scholarly journals The renal microcirculation in chronic kidney disease: novel diagnostic methods and therapeutic perspectives

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Shulin Li ◽  
Fei Wang ◽  
Dong Sun

AbstractChronic kidney disease (CKD) affects 8–16% of the population worldwide and is characterized by fibrotic processes. Understanding the cellular and molecular mechanisms underpinning renal fibrosis is critical to the development of new therapeutics. Microvascular injury is considered an important contributor to renal progressive diseases. Vascular endothelium plays a significant role in responding to physical and chemical signals by generating factors that help maintain normal vascular tone, inhibit leukocyte adhesion and platelet aggregation, and suppress smooth muscle cell proliferation. Loss of the rich capillary network results in endothelial dysfunction, hypoxia, and inflammatory and oxidative effects and further leads to the imbalance of pro- and antiangiogenic factors, endothelial cell apoptosis and endothelial-mesenchymal transition. New techniques, including both invasive and noninvasive techniques, offer multiple methods to observe and monitor renal microcirculation and guide targeted therapeutic strategies. A better understanding of the role of endothelium in CKD will help in the development of effective interventions for renal microcirculation improvement. This review focuses on the role of microvascular injury in CKD, the methods to detect microvessels and the novel treatments to ameliorate renal fibrosis.

2013 ◽  
Vol 305 (12) ◽  
pp. F1637-F1644 ◽  
Author(s):  
May Y. W. Wong ◽  
Sonia Saad ◽  
Carol Pollock ◽  
Muh Geot Wong

With better understanding of the molecular mechanisms underpinning chronic kidney disease, the roles of inflammation and fibrosis are becoming increasingly inseparable. The progression of renal disease is characterized by pathomorphological changes that consist of early inflammatory responses followed by tubulointerstitial fibrosis, tubular atrophy, and glomerular and vascular sclerosis. Currently available therapies that reduce hypertension, proteinuria, hyperglycemia, and interruption of the renin-angiotensin-aldosterone system are at best only partially effective. Hence, there remains a need to explore agents targeting nonrenin-angiotensin-aldosterone system pathways. In this review, we discuss mechanistic aspects in the physiological and pathological role of semicarbazide-sensitive amine oxidase, a protein enzyme involved in cellular trafficking and inflammation, with respect to the kidney. We explore the evidence for the use of semicarbazide-sensitive amine oxidase inhibitors as potential agents in renal fibrosis to delay the onset and progression of chronic kidney disease.


2019 ◽  
Vol 3 (9) ◽  
Author(s):  
Rattiyaporn Kanlaya ◽  
Visith Thongboonkerd

ABSTRACT Chronic kidney disease (CKD) is a common public health problem worldwide characterized by gradual decline of renal function over months/years accompanied by renal fibrosis and failure in tissue wound healing after sustained injury. Patients with CKD frequently present with profound signs/symptoms that require medical treatment, mostly culminating in hemodialysis and renal transplantation. To prevent CKD more efficiently, there is an urgent need for better understanding of the pathogenic mechanisms and molecular pathways of the disease pathogenesis and progression, and for developing novel therapeutic targets. Recently, several lines of evidence have shown that epigallocatechin-3-gallate (EGCG), an abundant phytochemical polyphenol derived from Camellia sinensis, might be a promising bioactive compound for prevention of CKD development/progression. This review summarizes current knowledge of molecular mechanisms underlying renoprotective roles of EGCG in CKD based on available preclinical evidence (from both in vitro and in vivo animal studies), particularly its antioxidant property through preservation of mitochondrial function and activation of Nrf2 (nuclear factor erythroid 2-related factor 2)/HO-1 (heme oxygenase-1) signaling, anti-inflammatory activity, and protective effect against epithelial mesenchymal transition. Finally, future perspectives, challenges, and concerns regarding its clinical use in CKD and renal fibrosis are discussed.


2019 ◽  
Vol 20 (14) ◽  
pp. 3567 ◽  
Author(s):  
Teresa Seccia ◽  
Brasilina Caroccia ◽  
Maria Piazza ◽  
Gian Paolo Rossi

Accumulating evidence indicates that epithelial-to-mesenchymal transition (EMT), originally described as a key process for organ development and metastasis budding in cancer, plays a key role in the development of renal fibrosis in several diseases, including hypertensive nephroangiosclerosis. We herein reviewed the concept of EMT and its role in renal diseases, with particular focus on hypertensive kidney disease, the second leading cause of end-stage renal disease after diabetes mellitus. After discussing the pathophysiology of hypertensive nephropathy, the ‘classic’ view of hypertensive nephrosclerosis entailing hyalinization, and sclerosis of interlobular and afferent arterioles, we examined the changes occurring in the glomerulus and tubulo-interstitium and the studies that investigated the role of EMT and its molecular mechanisms in hypertensive kidney disease. Finally, we examined the reasons why some studies failed to provide solid evidence for renal EMT in hypertension.


2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Byung Min Ye ◽  
Il Young Kim ◽  
Min Jeong Kim ◽  
Soo Bong Lee ◽  
Dong Won Lee ◽  
...  

Abstract Background and Aims Acute kidney injury (AKI) is an underestimated, yet important risk factor for the development of chronic kidney disease (CKD), which is characterized by the tubulointerstitial fibrosis and tubular epithelial-mesenchymal transition (EMT). Akt has been reported to be involved in renal fibrosis and EMT. Thus, we investigated the role of Akt1, one of the three Akt isoforms, in the murine model of AKI to CKD progression. Method We subjected the wild type and Akt1−/− mice to unilateral ischemia-reperfusion injury (UIRI). UIRI was induced by clamping the left renal artery for 30 min followed by reperfusion. After 6 weeks of UIRI, the renal fibrosis and EMT were assessed by histology, immunohistochemistry, and western blot. Results After 6 weeks after UIRI, we found that Akt1, not Akt2 or Akt3, was activated in UIRI-kidney. The tubulointerstitial fibrosis was significantly alleviated in Akt1−/− mice compared with the wild type (WT) mice. Besides, the deletion of Akt1 decreased the expression of the vimentin and α-SMA and increased the expression of E-cadherin, indicating the suppression of tubular EMT. However, there was no difference in the activity of TGF-β1/Smad signalling, which is the potent inducer of renal fibrosis and EMT, between WT mice and Akt1−/− mice. The deletion of Akt1 also increased the GSK-3β activity and decreased the expression of β-catenin, Snail, and twist1. Conclusion Our findings demonstrate that the deletion of Akt1 attenuates the renal fibrosis and tubular EMT independently of TGF-β1/Smad signalling during the AKI to CKD progression. Akt1 may be the therapeutic target against the AKI to CKD progression.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Jinxiu Hu ◽  
Jiao Qiao ◽  
Qun Yu ◽  
Bing Liu ◽  
Junhui Zhen ◽  
...  

Abstract Background Acute kidney injury (AKI), with a high morbidity and mortality, is recognized as a risk factor for chronic kidney disease (CKD). AKI-CKD transition has been regarded as one of the most pressing unmet needs in renal diseases. Recently, studies have showed that salt inducible kinase 1 (SIK1) plays a role in epithelial-mesenchymal transition (EMT) and inflammation, which are the hallmarks of AKI-CKD transition. However, whether SIK1 is involved in AKI-CKD transition and by what mechanism it regulates AKI-CKD transition remains unknown. Methods We firstly detected the expression of SIK1 in kidney tissues of AKI patients and AKI mice by immunohistochemistry staining, and then we established Aristolochic acid (AA)-induced AKI-CKD transition model in C57BL/6 mice and HK2 cells. Subsequently, we performed immunohistochemistry staining, ELISA, real-time PCR, Western blot, immunofluorescence staining and Transwell assay to explore the role and underlying mechanism of SIK1 on AKI-CKD transition. Results The expression of SIK1 was down-regulated in AKI patients, AKI mice, AA-induced AKI-CKD transition mice, and HK2 cells. Functional analysis revealed that overexpression of SIK1 alleviated AA-induced AKI-CKD transition and HK2 cells injury in vivo and in vitro. Mechanistically, we demonstrated that SIK1 mediated AA-induced AKI-CKD transition by regulating WNT/β-catenin signaling, the canonical pathway involved in EMT, inflammation and renal fibrosis. In addition, we discovered that inhibition of WNT/β-catenin pathway and its downstream transcription factor Twist1 ameliorated HK2 cells injury, delaying the progression of AKI-CKD transition. Conclusions Our study demonstrated, for the first time, a protective role of SIK1 in AKI-CKD transition by regulating WNT/β-catenin signaling pathway and its downstream transcription factor Twist1, which will provide novel insights into the prevention and treatment AKI-CKD transition in the future.


2021 ◽  
Vol 12 ◽  
Author(s):  
Myriam Dao ◽  
Helene François

Chronic kidney disease (CKD) concerns millions of individuals worldwide, with few therapeutic strategies available to date. Recent evidence suggests that the endocannabinoid system (ECS) could be a new therapeutic target to prevent CKD. ECS combines receptors, cannabinoid receptor type 1 (CB1R) and type 2 (CB2R), and ligands. The most prominent receptor within the kidney is CB1R, its endogenous local ligands being anandamide and 2-arachidonoylglycerol. Therefore, the present review focuses on the therapeutic potential of CB1R and not CB2R. In the normal kidney, CB1R is expressed in many cell types, especially in the vasculature where it contributes to the regulation of renal hemodynamics. CB1R could also participate to water and sodium balance and to blood pressure regulation but its precise role remains to decipher. CB1R promotes renal fibrosis in both metabolic and non-metabolic nephropathies. In metabolic syndrome, obesity and diabetes, CB1R inhibition not only improves metabolic parameters, but also exerts a direct role in preventing renal fibrosis. In non-metabolic nephropathies, its inhibition reduces the development of renal fibrosis. There is a growing interest of the industry to develop new CB1R antagonists without central nervous side-effects. Experimental data on renal fibrosis are encouraging and some molecules are currently under early-stage clinical phases (phases I and IIa studies). In the present review, we will first describe the role of the endocannabinoid receptors, especially CB1R, in renal physiology. We will next explore the role of endocannabinoid receptors in both metabolic and non-metabolic CKD and renal fibrosis. Finally, we will discuss the therapeutic potential of CB1R inhibition using the new pharmacological approaches. Overall, the new pharmacological blockers of CB1R could provide an additional therapeutic toolbox in the management of CKD and renal fibrosis from both metabolic and non-metabolic origin.


2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Sharma Pravesh ◽  
Kalyani Karnam ◽  
Kavita Sedmaki ◽  
Kirti Hira ◽  
Onkar Prakash Kulkarni

Abstract Background and Aims Role of epigenetic factors like histone deacetylases (HDACs) is largely unexplored in the pathogenesis of Nephrocalcinosis-related chronic kidney disease. The present study was performed to evaluate the functional role of HDAC5 in fibroblast activation in in-vitro and in a mouse model of Nephrocalcinosis-related chronic kidney disease. Method C57BL/6 male mice (6-7weeks old) were procured from registered CPCSEA breeder (Hyderabad, India), NRK49F cell line were generously provided by NCCS (Pune, India). All experimental procedures were approved by the animal ethics committee of the institute. Nephrolithiasis (oxalate nephropathy) was induced by feeding oxalate rich diet (Ssniff, Soest, Germany) to C57/BL6 mice for 10 days. NRK49F cells were stimulated with LPS (1µg/ml) for 2hrs followed by TGF-β (25ng/ml) for 24hrs. To achieve HDAC5 knockdown, the cells were pre-incubated with HDAC5 specific siRNA for 24hrs before stimulating with LPS. In another set of experiment, cells were incubated with 4-sodium phenyl butyrate(PBA) a non-selective HDAC5 and HDAC4 inhibitor at a dose of 1mM. 24 hrs after LPS stimulation cell lysates were analysed for protein expression of α-SMA, Collagen1a, KLF2, NLRP3, HDAC5 and β-actin. Mice fed with high oxalate diet were treated with PBA (500mg/kg) twice a day and on day 10 mice were sacrificed to analyse renal function and fibrosis parameters. Mice kidney tissues were analysed for crystal deposition (pizaalato staining), renal fibrosis (Picrosirius staining), and renal histology (H&E staining). The fibrosis markers were analysed by RT-PCR and immunoblotting. Renal function was determined by plasma BUN and creatinine analysis. Diet containing high oxalate with calcium was provided to mice in the control diet group (negative diet control). Results Renal fibroblasts (NRK 49F) stimulated with LPS and TGF-β showed upregulation of HDAC5, NLRP3, α-SMA, and Collagen-1a. Depletion of HDAC5 expression with HDAC5 siRNA significantly reduced expression of NLRP-3, α-SMA, and Collagen-1 in stimulated NRK49F cells. The expression of KLF2 in HDAC5 depleted cells was found to be upregulated. PBA treatment also reduced the expression of HDAC5 in NRK49F cells and had significant reduction in the expression of NLRP3 along with fibroblast activation markers, while KLF-2 expression was found to be up-regulated. Similar to our observation in stimulated renal fibroblasts, the expression of HDAC5 was found to be upregulated in mouse model of oxalate nephropathy. Treatment with PBA showed significant downregulation of HDAC5 in kidneys of the mice fed with high oxalate diet. Mice treated with PBA showed down regulated renal expression of NLRP3, Collagen1a, α-SMA, TGF-β in comparison to vehicle treated mice. Treatment with PBA showed significant upregulation of KLF2 expression in kidney. PBA treated mice showed significant renal protection during nephrolithiasis as indicated by reduced plasma BUN and creatinine levels. Oxalate crystal deposition index was similar in all high oxalate diet groups. PBA treatment showed overall better renal histological protection observed through H&E (reduced tubular injury score) and picrosirius staining (reduced collagen deposition). Conclusion HDAC5 knockdown or HDAC5 inhibition with PBA attenuates fibroblasts activation by inhibiting NLRP3 expression. Treatment with PBA in mice with Nephrocalcinosis-related chronic kidney disease showed significant protection against renal fibrosis with downregulation of NLRP3 expression. We propose HDAC5 as a novel regulator of fibroblast activation in Nephrocalcinosis-related chronic kidney disease.


2015 ◽  
Vol 308 (11) ◽  
pp. F1189-F1196 ◽  
Author(s):  
Stephanie Stangenberg ◽  
Hui Chen ◽  
Muh Geot Wong ◽  
Carol A. Pollock ◽  
Sonia Saad

The role of an adverse in utero environment in the programming of chronic kidney disease in the adult offspring is increasingly recognized. The cellular and molecular mechanisms linking the in utero environment and future disease susceptibility remain unknown. Maternal smoking is a common modifiable adverse in utero exposure, potentially associated with both mitochondrial dysfunction and epigenetic modification in the offspring. While studies are emerging that point toward a key role of mitochondrial dysfunction in acute and chronic kidney disease, it may have its origin in early development, becoming clinically apparent when secondary insults occur. Aberrant epigenetic programming may add an additional layer of complexity to orchestrate fibrogenesis in the kidney and susceptibility to chronic kidney disease in later life. In this review, we explore the evidence for mitochondrial dysfunction and epigenetic modification through aberrant DNA methylation as key mechanistic aspects of fetal programming of chronic kidney disease and discuss their potential use in diagnostics and targets for therapy.


2013 ◽  
Vol 2013 ◽  
pp. 1-7 ◽  
Author(s):  
Li Xiao ◽  
Ming Wang ◽  
Shikun Yang ◽  
Fuyou Liu ◽  
Lin Sun

The Wnt family of proteins belongs to a group of secreted lipid-modified glycoproteins with highly conserved cysteine residues. Prior results indicate that Wnt/β-catenin signaling plays a prominent role in cell differentiation, adhesion, survival, and apoptosis and is involved in organ development, tumorigenesis, and tissue fibrosis, among other functions. Accumulating evidence has suggested that Wnt/β-catenin exhibits a pivotal function in the progression of diabetic nephropathy (DN). In this review, we focused on discussing the dual role of Wnt/β-catenin in apoptosis and epithelial mesenchymal transition (EMT) formation of mesangial cells. Moreover, we also elucidated the effect of Wnt/β-catenin in podocyte dysfunction, tubular EMT formation, and renal fibrosis under DN conditions. In addition, the molecular mechanisms involved in this process are introduced. This information provides a novel molecular target of Wnt/β-catenin for the protection of kidney damage and in delay of the progression of DN.


Sign in / Sign up

Export Citation Format

Share Document