Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Cells ; 12(3)2023 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-36766751

RESUMEN

Through kidney transplantation, ischemia/reperfusion is known to induce tissular injury due to cell energy shortage, oxidative stress, and endoplasmic reticulum (ER) stress. ER stress stems from an accumulation of unfolded or misfolded proteins in the lumen of ER, resulting in the unfolded protein response (UPR). Adaptive UPR pathways can either restore protein homeostasis or can turn into a stress pathway leading to apoptosis. We have demonstrated that N1-guanyl-1,7-diamineoheptane (GC7), a specific inhibitor of eukaryotic Initiation Factor 5A (eIF5A) hypusination, confers an ischemic protection of kidney cells by tuning their metabolism and decreasing oxidative stress, but its role on ER stress was unknown. To explore this, we used kidney cells pretreated with GC7 and submitted to either warm or cold anoxia. GC7 pretreatment promoted cell survival in an anoxic environment concomitantly to an increase in xbp1 splicing and BiP level while eiF2α phosphorylation and ATF6 nuclear level decreased. These demonstrated a specific modulation of UPR pathways. Interestingly, the pharmacological inhibition of xbp1 splicing reversed the protective effect of GC7 against anoxia. Our results demonstrated that eIF5A hypusination inhibition modulates distinctive UPR pathways, a crucial mechanism for the protection against anoxia/reoxygenation.


Asunto(s)
Estrés del Retículo Endoplásmico , Isquemia , Riñón , Factores de Iniciación de Péptidos , Daño por Reperfusión , Humanos , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/fisiología , Hipoxia/genética , Hipoxia/metabolismo , Isquemia/genética , Isquemia/metabolismo , Riñón/irrigación sanguínea , Riñón/metabolismo , Factores de Iniciación de Péptidos/genética , Factores de Iniciación de Péptidos/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Respuesta de Proteína Desplegada , Factor 5A Eucariótico de Iniciación de Traducción
2.
Front Mol Biosci ; 9: 825028, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35252350

RESUMEN

Intracellular pH is a vital parameter that is maintained close to neutrality in all mammalian cells and tissues and acidic in most intracellular compartments. After presenting the main techniques used for intracellular an vesicular pH measurements we will briefly recall the main molecular mechanisms that affect and regulate intracellular pH. Following this we will discuss the large functional redundancy found in the transporters of H+ or acid-base equivalents. For this purpose, we will use mathematical modeling to simulate cellular response to persistent and/or transient acidification, in the presence of different transporters, single or in combination. We will also test the presence or absence of intracellular buffering. This latter section will highlight how modeling can yield fundamental insight into deep biological questions such as the utility of functional redundancy in natural selection.

3.
Cell Biosci ; 11(1): 219, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34952646

RESUMEN

Since the demonstration of its involvement in cell proliferation, the eukaryotic initiation factor 5A (eIF5A) has been studied principally in relation to the development and progression of cancers in which the isoform A2 is mainly expressed. However, an increasing number of studies report that the isoform A1, which is ubiquitously expressed in normal cells, exhibits novel molecular features that reveal its new relationships between cellular functions and organ homeostasis. At a first glance, eIF5A can be regarded, among other things, as a factor implicated in the initiation of translation. Nevertheless, at least three specificities: (1) its extreme conservation between species, including plants, throughout evolution, (2) its very special and unique post-translational modification through the activating-hypusination process, and finally (3) its close relationship with the polyamine pathway, suggest that the role of eIF5A in living beings remains to be uncovered. In fact, and beyond its involvement in facilitating the translation of proteins containing polyproline residues, eIF5A is implicated in various physiological processes including ischemic tolerance, metabolic adaptation, aging, development, and immune cell differentiation. These newly discovered physiological properties open up huge opportunities in the clinic for pathologies such as, for example, the ones in which the oxygen supply is disrupted. In this latter case, organ transplantation, myocardial infarction or stroke are concerned, and the current literature defines eIF5A as a new drug target with a high level of potential benefit for patients with these diseases or injuries. Moreover, the recent use of genomic and transcriptomic association along with metadata studies also revealed the implication of eIF5A in genetic diseases. Thus, this review provides an overview of eIF5A from its molecular mechanism of action to its physiological roles and the clinical possibilities that have been recently reported in the literature.

4.
Cancers (Basel) ; 13(10)2021 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-34065348

RESUMEN

Signaling, proliferation, and inflammation are dependent on K63-linked ubiquitination-conjugation of a chain of ubiquitin molecules linked via lysine 63. However, very little information is currently available about how K63-linked ubiquitination is subverted in cancer. The present study provides, for the first time, evidence that cadmium (Cd), a widespread environmental carcinogen, is a potent activator of K63-linked ubiquitination, independently of oxidative damage, activation of ubiquitin ligase, or proteasome impairment. We show that Cd induces the formation of protein aggregates that sequester and inactivate cylindromatosis (CYLD) and selective autophagy, two tumor suppressors that deubiquitinate and degrade K63-ubiquitinated proteins, respectively. The aggregates are constituted of substrates of selective autophagy-SQSTM1, K63-ubiquitinated proteins, and mitochondria. These protein aggregates also cluster double-membrane remnants, which suggests an impairment in autophagosome maturation. However, failure to eliminate these selective cargos is not due to alterations in the general autophagy process, as degradation of long-lived proteins occurs normally. We propose that the simultaneous disruption of CYLD and selective autophagy by Cd feeds a vicious cycle that further amplifies K63-linked ubiquitination and downstream activation of the NF-κB pathway, processes that support cancer progression. These novel findings link together impairment of selective autophagy, K63-linked ubiquitination, and carcinogenesis.

5.
Free Radic Biol Med ; 169: 258-270, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33892115

RESUMEN

Disorders characterized by ischemia/reperfusion (I/R) are the most common causes of debilitating diseases and death in stroke, cardiovascular ischemia, acute kidney injury or organ transplantation. In the latter example the I/R step defines both the amplitude of the damages to the graft and the functional recovery outcome. During transplantation the kidney is subjected to blood flow arrest followed by a sudden increase in oxygen supply at the time of reperfusion. This essential clinical protocol causes massive oxidative stress which is at the basis of cell death and tissue damage. The involvement of both reactive oxygen species (ROS) and nitric oxides (NO) has been shown to be a major cause of these cellular damages. In fact, in non-physiological situations, these species escape endogenous antioxidant control and dangerously accumulate in cells. In recent years, the objective has been to find clinical and pharmacological treatments to reduce or prevent the appearance of oxidative stress in ischemic pathologies. This is very relevant because, due to the increasing success of organ transplantation, clinicians are required to use limit organs, the preservation of which against oxidative stress is crucial for a better outcome. This review highlights the key actors in oxidative stress which could represent new pharmacological targets.


Asunto(s)
Trasplante de Riñón , Daño por Reperfusión , Antioxidantes/uso terapéutico , Humanos , Estrés Oxidativo , Especies Reactivas de Oxígeno
6.
Cell Death Dis ; 12(4): 283, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731685

RESUMEN

Inhibition of the eukaryotic initiation factor 5A activation by the spermidine analogue GC7 has been shown to protect proximal cells and whole kidneys against an acute episode of ischaemia. The highlighted mechanism involves a metabolic switch from oxidative phosphorylation toward glycolysis allowing cells to be transiently independent of oxygen supply. Here we show that GC7 decreases protein expression of the renal GLUT1 glucose transporter leading to a decrease in transcellular glucose flux. At the same time, GC7 modifies the native energy source of the proximal cells from glutamine toward glucose use. Thus, GC7 acutely and reversibly reprogrammes function and metabolism of kidney cells to make glucose its single substrate, and thus allowing cells to be oxygen independent through anaerobic glycolysis. The physiological consequences are an increase in the renal excretion of glucose and lactate reflecting a decrease in glucose reabsorption and an increased glycolysis. Such a reversible reprogramming of glucose handling and oxygen dependence of kidney cells by GC7 represents a pharmacological opportunity in ischaemic as well as hyperglycaemia-associated pathologies from renal origin.


Asunto(s)
Glucosa/metabolismo , Riñón/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Masculino , Ratones , Factor 5A Eucariótico de Iniciación de Traducción
8.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33400689

RESUMEN

The role of insulin receptor (IR) activated by hyperinsulinemia in obesity-induced kidney injury is not well understood. We hypothesized that activation of kidney proximal tubule epithelial IR contributes to obesity-induced kidney injury. We administered normal-fat diet (NFD) or high-fat diet (HFD) to control and kidney proximal tubule IR-knockout (KPTIRKO) mice for 4 months. Renal cortical IR expression was decreased by 60% in male and female KPTIRKO mice. Baseline serum glucose, serum creatinine, and the ratio of urinary albumin to creatinine (ACR) were similar in KPTIRKO mice compared to those of controls. On HFD, weight gain and increase in serum cholesterol were similar in control and KPTIRKO mice; blood glucose did not change. HFD increased the following parameters in the male control mice: renal cortical contents of phosphorylated IR and Akt, matrix proteins, urinary ACR, urinary kidney injury molecule-1-to-creatinine ratio, and systolic blood pressure. Renal cortical generation of hydrogen sulfide was reduced in HFD-fed male control mice. All of these parameters were ameliorated in male KPTIRKO mice. Interestingly, female mice were resistant to HFD-induced kidney injury in both genotypes. We conclude that HFD-induced kidney injury requires renal proximal tubule IR activation in male mice.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Túbulos Renales Proximales/metabolismo , Receptor de Insulina/metabolismo , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Animales , Epitelio/metabolismo , Femenino , Sulfuro de Hidrógeno/metabolismo , Resistencia a la Insulina , Corteza Renal/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/complicaciones , Obesidad/metabolismo , Receptor de Insulina/deficiencia , Receptor de Insulina/genética , Factores Sexuales , Transducción de Señal
9.
Int J Mol Sci ; 23(1)2021 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-35008578

RESUMEN

Lesions issued from the ischemia/reperfusion (I/R) stress are a major challenge in human pathophysiology. Of human organs, the kidney is highly sensitive to I/R because of its high oxygen demand and poor regenerative capacity. Previous studies have shown that targeting the hypusination pathway of eIF5A through GC7 greatly improves ischemic tolerance and can be applied successfully to kidney transplants. The protection process correlates with a metabolic shift from oxidative phosphorylation to glycolysis. Because the protein kinase B Akt is involved in ischemic protective mechanisms and glucose metabolism, we looked for a link between the effects of GC7 and Akt in proximal kidney cells exposed to anoxia or the mitotoxic myxothiazol. We found that GC7 treatment resulted in impaired Akt phosphorylation at the Ser473 and Thr308 sites, so the effects of direct Akt inhibition as a preconditioning protocol on ischemic tolerance were investigated. We evidenced that Akt inhibitors provide huge protection for kidney cells against ischemia and myxothiazol. The pro-survival effect of Akt inhibitors, which is reversible, implied a decrease in mitochondrial ROS production but was not related to metabolic changes or an antioxidant defense increase. Therefore, the inhibition of Akt can be considered as a preconditioning treatment against ischemia.


Asunto(s)
Hipoxia/tratamiento farmacológico , Riñón/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Animales , Antioxidantes/farmacología , Células Cultivadas , Hipoxia/metabolismo , Precondicionamiento Isquémico/métodos , Riñón/metabolismo , Metacrilatos/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación/efectos de los fármacos , Sustancias Protectoras/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Tiazoles/farmacología
10.
J Cereb Blood Flow Metab ; 41(5): 1080-1090, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32615885

RESUMEN

In eukaryotes, the polyamine pathway generates spermidine that activates the hypusination of the translation factor eukaryotic initiation factor 5A (eIF5A). Hypusinated-eIF5A modulates translation, elongation, termination and mitochondrial function. Evidence in model organisms like drosophila suggests that targeting polyamines synthesis might be of interest against ischemia. However, the potential of targeting eIF5A hypusination in stroke, the major therapeutic challenge specific to ischemia, is currently unknown. Using in vitro models of ischemic-related stress, we documented that GC7, a specific inhibitor of a key enzyme in the eIF5A activation pathway, affords neuronal protection. We identified the preservation of mitochondrial function and thereby the prevention of toxic ROS generation as major processes of GC7 protection. To represent a thoughtful opportunity of clinical translation, we explored whether GC7 administration reduces the infarct volume and functional deficits in an in vivo transient focal cerebral ischemia (tFCI) model in mice. A single GC7 pre- or post-treatment significantly reduces the infarct volume post-stroke. Moreover, GC7-post-treatment significantly improves mouse performance in the rotarod and Morris water-maze, highlighting beneficial effects on motor and cognitive post-stroke deficits. Our results identify the targeting of the polyamine-eIF5A-hypusine axis as a new therapeutic opportunity and new paradigm of research in stroke and ischemic diseases.


Asunto(s)
Guanina/análogos & derivados , Lisina/análogos & derivados , Mitocondrias/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/antagonistas & inhibidores , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Accidente Cerebrovascular/terapia , Animales , Conducta Animal/efectos de los fármacos , Cognición/efectos de los fármacos , Guanina/administración & dosificación , Guanina/farmacología , Guanina/uso terapéutico , Inyecciones Intraperitoneales , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/prevención & control , Lisina/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/ultraestructura , Modelos Animales , Neuroprotección/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Factores de Iniciación de Péptidos/efectos de los fármacos , Poliaminas/metabolismo , Proteínas de Unión al ARN/efectos de los fármacos , Especies Reactivas de Oxígeno/toxicidad , Accidente Cerebrovascular/metabolismo , Factor 5A Eucariótico de Iniciación de Traducción
11.
Nephrol Dial Transplant ; 36(6): 988-997, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33367789

RESUMEN

BACKGROUND: The nicotinamide adenine dinucleotide phosphate oxidase isoform 4 (Nox4) mediates reactive oxygen species (ROS) production and renal fibrosis in diabetic kidney disease (DKD) at the level of the podocyte. However, the mitochondrial localization of Nox4 and its role as a mitochondrial bioenergetic sensor has recently been reported. Whether Nox4 drives pathology in DKD within the proximal tubular compartment, which is densely packed with mitochondria, is not yet known. METHODS: We generated a proximal tubular-specific Nox4 knockout mouse model by breeding Nox4flox/flox mice with mice expressing Cre recombinase under the control of the sodium-glucose cotransporter-2 promoter. Subsets of Nox4ptKO mice and their Nox4flox/flox littermates were injected with streptozotocin (STZ) to induce diabetes. Mice were followed for 20 weeks and renal injury was assessed. RESULTS: Genetic ablation of proximal tubular Nox4 (Nox4ptKO) resulted in no change in renal function and histology. Nox4ptKO mice and Nox4flox/flox littermates injected with STZ exhibited the hallmarks of DKD, including hyperfiltration, albuminuria, renal fibrosis and glomerulosclerosis. Surprisingly, diabetes-induced renal injury was not improved in Nox4ptKO STZ mice compared with Nox4flox/flox STZ mice. Although diabetes conferred ROS overproduction and increased the mitochondrial oxygen consumption rate, proximal tubular deletion of Nox4 did not normalize oxidative stress or mitochondrial bioenergetics. CONCLUSIONS: Taken together, these results demonstrate that genetic deletion of Nox4 from the proximal tubules does not influence DKD development, indicating that Nox4 localization within this highly energetic compartment is dispensable for chronic kidney disease pathogenesis in the setting of diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Animales , Nefropatías Diabéticas/genética , Riñón , Túbulos Renales , Túbulos Renales Proximales , Ratones , NADP , NADPH Oxidasa 4/genética , NADPH Oxidasas/genética , Especies Reactivas de Oxígeno
12.
Am J Transplant ; 20(12): 3326-3340, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32400964

RESUMEN

The eIF5A hypusination inhibitor GC7 (N1-guanyl-1,7-diaminoheptane) was shown to protect from ischemic injuries. We hypothesized that GC7 could be useful for preconditioning kidneys from donors before transplantation. Using a preclinical porcine brain death (BD) donation model, we carried out in vivo evaluation of GC7 pretreatment (3 mg/kg iv, 5 minutes after BD) at the beginning of the 4h-donor management, after which kidneys were collected and cold-stored (18h in University of Wisconsin solution) and 1 was allotransplanted. Groups were defined as following (n = 6 per group): healthy (CTL), untreated BD (Vehicle), and GC7-treated BD (Vehicle + GC7). At the end of 4h-management, GC7 treatment decreased BD-induced markers, as radical oxygen species markers. In addition, GC7 increased expression of mitochondrial protective peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC1α) and antioxidant proteins (superoxyde-dismutase-2, heme oxygenase-1, nuclear factor [erythroid-derived 2]-like 2 [NRF2], and sirtuins). At the end of cold storage, GC7 treatment induced an increase of NRF2 and PGC1α mRNA and a better mitochondrial integrity/homeostasis with a decrease of dynamin- related protein-1 activation and increase of mitofusin-2. Moreover, GC7 treatment significantly improved kidney outcome during 90 days follow-up after transplantation (fewer creatininemia and fibrosis). Overall, GC7 treatment was shown to be protective for kidneys against BD-induced injuries during donor management and subsequently appeared to preserve antioxidant defenses and mitochondria homeostasis; these protective effects being accompanied by a better transplantation outcome.


Asunto(s)
Trasplante de Riñón , Daño por Reperfusión , Adenosina , Alopurinol , Animales , Muerte Encefálica , Glutatión , Insulina , Riñón/metabolismo , Trasplante de Riñón/efectos adversos , Soluciones Preservantes de Órganos , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN , Rafinosa , Daño por Reperfusión/etiología , Daño por Reperfusión/prevención & control , Porcinos
13.
Med Sci (Paris) ; 36(2): 147-152, 2020 Feb.
Artículo en Francés | MEDLINE | ID: mdl-32129751

RESUMEN

Ischemia is one of the major concerns of clinicians who are constantly confronted with it, both in surgical and pathological aspects. The consequences of ischemic stress are dramatic and can lead to organic, motor or cognitive disabilities. Currently, there is no identified specific molecular target whose targeting could be beneficial in this area. The drosophila melanogaster fly, used as a model animal, has made it possible to highlight a major advance by allowing the identification of a completely new pharmacological target whose inhibition significantly increases tolerance to hypoxia. Applied to a preclinical model of renal transplantation, this new approach significantly improves the functional recovery of the graft in the long term. This mini-synthesis retraces the steps that made it possible to transfer to higher mammals a concept highlighted in Drosophila that clearly shows, beyond basic research, the contribution that a model organism can make to the clinic.


TITLE: Un nouveau paradigme dans le traitement de l'ischémie - L'apport de la drosophile. ABSTRACT: L'ischémie est une des préoccupations majeures des cliniciens qui y sont confrontés en permanence, que ce soit aussi bien d'un point de vue chirurgical que pathologique. Les conséquences des stress ischémiques sont dramatiques et peuvent conduire à des incapacités organiques, motrices ou cognitives. Actuellement il n'existe aucune cible moléculaire identifiée dont le ciblage pourrait être bénéfique dans ce domaine. La mouche Drosophila melanogaster, utilisée comme animal modèle, a conduit à une avancée majeure en permettant l'identification d'une toute nouvelle cible pharmacologique dont l'inhibition augmente de façon importante la tolérance à l'hypoxie. Appliqué à un modèle préclinique de transplantation rénale, cette nouvelle approche améliore nettement la reprise fonctionnelle du greffon à long terme. Cette revue retrace les étapes qui ont permis de transférer chez le mammifère supérieur une cible mise en évidence chez la drosophile, ce qui montre clairement, au-delà de la recherche fondamentale, l'apport que peut apporter un organisme modèle à la clinique.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila melanogaster , Isquemia/terapia , Animales , Humanos , Isquemia/patología , Mamíferos , Investigación Biomédica Traslacional
14.
J Endocrinol ; 244(1): 189-200, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31697642

RESUMEN

Enhanced beta cell glycolytic and oxidative metabolism are necessary for glucose-induced insulin secretion. While several microRNAs modulate beta cell homeostasis, miR-375 stands out as it is highly expressed in beta cells where it regulates beta cell function, proliferation and differentiation. As glucose metabolism is central in all aspects of beta cell functioning, we investigated the role of miR-375 in this process using human and rat islets; the latter being an appropriate model for in-depth investigation. We used forced expression and repression of mR-375 in rat and human primary islet cells followed by analysis of insulin secretion and metabolism. Additionally, miR-375 expression and glucose-induced insulin secretion were compared in islets from rats at different developmental ages. We found that overexpressing of miR-375 in rat and human islet cells blunted insulin secretion in response to glucose but not to α-ketoisocaproate or KCl. Further, miR-375 reduced O2 consumption related to glycolysis and pyruvate metabolism, but not in response to α-ketoisocaproate. Concomitantly, lactate production was augmented suggesting that glucose-derived pyruvate is shifted away from mitochondria. Forced miR-375 expression in rat or human islets increased mRNA levels of pyruvate dehydrogenase kinase-4, but decreased those of pyruvate carboxylase and malate dehydrogenase1. Finally, reduced miR-375 expression was associated with maturation of fetal rat beta cells and acquisition of glucose-induced insulin secretion function. Altogether our findings identify miR-375 as an efficacious regulator of beta cell glucose metabolism and of insulin secretion, and could be determinant to functional beta cell developmental maturation.


Asunto(s)
Glucosa/metabolismo , Secreción de Insulina/genética , MicroARNs/metabolismo , Transducción de Señal/genética , Adulto , Animales , Femenino , Humanos , Islotes Pancreáticos/metabolismo , Masculino , Ratas , Ratas Wistar
15.
Cell Death Dis ; 10(12): 925, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804464

RESUMEN

Volume-regulated anion channels (VRAC) are chloride channels activated in response to osmotic stress to regulate cellular volume and also participate in other cellular processes, including cell division and cell death. Recently, members of the LRRC8 family have been identified as the main contributors of VRAC conductance. LRRC8/VRAC is permeable to chloride ions but also exhibits significant permeability to various substrates that vary strongly in charge and size. In this study, we explored the intriguing ability of LRRC8/VRAC to transport glutathione (GSH), the major cellular reactive oxygen species (ROS) scavenger, and its involvement in epithelial-to-mesenchymal transition (EMT), a cellular process in which cellular oxidative status is a crucial step. First, in HEK293-WT cells, we showed that a hypotonic condition induced LRRC8/VRAC-dependent GSH conductance (PGSH/PCl of ~0.1) and a marked decrease in intracellular GSH content. GSH currents and GSH intracellular decrease were both inhibited by DCPIB, an inhibitor of LRRC8/VRAC, and were not observed in HEK293-LRRC8A KO cells. Then, we induced EMT by exposing renal proximal tubule epithelial cells to the pleiotropic growth factor TGFß1, and we measured the contribution of LRRC8/VRAC in this process by measuring (i) EMT marker expression (assessed both at the gene and protein levels), (ii) cell morphology and (iii) the increase in migration ability. Interestingly, pharmacologic targeting of LRRC8/VRAC (DCPIB) or RNA interference-mediated inhibition (LRRC8A siRNA) attenuated the TGFß1-induced EMT response by controlling GSH and ROS levels. Interestingly, TGFß1 exposure triggered DCPIB-sensitive chloride conductance. These results suggest that LRRC8/VRAC, due to its native permeability to GSH and thus its ability to modulate ROS levels, plays a critical role in EMT and might contribute to other physiological and pathophysiological processes associated with oxidative stress.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Glutatión/metabolismo , Proteínas de la Membrana/genética , Factor de Crecimiento Transformador beta1/genética , Animales , Aniones/metabolismo , Glutatión/genética , Células HEK293 , Humanos , Presión Osmótica/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Canales Aniónicos Dependientes del Voltaje/genética , Canales Aniónicos Dependientes del Voltaje/metabolismo
16.
Hypertension ; 74(3): 526-535, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31352824

RESUMEN

The present study directly tested the hypothesis that the NHE3 (Na+/H+ exchanger 3) in the proximal tubules of the kidney is required for the development of Ang II (angiotensin II)-induced hypertension using PT-Nhe3-/- (proximal tubule-specific NHE3 knockout) mice. Specifically, PT-Nhe3-/- mice were generated using the SGLT2-Cre/Nhe3loxlox approach, whereas Ang II-induced hypertension was studied in 12 groups (n=5-12 per group) of adult male and female wild-type (WT) and PT-Nhe3-/- mice. Under basal conditions, systolic blood pressure, diastolic blood pressure, and mean arterial blood pressure were significantly lower in male and female PT-Nhe3-/- than WT mice (P<0.01). A high pressor, 1.5 mg/kg per day, intraperitoneal or a slow pressor dose of Ang II, 0.5 mg/kg per day, intraperitoneal for 2 weeks significantly increased systolic blood pressure, diastolic blood pressure, and mean arterial blood pressure in male and female WT mice (P<0.01), but the hypertensive response to Ang II was markedly attenuated in male and female PT-Nhe3-/- mice (P<0.01). Ang II impaired the pressure-natriuresis response in WT mice, whereas proximal tubule-specific deletion of NHE3 improved the pressure-natriuresis response in Ang II-infused PT-Nhe3-/- mice (P<0.01). AT1 receptor blocker losartan completely blocked Ang II-induced hypertension in both WT and PT-Nhe3-/- mice (P<0.01). However, inhibition of nitric oxide synthase with L-NG-Nitroarginine methyl ester had no effect on Ang II-induced hypertension in WT or PT-Nhe3-/- mice (not significant). Furthermore, Ang II-induced hypertension was significantly attenuated by an orally absorbable NHE3 inhibitor AVE0657. In conclusion, NHE3 in the proximal tubules of the kidney may be a therapeutical target in hypertension induced by Ang II or with increased NHE3 expression in the proximal tubules.


Asunto(s)
Angiotensina II/farmacología , Túbulos Renales Proximales/metabolismo , Losartán/administración & dosificación , Receptor de Angiotensina Tipo 1/metabolismo , Intercambiador 3 de Sodio-Hidrógeno/genética , Animales , Modelos Animales de Enfermedad , Femenino , Hipertensión/inducido químicamente , Hipertensión/tratamiento farmacológico , Hipertensión/fisiopatología , Inyecciones Intraperitoneales , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Noqueados , Distribución Aleatoria , Valores de Referencia , Intercambiadores de Sodio-Hidrógeno/metabolismo , Resultado del Tratamiento
17.
Expert Opin Ther Targets ; 23(6): 495-509, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31022355

RESUMEN

INTRODUCTION: Renal ischemia-reperfusion injury (IRI) is a significant clinical challenge faced by clinicians in a broad variety of clinical settings such as perioperative and intensive care. Renal IRI induced acute kidney injury (AKI) is a global public health concern associated with high morbidity, mortality, and health-care costs. Areas covered: This paper focuses on the pathophysiology of transplantation-related AKI and recent findings on cellular stress responses at the intersection of 1. The Unfolded protein response; 2. Mitochondrial dysfunction; 3. The benefits of mineralocorticoid receptor antagonists. Lastly, perspectives are offered to the readers. Expert opinion: Renal IRI is caused by a sudden and temporary impairment of blood flow to the organ. Defining the underlying cellular cascades involved in IRI will assist us in the identification of novel interventional targets to attenuate IRI with the potential to improve transplantation outcomes. Targeting mitochondrial function and cellular bioenergetics upstream of cellular damage may offer several advantages compared to targeting downstream inflammatory and fibrosis processes. An improved understanding of the cellular pathophysiological mechanisms leading to kidney injury will hopefully offer improved targeted therapies to prevent and treat the injury in the future.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Trasplante de Riñón/efectos adversos , Daño por Reperfusión/tratamiento farmacológico , Lesión Renal Aguda/etiología , Lesión Renal Aguda/fisiopatología , Animales , Humanos , Antagonistas de Receptores de Mineralocorticoides/farmacología , Mitocondrias/patología , Terapia Molecular Dirigida , Daño por Reperfusión/etiología , Daño por Reperfusión/fisiopatología
18.
Hypertension ; 72(6): 1328-1336, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30571224

RESUMEN

The present study directly tested the hypothesis that deletion of the NHE3 (Na+/H+ exchanger 3) selectively in the proximal tubules of the kidney lowers basal blood pressure by increasing the pressure-natriuresis response in mice. Adult male and female, age-matched wild-type (WT) littermates and proximal tubule-specific NHE3 knockout mice (PT- Nhe3-/-; n=6-16 per group) were studied for (1) basal phenotypes of electrolytes and pH, blood pressure, and kidney function; (2) the pressure-natriuresis response using the mesenteric, celiac, and abdominal arterial occlusion technique; and (3) the natriuretic responses to acute saline expansion (0.9% NaCl, 10% body weight, intraperitoneal) or 2-week of 2% NaCl diet. Under basal conditions, PT- Nhe3-/- mice showed significantly lower systolic, diastolic, and mean arterial blood pressure ( P<0.01) than WT mice ( P<0.01). PT- Nhe3-/- mice also exhibited significantly greater diuretic ( P<0.01) and natriuretic responses than WT mice ( P<0.01), without altering 24-hour fecal Na+ excretion, plasma pH, Na+, and bicarbonate levels. In response to increased renal perfusion pressure by 30 mm Hg, the pressure-natriuresis response increased 5-fold in WT mice ( P<0.01), but it increased 8-fold in PT- Nhe3-/- mice ( P<0.01). In response to 10% acute saline expansion or 2-week 2% NaCl diet, more pronounced natriuretic responses were demonstrated in PT- Nhe3-/- than WT mice ( P<0.01). Our results support the scientific premise and physiological relevance that NHE3 in the proximal tubules plays an essential role in maintaining basal blood pressure homeostasis, and genetic deletion of NHE3 selectively in the proximal tubules of the kidney lowers blood pressure by increasing the pressure natriuretic response.


Asunto(s)
Presión Sanguínea/fisiología , Túbulos Renales Proximales/metabolismo , Natriuresis/fisiología , Intercambiador 3 de Sodio-Hidrógeno/metabolismo , Animales , Riñón/metabolismo , Ratones , Ratones Noqueados , Cloruro de Sodio Dietético , Intercambiador 3 de Sodio-Hidrógeno/genética
19.
Cell Rep ; 22(13): 3468-3479, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29590616

RESUMEN

The tumor suppressor p53 limits tumorigenesis by inducing apoptosis, cell cycle arrest, and senescence. Although p53 is known to limit inflammation during tumor development, its role in regulating chronic lung inflammation is less well understood. To elucidate the function of airway epithelial p53 in such inflammation, we subjected genetically modified mice, whose bronchial epithelial club cells lack p53, to repetitive inhalations of lipopolysaccharide (LPS), an exposure that leads to severe chronic bronchitis and airway senescence in wild-type mice. Surprisingly, the club cell p53 knockout mice exhibited reduced airway senescence and bronchitis in response to chronic LPS exposure and were significantly protected from global lung destruction. Furthermore, pharmacological elimination of senescent cells also protected wild-type mice from chronic LPS-induced bronchitis. Our results implicate p53 in induction of club-cell senescence and correlate epithelial cell senescence of chronic airway inflammation and lung destruction.


Asunto(s)
Bronquios/metabolismo , Neumonía/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Bronquios/patología , Senescencia Celular/fisiología , Enfermedad Crónica , Progresión de la Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Neumonía/patología
20.
Front Pharmacol ; 8: 328, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28620305

RESUMEN

Chloride channels play an essential role in a variety of physiological functions and in human diseases. Historically, the field of chloride channels has long been neglected owing to the lack of powerful selective pharmacological agents that are needed to overcome the technical challenge of characterizing the molecular identities of these channels. Recently, members of the LRRC8 family have been shown to be essential for generating the volume-regulated anion channel (VRAC) current, a chloride conductance that governs the regulatory volume decrease (RVD) process. The inhibitory effects of six commonly used chloride channel inhibitors on VRAC/LRRC8-mediated chloride transport were tested in wild-type HEK-293 cells expressing LRRC8 proteins and devoid of other types of chloride channels (CFTR and ANO1/2). We explored the effectiveness of the inhibitors using the patch-clamp whole-cell approach and fluorescence-based quantification of cellular volume changes during hypotonic challenge. Both DCPIB and NFA inhibited VRAC current in a whole-cell configuration, with IC50 values of 5 ± 1 µM and 55 ± 2 µM, respectively. Surprisingly, GlyH-101 and PPQ-102, two CFTR inhibitors, also inhibited VRAC conductance at concentrations in the range of their current use, with IC50 values of 10 ± 1 µM and 20 ± 1 µM, respectively. T16Ainh-A01, a so-called specific inhibitor of calcium-activated Cl- conductance, blocked the chloride current triggered by hypo-osmotic challenge, with an IC50 of 6 ± 1 µM. Moreover, RVD following hypotonic challenge was dramatically reduced by these inhibitors. CFTRinh-172 was the only inhibitor that had almost no effect on VRAC/LRRC8-mediated chloride conductance. All inhibitors tested except CFTRinh-172 inhibited VRAC/LRRC8-mediated chloride conductance and cellular volume changes during hypotonic challenge. These results shed light on the apparent lack of chloride channel inhibitors specificity and raise the question of how these inhibitors actually block chloride conductances.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...