Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Am Soc Nephrol ; 29(10): 2482-2492, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30185468

RESUMEN

BACKGROUND: The major form of autosomal dominant polycystic kidney disease is caused by heterozygous mutations in PKD1, the gene that encodes polycystin-1 (PC1). Unlike PKD1 genes in the mouse and most other mammals, human PKD1 is unusual in that it contains two long polypyrimidine tracts in introns 21 and 22 (2.5 kbp and 602 bp, respectively; 97% cytosine and thymine). Although these polypyrimidine tracts have been shown to form thermodynamically stable segments of triplex DNA that can cause DNA polymerase stalling and enhance the local mutation rate, the efficiency of transcription and splicing across these cytosine- and thymine-rich introns has been unexplored. METHODS: We used RT-PCR and Western blotting (using an mAb to the N terminus) to probe splicing events over exons 20-24 in the mouse and human PKD1 genes as well as Nanopore sequencing to confirm the presence of multiple splice forms. RESULTS: Analysis of PC1 indicates that humans, but not mice, have a smaller than expected protein product, which we call Trunc_PC1. The findings show that Trunc_PC1 is the protein product of abnormal differential splicing across introns 21 and 22 and that 28.8%-61.5% of PKD1 transcripts terminate early. CONCLUSIONS: The presence of polypyrimidine tracts decreases levels of full-length PKD1 mRNA from normal alleles. In heterozygous individuals, low levels of full-length PC1 may reduce polycystin signaling below a critical "cystogenic" threshold.


Asunto(s)
Empalme Alternativo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/biosíntesis , Canales Catiónicos TRPP/genética , Adulto , Animales , Secuencia de Bases , Exones , Femenino , Humanos , Intrones , Masculino , Ratones , Persona de Mediana Edad , Mutación , Terminación de la Cadena Péptídica Traduccional/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Especificidad de la Especie , Canales Catiónicos TRPP/química , Adulto Joven
2.
Stem Cells ; 36(4): 501-513, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29271023

RESUMEN

Autophagy is a process essential for cell survival under stress condition. The patients with autosomal dominant polycystic kidney disease, which is caused by polycystin-1 or polycystin-2 (PKD2) mutation, display cardiovascular abnormalities and dysregulation in autophagy. However, it is unclear whether PKD2 plays a role in autophagy. In the present study, we explored the functional role of PKD2 in autophagy and apoptosis in human embryonic stem cell-derived cardiomyocytes. HES2 hESC line-derived cardiomyocytes (HES2-CMs) were transduced with adenoviral-based PKD2-shRNAs (Ad-PKD2-shRNAs), and then cultured with normal or glucose-free medium for 3 hours. Autophagy was upregulated in HES2-CMs under glucose starvation, as indicated by increased microtubule-associated protein 1 light chain 3-II level in immunoblots and increased autophagosome and autolysosome formation. Knockdown of PKD2 reduced the autophagic flux and increased apoptosis under glucose starvation. In Ca2+ measurement, Ad-PKD2-shRNAs reduced caffeine-induced cytosolic Ca2+ rise. Co-immunoprecipitation and in situ proximity ligation assay demonstrated an increased physical interaction of PKD2 with ryanodine receptor 2 (RyR2) under glucose starvation condition. Furthermore, Ad-PKD2-shRNAs substantially attenuated the starvation-induced activation of AMP-activated protein kinase (AMPK) and inactivation of mammalian target of rapamycin (mTOR). The present study for the first time demonstrates that PKD2 functions to promote autophagy under glucose starvation, thereby protects cardiomyocytes from apoptotic cell death. The mechanism may involve PKD2 interaction with RyR2 to alter Ca2+ release from sarcoplasmic reticulum, consequently modulating the activity of AMPK and mTOR, resulting in alteration of autophagy and apoptosis. Stem Cells 2018;36:501-513.


Asunto(s)
Autofagia , Glucosa/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Miocitos Cardíacos/metabolismo , Canales Catiónicos TRPP/biosíntesis , Apoptosis , Línea Celular , Glucosa/genética , Células Madre Embrionarias Humanas/citología , Humanos , Miocitos Cardíacos/citología , Canales Catiónicos TRPP/genética
3.
Nat Mater ; 16(11): 1112-1119, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28967916

RESUMEN

Polycystic kidney disease (PKD) is a life-threatening disorder, commonly caused by defects in polycystin-1 (PC1) or polycystin-2 (PC2), in which tubular epithelia form fluid-filled cysts. A major barrier to understanding PKD is the absence of human cellular models that accurately and efficiently recapitulate cystogenesis. Previously, we have generated a genetic model of PKD using human pluripotent stem cells and derived kidney organoids. Here we show that systematic substitution of physical components can dramatically increase or decrease cyst formation, unveiling a critical role for microenvironment in PKD. Removal of adherent cues increases cystogenesis 10-fold, producing cysts phenotypically resembling PKD that expand massively to 1-centimetre diameters. Removal of stroma enables outgrowth of PKD cell lines, which exhibit defects in PC1 expression and collagen compaction. Cyclic adenosine monophosphate (cAMP), when added, induces cysts in both PKD organoids and controls. These biomaterials establish a highly efficient model of PKD cystogenesis that directly implicates the microenvironment at the earliest stages of the disease.


Asunto(s)
Microambiente Celular , Modelos Biológicos , Organoides/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Línea Celular , AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Humanos , Organoides/patología , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Canales Catiónicos TRPP/biosíntesis , Canales Catiónicos TRPP/genética
4.
Anticancer Drugs ; 28(8): 852-860, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28723865

RESUMEN

Systemic therapy with cytotoxic agents remains one of the main treatment methods for non-small-cell lung cancer (NSCLC). Cisplatin is a commonly used chemotherapeutic agent, that, when combined with other drugs, is an effective treatment for NSCLC. However, effective cancer therapy is hindered by a patient's resistance to cisplatin. Unfortunately, the potential mechanism underlying such resistance remains unclear. In this study, we explored the mechanism of microRNA-106b-5p (miR-106b-5p), which is involved in the resistance to cisplatin in the A549 cell line of NSCLC. Quantitative real-time PCR was used to test the expression of miR-106-5p in the A549 and the A549/DDP cell line of NSCLC. The cell counting kit-8 assay was used to detect cell viability. Flow cytometry was used to measure cell cycle and cell apoptosis. Luciferase reporter assays and western blot were performed to confirm whether polycystic kidney disease-2 (PKD2) is a direct target gene of miR-106b-5p. Immunohistochemistry was performed to examine the distribution of PKD2 expression in patients who are sensitive and resistant to cisplatin. The experiments indicated that the expression of miR-106b-5p was significantly decreased in A549/DDP compared with that in A549. MiR-106b-5p affected the tolerance of cells to cisplatin by negatively regulating PKD2. Upregulation of miR-106b-5p or downregulation of PKD2 expression can cause A549/DDP cells to become considerably more sensitive to cisplatin. The results showed that miR-106b-5p enhanced the sensitivity of A549/DDP cells to cisplatin by targeting the expression of PKD2. These findings suggest that the use of miR-106b-5p may be a promising clinical strategy in the treatment of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Cisplatino/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , MicroARNs/biosíntesis , Canales Catiónicos TRPP/biosíntesis , Células A549 , Antineoplásicos/farmacología , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación hacia Abajo , Resistencia a Antineoplásicos , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/genética , MicroARNs/genética , Terapia Molecular Dirigida , Canales Catiónicos TRPP/genética
5.
J Clin Invest ; 127(5): 1772-1785, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28375157

RESUMEN

Dominantly inherited isolated polycystic liver disease (PCLD) consists of liver cysts that are radiologically and pathologically identical to those seen in autosomal dominant polycystic kidney disease, but without clinically relevant kidney cysts. The causative genes are known for fewer than 40% of PCLD index cases. Here, we have used whole exome sequencing in a discovery cohort of 102 unrelated patients who were excluded for mutations in the 2 most common PCLD genes, PRKCSH and SEC63, to identify heterozygous loss-of-function mutations in 3 additional genes, ALG8, GANAB, and SEC61B. Similarly to PRKCSH and SEC63, these genes encode proteins that are integral to the protein biogenesis pathway in the endoplasmic reticulum. We inactivated these candidate genes in cell line models to show that loss of function of each results in defective maturation and trafficking of polycystin-1, the central determinant of cyst pathogenesis. Despite acting in a common pathway, each PCLD gene product demonstrated distinct effects on polycystin-1 biogenesis. We also found enrichment on a genome-wide basis of heterozygous mutations in the autosomal recessive polycystic kidney disease gene PKHD1, indicating that adult PKHD1 carriers can present with clinical PCLD. These findings define genetic and biochemical modulators of polycystin-1 function and provide a more complete definition of the spectrum of dominant human polycystic diseases.


Asunto(s)
Quistes , Glucosiltransferasas , Heterocigoto , Hepatopatías , Mutación , Canales de Translocación SEC , Canales Catiónicos TRPP , Adulto , Animales , Proteínas de Unión al Calcio , Línea Celular Transformada , Quistes/genética , Quistes/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Femenino , Estudio de Asociación del Genoma Completo , Glucosidasas/genética , Glucosidasas/metabolismo , Glucosiltransferasas/genética , Glucosiltransferasas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hepatopatías/genética , Hepatopatías/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Chaperonas Moleculares , Proteínas de Unión al ARN , Canales de Translocación SEC/genética , Canales de Translocación SEC/metabolismo , Canales Catiónicos TRPP/biosíntesis , Canales Catiónicos TRPP/genética
6.
Oncotarget ; 8(8): 12800-12811, 2017 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-28077787

RESUMEN

In prostate cancer, androgen/androgen receptor (AR) and their downstream targets play key roles in all stages of disease progression. The protein kinase D (PKD) family, particularly PKD1, has been implicated in prostate cancer biology. Here, we examined the cross-regulation of PKD1 by androgen signaling in prostate cancer cells. Our data showed that the transcription of PKD1 was repressed by androgen in androgen-sensitive prostate cancer cells. Steroid depletion caused up regulation of PKD1 transcript and protein, an effect that was reversed by the AR agonist R1881 in a time- and concentration-dependent manner, thus identifying PKD1 as a novel androgen-repressed gene. Kinetic analysis indicated that the repression of PKD1 by androgen required the induction of a repressor protein. Furthermore, inhibition or knockdown of AR reversed AR agonist-induced PKD1 repression, indicating that AR was required for the suppression of PKD1 expression by androgen. Downstream of AR, we identified fibroblast growth factor receptor substrate 2 (FRS2) and its downstream MEK/ERK pathway as mediators of androgen-induced PKD1 repression. In summary, PKD1 was identified as a novel androgen-suppressed gene and could be downregulated by androgen through a novel AR/FRS2/MEK/ERK pathway. The upregulation of prosurvival PKD1 by anti-androgens may contribute to therapeutic resistance in prostate cancer treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Andrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/patología , Canales Catiónicos TRPP/biosíntesis , Western Blotting , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Androgénicos/metabolismo , Transducción de Señal/fisiología
7.
Neurochem Res ; 41(7): 1784-96, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27038933

RESUMEN

Retinal glial (Müller) cells possess an endogenous purinergic signal transduction cascade which normally prevents cellular swelling in osmotic stress. The cascade can be activated by osmotic or glutamate receptor-dependent ATP release. We determined whether activation of this cascade is altered in Müller cells of transgenic rats that suffer from a slow photoreceptor degeneration due to the expression of a truncated human cilia gene polycystin-2 (CMV-PKD21/703 HA). Age-matched Sprague-Dawley rats served as control. Retinal slices were superfused with a hypoosmotic solution (60 % osmolarity). Müller cells in retinas of PKD21/703 rats swelled immediately in hypoosmotic stress; this was not observed in control retinas. Pharmacological blockade of P2Y1 or adenosine A1 receptors induced osmotic swelling of Müller cells from control rats. The swelling induced by the P2Y1 receptor antagonist was mediated by induction of oxidative-nitrosative stress, mitochondrial dysfunction, production of inflammatory lipid mediators, and a sodium influx from the extracellular space. Exogenous VEGF or glutamate prevented the hypoosmotic swelling of Müller cells from PKD21/703 rats; this effect was mediated by activation of the purinergic signaling cascade. In neuroretinas of PKD21/703 rats, the gene expression levels of P2Y1 and A1 receptors, pannexin-1, connexin 45, NTPDases 1 and 2, and various subtypes of nucleoside transporters are elevated compared to control. The data may suggest that the osmotic swelling of Müller cells from PKD21/703 rats is caused by an abrogation of the osmotic ATP release while the glutamate-induced ATP release is functional. In the normal retina, ATP release and autocrine P2Y1 receptor activation serve to inhibit the induction of oxidative-nitrosative stress, mitochondrial dysfunction, and production of inflammatory lipid mediators, which otherwise will induce a sodium influx and cytotoxic Müller cell swelling under anisoosmotic conditions. Purinergic receptors may represent a target for the protection of retinal glial cells from mitochondrial oxidative stress.


Asunto(s)
Tamaño de la Célula , Células Ependimogliales/metabolismo , Receptor de Adenosina A1/fisiología , Receptores Purinérgicos P2Y1/fisiología , Retina/metabolismo , Canales Catiónicos TRPP/biosíntesis , Animales , Células Ependimogliales/patología , Regulación de la Expresión Génica , Humanos , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Retina/patología , Canales Catiónicos TRPP/genética
8.
Prostate ; 76(3): 245-58, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26764245

RESUMEN

BACKGROUND: African-American men with prostate cancer (PCa) present with higher-grade and -stage tumors compared to Caucasians. While the disparity may result from multiple factors, a biological basis is often strongly suspected. Currently, few well-characterized experimental model systems are available to study the biological basis of racial disparity in PCa. We report a validated in vitro cell line model system that could be used for the purpose. METHODS: We assembled a PCa cell line model that included currently available African-American PCa cell lines and LNCaP (androgen-dependent) and C4-2 (castration-resistant) Caucasian PCa cells. The utility of the cell lines in studying the biological basis of variance in a malignant phenotype was explored using a multiplex biomarker panel consisting of proteins that have been proven to play a role in the progression of PCa. The panel expression was evaluated by Western blot and RT-PCR in cell lines and validated in human PCa tissues by RT-PCR. As proof-of-principle to demonstrate the utility of our model in functional studies, we performed MTS viability assays and molecular studies. RESULTS: The dysregulation of the multiplex biomarker panel in primary African-American cell line (E006AA) was similar to metastatic Caucasian cell lines, which would suggest that the cell line model could be used to study an inherent aggressive phenotype in African-American men with PCa. We had previously demonstrated that Protein kinase D1 (PKD1) is a novel kinase that is down regulated in advanced prostate cancer. We established the functional relevance by over expressing PKD1, which resulted in decreased proliferation and epithelial mesenchymal transition (EMT) in PCa cells. Moreover, we established the feasibility of studying the expression of the multiplex biomarker panel in archived human PCa tissue from African-Americans and Caucasians as a prelude to future translational studies. CONCLUSION: We have characterized a novel in vitro cell line model that could be used to study the biological basis of disparity in PCa between African-Americans and Caucasians.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Negro o Afroamericano , Neoplasias de la Próstata/metabolismo , Canales Catiónicos TRPP/biosíntesis , Población Blanca , Negro o Afroamericano/genética , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Proliferación Celular/fisiología , Transición Epitelial-Mesenquimal/fisiología , Humanos , Masculino , Neoplasias de la Próstata/genética , Canales Catiónicos TRPP/genética , Población Blanca/genética
9.
Eur J Histochem ; 60(4): 2711, 2016 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-28076932

RESUMEN

The vascular smooth muscle cell (VSMC) phenotypic switch is a key pathophysiological change in various cardiovascular diseases, such as aortic dissection (AD), with a high morbidity. Polycystin-1 (PC1) is significantly downregulated in the VSMCs of AD patients. PC1 is an integral membrane glycoprotein and kinase that regulates different biological processes, including cell proliferation, apoptosis, and cell polarity. However, the role of PC1 in intracellular signaling pathways remains poorly understood. In this study, PC1 downregulation in VSMCs promoted the expression of SM22α, ACTA2 and calponin 1 (CNN1) proteins. Furthermore, PC1 downregulation in VSMCs upregulated phospho-MEK, phospho-ERK and myc, but did not change phospho-JNK and phospho-p38. These findings suggest that the MEK/ERK/myc signaling pathway is involved in PC1-mediated human VSMC phenotypic switch. Opposite results were observed when an ERK inhibitor was used in VSMCs downregulated by PC1. When the C-terminal domain of PC1 (PC1 C-tail) was overexpressed in VSMCs, the expression levels of phosphor-ERK, myc, SM22α, ACTA2 and CNN1 proteins were downregulated. The group with the overexpressed mutant protein (S4166A) in the PC1 C-tail showed similar results to the group with the downregulated PC1 in VSMCs. These results suggest that the Ser at the 4166 site in PC1 is crucial in the PC1 mediated MEK/ERK/myc signaling pathway, which might be the key pathophysiological cause of AD.


Asunto(s)
Rotura de la Aorta/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Canales Catiónicos TRPP/biosíntesis , Rotura de la Aorta/patología , Femenino , Humanos , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fosforilación
10.
J Clin Invest ; 125(5): 1955-67, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25844898

RESUMEN

The HSP40 cochaperone SEC63 is associated with the SEC61 translocon complex in the ER. Mutations in the gene encoding SEC63 cause polycystic liver disease in humans; however, it is not clear how altered SEC63 influences disease manifestations. In mice, loss of SEC63 induces cyst formation both in liver and kidney as the result of reduced polycystin-1 (PC1). Here we report that inactivation of SEC63 induces an unfolded protein response (UPR) pathway that is protective against cyst formation. Specifically, using murine genetic models, we determined that SEC63 deficiency selectively activates the IRE1α-XBP1 branch of UPR and that SEC63 exists in a complex with PC1. Concomitant inactivation of both SEC63 and XBP1 exacerbated the polycystic kidney phenotype in mice by markedly suppressing cleavage at the G protein-coupled receptor proteolysis site (GPS) in PC1. Enforced expression of spliced XBP1 (XBP1s) enhanced GPS cleavage of PC1 in SEC63-deficient cells, and XBP1 overexpression in vivo ameliorated cystic disease in a murine model with reduced PC1 function that is unrelated to SEC63 inactivation. Collectively, the findings show that SEC63 function regulates IRE1α/XBP1 activation, SEC63 and XBP1 are required for GPS cleavage and maturation of PC1, and activation of XBP1 can protect against polycystic disease in the setting of impaired biogenesis of PC1.


Asunto(s)
ADN Helicasas/fisiología , Proteínas de Unión al ADN/fisiología , Endorribonucleasas/metabolismo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Recesivo/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Canales Catiónicos TRPP/deficiencia , Factores de Transcripción/fisiología , Respuesta de Proteína Desplegada/fisiología , Animales , Línea Celular , ADN Helicasas/deficiencia , ADN Helicasas/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Glucosidasas/deficiencia , Glucosidasas/genética , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Chaperonas Moleculares , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Recesivo/metabolismo , Estructura Terciaria de Proteína , Empalme del ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción del Factor Regulador X , Canales Catiónicos TRPP/biosíntesis , Canales Catiónicos TRPP/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transfección , Proteína 1 de Unión a la X-Box
11.
Biochem Cell Biol ; 92(6): 547-54, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25367197

RESUMEN

Mutations in polycystin-1, polycystin-2, or fibrocystin account for autosomal dominant or recessive polycystic kidney disease. Renal cystogenesis is linked to abnormal localization and function of these cystoproteins in renal primary cilia. They are also expressed in extrarenal tissues in which their functions are unclear. Here we found that human type-II alveolar epithelial A549, airway submucosal Calu-3 cells, and rat bronchioles contain primary or multiple cilia in which we detected these cystoproteins. At sub-confluency, polycystin-1 was expressed on plasma membrane, while polycystin-2 was localized to the ER of resting cells. Both polycystins were detected on the spindle and mid-body of mitotic cells, while fibrocystin was on centrosome throughout cell cycle. Polycystins and fibrocystin may participate in regulating mucociliary sensing and transport within pulmonary airways.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Pulmón/metabolismo , Receptores de Superficie Celular/biosíntesis , Canales Catiónicos TRPP/biosíntesis , Animales , Ciclo Celular/fisiología , Línea Celular Tumoral , Centrosoma/metabolismo , Cilios/genética , Cilios/metabolismo , Cricetinae , Humanos , Pulmón/citología , Ratas , Receptores de Superficie Celular/genética , Canales Catiónicos TRPP/genética
12.
Oncotarget ; 5(16): 6867-84, 2014 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-25149539

RESUMEN

Over 80% of colon cancer development and progression is a result of the dysregulation of ß-catenin signaling pathway. Herein, for the first time, we demonstrate that a serine-threonine kinase, Protein Kinase D1 (PKD1), modulates the functions of ß-catenin to suppress colon cancer growth. Analysis of normal and colon cancer tissues reveals downregulation of PKD1 expression in advanced stages of colon cancer and its co-localization with ß-catenin in the colon crypts. This PKD1 downregulation corresponds with the aberrant expression and nuclear localization of ß-catenin. In-vitro investigation of the PKD1-ß-catenin interaction in colon cancer cells reveal that PKD1 overexpression suppresses cell proliferation and clonogenic potential and enhances cell-cell aggregation. We demonstrate that PKD1 directly interacts with ß-catenin and attenuates ß-catenin transcriptional activity by decreasing nuclear ß-catenin levels. Additionally, we show that inhibition of nuclear ß-catenin transcriptional activity is predominantly influenced by nucleus targeted PKD1. This subcellular modulation of ß-catenin results in enhanced membrane localization of ß-catenin and thereby increases cell-cell adhesion. Studies in a xenograft mouse model indicate that PKD1 overexpression delayed tumor appearance, enhanced necrosis and lowered tumor hypoxia. Overall, our results demonstrate a putative tumor-suppressor function of PKD1 in colon tumorigenesis via modulation of ß-catenin functions in cells.


Asunto(s)
Neoplasias del Colon/metabolismo , Factores de Transcripción TCF/metabolismo , Canales Catiónicos TRPP/metabolismo , beta Catenina/metabolismo , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Neoplasias del Colon/enzimología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Regulación hacia Abajo , Células HT29 , Xenoinjertos , Humanos , Masculino , Ratones , Transducción de Señal , Fracciones Subcelulares/enzimología , Fracciones Subcelulares/metabolismo , Canales Catiónicos TRPP/biosíntesis
13.
FASEB J ; 27(12): 4998-5009, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23985800

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 or PKD2, and it affects over 10 million people worldwide. It is characterized by cyst formation in the kidney, liver and pancreas. Dosage changes in PKD1/PKD2 are important in ADPKD pathogenesis; therefore, their expression and function has to be strictly regulated. However, how they are regulated remain poorly understood. Recent studies have linked PKD2 regulation to endoplasmic reticulum (ER) stress that is implicated in neuronal, cardiac, and renal diseases. One major ER stress downstream is phosphorylation of eukaryotic initiation factor eIF2α by kinase PERK, which attenuates global protein translation and enhances translation of selected proteins. Here, we showed in several mammalian cell lines that PKD2 protein expression is up-regulated by different stresses that all increase phosphorylated eIF2α (P-eIF2α). Increasing P-eIF2α by overexpression or inhibiting the phosphatase activity resulted in increased PKD2. PCR and polysome-binding assays showed that ER stress does not affect the PKD2 mRNA level but increase its binding with ribosomes, indicating that P-eIF2α translationally up-regulates PKD2. By mutation analysis, we found that the upstream open reading frame (uORF) in the 5'-untranslated region of PKD2 mRNA represses PKD2 translation. Thus, ER stress and P-eIF2α translationally up-regulates PKD2 through bypassing the inhibitory uORF.


Asunto(s)
Estrés del Retículo Endoplásmico , Biosíntesis de Proteínas , Canales Catiónicos TRPP/biosíntesis , Regulación hacia Arriba , Regiones no Traducidas 5' , Animales , Perros , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Células HEK293 , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Mutación , Polirribosomas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canales Catiónicos TRPP/genética
14.
Am J Physiol Renal Physiol ; 303(5): F689-99, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22759397

RESUMEN

Hepatocyte nuclear factor-1ß (HNF-1ß) is a transcription factor expressed in the kidney, liver, pancreas, and other organs. Mutations of HNF-1ß cause maturity-onset diabetes of the young type 5 (MODY5). The aims of this study were to investigate the functional roles of the HNF-1ß/suppressor of cytokine signaling-3 (SOCS-3) pathway in tubule damage after acute kidney injury (AKI) both in vivo and in vitro and to examine the effect of HNF-1ß on renal tubule formation. To clarify the significance of the HNF-1ß/SOCS-3 pathway in AKI, we used a rat ischemia/reperfusion (I/R) AKI model and cultured renal tubular cells (NRK-52E cells). Western blot analysis showed that HNF-1ß and polycystic kidney disease 2 (PKD2) expressions were increased at 3-12 h and 12-24 h after I/R, respectively. The expression level of SOCS-3 was decreased at 3-48 h. Immunohistological examination revealed that expression of HNF-1ß was increased in proximal tubules. Overexpression of HNF-1ß resulted in decreased SOCS-3 expression, activation of signal transducer and activator of transcription 3 (STAT3) and Erk, and increased [(3)H]thymidine uptake in the presence of hepatocyte growth factor. Furthermore, tubule formation in three-dimensional gels was inhibited by dominant-negative HNF-1ß. Our study shows that HNF-1ß is upregulated after AKI in proximal tubular cells and that HNF-1ß controls cellular proliferation and tubule formation by regulating SOCS-3 expression and STAT3/Erk activation. Therefore, the current study unravels the physiological and pathological significance of the HNF-1ß pathway in AKI.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Factor Nuclear 1-beta del Hepatocito/genética , Túbulos Renales/fisiología , Regeneración , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor Nuclear 1-beta del Hepatocito/biosíntesis , Masculino , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/fisiopatología , Factor de Transcripción STAT3/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Canales Catiónicos TRPP/biosíntesis , Regulación hacia Arriba
15.
J Neurochem ; 119(3): 497-506, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21883212

RESUMEN

Polycystic kidney disease 1-like 3 (Pkd1l3) is expressed specifically in sour-sensing type III taste cells that have synaptic contacts with afferent nerve fibers in circumvallate (CvP) and foliate papillae (FoP) located in the posterior region of the tongue, although not in fungiform papillae (FuP) or the palate. To visualize the gustatory neural pathways that originate from type III taste cells in CvP and FoP, we established transgenic mouse lines that express the transneuronal tracer wheat germ agglutinin (WGA) under the control of the mouse Pkd1l3 gene promoter/enhancer. The WGA transgene was accurately expressed in Pkd1l3-expressing type III taste cells in CvP and FoP. Punctate WGA protein signals appeared to be detected specifically in type III taste cells but not in other types of taste cells. WGA protein was transferred primarily to a subset of neurons located in close proximity to the glossopharyngeal (GL) nerve bundles in the nodose/petrosal ganglion (NPG). WGA signals were also observed in a small population of neurons in the geniculate ganglion (GG). This result demonstrates the anatomical connection between taste receptor cells (TRCs) in the FoP and the chorda tympani (CT) nerves. WGA protein was further conveyed to neurons in a rostro-central subdivision of the nucleus of the solitary tract (NST). These findings demonstrate that the approximately 10 kb 5'-flanking region of the mouse Pkd1l3 gene functions as a type III taste cell-specific promoter/enhancer. In addition, experiments using the pkd1l3-WGA transgenic mice reveal a sour gustatory pathway that originates from TRCs in the posterior region of the tongue.


Asunto(s)
Canales Catiónicos TRPP/biosíntesis , Papilas Gustativas/citología , Papilas Gustativas/metabolismo , Gusto/genética , Lengua/citología , Lengua/fisiología , Animales , Canales de Calcio , Regulación del Desarrollo de la Expresión Génica , Ganglio Geniculado/química , Ganglio Geniculado/citología , Ganglio Geniculado/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/química , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Ganglio Nudoso/química , Ganglio Nudoso/citología , Ganglio Nudoso/fisiología , Canales Catiónicos TRPP/química , Canales Catiónicos TRPP/genética , Papilas Gustativas/química , Lengua/química , Aglutininas del Germen de Trigo/biosíntesis , Aglutininas del Germen de Trigo/química , Aglutininas del Germen de Trigo/genética
16.
Hypertens Res ; 33(1): 37-42, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19893564

RESUMEN

The mechanisms underlying vascular complications in autosomal-dominant polycystic kidney disease (ADPKD) have not been fully elucidated. However, molecular components altered in Pkd mutant vascular smooth muscle cells (VSMCs) are gradually being identified. Pkd2(+/-) arterial smooth muscles show elevated levels of (1) phenylephrine (PE)-induced, Ca(2+)-independent vasocontraction and (2) smooth muscle alpha-actin (SMA) expression. As these two processes are heavily influenced by RhoA signaling and by cellular filamentous-to-globular (F/G)-actin dynamics, we examined PE-induced changes in RhoA activation and the F/G-SMA ratio in wild-type (wt) and Pkd2(+/-) VSMCs; we further tested the hypothesis that the abnormal response to PE and the resultant elevation in the F/G-SMA ratio contribute to the exuberant SMA expression in Pkd2(+/-) VSMCs. GTP-RhoA and F/G-SMA in mouse aortic media and primary cultured VSMCs were determined using RhoA activation and in vivo F-to-G-actin assays. Myocardin-related transcription factor-A (MRTF-A) (SMA transcription coactivator) was localized by immunofluorescence, nuclear MRTF-A quantified by western analysis using nuclear extracts and SMA expression by luciferase reporter assay. PE induced a >3-fold higher RhoA activation in Pkd2(+/-) than in wt VSMCs and higher levels of downstream p-LIMK and p-cofilin. Moreover, Pkd2(+/-) VSMCs showed a higher baseline and PE-induced F/G-SMA ratio. The F/G-SMA elevation enhanced nuclear translocation of MRTF-A, which upregulated SMA transcription. In summary, PE-induced RhoA hyperactivation and defects in F-to-G SMA balance likely have a role in the abnormal vasocontraction and SMA expression in Pkd2(+/-) arteries. These defects could potentially contribute to the genesis of vascular complications in ADPKD, thus providing new areas for further research and therapeutic targeting.


Asunto(s)
Actinas/biosíntesis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fenilefrina/farmacología , Canales Catiónicos TRPP/biosíntesis , Vasoconstrictores/farmacología , Proteína de Unión al GTP rhoA/metabolismo , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Técnica del Anticuerpo Fluorescente Indirecta , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Nucleares/metabolismo , Canales Catiónicos TRPP/genética , Transactivadores/metabolismo
17.
J Biol Chem ; 284(11): 7214-22, 2009 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-19098310

RESUMEN

The pathogenic mechanisms of human autosomal dominant polycystic kidney disease (ADPKD) have been well known to include the mutational inactivation of PKD2. Although haploinsufficiency and loss of heterozygosity at the Pkd2 locus can cause cyst formation in mice, polycystin-2 is frequently expressed in the renal cyst of human ADPKD, raising the possibility that deregulated activation of PKD2 may be associated with the cystogenesis of human ADPKD. To determine whether increased PKD2 expression is physiologically pathogenic, we generated PKD2-overexpressing transgenic mice. These mice developed typical renal cysts and an increase of proliferation and apoptosis, which are reflective of the human ADPKD phenotype. These manifestations were first observed at six months, and progressed with age. In addition, we found that ERK activation was induced by PKD2 overexpression via B-Raf signaling, providing a possible molecular mechanism of cystogenesis. In PKD2 transgenic mice, B-Raf/MEK/ERK sequential signaling was up-regulated. Additionally, the transgenic human polycystin-2 partially rescues the lethality of Pkd2 knock-out mice and therefore demonstrates that the transgene generated a functional product. Functional strengthening or deregulated activation of PKD2 may be a direct cause of ADPKD. The present study provides evidence for an in vivo role of overexpressed PKD2 in cyst formation. This transgenic mouse model should provide new insights into the pathogenic mechanism of human ADPKD.


Asunto(s)
Quistes/metabolismo , Riñón/metabolismo , Mutación , Riñón Poliquístico Autosómico Dominante/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal , Canales Catiónicos TRPP/biosíntesis , Animales , Apoptosis/genética , Proliferación Celular , Quistes/genética , Quistes/patología , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Riñón/patología , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fenotipo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Proteínas Proto-Oncogénicas B-raf/genética , Canales Catiónicos TRPP/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...