Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Eur J Ophthalmol ; : 11206721241226735, 2024 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-38263930

RESUMO

BACKGROUND: Central Serous Chorioretinopathy (CSCR) manifests as fluid accumulation between the neurosensory retina and the retinal pigment epithelium (RPE). Elevated levels of steroid hormones have been implicated in CSCR pathogenesis. This investigation aims to delineate the gene expression patterns of CSCR-associated risk and steroid receptors across human choroidal cell types and RPE cells to discern potential underlying mechanisms. METHODS: This study utilized a comprehensive query of transcriptomic data derived from non-pathological human choroid and RPE cells. FINDINGS: CSCR-associated genes such as PTPRB, CFH, and others are predominantly expressed in the choroidal endothelium as opposed to the RPE. The androgen receptor, encoded by the AR gene, demonstrates heightened expression in the macular endothelium compared to peripheral regions, unlike other steroid receptor genes. AR-expressing endothelial cells display an augmented responsiveness to Transforming growth factor beta (TGF-ß), indicating a propensity towards endothelial to mesenchymal transition (endMT) transcriptional profiling. INTERPRETATION: These results highlight the proclivity of CSCR to manifest primarily within the choroidal vasculature rather than the RPE, suggesting its categorization as a vascular eye disorder. This study accentuates the pivotal role of androgenic steroids, in addition to glucocorticoids. The observed linkage to TGF-ß-mediated endMT provides a potential mechanistic insight into the disease's etiology.

2.
Proc Natl Acad Sci U S A ; 121(4): e2317344121, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38241440

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic cause of chronic kidney disease and the fourth leading cause of end-stage kidney disease, accounting for over 50% of prevalent cases requiring renal replacement therapy. There is a pressing need for improved therapy for ADPKD. Recent insights into the pathophysiology of ADPKD revealed that cyst cells undergo metabolic changes that up-regulate aerobic glycolysis in lieu of mitochondrial respiration for energy production, a process that ostensibly fuels their increased proliferation. The present work leverages this metabolic disruption as a way to selectively target cyst cells for apoptosis. This small-molecule therapeutic strategy utilizes 11beta-dichloro, a repurposed DNA-damaging anti-tumor agent that induces apoptosis by exacerbating mitochondrial oxidative stress. Here, we demonstrate that 11beta-dichloro is effective in delaying cyst growth and its associated inflammatory and fibrotic events, thus preserving kidney function in perinatal and adult mouse models of ADPKD. In both models, the cyst cells with homozygous inactivation of Pkd1 show enhanced oxidative stress following treatment with 11beta-dichloro and undergo apoptosis. Co-administration of the antioxidant vitamin E negated the therapeutic benefit of 11beta-dichloro in vivo, supporting the conclusion that oxidative stress is a key component of the mechanism of action. As a preclinical development primer, we also synthesized and tested an 11beta-dichloro derivative that cannot directly alkylate DNA, while retaining pro-oxidant features. This derivative nonetheless maintains excellent anti-cystic properties in vivo and emerges as the lead candidate for development.


Assuntos
Cistos , Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Camundongos , Animais , Rim Policístico Autossômico Dominante/tratamento farmacológico , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Proliferação de Células , Doenças Renais Policísticas/metabolismo , Apoptose , Estresse Oxidativo , Cistos/metabolismo , DNA/metabolismo , Rim/metabolismo , Canais de Cátion TRPP/genética
3.
J Am Soc Nephrol ; 34(1): 110-121, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36270750

RESUMO

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in Pkd1 and Pkd2. They encode the polytopic integral membrane proteins polycystin-1 (PC1) and polycystin-2 (PC2), respectively, which are expressed on primary cilia. Formation of kidney cysts in ADPKD starts when a somatic second hit mechanism inactivates the wild-type Pkd allele. Approximately one quarter of families with ADPDK due to Pkd1 have germline nonsynonymous amino acid substitution (missense) mutations. A subset of these mutations is hypomorphic, retaining some residual PC1 function. Previous studies have shown that the highly conserved Ire1 α -XBP1 pathway of the unfolded protein response can modulate levels of functional PC1 in the presence of mutations in genes required for post-translational maturation of integral membrane proteins. We examine how activity of the endoplasmic reticulum chaperone-inducing transcription factor XBP1 affects ADPKD in a murine model with missense Pkd1 . METHODS: We engineered a Pkd1 REJ domain missense murine model, Pkd1 R2216W , on the basis of the orthologous human hypomorphic allele Pkd1 R2220W , and examined the effects of transgenic activation of XBP1 on ADPKD progression. RESULTS: Expression of active XBP1 in cultured cells bearing PC1 R2216W mutations increased levels and ciliary trafficking of PC1 R2216W . Mice homozygous for Pkd1 R2216W or heterozygous for Pkd1 R2216Win trans with a conditional Pkd1 fl allele exhibit severe ADPKD following inactivation in neonates or adults. Transgenic expression of spliced XBP1 in tubule segments destined to form cysts reduced cell proliferation and improved Pkd progression, according to structural and functional parameters. CONCLUSIONS: Modulating ER chaperone function through XBP1 activity improved Pkd in a murine model of PC1, suggesting therapeutic targeting of hypomorphic mutations.


Assuntos
Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Adulto , Camundongos , Humanos , Animais , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Modelos Animais de Doenças , Doenças Renais Policísticas/metabolismo , Mutação , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo
4.
J Cell Sci ; 133(24)2020 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-33199522

RESUMO

Approximately 15% of autosomal dominant polycystic kidney disease (ADPKD) is caused by variants in PKD2PKD2 encodes polycystin-2, which forms an ion channel in primary cilia and endoplasmic reticulum (ER) membranes of renal collecting duct cells. Elevated internal Ca2+ modulates polycystin-2 voltage-dependent gating and subsequent desensitization - two biophysical regulatory mechanisms that control its function at physiological membrane potentials. Here, we refute the hypothesis that Ca2+ occupancy of the polycystin-2 intracellular EF hand is responsible for these forms of channel regulation, and, if disrupted, results in ADPKD. We identify and introduce mutations that attenuate Ca2+-EF hand affinity but find channel function is unaltered in the primary cilia and ER membranes. We generated two new mouse strains that harbor distinct mutations that abolish Ca2+-EF hand association but do not result in a PKD phenotype. Our findings suggest that additional Ca2+-binding sites within polycystin-2 or Ca2+-dependent modifiers are responsible for regulating channel activity.


Assuntos
Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Animais , Cílios/metabolismo , Motivos EF Hand , Camundongos , Doenças Renais Policísticas/genética , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo
5.
Trends Mol Med ; 22(12): 997-999, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27793600

RESUMO

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 and PKD2, encoding polycystin-1 and polycystin-2, respectively. Optimizing the folding environment for polycystin-1 missense mutations may have a critical effect on the progression of ADPKD in animal models and could potentially lead to tangible therapeutic options for subgroups of ADPKD patients.


Assuntos
Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética , Animais , Descoberta de Drogas , Humanos , Mutação de Sentido Incorreto/efeitos dos fármacos , Rim Policístico Autossômico Dominante/tratamento farmacológico , Dobramento de Proteína/efeitos dos fármacos , Canais de Cátion TRPP/química
6.
Gastroenterology ; 147(6): 1417-28, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25194674

RESUMO

BACKGROUND & AIMS: Patients with cholestatic disease have increased systemic concentrations of bile acids (BAs) and profound pruritus. The G-protein-coupled BA receptor 1 TGR5 (encoded by GPBAR1) is expressed by primary sensory neurons; its activation induces neuronal hyperexcitability and scratching by unknown mechanisms. We investigated whether the transient receptor potential ankyrin 1 (TRPA1) is involved in BA-evoked, TGR5-dependent pruritus in mice. METHODS: Co-expression of TGR5 and TRPA1 in cutaneous afferent neurons isolated from mice was analyzed by immunofluorescence, in situ hybridization, and single-cell polymerase chain reaction. TGR5-induced activation of TRPA1 was studied in in HEK293 cells, Xenopus laevis oocytes, and primary sensory neurons by measuring Ca(2+) signals. The contribution of TRPA1 to TGR5-induced release of pruritogenic neuropeptides, activation of spinal neurons, and scratching behavior were studied using TRPA1 antagonists or Trpa1(-/-) mice. RESULTS: TGR5 and TRPA1 protein and messenger RNA were expressed by cutaneous afferent neurons. In HEK cells, oocytes, and neurons co-expressing TGR5 and TRPA1, BAs caused TGR5-dependent activation and sensitization of TRPA1 by mechanisms that required Gßγ, protein kinase C, and Ca(2+). Antagonists or deletion of TRPA1 prevented BA-stimulated release of the pruritogenic neuropeptides gastrin-releasing peptide and atrial natriuretic peptide B in the spinal cord. Disruption of Trpa1 in mice blocked BA-induced expression of Fos in spinal neurons and prevented BA-stimulated scratching. Spontaneous scratching was exacerbated in transgenic mice that overexpressed TRG5. Administration of a TRPA1 antagonist or the BA sequestrant colestipol, which lowered circulating levels of BAs, prevented exacerbated spontaneous scratching in TGR5 overexpressing mice. CONCLUSIONS: BAs induce pruritus in mice by co-activation of TGR5 and TRPA1. Antagonists of TGR5 and TRPA1, or inhibitors of the signaling mechanism by which TGR5 activates TRPA1, might be developed for treatment of cholestatic pruritus.


Assuntos
Ácidos e Sais Biliares/metabolismo , Colestase/metabolismo , Prurido/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Colestase/complicações , Modelos Animais de Doenças , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Peptídeo Liberador de Gastrina/metabolismo , Células HEK293 , Humanos , Camundongos Knockout , Peptídeos Natriuréticos/metabolismo , Neurônios Aferentes/citologia , Neurônios Aferentes/metabolismo , Nociceptores/metabolismo , Oócitos/citologia , Oócitos/metabolismo , Cultura Primária de Células , Prurido/etiologia , Receptores Acoplados a Proteínas G/genética , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/genética , Xenopus laevis
7.
J Vis Exp ; (89)2014 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-25045853

RESUMO

The described methods can be used to investigate the effect of proteases on ion channels, receptors, and other plasma membrane proteins heterologously expressed in Xenopus laevis oocytes. In combination with site-directed mutagenesis, this approach provides a powerful tool to identify functionally relevant cleavage sites. Proteolytic activation is a characteristic feature of the amiloride-sensitive epithelial sodium channel (ENaC). The final activating step involves cleavage of the channel's γ-subunit in a critical region potentially targeted by several proteases including chymotrypsin and plasmin. To determine the stimulatory effect of these serine proteases on ENaC, the amiloride-sensitive whole-cell current (ΔIami) was measured twice in the same oocyte before and after exposure to the protease using the two-electrode voltage-clamp technique. In parallel to the electrophysiological experiments, a biotinylation approach was used to monitor the appearance of γENaC cleavage fragments at the cell surface. Using the methods described, it was demonstrated that the time course of proteolytic activation of ENaC-mediated whole-cell currents correlates with the appearance of a γENaC cleavage product at the cell surface. These results suggest a causal link between channel cleavage and channel activation. Moreover, they confirm the concept that a cleavage event in γENaC is required as a final step in proteolytic channel activation. The methods described here may well be applicable to address similar questions for other types of ion channels or membrane proteins.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Técnicas de Patch-Clamp/métodos , Serina Proteases/metabolismo , Animais , Biotina/metabolismo , Canais Epiteliais de Sódio/genética , Feminino , Humanos , Mutagênese Sítio-Dirigida , Oócitos/enzimologia , Oócitos/metabolismo , Subunidades Proteicas , Xenopus laevis
8.
J Biol Chem ; 289(27): 19067-78, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24841206

RESUMO

Proteolytic activation is a unique feature of the epithelial sodium channel (ENaC). However, the underlying molecular mechanisms and the physiologically relevant proteases remain to be identified. The serine protease trypsin I can activate ENaC in vitro but is unlikely to be the physiologically relevant activating protease in ENaC-expressing tissues in vivo. Herein, we investigated whether human trypsin IV, a form of trypsin that is co-expressed in several extrapancreatic epithelial cells with ENaC, can activate human ENaC. In Xenopus laevis oocytes, we monitored proteolytic activation of ENaC currents and the appearance of γENaC cleavage products at the cell surface. We demonstrated that trypsin IV and trypsin I can stimulate ENaC heterologously expressed in oocytes. ENaC cleavage and activation by trypsin IV but not by trypsin I required a critical cleavage site (Lys-189) in the extracellular domain of the γ-subunit. In contrast, channel activation by trypsin I was prevented by mutating three putative cleavage sites (Lys-168, Lys-170, and Arg-172) in addition to mutating previously described prostasin (RKRK(178)), plasmin (Lys-189), and neutrophil elastase (Val-182 and Val-193) sites. Moreover, we found that trypsin IV is expressed in human renal epithelial cells and can increase ENaC-mediated sodium transport in cultured human airway epithelial cells. Thus, trypsin IV may regulate ENaC function in epithelial tissues. Our results show, for the first time, that trypsin IV can stimulate ENaC and that trypsin IV and trypsin I activate ENaC by cleavage at distinct sites. The presence of distinct cleavage sites may be important for ENaC regulation by tissue-specific proteases.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Proteólise , Tripsina/metabolismo , Sequência de Aminoácidos , Animais , Azetidinas/farmacologia , Benzilaminas/farmacologia , Sítios de Ligação , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio/química , Canais Epiteliais de Sódio/genética , Espaço Extracelular/metabolismo , Humanos , Rim/metabolismo , Dados de Sequência Molecular , Mutação , Oócitos/metabolismo , Estrutura Terciária de Proteína , Proteólise/efeitos dos fármacos , Xenopus laevis/genética
9.
J Gen Physiol ; 140(4): 375-89, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22966015

RESUMO

Proteolytic activation of the epithelial sodium channel (ENaC) involves cleavage of its γ subunit in a critical region targeted by several proteases. Our aim was to identify cleavage sites in this region that are functionally important for activation of human ENaC by plasmin and chymotrypsin. Sequence alignment revealed a putative plasmin cleavage site in human γENaC (K189) that corresponds to a plasmin cleavage site (K194) in mouse γENaC. We mutated this site to alanine (K189A) and expressed human wild-type (wt) αßγENaC and αßγ(K189A)ENaC in Xenopus laevis oocytes. The γ(K189A) mutation reduced but did not abolish activation of ENaC whole cell currents by plasmin. Mutating a putative prostasin site (γ(RKRK178AAAA)) had no effect on the stimulatory response to plasmin. In contrast, a double mutation (γ(RKRK178AAAA;K189A)) prevented the stimulatory effect of plasmin. We conclude that in addition to the preferential plasmin cleavage site K189, the putative prostasin cleavage site RKRK178 may serve as an alternative site for proteolytic channel activation by plasmin. Interestingly, the double mutation delayed but did not abolish ENaC activation by chymotrypsin. The time-dependent appearance of cleavage products at the cell surface nicely correlated with the stimulatory effect of chymotrypsin on ENaC currents in oocytes expressing wt or double mutant ENaC. Delayed proteolytic activation of the double mutant channel with a stepwise recruitment of so-called near-silent channels was confirmed in single-channel recordings from outside-out patches. Mutating two phenylalanines (FF174) in the vicinity of the prostasin cleavage site prevented proteolytic activation by chymotrypsin. This indicates that chymotrypsin preferentially cleaves at FF174. The close proximity of FF174 to the prostasin site may explain why mutating the prostasin site impedes channel activation by chymotrypsin. In conclusion, this study supports the concept that different proteases have distinct preferences for certain cleavage sites in γENaC, which may be relevant for tissue-specific proteolytic ENaC activation.


Assuntos
Quimotripsina/metabolismo , Canais Epiteliais de Sódio/química , Canais Epiteliais de Sódio/metabolismo , Fibrinolisina/metabolismo , Ativação do Canal Iônico , Motivos de Aminoácidos , Animais , Canais Epiteliais de Sódio/genética , Humanos , Mutação de Sentido Incorreto , Subunidades Proteicas/metabolismo , Proteólise , Serina Endopeptidases/farmacologia , Xenopus
10.
Pflugers Arch ; 464(4): 353-65, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22864553

RESUMO

Proteolytic processing of the amiloride-sensitive epithelial sodium channel (ENaC) by serine proteases is known to be important for channel activation. Inappropriate ENaC activation by proteases may contribute to the pathophysiology of cystic fibrosis and could be involved in sodium retention and the pathogenesis of arterial hypertension in the context of renal disease. We hypothesized that in addition to serine proteases, cathepsin proteases may activate ENaC. Cathepsin proteases belong to the group of cysteine proteases and play a pathophysiological role in inflammatory diseases. Under pathophysiological conditions, cathepsin-S (Cat-S) may reach ENaC in the apical membrane of epithelial cells. The aim of this study was to investigate the effect of purified Cat-S on human ENaC heterologously expressed in Xenopus laevis oocytes and on ENaC-mediated sodium transport in cultured M-1 mouse renal collecting duct cells. We demonstrated that Cat-S activates amiloride-sensitive whole-cell currents in ENaC-expressing oocytes. The stimulatory effect of Cat-S was preserved at pH 5. ENaC stimulation by Cat-S was associated with the appearance of a γENaC cleavage fragment at the plasma membrane indicating proteolytic channel activation. Mutating two valine residues (V182 and V193) in the critical region of γENaC prevented proteolytic activation of ENaC by Cat-S. Pre-incubation of the oocytes with the Cat-S inhibitor morpholinurea-leucine-homophenylalanine-vinylsulfone-phenyl (LHVS) prevented the stimulatory effect of Cat-S on ENaC. In contrast, LHVS had no effect on ENaC activation by the prototypical serine proteases trypsin and chymotrypsin. Cat-S also stimulated ENaC in differentiated renal epithelial cells. These findings demonstrate that the cysteine protease Cat-S can activate ENaC which may be relevant under pathophysiological conditions.


Assuntos
Catepsinas/metabolismo , Agonistas do Canal de Sódio Epitelial/farmacologia , Canais Epiteliais de Sódio/metabolismo , Amilorida/farmacologia , Sequência de Aminoácidos , Animais , Catepsinas/antagonistas & inibidores , Membrana Celular/metabolismo , Quimotripsina/metabolismo , Dipeptídeos/farmacologia , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Canais Epiteliais de Sódio/química , Canais Epiteliais de Sódio/genética , Humanos , Transporte de Íons , Camundongos , Dados de Sequência Molecular , Mutação , Proteólise , Sódio/metabolismo , Sulfonas/farmacologia , Tripsina/metabolismo , Valina/genética , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA