Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Gen Med ; 17: 2001-2009, 2024.
Article in English | MEDLINE | ID: mdl-38736672

ABSTRACT

Background: Lipoprotein(a) [Lp(a)] is a well-established risk factor for ischaemic stroke (IS). It is unclear whether Lp(a) is associated with IS in patients with atrial fibrillation (AF). The aim of this study is to explore the association between the concentration of Lp(a) and the risk of IS in AF patients, hope to find the potential risk factor for the IS in AF patients. Methods: This study is a retrospective cohort study. The screened AF patients between January 2017 and July 2021 were matched at 1:1 by the propensity score matching (PSM) method in the Second Affiliated Hospital of Nanchang University. Associations between Lp(a) and ischaemic stroke were analysed using logistic regression models, stratified analysis and sensitivity analysis. Statistical analyses were conducted using IBM SPSS software. Results: The number of enrolled participates is 2258, which contains 1129 non-AF patients and 1129 AF patients. Among IS patients, the median Lp(a) concentration was higher than that of controls (17.03 vs. 15.36 mg/dL, P = 0.032). The Spearman rank-order correlation coefficients revealed significant positive relationships between IS and Lp(a) (P = 0.032). In addition, a significant increase in IS risk was associated with Lp(a) levels >30.00 mg/dL in unadjusted model [OR:1.263, 95% CI(1.046-1.523), P = 0.015], model 1 [OR:1.284, 95% CI(1.062,1.552), P = 0.010], model 2 [OR: 1.297, 95% CI(1.07,1.573). P = 0.008], and model 3 [OR: 1.290, 95% CI (1.064, 1.562). P = 0.009]. The stratified analysis indicated that this correlation was not affected by female sex [1.484 (1.117, 1.972), P = 0.006], age ≤ 60 [1.864 (1.067-3.254), P=0.029], hypertension [1.359 (1.074, 1.721), P = 0.011], or non-coronary heart disease (CHD) [1.388 (1.108, 1.738), P = 0.004]. Conclusion: High levels of Lp(a) were significantly related to IS in AF patients and may be a potential risk factor in the onset of an IS in AF patients.

2.
Microvasc Res ; 154: 104689, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636926

ABSTRACT

Pathological retinal angiogenesis is not only the hallmark of retinopathies, but also a major cause of blindness. Guanylate binding protein 2 (GBP2) has been reported to be associated with retinal diseases such as diabetic retinopathy and hypoxic retinopathy. However, GBP2-mediated pathological retinal angiogenesis remains largely unknown. The present study aimed to investigate the role of GBP2 in pathological retinal angiogenesis and its underlying molecular mechanism. In this study, we established oxygen-induced retinopathy (OIR) mice model for in vivo study and hypoxia-induced angiogenesis in ARPE-19 cells for in vitro study. We demonstrated that GBP2 expression was markedly downregulated in the retina of mice with OIR and ARPE-19 cells treated with hypoxia, which was associated with pathological retinal angiogenesis. The regulatory mechanism of GBP2 in ARPE-19 cells was studied by GBP2 silencing and overexpression. The regulatory mechanism of GBP2 in the retina was investigated by overexpressing GBP2 in the retina of OIR mice. Mechanistically, GBP2 downregulated the expression and secretion of vascular endothelial growth factor (VEGFA) in ARPE-19 cells and retina of OIR mice. Interestingly, overexpression of GBP2 significantly inhibited neovascularization in OIR mice, conditioned medium of GBP2 overexpressing ARPE-19 cells inhibited angiogenesis in human umbilical vein endothelial cells (HUVECs). Furthermore, we confirmed that GBP2 downregulated VEGFA expression and angiogenesis by inhibiting the AKT/mTOR signaling pathway. Taken together, we concluded that GBP2 inhibited pathological retinal angiogenesis via the AKT/mTOR/VEGFA axis, thereby suggesting that GBP2 may be a therapeutic target for pathological retinal angiogenesis.


Subject(s)
Disease Models, Animal , GTP-Binding Proteins , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt , Retinal Neovascularization , Retinal Vessels , Signal Transduction , TOR Serine-Threonine Kinases , Vascular Endothelial Growth Factor A , Animals , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Humans , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/genetics , Retinal Neovascularization/prevention & control , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Cell Line , Retinal Vessels/metabolism , Retinal Vessels/pathology , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Mice , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Oxygen/metabolism , Cell Hypoxia
3.
Front Cardiovasc Med ; 10: 1237258, 2023.
Article in English | MEDLINE | ID: mdl-37823179

ABSTRACT

Familial hypercholesterolemia (FH) is a common hereditary cholesterol metabolic disease that usually leads to an increase in the level of low-density lipoprotein cholesterol in plasma and an increase in the risk of cardiovascular disease. The lack of disease screening and diagnosis often results in FH patients being unable to receive early intervention and treatment, which may mean early occurrence of cardiovascular disease. Thus, more requirements for FH identification and management have been proposed. Recently, machine learning (ML) has made great progress in the field of medicine, including many innovative applications in cardiovascular medicine. In this review, we discussed how ML can be used for FH screening, diagnosis and risk assessment based on different data sources, such as electronic health records, plasma lipid profiles and corneal radian images. In the future, research aimed at developing ML models with better performance and accuracy will continue to overcome the limitations of ML, provide better prediction, diagnosis and management tools for FH, and ultimately achieve the goal of early diagnosis and treatment of FH.

4.
Front Cardiovasc Med ; 10: 1237261, 2023.
Article in English | MEDLINE | ID: mdl-37621559

ABSTRACT

Background and aims: Familial hypercholesterolemia (FH) is becoming a global burden. However, it remains underdiagnosed and undertreated worldwide. This study aimed to observe the screening rate of FH patients and department distribution among hospitalized patients using different diagnostic criteria. Methods: A total of 45,410 inpatients with LDL-C ≥3.5 mmol/L between 2008 and 2019 were included from The Second Affiliated Hospital of Nanchang University. Inpatients are diagnosed and divided into groups by Dutch Lipid Clinic Network (DLCN) criteria, Chinese-modified DLCN criteria and Chinese expert consensus (CEC) criteria. Results: There were 172, 1,076 and 115 inpatients included in the DLCN group, Chinese-modified DLCN group and CEC group, respectively (screening rates: 0.38%, 2.37% and 0.25%). These FH patients had a very high risk of atherosclerotic cardiovascular disease (ASCVD) (55.7%-74.4%), especially in the DLCN group and CEC group (70.4%-74.4%). More than half of the patients were in the Department of Cardiology, and other high-risk departments included Neurology, Nephrology, Vascular Surgery, Otolaryngology & Head Neck Surgery and Traditional Chinese Medicine (24.35%-31.51%). Overall, hypertension, coronary heart disease, carotid arteriosclerosis, hepatic cyst, arrhythmia, and nonalcoholic fatty liver disease were common accompanying diseases with FH. Conclusions: It is necessary to establish appropriate diagnostic criteria and more positive treatment strategies for the FH inpatient population. In addition, promoting awareness of FH among doctors from other departments is also necessary. Therefore, developing a comprehensive management strategy for FH disease is very important.

6.
Hypertension ; 77(2): 405-416, 2021 02.
Article in English | MEDLINE | ID: mdl-33280408

ABSTRACT

Activation of PRR ([pro]renin receptor) contributes to enhancement of intrarenal RAS and renal medullary α-ENaC and thus elevated blood pressure during Ang II (angiotensin II) infusion. The goal of the present study was to test whether such action of PRR was mediated by sPRR (soluble PRR), generated by S1P (site-1 protease), a newly identified PRR cleavage protease. F1 B6129SF1/J mice were infused for 6 days with control or Ang II at 300 ng/kg per day alone or in combination with S1P inhibitor PF-429242 (PF), and blood pressure was monitored by radiotelemetry. S1P inhibition significantly attenuated Ang II-induced hypertension accompanied with suppressed urinary and renal medullary renin levels and expression of renal medullary but not renal cortical α-ENaC expression. The effects of S1P inhibition were all reversed by supplement with histidine-tagged sPRR termed as sPRR-His. Ussing chamber technique was performed to determine amiloride-sensitive short-circuit current, an index of ENaC activity in confluent mouse cortical collecting duct cell line cells exposed for 24 hours to Ang II, Ang II + PF, or Ang II + PF + sPRR-His. Ang II-induced ENaC activity was blocked by PF, which was reversed by sPRR-His. Together, these results support that S1P-derived sPRR mediates Ang II-induced hypertension through enhancement of intrarenal renin level and activation of ENaC.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure/drug effects , Hypertension/genetics , Proprotein Convertases/antagonists & inhibitors , Pyrrolidines/pharmacology , Receptors, Cell Surface/genetics , Animals , Hypertension/chemically induced , Hypertension/metabolism , Mice , Receptors, Cell Surface/metabolism , Renin/metabolism , Renin-Angiotensin System/drug effects , Serine Endopeptidases , Prorenin Receptor
7.
Am J Physiol Renal Physiol ; 319(5): F930-F940, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32865014

ABSTRACT

Adriamycin (ADR) administration in susceptible rodents such as the BALB/c mouse strain produces injury to the glomerulus mimicking human chronic kidney disease due to primary focal segmental glomerulosclerosis. The goal of the present study was to use this model to investigate antiproteinuric actions of the (pro)renin receptor decoy inhibitor PRO20. BALB/c mice were pretreated for 1 day with PRO20 at 500 µg·kg-1·day-1 via an osmotic minipump followed by a single injection of vehicle or ADR (10 mg/kg) via the tail vein. Albuminuria and renal function were analyzed at the fourth week post-ADR administration. ADR-treated mice exhibited severe proteinuria, hypoalbuminemia and hyperlipidemia, glomerulosclerosis, podocyte loss, tubulointerstitial fibrosis, and oxidative stress, accompanied by elevated urinary neutrophil gelatinase-associated lipocalin and kidney injury molecule-1, all of which were significantly attenuated by PRO20. Urinary and renal renin activity and angiotensin II were elevated by ADR and suppressed by PRO20. In parallel, urinary and renal H2O2 levels and renal NADPH oxidase 4 (Nox4) and transient receptor potential channel C6 (TRPC6) expression in response to ADR were all similarly suppressed. Taken together, the results of the present study provide the first evidence that PRO20 can protect against podocyte damage and interstitial fibrosis in ADR nephropathy by preventing activation of the intrarenal renin-angiotensin system and upregulation of Nox4 and TRPC6 expression. PRO20 may have a potential application in the treatment of ADR nephropathy.


Subject(s)
Kidney Diseases/drug therapy , Peptide Fragments/pharmacology , Renin-Angiotensin System/drug effects , Renin/metabolism , Angiotensin II/toxicity , Animals , Antihypertensive Agents/pharmacology , Doxorubicin/metabolism , Doxorubicin/pharmacology , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/metabolism , Hydrogen Peroxide/metabolism , Kidney Diseases/metabolism , Mice, Inbred BALB C , Peptide Fragments/metabolism , Podocytes/drug effects , Podocytes/metabolism , Protective Agents/pharmacology , Renin/drug effects , Renin/pharmacology
8.
Am J Physiol Renal Physiol ; 319(4): F647-F653, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32799674

ABSTRACT

It has been shown that cyclooxygenase (COX)-2-dependent activation of renal (pro)renin receptor (PRR) contributes to angiotensin II (ANG II)-induced hypertension. However, less is known about the involvement of this mechanism in ANG II-independent hypertension. The goal of the present study was to test whether or not COX-2-dependent upregulation of PRR serves as a universal mechanism contributing to ANG II-dependent and -independent hypertension. Here, we examined the association between renal COX-2 and PRR during deoxycorticosterone acetate (DOCA)-salt hypertension in rats. By immunoblot analysis and immunofluorescence, renal protein expression of PRR was remarkably upregulated by DOCA-salt treatment. Surprisingly, this upregulation of renal PRR expression was unaffected by a COX-2 inhibitor, celecoxib. To address the role of renal PRR to the pathogenesis of DOCA-salt hypertension, a decoy PRR inhibitor, PRO20, was infused to the renal medulla of uninephrectomized Sprague-Dawley rats for 14 days. Radiotelemetry demonstrated effective attenuation of DOCA-salt hypertension by intramedullary infusion of a PRR inhibitor, PRO20. In parallel, DOCA-salt-induced hypertrophy in the heart and kidney as well as proteinuria were improved, accompanied with blunted polydipsia and polyuria. In contrast, intravenous infusion of PRO20 was less effective in attenuating DOCA-salt hypertension and cardiorenal injury. Together, these results suggest that COX-2-independent activation of renal PRR contributes to DOCA-salt hypertension.


Subject(s)
Blood Pressure , Cyclooxygenase 2/metabolism , Desoxycorticosterone Acetate , Hypertension/enzymology , Kidney/enzymology , Receptors, Cell Surface/metabolism , Sodium Chloride, Dietary , Animals , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Disease Models, Animal , Enzyme Activation , Hypertension/chemically induced , Hypertension/physiopathology , Kidney/physiopathology , Male , Proteinuria/chemically induced , Proteinuria/enzymology , Proteinuria/physiopathology , Rats, Sprague-Dawley , Signal Transduction , Vacuolar Proton-Translocating ATPases
9.
JCI Insight ; 5(7)2020 04 09.
Article in English | MEDLINE | ID: mdl-32271168

ABSTRACT

The therapies available for management of obesity and associated conditions are limited, because they are often directed toward an individual component of metabolic syndrome and are associated with adverse effects. Here, we report the multifaceted therapeutic potential of histidine-tagged recombinant soluble (pro)renin receptor (sPRR), termed sPRR-His, in a mouse model of diet-induced obesity (DIO). In the DIO model, 2-week administration of sPRR-His lowered body weight and remarkably improved multiple metabolic parameters in the absence of fluid retention. Conversely, inhibition of endogenous sPRR production by PF429242 induced diabetes and insulin resistance, both of which were reversed by the sPRR-His supplement. At the cellular level, sPRR-His enhanced insulin-induced increases in glucose uptake via upregulation of phosphorylated AKT and protein abundance of glucose transporter 4. Promoter and gene expression analysis revealed PRR as a direct target gene of PPARγ. Adipocyte-specific PPARγ deletion induced severe diabetes and insulin resistance associated with reduced adipose PRR expression and circulating sPRR. The sPRR-His supplement in the null mice nearly normalized blood glucose and insulin levels. Additionally, sPRR-His treatment suppressed DIO-induced renal sodium-glucose cotransporter-2 (SGLT2) expression. Overall, sPRR-His exhibits a therapeutic potential in management of metabolic syndrome via interaction with PPARγ.


Subject(s)
Adipocytes/metabolism , Dietary Fats/adverse effects , Metabolic Syndrome/metabolism , Obesity/metabolism , PPAR gamma/metabolism , Receptors, Cell Surface/metabolism , Adipocytes/pathology , Animals , Dietary Fats/pharmacology , Disease Models, Animal , Male , Metabolic Syndrome/chemically induced , Mice , Obesity/chemically induced , Obesity/genetics , PPAR gamma/genetics , Receptors, Cell Surface/genetics , Prorenin Receptor
10.
Am J Physiol Renal Physiol ; 318(3): F817-F825, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31841392

ABSTRACT

We have previously shown that activation of (pro)renin receptor (PRR) induces epithelial Na+ channel (ENaC) activity in cultured collecting duct cells. Here, we examined the role of soluble PRR (sPRR), the cleavage product of PRR in ENaC regulation, and further tested its relevance to aldosterone signaling. In cultured mpkCCD cells, administration of recombinant histidine-tagged sPRR (sPRR-His) at 10 nM within minutes induced a significant and transient increase in the amiloride-sensitive short-circuit current as assessed using the Ussing chamber technique. The acute ENaC activation was blocked by the NADPH oxidase 1/4 inhibitor GKT137892 and siRNA against Nox4 but not the ß-catenin inhibitor ICG-001. In primary rat inner medullary collecting duct cells, administration of sPRR-His at 10 nM for 24 h induced protein expression of the α-subunit but not ß- or γ-subunits of ENaC, in parallel with upregulation of mRNA expression as well as promoter activity of the α-subunit. The transcriptional activation of α-ENaC was dependent on ß-catenin signaling. Consistent results obtained by epithelial volt ohmmeter measurement of equivalent current and Ussing chamber determination of short-circuit current showed that aldosterone-induced transepithelial Na+ transport was inhibited by the PRR decoy inhibitor PRO20 and PF-429242, an inhibitor of sPRR-generating enzyme site-1 protease, and the response was restored by the addition of sPRR-His. Medium sPRR was elevated by aldosterone and inhibited by PF-429242. Taken together, these results demonstrate that sPRR induces two phases of ENaC activation via distinct mechanisms and functions as a mediator of the natriferic action of aldosterone.


Subject(s)
Aldosterone/metabolism , Epithelial Sodium Channels , Kidney Tubules, Collecting/cytology , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Animals , Biological Transport , Cells, Cultured , Electrophysiological Phenomena , Epithelial Sodium Channel Blockers/administration & dosage , Epithelial Sodium Channel Blockers/pharmacology , Male , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Sodium/metabolism , Prorenin Receptor
11.
Front Physiol ; 10: 1157, 2019.
Article in English | MEDLINE | ID: mdl-31572210

ABSTRACT

We previously demonstrated that ureteral obstruction is associated with a urinary concentrating defect and reduced expression of renal aquaporins (AQPs), in which the renin-angiotensin system (RAS) may play an important role. The aims of the present study were to examine whether the renin inhibitor aliskiren could prevent the reduction in AQP expression and improve the urinary concentrating capacity in mice with bilateral ureteral obstruction (BUO) and BUO release. BUO was performed for 24 h, and BUO release was performed for 1 (B-R1D) or 3 days (B-R3D) with or without aliskiren treatment. Aliskiren prevented polyuria and decreased urine osmolality induced by B-R3D. In mice with BUO and BUO release, aliskiren attenuated the reduction in AQP2 protein and mRNA expression in the obstructed kidneys. B-R3D increased the protein expression of NLRP3 inflammasome components ASC, caspase-1, and interleukin-1ß in the obstructed kidneys, which was markedly prevented by aliskiren. Moreover, the NF-κB inhibitor Bay 11-7082 blocked NLRP3 inflammasome activation and attenuated the decrease in AQP2 protein expression in primary cultured rat inner medullary collecting duct cells treated with angiotensin II. These results indicate that the renin inhibitor aliskiren increases water channel AQP2 expression at least partially by suppressing NLRP3 inflammasome activation in the obstructed kidneys of mice with BUO and BUO release.

12.
JCI Insight ; 4(7)2019 04 04.
Article in English | MEDLINE | ID: mdl-30944256

ABSTRACT

The antidiuretic hormone vasopressin (AVP), acting through its type 2 receptor (V2R) in the collecting duct (CD), critically controls urine concentrating capability. Here, we report that site-1 protease-derived (S1P-derived) soluble (pro)renin receptor (sPRR) participates in regulation of fluid homeostasis via targeting V2R. In cultured inner medullary collecting duct (IMCD) cells, AVP-induced V2R expression was blunted by a PRR antagonist, PRO20; a PRR-neutralizing antibody; or a S1P inhibitor, PF-429242. In parallel, sPRR release was increased by AVP and reduced by PF-429242. Administration of histidine-tagged sPRR, sPRR-His, stimulated V2R expression and also reversed the inhibitory effect of PF-429242 on the expression induced by AVP. PF-429242 treatment in C57/BL6 mice impaired urine concentrating capability, which was rescued by sPRR-His. This observation was recapitulated in mice with renal tubule-specific deletion of S1P. During the pharmacological or genetic manipulation of S1P alone or in combination with sPRR-His, the changes in urine concentration were paralleled with renal expression of V2R and aquaporin-2 (AQP2). Together, these results support that S1P-derived sPRR exerts a key role in determining renal V2R expression and, thus, urine concentrating capability.


Subject(s)
Kidney Concentrating Ability/physiology , Kidney Tubules, Collecting/metabolism , Proton-Translocating ATPases/metabolism , Receptors, Cell Surface/metabolism , Receptors, Vasopressin/metabolism , Animals , Antidiuretic Hormone Receptor Antagonists/pharmacology , Aquaporin 2/genetics , Cells, Cultured , Epithelial Cells , Kidney Concentrating Ability/drug effects , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Male , Mice , Mice, Knockout , Models, Animal , Peptide Fragments/pharmacology , Primary Cell Culture , Proprotein Convertases/antagonists & inhibitors , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , Pyrrolidines/pharmacology , Rats , Receptors, Vasopressin/genetics , Renin/metabolism , Renin/pharmacology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Urothelium/cytology , Vacuolar Proton-Translocating ATPases
13.
FASEB J ; 33(1): 469-483, 2019 01.
Article in English | MEDLINE | ID: mdl-30036087

ABSTRACT

Increasing evidence supports the important role of H2S in renal physiology and the pathogenesis of kidney injury. Whether H2S regulates water metabolism in the kidney and the potential mechanism are still unknown. The present study was conducted to determine the role of H2S in urine concentration. Inhibition of both cystathionine-γ-lyase (CSE) and cystathionine-ß-synthase (CBS), 2 major enzymes for endogenous H2S production, with propargylglycine (PPG) and amino-oxyacetate (AOAA), respectively, caused increased urine output and reduced urine osmolality in mice that was associated with decreased expression of aquaporin (AQP)-2 in the renal inner medulla. Mice treated with both PPG and AOAA developed a urine concentration defect in response to dehydration that was accompanied by reduced AQP-2 protein expression. Inhibition of CSE alone was associated with a mild decrease in AQP-2 protein level in the renal medulla of heterozygous CBS mice. GYY4137, a slow H2S donor, markedly improved urine concentration and prevented the down-regulation of renal AQP-2 protein expression in mice with lithium-induced nephrogenic diabetes insipidus (NDI). GYY4137 significantly increased cAMP levels in cell lysates prepared from inner medullary collecting duct (IMCD) suspensions. AQP-2 protein expression was also upregulated, but was significantly inhibited by the adenyl cyclase inhibitor MDL12330A or the PKA inhibitor H89, but not the vasopressin 2 receptor (V2R) antagonist tolvaptan. Inhibition of endogenous H2S production impaired urine concentration in mice, whereas an exogenous H2S donor improved urine concentration in lithium-induced NDI by increasing AQP-2 expression in the collecting duct principal cells. H2S upregulated AQP-2 protein expression, probably via the cAMP-PKA pathway.-Luo, R., Hu, S., Liu, Q., Han, M., Wang, F., Qiu, M., Li, S., Li, X., Yang, T., Fu, X., Wang, W., Li, C. Hydrogen sulfide upregulates renal AQP-2 protein expression and promotes urine concentration.


Subject(s)
Aquaporin 2/metabolism , Cystathionine beta-Synthase/physiology , Cystathionine gamma-Lyase/physiology , Hydrogen Sulfide/pharmacology , Kidney Medulla/metabolism , Urination/drug effects , Urine/chemistry , Alkynes/metabolism , Aminooxyacetic Acid/metabolism , Animals , Gasotransmitters/pharmacology , Glycine/analogs & derivatives , Glycine/metabolism , Kidney Medulla/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Urinalysis
14.
Am J Physiol Renal Physiol ; 315(2): F199-F210, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29592526

ABSTRACT

The molecular mechanisms of melamine-induced renal toxicity have not been fully understood. The purpose of the study aimed to investigate whether melamine and cyanuric acid induced NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation in the kidney, which may contribute to abnormal water and sodium handling in a rat model. Wistar rats received melamine (Mel; 200 mg·kg body wt-1·day-1), cyanuric acid (CA; 200 mg·kg body wt-1·day-1), or Mel plus CA (Mel + CA; 100 mg·kg body wt-1·day-1, each) for 2 wk. Mel + CA caused damaged tubular epithelial structure and organelles, dilated tubular lumen, and inflammatory responses. Crystals were observed in urine and serum specimen, also in the lumen of dilated distal renal tubules. The combined ingestion of Mel and CA in rats caused a markedly impaired urinary concentration, which was associated with reduced protein expression of aquaporin (AQP)1, 2, and 3 in inner medulla and α-Na-K-ATPase and Na-K-2Cl transporters in cortex and outer medulla. Mel + CA treatment was associated with increased protein expression of CD3 and mRNA levels of CD68 and F4/80 as well as phosphorylation of NF-κB in the kidney. Mel + CA treatment increased protein and mRNA expression of NLRP3 inflammasome components apoptosis-associated speck-like protein containing a caspase recruitment domain, caspase-1, and IL-1ß in the inner medulla of rats. NF-κB inhibitor Bay 11-7082 reduced IL-1ß expression induced by Mel + CA and prevented downregulation of AQP2 in inner medullary collecting duct cell suspensions. In conclusion, Mel + CA treatment caused urinary-concentrating defects and reduced expression of renal AQPs and key sodium transporters, which is likely due to the inflammatory responses and activation of NLRP3 inflammasome induced by crystals formed in the kidney.


Subject(s)
Inflammasomes/metabolism , Kidney/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Polyuria/metabolism , Triazines , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Aquaporins/metabolism , CARD Signaling Adaptor Proteins/metabolism , CD3 Complex/metabolism , Caspase 1/metabolism , Interleukin-1beta/metabolism , Kidney/pathology , Kidney/physiopathology , Kidney Concentrating Ability , Male , NF-kappa B/metabolism , Phosphorylation , Polyuria/chemically induced , Polyuria/pathology , Polyuria/physiopathology , Rats, Wistar , Signal Transduction , Sodium-Potassium-Chloride Symporters/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism
15.
Am J Physiol Renal Physiol ; 312(1): F121-F133, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27784694

ABSTRACT

Microsomal prostaglandin E2 synthase-1 (mPGES-1), an inducible enzyme that converts prostaglandin H2 to prostaglandin E2 (PGE2), plays an important role in a variety of inflammatory diseases. We investigated the contribution of mPGES-1 to renal fibrosis and inflammation in unilateral ureteral obstruction (UUO) for 7 days using wild-type (WT) and mPGES-1 knockout (KO) mice. UUO induced increased mRNA and protein expression of mPGES-1 and cyclooxygenase-2 in WT mice. UUO was associated with increased renal PGE2 content and upregulated PGE2 receptor (EP) 4 expression in obstructed kidneys of both WT and mPGES-1 KO mice; EP4 expression levels were higher in KO mice with UUO than those in WT mice. Protein expression of NLRP3 inflammasome components ASC and interleukin-1ß was significantly increased in obstructed kidneys of KO mice compared with that in WT mice. mRNA expression levels of fibronectin, collagen III, and transforming growth factor-ß1 (TGF-ß1) were significantly higher in obstructed kidneys of KO mice than that in WT mice. In KO mice, protein expression of fibronectin and collagen III was markedly increased in obstructed kidneys compared with WT mice, which was associated with increased phosphorylation of protein kinase B (AKT). EP4 agonist CAY10598 attenuated increased expression of collagen I and fibronectin induced by TGF-ß1 in inner medullary collecting duct 3 cells. Moreover, CAY10598 prevented the activation of NLRP3 inflammasomes induced by angiotensin II in human proximal tubule cells (HK2). In conclusion, these findings suggested that mPGES-1 exerts a potentially protective effect against renal fibrosis and inflammation induced by UUO in mice.


Subject(s)
Inflammasomes/metabolism , Inflammation/metabolism , Prostaglandin-E Synthases/deficiency , Prostaglandin-E Synthases/metabolism , Ureteral Obstruction/metabolism , Animals , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Disease Models, Animal , Fibronectins/metabolism , Fibrosis/drug therapy , Fibrosis/metabolism , Inflammasomes/drug effects , Kidney/drug effects , Kidney/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , Pyrrolidinones/pharmacology , Tetrazoles/pharmacology , Ureteral Obstruction/pathology
16.
Am J Physiol Renal Physiol ; 311(4): F763-F776, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27385737

ABSTRACT

Endoplasmic reticulum (ER) stress has been implicated in some types of glomerular and tubular disorders. The objectives of this study were to elucidate the role of ER stress in lithium-induced nephrogenic diabetes insipidus (NDI) and to investigate whether attenuation of ER stress by 4-phenylbutyric acid (4-PBA) improves urinary concentrating defect in lithium-treated rats. Wistar rats received lithium (40 mmol/kg food), 4-PBA (320 mg/kg body wt by gavage every day), or no treatment (control) for 2 wk, and they were dehydrated for 24 h before euthanasia. Lithium treatment resulted in increased urine output and decreased urinary osmolality, which was significantly improved by 4-PBA. 4-PBA also prevented reduced protein expression of aquaporin-2 (AQP2), pS256-AQP2, and pS261-AQP2 in the inner medulla of kidneys from lithium-treated rats after 24-h dehydration. Lithium treatment resulted in increased expression of ER stress markers in the inner medulla, which was associated with dilated cisternae and expansion of ER in the inner medullary collecting duct (IMCD) principal cells. Confocal immunofluorescence studies showed colocalization of a molecular chaperone, binding IgG protein (BiP), with AQP2 in principal cells. Immunohistochemistry demonstrated increased intracellular expression of BiP and decreased AQP2 expression in IMCD principal cells of kidneys from lithium-treated rats. 4-PBA attenuated expression of ER stress markers and recovered ER morphology. In IMCD suspensions isolated from lithium-treated rats, 4-PBA incubation was also associated with increased AQP2 expression and ameliorated ER stress. In conclusion, in experimental lithium-induced NDI, 4-PBA improved the urinary concentrating defect and increased AQP2 expression, likely via attenuating ER stress in IMCD principal cells.


Subject(s)
Butylamines/therapeutic use , Diabetes Insipidus, Nephrogenic/drug therapy , Endoplasmic Reticulum Stress/drug effects , Animals , Aquaporin 2/metabolism , Butylamines/pharmacology , Diabetes Insipidus, Nephrogenic/chemically induced , Diabetes Insipidus, Nephrogenic/metabolism , Kidney/drug effects , Kidney/metabolism , Lithium , Male , Rats , Rats, Wistar , Treatment Outcome , Urination/drug effects
17.
J Am Soc Nephrol ; 27(10): 3063-3078, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26940090

ABSTRACT

Evidence supports an important role of Ca2+ release-activated Ca2+ channel protein 1 (Orai1)-mediated Ca2+ entry in the development of renal fibrosis, a common pathologic feature of CKDs that lead to ESRD, but the molecular mechanisms remain unclear. We determined the role of Orai1 calcium channel in renal fibrosis induced by high-fat diet and by unilateral ureteral obstruction. Mouse kidneys with fibrosis had higher levels of Orai1 protein expression than did kidneys without fibrosis. In vivo knockdown of Orai1 with adenovirus harboring Orai1-short hairpin RNA or inhibition of Orai1 with SKF96365 dramatically prevented renal fibrosis and significantly decreased protein expression of fibronectin, α­smooth muscle actin, and TGF­ß1 in the kidney cortex of ApoE-/- mice on a high-fat diet and in the obstructed kidneys of mice with unilateral ureteral obstruction. Compared with kidney biopsy specimens of patients with glomerular minimal change disease, those of patients with fibrotic nephropathy had higher expression levels of Orai1. In cultured human proximal tubule epithelial cells (HK2), knockdown of Orai1 Ca2+ channel with adenovirus-Orai1-short hairpin RNA markedly inhibited TGF-ß1-induced intracellular Ca2+ influx and phosphorylation of smad2/3. Knockdown or blockade of the Orai1 Ca2+ channel in HK2 cells also prevented epithelial-to-mesenchymal transition induced by TGF­ß1. In conclusion, blockade of the Orai1 Ca2+ channel prevented progression of renal fibrosis in mice, likely by suppressing smad2/3 phosphorylation and TGF-ß1-induced epithelial-to-mesenchymal transition. These results render the Orai1 Ca2+ channel a potential therapeutic target against renal fibrosis.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Calcium/metabolism , Imidazoles/pharmacology , Kidney/pathology , ORAI1 Protein/antagonists & inhibitors , Animals , Cells, Cultured , Fibrosis/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
Am J Physiol Renal Physiol ; 310(5): F351-63, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26672616

ABSTRACT

Obesity-related kidney disease is related to caloric excess promoting deleterious cellular responses. Accumulation of saturated free fatty acids in tubular cells produces lipotoxicity involving significant cellular dysfunction and injury. The objectives of this study were to elucidate the role of renin-angiotensin system (RAS) activation in saturated fatty acid-induced endoplasmic reticulum (ER) stress in cultured human proximal tubule epithelial cells (HK2) and in mice fed with a high-fat diet. Treatment with saturated fatty acid palmitic acid (PA; 0.8 mM) for 24 h induced ER stress in HK2, leading to an unfolded protein response as reflected by increased expressions of the ER chaperone binding immunoglobulin protein (BiP) and proapoptotic transcription factor C/EBP homologous protein (CHOP) protein as evaluated by immunoblotting. PA treatment also induced increased protein expression of inositol requiring protein 1α (IRE1α), phosphorylated eukaryotic initiation factor-α (eIF2α), and activating transcription factor 4 (ATF4) as well as activation of caspase-3. PA treatment was associated with increased angiotensin II levels in cultured medium. The angiotensin II type 1 receptor (AT1R) blocker valsartan or renin inhibitor aliskiren dramatically suppressed PA-induced upregulation of BiP, CHOP, IRE1α, p-eIF2α, and ATF4 in HK2 cells. In contrast, valsartan or aliskiren did not prevent ER stress induced by tunicamycin. C57BL/6 mice fed with a high-fat diet for 14 wk exhibited increased protein expressions of BiP and CHOP compared with control mice, which were significantly attenuated by the valsartan treatment. Increased angiotensin II levels in serum and urine were observed in mice fed with a high-fat diet when compared with controls. It is suggested that the intrarenal RAS activation may play an important role in diabetic kidney injury via mediating ER stress induced by saturated fatty acid.


Subject(s)
Diet, High-Fat , Endoplasmic Reticulum Stress/drug effects , Kidney Diseases/metabolism , Kidney Tubules, Proximal/drug effects , Palmitic Acid/pharmacology , Renin-Angiotensin System/drug effects , Amides/pharmacology , Angiotensin II/blood , Angiotensin II/urine , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Fumarates/pharmacology , Gene Expression Regulation , Heat-Shock Proteins/metabolism , Humans , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice, Inbred C57BL , RNA, Messenger/metabolism , Renin-Angiotensin System/genetics , Signal Transduction/drug effects , Transcription Factor CHOP/metabolism , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects , Valsartan/pharmacology
19.
Am J Physiol Renal Physiol ; 308(8): F910-22, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25694485

ABSTRACT

Ureteral obstruction is associated with reduced expression of renal aquaporins (AQPs), urinary concentrating defects, and an enhanced inflammatory response, in which the renin-angiotensin system (RAS) may play an important role. We evaluated whether RAS blockade by a direct renin inhibitor, aliskiren, would prevent the decreased renal protein expression of AQPs in a unilateral ureteral obstruction (UUO) model and what potential mechanisms may be involved. UUO was performed for 3 days (3UUO) and 7 days (7UUO) in C57BL/6 mice with or without aliskiren injection. In 3UUO and 7UUO mice, aliskiren abolished the reduction of AQP2 protein expression but not AQP1, AQP3, and AQP4. mRNA levels of renal AQP2 and vasopressin type 2 receptor were decreased in obstructed kidneys of 7UUO mice, which were prevented by aliskiren treatment. Aliskiren treatment was also associated with a reduced inflammatory response in obstructed kidneys of UUO mice. Aliskiren significantly decreased mRNA levels of several proinflammatory factors, such as transforming growth factor-ß and tumor necrosis factor-α, seen in obstructed kidneys of UUO mice. Interestingly, mRNA and protein levels of the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome components apoptosis-associated speck-like protein containing a caspase recruitment domain, caspase-1, and IL-1ß were dramatically increased in obstructed kidneys of 7UUO mice, which were significantly suppressed by aliskiren. In primary cultured inner medullary collecting duct cells, IL-1ß significantly decreased AQP2 expression. In conclusions, RAS blockade with the direct renin inhibitor aliskiren increased water channel AQP2 expression in obstructed kidneys of UUO mice, at least partially by preventing NLRP3 inflammasome activation in association with ureteral obstruction.


Subject(s)
Amides/pharmacology , Aquaporin 2/drug effects , Fumarates/pharmacology , Inflammasomes/antagonists & inhibitors , Kidney Diseases/prevention & control , Kidney/drug effects , Ureteral Obstruction/drug therapy , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Aquaporin 2/genetics , Aquaporin 2/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Cytoprotection , Disease Models, Animal , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Kidney/metabolism , Kidney Diseases/etiology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Male , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , RNA, Messenger/metabolism , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Renin/antagonists & inhibitors , Renin/metabolism , Renin-Angiotensin System/drug effects , Time Factors , Up-Regulation , Ureteral Obstruction/complications , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...