SciELO - Scientific Electronic Library Online

 
vol.73 issue2An apparent life threatening secondary to long Qt syndromeInter-society consensus for the management of respiratory infections: acute bronchitis and chronic obstructive pulmonary disease author indexsubject indexarticles search
Home Pagealphabetic serial listing  

Services on Demand

Journal

Article

Indicators

  • Have no cited articlesCited by SciELO

Related links

  • Have no similar articlesSimilars in SciELO

Share


Medicina (Buenos Aires)

Print version ISSN 0025-7680

Medicina (B. Aires) vol.73 no.2 Ciudad Autónoma de Buenos Aires Apr. 2013

 

ARTÍCULO ESPECIAL

Polycystin-1 C terminus cleavage and its relation with polycystin-2, two proteins involved in polycystic kidney disease

 

Claudia A. Bertuccio1, Michael J. Caplan2

1Department of Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, Pennsylvania USA.
2Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA

Postal address: Dra. Claudia A. Bertuccio, University of Pittsburgh, 3500 Terrace St., S331 BST, Pittsburgh, PA 15261, USA
Fax: 1-(412) 648-8330 e-mail: cab199@pitt.edu

 


Abstract
Autosomal dominant polycystic kidney disease (ADPKD), a most common genetic cause of chronic renal failure, is characterized by the progressive development and enlargement of cysts in kidneys and other organs. The cystogenic process is highly complex and involves a high proliferative rate, increased apoptosis, altered protein sorting, changed secretory characteristics, and disorganization of the extracellular matrix. ADPKD is caused by mutations in the genes encoding polycystin-1 (PC-1) or polycystin-2 (PC-2). PC-1 undergoes multiple cleavages that intervene in several signaling pathways involved in cellular proliferation and differentiation mechanisms. One of these cleavages releases the cytoplasmic C-terminal tail of PC-1. In addition, the C-terminal cytoplasmic tails of PC-1 and PC-2 interact in vitro and in vivo. The purpose of this review is to summarize recent literature that suggests that PC-1 and PC-2 may function through a common signaling pathway necessary for normal tubulogenesis. We hope that a better understanding of PC-1 and PC-2 protein function will lead to progress in diagnosis and treatment for ADPKD.

Key words: Polycystin-1 (PC-1); Polycystin-2 (PC-2); C-terminal tail (CTT) cleavage of polycystin-1; Primary cilium; MTOR; CAMP; Ca2+

Resumen
Clivaje del C-terminal de policistina-1 y su relación con policistina-2, dos proteínas involucradas en la poliquistosis renal. La poliquistosis renal autosómica dominante (ADPKD por sus siglas en inglés) es una causa genética muy común de falla renal crónica que se caracteriza por el progresivo desarrollo y agrandamiento de quistes en los riñones y en otros órganos. El proceso de cistogénesis comprende incrementos en la proliferación y muerte celular por apoptosis, así como alteraciones en la distribución intracelular de proteínas, el movimiento transcelular de solutos y organización de la matriz extracelular. ADPKD es causada por mutaciones en los genes que codifican para policistina-1 (PC-1) o policistina-2 (PC-2). PC-1 puede sufrir múltiples clivajes y los fragmentos generados intervienen en diferentes cascadas de señalización involucradas en mecanismos de proliferación y diferenciación celular. Uno de estos clivajes libera el extremo C-terminal citoplasmático de la PC-1. Se ha demostrado que los extremos C-terminal citoplasmático de PC-1 y PC-2 pueden interactuar tanto in vitro como in vivo. El propósito de esta revisión es resumir la literatura más reciente que sugiere que PC-1 y PC-2 pueden funcionar a través de una cascada de señalización común necesaria para la tubulogénesis normal. Creemos que una mejor comprensión de los mecanismos moleculares de acción de PC-1 y PC-2 contribuirán al progreso en el diagnóstico y tratamiento de ADPKD.

Palabras clave: Policistina-1 (PC-1); Policistina-2 (PC-2); Clivaje del extremo C-terminal (CTT) de policistina-1; Cilia primaria; MTOR; CAMP; Ca2+


 

Polycystin 1 and 2: structure, localization and cleavage

Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common monogenic disorders, and globally is the third most common cause of end stage kidney disease in humans. It is characterized by the slow development of multiple bilateral cysts in the kidneys, resulting in progressive renal failure requiring the need for renal replacement therapy in 50% of the patients by the time they are 60 years old. Furthermore, ADPKD is a multisystem disease that also causes cysts in the liver, pancreas, and other organs, aneurysms of the cerebral artery, cardiac valvular insufficiency and colonic diverticula1. Most cases of ADPKD are caused by mutations in two separate genes, PKD1 and PKD2, which encode the polycystin-1 (PC-1) and polycystin-2 (PC-2) proteins, respectively2, 3.
Regulated intramembrane proteolysis (RIP) is an evolutionarily conserved process by which transmembrane proteins are cleaved to release cytoplasmic domains or luminal/extracellular domains. Released cytosolic domains can translocate to the nucleus to regulate transcription
of genes involved in apoptosis, inflammation, lipid metabolism and neurogenesis. To date, numerous integral membrane proteins have been shown to release a soluble cleavage fragment, and included on this list is PC-14. PC-1 is an integral membrane protein of 4,302 amino acids with a large extracellular region containing sequences with homology to domains that are usually involved in protein-protein or protein-carbohydrate interactions5, 6. This segment is followed by 11 transmembrane (TM) domains and a short cytoplasmic C-terminal domain. Moreover, PC-1 undergoes an autocatalytic cleavage at a site that is homologous to a G-protein coupled receptor proteolytic site (GPS)7. This GPS motif is localized in the extracellular domain immediately proximal to the first TM domain. This cleavage essentially removes the entire extracellular N-terminus of the protein (~350 kDa fragment, NTF) and generates a ~150 kDa C-terminal fragment that is composed of all of the TM domains and the C-terminal tail of PC-1 (CTF). Furthermore, the entire C-terminal tail (CTT) of PC-1 can be proteolytically released from the membrane and can translocate to the nucleus, where it may be able to regulate the expression of genes involved in cellular proliferation and differentiation7-10 (Fig. 1). Finally, Low et al. observed a 17 kDa cleavage product, which is produced by a different cleavage event in the C-terminal half of the PC1 tail. This small fragment is released from the plasma membrane and targets the nucleus in association with STAT transcription factors9 (Fig. 1). In summary, the CTT of PC-1 undergoes two different cleavages, each of which generates fragments that participate in dissimilar signaling processes. These processes will be reviewed below.


Fig. 1.- The predicted topology of polycystin-1 (PC-1). PC-1 has a large extracellular region (NTF) with a highly conserved G protein-coupled receptor proteolytic cleavage site (GPS). This segment is followed by 11 transmembrane (TM) domains and a short cytoplasmic C-terminal domain (CTF). The CTT of PC-1 undergoes two different cleavages, each of which generates fragments that participate in dissimilar signaling processes. E: extracellular space, I: intracellular space. The thick gray rectangle indicates the membrane bilayer.

PC-2 is a 968 amino acid protein that contains six TM domains with an ion conducting pore that resides between the TM5 and TM6 domains. Both the N- and C-termini are predicted to be intracellular12 (Fig. 2). Immunocytochemical colocalization, subcellular fractionation and endoglycosidase H analyses have established that epithelial cells express a large fraction of PC-2 in the membranes of the endoplasmic reticulum (ER)13. It has been demonstrated that the cytoplasmic tail of PC-2 contains signals that are necessary for ER retention13 and in the ER, PC-2 can act as a Ca2+-activated Ca2+-release channel14.


Fig. 2.- The predicted topology of polycystin-2 (PC-2). PC-2 has an ER endoplasmic reticulum retention signal (ER). The short cytoplasmic tail of PC1 contains a coiled-coil region that couples with the COOH-terminal region of PC-2. The PC-1/ PC-2 interaction was implicated in common cellular signaling pathways. Proteins are not drawn to scale. The numbers represent the predicted transmembrane domains. E: extracellular space, I: intracellular space. The thick gray rectangle indicates the membrane bilayer.

Role of polycystin 1/polycystin 2 interaction

Patients with mutations in PKD1 and those with mutations in PKD2 genes have a similar clinical phenotype, and both PKD1-/- and PKD2-/- mice develop kidney and liver cysts resembling the human phenotype15, 16. These studies suggested that PC-1 and PC-2 are implicated in common cellular signaling pathways. Experiments conducted both in vitro and in vivo have shown that PC-2 directly interacts with PC-117-20 (Fig. 2). It has been described that the C-terminal domain of PC-1 contains a coiled-coil motif that binds to the C-terminal domain of PC-2. Hanoaka et al., have provided evidence that the co-expression of PC-1 and PC-2 in mammalian cells results in the appearance of a unique non-selective Ca2+-permeable cation channel on the plasma membrane18, 21, and that the deletion of the carboxyl-terminus of either protein destroys the PC-1/PC-2 interaction and fails to generate Ca2+ currents. In artificial lipid bilayers, recombinant PC-2 can reconstitute Ca2+ channel activity in the absence of PC-1, but PC-1 appears to stabilize or activate PC-2 channel activity in this system22. In addition, co-expression and co-assembly of PC-2 with PC-1 have been shown to dislodge PC-2 from the ER and to allow its relocalization to the cell surface in mammalian cells18. Whether PC-1 can reconstitute channel activity by itself by forming part of a channel pore and/or act as a chaperone protein remains unclear.
The primary cilium is a nonmotile hair-like structure localized in the apical surface of most tubular epithelial cells. It has been observed that both PC-1 and PC-2 localize to the primary cilium of renal epithelial cells and vascular endothelial cells23-25. Recent advances support the hypothesis that in the primary cilium the large extracellular N-terminal domain of PC-1 functions as a sensing antenna for mechanical26 or chemical stimuli27. Mechanical stimuli in the cilia, in response to flow rates comparable to those observed in renal tubules, may induce conformational changes of PC-1, therefore activating its binding partner PC-2, which could mediate Ca2+ entry into the cell. This calcium entry could then amplified by Ca2+ release from IP3- and/or ryanodine-sensitive stores and spread to neighboring cells through gap junction communication23.
The CTT of PC-1 and PC-2 have also been linked to other signaling pathways. The C-terminus of PC-1 has been shown to induce JNK/ activator protein-1 (AP-1) transcription via the activation of heterotrimeric G proteins and PKC a28, while PC-2 expression upregulated JNK/AP-1 upon activation of PKC e29. Additionally, PC-1 activates the Janus kinase (JAK2) and Signal Transducers and Activators of Transcription (STAT)1/3 pathway in a process that requires PC-2 and the up-regulation of p21waf1, leading to cell cycle arrest in G0/G130. The PC1/PC2 complex also interacts with the adherens junction protein E-cadherin and its associated cytoplasmic catenins in lateral membranes of renal tubular cells31, 32. Thus, a delicate relationship between PC-1 and PC-2 may regulate processes involved in the control of different cellular programs such as proliferation, differentiation, apoptosis29, tubulogenesis31, adhesion and maturation32, which are all essential steps of kidney morphogenesis.

Distal proteolytic cleavage of CTT and its role in ADPK

As mentioned above, PC-1 undergoes another proteolytic cleavage in the distal half of the CTT9. The same group proposed that under normal luminal fluid flow conditions, PC-1 localizes to primary cilia, where it is in a complex with P100 and STAT6. Renal injury, during which the luminal fluid flow is strongly decreased or absent, will trigger cleavage of the second half of PC1 tail by a yet unknown mechanism, and this is accompanied by activation of STAT6 by its tyrosine phosphorylation, translocation of the PC-1 tail/STAT6/P100 complex from the cilia to nuclei and induction of gene expression9 (Fig. 1). The same authors showed that the cleaved nuclear PC-1 tail is also prominent in ADPKD cysts together with an upregulation of STAT6 and P1009.
The mTOR (mammalian target of rapamycin) is a conserved serine/threonine kinase that is involved in cell growth regulation, proliferation, regulation of the cellular cytoskeleton and cell survival33-35. The mTOR can be inhibited by the fungal metabolite rapamycin. The two major regulators of the mTOR are TSC1 and TSC2, the genes that encode hamartin and tuberin, respectively and are mutated in Tuberous Sclerosis Complex (TSC)36. The mTOR is activated by the small GTPase Rheb when it is in a GTP-bound state. The tuberin/hamartin complex favors a GTP-to-GDP conversion of Rheb, which results in the inactivation of Rheb and the inhibition of mTOR35. Disruption of the tuberin-hamartin complex signaling, leads to relief of this inhibition and therefore mTOR activation.
Molecular analysis has revealed that, the PKD1 and TSC2 genes are located very close to each other on the human chromosome 1637. Large deletions of chromosome 16 affecting both the TSC2 and the PKD1 gene produce more severe and earlier onset polycystic kidney disease than is seen in patients with PKD1 mutations alone37, 38 and this new syndrome is called TSC/PKD37. In addition, it has been observed that cells that are null for tuberin exhibit a defect in the trafficking of PC1 to the plasma membrane39. These observations suggest that PC1 and tuberin have a functional link and may act together in the same pathway. It has been recently reported that the CTT of PC1 interacts, directly or indirectly, with tuberin and also with mTOR40. In addition, the same authors showed that mTOR is upregulated in the cystic epithelia of ADPKD tissues and PC1-null model40. Treatment of "orpk" mice (a mouse model with a defect in the ciliary polaris gene) or treatment of "bpk" mice (with a defect in the ciliary bicaudal-C gene) with rapamycin resulted in a dramatic inhibition of renal cyst growth and preservation of renal function40. ADPKD patients who underwent renal transplantation and were receiving rapamycin to prevent rejection exhibited a significant reduction in the renal volume of the polycystic kidney that had not been removed40. Interestingly, the damage produced by ureteral obstruction, during which fluid flow essentially ceases and the damage is caused by the proliferative and profibrotic response of the renal epithelial cells, can be prevented by treating the animals with rapamycin41. Lieberthal et al. found that ischemia-reperfusion injury induces dramatic upregulation of mTOR activity in the kidney and that rapamycin treatment prevented the proliferation and induction of apoptosis42. All of these results are consistent with the following model:
1) In normal renal tubule epithelial cells, luminal fluid flow stimulates primary cilia and PC1 remains intact and localized to primary cilia and the lateral plasma membranes. Under those conditions, PC-1 is a constitutive partner of the tuberin/hamartin complex that is required for the inactivation of Rheb and the repression of mTOR43. As long as PC-1 forms a complex with P100 and STAT6 in the primary cilia, mTOR and STAT6 activities are suppressed, resulting in a normal cell proliferation.
2) During ischemic injury or ureteral obstruction, fluid flow essentially ceases. An important function of primary cilia is to sense the lack of luminal fluid flow associated with renal injury. The lack of ciliary mechano-stimulation leads to proteolytic cleavage of the second half of PC1 tail, which translocates to the nucleus. This disruption of PC1 would lead to the inability of CTT PC1 to assemble with tuberin/hamartin inhibitory complex, leading to dysregulation of mTOR. PC1 tail cleavage also results in the cessation of STAT6 inhibition through its retention on the cilia, resulting instead in its translocation to the nuclei and induction of gene expression. The constitutive activation of the mTOR and STAT6 pathways produce cyst growth and an aberrant proliferation required for injury repair9, 40.

Association between the PC1 CTT and both intracellular cAMP and Ca2+ in ADPK

The 'PKD phenotype', which includes enlargement and formation of renal cysts as a consequence of net fluid secretion into the cysts and increased cell proliferation in the cyst wall, were simulated by the overexpression of the entire cytosolic C-terminal tail of PC1 in mouse cortical collecting duct cells44. There is a considerable body of literature providing evidence that cAMP-stimulated chloride transport is the driving force for the fluid secretion associated with cyst filling45-47. Besides, it was demonstrated that cAMP is involved in stimulating the abnormal proliferation of cystic epithelial cells. Studies from Wallace's laboratory demonstrated that primary cells from various ADPKD cysts had significantly lower basal levels of intracellular calcium. These ADPKD cells responded to cAMP with increased cell proliferation. This response to cAMP could be normalized by re-introducing calcium to these cyst epithelial cells48. Additionally, Yamaguchi et al. observed that a combination of restriction in intracellular Ca2+ and elevated cAMP in normal renal epithelial cells can lead to abnormal cell proliferation49 (Fig. 1). These authors observed that in immortalized M-1 collecting duct cells, the calcium restriction caused an inhibition of the phosphatidylinositol 3-kinase/Akt pathway, which relieves the inhibition of B-Raf and allows cAMP to activate PKA, which activates Ras. Ras can then activate the B-Raf/MEK/ERK pathway, causing an increased rate of cell proliferation. M-1 cells stably overexpressing polycystin-1 CTT exhibited a cAMP growth-stimulated phenotype involving B-Raf and ERK activation and it was reversed by the addition of a calcium ionophore.
Interestingly, in ADPKD cyst cells, Xu et al. observed equivalent whole-cell levels of PC1 and PC2 to those found in normal epithelial cells, but the primary cilium in the cyst cells lacked any detectable PC1 and presented reduced PC-2 expression. ADPKD cysts cells also exhibited no flow-sensitive elevation of intracellular Ca2+ concentration at any level of shear stress and were characterized by reduced endoplasmic reticulum Ca2+ stores and reduced capacitative Ca2+ entry50. Igarashi´s group demonstrated that primary cilia of renal epithelial cells contain a cAMP-regulating protein complex, including PC2 and PDE4C. Under normal conditions, PDE4C promotes the hydrolysis of cAMP, and PC2 acts as a Ca2+ entry channel and mediates the local accumulation of Ca2+ that inhibits the activity of Ca2+ -sensitive adenylyl cyclase 5/651. These findings support the concept that ADPKD is associated with a disruption in calcium homeostasis and accumulation of intracellular cAMP and these processes also have a central role in the primary cilium.
Similarly to renal epithelial cells, vascular endothelial cells sense blood flow, which induces an increase in intracellular Ca2+ concentration followed by the endothelial release of nitric oxide leading to vasodilation52. Nauli´s group showed that the primary cilia of endothelial cells respond to shear stress and expresses PC-2 and PC-1. Furthermore, the same group observed that endothelial cells from Pkd2-knockout or Pkd1-knockout mice exhibit impairments in the increase in intracellular Ca2+ concentration and release of nitric oxide induced by flow24, 53. In addition, the PC-1 protein expressed in endothelial cells undergoes a C-terminal cleavage, resulting in loss of receptiveness of cells to high fluid shear24. This study suggests that in patients with high blood pressure, the cilia may be not able to sense changes in blood pressure because PC-1 is proteolytically inactivated by high fluid-shear stress, which may in turn produce a malfunction in the autoregulation of the local circulatory system and may increase the possibility of the localized blood vessel injuries characteristic of ADPKD.

Presence of PC-2 activates PC1 CTT cleavage and modulates cAMP and Ca2+ -independent signaling pathway

In addition to the findings of Weimbs' group, Chauvet et al. observed that blocking the sensing of the urinary flow in vivo, either by occluding a ureter in WT animals or by genetically abolishing cilia formation in renal cyst of epithelial cells, led to an increase in total CTT cleavage, as evidenced by the accumulation of the CTT fragment in the nuclei of renal epithelial cells8. In addition, several groups have shown that PC-2 is expressed at higher levels in cyst-lining epithelium of ADPKD kidneys54-56. As mentioned previously, PC-1 and PC-2 are involved in common signaling pathways and they interact physically with each other17-20. Nevertheless, it still remains unclear whether high levels of PC-2 and/or the interaction of PC-2 with PC-1 may regulate the cleavage and nuclear translocation of the PC-1 CTT.
To pursue these results further, we decided to address some outstanding and fundamental questions: 1) is PC-2 able to modulate the PC-1 CTT cleavage? 2) is the PC-1/PC-2 interaction necessary for the increase in CTT cleavage? And finally 3) is the PC-1 CTT cleavage modulated by the extracellular and/or intracellular calcium and cAMP levels? In order to study the CTT cleavage of the PC-1 protein we generated a fusion protein in which a chimera of the Gal4 transcription factor and the VP16 transcriptional activator was inserted into the full length PC-1 sequence downstream of the intracellular CTT domain (PKDgalvp). Our strategy included transfecting COS-7 cells with PKDgalvp together with a Gal4-driven Firefly luciferase reporter gene. We expected that if the CTT of PC-1 is cleaved, it will translocate to the nucleus, and there it would drive the expression of the luciferase gene that is under the control of the Gal4 promoter (Fig. 3). The magnitude of luciferase expression can be measured using a quantitative luminescence assay57. To determine whether PC-2 increases CTT cleavage and its nuclear traslocation, we coexpressed PC-2 and the PKDgalvp construct in COS-7 cells. We showed that both luciferase activity and the expression of CTT were significantly increased in the presence of PC-2 without changes in PC-1 full length expression.


Fig. 3.- Schematic representation of the PC-1 tagged at its C-terminal tail (CTT) with HA-epitope/galvp and at its N-terminus with a Flag epitope (PKDgalvp), and schematic representation of the luciferase reporter assay. Proteolytic cleavage of the PC-1 fusion protein releases the CTT with the Gal4-VP16, which translocates to the nucleus (N) and activates a Gal4-responsive luciferase reporter gene. Therefore, luciferase expression correlates with PC-1 CTT cleavage. The CTT cleavage requires the presence of PC-2, however, it does not involve the PC-2/PC-1 interaction nor is it regulated by the presence of cAMP nor Ca2+.

To answer our second question, we used a series of truncated mutant constructs of PC-2 that were generated by introducing premature termination codons into the C-terminus of the cDNA encoding PC-2. The PC-2 mutant constructs which were incapable of associating with PC-119 stimulated PC-1 CTT cleavage to the same extent as full length PC-2, suggesting that the interaction between PC-1 and PC-2 is not necessary to elicit PC-1 CTT cleavage.
Finally, as we mentioned above, Ca2+ homeostasis and intracellular cAMP are connected to the CTT PC-1 cleavage in renal epithelial and vascular endothelial cells. Therefore, we wanted to determine whether the Ca2+ channel function of PC-2, Ca2+ and/or cAMP intracellular concentrations are important for CTT cleavage regulation. Thus, we altered extracellular and intracellular Ca2+ levels through several different treatments but we were unable to detect any changes in luciferase activity. Furthermore, when we increased the intracellular levels of cAMP with
forskolin, an adenylate cyclase activator, IBMX, a phosphodiesterase inhibitor or 8-Br-cAMP, an analogue of cAMP, we observed that the PC-2-enhancement of luciferase activity was not altered by any of these treatments (Bertuccio and Caplan, unpublished data).
Taken together, these data suggest a role for PC-2 in the regulation of the cleavage and nuclear translocation of the CTT of PC-1. Although this cleavage requires the presence of PC-2, it does not involve the PC-2/PC-1 interaction nor is it directly regulated by the presence of cAMP nor Ca2+ (Fig. 3), suggesting that the release of the CTT occurs through a novel mechanism that has not been described before.

Discussion

A key event of cystogenesis is the high rate of proliferation and poor differentiation of renal epithelial cells. However, up to now, no specific treatment is available to prevent these processes in ADPKD. The goal in developing therapies for ADPKD is to find treatments that slow cyst enlargement and preserve the renal parenchyma surrounding the cyst, thus conserving kidney function. Several studies have shown that the treatment with rapamycin alleviates cyst enlargement in murine models. Unfortunately, the use of mTOR inhibitors in clinical trials failed to show a clinical benefit in ADPKD patients58, 59.
How the primary cilium, along with PC-1 and PC-2, transmit signals to the nucleus to modulate DNA replication and cell division is not completely understood. We believe that studying and identifying the role and function of PC-2 in this process may help in the understanding of the molecular mechanism responsible for this disease and may help to identify novel therapeutic strategies in the treatment of ADPKD in the future.

Conflicts of interest: No conflicts of interest, financial or otherwise, are declared by the authors.

References

1. Gabow PA. Autosomal dominant polycystic kidney disease. N Engl J Med 1993; 329: 332-42.         [ Links ]

2. Burn TC, Connors TD, Dackowski WR, et al. Analysis of the genomic sequence for the autosomal dominant polycystic kidney disease (PKD1) gene predicts the presence of a leucine-rich repeat. The American PKD1 Consortium (APKD1 Consortium). Hum Mol Genet 1995; 4: 575-82.         [ Links ]

3. Mochizuki T, Wu G, Hayashi T, et al. PKD2, a gene for polycystic kidney disease that encodes an integral membrane protein. Science 1996; 272: 1339-42.         [ Links ]

4. Lal M, Caplan M. Regulated intramembrane proteolysis: signaling pathways and biological functions. Physiology (Bethesda) 2011; 26: 34-44.         [ Links ]

5. Hughes J, Ward C, Peral B, et al. The polycystic kidney disease 1 (PKD1) gene encodes a novel protein with multiple cell recognition domains. Nat Genet 1995; 10: 151-60.         [ Links ]

6. Sandford R, Sgotto B, Aparicio S, et al. Comparative analysis of the polycystic kidney disease 1 (PKD1) gene reveals an integral membrane glycoprotein with multiple evolutionary conserved domains. Hum Mol Genet 1997; 6: 1483-9.         [ Links ]

7. Qian F, Boletta A, Bhunia AK, et al. Cleavage of polycystin-1 requires the receptor for egg jelly domain and is disrupted by human autosomal-dominant polycystic kidney disease 1-associated mutations. Proc Natl Acad Sci USA 2002; 99: 16981-6.         [ Links ]

8. Chauvet V, Tian X, Husson H, et al. Mechanical stimuli induce cleavage and nuclear translocation of the polycystin-1 C terminus. J Clin Invest 2004; 114: 1433-43.         [ Links ]

9. Low SH, Vasanth S, Larson CH, et al. Polycystin-1, STAT6, and P100 function in a pathway that transduces ciliary mechanosensation and is activated in polycystic kidney disease. Dev Cell 2006; 10: 57-69.         [ Links ]

10. Talbot JJ, Shillingford JM, Vasanth S, et al. Polycystin-1 regulates STAT activity by a dual mechanism. Proc Natl Acad Sci USA 2011; 108: 7985-90.         [ Links ]

11. Merrick D, Baggs JE, Yu Z, et al. The ?-secretase cleavage product of Polycystin-1 regulates TCF and CHOP-mediated transcriptional activation through a p300-dependent mechanism. Dev Cell 2012; 22: 197-210.         [ Links ]

12. Parfrey PS, Bear JC, Morgan J, et al. The diagnosis and prognosis of autosomal dominant polycystic kidney disease. New Engl J Med 1990; 323: 1085-90.         [ Links ]

13. Cai Y, Maeda Y, Cedzich A, et al. Identification and characterization of polycystin-2, the PKD2 gene product. J Biol Chem 1999; 274: 28557-65.         [ Links ]

14. Koulen P, Cai Y, Geng L, et al. Polycystin-2 is an intracellular calcium release channel. Cell Biol 2002; 4: 191-7.         [ Links ]

15. Lu W, Zhou J. Perinatal lethality with kidney and pancreas defects in mice with a targeted Pkd1 mutation. Nat Genet 1997; 17: 179-81.         [ Links ]

16. Wu G, D'Agati V, Cai Y, et al. Somatic inactivation of Pkd2 results in polycystic kidney disease. Cell 1998; 93:177-88.         [ Links ]

17. Newby LJ, Streets AJ, Zhao Y, et al. Identification, characterization, and localization of a novel kidney polycystin-1-polycystin-2 complex. J Biol Chem 2002; 277: 20763-73.         [ Links ]

18. Hanaoka K, Qian F, Boletta A, et al. Co-assembly of polycystin-1 and -2 produces unique cation-permeable currents. Nature 2000; 408: 990-4.         [ Links ]

19. Qian F, Germino FJ, Cai Y, et al. PKD1 interacts with PKD2 through a probable coiled-coil domain: Nat Genet 1997; 16: 179-83.         [ Links ]

20. Tsiokas L, Kim E, Arnould T, et al. Homo and heterodimeric interactions between the gene products of PKD1 and PKD2. Proc Natl Acad Sci USA 1997; 94: 6965-70.         [ Links ]

21. González-Perrett S, Kim K, Ibarra C, et al. Polycystin-2, the protein mutated in autosomal dominant polycystic kidney disease (ADPKD), is a Ca2+-permeable nonselective cation channel. Proc Natl Acad Sci USA 2001; 98: 1182-7.         [ Links ]

22. Xu GM, González-Perrett S, Essafi M, et al. Polycystin-1 activates and stabilizes the polycystin-2 channel. J Biol Chem 2003; 278: 1457-62.         [ Links ]

23. Nauli SM, Alenghat FJ, Luo Y, et al. Polycystins 1 and 2 mediate mechanosensation in the primary cilium of kidney cells. Nat Genet 2003; 33: 129-37.         [ Links ]

24. Nauli SM, Kawanabe Y, Kaminski J, et al. Endothelial cilia are fluid shear sensors that regulate calcium signaling and nitric oxide production through polycystin-1. Circulation 2008; 117, 1161-71.         [ Links ]

25. Aboualaiwi W, Takahashi M, Mell B, et al. Ciliary polycystin-2 is a mechanosensitive calcium channel involved in nitric oxide signaling cascades. Circ Res 2009; 104, 860-9.         [ Links ]

26. Praetorius HA, Spring KR. Bending the MDCK cell primary cilium increases intracellular calcium. J Membr Biol 2001; 184: 71-9.         [ Links ]

27. Praetorius HA, Praetorius J, Nielsen S, et al. Beta1-integrins in the primary cilium of MDCK cells potentiate fibronectininduced Ca2+ signaling. Am J Physiol Renal Physiol 2004; 287: F969-78.         [ Links ]

28. Parnell SC, Magenheimer BS, Maser RL, et al. The polycystic kidney disease-1 protein, polycystin-1, binds and activates heterotrimeric G-proteins in vitro. Biochem Biophys Res Comm 1998; 251, 625-31.         [ Links ]

29. Arnould T, Sellin L, Benzing T, et al. Cellular activation triggered by the autosomal dominant polycystic kidney disease gene product PKD2. Mol Cell Biol 1999; 19, 3423-34.         [ Links ]

30. Bhunia AK, Piontek K, Boletta A, et al. PKD1 induces p21(waf1) and regulation of the cell cycle via direct activation of the JAK-STAT signaling pathway in a process requiring PKD2. Cell 2002; 109: 157-68.         [ Links ]

31. Gumbiner BM. Cell adhesion: the molecular basis of tissue architecture and morphogenesis. Cell 1996; 84: 345-57.         [ Links ]

32. van Adelsberg J. Polycystin-1 interacts with E-cadherin and the catenins-clues to the pathogenesis of cyst formation in ADPKD? Nephrol Dial Transplant 2000; 15: 1-2.         [ Links ]

33. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell 2006; 124: 471-84.         [ Links ]

34. Tee AR, Blenis J. mTOR, translational control and human disease. Semin Cell Dev Biol 2005; 16: 29-37.         [ Links ]

35. Huang J, Manning BD. The TSC1-TSC2 complex: a molecular switchboard controlling cell growth. Biochem J 2008; 412: 179-90.         [ Links ]

36. Crino PB, Nathanson KL, Henske EP. The tuberous sclerosis complex. N Engl J Med 2006; 355: 1345-56.         [ Links ]

37. Brook-Carter PT, Peral B, Ward CJ, et al. Deletion of the TSC2 and PKD1 genes associated with severe infantile polycystic kidney disease-a contiguous gene syndrome. Nat Genet 1994; 8: 328-32.         [ Links ]

38. Sampson JR, Maheshwar MM, Aspinwall R, et al. Renal cystic disease in tuberous sclerosis: role of the polycystic kidney disease 1 gene. Am J Hum Genet 1997; 61: 843-51.         [ Links ]

39. Kleymenova E, Ibraghimov-Beskrovnaya O, Kugoh H, et al. Tuberin-dependent membrane localization of polycystin-1: a functional link between polycystic kidney disease and the TSC2 tumor suppressor gene. Mol Cell 2001; 7: 823-32.         [ Links ]

40. Shillingford JM, Murcia NS, Larson CH, et al. From the Cover: The mTOR pathway is regulated by polycystin-1, and its inhibition reverses renal cystogenesis in polycystic kidney disease. Proc Natl Acad Sci USA 2006; 103: 5466-71.         [ Links ]

41. Wu MJ, Wen MC, Chiu YT, et al. Rapamycin attenuates unilateral ureteral obstruction-induced renal fibrosis. Kidney Int 2006; 69: 2029-36.         [ Links ]

42. Lieberthal W, Fuhro R, Andry CC, et al. Rapamycin impairs recovery from acute renal failure: role of cell-cycle arrest and apoptosis of tubular cells. Am J Physiol Renal Physiol 2001; 281: F693-706.         [ Links ]

43. Dere R, Wilson PD, Sandford RN, et al. Carboxy terminal tail of polycystin-1 regulates localization of TSC2 to repress mTOR. PLoS One 2010; 5: e9239.         [ Links ]

44. Sutters M, Yamaguchi T, Maser RL, et al. Polycystin-1 transforms the cAMP growth-responsive phenotype of M-1 cells. Kidney Int 2001; 60: 484-94.         [ Links ]

45. Wallace DP, Grantham JJ, Sullivan LP. Chloride and fluid secretion by cultured human polycystic kidney cells. Kidney Int 1996; 50: 1327-36.         [ Links ]

46. Hanaoka K, Devuyst O, Schwiebert EM, et al. A role for CFTR in human autosomal dominant polycystic kidney disease. Am J Physiol 1996; 270: C389-99.         [ Links ]

47. Sullivan LP, Wallace DP, Grantham JJ. Chloride and fluid secretion in polycystic kidney disease. J Am Soc Nephrol 1998; 9: 903-16.         [ Links ]

48. Yamaguchi T, Hempson SJ, Reif GA, et al. Calcium restores a normal proliferation phenotype in human polycystic kidney disease epithelial cells. J Am Soc Nephrol 2006; 17: 178-87.         [ Links ]

49. Yamaguchi T, Nagao S, Wallace DP, et al. Cyclic AMP activates B-Raf and ERK in cyst epithelial cells from autosomal-dominant polycystic kidneys. Kidney Int 2003; 63: 1983-94.         [ Links ]

50. Xu C, Rossetti S, Jiang L, et al. Human ADPKD primary cyst epithelial cells with a novel, single codon deletion in the PKD1 gene exhibit defective ciliary polycystin localization and loss of flow induced Ca2+ signaling. Am J Physiol Renal Physiol 2007; 292: F930-45.         [ Links ]

51. Choia Y, Suzukia A, Hajarnisa S, et al. Polycystin-2 and phosphodiesterase 4C are components of a ciliary A-kinase anchoring protein complex that is disrupted in cystic kidney diseases. Proc Natl Acad Sci USA 2011; 108: 26, 10679-84.         [ Links ]

52. Hahn C, Schwartz MA. Mechanotransduction in vascular physiology and atherogenesis. Nat Rev Mol Cell Biol 2009; 10: 53-62.         [ Links ]

53. Nauli SM, Jin X, Hierck BP. The mechanosensory role of primary cilia in vascular hypertension. Int J Vasc Med 2011; 376281. doi: 10.1155/2011/376281. Epub 2011 Jun 16.         [ Links ]

54. Palsson R, Sharma CP, Kim K, et al. Characterization and cell distribution of polycystin, the product of autosomal dominant polycystic kidney disease gene 1. Mol Med 1996; 2, 702-11.         [ Links ]

55. Geng L, Segal Y, Peissel B, et al. Identification and localization of polycystin, the PKD1 gene product. J Clin Invest 1996; 98, 2674-82.         [ Links ]

56. Griffin MD, Torres VE, Grande JP, Kumar R. Immunolocalization of polycystin in human tissues and cultured cells. Proc Assoc Am Physicians 1996; 108, 185-97.         [ Links ]

57. Bertuccio CA, Chapin HC, Cai Y, et al. Polycystin-1 C-terminal cleavage is modulated by polycystin-2 expression. J Biol Chem 2009; 284: 21011-26.         [ Links ]

58. Serra AL, Poster D, Kistler AD, et al. Sirolimus and kidney growth in autosomal dominant polycystic kidney disease. N Engl J Med 2010; 363: 820-29.         [ Links ]

59. Walz G, Budde K, Mannaa M, et al. Everolimus in patients with autosomal dominant polycystic kidney disease. N Engl J Med 2010; 363: 830-40.         [ Links ]

Recibido: 23-VIII-2011
Aceptado: 19-XII-2012

Creative Commons License All the contents of this journal, except where otherwise noted, is licensed under a Creative Commons Attribution License