Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
3.
Sci Rep ; 12(1): 7763, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35546171

RESUMEN

Apelin exists in many isoforms, both in the circulation and in specific tissues. Apelin peptides have a short half-life but preservation before measurement is scarcely studied. Reproducible mass spectrometry methods to specifically measure a broad range of apelinergic peptide isoforms are currently lacking. A sample protocol to conserve apelinergic peptides in the preanalytical phase and a high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) method to measure apelinergic isoforms was developed. Apelin was measured in plasma. For validation, human embryonic kidney (HEK) cells transfected with cDNA for preproapelin were used. Results were compared with a validated radioimmunoassay (RIA) method. Acidifying plasma to pH 2.5 improves post-sampling stability of apelin. HPLC-MS/MS was unable to detect apelin isoforms in plasma of healthy volunteers (n = 16) and chronic kidney disease patients (n = 4). RIA could detect apelin in concentrations between 71 and 263 fmol/l in 10 healthy volunteers. An optimized preanalytical protocol was developed. A sensitive and specific HPLC-MS/MS method failed to detect apelin in human plasma. Apelin-36 was detected in HEK cells transfected with cDNA for preproapelin. Currently, RIA with relatively selective antibodies is the best alternative for the measurement of apelin but novel sensitive and specific methods are needed.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular , Espectrometría de Masas en Tándem , Apelina , Receptores de Apelina , ADN Complementario , Humanos , Ligandos , Péptidos , Isoformas de Proteínas , Espectrometría de Masas en Tándem/métodos
4.
Pediatr Nephrol ; 37(9): 1945-1955, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-34988697

RESUMEN

The clinical course of autosomal dominant polycystic kidney disease (ADPKD) starts in childhood. Evidence of the beneficial impact of early nephron-protective strategies and lifestyle modifications on ADPKD prognosis is accumulating. Recent studies have described the association of overweight and obesity with rapid disease progression in adults with ADPKD. Moreover, defective glucose metabolism and metabolic reprogramming have been reported in distinct ADPKD models highlighting these pathways as potential therapeutic targets in ADPKD. Several "metabolic" approaches are currently under evaluation in adults, including ketogenic diet, food restriction, and metformin therapy. No data are available on the impact of these approaches in childhood thus far. Yet, according to World Health Organization (WHO), we are currently facing a childhood obesity crisis with an increased prevalence of overweight/obesity in the pediatric population associated with a cardio-metabolic risk profile. The present review summarizes the knowledge about the role of glucose metabolism in the pathophysiology of ADPKD and underscores the possible harm of overweight and obesity in ADPKD especially in terms of long-term cardiovascular outcomes and renal prognosis.


Asunto(s)
Obesidad Infantil , Riñón Poliquístico Autosómico Dominante , Adulto , Niño , Progresión de la Enfermedad , Glucosa/uso terapéutico , Humanos , Riñón , Sobrepeso/complicaciones , Obesidad Infantil/complicaciones , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/terapia
5.
Nephrol Dial Transplant ; 37(12): 2314-2326, 2022 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33744967

RESUMEN

The apelinergic system (AS) is a novel pleiotropic system with an essential role in renal and cardiovascular physiology and disease, including water homeostasis and blood pressure regulation. It consists of two highly conserved peptide ligands, apelin and apela, and a G-protein-coupled apelin receptor. The two ligands have many isoforms and a short half-life and exert both similar and divergent effects. Vasopressin, apelin and their receptors colocalize in hypothalamic regions essential for body fluid homeostasis and interact at the central and renal levels to regulate water homeostasis and diuresis in inverse directions. In addition, the AS and renin-angiotensin system interact both systemically and in the kidney, with implications for the cardiovascular system. A role for the AS in diverse pathological states, including disorders of sodium and water balance, hypertension, heart failure, pre-eclampsia, acute kidney injury, sepsis and diabetic nephropathy, has recently been reported. Furthermore, several metabolically stable apelin analogues have been developed, with potential applications in diverse diseases. We review here what is currently known about the physiological functions of the AS, focusing on renal, cardiovascular and metabolic homeostasis, and the role of the AS in associated diseases. We also describe several hurdles and research opportunities worthy of the attention of the nephrology community.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular , Enfermedades Renales , Receptores Acoplados a Proteínas G , Humanos , Apelina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/metabolismo , Ligandos , Receptores Acoplados a Proteínas G/metabolismo , Agua/metabolismo
6.
Int J Mol Sci ; 22(24)2021 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-34948309

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is mainly caused by deficiency of polycystin-1 (PC1) or polycystin-2 (PC2). Altered autophagy has recently been implicated in ADPKD progression, but its exact regulation by PC1 and PC2 remains unclear. We therefore investigated cell death and survival during nutritional stress in mouse inner medullary collecting duct cells (mIMCDs), either wild-type (WT) or lacking PC1 (PC1KO) or PC2 (PC2KO), and human urine-derived proximal tubular epithelial cells (PTEC) from early-stage ADPKD patients with PC1 mutations versus healthy individuals. Basal autophagy was enhanced in PC1-deficient cells. Similarly, following starvation, autophagy was enhanced and cell death reduced when PC1 was reduced. Autophagy inhibition reduced cell death resistance in PC1KO mIMCDs to the WT level, implying that PC1 promotes autophagic cell survival. Although PC2 expression was increased in PC1KO mIMCDs, PC2 knockdown did not result in reduced autophagy. PC2KO mIMCDs displayed lower basal autophagy, but more autophagy and less cell death following chronic starvation. This could be reversed by overexpression of PC1 in PC2KO. Together, these findings indicate that PC1 levels are partially coupled to PC2 expression, and determine the transition from renal cell survival to death, leading to enhanced survival of ADPKD cells during nutritional stress.


Asunto(s)
Autofagia/fisiología , Muerte Celular/fisiología , Inanición/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Línea Celular , Células Epiteliales/metabolismo , Humanos , Túbulos Renales Proximales/metabolismo , Ratones , Riñón Poliquístico Autosómico Dominante/metabolismo
7.
Cells ; 10(8)2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34440683

RESUMEN

Apolipoprotein L1 (APOL1) high-risk genotypes (HRG), G1 and G2, increase the risk of various non-diabetic kidney diseases in the African population. To date, the precise mechanisms by which APOL1 risk variants induce injury on podocytes and other kidney cells remain unclear. Trying to unravel these mechanisms, most studies have used animal or cell models created by gene editing. We developed and characterised conditionally immortalised human podocyte cell lines derived from urine of a donor carrying APOL1 HRG G2/G2. Following induction of APOL1 expression by polyinosinic-polycytidylic acid (poly(I:C)), we assessed functional features of APOL1-induced podocyte dysfunction. As control, APOL1 wild type (G0/G0) podocyte cell line previously generated from a Caucasian donor was used. Upon exposure to poly(I:C), G2/G2 and G0/G0 podocytes upregulated APOL1 expression resulting in podocytes detachment, decreased cells viability and increased apoptosis rate in a genotype-independent manner. Nevertheless, G2/G2 podocyte cell lines exhibited altered features, including upregulation of CD2AP, alteration of cytoskeleton, reduction of autophagic flux and increased permeability in an in vitro model under continuous perfusion. The human APOL1 G2/G2 podocyte cell model is a useful tool for unravelling the mechanisms of APOL1-induced podocyte injury and the cellular functions of APOL1.


Asunto(s)
Apolipoproteína L1/metabolismo , Modelos Biológicos , Adulto , Apolipoproteína L1/genética , Autofagia/efectos de los fármacos , Adhesión Celular , Línea Celular , Preescolar , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Femenino , Genotipo , Humanos , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Podocitos/citología , Podocitos/metabolismo , Poli I-C/farmacología , Regulación hacia Arriba/efectos de los fármacos
8.
Kidney Int Rep ; 6(6): 1687-1698, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34169210

RESUMEN

INTRODUCTION: Autosomal dominant polycystic kidney disease (ADPKD) causes kidney failure typically in adulthood, but the disease starts in utero. Copeptin, epidermal growth factor (EGF), and monocyte chemoattractant protein-1 (MCP-1) are associated with severity and hold prognostic value in adults but remain unstudied in the early disease stage. Kidneys from adults with ADPKD exhibit macrophage infiltration, and a prominent role of MCP-1 secretion by tubular epithelial cells is suggested from rodent models. METHODS: In a cross-sectional study, plasma copeptin, urinary EGF, and urinary MCP-1 were evaluated in a pediatric ADPKD cohort and compared with age-, sex-, and body mass index (BMI)-matched healthy controls. MCP-1 was studied in mouse collecting duct cells, human proximal tubular cells, and fetal kidney tissue. RESULTS: Fifty-three genotyped ADPKD patients and 53 controls were included. The mean (SD) age was 10.4 (5.9) versus 10.5 (6.1) years (P = 0.543), and the estimated glomerular filtration rate (eGFR) was 122.7 (39.8) versus 114.5 (23.1) ml/min per 1.73 m2 (P = 0.177) in patients versus controls, respectively. Plasma copeptin and EGF secretion were comparable between groups. The median (interquartile range) urinary MCP-1 (pg/mg creatinine) was significantly higher in ADPKD patients (185.4 [213.8]) compared with controls (154.7 [98.0], P = 0.010). Human proximal tubular cells with a heterozygous PKD1 mutation and mouse collecting duct cells with a PKD1 knockout exhibited increased MCP-1 secretion. Human fetal ADPKD kidneys displayed prominent MCP-1 immunoreactivity and M2 macrophage infiltration. CONCLUSION: An increase in tubular MCP-1 secretion is an early event in ADPKD. MCP-1 is an early disease severity marker and a potential treatment target.

9.
Int J Mol Sci ; 21(19)2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-33003356

RESUMEN

Renal ischemia-reperfusion (IR) injury leading to cell death is a major cause of acute kidney injury, contributing to morbidity and mortality. Autophagy counteracts cell death by removing damaged macromolecules and organelles, making it an interesting anchor point for treatment strategies. However, autophagy is also suggested to enhance cell death when the ischemic burden is too strong. To investigate whether the role of autophagy depends on the severity of ischemic stress, we analyzed the dynamics of autophagy and apoptosis in an IR rat model with mild (45 min) or severe (60 min) renal ischemia. Following mild IR, renal injury was associated with reduced autophagy, enhanced mammalian target of rapamycin (mTOR) activity, and apoptosis. Severe IR, on the other hand, was associated with a higher autophagic activity, independent of mTOR, and without affecting apoptosis. Autophagy stimulation by trehalose injected 24 and 48 h prior to onset of severe ischemia did not reduce renal injury markers nor function, but reduced apoptosis and restored tubular dilation 7 days post reperfusion. This suggests that trehalose-dependent autophagy stimulation enhances tissue repair following an IR injury. Our data show that autophagy dynamics are strongly dependent on the severity of IR and that trehalose shows the potential to trigger autophagy-dependent repair processes following renal IR injury.


Asunto(s)
Lesión Renal Aguda/genética , Autofagia/genética , Daño por Reperfusión/genética , Serina-Treonina Quinasas TOR/genética , Lesión Renal Aguda/patología , Animales , Apoptosis/genética , Modelos Animales de Enfermedad , Humanos , Riñón/metabolismo , Riñón/patología , Ratas , Daño por Reperfusión/patología
10.
Nature ; 578(7795): 419-424, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31996848

RESUMEN

ATP13A2 (PARK9) is a late endolysosomal transporter that is genetically implicated in a spectrum of neurodegenerative disorders, including Kufor-Rakeb syndrome-a parkinsonism with dementia1-and early-onset Parkinson's disease2. ATP13A2 offers protection against genetic and environmental risk factors of Parkinson's disease, whereas loss of ATP13A2 compromises lysosomes3. However, the transport function of ATP13A2 in lysosomes remains unclear. Here we establish ATP13A2 as a lysosomal polyamine exporter that shows the highest affinity for spermine among the polyamines examined. Polyamines stimulate the activity of purified ATP13A2, whereas ATP13A2 mutants that are implicated in disease are functionally impaired to a degree that correlates with the disease phenotype. ATP13A2 promotes the cellular uptake of polyamines by endocytosis and transports them into the cytosol, highlighting a role for endolysosomes in the uptake of polyamines into cells. At high concentrations polyamines induce cell toxicity, which is exacerbated by ATP13A2 loss due to lysosomal dysfunction, lysosomal rupture and cathepsin B activation. This phenotype is recapitulated in neurons and nematodes with impaired expression of ATP13A2 or its orthologues. We present defective lysosomal polyamine export as a mechanism for lysosome-dependent cell death that may be implicated in neurodegeneration, and shed light on the molecular identity of the mammalian polyamine transport system.


Asunto(s)
Lisosomas/metabolismo , Poliaminas/metabolismo , ATPasas de Translocación de Protón/deficiencia , ATPasas de Translocación de Protón/genética , Animales , Biocatálisis , Transporte Biológico , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Catepsina B/metabolismo , Citosol/metabolismo , Modelos Animales de Enfermedad , Endocitosis , Humanos , Lisosomas/patología , Ratones , Mutación , Neuronas/metabolismo , Fenotipo , Poliaminas/toxicidad , ATPasas de Translocación de Protón/metabolismo , Espermidina/metabolismo , Espermina/metabolismo
11.
Pediatr Nephrol ; 34(10): 1697-1715, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30215095

RESUMEN

Several animal- and human-derived models are used in autosomal dominant polycystic kidney disease (ADPKD) research to gain insight in the disease mechanism. However, a consistent correlation between animal and human ADPKD models is lacking. Therefore, established human-derived models are relevant to affirm research results and translate findings into a clinical set-up. In this review, we give an extensive overview of the existing human-based cell models. We discuss their source (urine, nephrectomy and stem cell), immortalisation procedures, genetic engineering, kidney segmental origin and characterisation with nephron segment markers. We summarise the most studied pathways and lessons learned from these different ADPKD models. Finally, we issue recommendations for the derivation of human-derived cell lines and for experimental set-ups with these cell lines.


Asunto(s)
Riñón/fisiopatología , Riñón Poliquístico Autosómico Dominante/fisiopatología , Antagonistas de los Receptores de Hormonas Antidiuréticas/farmacología , Antagonistas de los Receptores de Hormonas Antidiuréticas/uso terapéutico , Señalización del Calcio , Línea Celular , Proliferación Celular , Cilios/patología , Ensayos Clínicos como Asunto , Glucosidasas/genética , Humanos , Riñón/citología , Riñón/efectos de los fármacos , Riñón/patología , Mutación , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Cultivo Primario de Células/métodos , Serina-Treonina Quinasas TOR/metabolismo , Canales Catiónicos TRPP/genética , Tolvaptán/farmacología , Tolvaptán/uso terapéutico , Resultado del Tratamiento
12.
Transplantation ; 101(11): e330-e336, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28704336

RESUMEN

BACKGROUND: Early detection of acute kidney injury (AKI), a common condition with a high mortality risk, can be facilitated by specific and reliable biomarkers. Villin 1, a protein typically found in the brush borders of proximal tubular cells, has been detected in urine of patients with AKI, but its possible release in plasma remains unexplored. METHODS: We measured the presence of villin 1 by immunohistochemistry on kidney sections and by Western blotting in plasma samples from rats subjected to renal ischemia-reperfusion injury, pigs subjected to renal transplantation and liver transplantation patients that developed AKI. Moreover, rats were treated with necrostatin-1, an inhibitor of programmed necrosis (necroptosis), which occurs in renal tubular cells during AKI. Villin 1 levels were compared with other renal injury markers (creatinine, aspartate transaminase, and heart-type fatty acid binding protein). RESULTS: During AKI, plasmatic villin 1 levels corresponded with the severity of kidney injury and dysfunction. Its detection in plasma was associated with a redistribution in the kidney tissue. Unlike the levels of other markers, plasmatic villin 1 decreased already after a short (3 hours) treatment with necrostatin-1 during renal ischemia-reperfusion injury. The presence of plasmatic villin 1 was confirmed in patients who experienced AKI after liver transplantation. CONCLUSIONS: Villin 1 is released in plasma during AKI and shows potential as an early marker for proximal tubular injury/necrosis and warrants further investigation.


Asunto(s)
Lesión Renal Aguda/sangre , Túbulos Renales Proximales/metabolismo , Proteínas de Microfilamentos/sangre , Daño por Reperfusión/sangre , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Aspartato Aminotransferasas/sangre , Biomarcadores/sangre , Creatinina/sangre , Modelos Animales de Enfermedad , Diagnóstico Precoz , Proteína 3 de Unión a Ácidos Grasos , Proteínas de Unión a Ácidos Grasos/sangre , Femenino , Imidazoles/farmacología , Indoles/farmacología , Trasplante de Riñón/efectos adversos , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Trasplante de Hígado/efectos adversos , Necrosis , Valor Predictivo de las Pruebas , Ratas Sprague-Dawley , Daño por Reperfusión/etiología , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Sus scrofa , Factores de Tiempo
13.
Sci Rep ; 7: 41408, 2017 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-28134274

RESUMEN

Superparamagnetic iron oxide nanoparticles (SPIONs) have mainly been used as cellular carriers for genes and therapeutic products, while their use in subcellular organelle isolation remains underexploited. We engineered SPIONs targeting distinct subcellular compartments. Dimercaptosuccinic acid-coated SPIONs are internalized and accumulate in late endosomes/lysosomes, while aminolipid-SPIONs reside at the plasma membrane. These features allowed us to establish standardized magnetic isolation procedures for these membrane compartments with a yield and purity permitting proteomic and lipidomic profiling. We validated our approach by comparing the biomolecular compositions of lysosomes and plasma membranes isolated from wild-type and Niemann-Pick disease type C1 (NPC1) deficient cells. While the accumulation of cholesterol and glycosphingolipids is seen as a primary hallmark of NPC1 deficiency, our lipidomics analysis revealed the buildup of several species of glycerophospholipids and other storage lipids in selectively late endosomes/lysosomes of NPC1-KO cells. While the plasma membrane proteome remained largely invariable, we observed pronounced alterations in several proteins linked to autophagy and lysosomal catabolism reflecting vesicular transport obstruction and defective lysosomal turnover resulting from NPC1 deficiency. Thus the use of SPIONs provides a major advancement in fingerprinting subcellular compartments, with an increased potential to identify disease-related alterations in their biomolecular compositions.


Asunto(s)
Metabolismo de los Lípidos , Lisosomas/patología , Glicoproteínas de Membrana/deficiencia , Proteómica , Autofagosomas/metabolismo , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Dextranos/química , Endosomas/metabolismo , Técnicas de Inactivación de Genes , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lisosomas/metabolismo , Nanopartículas de Magnetita/química , Glicoproteínas de Membrana/metabolismo , Nanopartículas/ultraestructura , Proteína Niemann-Pick C1 , Proteoma/metabolismo , Esteroles/metabolismo , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/ultraestructura
14.
PLoS One ; 12(1): e0169331, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28060943

RESUMEN

INTRODUCTION: The farnesoid X receptor (FXR) is abundantly expressed in the ileum, where it exerts an enteroprotective role as a key regulator of intestinal innate immunity and homeostasis, as shown in pre-clinical models of inflammatory bowel disease. Since intestinal ischemia reperfusion injury (IRI) is characterized by hyperpermeability, bacterial translocation and inflammation, we aimed to investigate, for the first time, if the FXR-agonist obeticholic acid (OCA) could attenuate intestinal ischemia reperfusion injury. MATERIAL AND METHODS: In a validated rat model of intestinal IRI (laparotomy + temporary mesenteric artery clamping), 3 conditions were tested (n = 16/group): laparotomy only (sham group); ischemia 60min+ reperfusion 60min + vehicle pretreatment (IR group); ischemia 60min + reperfusion 60min + OCA pretreatment (IR+OCA group). Vehicle or OCA (INT-747, 2*30mg/kg) was administered by gavage 24h and 4h prior to IRI. The following end-points were analyzed: 7-day survival; biomarkers of enterocyte viability (L-lactate, I-FABP); histology (morphologic injury to villi/crypts and villus length); intestinal permeability (Ussing chamber); endotoxin translocation (Lipopolysaccharide assay); cytokines (IL-6, IL-1-ß, TNFα, IFN-γ IL-10, IL-13); apoptosis (cleaved caspase-3); and autophagy (LC3, p62). RESULTS: It was found that intestinal IRI was associated with high mortality (90%); loss of intestinal integrity (structurally and functionally); increased endotoxin translocation and pro-inflammatory cytokine production; and inhibition of autophagy. Conversely, OCA-pretreatment improved 7-day survival up to 50% which was associated with prevention of epithelial injury, preserved intestinal architecture and permeability. Additionally, FXR-agonism led to decreased pro-inflammatory cytokine release and alleviated autophagy inhibition. CONCLUSION: Pretreatment with OCA, an FXR-agonist, improves survival in a rodent model of intestinal IRI, preserves the gut barrier function and suppresses inflammation. These results turn FXR into a promising target for various conditions associated with intestinal ischemia.


Asunto(s)
Mucosa Intestinal/metabolismo , Intestinos/irrigación sanguínea , Receptores Citoplasmáticos y Nucleares/metabolismo , Daño por Reperfusión/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Biomarcadores , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/farmacología , Modelos Animales de Enfermedad , Endotoxinas/metabolismo , Íleon/irrigación sanguínea , Íleon/efectos de los fármacos , Íleon/metabolismo , Íleon/patología , Mediadores de Inflamación/metabolismo , Intestinos/efectos de los fármacos , Intestinos/patología , Masculino , Permeabilidad , Ratas , Receptores Citoplasmáticos y Nucleares/agonistas , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/mortalidad , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos
15.
Oxid Med Cell Longev ; 2017: 7120962, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29410735

RESUMEN

Many factors during the transplantation process influence posttransplant graft function and survival, including donor type and age, graft preservation methods (cold storage, machine perfusion), and ischemia-reperfusion injury. Successively, they will lead to cellular and molecular alterations that determine cell and ultimately organ fate. Oxidative stress and autophagy are implicated in posttransplant outcome since they are both affected by the stress responses triggered in each step (donor, preservation, and recipient) of the transplantation process. Furthermore, oxidative stress influences autophagy and vice versa. Interestingly, both processes have positive as well as negative effects on graft outcome, suggesting they are tightly linked during the transplantation process. In this review, we discuss the importance, regulation and crosstalk of oxidative signals, and autophagy in the field of transplantation medicine.


Asunto(s)
Autofagia/genética , Especies Reactivas de Oxígeno/metabolismo , Trasplante/métodos , Humanos
16.
BMC Cell Biol ; 17 Suppl 1: 20, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27229147

RESUMEN

Connexins mediate intercellular communication by assembling into hexameric channel complexes that act as hemichannels and gap junction channels. Most connexins are characterized by a very rapid turn-over in a variety of cell systems. The regulation of connexin turn-over by phosphorylation and ubiquitination events has been well documented. Moreover, different pathways have been implicated in connexin degradation, including proteasomal and lysosomal-based pathways. Only recently, autophagy emerged as an important connexin-degradation pathway for different connexin isoforms. As such, conditions well known to induce autophagy have an immediate impact on the connexin-expression levels. This is not only limited to experimental conditions but also several pathophysiological conditions associated with autophagy (dys)function affect connexin levels and their presence at the cell surface as gap junctions. Finally, connexins are not only substrates of autophagy but also emerge as regulators of the autophagy process. In particular, several connexin isoforms appear to recruit pre-autophagosomal autophagy-related proteins, including Atg16 and PI3K-complex components, to the plasma membrane, thereby limiting their availability and capacity for regulating autophagy.


Asunto(s)
Autofagia , Conexinas/metabolismo , Animales , Humanos , Lisosomas/metabolismo , Modelos Biológicos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis
17.
Pediatr Nephrol ; 31(5): 737-52, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26141928

RESUMEN

Autophagy is the cell biology process in which cytoplasmic components are degraded in lysosomes to maintain cellular homeostasis and energy production. In the healthy kidney, autophagy plays an important role in the homeostasis and viability of renal cells such as podocytes and tubular epithelial cells and of immune cells. Recently, evidence is mounting that (dys)regulation of autophagy is implicated in the pathogenesis of various renal diseases, and might be an attractive target for new renoprotective therapies. In this review, we provide an overview of the role of autophagy in kidney physiology and kidney diseases.


Asunto(s)
Autofagia , Células Epiteliales/patología , Enfermedades Renales/patología , Riñón/patología , Animales , Autofagia/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/fisiopatología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Terapia Molecular Dirigida , Transducción de Señal , Agentes Urológicos/uso terapéutico
18.
Autophagy ; 11(10): 1944-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26291777

RESUMEN

ITPRs (inositol 1,4,5-trisphosphate receptors), the main endoplasmic reticulum (ER) Ca(2+)-release channels, were originally proposed as suppressors of autophagy. Yet, new evidence has accumulated over recent years supporting a crucial, stimulatory role for ITPRs in driving the autophagic flux. Here, we provide an integrated view on how ITPR-mediated Ca(2+) signaling can have a dual impact on autophagy, depending on the characteristics of the spatio-temporal Ca(2+) signals, including the existence of ER-mitochondrial and ER-lysosomal Ca(2+) signaling microdomains.


Asunto(s)
Autofagia/fisiología , Señalización del Calcio/fisiología , Retículo Endoplásmico/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Animales , Calcio/metabolismo , Humanos
19.
Am J Kidney Dis ; 66(4): 699-709, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26169721

RESUMEN

Autophagy, an evolutionary conserved intracellular lysosome-dependent catabolic process, is an important mechanism for cellular homeostasis and survival during pathologic stress conditions in the kidney, such as ischemia-reperfusion injury (IRI). However, stimulation of autophagy has been described to both improve and exacerbate IRI in the kidney. We summarize the current understanding of autophagy in renal IRI and discuss possible reasons for these contradictory findings. Furthermore, we hypothesize that autophagy plays a dual role in renal IRI, having both protective and detrimental properties, depending on the duration of the ischemic period and the phase of the IRI process. Finally, we discuss the influence of currently used diuretics and immunosuppressive drugs on autophagy, underscoring the need to clarify the puzzling role of autophagy in renal IRI.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Trasplante de Riñón/efectos adversos , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Animales , Autofagia/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Rechazo de Injerto , Supervivencia de Injerto , Humanos , Inmunosupresores/administración & dosificación , Trasplante de Riñón/métodos , Masculino , Ratones , Ratones Noqueados , Ratas , Daño por Reperfusión/fisiopatología , Medición de Riesgo , Sensibilidad y Especificidad
20.
Pflugers Arch ; 466(8): 1591-604, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24193408

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is caused by loss-of-function mutations in either PKD1 or PKD2 genes, which encode polycystin-1 (TRPP1) and polycystin-2 (TRPP2), respectively. Increased activity of the mammalian target of rapamycin (mTOR) pathway has been shown in PKD1 mutants but is less documented for PKD2 mutants. Clinical trials using mTOR inhibitors were disappointing, while the AMP-activated kinase (AMPK) activator, metformin is not yet tested in patients. Here, we studied the mTOR activity and its upstream pathways in several human and mouse renal cell models with either siRNA or stable knockdown and with overexpression of TRPP2. Our data reveal for the first time differences between TRPP1 and TRPP2 deficiency. In contrast to TRPP1 deficiency, TRPP2-deficient cells did neither display excessive activation of the mTOR-kinase complex nor inhibition of AMPK activity, while ERK1/2 and Akt activity were similarly affected among TRPP1- and TRPP2-deficient cells. Furthermore, cell proliferation was more pronounced in TRPP1 than in TRPP2-deficient cells. Interestingly, combining low concentrations of rapamycin and metformin was more effective for inhibiting mTOR complex 1 activity in TRPP1-deficient cells than either drug alone. Our results demonstrate a synergistic effect of a combination of low concentrations of drugs suppressing the increased mTOR activity in TRPP1-deficient cells. This novel insight can be exploited in future clinical trials to optimize the efficiency and avoiding side effects of drugs in the treatment of ADPKD patients with PKD1 mutations. Furthermore, as TRPP2 deficiency by itself did not affect mTOR signaling, this may underlie the differences in phenotype, and genetic testing has to be considered for selecting patients for the ongoing trials.


Asunto(s)
Metformina/farmacología , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Canales Catiónicos TRPP/deficiencia , Animales , Sinergismo Farmacológico , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA