Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(40): 19983-19988, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31527264

RESUMO

Pancreatic ß cells store insulin within secretory granules which undergo exocytosis upon elevation of blood glucose levels. Crinophagy and autophagy are instead responsible to deliver damaged or old granules to acidic lysosomes for intracellular degradation. However, excessive consumption of insulin granules can impair ß cell function and cause diabetes. Atp6ap2 is an essential accessory component of the vacuolar ATPase required for lysosomal degradative functions and autophagy. Here, we show that Cre recombinase-mediated conditional deletion of Atp6ap2 in mouse ß cells causes a dramatic accumulation of large, multigranular vacuoles in the cytoplasm, with reduction of insulin content and compromised glucose homeostasis. Loss of insulin stores and gigantic vacuoles were also observed in cultured insulinoma INS-1 cells upon CRISPR/Cas9-mediated removal of Atp6ap2. Remarkably, these phenotypic alterations could not be attributed to a deficiency in autophagy or acidification of lysosomes. Together, these data indicate that Atp6ap2 is critical for regulating the stored insulin pool and that a balanced regulation of granule turnover is key to maintaining ß cell function and diabetes prevention.


Assuntos
Deleção de Genes , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , ATPases Translocadoras de Prótons/genética , Receptores de Superfície Celular/genética , Animais , Autofagia , Sistemas CRISPR-Cas , Citosol/metabolismo , Feminino , Inativação Gênica , Insulinoma/metabolismo , Lisossomos/metabolismo , Masculino , Camundongos , Fenótipo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Ratos , Receptores de Superfície Celular/metabolismo , Receptores de Estrogênio/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Vacúolos/metabolismo
2.
Pflugers Arch ; 473(8): 1229-1246, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34228176

RESUMO

The ATP6ap2 (Pro)renin receptor protein associates with H+-ATPases which regulate organellar, cellular, and systemic acid-base homeostasis. In the kidney, ATP6ap2 colocalizes with H+-ATPases in various cell types including the cells of the proximal tubule. There, H+-ATPases are involved in receptor-mediated endocytosis of low molecular weight proteins via the megalin/cubilin receptors. To study ATP6ap2 function in the proximal tubule, we used an inducible shRNA Atp6ap2 knockdown rat model (Kd) and an inducible kidney-specific Atp6ap2 knockout mouse model. Both animal lines showed higher proteinuria with elevated albumin, vitamin D binding protein, and procathepsin B in urine. Endocytosis of an injected fluid-phase marker (FITC- dextran, 10 kDa) was normal whereas processing of recombinant transferrin, a marker for receptor-mediated endocytosis, to lysosomes was delayed. While megalin and cubilin expression was unchanged, abundance of several subunits of the H+-ATPase involved in receptor-mediated endocytosis was reduced. Lysosomal integrity and H+-ATPase function are associated with mTOR signaling. In ATP6ap2, KO mice mTOR and phospho-mTOR appeared normal but increased abundance of the LC3-B subunit of the autophagosome was observed suggesting a more generalized impairment of lysosomal function in the absence of ATP6ap2. Hence, our data suggests a role for ATP6ap2 for proximal tubule function in the kidney with a defect in receptor-mediated endocytosis in mice and rats.


Assuntos
Túbulos Renais Proximais/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Receptores de Superfície Celular/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Endocitose , Feminino , Lisossomos/metabolismo , Masculino , Camundongos Knockout , Ratos Transgênicos
3.
Am J Physiol Renal Physiol ; 315(3): F521-F534, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29667908

RESUMO

The prorenin receptor (PRR) was originally proposed to be a member of the renin-angiotensin system (RAS); however, recent work questioned their association. The present paper describes a functional link between the PRR and RAS in the renal juxtaglomerular apparatus (JGA), a classic anatomical site of the RAS. PRR expression was found in the sensory cells of the JGA, the macula densa (MD), and immunohistochemistry-localized PRR to the MD basolateral cell membrane in mouse, rat, and human kidneys. MD cell PRR activation led to MAP kinase ERK1/2 signaling and stimulation of PGE2 release, the classic pathway of MD-mediated renin release. Exogenous renin or prorenin added to the in vitro microperfused JGA-induced acute renin release, which was inhibited by removing the MD or by the administration of a PRR decoy peptide. To test the function of MD PRR in vivo, we established a new mouse model with inducible conditional knockout (cKO) of the PRR in MD cells based on neural nitric oxide synthase-driven Cre-lox recombination. Deletion of the MD PRR significantly reduced blood pressure and plasma renin. Challenging the RAS by low-salt diet + captopril treatment caused further significant reductions in blood pressure, renal renin, cyclooxygenase-2, and microsomal PGE synthase expression in cKO vs. wild-type mice. These results suggest that the MD PRR is essential in a novel JGA short-loop feedback mechanism, which is integrated within the classic MD mechanism to control renin synthesis and release and to maintain blood pressure.


Assuntos
Pressão Sanguínea , Sistema Justaglomerular/enzimologia , ATPases Translocadoras de Prótons/metabolismo , Receptores de Superfície Celular/metabolismo , Sistema Renina-Angiotensina , Renina/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Técnicas Biossensoriais , Pressão Sanguínea/efeitos dos fármacos , Captopril/farmacologia , Ciclo-Oxigenase 2/metabolismo , Dieta Hipossódica , Dinoprostona/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Sistema Justaglomerular/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prostaglandina-E Sintases/metabolismo , ATPases Translocadoras de Prótons/deficiência , ATPases Translocadoras de Prótons/genética , Ratos Sprague-Dawley , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Sistema Renina-Angiotensina/efeitos dos fármacos , Via Secretória , Transdução de Sinais , ATPases Vacuolares Próton-Translocadoras/genética , Receptor de Pró-Renina
4.
Blood ; 126(4): 504-7, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26063165

RESUMO

The (pro)renin receptor (PRR) was originally thought to be important for regulating blood pressure via the renin-angiotensin system. However, it is now emerging that PRR has instead a generic role in cellular development. Here, we have specifically deleted PRR from T cells. T-cell-specific PRR-knockout mice had a significant decrease in thymic cellularity, corresponding with a 100-fold decrease in the number of CD4(+) and CD8(+) thymocytes, and a large increase in double-negative (DN) precursors. Gene expression analysis on sorted DN3 thymocytes indicated that PRR-deficient thymocytes have perturbations in key cellular pathways essential at the DN3 stage, including transcription and translation. Further characterization of DN T-cell progenitors leads us to propose that PRR deletion affects thymocyte survival and development at multiple stages; from DN3 through to DN4, double-positive, and single-positive CD4 and CD8. Our study thus identifies a new role for PRR in T-cell development.


Assuntos
Diferenciação Celular , Receptores de Superfície Celular/fisiologia , Subpopulações de Linfócitos T/citologia , Timócitos/citologia , Animais , Feminino , Citometria de Fluxo , Integrases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timócitos/imunologia , Timócitos/metabolismo , Receptor de Pró-Renina
5.
Am J Physiol Renal Physiol ; 308(5): F487-99, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25503726

RESUMO

The renin-angiotensin system (RAS) is involved in inflammation. The signaling via the ANG II type 1 receptor in human lymphocytes and monocytes, which play key roles in pathophysiology of glomerulonephritis (GN), can enhance inflammation. However, the role of the (pro)renin receptor [(P)RR], a component of the RAS, in inflammatory reactions is unknown. We assessed whether (P)RR is expressed in human lymphocytes and monocytes by RT-PCR, Western blotting, flow cytometry, and immunohistochemistry, and whether (P)RR functions in inflammation. (P)RR mRNA and protein were expressed in human peripheral blood mononuclear cells (PBMCs). Flow cytometric analysis revealed high expression of (P)RR on monocytes. (P)RR was present on PBMCs, infiltrating lymphocytes, and macrophages around glomeruli with a crescent in anti-neutrophil cytoplasmic antibody (ANCA)-associated GN. Renin stimulation of PBMCs from healthy subjects in the presence of the ANG II type 1 receptor and ANG II type 2 receptor blockers induced ERK1/2 phosphorylation and release of IL-6 and expression of cyclooxygenase-2 (COX-2). The increases in cytokine release and COX-2 expression were inhibited in the presence of an ERK1/2 inhibitor. (P)RR knockdown by small interfering RNA in U937 cells, a human leukemic monocyte lymphoma cell line, significantly decreased ERK1/2 phosphorylation after renin stimulation. Thus (P)RR expressed in human inflammatory cells might contribute to inflammation in ANCA-associated GN.


Assuntos
Linfócitos/metabolismo , Monócitos/metabolismo , Receptores de Superfície Celular/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Adulto , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Citometria de Fluxo , Glomerulonefrite/metabolismo , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Fosforilação , Renina , Células U937 , Adulto Jovem
6.
Lab Invest ; 94(8): 863-72, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25046440

RESUMO

Binding of renin and prorenin to the (pro)renin receptor (PRR) increases their enzymatic activity and upregulates the expression of pro-fibrotic genes in vitro. Expression of PRR is increased in the heart and kidney of hypertensive and diabetic animals, but its causative role in organ damage is still unclear. To determine whether increased expression of PRR is sufficient to induce cardiac or renal injury, we generated a mouse that constitutively overexpresses PRR by knocking-in the Atp6ap2/PRR gene in the hprt locus under the control of a CMV immediate early enhancer/chicken beta-actin promoter. Mice were backcrossed in the C57Bl/6 and FVB/N strain and studied at the age of 12 months. In spite of a 25- to 80-fold renal and up to 400-fold cardiac increase in Atp6ap2/PRR expression, we found no differences in systolic blood pressure or albuminuria between wild-type and PRR overexpressing littermates. Histological examination did not show any renal or cardiac fibrosis in mutant mice. This was supported by real-time PCR analysis of inflammatory markers as well as of pro-fibrotic genes in the kidney and collagen in cardiac tissue. To determine whether the concomitant increase of renin would trigger fibrosis, we treated PRR overexpressing mice with the angiotensin receptor-1 blocker losartan over a period of 6 weeks. Renin expression increased eightfold in the kidney but no renal injury could be detected. In conclusion, our results suggest no major role for PRR in organ damage per se or related to its function as a receptor of renin.


Assuntos
Ventrículos do Coração/metabolismo , Hipertensão/metabolismo , Rim/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Receptores de Superfície Celular/metabolismo , Insuficiência Renal/metabolismo , Disfunção Ventricular/metabolismo , Albuminúria/etiologia , Albuminúria/metabolismo , Albuminúria/patologia , Albuminúria/urina , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Feminino , Fibrose , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/patologia , Hemizigoto , Heterozigoto , Homozigoto , Hipertensão/etiologia , Hipertensão/patologia , Hipertensão/urina , Mediadores da Inflamação/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Losartan/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , ATPases Translocadoras de Prótons/genética , Receptores de Superfície Celular/agonistas , Receptores de Superfície Celular/genética , Insuficiência Renal/induzido quimicamente , Insuficiência Renal/etiologia , Insuficiência Renal/patologia , Renina/química , Renina/metabolismo , Regulação para Cima/efeitos dos fármacos , Disfunção Ventricular/induzido quimicamente , Disfunção Ventricular/etiologia , Disfunção Ventricular/patologia , Receptor de Pró-Renina
7.
Am J Physiol Renal Physiol ; 303(7): F1037-48, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22791343

RESUMO

The role of the renin-angiotensin system in chronic kidney disease involves multiple peptides and receptors. Exerting antipodal pathophysiological mechanisms, renin inhibition and AT(1) antagonism ameliorate renal damage. However, it is unclear which mechanism exerts better nephroprotection. We compared the renin inhibitor aliskiren with the AT(1) antagonist losartan in mice with chronic kidney disease due to renal ablation. Doses were adjusted to equipotent inhibition of the renin-angiotensin system, determined via a dose-response quantifying plasma and renal renin expression. Six-week treatment with either 500 mg/l drinking water losartan or 50 mg·kg(-1)·day(-1) aliskiren significantly decreased albuminuria, glomerular damage, and transcription rates of renal injury markers to a similar extent. An array analysis comparing renal gene expression of losartan- and aliskiren-treated mice evaluating >34,000 transcripts demonstrated regulation for 14 genes only, with small differences. No superior nephroprotection was found by combining losartan and aliskiren. Compared with plasma concentrations, aliskiren accumulated ∼7- to 29-fold in the heart, liver, lung, and spleen and ∼156-fold in the kidney. After withdrawal, plasma concentrations dropped to zero within 24 h, whereas renal tissue concentrations declined slowly over days. Withdrawal of aliskiren in mice with chronic kidney disease revealed a significantly delayed re-increase in albuminuria compared with withdrawal of losartan. This study demonstrates equieffective nephroprotection of renin inhibition and AT(1) antagonism in mice with chronic kidney disease without additional benefit of combination therapy. These observations underscore the pivotal role of targeting ANG II to reduce renal injury.


Assuntos
Amidas/uso terapêutico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Fumaratos/uso terapêutico , Rim/efeitos dos fármacos , Losartan/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Sistema Renina-Angiotensina/efeitos dos fármacos , Renina/antagonistas & inibidores , Albuminúria/tratamento farmacológico , Albuminúria/metabolismo , Albuminúria/fisiopatologia , Amidas/farmacologia , Angiotensina II/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fumaratos/farmacologia , Expressão Gênica/efeitos dos fármacos , Rim/metabolismo , Losartan/farmacologia , Masculino , Camundongos , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/fisiopatologia , Renina/metabolismo , Resultado do Tratamento
8.
J Am Soc Nephrol ; 22(12): 2193-202, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22034640

RESUMO

The prorenin receptor (PRR) is highly expressed in podocytes, but its role in the maintenance of podocyte function is unknown. Here we generated podocyte-specific PRR-knockout mice and found that these animals died between 2 to 3 wk after birth. Within 14 d, PRR-knockout mice developed nephrotic syndrome, albuminuria with podocyte foot-process fusion, and cytoskeletal changes. Podocyte-specific PRR deletion also led to disturbed processing of multivesicular bodies and enrichment of autophagosomal (LC3) and lysosomal (LAMP2) markers, indicating a functional block in autophagosome-lysosome fusion and an overload of the proteasomal protein-degradation machinery. In vitro, PRR knockdown and pharmacologic blockade of vacuolar H(+)-ATPases, which associate with the PRR, increased vesicular pH, led to accumulation of LC3-positive and LAMP2-positive vesicles and altered the cytoskeleton. Taken together, these results suggest that the PRR is essential for podocyte function and survival by maintaining autophagy and protein-turnover machinery. Furthermore, PRR contributes to the control of lysosomal pH, which is important for podocyte survival and cytoskeletal integrity.


Assuntos
Autofagia/fisiologia , Podócitos/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Sobrevivência Celular , Feminino , Camundongos , Receptor de Pró-Renina
9.
Clin Sci (Lond) ; 120(5): 169-78, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21087212

RESUMO

PRR [(pro)renin receptor] was named after its biological characteristics, namely the binding of renin and of its inactive precursor prorenin, that triggers intracellular signalling involving ERK (extracellular-signal-regulated kinase) 1/2. However the gene encoding for PRR is named ATP6ap2 (ATPase 6 accessory protein 2) because PRR was initially found as a truncated form co-purifying with V-ATPase (vacuolar H+-ATPase). There are now data showing that this interaction is not only physical, but also functional in the kidney and the heart. However, the newest and most fascinating development of PRR is its involvement in both the canonical Wnt/ß-catenin and non-canonical Wnt/PCP (planar cell polarity) pathways, which are essential for adult and embryonic stem cell biology, embryonic development and disease, including cancer. In the Wnt/ß-catenin pathway, it has been shown that PRR acts as an adaptor between the Wnt receptor LRP5/6 (low-density lipoprotein receptor-related protein 5/6) and Fz (frizzled) and that the proton gradient generated by the V-ATPase in endosomes is necessary for LRP5/6 phosphorylation and ß-catenin activation. In the Wnt/PCP pathway, PRR binds to Fz and controls its asymetrical subcellular distribution and therefore the polarization of the cells in a plane of a tissue. These essential cellular functions of PRR are independent of renin and open new avenues on the pathophysiological role of PRR. The present review will summarize our knowledge of (pro)renin-dependent functions of PRR and will discuss the newly recognized functions of PRR related to the V-ATPase and to Wnt signalling.


Assuntos
Receptores de Superfície Celular/fisiologia , Renina/fisiologia , Animais , Doenças Cardiovasculares/fisiopatologia , Humanos , Nefropatias/fisiopatologia , Transdução de Sinais/fisiologia , ATPases Vacuolares Próton-Translocadoras/fisiologia , Proteínas Wnt/fisiologia , Receptor de Pró-Renina
10.
Curr Hypertens Rep ; 13(1): 79-85, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21125352

RESUMO

The (pro)renin receptor, PRR, was initially characterized as a component of the renin-angiotensin system (RAS). PRR-bound renin and prorenin display increased enzymatic activity, and binding activates intracellular signaling, upregulating the expression of profibrotic proteins. As a consequence, most studies set out to demonstrate a role of PRR in hypertension, cardiovascular and renal diseases, and organ damage, and to identify PRR as a therapeutic target to optimize RAS blockade. The results of animal studies were disappointing and did not convincingly establish PRR as major player in hypertension or in organ damage, although human studies suggested a link between a polymorphism in the PRR gene and blood pressure. New data now suggest that PRR is functionally linked to the vacuolar proton-ATPase and, quite unexpectedly, that PRR is necessary to Wnt signaling pathways that are essential (independently of renin) for adult and embryonic stem cell biology, embryonic development, and diseases including cancer, thereby opening new perspectives on the pathophysiological roles of PRR.


Assuntos
Hipertensão/tratamento farmacológico , Receptores de Superfície Celular/antagonistas & inibidores , Renina/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Humanos , Hipertensão/patologia , Nefropatias/metabolismo , Nefropatias/patologia , Sistema Renina-Angiotensina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , ATPases Vacuolares Próton-Translocadoras/efeitos dos fármacos , ATPases Vacuolares Próton-Translocadoras/metabolismo , Proteínas Wnt/efeitos dos fármacos , Proteínas Wnt/metabolismo , Receptor de Pró-Renina
11.
J Am Soc Nephrol ; 21(1): 18-23, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19917780

RESUMO

The (pro)renin receptor (PRR) binds renin and prorenin, its proenzyme inactive form. Receptor-bound prorenin becomes enzymatically active and binding then activates the MAP kinases ERK1/2 and p38 pathways, leading to upregulation of profibrotic and cyclooxygenase-2 genes independent of angiotensin II generation. These characteristics explain the interest in the potential role of PRR in organ damage in diseases associated with activation of the renin-angiotensin system (RAS), in particular hypertension and diabetes. Although identification of PRR has improved our understanding of the physiology of the tissue RAS, its role in pathology is far from clear. Transgenic animals overexpressing PRR ubiquitously or selectively in smooth-muscle cells develop high BP or glomerulosclerosis, and increased expression of PRR is reported in models of hypertension or kidney damage. However, definitive proof is still lacking for a role for PRR in disease, or by showing improvement of disease by tissue-specific ablation of PRR or by administration of a specific PRR antagonist. Furthermore, the early embryonic lethality seen in PRR-null mice suggests PRR has additional essential cellular functions we do not understand.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Receptores de Superfície Celular/fisiologia , Sistema Renina-Angiotensina/fisiologia , Animais , Modelos Animais de Doenças , Humanos , Hipertensão/fisiopatologia , Nefropatias/fisiopatologia , Camundongos , Camundongos Knockout , Receptores de Superfície Celular/genética , Transdução de Sinais/fisiologia , Receptor de Pró-Renina
12.
Front Cardiovasc Med ; 8: 725203, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35004870

RESUMO

Objective: Elevated plasma cholesterol concentrations contributes to ischemic cardiovascular diseases. Recently, we showed that inhibiting hepatic (pro)renin receptor [(P)RR] attenuated diet-induced hypercholesterolemia and hypertriglyceridemia in low-density lipoprotein receptor (LDLR) deficient mice. The purpose of this study was to determine whether inhibiting hepatic (P)RR could attenuate atherosclerosis. Approach and Results: Eight-week-old male LDLR-/- mice were injected with either saline or N-acetylgalactosamine-modified antisense oligonucleotides (G-ASOs) primarily targeting hepatic (P)RR and were fed a western-type diet (WTD) for 16 weeks. (P)RR G-ASOs markedly reduced plasma cholesterol concentrations from 2,211 ± 146 to 1,128 ± 121 mg/dL. Fast protein liquid chromatography (FPLC) analyses revealed that cholesterol in very low-density lipoprotein (VLDL) and intermediate density lipoprotein (IDL)/LDL fraction were potently reduced by (P)RR G-ASOs. Moreover, (P)RR G-ASOs reduced plasma triglyceride concentrations by more than 80%. Strikingly, despite marked reduction in plasma lipid concentrations, atherosclerosis was not reduced but rather increased in these mice. Further testing in ApoE-/- mice confirmed that (P)RR G-ASOs reduced plasma lipid concentrations but not atherosclerosis. Transcriptomic analysis of the aortas revealed that (P)RR G-ASOs induced the expression of the genes involved in immune responses and inflammation. Further investigation revealed that (P)RR G-ASOs also inhibited (P)RR in macrophages and in enhanced inflammatory responses to exogenous stimuli. Moreover, deleting the (P)RR in macrophages resulted in accelerated atherosclerosis in WTD fed ApoE-/- mice. Conclusion: (P)RR G-ASOs reduced the plasma lipids in atherosclerotic mice due to hepatic (P)RR deficiency. However, augmented pro-inflammatory responses in macrophages due to (P)RR downregulation counteracted the beneficial effects of lowered plasma lipid concentrations on atherosclerosis. Our study demonstrated that hepatic (P)RR and macrophage (P)RR played a counteracting role in atherosclerosis.

13.
J Nephrol ; 23(5): 508-13, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20383869

RESUMO

The discovery of a (pro)renin receptor ((P)RR) and the introduction of renin inhibitors in the clinic has brought prorenin, the inactive proenzyme form of renin, back into the spotlight. The (P)RR binds both renin and its inactive precursor prorenin, and their binding triggers intracellular signaling that up-regulates the expression of profibrotic genes. Furthermore, binding of prorenin unmasks its active site and endows prorenin with angiotensin I-generating activity. Many studies have attempted to establish a link between (P)RR and hypertension, (P)RR and tissue fibrosis associated with hypertension and with diabetic nephropathy. Models of transgenic rats overexpressing (P)RR develop high blood pressure and have glomerulosclerosis, suggesting a link between increased (P)RR and these pathologies, but no definite proof of any role of (P)RR in other models of cardiovascular or renal diseases could be established because of the absence of any specific (P)RR antagonist and of tissue-specific (P)RR null mice. Nevertheless, a study in a large cohort of Japanese men has shown a correlation between a polymorphism in the (P)RR gene and increased ambulatory blood pressure. Finally, a mutation in the (P)RR gene is responsible for mental retardation and epilepsy, indicating that (P)RR is essential during brain development.


Assuntos
Doenças Cardiovasculares/etiologia , Nefropatias/etiologia , Receptores de Superfície Celular/fisiologia , Animais , Humanos , Camundongos , Modelos Animais , Ratos , Receptores de Superfície Celular/química , Transdução de Sinais , Receptor de Pró-Renina
14.
Am J Physiol Regul Integr Comp Physiol ; 297(2): R250-7, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19474391

RESUMO

The (pro)renin receptor [(P)RR] plays a pivotal role in the renin-angiotensin system. Experimental models emphasize the role of (P)RR in organ damage associated with hypertension and diabetes. However, a mutation of the (P)RR gene, resulting in frame deletion of exon 4 [Delta4-(P)RR] is associated with X-linked mental retardation (XLMR) and epilepsy pointing to a novel role of (P)RR in brain development and cognitive function. We have studied (P)RR expression in mouse brain, as well as the effect of transfection of Delta4-(P)RR on neuronal differentiation of rat neuroendocrine PC-12 cells induced by nerve growth factor (NGF). In situ hybridization showed a wide distribution of (P)RR, including in key regions involved in the regulation of blood pressure and body fluid homeostasis. In mouse neurons, the receptor is on the plasma membrane and in synaptic vesicles, and stimulation by renin provokes ERK1/2 phosphorylation. In PC-12 cells, (P)RR localized mainly in the Golgi and in endoplasmic reticulum and redistributed to neurite projections during NGF-induced differentiation. In contrast, Delta4-(P)RR remained cytosolic and inhibited NGF-induced neuronal differentiation and ERK1/2 activation. Cotransfection of PC-12 cells with (P)RR and Delta4-(P)RR cDNA resulted in altered localization of (P)RR and inhibited (P)RR redistribution to neurite projections upon NGF stimulation. Furthermore, (P)RR dimerized with itself and with Delta4-(P)RR, suggesting that the XLMR and epilepsy phenotype resulted from a dominant-negative effect of Delta4-(P)RR, which coexists with normal transcript in affected males. In conclusion, our results show that (P)RR is expressed in mouse brain and suggest that the XLMR and epilepsy phenotype might result from a dominant-negative effect of the Delta4-(P)RR protein.


Assuntos
Diferenciação Celular , Neurônios/citologia , Receptores de Superfície Celular/fisiologia , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Epilepsia/genética , Epilepsia/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Deleção de Genes , Humanos , Deficiência Intelectual Ligada ao Cromossomo X/genética , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Camundongos , Camundongos Endogâmicos , Fator de Crescimento Neural/farmacologia , Neuritos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Organelas/metabolismo , Células PC12 , Multimerização Proteica/fisiologia , Transporte Proteico/efeitos dos fármacos , Células Piramidais/metabolismo , Ratos , Renina/farmacologia , Vesículas Sinápticas/metabolismo , Sinaptofisina/metabolismo , Transfecção , Receptor de Pró-Renina
15.
Arterioscler Thromb Vasc Biol ; 28(6): 1151-7, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18388329

RESUMO

OBJECTIVE: Renin inhibition with aliskiren has been reported to cause a greater rise in renin than other types of renin-angiotensin system blockade, thereby potentially leading to angiotensin generation or stimulation of the human (pro)renin receptor (h(P)RR). Here we studied whether this rise in renin is attributable to an aliskiren-induced change in the prorenin conformation, allowing its detection in renin assays, or a change in renin/prorenin clearance. We also investigated whether aliskiren affects (pro)renin binding to its receptors, using rat aortic vascular smooth muscle cells (VSMCs) overexpressing the h(P)RR. METHODS AND RESULTS: A 48-hour incubation with aliskiren at 4 degrees C converted the prorenin conformation from "closed" to "open," thus allowing its recognition in active site-directed renin assays. VSMCs accumulated (pro)renin through binding to mannose 6-phosphate receptors (M6PRs) and h(P)RRs. Aliskiren did not affect binding at 4 degrees C. At 37 degrees C, aliskiren increased (pro)renin accumulation up to 40-fold, and M6PR blockade prevented this. Aliskiren increased the intracellular half life of prorenin 2 to 3 times. CONCLUSIONS: Aliskiren allows the detection of prorenin as renin, and decreases renin/prorenin clearance. Both phenomena may contribute to the "renin" surge during aliskiren treatment, but because they depend on aliskiren binding, they will not result in angiotensin generation. Aliskiren does not affect (pro)renin binding to its receptors.


Assuntos
Amidas/metabolismo , Anti-Hipertensivos/metabolismo , Aorta/metabolismo , Fumaratos/metabolismo , Músculo Liso Vascular/metabolismo , Renina/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Amidas/farmacologia , Animais , Animais Geneticamente Modificados , Anti-Hipertensivos/farmacologia , Aorta/citologia , Aorta/efeitos dos fármacos , Células Cultivadas , Feminino , Fumaratos/farmacologia , Meia-Vida , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Ligação Proteica , Ratos , Receptor IGF Tipo 2/metabolismo , Receptores de Superfície Celular/metabolismo , Renina/antagonistas & inibidores , ATPases Vacuolares Próton-Translocadoras/metabolismo , Receptor de Pró-Renina
16.
J Mol Med (Berl) ; 86(6): 643-6, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18322668

RESUMO

Two (pro)renin receptors have been characterized so far, the mannose-6-phosphate receptor (M6P-R) and a specific receptor called (P)RR for (pro)renin receptor. Each receptor controls a different aspect of renin and prorenin metabolism. The M6P-R is a clearance receptor, whereas (P)RR mediates their cellular effects by activating intracellular signaling and up-regulating gene expression. Moreover, binding to (P)RR increases renin enzymatic activity and fully activates prorenin, the inactive proenzyme form of renin. Experimental models suggest that increased (P)RR synthesis and/or activation may be relevant to diseases, especially to high blood pressure, to cardiac fibrosis associated with hypertension, and to diabetic nephropathy.


Assuntos
Receptores de Superfície Celular/metabolismo , Animais , Doenças Cardiovasculares/metabolismo , Humanos , Nefropatias/metabolismo , Receptores de Superfície Celular/química , Renina/metabolismo , Receptor de Pró-Renina
17.
Curr Opin Pharmacol ; 8(2): 127-32, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18243793

RESUMO

The (pro)renin receptor [(P)RR] is a single trans-membrane domain receptor that mediates renin and prorenin specific effects. The receptor acts as co-factor for renin and prorenin by increasing their enzymatic activity on the cell-surface and it activates the mitogen activated protein kinases ERK1/2 cascade leading to cell proliferation and to up-regulation of profibrotic genes expression. Studies in genetically modified animals over-expressing (P)RR suggest a direct role for (P)RR cardiovascular and renal pathologies since rats over-expressing (P)RR in vascular smooth-muscle cells develop high blood pressure and those with an ubiquitous over-expression of (P)RR have glomerulosclerosis and proteinuria. A peptide called "handle region peptide" (HRP) mimicking part of the prosegment of prorenin was claimed to block prorenin binding to (P)RR and its activation. The mechanism of action of HRP and its specificity for (P)RR remains very controversial although infusion of this peptide gave spectacular results by preventing diabetic nephropathy in angiotensin II type1a receptor-deficient mice. In contrast to the other components of the renin angiotensin system, (P)RR is necessary to cell survival and proliferation and a mutation of (P)RR is associated with mental retardation and epilepsy, pointing to an essential role of (P)RR in brain development. The (pro)renin receptor is a more complex protein than anticipated and in depth studies of its functions that are likely not restricted to the renin angiotensin system are needed especially in the perspective of the design of a (P)RR blocker.


Assuntos
Receptores de Superfície Celular/metabolismo , Renina/metabolismo , Animais , Doenças Cardiovasculares/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Nefropatias/fisiopatologia , Receptores de Superfície Celular/fisiologia , Sistema Renina-Angiotensina/fisiologia , Receptor de Pró-Renina
18.
J Clin Invest ; 129(5): 2145-2162, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30985297

RESUMO

Vacuolar H+-ATPase-dependent (V-ATPase-dependent) functions are critical for neural proteostasis and are involved in neurodegeneration and brain tumorigenesis. We identified a patient with fulminant neurodegeneration of the developing brain carrying a de novo splice site variant in ATP6AP2 encoding an accessory protein of the V-ATPase. Functional studies of induced pluripotent stem cell-derived (iPSC-derived) neurons from this patient revealed reduced spontaneous activity and severe deficiency in lysosomal acidification and protein degradation leading to neuronal cell death. These deficiencies could be rescued by expression of full-length ATP6AP2. Conditional deletion of Atp6ap2 in developing mouse brain impaired V-ATPase-dependent functions, causing impaired neural stem cell self-renewal, premature neuronal differentiation, and apoptosis resulting in degeneration of nearly the entire cortex. In vitro studies revealed that ATP6AP2 deficiency decreases V-ATPase membrane assembly and increases endosomal-lysosomal fusion. We conclude that ATP6AP2 is a key mediator of V-ATPase-dependent signaling and protein degradation in the developing human central nervous system.


Assuntos
Sistema Nervoso Central/fisiopatologia , Doenças Neurodegenerativas/diagnóstico por imagem , Doenças Neurodegenerativas/genética , Células-Tronco Pluripotentes/metabolismo , Receptores de Superfície Celular/genética , ATPases Vacuolares Próton-Translocadoras/genética , Adolescente , Processamento Alternativo , Animais , Apoptose , Encéfalo/diagnóstico por imagem , Morte Celular , Diferenciação Celular , Sobrevivência Celular , Pré-Escolar , Deleção de Genes , Variação Genética , Células HEK293 , Células HeLa , Humanos , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , ATPases Translocadoras de Prótons/genética , ATPases Translocadoras de Prótons/fisiologia , Receptores de Superfície Celular/fisiologia , ATPases Vacuolares Próton-Translocadoras/fisiologia
19.
Sci Rep ; 8(1): 2648, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29422602

RESUMO

TGF-ß1, which can cause renal tubular injury through a vacuolar-type H+-ATPase (V-ATPase)-mediated pathway, is induced by the glucose degradation product methylglyoxal to yield peritoneal injury and fibrosis. The present study investigated the roles of V-ATPase and its accessory protein, the (pro)renin receptor, in peritoneal fibrosis during peritoneal dialysis. Rats daily administered 20 mM methylglyoxal intraperitoneally developed significant peritoneal fibrosis after 7 days with increased expression of TGF-ß and V-ATPase, which was reduced by the inhibition of V-ATPase with co-administration of 100 mM bafilomycin A1. The (pro)renin receptor and V-ATPase were expressed in acidic organelles and cell membranes of human peritoneal mesothelial cells. TGF-ß1 upregulated the expression of collagens, α-SMA, and EDA-fibronectin, together with ERK1/2 phosphorylation, which was reduced by inhibition of V-ATPase, (pro)renin receptor, or the MAPK pathway. Fibronectin and the soluble (pro)renin receptor were excreted from cells by acidic organelle trafficking in response to TGF-ß1; this excretion was also suppressed by inhibition of V-ATPase. Soluble (pro)renin receptor concentrations in effluents of patients undergoing peritoneal dialysis were associated with the dialysate-to-plasma ratio of creatinine. Together, these results demonstrate a novel fibrosis mechanism through the (pro)renin receptor and V-ATPase in the acidic organelles of peritoneal mesothelial cells.


Assuntos
Organelas/metabolismo , Fibrose Peritoneal/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Idoso , Animais , Movimento Celular/efeitos dos fármacos , Soluções para Diálise/metabolismo , Epitélio/metabolismo , Feminino , Fibrose/metabolismo , Fibrose/patologia , Glucose/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Rim/metabolismo , Masculino , Pessoa de Meia-Idade , Cavidade Peritoneal/fisiologia , Diálise Peritoneal/efeitos adversos , Diálise Peritoneal/métodos , Fibrose Peritoneal/patologia , Peritônio/metabolismo , Transporte Proteico , Ratos , Ratos Wistar , Receptores de Superfície Celular/metabolismo , Renina/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
20.
J Clin Invest ; 109(11): 1417-27, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12045255

RESUMO

Renin is an aspartyl protease essential for the control of blood pressure and was long suspected to have cellular receptors. We report the expression cloning of the human renin receptor complementary DNA encoding a 350-amino acid protein with a single transmembrane domain and no homology with any known membrane protein. Transfected cells stably expressing the receptor showed renin- and prorenin-specific binding. The binding of renin induced a fourfold increase of the catalytic efficiency of angiotensinogen conversion to angiotensin I and induced an intracellular signal with phosphorylation of serine and tyrosine residues associated to an activation of MAP kinases ERK1 and ERK2. High levels of the receptor mRNA are detected in the heart, brain, placenta, and lower levels in the kidney and liver. By confocal microscopy the receptor is localized in the mesangium of glomeruli and in the subendothelium of coronary and kidney artery, associated to smooth muscle cells and colocalized with renin. The renin receptor is the first described for an aspartyl protease. This discovery emphasizes the role of the cell surface in angiotensin II generation and opens new perspectives on the tissue renin-angiotensin system and on renin effects independent of angiotensin II.


Assuntos
Angiotensina II/biossíntese , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/fisiologia , Renina/metabolismo , ATPases Vacuolares Próton-Translocadoras , Sequência de Aminoácidos , Angiotensina I/biossíntese , Sequência de Bases , Northern Blotting , Cálcio/metabolismo , Divisão Celular , Clonagem Molecular , Reagentes de Ligações Cruzadas/farmacologia , AMP Cíclico/metabolismo , DNA/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Precursores Enzimáticos/metabolismo , Biblioteca Gênica , Mesângio Glomerular/citologia , Humanos , Cinética , Microscopia Confocal , Microscopia de Fluorescência , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Fosforilação , Testes de Precipitina , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/biossíntese , Fatores de Tempo , Distribuição Tecidual , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa