9.
Biochim Biophys Acta ; 1464(1): 69-82, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10704921

ABSTRACT

A synthetic Cl(-) channel-forming peptide, C-K4-M2GlyR, applied to the apical membrane of human epithelial cell monolayers induces transepithelial Cl(-) and fluid secretion. The sequence of the core peptide, M2GlyR, corresponds to the second membrane-spanning region of the glycine receptor, a domain thought to line the pore of the ligand-gated Cl(-) channel. Using a pharmacological approach, we show that the flux of Cl(-) through the artificial Cl(-) channel can be regulated by modulating basolateral K(+) efflux through Ca(2+)-dependent K(+) channels. Application of C-K4-M2GlyR to the apical surface of monolayers composed of human colonic cells of the T84 cell line generated a sustained increase in short-circuit current (I(SC)) and caused net fluid secretion. The current was inhibited by the application of clotrimazole, a non-specific inhibitor of K(+) channels, and charybdotoxin, a potent inhibitor of Ca(2+)-dependent K(+) channels. Direct activation of these channels with 1-ethyl-2-benzimidazolinone (1-EBIO) greatly amplified the Cl(-) secretory current induced by C-K4-M2GlyR. The effect of the combination of C-K4-M2GlyR and 1-EBIO on I(SC) was significantly greater than the sum of the individual effects of the two compounds and was independent of cAMP. Treatment with 1-EBIO also increased the magnitude of fluid secretion induced by the peptide. The cooperative action of C-K4-M2GlyR and 1-EBIO on I(SC) was attenuated by Cl(-) transport inhibitors, by removing Cl(-) from the bathing solution and by basolateral treatment with K(+) channel blockers. These results indicate that apical membrane insertion of Cl(-) channel-forming peptides such as C-K4-M2GlyR and direct activation of basolateral K(+) channels with benzimidazolones may coordinate the apical Cl(-) conductance and the basolateral K(+) conductance, thereby providing a pharmacological approach to modulating Cl(-) and fluid secretion by human epithelia deficient in cystic fibrosis transmembrane conductance regulator Cl(-) channels.


Subject(s)
Chloride Channels/metabolism , Epithelial Cells/drug effects , Peptides/pharmacology , Amino Acid Sequence , Benzimidazoles/pharmacology , Calcium/metabolism , Calcium Channel Agonists/pharmacology , Cell Line , Chlorides/antagonists & inhibitors , Chlorides/metabolism , Clotrimazole/pharmacology , Cyclic AMP/metabolism , Cystic Fibrosis/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Electric Conductivity , Epithelial Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Molecular Sequence Data , Potassium Channel Blockers , Potassium Channels/metabolism
10.
Am J Kidney Dis ; 35(2): 221-6, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10676720

ABSTRACT

Epithelial proliferation, extracellular matrix remodeling, and interstitial inflammation are central elements in the pathogenesis of slowly progressive polycystic kidney disorders. Probucol, an antioxidant that lowers plasma cholesterol, has been shown to decrease smooth muscle cell proliferation and macrophage accumulation in blood vessels and to prevent restenosis after coronary angioplasty. We determined in 30-day-old male BDF1-pcy hybrid mice (derived from mating DBA/2FG-pcy and C57BL/6FG-pcy) the effect of probucol administered in the diet (1%) for 200 days on kidney weight relative to body weight (KW/BW), cyst expansion, renal interstitial fibrosis, and serum urea nitrogen (SUN) concentration. Animals were fed a moderately high-protein diet (HPD, 36%) to accentuate the development of renal cysts and to promote interstitial fibrosis. Probucol decreased serum cholesterol from 68 to 16 mg/dL but had no effect on food intake or body weight. Probucol decreased relative kidney size from 4.16% +/- 0.55% to 2.64% +/- 0.12% KW/BW (P < 0.01), SUN from 30.5 +/- 1.8 to 25.9 +/- 1.0 mg/dL (P < 0.05), cystic index from 2.45 +/- 0.11 to 1.36 +/- 0.10 (P < 0.01), and fibrosis index from 2.40 +/- 0.11 to 1.82 +/- 0.08 (P < 0.01). We conclude that probucol ameliorates the progressive deterioration in renal function and structure in pcy mice ingesting a relatively high level of dietary protein.


Subject(s)
Anticholesteremic Agents/therapeutic use , Polycystic Kidney Diseases/drug therapy , Probucol/therapeutic use , Animals , Disease Models, Animal , Disease Progression , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Time Factors
12.
Physiol Rev ; 78(4): 1165-91, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9790573

ABSTRACT

In autosomal dominant polycystic kidney disease (ADPKD), the genetic defect results in the slow growth of a multitude of epithelial cysts within the renal parenchyma. Cysts originate within the glomeruli and all tubular structures, and their growth is the result of proliferation of incompletely differentiated epithelial cells and the accumulation of fluid within the cysts. The majority of cysts disconnect from tubular structures as they grow but still accumulate fluid within the lumen. The fluid accumulation is the result of secretion of fluid driven by active transepithelial Cl- secretion. Proliferation of the cells and fluid secretion are activated by agonists of the cAMP signaling pathway. The transport mechanisms involved include the cystic fibrosis transmembrane conductance regulator (CFTR) present in the apical membrane of the cystic cells and a bumetanide-sensitive transporter located in the basolateral membrane. A lipid factor, called cyst activating factor, has been found in the cystic fluid. Cyst activating factor stimulates cAMP production, proliferation, and fluid secretion by cultured renal epithelial cells and also is a chemotactic agent. Cysts also appear in the intrahepatic biliary tree in ADPKD. Normal ductal cells secrete Cl- and HCO3-. The cystic ductal cell also secretes Cl-, but HCO3- secretion is diminished, probably as the result of a lower population of Cl-/HCO3- exchangers in the apical membrane as compared with the normal cells. Some segments of the normal renal tubule are also capable of utilizing CFTR to secrete Cl-, particularly the inner medullary collecting duct. The ability of Madin-Darby canine kidney cells and normal human kidney cortex cells to form cysts in culture and to secrete fluid and the functional similarities between these incompletely differentiated, proliferative cells and developing cells in the intestinal crypt and in the fetal lung have led us to suggest that Cl- and fluid secretion may be a common property of at least some renal epithelial cells in an intermediate stage of development. The genetic defect in ADPKD may not directly affect membrane transport mechanisms but rather may arrest the development of certain renal epithelial cells in an incompletely differentiated, proliferative stage.


Subject(s)
Epithelial Cells/metabolism , Polycystic Kidney, Autosomal Recessive/metabolism , Animals , Biological Transport , Cysts/complications , Cysts/metabolism , Disease Models, Animal , Dogs , Humans , Liver Diseases/complications , Liver Diseases/metabolism , Polycystic Kidney, Autosomal Recessive/complications , Polycystic Kidney, Autosomal Recessive/genetics
15.
Am J Kidney Dis ; 30(5): 703-9, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9370187

ABSTRACT

Evidence from in vitro studies indicates that increased proliferation of epithelial cells and secretion of fluid by these cells may be important factors in the progressive enlargement of renal cysts. The rate of cellular proliferation and fluid secretion by cyst epithelium in vitro can be strikingly accelerated by cyclic adenosine 3'5' monophosphate (cAMP) and agonists that lead to the production of this nucleotide. The extent to which renal cAMP content is increased in polycystic kidneys is unknown. In the current study, we determined the amount of this nucleotide in intact kidneys, cyst fluid, plasma, and urine in nonazotemic mice (DBA/2FG-pcy/pcy) with a slowly progressive form of inherited polycystic kidney disease (PKD). In 45 pcy/pcy mice studied 20, 45, or 70 days after birth, the total kidney cAMP content was 0.22 +/- 0.01, 0.46 +/- 0.02, and 0.90 +/- 0.05 pmol/mg tissue, respectively. By contrast, in 37 control DBA/2J mice the levels of cAMP at identical times remained relatively constant at 0.22 +/- 0.01, 0.21 +/- 0.01, and 0.29 +/- 0.01 pmol/mg tissue, respectively. In 70-day-old nonazotemic pcy/pcy mice with normal serum levels of parathyroid hormone, cAMP generated by the kidneys (nephrogenous cAMP) was 22.9 +/- 2.8 nmol/100 mL creatinine clearance, compared with 6.5 +/- 1.3 in normal animals of the same age (P < 0.001). The cyst fluids of 70-day-old pcy/pcy mice contained a lipid that increased transepithelial secretion of fluid by MDCK monolayers from a baseline of 0.012 +/- 0.002 to 0.136 +/- 0.008 microL/cm2/hr (P < 0.05). This lipid also stimulated cellular proliferation by monolayers of cultured MDCK and LLC-PK1 cells 2.5- and 7.9-fold (P < .05), respectively, and stimulated cAMP accumulation by these cells 1.6- and 2.0-fold (P < .05), respectively. These studies illustrate that renal cAMP production and excretion increase in concert with the cystic enlargement of the kidneys in DBA/2FG-pcy/pcy mice and identify a lipid cAMP agonist in murine renal cystic kidney disease.


Subject(s)
Cyclic AMP/metabolism , Disease Models, Animal , Kidney/metabolism , Polycystic Kidney Diseases/metabolism , Aging/metabolism , Analysis of Variance , Animals , Cell Division , Cell Line , Cells, Cultured , Cyclic AMP/analysis , Disease Progression , Kidney/cytology , LLC-PK1 Cells/cytology , LLC-PK1 Cells/metabolism , Male , Mice , Mice, Inbred DBA , Swine
16.
Proc Assoc Am Physicians ; 109(4): 397-408, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9220537

ABSTRACT

Advanced autosomal dominant polycystic kidney disease (ADPKD) is characterized morphologically by massive cyst enlargement, moderate interstitial infiltration with mononuclear cells, and extensive fibrosis. In patients affected by a common genotype (PKD1), it has been suggested that the progressive decline in renal function that transpires over a highly variable time course may be due to endogenous or exogenous epigenetic factors. We have postulated that a neutral lipid, discovered in human cyst fluid and stimulating the rates of transepithelial fluid secretion and cellular proliferation of renal epithelial cells in vitro may have a potential role in cyst growth and the progressive decline of kidney function. In this study, we used thin-layer chromatography (TLC) and high-performance TLC (HPTLC) to determine whether lipid extracts of human cyst fluid stimulated monocyte chemotaxis in vitro. Monocyte chemotactic activity, determined by the transmembrane migration of murine RAW 264.7 cells, was stimulated (delta 26.0 +/- 1.5 optical density units) by a lipid fraction less polar than sphingosine but more polar by TLC and HPTLC than 1-monooleoylglycerol. A high level of secretagogue activity was detected in this fraction (delta 0.336 +/- 0.022 microliter/cm2 1 hr) and to a lesser extent (delta 0.253 +/- 0.022 microliter/cm2/hr) in a neighboring fraction that encompassed the 1-monooleoylglycerol standard. Cyst fluid with undetectable secretagogue activity had a monocyte chemotactic-activity level only 18% as great as fluids with high levels of secretagogue activity. The secretagogue and chemotactic activities in TLC-HPTLC fractions were resistant to treatment with KOH, but both were diminished by HCl, borohydride, or periodate. Rat proximal tubule cultures incubated with oleate complexed with albumin elaborated secretagogue and chemotactic activities in the conditioned medium, with TLC-HPTLC mobility characteristics similar to the biologically active cyst fluid lipids. On the basis of these studies, we conclude that human cyst fluids harbor potent secretagogue and chemotactic lipids that may have a role in determining the functional course of ADPKD. On the basis of preliminary chemical characterizations, we suggest that the secretatogue and monocyte chemotactic activities of cyst fluid may reflect the action of lipid molecules of similar structure, the source of which may be renal epithelial cells.


Subject(s)
Chemotaxis, Leukocyte/physiology , Cysts/metabolism , Lipids/physiology , Polycystic Kidney, Autosomal Dominant/physiopathology , Animals , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Cysts/chemistry , Endotoxins/analysis , Exudates and Transudates/chemistry , Humans , Lipids/analysis , Mice , Monocytes/physiology , Polycystic Kidney, Autosomal Dominant/pathology , Prostaglandins/analysis , Rats , Rats, Inbred Lew , Sensitivity and Specificity
17.
Am J Physiol ; 272(5 Pt 1): C1672-9, 1997 May.
Article in English | MEDLINE | ID: mdl-9176159

ABSTRACT

M2GlyR is a synthetic 23-amino acid peptide that mimics the second membrane-spanning region of the alpha-subunit of the postsynaptic glycine receptor. This peptide has been shown to form an anion-selective channel in phospholipid bilayers. We have investigated the possibility that the peptide may incorporate into the apical membrane of secretory epithelia and induce the secretion of Cl- and water. We improved the solubility of this peptide by adding four lysine residues to the carboxy terminus, C-K4-M2GlyR, and assayed its channel-forming activity using a subculture of Madin-Darby canine kidney (MDCK) cells. The addition of 100 microM C-K4-M2GlyR to the apical surface of MDCK monolayers significantly increased short-circuit current (Ise), hyperpolarized transepithelial potential difference, and induced fluid secretion. The increase in Ise was inhibited by 100 microM bumetanide and by Cl- channel inhibitors. The effectiveness of the channel blockers followed the sequence niflumic acid > or = 5-nitro-2-(3-phenylpropylamino)benzoate > diphenylamine-2-carboxylate (DPC) > glibenclamide. The effect of the peptide was not inhibited by 4.4'-diisothiocyanostilbene-2-2'-disulfonic acid. Removing Cl from the bathing solutions also inhibited the effect of the peptide. The Cl- efflux pathway induced by C-K4-M2GlyR differs from the native pathway activated by the adenosine 3',5'-cyclic monophosphate (cAMP) agonist, forskolin. First, intracellular cAMP levels were unaffected. Second, the concentration of DPC required to inhibit the effect of the peptide was much lower than that needed to block the forskolin response (100 microM vs. 3 mM). These results support the hypothesis that the synthetic peptide C-K4-M2GlyR can from Cl -selective channels in the apical membrane of secretory epithelial cells and can induce sustained transepithelial secretion of Cl- and fluid.


Subject(s)
Chloride Channels/drug effects , Chloride Channels/genetics , Glycine/pharmacology , Ion Channel Gating , Kidney/metabolism , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Biological Transport , Body Fluids/metabolism , Bumetanide/pharmacology , Cell Line , Chloride Channels/antagonists & inhibitors , Chlorides/metabolism , Dogs , Electrophysiology , Epithelial Cells , Epithelium/metabolism , Epithelium/physiology , Extracellular Space/metabolism , Kidney/cytology , Kidney/physiology , Molecular Sequence Data , Peptide Fragments/chemical synthesis
18.
Am J Kidney Dis ; 29(3): 435-44, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9041221

ABSTRACT

Woo et al (Nature 368:750-753) reported that parenteral administration of paclitaxel arrested the striking renal enlargement and prolonged life in C57BL/6J-cpk/cpk mice with a rapidly progressive form of polycystic kidney disease (PKD). In the current study, we sought to determine whether paclitaxel could alter the progression of other forms of hereditary PKD in rodents. Paclitaxel was administered by intraperitoneal injection to C57BL/6J-cpk/cpk mice and Han:SPRD-Cy/Cy rats with rapidly progressive PKD and to DBA/2FG-pcy/pcy mice and Han:SPRD-Cy/+ rats with slowly progressive PKD. Paclitaxel (150 micrograms/wk) prolonged the survival of cpk/cpk mice from 24.5 days to more than 65 days and decreased kidney weight relative to body weight from 16.5% at 21 days of age to 8.2% at more than 65 days of age. Mortality attributable to paclitaxel was 12%. By contrast, the administration of paclitaxel (0.1 to 15 mg/kg/wk) to 7- to 10-day-old Han:SPRD-Cy/Cy rats with rapidly progressive PKD had no effect on the course of the disease; moreover, paclitaxel caused severe side effects and premature death in all the Cy/Cy animals. Heterozygous male Cy/+ rats develop slowly progressive renal enlargement and azotemia. Paclitaxel, administered at 7, 15, or 27 mg/kg/wk to male Cy/+ rats from 4 until 10 weeks of age, reduced body weight gain, had an inconsistent effect on kidney weight relative to body weight, and had no effect on the serum urea nitrogen concentration. Mortality associated with the 7, 15, and 27 mg/kg/wk doses of paclitaxel was 0%, 15.4%, and 28.5%, respectively. DBA/2FG-pcy/pcy mice of either sex developed slowly progressive renal enlargement and azotemia. The administration of paclitaxel (100 to 150 micrograms/wk) from 2 to 10 weeks of age to DBA/2FG-pcy/pcy mice with cystic disease had no effect on the increase in kidney weight or on the level of serum urea nitrogen in comparison to untreated cystic animals. Mortality associated with 100- and 150-micrograms/wk doses of paclitaxel was 0% and 20%, respectively. We conclude that paclitaxel diminished the rate of renal enlargement and increased the life span of cpk/cpk mice but not Cy/Cy rats with rapidly progressive forms of PKD. Paclitaxel had no apparent benefit in Cy/+ rats nor pcy/pcy mice with slowly progressive PKD. On the basis of these studies in rodents, it appears that paclitaxel has limited potential usefulness as a therapeutic agent in the treatment of PKD.


Subject(s)
Paclitaxel/therapeutic use , Polycystic Kidney Diseases/drug therapy , Renal Agents/therapeutic use , Animals , Disease Progression , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Heterozygote , Homozygote , Kidney/drug effects , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/mortality , Polycystic Kidney Diseases/pathology , Random Allocation , Rats , Rats, Inbred Strains
20.
Trans Am Clin Climatol Assoc ; 108: 165-70; discussion 170-2, 1997.
Article in English | MEDLINE | ID: mdl-9108675

ABSTRACT

Polycystic kidney disorders are more common than once appreciated, are contributors to significant morbidity, are potentially fatal and are costly to treat. In the past few years much progress has been made toward understanding the pathogenesis of renal cystic disorders. The dominantly inherited disorders are initiated by mutations within genes located in chromosomes 16 and 4 (ADPKD) that cause the kidneys to enlarge several-fold greater than normal. This enlargement is owing to the proliferation of epithelial cells in tubule segments, to the accumulation of fluid within the dilated tubule segment created by the proliferating cells, and to remodelling of the extracellular matrix. The focal beginning of ADPKD in a relatively few renal tubules suggests that the cells in the walls of cysts may reflect clonal growth and that this aberrant proliferation may be secondary to a somatic "second hit" process. The rate at which the cysts enlarge appears to depend on endocrine, paracrine and autocrine factors that drive cellular proliferation and transepithelial fluid secretion within the cysts. The presence of the renal cysts within certain kidneys appears to provoke interstitial inflammation and apoptosis that contribute to fibrosis and renal insufficiency in approximately one-half of persons with ADPKD. Why renal cysts do not cause renal failure in the other one-half of patients with polycystic kidneys is a provocative question that awaits further study.


Subject(s)
Polycystic Kidney, Autosomal Dominant/etiology , Body Fluids/metabolism , Chlorides/metabolism , Cyclic AMP/metabolism , Extracellular Matrix/metabolism , Genotype , Humans , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Polycystic Kidney, Autosomal Dominant/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL