American journal of physiology. Renal physiology最新文献

筛选
英文 中文
Deletion of AT1a receptors selectively in the proximal tubules alters the hypotensive and natriuretic response to ANP via NPRA/cGMP/NO Signaling. 通过NPRA/cGMP/NO信号传导,选择性缺失近端肾小管中的AT1a受体可改变对ANP的降压和利钠反应。
American journal of physiology. Renal physiology Pub Date : 2024-10-03 DOI: 10.1152/ajprenal.00160.2024
Xiao Chun Li, Chih-Hong Wang, Rumana Hassan, Akemi Sato, Ryosuke Sato, Jia L Zhuo
{"title":"Deletion of AT<sub>1a</sub> receptors selectively in the proximal tubules alters the hypotensive and natriuretic response to ANP via NPR<sub>A</sub>/cGMP/NO Signaling.","authors":"Xiao Chun Li, Chih-Hong Wang, Rumana Hassan, Akemi Sato, Ryosuke Sato, Jia L Zhuo","doi":"10.1152/ajprenal.00160.2024","DOIUrl":"https://doi.org/10.1152/ajprenal.00160.2024","url":null,"abstract":"<p><p>In the kidney, vasoactive peptide hormones angiotensin II (Ang II), via AT<sub>1a</sub> receptors, and atrial natriuretic peptide (ANP), via NPR<sub>A</sub> receptors, reportedly play counteracting roles to regulate proximal tubule Na<sup>+</sup> reabsorption and maintain blood pressure homeostasis. However, how AT<sub>1a</sub> and NPR<sub>A</sub> receptors interact in the proximal tubules and whether deletion of AT<sub>1</sub> (AT<sub>1a</sub>) receptors selectively in the proximal tubules alters the hypotensive and natriuretic effects of ANP) have not been studied previously. The present study used a novel mouse model with proximal tubule-specific knockout of AT<sub>1a</sub> receptors to test the hypothesis that deletion of AT<sub>1a</sub> receptors selectively in the proximal tubules augments the hypotensive and natriuretic responses to ANP. Basal blood pressure was about 16 ± 3 mmHg lower, fractional proximal tubule Na<sup>+</sup> reabsorption was significantly lower, whereas 24 h urinary Na<sup>+</sup> excretion was significantly higher in PT-<i>Agtr1a</i><sup>-/-</sup> than in wild-type mice (<i>P</i><0.01). Infusion of ANP for 2 weeks (0.5 mg/kg/day, i.p.) further significantly decreased blood pressure and increased the natriuretic response in PT-<i>Agtr1a</i><sup>-/-</sup> mice by inhibiting proximal tubule Na<sup>+</sup> reabsorption (<i>P</i><0.01). These augmented hypotensive and natriuretic responses to ANP in PT-<i>Agtr1a</i><sup>-/-</sup> mice were associated with increased plasma and kidney cGMP levels (<i>P</i><0.01), kidney cortical NPR<sub>A</sub> and NPR<sub>C</sub> mRNA expression (<i>P</i><0.01), total and phosphorylated endothelial nitric oxide synthase (eNOS) (<i>P</i><0.01), and urinary nitric oxide (NO) excretion (<i>P</i><0.01). Taken together, the results of the present study support important physiological roles of Ang II/AT<sub>1a</sub> and ANP/NPR<sub>A</sub> signaling pathways in the proximal tubules to regulate proximal tubule reabsorption and maintain blood pressure homeostasis.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373701","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chronic central nervous system leptin administration attenuates kidney dysfunction and injury in a model of ischemia/reperfusion-induced acute kidney injury. 在缺血/再灌注诱发急性肾损伤模型中,长期服用中枢神经系统瘦素可减轻肾功能障碍和损伤。
American journal of physiology. Renal physiology Pub Date : 2024-10-03 DOI: 10.1152/ajprenal.00158.2024
Jussara M do Carmo, John E Hall, Luzia N S Furukawa, Viktoria Woronik, Xuemei Dai, Emily Ladnier, Zhen Wang, Ana C M Omoto, Alan Mouton, Xuan Li, Emilio M Luna-Suarez, Alexandre A da Silva
{"title":"Chronic central nervous system leptin administration attenuates kidney dysfunction and injury in a model of ischemia/reperfusion-induced acute kidney injury.","authors":"Jussara M do Carmo, John E Hall, Luzia N S Furukawa, Viktoria Woronik, Xuemei Dai, Emily Ladnier, Zhen Wang, Ana C M Omoto, Alan Mouton, Xuan Li, Emilio M Luna-Suarez, Alexandre A da Silva","doi":"10.1152/ajprenal.00158.2024","DOIUrl":"https://doi.org/10.1152/ajprenal.00158.2024","url":null,"abstract":"<p><p>In the present study we examined whether chronic intracerebroventricular (ICV) leptin administration protects against ischemia/reperfusion (I/R)-induced acute kidney injury (AKI). Twelve-week-old male rats were implanted with an ICV cannula into the right lateral ventricle and 8-10 days after surgery, leptin (0.021 mg/hr, n=8) or saline vehicle (0.5 ml/h, n=8) was infused via osmotic minipump connected to the ICV cannula for 12 days. On day 8 of leptin or vehicle infusion, rats were submitted to unilateral ischemia/reperfusion (UIR) by clamping the left pedicle for 30 min. To control for leptin-induced reductions in food intake, the vehicle-treated group was pair-fed (UIR-PF) to match the same amount of food consumed by leptin-treated (UIR-Leptin) rats. On the 12<sup>th</sup> day of leptin or vehicle infusion (4<sup>th</sup> day after AKI), single-left kidney glomerular filtration rate (GFR) was measured, blood samples were collected to quantify white blood cells, and kidneys were collected for histological assessment of injury. UIR-Leptin treated rats showed reduced right and left kidney weights (right: 1040±24 vs. 1281±36 mg; left: 1127±71 vs. 1707±45 mg, for UIR-Leptin and UIR-PF, respectively). ICV leptin infusion improved GFR (0.50±0.06 vs. 0.13±0.03 ml/min/g KW) and reduced kidney injury scores. ICV leptin treatment also attenuated the reduction in circulating adiponectin levels that was observed in UIR-PF rats, and increased circulating white blood cells count compared to UIR-PF rats (16.3±1.3 vs. 9.8±0.6 k/mL). Therefore, we show that leptin, via its actions on the central nervous system, confers significant protection against major kidney dysfunction and injury in a model of ischemia/reperfusion-induced AKI.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373700","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Matrix metalloproteinases in kidney homeostasis and diseases: an update. 基质金属蛋白酶在肾脏稳态和疾病中的作用:最新进展。
American journal of physiology. Renal physiology Pub Date : 2024-10-03 DOI: 10.1152/ajprenal.00179.2024
Roderick J Tan, Youhua Liu
{"title":"Matrix metalloproteinases in kidney homeostasis and diseases: an update.","authors":"Roderick J Tan, Youhua Liu","doi":"10.1152/ajprenal.00179.2024","DOIUrl":"https://doi.org/10.1152/ajprenal.00179.2024","url":null,"abstract":"<p><p>Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases with important roles in kidney homeostasis and pathology. While capable of collectively degrading each component of the extracellular matrix, MMPs also degrade nonmatrix substrates to regulate inflammation, epithelial plasticity, proliferation, apoptosis, and angiogenesis. More recently, intriguing mechanisms that directly alter podocyte biology have been described. There is now irrefutable evidence for MMP dysregulation in many types of kidney disease including acute kidney injury, diabetic and hypertensive nephropathy, polycystic kidney disease and Alport syndrome. This updated review will detail the complex biology of MMPs in kidney disease.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373702","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Optimized protocol for the multi-omics processing of cryopreserved human kidney tissue. 低温保存的人体肾组织多组学处理优化方案。
American journal of physiology. Renal physiology Pub Date : 2024-10-03 DOI: 10.1152/ajprenal.00404.2023
Sydney E Gies, Sonja Hänzelmann, Dominik Kylies, Moritz Lassé, Simon Lagies, Fabian Hausmann, Robin Khatri, Nikolay Zolotarev, Manuela Poets, Tianran Zhang, Fatih Demir, Anja M Billing, Josephine Quaas, Elisabeth Meister, Jonas Engesser, Anne K Mühlig, Shun Lu, Shuya Liu, Silvia Chilla, Ilka Edenhofer, Jan Czogalla, Fabian Braun, Bernd Kammerer, Victor G Puelles, Stefan Bonn, Markus M Rinschen, Maja Lindenmeyer, Tobias B Huber
{"title":"Optimized protocol for the multi-omics processing of cryopreserved human kidney tissue.","authors":"Sydney E Gies, Sonja Hänzelmann, Dominik Kylies, Moritz Lassé, Simon Lagies, Fabian Hausmann, Robin Khatri, Nikolay Zolotarev, Manuela Poets, Tianran Zhang, Fatih Demir, Anja M Billing, Josephine Quaas, Elisabeth Meister, Jonas Engesser, Anne K Mühlig, Shun Lu, Shuya Liu, Silvia Chilla, Ilka Edenhofer, Jan Czogalla, Fabian Braun, Bernd Kammerer, Victor G Puelles, Stefan Bonn, Markus M Rinschen, Maja Lindenmeyer, Tobias B Huber","doi":"10.1152/ajprenal.00404.2023","DOIUrl":"https://doi.org/10.1152/ajprenal.00404.2023","url":null,"abstract":"<p><p>Biobanking of tissue from clinically obtained kidney biopsies for later use with multi-omic and imaging techniques is an inevitable step to overcome the need of disease model systems and towards translational medicine. Hence, collection protocols ensuring integration into daily clinical routines using preservation media not requiring liquid nitrogen but instantly preserving kidney tissue for clinical and scientific analyses are of paramount importance. Thus, we modified a robust single nucleus dissociation protocol for kidney tissue stored snap frozen or in the preservation media RNA<i>later</i>and CellCover. Using porcine kidney tissue as surrogate for human kidney tissue, we conducted single nucleus RNA sequencing with the Chromium 10X Genomics platform. The resulting data sets from each storage condition were analyzed to identify any potential variations in transcriptomic profiles. Furthermore, we assessed the suitability of the preservation media for additional analysis techniques (proteomics, metabolomics) and the preservation of tissue architecture for histopathological examination including immunofluorescence staining. In this study, we show that in daily clinical routines the RNA<i>later</i> facilitates the collection of highly preserved human kidney biopsies and enables further analysis with cutting-edge techniques like single nucleus RNA sequencing, proteomics, and histopathological evaluation. Only metabolome analysis is currently restricted to snap frozen tissue. This work will contribute to build tissue biobanks with well-defined cohorts of the respective kidney disease that can be deeply molecularly characterized, opening new horizons for the identification of unique cells, pathways and biomarkers for the prevention, early identification, and targeted therapy of kidney diseases.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bayesian mapping of protein kinases to vasopressin-regulated phosphorylation sites in renal collecting duct. 蛋白激酶与肾集合管中血管加压素调节的磷酸化位点的贝叶斯图谱。
American journal of physiology. Renal physiology Pub Date : 2024-10-01 Epub Date: 2024-07-18 DOI: 10.1152/ajprenal.00142.2024
Venkatesh Deshpande, Euijung Park, Nipun U Jayatissa, Shaza Khan, Raymond Mejia, Chin-Rang Yang, Chung-Lin Chou, Viswanathan Raghuram, Mark A Knepper
{"title":"Bayesian mapping of protein kinases to vasopressin-regulated phosphorylation sites in renal collecting duct.","authors":"Venkatesh Deshpande, Euijung Park, Nipun U Jayatissa, Shaza Khan, Raymond Mejia, Chin-Rang Yang, Chung-Lin Chou, Viswanathan Raghuram, Mark A Knepper","doi":"10.1152/ajprenal.00142.2024","DOIUrl":"10.1152/ajprenal.00142.2024","url":null,"abstract":"<p><p>Vasopressin controls water permeability in the renal collecting duct by regulating the water channel protein, aquaporin-2 (AQP2). Phosphoproteomic studies have identified multiple proteins that undergo phosphorylation changes in response to vasopressin. The kinases responsible for the phosphorylation of most of these sites have not been identified. Here, we use large-scale Bayesian data integration to predict the responsible kinases for 51 phosphoproteomically identified vasopressin-regulated phosphorylation sites in the renal collecting duct. To do this, we applied Bayes' rule to rank the 515 known mammalian protein kinases for each site. Bayes' rule was applied recursively to integrate each of the seven independent datasets, each time using the posterior probability vector of a given step as the prior probability vector of the next step. In total, 30 of the 33 phosphorylation sites that increase with vasopressin were predicted to be phosphorylated by protein kinase A (PKA) catalytic subunit-α, consistent with prior studies implicating PKA in vasopressin signaling. Eighteen of the vasopressin-regulated phosphorylation sites were decreased in response to vasopressin and all but three of these sites were predicted to be targets of extracellular signal-regulated kinases, ERK1 and ERK2. This result implies that ERK1 and ERK2 are inhibited in response to vasopressin V2 receptor occupation, secondary to PKA activation. The six phosphorylation sites not predicted to be phosphorylated by PKA or ERK1/2 are potential targets of other protein kinases previously implicated in aquaporin-2 regulation, including cyclin-dependent kinase 18 (CDK18), calmodulin-dependent kinase 2δ (CAMK2D), AMP-activated kinase catalytic subunit-α-1 (PRKAA1) and CDC42 binding protein kinase β (CDC42BPB).<b>NEW & NOTEWORTHY</b> Vasopressin regulates water transport in the renal collecting duct in part through phosphorylation or dephosphorylation of proteins that regulate aquaporin-2. Prior studies have identified 51 vasopressin-regulated phosphorylation sites in 45 proteins. This study uses Bayesian data integration techniques to combine information from multiple prior proteomics and transcriptomics studies to predict the protein kinases that phosphorylate the 51 sites. Most of the regulated sites were predicted to be phosphorylated by protein kinase A or ERK1/ERK2.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141725272","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of NET formation and the renoprotective effect of degraded NETs in lupus nephritis. 鉴定狼疮性肾炎中 NETs 的形成和降解 NETs 对肾脏的保护作用。
American journal of physiology. Renal physiology Pub Date : 2024-10-01 Epub Date: 2024-08-29 DOI: 10.1152/ajprenal.00122.2024
Yong Jin, Yutong Wang, Xu Ma, Hongbin Li, Manling Zhang
{"title":"Identification of NET formation and the renoprotective effect of degraded NETs in lupus nephritis.","authors":"Yong Jin, Yutong Wang, Xu Ma, Hongbin Li, Manling Zhang","doi":"10.1152/ajprenal.00122.2024","DOIUrl":"10.1152/ajprenal.00122.2024","url":null,"abstract":"<p><p>To explore molecular biomarkers associated with the pathophysiology and therapy of lupus nephritis (LN), we conducted a joint analysis of transcriptomic data from 40 peripheral blood mononuclear cells (PBMCs) (GSE81622) and 21 kidney samples (GSE112943) from the Gene Expression Omnibus database using bioinformatics. A total of 976 and 2,427 differentially expressed genes (DEGs) were identified in PBMCs and renal tissues. Seven and two functional modules closely related to LN were identified. Further enrichment analysis revealed that the neutrophil activation pathway was highly active in both PBMCs and the kidney. Subsequently, 16 core genes closely associated with LN were verified by protein-protein interaction screening and quantitative PCR. In vitro cell models and MRL/lpr mouse models confirmed that the abnormal expression of these core genes was closely linked to neutrophil extracellular traps (NETs) generated by neutrophil activation, while degradation of NETs led to downregulation of core gene expression, thereby improving pathological symptoms of LN. Therefore, identification of patients with systemic lupus erythematosus exhibiting abnormal expression patterns for these core genes may serve as a useful indicator for kidney involvement. In addition, targeting neutrophils to modulate their activation levels and inhibit aberrant expression of these genes represents a potential therapeutic strategy for treating LN. <b>NEW & NOTEWORTHY</b> The mechanisms by which immune cells cause kidney injury in lupus nephritis are poorly understood. We integrated and analyzed the transcriptomic features of PBMCs and renal tissues from the GEO database to identify key molecular markers associated with neutrophil activation. We confirmed that neutrophil extracellular traps (NETs) formed by neutrophil activation promoted the upregulation of key genes in cell and animal models. Targeted degradation of NETs significantly ameliorated kidney injury in MRL/lpr mice.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142115830","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Histone deacetylase expression following cisplatin-induced acute kidney injury in male and female mice. 顺铂诱发雌雄小鼠急性肾损伤后组蛋白去乙酰化酶的表达。
American journal of physiology. Renal physiology Pub Date : 2024-10-01 Epub Date: 2024-08-08 DOI: 10.1152/ajprenal.00132.2024
Huy Nguyen, Anabelle Gales, Sureena Monteiro-Pai, Ariana S Oliver, Nicholas Harris, Anna D Montgomery, Stephanie Franzén, Malgorzata Kasztan, Kelly A Hyndman
{"title":"Histone deacetylase expression following cisplatin-induced acute kidney injury in male and female mice.","authors":"Huy Nguyen, Anabelle Gales, Sureena Monteiro-Pai, Ariana S Oliver, Nicholas Harris, Anna D Montgomery, Stephanie Franzén, Malgorzata Kasztan, Kelly A Hyndman","doi":"10.1152/ajprenal.00132.2024","DOIUrl":"10.1152/ajprenal.00132.2024","url":null,"abstract":"<p><p>The chemotherapeutic agent cisplatin accumulates in the kidneys, leading to acute kidney injury (AKI). Preclinical and clinical studies have demonstrated sex-dependent outcomes of cisplatin-AKI. Deranged histone deacetylase (HDAC) activity is hypothesized to promote the pathogenesis of male murine cisplatin-AKI; however, it is unknown whether there are sex differences in the kidney HDACs. We hypothesized that there would be sex-specific <i>Hdac</i> expression, localization, or enzymatic activity, which may explain sexual dimorphic responses to cisplatin-AKI. In normal human kidney RNA samples, <i>HDAC10</i> was significantly greater in the kidneys of women compared with men, whereas <i>HDAC1</i>, <i>HDAC6</i>, <i>HDAC10</i>, and <i>HDAC11</i> were differentially expressed between the kidney cortex and medulla, regardless of sex. In a murine model of cisplatin-AKI (3 days after a 15 mg/kg injection), we found few sex- or cisplatin-related differences in <i>Hdac</i> kidney transcripts among the mice. Although <i>Hdac9</i> was significantly greater in female mice compared with male mice, HDAC9 protein localization did not differ. <i>Hdac7</i> transcripts were greater in the inner medulla of cisplatin-AKI mice, regardless of sex, and this agreed with a greater HDAC7 abundance. HDAC activity within the cortex, outer medulla, and inner medulla was significantly lower in cisplatin-AKI mice but did not differ between the sexes. In agreement with these findings, a class I HDAC inhibitor did not improve kidney injury or function. In conclusion, even though cisplatin-AKI was evident and there were transcript level differences among the different kidney regions in this model, there were few sex- or cisplatin-dependent effects on kidney HDAC localization or activity.<b>NEW & NOTEWORTHY</b> Kidney histone deacetylases (HDACs) are abundant in male and female mice, and the inner medulla has the greatest HDAC activity. A low dose of cisplatin caused acute kidney injury (AKI) in these mice, but there were few changes in kidney HDACs at the RNA/protein/activity level. A class I HDAC inhibitor failed to improve AKI outcomes. Defining the HDAC isoform, cellular source, and interventional timing is necessary to determine whether HDAC inhibition is a therapeutic strategy to prevent cisplatin-AKI in both sexes.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141908615","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Peripheral dopamine suppression and elevated cystatin C in early diabetic nephropathy in spontaneously diabetic rats. 自发性糖尿病大鼠早期糖尿病肾病的外周多巴胺抑制和胱抑素 C 升高
American journal of physiology. Renal physiology Pub Date : 2024-10-01 Epub Date: 2024-08-01 DOI: 10.1152/ajprenal.00180.2023
Shoichiro Horita, Guy Watanabe, Shingen Misaka, Shu Taira, Mamoru Satoh, Yuko Maejima, Kenju Shimomura, Michio Shimabukuro, Junichiro James Kazama, Shuichi Shigetomi
{"title":"Peripheral dopamine suppression and elevated cystatin C in early diabetic nephropathy in spontaneously diabetic rats.","authors":"Shoichiro Horita, Guy Watanabe, Shingen Misaka, Shu Taira, Mamoru Satoh, Yuko Maejima, Kenju Shimomura, Michio Shimabukuro, Junichiro James Kazama, Shuichi Shigetomi","doi":"10.1152/ajprenal.00180.2023","DOIUrl":"10.1152/ajprenal.00180.2023","url":null,"abstract":"<p><p>Intrarenal dopamine plays a protective role against the development of diabetic nephropathy during the early stages of the disease. In streptozotocin-induced diabetic mice with renal-specific catechol-<i>O</i>-methyl transferase knockout, intrarenal dopamine was found to suppress glomerular hyperfiltration, reduce oxidative stress and inflammation, and inhibit fibrosis. However, although dopamine activation in streptozotocin-induced diabetic models has been shown to provide renal protection, the role of dopamine in models of naturally induced diabetes mellitus is still unclear. In the present study, we orally administered 10 mg/kg benserazide, a peripheral decarboxylase inhibitor, to spontaneously diabetic Torii rats daily to investigate the activation of the renal dopaminergic system during the progression of diabetic nephropathy. Our findings show that peripheral dopamine decreased urinary 8-iso-prostaglandin F<sub>2α</sub> and suppressed increases in plasma cystatin C levels. This study demonstrates that a reduction in peripheral dopamine can exacerbate renal dysfunction, even in the early stages of diabetic nephropathy characterized by glomerular hyperfiltration, thereby clarifying the pivotal role of endogenous peripheral dopamine in modulating oxidative stress and kidney performance.<b>NEW & NOTEWORTHY</b> By administering a peripheral decarboxylase inhibitor, we revealed that peripheral dopamine inhibits both the increase in urinary 8-iso-prostaglandin F<sub>2α</sub>, an oxidative stress marker, and the increase in plasma cystatin C, an early renal dysfunction marker, even in the early stages of diabetic nephropathy characterized by glomerular hyperfiltration. By visualizing renal dopamine precursor distribution, we highlighted the role of endogenous renal dopamine in oxidative stress and renal function following the onset of glomerular hyperfiltration.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141876941","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Optimizing renal transporter immunodetection: consequences of freeze-thaw during sample preparation. 优化肾脏转运体免疫检测:样品制备过程中的冻融后果。
American journal of physiology. Renal physiology Pub Date : 2024-10-01 Epub Date: 2024-08-29 DOI: 10.1152/ajprenal.00210.2024
Hannah L Hartman-Houstman, Donna L Ralph, Jonathan W Nelson, Lawrence G Palmer, Jessica E Faulkner, Jennifer C Sullivan, Desmond M Moronge, Alicia A McDonough
{"title":"Optimizing renal transporter immunodetection: consequences of freeze-thaw during sample preparation.","authors":"Hannah L Hartman-Houstman, Donna L Ralph, Jonathan W Nelson, Lawrence G Palmer, Jessica E Faulkner, Jennifer C Sullivan, Desmond M Moronge, Alicia A McDonough","doi":"10.1152/ajprenal.00210.2024","DOIUrl":"10.1152/ajprenal.00210.2024","url":null,"abstract":"<p><p>Renal transporters (cotransporters, channels, and claudins) mediate homeostasis of fluids and electrolytes and are targets of hormonal and therapeutic regulators. Assessing renal transporter abundance with antibody probes by immunoblotting is an essential tool for mechanistic studies. Although journals require authors to demonstrate antibody specificity, there are no consensus guidelines for kidney sample preparation leading to lab-to-lab variability in immunoblot results. In this study, we determined the impact of sample preparation, specifically freeze-thawed (Frozen) versus freshly processed (Fresh) kidneys (female and male rats and mice) on immunoblot signal detection of 15 renal transporters and the impact of protease inhibitors during homogenization. In female Sprague-Dawley rat kidneys homogenized with aprotinin, Na<sub>2</sub>EDTA, PMSF, and phosphatase inhibitors, immunodetection signals were ∼50% lower in Frozen versus Fresh samples for most transporters. Inclusion of additional inhibitors (Roche cOmplete Protease Inhibitor, \"+\") only partially increased transporter immunoblot signals to near Fresh levels. In male Sprague-Dawley rats, immunoblot signal density was lower in Frozen+ versus Fresh+ despite additional inhibitors. In C57BL/6 male mice, immunoblot signals from proximal tubule transporters were lower in Frozen versus Fresh by ∼25-50% and greater in Frozen+. In contrast, immunodetection signal was equivalent in female Frozen+ versus female Fresh+ for claudin 2, villin, AQP1, NKCC2, NCC, ENaCβ, ENaCɣ, claudin 7, AQP2, NKAα1, and NKAβ1. Thus, kidney sample preparation variables, including freeze-thaw and protease inhibition, have substantial transporter-specific effects on quantification of renal transporter abundance by immunoblot. These findings underscore the critical importance of assessing and reporting the impact of sample preparation protocols on transporter recovery to ensure robust rigor and reproducibility. <b>NEW & NOTEWORTHY</b> Freeze-thawing kidneys before homogenization is widely accepted in renal research. This study demonstrates that if kidneys are freeze-thawed just once before homogenization, immunoblot signals are reduced in a transporter-specific manner in rats and mice dependent on sex and that immunoblot signals can be partially recovered by adding additional protease inhibitors. These findings underscore the critical importance of assessing the impact of sample preparation, including freeze-thaw versus fresh, to ensure robust rigor and reproducibility.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142115816","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Actin cytoskeleton and associated myosin motors within the renal epithelium. 肾上皮细胞内的肌动蛋白细胞骨架和相关肌球蛋白马达。
American journal of physiology. Renal physiology Pub Date : 2024-10-01 Epub Date: 2024-07-25 DOI: 10.1152/ajprenal.00078.2024
Brook W Busselman, Ishara Ratnayake, Mark R Terasaki, Vedant P Thakkar, Arooba Ilyas, Karla L Otterpohl, Jenna L Zimmerman, Indra Chandrasekar
{"title":"Actin cytoskeleton and associated myosin motors within the renal epithelium.","authors":"Brook W Busselman, Ishara Ratnayake, Mark R Terasaki, Vedant P Thakkar, Arooba Ilyas, Karla L Otterpohl, Jenna L Zimmerman, Indra Chandrasekar","doi":"10.1152/ajprenal.00078.2024","DOIUrl":"10.1152/ajprenal.00078.2024","url":null,"abstract":"<p><p>This review highlights the complexity of renal epithelial cell membrane architectures and organelles through careful review of ultrastructural and physiological studies published over the past several decades. We also showcase the vital roles played by the actin cytoskeleton and actin-associated myosin motor proteins in regulating cell type-specific physiological functions within the cells of the renal epithelium. The purpose of this review is to provide a fresh conceptual framework to explain the structure-function relationships that exist between the actin cytoskeleton, organelle structure, and cargo transport within the mammalian kidney. With recent advances in technologies to visualize the actin cytoskeleton and associated proteins within intact kidneys, it has become increasingly imperative to reimagine the functional roles of these proteins in situ to provide a rationale for their unique, cell type-specific functions that are necessary to establish and maintain complex physiological processes.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141763108","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信