American journal of physiology. Renal physiology最新文献

筛选
英文 中文
Cell death induced by acute kidney injury: A perspective on the contributions of accidental and programmed cell death. 急性肾损伤诱导的细胞死亡:透视意外死亡和程序性细胞死亡的贡献
American journal of physiology. Renal physiology Pub Date : 2024-04-25 DOI: 10.1152/ajprenal.00275.2023
Mi Ra Noh, Babu Padanilam
{"title":"Cell death induced by acute kidney injury: A perspective on the contributions of accidental and programmed cell death.","authors":"Mi Ra Noh, Babu Padanilam","doi":"10.1152/ajprenal.00275.2023","DOIUrl":"https://doi.org/10.1152/ajprenal.00275.2023","url":null,"abstract":"The involvement of cell death in AKI is linked to multiple factors including nucleotide depletion, electrolyte imbalance, reactive oxygen species, endonucleases, disturbance of mitochondrial integrity, and activation of several cell death pathway components. Since our review in 2003, discussing the relative contributions of apoptosis and necrosis, several other forms of cell death have been identified and are shown to contribute to acute kidney injury (AKI). Currently, these various forms of cell death can be fundamentally divided into accidental cell death (ACD) and regulated or programmed cell death (RCD/PCD) based on functional aspects. Several death initiator and effector molecules, switch molecules that may act as signaling components triggering either death or protective mechanisms or alternate cell death pathways have been identified as part of the machinery. Intriguingly, several of these cell death pathways share components and signaling pathways suggesting complementary or compensatory functions. Thus defining the crosstalk between distinct cell death pathways and identifying the unique molecular effectors for each type of cell death may be required to develop novel strategies to prevent cell death. Further, depending on the multiple forms of cell death simultaneously induced in different AKI settings, strategies for combination therapies that block multiple cell death pathways need to be developed to completely prevent injury, cell death and renal function. This review highlights the various cell death pathways, crosstalk and interactions between different cell death modalities in AKI.","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140658401","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
The degree of aortic occlusion in the setting of trauma alters the extent of acute kidney injury associated with mitochondrial preservation. 创伤时主动脉闭塞的程度会改变与线粒体保存相关的急性肾损伤的程度。
American journal of physiology. Renal physiology Pub Date : 2024-04-01 Epub Date: 2024-03-07 DOI: 10.1152/ajprenal.00323.2023
Biebele Abel, John Mares, Justin Hutzler, Babita Parajuli, Lalitha Kurada, Joseph M White, Brandon W Propper, Ian J Stewart, David M Burmeister
{"title":"The degree of aortic occlusion in the setting of trauma alters the extent of acute kidney injury associated with mitochondrial preservation.","authors":"Biebele Abel, John Mares, Justin Hutzler, Babita Parajuli, Lalitha Kurada, Joseph M White, Brandon W Propper, Ian J Stewart, David M Burmeister","doi":"10.1152/ajprenal.00323.2023","DOIUrl":"10.1152/ajprenal.00323.2023","url":null,"abstract":"<p><p>Resuscitative endovascular balloon occlusion of the aorta (REBOA) is used to control noncompressible hemorrhage not addressed with traditional tourniquets. However, REBOA is associated with acute kidney injury (AKI) and subsequent mortality in severely injured trauma patients. Here, we investigated how the degree of aortic occlusion altered the extent of AKI in a porcine model. Female Yorkshire-cross swine (<i>n</i> = 16, 68.1 ± 0.7 kg) were anesthetized and had carotid and bilateral femoral arteries accessed for REBOA insertion and distal and proximal blood pressure monitoring. Through a laparotomy, a 6-cm liver laceration was performed and balloon inflation was performed in <i>zone 1</i> of the aorta for 90 min, during which animals were randomized to target distal mean arterial pressures of 25 or 45 mmHg via balloon volume adjustment. Blood draws were taken at baseline, end of occlusion, and time of death, at which point renal tissues were harvested 6 h after balloon deflation for histological and molecular analyses. Renal blood flow was lower in the 25-mmHg group (48.5 ± 18.3 mL/min) than in the 45-mmHg group (177.9 ± 27.2 mL/min) during the occlusion phase, which recovered and was not different after balloon deflation. AKI was more severe in the 25-mmHg group, as evidenced by circulating creatinine, blood urea nitrogen, and urinary neutrophil gelatinase-associated lipocalin. The 25-mmHg group had increased tubular necrosis, lower renal citrate synthase activity, increased tissue and circulating syndecan-1, and elevated systemic inflammatory cytokines. The extent of renal ischemia-induced AKI is associated with the magnitude of mitochondrial biomass and systemic inflammation, highlighting potential mechanistic targets to combine with partial REBOA strategies to prevent AKI.<b>NEW & NOTEWORTHY</b> Large animal models of ischemia-reperfusion acute kidney injury (IR-AKI) are lacking. This report establishes a titratable IR-AKI model in swine in which a balloon catheter can be used to alter distal pressures experienced by the kidney, thus controlling renal blood flow. Lower blood flow results in greater renal dysfunction and structural damage, as well as lower mitochondrial biomass, elevated systemic inflammation, and vascular dysfunction.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140051272","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
Is fibroblast growth factor 23 the main culprit for cardiovascular disease in chronic kidney disease? 成纤维细胞生长因子 23 是慢性肾病心血管疾病的罪魁祸首吗?
American journal of physiology. Renal physiology Pub Date : 2024-04-01 Epub Date: 2024-01-11 DOI: 10.1152/ajprenal.00379.2023
Carsten A Wagner, Isabel Rubio Aliaga, Daniela Egli-Spichtig
{"title":"Is fibroblast growth factor 23 the main culprit for cardiovascular disease in chronic kidney disease?","authors":"Carsten A Wagner, Isabel Rubio Aliaga, Daniela Egli-Spichtig","doi":"10.1152/ajprenal.00379.2023","DOIUrl":"10.1152/ajprenal.00379.2023","url":null,"abstract":"","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139418716","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
Rodent models of AKI and AKI-CKD transition: an update in 2024. AKI 和 AKI-CKD 转换的啮齿动物模型:2024 年的更新。
American journal of physiology. Renal physiology Pub Date : 2024-04-01 Epub Date: 2024-02-01 DOI: 10.1152/ajprenal.00402.2023
Ying Fu, Yu Xiang, Qingqing Wei, Daria Ilatovskaya, Zheng Dong
{"title":"Rodent models of AKI and AKI-CKD transition: an update in 2024.","authors":"Ying Fu, Yu Xiang, Qingqing Wei, Daria Ilatovskaya, Zheng Dong","doi":"10.1152/ajprenal.00402.2023","DOIUrl":"10.1152/ajprenal.00402.2023","url":null,"abstract":"<p><p>Despite known drawbacks, rodent models are essential tools in the research of renal development, physiology, and pathogenesis. In the past decade, rodent models have been developed and used to mimic different etiologies of acute kidney injury (AKI), AKI to chronic kidney disease (CKD) transition or progression, and AKI with comorbidities. These models have been applied for both mechanistic research and preclinical drug development. However, current rodent models have their limitations, especially since they often do not fully recapitulate the pathophysiology of AKI in human patients, and thus need further refinement. Here, we discuss the present status of these rodent models, including the pathophysiologic compatibility, clinical translational significance, key factors affecting model consistency, and their main limitations. Future efforts should focus on establishing robust models that simulate the major clinical and molecular phenotypes of human AKI and its progression.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11208034/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139652395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Podocytes from hypertensive and obese mice acquire an inflammatory, senescent, and aged phenotype. 来自高血压和肥胖小鼠的荚膜细胞获得炎症、衰老和老化表型
American journal of physiology. Renal physiology Pub Date : 2024-04-01 Epub Date: 2024-02-29 DOI: 10.1152/ajprenal.00417.2023
Sierra R McKinzie, Natalya Kaverina, Robert Allen Schweickart, Christopher P Chaney, Diana G Eng, Beatriz Maria Veloso Pereira, Bryan Kestenbaum, Jeffrey W Pippin, Oliver Wessely, Stuart J Shankland
{"title":"Podocytes from hypertensive and obese mice acquire an inflammatory, senescent, and aged phenotype.","authors":"Sierra R McKinzie, Natalya Kaverina, Robert Allen Schweickart, Christopher P Chaney, Diana G Eng, Beatriz Maria Veloso Pereira, Bryan Kestenbaum, Jeffrey W Pippin, Oliver Wessely, Stuart J Shankland","doi":"10.1152/ajprenal.00417.2023","DOIUrl":"10.1152/ajprenal.00417.2023","url":null,"abstract":"<p><p>Patients with hypertension or obesity can develop glomerular dysfunction characterized by injury and depletion of podocytes. To better understand the molecular processes involved, young mice were treated with either deoxycorticosterone acetate (DOCA) or fed a high-fat diet (HFD) to induce hypertension or obesity, respectively. The transcriptional changes associated with these phenotypes were measured by unbiased bulk mRNA sequencing of isolated podocytes from experimental models and their respective controls. Key findings were validated by immunostaining. In addition to a decrease in canonical proteins and reduced podocyte number, podocytes from both hypertensive and obese mice exhibited a sterile inflammatory phenotype characterized by increases in NLR family pyrin domain containing 3 (NLRP3) inflammasome, protein cell death-1, and Toll-like receptor pathways. Finally, although the mice were young, podocytes in both models exhibited increased expression of senescence and aging genes, including genes consistent with a senescence-associated secretory phenotype. However, there were differences between the hypertension- and obesity-associated senescence phenotypes. Both show stress-induced podocyte senescence characterized by increased <i>p21</i> and <i>p53</i>. Moreover, in hypertensive mice, this is superimposed upon age-associated podocyte senescence characterized by increased <i>p16</i> and <i>p19</i>. These results suggest that senescence, aging, and inflammation are critical aspects of the podocyte phenotype in experimental hypertension and obesity in mice.<b>NEW & NOTEWORTHY</b> Hypertension and obesity can lead to glomerular dysfunction in patients, causing podocyte injury and depletion. Here, young mice given deoxycorticosterone acetate or a high-fat diet to induce hypertension or obesity, respectively. mRNA sequencing of isolated podocytes showed transcriptional changes consistent with senescence, a senescent-associated secretory phenotype, and aging, which was confirmed by immunostaining. Ongoing studies are determining the mechanistic roles of the accelerated aging podocyte phenotype in experimental hypertension and obesity.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11208020/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139992004","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Human soluble prorenin receptor expressed in mouse renal collecting duct shows sex-specific effect on cardiorenal function. 在小鼠肾集合管中表达的人可溶性肾素受体对心肾功能有性别特异性影响
American journal of physiology. Renal physiology Pub Date : 2024-04-01 Epub Date: 2024-02-22 DOI: 10.1152/ajprenal.00375.2023
Gertrude Arthur, Audrey Poupeau, Katherine Biel, Jeffrey L Osborn, Ming Gong, Terry D Hinds, Volkhard Lindner, Analia S Loria
{"title":"Human soluble prorenin receptor expressed in mouse renal collecting duct shows sex-specific effect on cardiorenal function.","authors":"Gertrude Arthur, Audrey Poupeau, Katherine Biel, Jeffrey L Osborn, Ming Gong, Terry D Hinds, Volkhard Lindner, Analia S Loria","doi":"10.1152/ajprenal.00375.2023","DOIUrl":"10.1152/ajprenal.00375.2023","url":null,"abstract":"<p><p>Soluble prorenin receptor (sPRR), a component of the renin-angiotensin system (RAS), has been identified as a plasma biomarker for hypertension and cardiovascular diseases in humans. Despite studies showing that sPRR in the kidney is produced by tubular cells in the renal collecting duct (CD), its biological actions modulating cardiorenal function in physiological conditions remain unknown. Therefore, the objective of our study was to investigate whether CD-derived human sPRR (HsPRR) expression influences cardiorenal function and examine sex and circadian differences. Thus, we investigated the status of the intrarenal RAS, water and electrolyte balance, renal filtration capacity, and blood pressure (BP) regulation in CD-HsPRR and control (CTL) mice. CD-HsPRR mice were generated by breeding human sPRR-Myc-tag mice with Hoxb7/Cre mice. Renal sPRR expression increased in CD-HsPRR mice, but circulating sPRR and RAS levels were unchanged compared with CTL mice. Only female littermates expressing CD-HsPRR showed <i>1</i>) increased 24-h BP, <i>2</i>) an impaired BP response to an acute dose of losartan and attenuated angiotensin II (ANG II)-induced hypertension, <i>3</i>) reduced angiotensin-converting enzyme activity and ANG II content in the renal cortex, and <i>4</i>) decreased glomerular filtration rate, with no changes in natriuresis and kaliuresis despite upregulation of the β-subunit of the epithelial Na<sup>+</sup> channel in the renal cortex. These cardiorenal alterations were displayed only during the active phase of the day. Taken together, these data suggest that HsPRR could interact with ANG II type 1 receptors mediating sex-specific, ANG II-independent renal dysfunction and a prohypertensive phenotype in a sex-specific manner.<b>NEW & NOTEWORTHY</b> We successfully generated a humanized mouse model that expresses human sPRR in the collecting duct. Collecting duct-derived human sPRR did not change circulating sPRR and RAS levels but increased daytime BP in female mice while showing an attenuated angiotensin II-dependent pressor response. These findings may aid in elucidating the mechanisms by which women show uncontrolled BP in response to antihypertensive treatments targeting the RAS, improving approaches to reduce uncontrolled BP and chronic kidney disease incidences in women.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11208026/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139934609","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Distinct developmental reprogramming footprint of macrophages during acute kidney injury across species. 急性肾损伤期间不同物种巨噬细胞不同的发育重编程足迹
American journal of physiology. Renal physiology Pub Date : 2024-04-01 Epub Date: 2024-02-15 DOI: 10.1152/ajprenal.00013.2024
Michal Mrug, Elias Mrug, Frida Rosenblum, Jiandong Chen, Xiangqin Cui, Anupam Agarwal, Abolfazl Zarjou
{"title":"Distinct developmental reprogramming footprint of macrophages during acute kidney injury across species.","authors":"Michal Mrug, Elias Mrug, Frida Rosenblum, Jiandong Chen, Xiangqin Cui, Anupam Agarwal, Abolfazl Zarjou","doi":"10.1152/ajprenal.00013.2024","DOIUrl":"10.1152/ajprenal.00013.2024","url":null,"abstract":"<p><p>Acute kidney injury (AKI) is a common finding in hospitalized patients, particularly those who are critically ill. The development of AKI is associated with several adverse outcomes including mortality, morbidity, progression to chronic kidney disease, and an increase in healthcare expenditure. Despite the well-established negative impact of AKI and rigorous efforts to better define, identify, and implement targeted therapies, the overall approach to the treatment of AKI continues to principally encompass supportive measures. This enduring challenge is primarily due to the heterogeneous nature of insults that activate many independent and overlapping molecular pathways. Consequently, it is evident that the identification of common mechanisms that mediate the pathogenesis of AKI, independent of etiology and engaged pathophysiological pathways, is of paramount importance and could lead to the identification of novel therapeutic targets. To better distinguish the commonly modulated mechanisms of AKI, we explored the transcriptional characteristics of human kidney biopsies from patients with acute tubular necrosis (ATN), and acute interstitial nephritis (AIN) using a NanoString inflammation panel. Subsequently, we used publicly available single-cell transcriptional resources to better interpret the generated transcriptional findings. Our findings identify robust acute kidney injury (AKI-induced) developmental reprogramming of macrophages (MΦ) with the expansion of <i>C1Q</i><sup>+</sup>, <i>CD163</i><sup>+</sup> MΦ that is independent of the etiology of AKI and conserved across mouse and human species. These results would expand the current understanding of the pathophysiology of AKI and potentially offer novel targets for additional studies to enhance the translational transition of AKI research.<b>NEW & NOTEWORTHY</b> Our findings identify robust acute kidney injury (AKI)-induced developmental reprogramming of macrophages (MΦ) with the expansion of <i>C1Q</i><sup>+</sup>, <i>CD163</i><sup>+</sup> MΦ that is independent of the etiology of AKI and conserved across mouse and human species.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11208015/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139736919","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Liver and spleen predominantly mediate calciprotein particle clearance in a rat model of chronic kidney disease. 在慢性肾病大鼠模型中,肝脏和脾脏主要介导钙蛋白颗粒的清除。
American journal of physiology. Renal physiology Pub Date : 2024-04-01 Epub Date: 2024-02-29 DOI: 10.1152/ajprenal.00239.2023
Lara W Zeper, Caro Bos, Pieter A Leermakers, Gerben M Franssen, René Raavé, Joost G J Hoenderop, Jeroen H F de Baaij
{"title":"Liver and spleen predominantly mediate calciprotein particle clearance in a rat model of chronic kidney disease.","authors":"Lara W Zeper, Caro Bos, Pieter A Leermakers, Gerben M Franssen, René Raavé, Joost G J Hoenderop, Jeroen H F de Baaij","doi":"10.1152/ajprenal.00239.2023","DOIUrl":"10.1152/ajprenal.00239.2023","url":null,"abstract":"<p><p>Calciprotein particles (CPPs) provide an efficient mineral buffering system to prevent the complexation of phosphate and calcium in the circulation. However, in chronic kidney disease (CKD), the phosphate load exceeds the mineral buffering capacity, resulting in the formation of crystalline CPP2 particles. CPP2 have been associated with cardiovascular events and mortality. Moreover, CPP2 have been demonstrated to induce calcification in vitro. In this study, we examined the fate of CPP2 in a rat model of CKD. Calcification was induced in Sprague-Dawley rats by 5/6 nephrectomy (5/6-Nx) combined with a high-phosphate diet. Control rats received sham surgery and high-phosphate diet. Twelve weeks after surgery, kidney failure was significantly induced in 5/6-Nx rats as determined by enhanced creatinine and urea plasma levels and abnormal kidney histological architecture. Subsequently, radioactive and fluorescent (FITC)-labeled CPP2 ([<sup>89</sup>Zr]Zr-CPP2-FITC) were injected intravenously to determine clearance in vivo. Using positron emission tomography scans and radioactive biodistribution measurements, it was demonstrated that [<sup>89</sup>Zr]Zr-CPP2-FITC are mainly present in the liver and spleen in both 5/6-Nx and sham rats. Immunohistochemistry showed that [<sup>89</sup>Zr]Zr-CPP2-FITC are predominantly taken up by Kupffer cells and macrophages. However, [<sup>89</sup>Zr]Zr-CPP2-FITC could also be detected in hepatocytes. In the different parts of the aorta and in the blood, low values of [<sup>89</sup>Zr]Zr-CPP2-FITC were detectable, independent of the presence of calcification. CPP2 are cleared rapidly from the circulation by the liver and spleen in a rat model of CKD. In the liver, Kupffer cells, macrophages, and hepatocytes contribute to CPP2 clearance.<b>NEW & NOTEWORTHY</b> Calciprotein particles (CPPs) buffer calcium and phosphate in the blood to prevent formation of crystals. In CKD, increased phosphate levels may exceed the buffering capacity of CPPs, resulting in crystalline CPPs that induce calcification. This study demonstrates that labeled CPPs are predominantly cleared from the circulation in the liver by Kupffer cells, macrophages, and hepatocytes. Our results suggest that targeting liver CPP clearance may reduce the burden of crystalline CPP in the development of vascular calcification.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139992081","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
Sex differences in sympathetic activity and pulse wave velocity in adults with chronic kidney disease. 慢性肾脏病(CKD)成人交感神经活动和血管僵硬度的性别差异。
American journal of physiology. Renal physiology Pub Date : 2024-04-01 Epub Date: 2024-02-22 DOI: 10.1152/ajprenal.00308.2023
Matias G Zanuzzi, Jinhee Jeong, Dana R DaCosta, Jeanie Park
{"title":"Sex differences in sympathetic activity and pulse wave velocity in adults with chronic kidney disease.","authors":"Matias G Zanuzzi, Jinhee Jeong, Dana R DaCosta, Jeanie Park","doi":"10.1152/ajprenal.00308.2023","DOIUrl":"10.1152/ajprenal.00308.2023","url":null,"abstract":"<p><p>Chronic kidney disease (CKD) is characterized by sympathetic nervous system (SNS) overactivity that contributes to increased vascular stiffness and cardiovascular risk. Although it is well established that SNS activity and vascular stiffness are substantially elevated in CKD, whether sex differences in autonomic and vascular function exist in CKD remains unknown. We tested the hypothesis that compared with females, males with CKD have higher baseline sympathetic activity that is related to increased arterial stiffness. One hundred twenty-nine participants (96 males and 33 females) with CKD stages III and IV were recruited and enrolled. During two separate study visits, vascular stiffness was assessed by measuring carotid-to-femoral pulse wave velocity (cfPWV), and resting muscle sympathetic nerve activity (MSNA) was measured by microneurography. Males with CKD had higher resting MSNA compared with females with CKD (68 ± 16 vs. 55 ± 14 bursts/100 heart beats, <i>P</i> = 0.005), whereas there was no difference in cfPWV between the groups (<i>P</i> = 0.248). Resting MSNA was not associated with cfPWV in both males and females. In conclusion, males with CKD have higher resting sympathetic activity compared with females with CKD. However, there was no difference in vascular stiffness between the sexes. There was no correlation between resting MSNA and cfPWV, suggesting that non-neural mechanisms may play a greater role in the progression of vascular stiffness in CKD, particularly in females.<b>NEW & NOTEWORTHY</b> Males with chronic kidney disease (CKD) have higher resting muscle sympathetic nerve activity (MSNA) compared with females. There was no correlation between MSNA and carotid-to-femoral pulse wave velocity (cfPWV), suggesting that non-neural mechanisms may play a greater role in the progression of vascular stiffness in CKD. Sex differences in SNS activity may play a mechanistic role in observations from epidemiological studies suggesting greater cardiovascular risk in males compared with females with CKD.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11208017/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139934610","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
C-terminal fragment of fibroblast growth factor 23 improves heart function in murine models of high intact fibroblast growth factor 23. 成纤维细胞生长因子(FGF)-23 的 C 端片段可改善高完整 FGF23 小鼠模型的心脏功能。
American journal of physiology. Renal physiology Pub Date : 2024-04-01 Epub Date: 2024-02-01 DOI: 10.1152/ajprenal.00298.2023
Ming Chang Hu, James A Reneau, Mingjun Shi, Masaya Takahashi, Gaozhi Chen, Moosa Mohammadi, Orson W Moe
{"title":"C-terminal fragment of fibroblast growth factor 23 improves heart function in murine models of high intact fibroblast growth factor 23.","authors":"Ming Chang Hu, James A Reneau, Mingjun Shi, Masaya Takahashi, Gaozhi Chen, Moosa Mohammadi, Orson W Moe","doi":"10.1152/ajprenal.00298.2023","DOIUrl":"10.1152/ajprenal.00298.2023","url":null,"abstract":"<p><p>Cardiovascular disease (CVD) is the major cause of death in chronic kidney disease (CKD) and is associated with high circulating fibroblast growth factor (FGF)23 levels. It is unresolved whether high circulating FGF23 is a mere biomarker or pathogenically contributes to cardiomyopathy. It is also unknown whether the C-terminal FGF23 peptide (cFGF23), a natural FGF23 antagonist proteolyzed from intact FGF23 (iFGF23), retards CKD progression and improves cardiomyopathy. We addressed these questions in three murine models with high endogenous FGF23 and cardiomyopathy. First, we examined wild-type (WT) mice with CKD induced by unilateral ischemia-reperfusion and contralateral nephrectomy followed by a high-phosphate diet. These mice were continuously treated with intraperitoneal implanted osmotic minipumps containing either iFGF23 protein to further escalate FGF23 bioactivity, cFGF23 peptide to block FGF23 signaling, vehicle, or scrambled peptide as negative controls. Exogenous iFGF23 protein given to CKD mice exacerbated pathological cardiac remodeling and CKD progression, whereas cFGF23 treatment improved heart and kidney function, attenuated fibrosis, and increased circulating soluble Klotho. WT mice without renal insult placed on a high-phosphate diet and homozygous Klotho hypomorphic mice, both of whom develop moderate CKD and clear cardiomyopathy, were treated with cFGF23 or vehicle. Mice treated with cFGF23 in both models had improved heart and kidney function and histopathology. Taken together, these data indicate high endogenous iFGF23 is not just a mere biomarker but pathogenically deleterious in CKD and cardiomyopathy. Furthermore, attenuation of FGF23 bioactivity by cFGF23 peptide is a promising therapeutic strategy to protect the kidney and heart from high FGF23 activity.<b>NEW & NOTEWORTHY</b> There is a strong correlation between cardiovascular morbidity and high circulating fibroblast growth factor 23 (FGF23) levels, but causality was never proven. We used a murine chronic kidney disease (CKD) model to show that intact FGF23 (iFGF23) is pathogenic and contributes to both CKD progression and cardiomyopathy. Blockade of FGF23 signaling with a natural proteolytic product of iFGF23, C-terminal FGF23, alleviated kidney and cardiac histology, and function in three separate murine models of high endogenous FGF23.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11208029/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139652394","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","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