Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 17(4): e1009417, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33861800

RESUMEN

Macrophages are important drivers of pathogenesis and progression to AIDS in HIV infection. The virus in the later phases of the infection is often predominantly macrophage-tropic and this tropism contributes to a chronic inflammatory and immune activation state that is observed in HIV patients. Pattern recognition receptors of the innate immune system are the key molecules that recognise HIV and mount the inflammatory responses in macrophages. The innate immune response against HIV-1 is potent and elicits caspase-1-dependent pro-inflammatory cytokine production of IL-1ß and IL-18. Although, NLRP3 has been reported as an inflammasome sensor dictating this response little is known about the pattern recognition receptors that trigger the "priming" signal for inflammasome activation, the NLRs involved or the HIV components that trigger the response. Using a combination of siRNA knockdowns in monocyte derived macrophages (MDMs) of different TLRs and NLRs as well as chemical inhibition, it was demonstrated that HIV Vpu could trigger inflammasome activation via TLR4/NLRP3 leading to IL-1ß/IL-18 secretion. The priming signal is triggered via TLR4, whereas the activation signal is triggered by direct effects on Kv1.3 channels, causing K+ efflux. In contrast, HIV gp41 could trigger IL-18 production via NAIP/NLRC4, independently of priming, as a one-step inflammasome activation. NAIP binds directly to the cytoplasmic tail of HIV envelope protein gp41 and represents the first non-bacterial ligand for the NAIP/NLRC4 inflammasome. These divergent pathways represent novel targets to resolve specific inflammatory pathologies associated with HIV-1 infection in macrophages.


Asunto(s)
Infecciones por VIH/virología , Inflamasomas/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/virología , Fragmentos de Péptidos/metabolismo , Comunicación Celular/genética , Comunicación Celular/inmunología , Expresión Génica/genética , Expresión Génica/inmunología , Infecciones por VIH/metabolismo , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inflamasomas/metabolismo , Macrófagos/inmunología , Proteína Inhibidora de la Apoptosis Neuronal/genética , Transducción de Señal/inmunología
2.
Am J Transplant ; 21(6): 2188-2199, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33098231

RESUMEN

Transplantation is the optimal treatment for most patients with end-stage kidney disease but organ shortage is a major challenge. Normothermic machine perfusion (NMP) has been used to recondition marginal organs; however, mechanisms by which NMP might benefit organs are not well understood. Using pairs of human kidneys obtained from the same donor, we compared the effect of NMP with that of cold storage on the global kidney transcriptome. We found that cold storage led to a global reduction in gene expression, including inflammatory pathway genes and those required for energy generation processes, such as oxidative phosphorylation (OXPHOS). In contrast, during NMP, there was marked upregulation OXPHOS genes, but also of a number of immune and inflammatory pathway genes. Using biopsies from kidneys undergoing NMP that were subsequently transplanted, we found that higher inflammatory gene expression occurred in organs with prolonged delayed graft function (DGF). Therefore, we used a hemoadsorber (HA) to remove pro-inflammatory cytokines. This attenuated inflammatory gene expression increased OXPHOS pathway genes and had potentially clinically important effects in reducing the expression of a DGF-associated gene signature. Together, our data suggest that adsorption of pro-inflammatory mediators from the perfusate represents a potential intervention which may improve organ viability.


Asunto(s)
Funcionamiento Retardado del Injerto , Trasplante de Riñón , Citocinas/genética , Funcionamiento Retardado del Injerto/genética , Supervivencia de Injerto , Humanos , Riñón , Preservación de Órganos , Perfusión , Donantes de Tejidos
3.
Hum Mol Genet ; 27(19): 3313-3324, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29931260

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the growth of renal cysts that ultimately destroy kidney function. Mutations in the PKD1 and PKD2 genes cause ADPKD. Their protein products, polycystin-1 (PC1) and polycystin-2 (PC2) have been proposed to form a calcium-permeable receptor-channel complex; however the mechanisms by which they function are almost completely unknown. Most mutations in PKD1 are truncating loss-of-function mutations or affect protein biogenesis, trafficking or stability and reveal very little about the intrinsic biochemical properties or cellular functions of PC1. An ADPKD patient mutation (L4132Δ or ΔL), resulting in a single amino acid deletion in a putative G-protein binding region of the PC1 C-terminal cytosolic tail, was found to significantly decrease PC1-stimulated, G-protein-dependent signaling in transient transfection assays. Pkd1ΔL/ΔL mice were embryo-lethal suggesting that ΔL is a functionally null mutation. Kidney-specific Pkd1ΔL/cond mice were born but developed severe, postnatal cystic disease. PC1ΔL protein expression levels and maturation were comparable to those of wild type PC1, and PC1ΔL protein showed cell surface localization. Expression of PC1ΔL and PC2 complexes in transfected CHO cells failed to support PC2 channel activity, suggesting that the role of PC1 is to activate G-protein signaling to regulate the PC1/PC2 calcium channel.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/genética , Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/genética , Animales , Células CHO , Canales de Calcio/genética , Cilios/genética , Cilios/patología , Cricetulus , Humanos , Riñón/patología , Ratones , Mutación , Riñón Poliquístico Autosómico Dominante/patología , Dominios Proteicos/genética , Transducción de Señal
4.
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
5.
Kidney Int ; 92(5): 1041-1043, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29055424

RESUMEN

In this issue of Kidney International, Outeda et al. present a new epitope-tagged allele of murine Pkhd1 that allows the monitoring of functional fibrocystin in vivo from the extreme C-terminus of the molecule. This work also shows that the removal of two-thirds of the intracellular tail of fibrocystin does not result in cystogenesis in either the liver or kidney, with major implications for our understanding of Pkhd1 function and polycystic kidney disease in general.


Asunto(s)
Alelos , Receptores de Superficie Celular/genética , Animales , Epítopos , Humanos , Riñón , Ratones , Riñón Poliquístico Autosómico Recesivo/genética
6.
Radiology ; 284(3): 694-705, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28128707

RESUMEN

Purpose To investigate the utility of magnetic resonance (MR) elastography-derived mechanical properties in the discrimination of hepatic inflammation and fibrosis in the early stages of chronic liver diseases. Materials and Methods All studies were approved by the institutional animal care and use committee. A total of 187 animals were studied, including 182 mice and five pigs. These animals represented five different liver diseases with a varying combination and extent of hepatic inflammation, fibrosis, congestion, and portal hypertension. Multifrequency three-dimensional MR elastography was performed, and shear stiffness, storage modulus, shear loss modulus, and damping ratio were calculated for all animals. Necroinflammation, fibrosis, and portal pressure were either histologically scored or biochemically and physically quantified in all animals. Two-sided Welch t tests were used to evaluate mean differences between disease and control groups. Spearman correlation analyses were used to evaluate the relationships between mechanical parameters and quantitative fibrosis extent (hydroxyproline concentration) and portal pressure. Results Liver stiffness and storage modulus increased with progressively developed fibrosis and portal hypertension (mean stiffness at 80 Hz and 48-week feeding, 0.51 kPa ± 0.12 in the steatohepatitis group vs 0.29 kPa ± 0.01 in the control group; P = .02). Damping ratio and shear loss modulus can be used to distinguish inflammation from fibrosis at early stages of disease, even before the development of histologically detectable necroinflammation and fibrosis (mean damping ratio at 80 Hz and 20-week feeding, 0.044 ± 0.012 in the steatohepatitis group vs 0.014 ± 0.008 in the control group; P < .001). Damping ratio and liver stiffness vary differently with respect to cause of portal hypertension (ie, congestion- or cirrhosis-induced hypertension). These differentiation abilities have frequency-dependent variations. Conclusion Liver stiffness and damping ratio measurements can extend hepatic MR elastography to potentially enable assessment of necroinflammatory, congestive, and fibrotic processes of chronic liver diseases. © RSNA, 2017 Online supplemental material is available for this article.


Asunto(s)
Diagnóstico por Imagen de Elasticidad/métodos , Hepatitis/diagnóstico por imagen , Cirrosis Hepática/diagnóstico por imagen , Hígado/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Animales , Tetracloruro de Carbono/toxicidad , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/diagnóstico por imagen , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/fisiopatología , Femenino , Hepatitis/patología , Hepatitis/fisiopatología , Hígado/patología , Hígado/fisiopatología , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , Masculino , Ratones , Porcinos
7.
Am J Hematol ; 92(6): 536-541, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28295502

RESUMEN

Immunoglobulin light chain (AL) amyloidosis is a fatal complication of B-cell proliferation secondary to deposition of amyloid fibrils in various organs. Urinary exosomes (UEX) are the smallest of the microvesicles excreted in the urine. Previously, we found UEX of patients with AL amyloidosis contained immunoglobulin light chain (LC) oligomers that patients with multiple myeloma did not have. To further explore the role of the LC oligomers, UEX was isolated from an AL amyloidosis patient with progressive renal disease despite achieving a complete response. LC oligomers were identified. Mass spectrometry (MS) of the UEX and serum identified two monoclonal lambda LCs. Proteomics of the trypsin digested amyloid fragments in the kidney by laser microdissection and MS analysis identified a λ6 LC. The cDNA from plasma cell clone was from the IGLV- 6-57 family and it matched the amino acid sequences of the amyloid peptides. The predicted mass of the peptide product of the cDNA matched the mass of one of the two LCs identified in the UEX and serum. UEX combined with MS were able to identify 2 monoclonal lambda LCs that current clinical methods could not. It also identified the amyloidogenic LC which holds potential for response assessment in the future.


Asunto(s)
Amiloidosis/complicaciones , Amiloidosis/metabolismo , Exosomas/metabolismo , Cadenas Ligeras de Inmunoglobulina/metabolismo , Proteinuria/diagnóstico , Proteinuria/etiología , Adulto , Anciano , Secuencia de Aminoácidos , Amiloidosis/genética , Femenino , Humanos , Cadenas Ligeras de Inmunoglobulina/química , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/orina , Amiloidosis de Cadenas Ligeras de las Inmunoglobulinas , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Peso Molecular , Agregado de Proteínas , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/orina , Análisis de Secuencia de ADN
8.
Prehosp Emerg Care ; 21(5): 567-574, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28418753

RESUMEN

STUDY OBJECTIVE: Invasively monitoring blood pressure through the IO device has not been thoroughly demonstrated. This study attempted to establish baseline values of IO pressure in a healthy human population. METHODS: This was a prospective, healthy volunteer, observational study. Participants had two IO devices placed (humerus and tibia), and participant IO pressures, vital signs, and pain scores were monitored for up to 60 minutes. Participants were contacted at 24-hours and 7 days post-testing to assess for adverse events. Summary statistics were calculated for systolic, diastolic, and mean humeral and tibial IO pressure. The ratio of IO to non-invasive blood pressure was calculated, and Bland Altman plots were created. The slope (linear) of the mean humeral and the tibial IO pressures were also calculated. RESULTS: Fifteen subjects were enrolled between April and July 2015. Fourteen of 15 humeral IOs were placed successfully (93.3%) and all 15 of the tibial IOs were placed successfully. Mean tibial systolic, diastolic, and mean IO pressure were 55.8 ± 27.9, 49.3 ± 27.1, and 48.4 ± 29.4 mm Hg, respectively. Humeral systolic, diastolic, and mean IO pressure were 32.9 ± 16.0, 27.4 ± 15.2, and 24.5 ± 14.3 mm Hg. The mean tibial IO pressure was 52.5% ± 32.0% of external cuff pressure ratio. The mean humeral IO pressure was 26.5% ± 15.2% of the external mean blood pressure. The Bland Altman plots showed an inconsistent relationship between the systolic and diastolic cuff pressure and the IO pressures. We observed a 1% per minute decrease in IO pressure from the initial placement until the final reading. CONCLUSIONS: Intraosseous pressure readings can be obtained in healthy human volunteers. However, absolute IOP values were not consistent between subjects. Future research may determine how IO pressure can be used to guide therapy in ill and injured patients.


Asunto(s)
Determinación de la Presión Sanguínea/métodos , Infusiones Intraóseas/métodos , Adulto , Presión Sanguínea/fisiología , Determinación de la Presión Sanguínea/efectos adversos , Servicios Médicos de Urgencia/métodos , Femenino , Voluntarios Sanos , Humanos , Húmero , Infusiones Intraóseas/efectos adversos , Infusiones Intraóseas/instrumentación , Masculino , Persona de Mediana Edad , Dolor/etiología , Dimensión del Dolor , Estudios Prospectivos , Tibia , Adulto Joven
9.
J Am Soc Nephrol ; 27(5): 1312-20, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26374610

RESUMEN

Aberrant intracellular calcium levels and increased cAMP signaling contribute to the development of polycystic kidney disease (PKD). cAMP can be hydrolyzed by various phosphodiesterases (PDEs). To examine the role of cAMP hydrolysis and the most relevant PDEs in the pathogenesis of PKD, we examined cyst development in Pde1- or Pde3-knockout mice on the Pkd2(-/WS25) background (WS25 is an unstable Pkd2 allele). These PDEs were selected because of their importance in cross-talk between calcium and cyclic nucleotide signaling (PDE1), control of cell proliferation and cystic fibrosis transmembrane conductance regulator (CFTR) -driven fluid secretion (PDE3), and response to vasopressin V2 receptor activation (both). In Pkd2(-/WS25) mice, knockout of Pde1a, Pde1c, or Pde3a but not of Pde1b or Pde3b aggravated the development of PKD and was associated with higher levels of protein kinase A-phosphorylated (Ser133) cAMP-responsive binding protein (P-CREB), activating transcription factor-1, and CREB-induced CRE modulator proteins in kidney nuclear preparations. Immunostaining also revealed higher expression of P-CREB in Pkd2(-/) (WS25);Pde1a(-/-), Pkd2(-) (/WS25);Pde1c(-/-), and Pkd2(-/) (WS25);Pde3a(-/-) kidneys. The cystogenic effect of desmopressin administration was markedly enhanced in Pkd2(-/WS25);Pde3a(-/-) mice, despite PDE3 accounting for only a small fraction of renal cAMP PDE activity. These observations show that calcium- and calmodulin-dependent PDEs (PDE1A and PDE1C) and PDE3A modulate the development of PKD, possibly through the regulation of compartmentalized cAMP pools that control cell proliferation and CFTR-driven fluid secretion. Treatments capable of increasing the expression or activity of these PDEs may, therefore, retard the development of PKD.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/fisiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/fisiología , Enfermedades Renales Poliquísticas/enzimología , Animales , Femenino , Masculino , Ratones , Ratones Noqueados , Enfermedades Renales Poliquísticas/etiología , Índice de Severidad de la Enfermedad
10.
Cell Mol Life Sci ; 72(12): 2415-29, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25650235

RESUMEN

Dysfunction of many ciliary proteins has been linked to a list of diseases, from cystic kidney to obesity and from hypertension to mental retardation. We previously proposed that primary cilia are unique communication organelles that function as microsensory compartments that house mechanosensory molecules. Here we report that primary cilia exhibit membrane swellings or ciliary bulbs, which based on their unique ultrastructure and motility, could be mechanically regulated by fluid-shear stress. Together with the ultrastructure analysis of the swelling, which contains monosialodihexosylganglioside (GM3), our results show that ciliary bulb has a distinctive set of functional proteins, including GM3 synthase (GM3S), bicaudal-c1 (Bicc1), and polycystin-2 (PC2). In fact, results from our cilia isolation demonstrated for the first time that GM3S and Bicc1 are members of the primary cilia proteins. Although these proteins are not required for ciliary membrane swelling formation under static condition, fluid-shear stress induced swelling formation is partially modulated by GM3S. We therefore propose that the ciliary bulb exhibits a sensory function within the mechano-ciliary structure. Overall, our studies provided an important step towards understanding the ciliary bulb function and structure.


Asunto(s)
Membrana Celular/fisiología , Cilios/fisiología , Células Epiteliales/metabolismo , Riñón/metabolismo , Mecanotransducción Celular/fisiología , Proteínas de Unión al ARN/metabolismo , Sialiltransferasas/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Células Epiteliales/citología , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Riñón/citología , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Sialiltransferasas/antagonistas & inhibidores , Sialiltransferasas/genética , Porcinos , Canales Catiónicos TRPP/antagonistas & inhibidores , Canales Catiónicos TRPP/genética
11.
J Am Soc Nephrol ; 26(7): 1661-70, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25475747

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of ESRD. Affected individuals inherit a defective copy of either PKD1 or PKD2, which encode polycystin-1 (PC1) or polycystin-2 (PC2), respectively. PC1 and PC2 are secreted on urinary exosome-like vesicles (ELVs) (100-nm diameter vesicles), in which PC1 is present in a cleaved form and may be complexed with PC2. Here, label-free quantitative proteomic studies of urine ELVs in an initial discovery cohort (13 individuals with PKD1 mutations and 18 normal controls) revealed that of 2008 ELV proteins, 9 (0.32%) were expressed at significantly different levels in samples from individuals with PKD1 mutations compared to controls (P<0.03). In samples from individuals with PKD1 mutations, levels of PC1 and PC2 were reduced to 54% (P<0.02) and 53% (P<0.001), respectively. Transmembrane protein 2 (TMEM2), a protein with homology to fibrocystin, was 2.1-fold higher in individuals with PKD1 mutations (P<0.03). The PC1/TMEM2 ratio correlated inversely with height-adjusted total kidney volume in the discovery cohort, and the ratio of PC1/TMEM2 or PC2/TMEM2 could be used to distinguish individuals with PKD1 mutations from controls in a confirmation cohort. In summary, results of this study suggest that a test measuring the urine exosomal PC1/TMEM2 or PC2/TMEM2 ratio may have utility in diagnosis and monitoring of polycystic kidney disease. Future studies will focus on increasing sample size and confirming these studies. The data were deposited in the ProteomeXchange (identifier PXD001075).


Asunto(s)
Exosomas/metabolismo , Mutación , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/metabolismo , Adulto , Biomarcadores/metabolismo , Western Blotting , Estudios de Casos y Controles , Estudios de Factibilidad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Fallo Renal Crónico/etiología , Fallo Renal Crónico/fisiopatología , Masculino , Riñón Poliquístico Autosómico Dominante/complicaciones , Riñón Poliquístico Autosómico Dominante/diagnóstico , Riñón Poliquístico Autosómico Dominante/genética , Proteómica/métodos , Valores de Referencia , Sensibilidad y Especificidad , Canales Catiónicos TRPP/genética , Urinálisis , Adulto Joven
12.
BMC Nephrol ; 16: 190, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26577187

RESUMEN

BACKGROUND: Glomerular diseases are potentially fatal, requiring aggressive interventions and close monitoring. Urine is a readily-accessible body fluid enriched in molecular signatures from the kidney and therefore particularly suited for routine clinical analysis as well as development of non-invasive biomarkers for glomerular diseases. METHODS: The Nephrotic Syndrome Study Network (NEPTUNE; ClinicalTrials.gov Identifier NCT01209000) is a North American multicenter collaborative consortium established to develop a translational research infrastructure for nephrotic syndrome. This includes standardized urine collections across all participating centers for the purpose of discovering non-invasive biomarkers for patients with nephrotic syndrome due to minimal change disease, focal segmental glomerulosclerosis, and membranous nephropathy. Here we describe the organization and methods of urine procurement and banking procedures in NEPTUNE. RESULTS: We discuss the rationale for urine collection and storage conditions, and demonstrate the performance of three experimental analytes (neutrophil gelatinase-associated lipocalin [NGAL], retinol binding globulin, and alpha-1 microglobulin) under these conditions with and without urine preservatives (thymol, toluene, and boric acid). We also demonstrate the quality of RNA and protein collected from the urine cellular pellet and exosomes. CONCLUSIONS: The urine collection protocol in NEPTUNE allows robust detection of a wide range of proteins and RNAs from urine supernatant and pellets collected longitudinally from each patient over 5 years. Combined with the detailed clinical and histopathologic data, this provides a unique resource for exploration and validation of new or accepted markers of glomerular diseases. TRIAL REGISTRATION: ClinicalTrials.gov Identifier NCT01209000.


Asunto(s)
Bancos de Muestras Biológicas/organización & administración , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/orina , Proteinuria/orina , Conservación de Tejido/métodos , Toma de Muestras de Orina/métodos , Biomarcadores/sangre , Femenino , Humanos , Masculino , Proteinuria/diagnóstico , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Estados Unidos
13.
J Am Soc Nephrol ; 25(10): 2222-30, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24700876

RESUMEN

Substantial evidence indicates the importance of elevated cAMP in polycystic kidney disease (PKD). Accumulation of cAMP in cystic tissues may be, in part, caused by enhanced adenylyl cyclase activity, but inhibition of cAMP degradation by phosphodiesterases (PDE) likely has an important role, because cAMP is inactivated much faster than it is synthesized. PDE1 is the only PDE family activated by Ca(2+), which is reduced in PKD cells. To assess the contribution of the PDE1A subfamily to renal cyst formation, we examined the expression and function of PDE1A in zebrafish. We identified two splice isoforms with alternative starts corresponding to human PDE1A1 and PDE1A4. Expression of the two isoforms varied in embryos and adult tissues, and both isoforms hydrolyzed cAMP with Ca(2+)/calmodulin dependence. Depletion of PDE1A in zebrafish embryos using splice- and translation-blocking morpholinos (MOs) caused pronephric cysts, hydrocephalus, and body curvature. Human PDE1A RNA and the PKA inhibitors, H89 and Rp-cAMPS, partially rescued phenotypes of pde1a morphants. Additionally, MO depletion of PDE1A aggravated phenotypes in pkd2 morphants, causing more severe body curvature, and human PDE1A RNA partially rescued pkd2 morphant phenotypes, pronephric cysts, hydrocephalus, and body curvature. Together, these data indicate the integral role of PDE1A and cAMP signaling in renal development and cystogenesis, imply that PDE1A activity is altered downstream of polycystin-2, and suggest that PDE1A is a viable drug target for PKD.


Asunto(s)
AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Riñón/embriología , Enfermedades Renales Poliquísticas/enzimología , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Embrión no Mamífero/enzimología , Humanos , Hidrocefalia/etiología , Datos de Secuencia Molecular , Fenotipo , Enfermedades Renales Poliquísticas/etiología , Canales Catiónicos TRPP/metabolismo , Pez Cebra
14.
Nat Genet ; 38(2): 191-6, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16415887

RESUMEN

Meckel-Gruber syndrome is a severe autosomal, recessively inherited disorder characterized by bilateral renal cystic dysplasia, developmental defects of the central nervous system (most commonly occipital encephalocele), hepatic ductal dysplasia and cysts and polydactyly. MKS is genetically heterogeneous, with three loci mapped: MKS1, 17q21-24 (ref. 4); MKS2, 11q13 (ref. 5) and MKS3 (ref. 6). We have refined MKS3 mapping to a 12.67-Mb interval (8q21.13-q22.1) that is syntenic to the Wpk locus in rat, which is a model with polycystic kidney disease, agenesis of the corpus callosum and hydrocephalus. Positional cloning of the Wpk gene suggested a MKS3 candidate gene, TMEM67, for which we identified pathogenic mutations for five MKS3-linked consanguineous families. MKS3 is a previously uncharacterized, evolutionarily conserved gene that is expressed at moderate levels in fetal brain, liver and kidney but has widespread, low levels of expression. It encodes a 995-amino acid seven-transmembrane receptor protein of unknown function that we have called meckelin.


Asunto(s)
Anomalías Múltiples/genética , Mutación/genética , Proteínas/genética , Ratas Mutantes/genética , Animales , Secuencia de Bases , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Exones/genética , Femenino , Marcadores Genéticos , Haplotipos , Humanos , Intrones/genética , Masculino , Proteínas de la Membrana , Datos de Secuencia Molecular , Defectos del Tubo Neural/genética , Linaje , Mapeo Físico de Cromosoma , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Síndrome
15.
Kidney Int ; 85(5): 1225-37, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24196483

RESUMEN

Urinary exosome-like vesicles (ELVs) are a heterogenous mixture (diameter 40-200 nm) containing vesicles shed from all segments of the nephron including glomerular podocytes. Contamination with Tamm-Horsfall protein (THP) oligomers has hampered their isolation and proteomic analysis. Here we improved ELV isolation protocols employing density centrifugation to remove THP and albumin, and isolated a glomerular membranous vesicle (GMV)-enriched subfraction from 7 individuals identifying 1830 proteins and in 3 patients with glomerular disease identifying 5657 unique proteins. The GMV fraction was composed of podocin/podocalyxin-positive irregularly shaped membranous vesicles and podocin/podocalyxin-negative classical exosomes. Ingenuity pathway analysis identified integrin, actin cytoskeleton, and Rho GDI signaling in the top three canonical represented signaling pathways and 19 other proteins associated with inherited glomerular diseases. The GMVs are of podocyte origin and the density gradient technique allowed isolation in a reproducible manner. We show many nephrotic syndrome proteins, proteases, and complement proteins involved in glomerular disease are in GMVs and some were only shed in the disease state (nephrin, TRPC6, INF2 and phospholipase A2 receptor). We calculated sample sizes required to identify new glomerular disease biomarkers, expand the ELV proteome, and provide a reference proteome in a database that may prove useful in the search for biomarkers of glomerular disease.


Asunto(s)
Exosomas/química , Membrana Basal Glomerular/química , Enfermedades Renales/orina , Podocitos/química , Proteinuria/orina , Proteómica/métodos , Urinálisis , Orina/química , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Biomarcadores/orina , Estudios de Casos y Controles , Centrifugación por Gradiente de Densidad , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Enfermedades Renales/diagnóstico , Masculino , Datos de Secuencia Molecular , Proteinuria/diagnóstico , Adulto Joven
16.
Arterioscler Thromb Vasc Biol ; 33(5): 1006-13, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23430615

RESUMEN

OBJECTIVE: Endothelial outgrowth cells (EOC) decrease inflammation and improve endothelial repair. Inflammation aggravates kidney injury in renal artery stenosis (RAS), and may account for its persistence upon revascularization. We hypothesized that EOC would decrease inflammatory (M1) macrophages and improve renal recovery in RAS. APPROACH AND RESULTS: Pigs with 10 weeks of RAS were studied 4 weeks after percutaneous transluminal renal angioplasty (PTRA+stenting) or sham, with or without adjunct intrarenal delivery of autologous EOC (10×10(6)), and compared with similarly treated normal controls (n=7 each). Single-kidney function, microvascular and tissue remodeling, inflammation, oxidative stress, and fibrosis were evaluated. Four weeks after PTRA, EOC were engrafted in injected RAS-kidneys. Stenotic-kidney glomerular filtration rate was restored in RAS+EOC, RAS+PTRA, and RAS+PTRA+EOC pigs, whereas stenotic-kidney blood flow and angiogenesis were improved and fibrosis attenuated only in EOC-treated pigs. Furthermore, EOC increased cell proliferation and decreased the ratio of M1 (inflammatory)/M2 (reparative) macrophages, as well as circulating levels and stenotic-kidney release of inflammatory cytokines. Cultured-EOC released microvesicles in vitro and induced phenotypic switch (M1-to-M2) in cultured monocytes, which was inhibited by vascular endothelial growth factor blockade. Finally, a single intrarenal injection of rh-vascular endothelial growth factor (0.05 µg/kg) in 7 additional RAS pigs also restored M1/M2 ratio 4 weeks later. CONCLUSIONS: Intrarenal infusion of EOC after PTRA induced a vascular endothelial growth factor-mediated attenuation in macrophages inflammatory phenotype, preserved microvascular architecture and function, and decreased inflammation and fibrosis in the stenotic kidney, suggesting a novel mechanism and therapeutic potential for adjunctive EOC delivery in experimental RAS to improve PTRA outcomes.


Asunto(s)
Células Endoteliales/fisiología , Riñón/fisiopatología , Macrófagos/fisiología , Obstrucción de la Arteria Renal/fisiopatología , Animales , Proliferación Celular , Fibrosis , Hemodinámica , Inflamación/etiología , Riñón/patología , Activación de Macrófagos , Fenotipo , Porcinos , Factor A de Crecimiento Endotelial Vascular/farmacología
17.
Gastroenterology ; 142(3): 622-633.e4, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22155366

RESUMEN

BACKGROUND & AIMS: In polycystic kidney disease and polycystic liver disease (PLD), the normally nonproliferative hepato-renal epithelia acquire a proliferative, cystic phenotype that is linked to overexpression of cell division cycle 25 (Cdc25)A phosphatase and cell-cycle deregulation. We investigated the effects of Cdc25A inhibition in mice and rats via genetic and pharmacologic approaches. METHODS: Cdc25A(+/-) mice (which have reduced levels of Cdc25A) were cross-bred with polycystic kidney and hepatic disease 1 (Pkhd1(del2/del2)) mice (which have increased levels of Cdc25A and develop hepatic cysts). Cdc25A expression was analyzed in livers of control and polycystic kidney (PCK) rats, control and polycystic kidney 2 (Pkd2(ws25/-)) mice, healthy individuals, and patients with PLD. We examined effects of pharmacologic inhibition of Cdc25A with vitamin K3 (VK3) on the cell cycle, proliferation, and cyst expansion in vitro; hepato-renal cystogenesis in PCK rats and Pkd2(ws25/-)mice; and expression of Cdc25A and the cell-cycle proteins regulated by Cdc25A. We also examined the effects of the Cdc25A inhibitor PM-20 on hepato-renal cystogenesis in Pkd2(ws25/-) mice. RESULTS: Liver weights and hepatic and fibrotic areas were decreased by 32%-52% in Cdc25A(+/-):Pkhd1(del2/del2) mice, compared with Pkhd1(del2/del2) mice. VK3 altered the cell cycle and reduced proliferation of cultured cholangiocytes by 32%-83% and decreased growth of cultured cysts by 23%-67%. In PCK rats and Pkd2(ws25/-) mice, VK3 reduced liver and kidney weights and hepato-renal cystic and fibrotic areas by 18%-34%. PM-20 decreased hepato-renal cystogenesis in Pkd2(ws25/-) mice by 15%. CONCLUSIONS: Cdc25A inhibitors block cell-cycle progression and proliferation, reduce liver and kidney weights and cyst growth in animal models of polycystic kidney disease and PLD, and might be developed as therapeutics for these diseases.


Asunto(s)
Quistes/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Riñón/efectos de los fármacos , Hepatopatías/tratamiento farmacológico , Hígado/efectos de los fármacos , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Vitamina K 3/farmacología , Fosfatasas cdc25/antagonistas & inhibidores , Animales , Conductos Biliares Intrahepáticos/efectos de los fármacos , Conductos Biliares Intrahepáticos/enzimología , Conductos Biliares Intrahepáticos/patología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quistes/enzimología , Quistes/genética , Quistes/patología , Modelos Animales de Enfermedad , Humanos , Riñón/enzimología , Riñón/patología , Hígado/enzimología , Hígado/patología , Hepatopatías/enzimología , Hepatopatías/genética , Hepatopatías/patología , Ratones , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , Riñón Poliquístico Autosómico Recesivo/enzimología , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/patología , Ratas , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Factores de Tiempo , Regulación hacia Arriba , Fosfatasas cdc25/deficiencia , Fosfatasas cdc25/genética , Fosfatasas cdc25/metabolismo
18.
J Am Soc Nephrol ; 23(5): 915-33, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22383692

RESUMEN

Mutations in two large multi-exon genes, PKD1 and PKD2, cause autosomal dominant polycystic kidney disease (ADPKD). The duplication of PKD1 exons 1-32 as six pseudogenes on chromosome 16, the high level of allelic heterogeneity, and the cost of Sanger sequencing complicate mutation analysis, which can aid diagnostics of ADPKD. We developed and validated a strategy to analyze both the PKD1 and PKD2 genes using next-generation sequencing by pooling long-range PCR amplicons and multiplexing bar-coded libraries. We used this approach to characterize a cohort of 230 patients with ADPKD. This process detected definitely and likely pathogenic variants in 115 (63%) of 183 patients with typical ADPKD. In addition, we identified atypical mutations, a gene conversion, and one missed mutation resulting from allele dropout, and we characterized the pattern of deep intronic variation for both genes. In summary, this strategy involving next-generation sequencing is a model for future genetic characterization of large ADPKD populations.


Asunto(s)
Mutación , Riñón Poliquístico Autosómico Dominante/genética , Análisis de Secuencia de ADN/métodos , Canales Catiónicos TRPP/genética , Procesamiento Automatizado de Datos , Humanos , Reacción en Cadena de la Polimerasa
19.
Nat Genet ; 30(3): 259-69, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11919560

RESUMEN

Autosomal recessive polycystic kidney disease (ARPKD) is characterized by dilation of collecting ducts and by biliary dysgenesis and is an important cause of renal- and liver-related morbidity and mortality. Genetic analysis of a rat with recessive polycystic kidney disease revealed an orthologous relationship between the rat locus and the ARPKD region in humans; a candidate gene was identified. A mutation was characterized in the rat and screening the 66 coding exons of the human ortholog (PKHD1) in 14 probands with ARPKD revealed 6 truncating and 12 missense mutations; 8 of the affected individuals were compound heterozygotes. The PKHD1 transcript, approximately 16 kb long, is expressed in adult and fetal kidney, liver and pancreas and is predicted to encode a large novel protein, fibrocystin, with multiple copies of a domain shared with plexins and transcription factors. Fibrocystin may be a receptor protein that acts in collecting-duct and biliary differentiation.


Asunto(s)
Mutación , Riñón Poliquístico Autosómico Recesivo/genética , Receptores de Superficie Celular/genética , Adulto , Secuencia de Aminoácidos , Animales , Clonación Molecular , Femenino , Pruebas Genéticas , Heterocigoto , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Riñón Poliquístico Autosómico Recesivo/diagnóstico , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/química , Homología de Secuencia de Aminoácido
20.
Cells ; 12(17)2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37681898

RESUMEN

The PKD1 gene, encoding protein polycystin-1 (PC1), is responsible for 85% of cases of autosomal dominant polycystic kidney disease (ADPKD). PC1 has been shown to be present in urinary exosome-like vesicles (PKD-ELVs) and lowered in individuals with germline PKD1 mutations. A label-free mass spectrometry comparison of urinary PKD-ELVs from normal individuals and those with PKD1 mutations showed that several proteins were reduced to a degree that matched the decrease observed in PC1 levels. Some of these proteins, such as polycystin-2 (PC2), may be present in a higher-order multi-protein assembly with PC1-the polycystin complex (PCC). CU062 (Q9NYP8) is decreased in ADPKD PKD-ELVs and, thus, is a candidate PCC component. CU062 is a small glycoprotein with a signal peptide but no transmembrane domain and can oligomerize with itself and interact with PC1. We investigated the localization of CU062 together with PC1 and PC2 using immunofluorescence (IF). In nonconfluent cells, all three proteins were localized in close proximity to focal adhesions (FAs), retraction fibers (RFs), and RF-associated extracellular vesicles (migrasomes). In confluent cells, primary cilia had PC1/PC2/CU062 + extracellular vesicles adherent to their plasma membrane. In cells exposed to mitochondrion-decoupling agents, we detected the development of novel PC1/CU062 + ring-like structures that entrained swollen mitochondria. In contact-inhibited cells under mitochondrial stress, PC1, PC2, and CU062 were observed on large, apically budding extracellular vesicles, where the proteins formed a reticular network on the membrane. CU062 interacts with PC1 and may have a role in the identification of senescent mitochondria and their extrusion in extracellular vesicles.


Asunto(s)
Vesículas Extracelulares , Riñón Poliquístico Autosómico Dominante , Humanos , Genes Reguladores , Mitocondrias , Canales Catiónicos TRPP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA