Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Am J Physiol Cell Physiol ; 326(6): C1683-C1696, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38646785

RESUMEN

Renovascular hypertension (RVHT) is characterized by renal artery stenosis and overactivated renin-angiotensin system (RAS). Apelin, known for its negative modulation of RAS, has protective effects against cardiovascular diseases. The role and mechanisms of the primary active form of apelin, apelin-13, in RVHT are unclear. In this study, male Sprague-Dawley rats were divided into control, two-kidney one-clip (2K1C) model, and 2K1C with apelin-13 treatment groups. Renin expression was analyzed using immunohistochemistry and molecular techniques. Full-length (pro)renin receptor (fPRR) and soluble PRR (sPRR) levels were assessed via Western blotting, and cAMP levels were measured using ELISA. Plasma renin content, plasma renin activity (PRA), angiotensin II (ANG II), and sPRR levels were determined by ELISA. Human Calu-6 and mouse As4.1 cells were used to investigate renin production mechanisms. The 2K1C model exhibited increased systolic blood pressure, plasma renin content, PRA, sPRR, and ANG II levels, while apelin-13 treatment reduced these elevations. Apelin-13 inhibited cAMP production, renin mRNA expression, protein synthesis, and PRR/sPRR protein expression in renal tissue. In Calu-6 cells, cAMP-induced fPRR and site-1 protease (S1P)-derived sPRR expression, which was blocked by cAMP-responsive element-binding protein (CREB) inhibition. Apelin-13 suppressed cAMP elevation, CREB phosphorylation, fPRR/sPRR protein expression, and renin production. Recombinant sPRR (sPRR-His) stimulated renin production, which was inhibited by the PRR decoy peptide PRO20 and S1P inhibitor PF429242. These findings suggest that apelin-13 inhibits plasma renin expression through the cAMP/PKA/sPRR pathway, providing a potential therapeutic approach for RVHT. Understanding the regulation of renin production is crucial for developing effective treatments.NEW & NOTEWORTHY Our research elucidated that apelin-13 inhibits renin production through the cAMP/PKA/soluble (pro)renin receptor pathway, presenting a promising therapeutic approach for renovascular hypertension (RVHT) by targeting renin expression mechanisms. These findings underscore the potential of apelin-13 as a novel strategy to address RVHT.


Asunto(s)
Hipertensión Renovascular , Péptidos y Proteínas de Señalización Intercelular , Ratas Sprague-Dawley , Renina , Animales , Renina/metabolismo , Renina/genética , Masculino , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Ratas , Humanos , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/tratamiento farmacológico , Hipertensión Renovascular/genética , Ratones , Sistema Renina-Angiotensina/efectos de los fármacos , Riñón/metabolismo , Receptor de Prorenina , Angiotensina II/metabolismo , AMP Cíclico/metabolismo , Presión Sanguínea/efectos de los fármacos , Transducción de Señal , Línea Celular , Modelos Animales de Enfermedad , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo
2.
Hypertens Res ; 47(6): 1627-1641, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38605139

RESUMEN

Preeclampsia is classified as new-onset hypertension coupled with gross endothelial dysfunction. Placental (pro)renin receptor ((P)RR) and plasma soluble (P)RR (s(P)RR) are elevated in patients with preeclampsia. Thus, we aimed to interrogate the role (P)RR may play in the pathogenesis of preeclampsia. Human uterine microvascular endothelial cells (HUtMECs, n = 4) were cultured with either; vehicle (PBS), 25-100 nM recombinant s(P)RR, or 10 ng/ml TNF-a (positive control) for 24 h. Conditioned media and cells were assessed for endothelial dysfunction markers via qPCR, ELISA, and immunoblot. Angiogenic capacity was assessed through tube formation and adhesion assays. Additionally, pregnant rats were injected with an adenovirus overexpressing s(P)RR from mid-pregnancy (day 8.5), until term (n = 6-7 dams/treatment). Maternal and fetal tissues were assessed. HUtMECs treated with recombinant s(P)RR displayed increased expression of endothelial dysfunction makers including vascular cell adhesion molecule-1, intracellular adhesion molecule-1, and endothelin-1 mRNA expression (P = 0.003, P = 0.001, P = 0.009, respectively), along with elevated endothelin-1 protein secretion (P < 0.001) compared with controls. Recombinant s(P)RR impaired angiogenic capacity decreasing the number of branches, total branch length, and mesh area (P < 0.001, P = 0.004, and P = 0.009, respectively), while also increasing vascular adhesion (P = 0.032). +ADV rats exhibited increased systolic (P = 0.001), diastolic (P = 0.010), and mean arterial pressures (P = 0.012), compared with -ADV pregnancies. Renal arteries from +ADV-treated rats had decreased sensitivity to acetylcholine-induced relaxation (P = 0.030), compared with -ADV pregnancies. Our data show that treatment with s(P)RR caused hypertension and growth restriction in vivo and caused marked endothelial dysfunction in vitro. These findings demonstrate the significant adverse actions of s(P)RR on vascular dysfunction that is characteristic of the preeclamptic phenotype.


Asunto(s)
Células Endoteliales , Preeclampsia , Receptores de Superficie Celular , Embarazo , Femenino , Animales , Preeclampsia/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/genética , Humanos , Ratas , Células Endoteliales/metabolismo , Ratas Sprague-Dawley , Fenotipo , Células Cultivadas , Receptor de Prorenina , Placenta/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Útero/irrigación sanguínea , Útero/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo
3.
Obes Facts ; 16(4): 335-343, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37231878

RESUMEN

INTRODUCTION: High soluble (pro)renin receptor (s[P]RR) level in circulation is reported in obese patients; however, it is unclear which body composition components are responsible for it. In this study, the authors examined blood s(P)RR levels and ATP6AP2 gene expression levels in visceral and subcutaneous adipose tissue (VAT, SAT) in severely obese patients who underwent laparoscopic sleeve gastrectomy (LSG), with the aim of clarifying the relationship with body composition and metabolic factors. METHODS: Seventy five cases who underwent LSG between 2011 and 2015 and were postoperatively followed-up for 12 months at the Toho University Sakura Medical Center were included in the analysis of the cross-sectional survey at baseline, and 33 cases were included in the analysis of the longitudinal survey during the 12 months after LSG. We evaluated body composition, glycolipid parameters, liver/renal function, as well as serum s(P)RR level and ATP6AP2 mRNA expression level in VAT and SAT. RESULTS: The mean serum s(P)RR level at baseline was 26.1 ng/mL, this value was considered higher than values in healthy subjects. There was no significant difference in the expression level of ATP6AP2 mRNA between VAT and SAT. At baseline, multiple regression analysis for the association between s(P)RR and variables identified that visceral fat area, HOMA2-IR, and UACR showed the independent relationships with s(P)RR. During the 12 months after LSG, body weight, serum s(P)RR level showed a significant decrease (from 30.0 ± 7.0 to 21.9 ± 4.3). Multiple regression analysis for the association between the change in s(P)RR and variables showed that changes in visceral fat area, and alanine transaminase were independently related to the change in s(P)RR. CONCLUSION: This study showed that blood s(P)RR level was high in severely obese patients, decreased with weight loss by LSG, and was associated with visceral fat area in both pre- and postoperative changes. The results suggest that blood s(P)RR levels in obese patients may reflect the involvement of visceral adipose (P)RR in insulin resistance and renal damage mechanisms associated with obesity.


Asunto(s)
Resistencia a la Insulina , Obesidad Mórbida , Humanos , Obesidad Mórbida/complicaciones , Obesidad Mórbida/cirugía , Obesidad Mórbida/metabolismo , Adiposidad , Receptor de Prorenina , Estudios Transversales , Obesidad/complicaciones , Obesidad/cirugía , Obesidad/metabolismo , Grasa Intraabdominal/metabolismo , Riñón/metabolismo
4.
Am J Physiol Renal Physiol ; 324(1): F1-F11, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36302140

RESUMEN

Soluble (pro)renin receptor (sPRR), the extracellular domain of (pro)renin receptor (PRR), is primarily generated by site-1 protease and furin. It has been reported that sPRR functions as an important regulator of intrarenal renin contributing to angiotensin II (ANG II)-induced hypertension. Relatively, less is known for the function of sPRR in ANG II-independent hypertension such as mineralocorticoid excess. In the present study, we used a novel mouse model with mutagenesis of the cleavage site in PRR (termed as PRRR279V/L282V or mutant) to examine the phenotype during aldosterone (Aldo)-salt treatment. The hypertensive response of mutant mice to Aldo-salt treatment was blunted in parallel with the attenuated response of plasma volume expansion and renal medullary α-epithelial Na+ channel expression. Moreover, Aldo-salt-induced hypertrophy in the heart and kidney as well as proteinuria were improved, accompanied by blunted polydipsia and polyuria. Together, these results represent strong evidence favoring endogenous sPRR as a mediator of Aldo-salt-induced hypertension and renal injury.NEW & NOTEWORTHY We used a novel mouse model with mutagenesis of the cleavage site of PRR to support soluble PRR as an essential mediator of aldosterone-salt-induced hypertension and also as a potential therapeutic target for patients with mineralocorticoid excess. We firstly report that soluble PRR-dependent pathway medicates the Na+-retaining action of aldosterone in the distal nephron, which opens up a new area for a better understanding of the molecular basis of renal handling of Na+ balance and blood pressure.


Asunto(s)
Aldosterona , Hipertensión , Ratones , Animales , Aldosterona/metabolismo , Receptor de Prorenina , Mineralocorticoides , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/metabolismo , Riñón/metabolismo , Cloruro de Sodio Dietético/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina , Angiotensina II/farmacología , Sodio/metabolismo , Mutagénesis
5.
Nephron ; 147(3-4): 234-243, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35871512

RESUMEN

The (pro)renin receptor (PRR) was originally cloned as a specific single-transmembrane receptor for prorenin and renin and has now emerged as a multifunctional protein implicated in a wide variety of developmental and physiopathological processes. Activation of PRR in the kidney causes Na+ and water retention, contributing to elevation of blood pressure in response to various hypertensive stimuli. Part of the renal action of PRR depends on activation of intrarenal renin-angiotensin system. In recent years, accumulating evidence suggests that the prohypertensive action of renal PRR was largely mediated by production of the 28-kDa soluble (pro)renin receptor through protease-mediated cleavage of the extracellular domain of PRR. The generation of multiple isoforms of PRR due to the protease-mediated cleavage partially explains diversified actions of PRR. The current review will summarize recent advances in understanding the roles of sPPR in animal models of hypertension.


Asunto(s)
Hipertensión , Receptor de Prorenina , Animales , Receptores de Superficie Celular , Hipertensión/metabolismo , Riñón/patología , Sistema Renina-Angiotensina , Renina/metabolismo
6.
Am J Physiol Renal Physiol ; 323(5): F507-F514, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36074917

RESUMEN

(Pro)renin receptor (PRR), also termed ATPase H+-transporting accessory protein 2 (ATP6AP2), is a type I transmembrane receptor and is capable of binding and activating prorenin and renin. Apart from its association with the renin-angiotensin system, PRR has been implicated in diverse developmental, physiological, and pathophysiological processes. Within the kidney, PRR is predominantly expressed in the distal nephron, particularly the intercalated cells, and activation of renal PRR contributes to renal injury in various rodent models of chronic kidney disease. Moreover, recent evidence demonstrates that PRR is primarily cleaved by site-1 protease to produce 28-kDa soluble PRR (sPRR). sPRR seems to mediate most of the known pathophysiological functions of renal PRR through modulating the activity of the intrarenal renin-angiotensin system and provoking proinflammatory and profibrotic responses. Not only does sPRR activate renin, but it also directly binds and activates the angiotensin II type 1 receptor. This review summarizes recent advances in understanding the roles and mechanisms of sPRR in the context of renal pathophysiology.


Asunto(s)
Insuficiencia Renal Crónica , ATPasas de Translocación de Protón Vacuolares , Humanos , Renina/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Superficie Celular/metabolismo , Sistema Renina-Angiotensina , Riñón/metabolismo , Insuficiencia Renal Crónica/metabolismo , Biomarcadores/metabolismo , Adenosina Trifosfatasas , ATPasas de Translocación de Protón Vacuolares/metabolismo
7.
Biomedicines ; 10(8)2022 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-36009420

RESUMEN

Symptomatic heart failure with reduced ejection fraction (HFrEF) is characterized by edema and chronic pathological activation of the classical renin-angiotensin-aldosterone system (RAAS). The soluble (pro)renin receptor (s(P)RR) is released into circulation by proteolytic cleavage of tissue expressed (P)RR and is a candidate biomarker of RAAS activation. However, previous studies linked elevated levels of s(P)RR in patients with HFrEF to renal dysfunction. Utilizing prospectively enrolled patients with comparable rEF, we show that increased plasma levels of s(P)RR are associated with symptomatic HF (characterized by edema), independent of chronic renal dysfunction. We also found that s(P)RR levels were positively correlated with patient plasma renin activity (PRA). Normotensive mice with dilated cardiomyopathy (DCM) and HFrEF, without renal dysfunction, showed plasma s(P)RR and PRA patterns similar to human HFrEF patients. Plasma s(P)RR levels positively correlated with PRA and systemic edema, but not with EF, resembling findings in patients with HFrEF without chronic kidney dysfunction. In female DCM mice with elevated PRA levels and plasma s(P)RR levels, a randomized, blinded trial comparing the direct renin inhibitor, aliskiren vs. vehicle control, showed that direct renin inhibition normalized PRA, lowered s(P)RR, and prevented symptomatic HFrEF. Considered in light of previous findings, these data suggest that, in HFrEF, in the absence of renal dysfunction, elevation of plasma s(P)RR levels is caused by increased PRA and associated with the development of systemic edema.

8.
J Cardiovasc Dev Dis ; 9(5)2022 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-35621841

RESUMEN

This study aims to compare soluble (pro)renin receptor [s(P)RR] levels between black and white adults and to explore the associations of left ventricular (LV) structure and function with s(P)RR in the total and ethnicity-stratified groups. The study sample included 1172 apparently healthy black (n = 587) and white (n = 585) participants of the African-PREDICT study aged 20−30 years. Echocardiography was performed to determine relative wall thickness (RWT), LV mass index, LV ejection fraction and stroke volume index (SVi). s(P)RR was analyzed from serum samples, while plasma renin activity-surrogate (PRA-S) and eq angiotensin II were determined using the RAS™ Fingerprint. s(P)RR was higher in the white participants compared to the black participants (p < 0.001). In multivariable-adjusted linear regression analyses, we observed a positive association between RWT and s(P)RR (ß = 0.141; p = 0.005) and negative associations of LV ejection fraction (ß = −0.123; p = 0.016) and SVi (ß = −0.144; p = 0.004) with s(P)RR only in white adults. Higher s(P)RR observed in white vs. black participants was associated with higher RWT and poorer LV function only in young white adults but not in their black counterparts. These results suggest that s(P)RR may contribute to LV remodeling and dysfunction in white populations due to its role in volume−pressure regulation and its proinflammatory as well as profibrotic effects.

10.
Clin Sci (Lond) ; 135(13): 1627-1630, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34240733

RESUMEN

This commentary highlights the study entitled 'Soluble (pro)renin receptor induces endothelial dysfunction and hypertension in mice with diet-induced obesity via activation of angiotensin II type 1 receptor' presented by Fu et al. published in Clinical Science (Clin Sci (Lond) (2021) 135(6), https://doi.org/10.1042/CS20201047). The authors evaluated the role of the soluble (pro)renin receptor (sPRR), a cleavage product of the prorenin receptor (PRR) by the site 1 protease, as a ligand for angiotensin II type 1 receptor (AT1R). They presented for the first time that sPRR directly interacts with AT1R, causing nuclear factor-κB activation, inflammation, apoptosis, and endothelial dysfunction in primary human umbilical vein endothelial cells (HUVECs). Furthermore, the interaction between sPRR and AT1R was responsible for endothelial dysfunction and hypertension in diet-induced obesity mice. These results provide a potential mechanism for obesity-induced endothelial dysfunction and hypertension. Thus, the sPRR/AT1R complex may be a novel therapeutic target for cardiovascular diseases associated with endothelial dysfunction.


Asunto(s)
Receptor de Angiotensina Tipo 1 , Renina , Animales , Células Endoteliales/metabolismo , Ligandos , Ratones , Renina/metabolismo , Sistema Renina-Angiotensina
12.
Placenta ; 116: 43-50, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34020806

RESUMEN

The (pro)renin receptor ((P)RR) is a multi-functional protein that can be proteolytically cleaved and released in a soluble form (s(P)RR). Recently, the (P)RR and s(P)RR have become of interest in pregnancy and its associated pathologies. This is because the (P)RR not only activates tissue renin angiotensin systems, but it is also an integral component of vacuolar-ATPase, activates the wingless/integrated (Wnt)/ß-catenin and extracellular signal regulated kinases 1 and 2/mitogen-activated protein kinase signalling pathways, and stabilises the ß subunit of pyruvate dehydrogenase. Additionally, s(P)RR is detected in plasma and urine, and maternal plasma levels are elevated in pregnancy complications including fetal growth restriction, preeclampsia and gestational diabetes mellitus. Therefore, s(P)RR has potential as a biomarker for these pregnancy pathologies. Preliminary functional findings suggest that s(P)RR may be important for regulating fluid balance, inflammation and blood pressure, all of which contribute to a successful pregnancy. The (P)RR and s(P)RR regulate pathways that are known to be important in maintaining pregnancy, however their role in the physiological context of pregnancy is poorly characterised. This review summarises the known and potential functions of the (P)RR and s(P)RR in pregnancy, and how their dysregulation may contribute to pregnancy complications. It also highlights the need for further research into the source and function of s(P)RR in pregnancy. Soluble (P)RR levels could be indicative of placental, kidney or liver dysfunction and therefore be a novel clinical biomarker, or therapeutic target, to improve the detection and treatment of pregnancy pathologies.


Asunto(s)
Diabetes Gestacional/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Femenino , Humanos , Embarazo , Receptor de Prorenina
13.
Clin Sci (Lond) ; 135(6): 793-810, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33625485

RESUMEN

Until now, renin-angiotensin system (RAS) hyperactivity was largely thought to result from angiotensin II (Ang II)-dependent stimulation of the Ang II type 1 receptor (AT1R). Here we assessed the role of soluble (pro)renin receptor (sPRR), a product of site-1 protease-mediated cleavage of (pro)renin receptor (PRR), as a possible ligand of the AT1R in mediating: (i) endothelial cell dysfunction in vitro and (ii) arterial dysfunction in mice with diet-induced obesity. Primary human umbilical vein endothelial cells (HUVECs) treated with a recombinant histidine-tagged sPRR (sPRR-His) exhibited IκBα degradation concurrent with NF-κB p65 activation. These responses were secondary to sPRR-His evoked elevations in Nox4-derived H2O2 production that resulted in inflammation, apoptosis and reduced NO production. Each of these sPRR-His-evoked responses was attenuated by AT1R inhibition using Losartan (Los) but not ACE inhibition using captopril (Cap). Further mechanistic exploration revealed that sPRR-His activated AT1R downstream Gq signaling pathway. Immunoprecipitation coupled with autoradiography experiments and radioactive ligand competitive binding assays indicate sPRR directly interacts with AT1R via Lysine199 and Asparagine295. Important translational relevance was provided by findings from obese C57/BL6 mice that sPRR-His evoked endothelial dysfunction was sensitive to Los. Besides, sPRR-His elevated blood pressure in obese C57/BL6 mice, an effect that was reversed by concurrent treatment with Los but not Cap. Collectively, we provide solid evidence that the AT1R mediates the functions of sPRR during obesity-related hypertension. Inhibiting sPRR signaling should be considered further as a potential therapeutic intervention in the treatment and prevention of cardiovascular disorders involving elevated blood pressure.


Asunto(s)
Hipertensión/fisiopatología , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Animales , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Captopril/farmacología , Dieta Alta en Grasa/efectos adversos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Peróxido de Hidrógeno , Losartán/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad , Sistema Renina-Angiotensina/efectos de los fármacos , Receptor de Prorenina
14.
Am J Physiol Renal Physiol ; 319(5): F941-F953, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32865015

RESUMEN

Tubulointerstitial fibrosis has been regarded as a critical event in the pathogenesis of chronic kidney disease. The soluble form of (pro)renin receptor (sPRR), generated by site-1 protease (S1P) cleavage of full-length PRR, can be detected in biological fluid and elevated under certain pathological conditions. The present study was designed to evaluate the potential role of sPRR in the regulation of the fibrotic response in a cultured human renal proximal tubular cell line (HK-2 cells) in the setting of transforming growth factor (TGF)-ß or sPRR-His treatment. The TGF-ß-induced fibrotic response of HK-2 cells was indicated by upregulation of fibronectin (FN) expression; meanwhile, TGF-ß could also induce the generation of sPRR, due to enhanced cleavage of full-length PRR. To explore the role of sPRR in the fibrotic response of HK-2 cells, we blocked the production of sPRR with a the S1P inhibitor PF429242 and found that PF429242 remarkably suppressed TGF-ß-induced sPRR generation and FN expression in HK-2 cells. Administration of sPRR-His restored the PF429242-attenuated FN expression in HK-2 cells, indicating that sPRR could promote the TGF-ß-induced fibrotic response. Furthermore, sPRR-His alone also increased the abundance of FN in HK-2 cells. These data suggested that sPRR was sufficient and necessary for the TGF-ß-induced fibrotic response of HK-2 cells. Mechanistically, sPRR activated the AKT and ß-catenin pathway in HK-2 cells, and blockade of the AKT or ß-catenin pathway significantly abrogated sPRR-induced FN and Snail expression. Taking together, sPRR promoted the fibrotic response of HK-2 cells by activating Akt/ß-catenin/Snail signaling, and it may serve as a potential therapeutic target in renal fibrosis.


Asunto(s)
Células Epiteliales/metabolismo , Túbulos Renales Proximales/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo , Humanos , Riñón/metabolismo , Receptores de Superficie Celular/metabolismo , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
15.
Placenta ; 101: 49-56, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32920451

RESUMEN

INTRODUCTION: The (pro)renin receptor (ATP6AP2) is cleaved and released as soluble ATP6AP2 (sATP6AP2). The sATP6AP2 is detected in plasma and urine and is elevated in women with gestational diabetes and preeclampsia. The source and cleavage pathway of sATP6AP2 in pregnancy is unknown. The syncytiotrophoblast is the major placental secretory layer and is in direct contact with maternal blood. Both FURIN and Site 1 protease (MBTPS1) cleave sATP6AP2 in non-placental cells. We postulated that ATP6AP2 was cleaved by FURIN and/or MBTPS1 and that sATP6AP2 is secreted by the placental syncytiotrophoblast. METHODS: Term primary trophoblast cells were transfected with FURIN siRNA, negative control siRNA or vehicle. In a separate experiment, primary trophoblasts were treated with a pro-protein convertase inhibitor (DEC-RVKR-CMK), an MBTPS1 inhibitor (PF 429242) or vehicle. Trophoblasts were left to spontaneously syncytialise before cells and supernatants were collected and intracellular and extracellular sATP6AP2 levels analysed by immunoblot. RESULTS: sATP6AP2 is secreted by placental trophoblasts. Levels of intra and extra-cellular sATP6AP2 decrease with syncytialisation (P = 0.01 and P = 0.02, respectively), as do FURIN mRNA (P = 0.0003) and protein (P = 0.0007). FURIN siRNA decreased FURIN mRNA and protein levels (both P < 0.0001). Neither FURIN siRNA or PF 429242 affected sATP6AP2 levels. DEC-RVKR-CMK significantly decreased extracellular sATP6AP2 protein levels (P = 0.02). DISCUSSION: Soluble ATP6AP2 is secreted by placental trophoblasts and levels decrease with syncytialisation. DEC-RVKR-CMK, a broad inhibitor of pro-protein convertases reduced extracellular sATP6AP2 levels, but FURIN siRNA and MBTPS1 inhibition had no effect. Hence, a convertase other than FURIN or MBTPS1 is most likely responsible for placental sATP6AP2 secretion.


Asunto(s)
Furina/metabolismo , Proproteína Convertasas/metabolismo , Receptores de Superficie Celular/metabolismo , Serina Endopeptidasas/metabolismo , Trofoblastos/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Humanos , Cultivo Primario de Células
16.
Peptides ; 111: 152-157, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29660382

RESUMEN

Soluble (pro)renin receptor [s(P)RR], which is generated from cleavage of (P)RR, can be detected in plasma and urine. s(P)RR levels can reflect the severity of some diseases, such as renal lesions, gestational diabetes mellitus or hypertension, and obstructive sleep apnea syndrome. However, the relationship between s(P)RR levels and the severity of chronic heart failure remains undetermined. We studied s(P)RR levels in 118 patients with chronic heart failure with reduced ejection fraction (HFrEF), including 86 without renal dysfunction (HF) and 32 with renal dysfunction (HF + RF), and 28 healthy subjects (HS) to reveal the relationship between s(P)RR levels and other HFrEF parameters. Plasma s(P)RR levels were 22.2 ±â€¯4.1 ng/mL (HS), 26.4 ±â€¯5.3 ng/ mL (HF) and 30.0 ±â€¯5.3 ng/mL (HF + RF). Plasma s(P)RR levels were significantly higher in the HF group than in the HS group (P < 0.001) and even more increased in the HF + RF group (P < 0.001 vs. the HS group and P < 0.05 vs. the HF group). Multivariate regression analysis revealed that the left ventricular mass index (LVMI) and estimated glomerular filtration rate (eGFR) were independently related to s(P)RR levels in HFrEF patients. In conclusion, high plasma s(P)RR levels are associated with left ventricular remodeling and, especially, with renal dysfunction. Therefore, s(P)RR is a promising evaluative indicator for the severity of HFrEF patients.


Asunto(s)
Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/fisiopatología , Receptores de Superficie Celular/sangre , ATPasas de Translocación de Protón Vacuolares/sangre , Disfunción Ventricular Izquierda/sangre , Disfunción Ventricular Izquierda/fisiopatología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Tasa de Filtración Glomerular/fisiología , Humanos , Modelos Lineales , Masculino , Persona de Mediana Edad , Análisis Multivariante , Volumen Sistólico , Remodelación Ventricular/fisiología
17.
Biomed Rep ; 9(3): 247-252, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30271601

RESUMEN

(Pro)renin receptor [(P)RR] has been implicated in diverse biological processes through binding to its ligands, which include renin, prorenin, Wnt signaling molecules and subunits of vascular H+-ATPase. Recent studies have reported that (P)RR is implicated in pathophysiological conditions including retinopathy and pancreatic ductal adenocarcinoma, and the soluble form of this receptor [s(P)RR] is considered as a useful biomarker for diseases. The present study examined the effect of aliskiren, the first orally active direct renin inhibitor, on the protein levels of (P)RR using cultured human umbilical vein endothelial cells (HUVECs). The cells were treated with or without aliskiren (10 nM) at 37°C for different durations (0, 8, 16 and 24 h). Aliskiren-treated HUVECs exhibited reduced proliferation compared with those treated without the drug. Furthermore, aliskiren treatment decreased not only the level of exogenous prorenin that bound to the membranes of HUVECs, but also the renin activity derived from this binding activity. These results indicate that the quantity of full-length (P)RR was reduced by aliskiren treatment, and furthermore, that the level of s(P)RR released from HUVECs was decreased with the treatment. Recent study has reported that s(P)RR exerted antidiuretic function. The current study suggests that the levels of s(P)RR, as a potential antidiuretic molecule and prospective disease biomarker, may be decreased during anti-hypertensive treatments with aliskiren.

18.
Am J Physiol Renal Physiol ; 315(5): F1416-F1421, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30019932

RESUMEN

The antidiuretic hormone vasopressin (VP) is produced by the hypothalamus and is stored and secreted from the posterior pituitary. VP acts via VP type 2 receptors (V2Rs) on the basolateral membrane of principal cells of the collecting duct (CD) to regulate fluid permeability. The VP-evoked endocrine pathway is essential in determining urine concentrating capability. For example, a defect in any component of the VP signaling pathway can result in polyuria, polydipsia, and hypotonic urine, collectively termed diabetes insipidus (DI). A lack of VP production precipitates central diabetes insipidus (CDI), which can be managed effectively by VP supplementation. A majority of cases of nephrogenic diabetes insipidus (NDI) result from V2R mutations that impair receptor sensitivity. No specific therapy is currently available for management of NDI. Evidence is evolving that (pro)renin receptor (PRR), a newly identified member of the renin-angiotensin system, is capable of regulating VP production and action. As such, PRR should be considered strongly as a therapeutic target for treating CDI and NDI. The current review will summarize recent advances in understanding the physiology of renal and central PRR as it relates to the two types of DI.


Asunto(s)
Fármacos Antidiuréticos/uso terapéutico , Diabetes Insípida/tratamiento farmacológico , Diuresis/efectos de los fármacos , Riñón/efectos de los fármacos , Receptores de Superficie Celular/uso terapéutico , Sistema Renina-Angiotensina/efectos de los fármacos , Animales , Diabetes Insípida/enzimología , Diabetes Insípida/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Riñón/enzimología , Riñón/patología , Mutación , Fenotipo , Receptores de Superficie Celular/metabolismo , Receptores de Vasopresinas/genética , Vasopresinas/metabolismo , Receptor de Prorenina
19.
Metab Syndr Relat Disord ; 16(4): 174-182, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29649379

RESUMEN

BACKGROUND: Plasma concentrations of soluble (pro)renin receptor [s(P)RR], which are elevated in patients with obstructive sleep apnea (OSA), have not been studied in morbid obesity. The aim of this study is to clarify effects of bariatric surgery on plasma s(P)RR concentrations and identify associated factors for their changes in OSA patients with morbid obesity. METHODS: Twenty-three patients with OSA complicated by morbid obesity (10 men and 13 women; body mass index, 40.7 ± 6.16 kg/m2) without chronic kidney disease were followed up after bariatric surgery. Overnight polysomnography (PSG) was performed before surgery, and 4 and 24 weeks after surgery. Plasma s(P)RR concentrations were measured each morning after PSG. RESULTS: Preoperative plasma s(P)RR concentrations showed significant positive correlations with serum creatinine (P < 0.05), arousal index (P < 0.01), apnea-hypopnea index (AHI) (P < 0.05), apnea index (P < 0.005), and desaturation index (P < 0.05), and a significant inverse correlation with an estimated glomerular filtration rate (P < 0.05). With the improvement of these PSG parameters, plasma s(P)RR concentrations significantly decreased from 15.3 ± 3.6 to 12.5 ± 2.7 ng/mL 4 weeks after surgery, which further decreased to 11.4 ± 2.4 ng/mL 24 weeks after surgery. The association observed before surgery between plasma s(P)RR concentrations and the PSG parameters was not seen after surgery. CONCLUSIONS: Bariatric surgery in patients with OSA complicated by morbid obesity decreased plasma s(P)RR concentrations. The most associated factors for their changes were arousal index, AHI, apnea index, and desaturation index.


Asunto(s)
Cirugía Bariátrica , Obesidad Mórbida/sangre , Obesidad Mórbida/cirugía , Receptores de Superficie Celular/sangre , Apnea Obstructiva del Sueño/sangre , ATPasas de Translocación de Protón Vacuolares/sangre , Adulto , Anciano , Índice de Masa Corporal , Femenino , Tasa de Filtración Glomerular , Humanos , Fallo Renal Crónico/sangre , Masculino , Persona de Mediana Edad , Polisomnografía , Apnea Obstructiva del Sueño/cirugía
20.
J Biochem ; 161(4): 369-379, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28013223

RESUMEN

The extracellular domain of the (pro)renin receptor [(P)RR] is cleaved to generate the soluble form of (P)RR [s(P)RR]. Multiple clinical studies have revealed the association between serum/plasma s(P)RR levels and certain diseases, thereby suggesting a potential role for s(P)RR as a disease biomarker. Here, we investigated whether site-1 protease (S1P) is responsible for cleaving (P)RR to generate s(P)RR. Reduction of endogenous S1P with siRNA attenuated s(P)RR generation in Chinese hamster ovary (CHO) cells exogenously expressing human (P)RR with a C-terminal decahistidine tag [CHO/h(P)RR-10His cells]; conversely, overexpression of S1P by transient transfection increased s(P)RR generation. The S1P inhibitor PF429242 suppressed s(P)RR generation in CHO/h(P)RR-10His and human cervical carcinoma HeLa cells; however, the ADAM inhibitor GM6001 had no effect. The furin inhibitor Dec-RVKR-CMK had no effect on the amount of s(P)RR, but caused a slight increase in the size of the s(P)RR. Moreover, the reversible vesicle-trafficking inhibitor brefeldin A (BFA) enhanced the generation of large-sized s(P)RR; PF429242, but not Dec-RVKR-CMK, suppressed this BFA-induced s(P)RR formation. The size of s(P)RR generated during BFA treatment was reduced after removal of BFA; Dec-RVKR-CMK, but not PF429242, suppressed this conversion. Together, these results suggest that s(P)RR is generated by sequential processing by S1P and furin.


Asunto(s)
Proproteína Convertasas/metabolismo , Receptores de Superficie Celular/metabolismo , Serina Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Animales , Brefeldino A/farmacología , Células CHO , Cricetinae , Cricetulus , Furina/metabolismo , Células HeLa , Humanos , Immunoblotting , Modelos Biológicos , Proproteína Convertasas/antagonistas & inhibidores , Proproteína Convertasas/genética , Inhibidores de Proteasas/farmacología , Interferencia de ARN , Receptores de Superficie Celular/genética , Homología de Secuencia de Aminoácido , Serina Endopeptidasas/genética , Solubilidad , Receptor de Prorenina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA