Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.474
Filter
1.
Cell Death Dis ; 15(6): 397, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844455

ABSTRACT

Integrin αvß6 holds promise as a therapeutic target for organ fibrosis, yet targeted therapies are hampered by concerns over inflammatory-related side effects. The role of αvß6 in renal inflammation remains unknown, and clarifying this issue is crucial for αvß6-targeted treatment of chronic kidney disease (CKD). Here, we revealed a remarkable positive correlation between overexpressed αvß6 in proximal tubule cells (PTCs) and renal inflammation in CKD patients and mouse models. Notably, knockout of αvß6 not only significantly alleviated renal fibrosis but also reduced inflammatory responses in mice, especially the infiltration of pro-inflammatory macrophages. Furthermore, conditional knockout of αvß6 in PTCs in vivo and co-culture of PTCs with macrophages in vitro showed that depleting αvß6 in PTCs suppressed the migration and pro-inflammatory differentiation of macrophages. Screening of macrophage activators showed that αvß6 in PTCs activates macrophages via secreting IL-34. IL-34 produced by PTCs was significantly diminished by αvß6 silencing, and reintroduction of IL-34 restored macrophage activities, while anti-IL-34 antibody restrained macrophage activities enhanced by αvß6 overexpression. Moreover, RNA-sequencing of PTCs and verification experiments demonstrated that silencing αvß6 in PTCs blocked hypoxia-stimulated IL-34 upregulation and secretion by inhibiting YAP expression, dephosphorylation, and nuclear translocation, which resulted in the activation of Hippo signaling. While application of a YAP agonist effectively recurred IL-34 production by PTCs, enhancing the subsequent macrophage migration and activation. Besides, reduced IL-34 expression and YAP activation were also observed in global or PTCs-specific αvß6-deficient injured kidneys. Collectively, our research elucidates the pro-inflammatory function and YAP/IL-34/macrophage axis-mediated mechanism of αvß6 in renal inflammation, providing a solid rationale for the use of αvß6 inhibition to treat kidney inflammation and fibrosis.


Subject(s)
Integrins , Macrophages , Mice, Knockout , Renal Insufficiency, Chronic , Animals , Macrophages/metabolism , Mice , Humans , Integrins/metabolism , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/metabolism , Inflammation/pathology , Inflammation/metabolism , Male , Antigens, Neoplasm/metabolism , Mice, Inbred C57BL , Signal Transduction , Disease Models, Animal , YAP-Signaling Proteins/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Fibrosis
2.
Ren Fail ; 46(2): 2359638, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38832484

ABSTRACT

Emerging data have revealed that damage to tubular epithelial cell is a driving force in the progression of diabetic kidney disease (DKD). However, the specific mechanisms by which lipotoxicity contributes to the injury of these cells, thereby influencing the development of DKD, are yet to be fully understood. Here, we analyzed the GSE 30529 microarray datasets of human tubulointerstitial tissue samples from the Gene Expression Omnibus database (GEO). Concurrently, we conducted RNA-sequencing on palmitic acid (PA)-treated human renal proximal tubule epithelial cells (HK2 cells). After normalization, the differentially expressed genes (DEGs) were screened by R software and gene ontology (GO) enrichment analysis was conducted, and lysosomal-associated protein transmembrane 5 (LAPTM5) was finally selected. Our findings indicate that the expression of LAPTM5 was obviously increased in DKD patients, and the correlation between LAPTM5, and other clinical parameters of DKD was analyzed using the Spearman correlation analysis. The potential of LAPTM5 as a prognostic biomarker for DKD was further consolidated through receiver operating characteristic (ROC) analysis. To further verify the function of LAPTM5, we established mouse or in vitro systems mimicking DKD. The results showed that a consistent upregulation of LAPTM5, which was also found to be linked with inflammatory mediators within the context of DKD. Additionally, LAPTM5 silencing significantly downregulated mRNA expression of inflammatory factors in PA-treated HK2 cells. These results indicate that LAPTM5 is a potential biomarker and therapeutic treatment target for DKD. This discovery paves the way for future research and development of targeted interventions aimed at mitigating the progression of this prevalent condition.


Subject(s)
Computational Biology , Diabetic Nephropathies , Membrane Proteins , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Humans , Animals , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Cell Line , Palmitic Acid/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Male , Mice, Inbred C57BL , Up-Regulation , Biomarkers/metabolism
3.
Ren Fail ; 46(1): 2347462, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38832497

ABSTRACT

Diabetic nephropathy (DN) is one of the most serious and frequent complications among diabetes patients and presently constitutes vast the cases of end-stage renal disease worldwide. Tubulointerstitial fibrosis is a crucial factor related to the occurrence and progression of DN. Oridonin (Ori) is a diterpenoid derived from rubescens that has diverse pharmacological properties. Our previous study showed that Ori can protect against DN by decreasing the inflammatory response. However, whether Ori can alleviate renal fibrosis in DN remains unknown. Here, we investigated the mechanism through which Ori affects the Wnt/ß-catenin signaling pathway in diabetic rats and human proximal tubular epithelial cells (HK-2) exposed to high glucose (HG) levels. Our results revealed that Ori treatment markedly decreased urinary protein excretion levels, improved renal function and alleviated renal fibrosis in diabetic rats. In vitro, HG treatment increased the migration of HK-2 cells while reducing their viability and proliferation rate, and treatment with Ori reversed these changes. Additionally, the knockdown of ß-catenin arrested cell migration and reduced the expression levels of Wnt/ß-catenin signaling-related molecules (Wnt4, p-GSK3ß and ß-catenin) and fibrosis-related molecules (α-smooth muscle actin, collagen I and fibronectin), and Ori treatment exerted an effect similar to that observed after the knockdown of ß-catenin. Furthermore, the combination of Ori treatment and ß-catenin downregulation exerted more pronounced biological effects than treatment alone. These findings may provide the first line of evidence showing that Ori alleviates fibrosis in DN by inhibiting the Wnt/ß-catenin signaling pathway and thereby reveal a novel therapeutic avenue for treating tubulointerstitial fibrosis.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Diterpenes, Kaurane , Fibrosis , Rats, Sprague-Dawley , Wnt Signaling Pathway , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/etiology , Wnt Signaling Pathway/drug effects , Animals , Diterpenes, Kaurane/pharmacology , Diterpenes, Kaurane/therapeutic use , Rats , Fibrosis/drug therapy , Humans , Male , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Cell Line , beta Catenin/metabolism , Cell Movement/drug effects , Kidney/pathology , Kidney/drug effects , Cell Proliferation/drug effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/metabolism
4.
FASEB J ; 38(10): e23688, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38780519

ABSTRACT

Diabetic nephropathy (DN) is a major cause of chronic kidney disease. Microalbuminuria is currently the most common non-invasive biomarker for the early diagnosis of DN. However, renal structural damage may have advanced when albuminuria is detected. In this study, we sought biomarkers for early DN diagnosis through proteomic analysis of urinary extracellular vesicles (uEVs) from type 2 diabetic model rats and normal controls. Isocitrate dehydrogenase 1 (IDH1) was significantly increased in uEVs from diabetic model rats at the early stage despite minimal differences in albuminuria between the groups. Calorie restriction significantly suppressed the increase in IDH1 in uEVs and 24-hour urinary albumin excretion, suggesting that the increase in IDH1 in uEVs was associated with the progression of DN. Additionally, we investigated the origin of IDH1-containing uEVs based on their surface sugar chains. Lectin affinity enrichment and immunohistochemical staining showed that IDH1-containing uEVs were derived from proximal tubules. These findings suggest that the increase in IDH1 in uEVs reflects pathophysiological alterations in the proximal tubules and that IDH1 in uEVs may serve as a potential biomarker of DN in the proximal tubules.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Extracellular Vesicles , Isocitrate Dehydrogenase , Kidney Tubules, Proximal , Up-Regulation , Animals , Isocitrate Dehydrogenase/metabolism , Isocitrate Dehydrogenase/genetics , Extracellular Vesicles/metabolism , Rats , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Diabetes Mellitus, Type 2/urine , Diabetes Mellitus, Type 2/metabolism , Male , Diabetic Nephropathies/urine , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/urine , Rats, Sprague-Dawley , Biomarkers/urine , Biomarkers/metabolism
5.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731829

ABSTRACT

Kidney ischemia and reperfusion injury (IRI) is a significant contributor to acute kidney injury (AKI), characterized by tubular injury and kidney dysfunction. Salvador family WW domain containing protein 1 (SAV1) is a key component of the Hippo pathway and plays a crucial role in the regulation of organ size and tissue regeneration. However, whether SAV1 plays a role in kidney IRI is not investigated. In this study, we investigated the role of SAV1 in kidney injury and regeneration following IRI. A proximal tubule-specific knockout of SAV1 in kidneys (SAV1ptKO) was generated, and wild-type and SAV1ptKO mice underwent kidney IRI or sham operation. Plasma creatinine and blood urea nitrogen were measured to assess kidney function. Histological studies, including periodic acid-Schiff staining and immunohistochemistry, were conducted to assess tubular injury, SAV1 expression, and cell proliferation. Western blot analysis was employed to assess the Hippo pathway-related and proliferation-related proteins. SAV1 exhibited faint expression in the proximal tubules and was predominantly expressed in the connecting tubule to the collecting duct. At 48 h after IRI, SAV1ptKO mice continued to exhibit severe kidney dysfunction, compared to attenuated kidney dysfunction in wild-type mice. Consistent with the functional data, severe tubular damage induced by kidney IRI in the cortex was significantly decreased in wild-type mice at 48 h after IRI but not in SAV1ptKO mice. Furthermore, 48 h after IRI, the number of Ki67-positive cells in the cortex was significantly higher in wild-type mice than SAV1ptKO mice. After IRI, activation and expression of Hippo pathway-related proteins were enhanced, with no significant differences observed between wild-type and SAV1ptKO mice. Notably, at 48 h after IRI, protein kinase B activation (AKT) was significantly enhanced in SAV1ptKO mice compared to wild-type mice. This study demonstrates that SAV1 deficiency in the kidney proximal tubule worsens the injury and delays kidney regeneration after IRI, potentially through the overactivation of AKT.


Subject(s)
Acute Kidney Injury , Cell Cycle Proteins , Kidney Tubules, Proximal , Mice, Knockout , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/etiology , Acute Kidney Injury/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Male , Cell Proliferation , Signal Transduction , Hippo Signaling Pathway , Mice, Inbred C57BL , Disease Models, Animal
6.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38732005

ABSTRACT

In calcium nephrolithiasis (CaNL), most calcium kidney stones are identified as calcium oxalate (CaOx) with variable amounts of calcium phosphate (CaP), where CaP is found as the core component. The nucleation of CaP could be the first step of CaP+CaOx (mixed) stone formation. High urinary supersaturation of CaP due to hypercalciuria and an elevated urine pH have been described as the two main factors in the nucleation of CaP crystals. Our previous in vivo findings (in mice) show that transient receptor potential canonical type 3 (TRPC3)-mediated Ca2+ entry triggers a transepithelial Ca2+ flux to regulate proximal tubular (PT) luminal [Ca2+], and TRPC3-knockout (KO; -/-) mice exhibited moderate hypercalciuria and microcrystal formation at the loop of Henle (LOH). Therefore, we utilized TRPC3 KO mice and exposed them to both hypercalciuric [2% calcium gluconate (CaG) treatment] and alkalineuric conditions [0.08% acetazolamide (ACZ) treatment] to generate a CaNL phenotype. Our results revealed a significant CaP and mixed crystal formation in those treated KO mice (KOT) compared to their WT counterparts (WTT). Importantly, prolonged exposure to CaG and ACZ resulted in a further increase in crystal size for both treated groups (WTT and KOT), but the KOT mice crystal sizes were markedly larger. Moreover, kidney tissue sections of the KOT mice displayed a greater CaP and mixed microcrystal formation than the kidney sections of the WTT group, specifically in the outer and inner medullary and calyceal region; thus, a higher degree of calcifications and mixed calcium lithiasis in the kidneys of the KOT group was displayed. In our effort to find the Ca2+ signaling pathophysiology of PT cells, we found that PT cells from both treated groups (WTT and KOT) elicited a larger Ca2+ entry compared to the WT counterparts because of significant inhibition by the store-operated Ca2+ entry (SOCE) inhibitor, Pyr6. In the presence of both SOCE (Pyr6) and ROCE (receptor-operated Ca2+ entry) inhibitors (Pyr10), Ca2+ entry by WTT cells was moderately inhibited, suggesting that the Ca2+ and pH levels exerted sensitivity changes in response to ROCE and SOCE. An assessment of the gene expression profiles in the PT cells of WTT and KOT mice revealed a safeguarding effect of TRPC3 against detrimental processes (calcification, fibrosis, inflammation, and apoptosis) in the presence of higher pH and hypercalciuric conditions in mice. Together, these findings show that compromise in both the ROCE and SOCE mechanisms in the absence of TRPC3 under hypercalciuric plus higher tubular pH conditions results in higher CaP and mixed crystal formation and that TRPC3 is protective against those adverse effects.


Subject(s)
Calcium Oxalate , Hypercalciuria , Kidney Calculi , Mice, Knockout , Animals , Hypercalciuria/metabolism , Hypercalciuria/genetics , Hydrogen-Ion Concentration , Mice , Calcium Oxalate/metabolism , Kidney Calculi/metabolism , Kidney Calculi/etiology , Kidney Calculi/pathology , Calcium Phosphates/metabolism , Nephrolithiasis/metabolism , Nephrolithiasis/genetics , Nephrolithiasis/pathology , Calcium/metabolism , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Disease Models, Animal , Mice, Inbred C57BL , Acetazolamide/pharmacology
7.
Cell Mol Life Sci ; 81(1): 244, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38814462

ABSTRACT

Four-and-a-half LIM domains protein 2 (FHL2) is an adaptor protein that may interact with hypoxia inducible factor 1α (HIF-1α) or ß-catenin, two pivotal protective signaling in acute kidney injury (AKI). However, little is known about the regulation and function of FHL2 during AKI. We found that FHL2 was induced in renal tubular cells in patients with acute tubular necrosis and mice model of ischemia-reperfusion injury (IRI). In cultured renal proximal tubular cells (PTCs), hypoxia induced FHL2 expression and promoted the binding of HIF-1 to FHL2 promoter. Compared with control littermates, mice with PTC-specific deletion of FHL2 gene displayed worse renal function, more severe morphologic lesion, more tubular cell death and less cell proliferation, accompanying by downregulation of AQP1 and Na, K-ATPase after IRI. Consistently, loss of FHL2 in PTCs restricted activation of HIF-1 and ß-catenin signaling simultaneously, leading to attenuation of glycolysis, upregulation of apoptosis-related proteins and downregulation of proliferation-related proteins during IRI. In vitro, knockdown of FHL2 suppressed hypoxia-induced activation of HIF-1α and ß-catenin signaling pathways. Overexpression of FHL2 induced physical interactions between FHL2 and HIF-1α, ß-catenin, GSK-3ß or p300, and the combination of these interactions favored the stabilization and nuclear translocation of HIF-1α and ß-catenin, enhancing their mediated gene transcription. Collectively, these findings identify FHL2 as a direct downstream target gene of HIF-1 signaling and demonstrate that FHL2 could play a critical role in protecting against ischemic AKI by promoting the activation of HIF-1 and ß-catenin signaling through the interactions with its multiple protein partners.


Subject(s)
Acute Kidney Injury , Kidney Tubules, Proximal , LIM-Homeodomain Proteins , Muscle Proteins , Reperfusion Injury , Transcription Factors , beta Catenin , Animals , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Muscle Proteins/metabolism , Muscle Proteins/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/genetics , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/genetics , Mice , beta Catenin/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Male , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Signal Transduction , Mice, Inbred C57BL , Mice, Knockout , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Cell Proliferation , Apoptosis
8.
Mol Biol Rep ; 51(1): 620, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38709349

ABSTRACT

BACKGROUND: Recent years of evidence suggest the crucial role of renal tubular cells in developing diabetic kidney disease. Scopoletin (SCOP) is a plant-based coumarin with numerous biological activities. This study aimed to determine the effect of SCOP on renal tubular cells in developing diabetic kidney disease and to elucidate mechanisms. METHODS AND RESULTS: In this study, SCOP was evaluated in vitro using renal proximal tubular (HK-2) cells under hyperglycemic conditions to understand its mechanism of action. In HK-2 cells, SCOP alleviated the high glucose-generated reactive oxygen species (ROS), restored the levels of reduced glutathione, and decreased lipid peroxidation. High glucose-induced alteration in the mitochondrial membrane potential was markedly restored in the SCOP-treated cells. Moreover, SCOP significantly reduced the high glucose-induced apoptotic cell population in the Annexin V-FITC flow cytometry study. Furthermore, high glucose markedly elevated the mRNA expression of fibrotic and extracellular matrix (ECM) components, namely, transforming growth factor (TGF)-ß, alfa-smooth muscle actin (α-SMA), collagen I, and collagen III, in HK-2 cells compared to the untreated cells. SCOP treatment reduced these mRNA expressions compared to the high glucose-treated cells. Collagen I and TGF-ß protein levels were also significantly reduced in the SCOP-treated cells. Further findings in HK-2 cells revealed that SCOP interfered with the epithelial-mesenchymal transition (EMT) in the high glucose-treated HK-2 cells by normalizing E-cadherin and downregulating the vimentin and α-SMA proteins. CONCLUSIONS: In conclusion, SCOP modulates the high glucose-generated renal tubular cell oxidative damage and accumulation of ECM components and may be a promising molecule against diabetic nephropathy.


Subject(s)
Diabetic Nephropathies , Epithelial-Mesenchymal Transition , Glucose , Kidney Tubules, Proximal , Oxidative Stress , Reactive Oxygen Species , Scopoletin , Humans , Epithelial-Mesenchymal Transition/drug effects , Glucose/metabolism , Glucose/pharmacology , Glucose/toxicity , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Oxidative Stress/drug effects , Scopoletin/pharmacology , Cell Line , Reactive Oxygen Species/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/drug therapy , Apoptosis/drug effects , Fibrosis , Membrane Potential, Mitochondrial/drug effects , Lipid Peroxidation/drug effects
9.
Sci Rep ; 14(1): 9357, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38653823

ABSTRACT

The advent of micro-physiological systems (MPS) in biomedical research has enabled the introduction of more complex and relevant physiological into in vitro models. The recreation of complex morphological features in three-dimensional environments can recapitulate otherwise absent dynamic interactions in conventional models. In this study we developed an advanced in vitro Renal Cell Carcinoma (RCC) that mimics the interplay between healthy and malignant renal tissue. Based on the TissUse Humimic platform our model combines healthy renal proximal tubule epithelial cells (RPTEC) and RCC. Co-culturing reconstructed RPTEC tubules with RCC spheroids in a closed micro-perfused circuit resulted in significant phenotypical changes to the tubules. Expression of immune factors revealed that interleukin-8 (IL-8) and tumor necrosis factor-alfa (TNF-α) were upregulated in the non-malignant cells while neutrophil gelatinase-associated lipocalin (NGAL) was downregulated in both RCC and RPTEC. Metabolic analysis showed that RCC prompted a shift in the energy production of RPTEC tubules, inducing glycolysis, in a metabolic adaptation that likely supports RCC growth and immunogenicity. In contrast, RCC maintained stable metabolic activity, emphasizing their resilience to external factors. RNA-seq and biological process analysis of primary RTPTEC tubules demonstrated that the 3D tubular architecture and MPS conditions reverted cells to a predominant oxidative phosphorylate state, a departure from the glycolytic metabolism observed in 2D culture. This dynamic RCC co-culture model, approximates the physiology of healthy renal tubules to that of RCC, providing new insights into tumor-host interactions. Our approach can show that an RCC-MPS can expand the complexity and scope of pathophysiology and biomarker studies in kidney cancer research.


Subject(s)
Carcinoma, Renal Cell , Coculture Techniques , Epithelial Cells , Kidney Neoplasms , Kidney Tubules, Proximal , Humans , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Cell Line, Tumor , Lipocalin-2/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
10.
Biomed Pharmacother ; 175: 116666, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677246

ABSTRACT

Flavored e-liquid use has become popular among e-cigarette users recently, but the effects of such products outside the lung are not well characterized. In this work, acute exposure to the popular flavoring cinnamaldehyde (CIN) was performed on human proximal tubule (HK-2) kidney cells. Cells were exposed to 0-100 µM CIN for 24-48 h and cellular stress responses were assessed. Mitochondrial viability via MTT assay was significantly decreased at 20 µM for 24 and 48 h exposure. Seahorse XFp analysis showed significantly decreased mitochondrial energy output at 20 µM by 24 h exposure, in addition to significantly reduced ATP Synthase expression. Seahorse analysis also revealed significantly decreased glycolytic function at 20 µM by 24 h exposure, suggesting inability of glycolytic processes to compensate for reduced mitochondrial energy output. Cleaved caspase-3 expression, a mediator of apoptosis, was significantly increased at the 24 h mark. C/EBP homologous protein (CHOP) expression, a mediator of ER-induced apoptosis, was induced by 48 h and subsequently lost at the highest concentration of 100 µM. This decrease was accompanied by a simultaneous decrease in its downstream target cleaved caspase-3 at the 48 h mark. The autophagy marker microtubule-associated protein 1 A/1B light chain 3 (LC3B-I and LC3B-II) expression was significantly increased at 100 µM by 24 h. Autophagy-related 7 (ATG7) protein and mitophagy-related proteins PTEN-induced putative kinase 1 (PINK1) and PARKIN expression were significantly reduced at 24 and 48 h exposure. These results indicate acute exposure to CIN in the kidney HK-2 model induces mitochondrial dysfunction and cellular stress responses.


Subject(s)
Acrolein , Apoptosis , Flavoring Agents , Kidney Tubules, Proximal , Mitochondria , Humans , Acrolein/pharmacology , Acrolein/analogs & derivatives , Acrolein/toxicity , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Flavoring Agents/toxicity , Flavoring Agents/pharmacology , Cell Line , Mitochondria/drug effects , Mitochondria/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Stress, Physiological/drug effects , Cell Survival/drug effects , Endoplasmic Reticulum Stress/drug effects , Glycolysis/drug effects , Caspase 3/metabolism
11.
Am J Physiol Renal Physiol ; 326(5): F827-F838, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38482555

ABSTRACT

In the aftermath of acute kidney injury (AKI), surviving proximal tubule epithelia repopulate injured tubules to promote repair. However, a portion of cells fail to repair [termed failed-repair proximal tubule cells (FR-PTCs)] and exert ongoing proinflammatory and profibrotic effects. To better understand the molecular drivers of the FR-PTC state, we reanalyzed a mouse ischemia-reperfusion injury single-nucleus RNA-sequencing (snRNA-seq) atlas to identify Traf2 and Nck interacting kinase (Tnik) to be exclusively expressed in FR-PTCs but not in healthy or acutely injured proximal tubules after AKI (2 and 6 wk) in mice. We confirmed expression of Tnik protein in injured mouse and human tissues by immunofluorescence. Then, to determine the functional role of Tnik in FR-PTCs, we depleted TNIK with siRNA in two human renal proximal tubule epithelial cell lines (primary and immortalized hRPTECs) and analyzed each by bulk RNA-sequencing. Pathway analysis revealed significant upregulation of inflammatory signaling pathways, whereas pathways associated with differentiated proximal tubules such as organic acid transport were significantly downregulated. TNIK gene knockdown drove reduced cell viability and increased apoptosis, including differentially expressed poly(ADP-ribose) polymerase (PARP) family members, cleaved PARP-1 fragments, and increased annexin V binding to phosphatidylserine. Together, these results indicate that Tnik upregulation in FR-PTCs acts in a compensatory fashion to suppress inflammation and promote proximal tubule epithelial cell survival after injury. Modulating TNIK activity may represent a prorepair therapeutic strategy after AKI.NEW & NOTEWORTHY The molecular drivers of successful and failed repair in the proximal tubule after acute kidney injury (AKI) are incompletely understood. We identified Traf2 and Nck interacting kinase (Tnik) to be exclusively expressed in failed-repair proximal tubule cells after AKI. We tested the effect of siTNIK depletion in two proximal tubule cell lines followed by bulk RNA-sequencing analysis. Our results indicate that TNIK acts to suppress inflammatory signaling and apoptosis in injured renal proximal tubule epithelial cells to promote cell survival.


Subject(s)
Acute Kidney Injury , Apoptosis , Epithelial Cells , Kidney Tubules, Proximal , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Animals , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 2/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/genetics , Signal Transduction , Disease Models, Animal , Mice , Mice, Inbred C57BL , Cell Line , Inflammation/metabolism , Inflammation/pathology , Male
12.
Am J Physiol Renal Physiol ; 326(6): F981-F987, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38545650

ABSTRACT

Cystinosis is an autosomal recessive lysosomal storage disorder, caused by mutations in the CTNS gene, resulting in an absent or altered cystinosin (CTNS) protein. Cystinosin exports cystine out of the lysosome, with a malfunction resulting in cystine accumulation and a defect in other cystinosin-mediated pathways. Cystinosis is a systemic disease, but the kidneys are the first and most severely affected organs. In the kidney, the disease initially manifests as a generalized dysfunction in the proximal tubules (also called renal Fanconi syndrome). MFSD12 is a lysosomal cysteine importer that directly affects the cystine levels in melanoma cells, HEK293T cells, and cystinosis patient-derived fibroblasts. In this study, we aimed to evaluate MFSD12 mRNA levels in cystinosis patient-derived proximal tubular epithelial cells (ciPTECs) and to study the effect of MFSD12 knockout on cystine levels. We showed similar MFSD12 mRNA expression in patient-derived ciPTECs in comparison with the control cells. CRISPR MFSD12 knockout in a patient-derived ciPTEC (CTNSΔ57kb) resulted in significantly reduced cystine levels. Furthermore, we evaluated proximal tubular reabsorption after injection of mfsd12a translation-blocking morpholino (TB MO) in a ctns-/- zebrafish model. This resulted in decreased cystine levels but caused a concentration-dependent increase in embryo dysmorphism. Furthermore, the mfsd12a TB MO injection did not improve proximal tubular reabsorption or megalin expression. In conclusion, MFSD12 mRNA depletion reduced cystine levels in both tested models without improvement of the proximal tubular function in the ctns-/- zebrafish embryo. In addition, the apparent toxicity of higher mfsd12a TB MO concentrations on the zebrafish development warrants further evaluation.NEW & NOTEWORTHY In this study, we show that MFSD12 depletion with either CRISPR/Cas9-mediated gene editing or a translation-blocking morpholino significantly reduced cystine levels in cystinosis ciPTECs and ctns-/- zebrafish embryos, respectively. However, we observed no improvement in the proximal tubular reabsorption of dextran in the ctns-/- zebrafish embryos injected with mfsd12a translation-blocking morpholino. Furthermore, a negative effect of the mfsd12a morpholino on the zebrafish development warrants further investigation.


Subject(s)
Cystine , Cystinosis , Disease Models, Animal , Kidney Tubules, Proximal , Zebrafish , Animals , Zebrafish/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Cystinosis/metabolism , Cystinosis/genetics , Cystinosis/pathology , Humans , Cystine/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Epithelial Cells/metabolism , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , CRISPR-Cas Systems
13.
Ren Fail ; 46(1): 2283587, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38374684

ABSTRACT

Background: Light-chain proximal tubulopathy (LCPT) is a rare disease characterized by the accumulation of monoclonal light chains within proximal tubular cells. This study aimed to investigate the clinical characteristics of LCPT from a single Chinese nephrology referral center.Methods: Patients with kidney biopsy-proven isolated LCPT between 2016 and 2022 at Peking University First Hospital were retrospectively included. Clinical data, kidney pathological type, treatment, and prognosis were analyzed.Results: Nineteen patients were enrolled, the mean age at diagnosis was 57 ± 11 and the sex ratio was 6/13 (female/male). Mean proteinuria was 2.44 ± 1.89 g/24 hr and the mean estimated glomerular filtration rate (eGFR) at the point of biopsy was 59.640 ± 27.449 ml/min/1.73 m2. κ-restriction (84%) was dominant among LCPTs. An abnormal free light chain ratio was observed in 86% of the patients. Proximal tubulopathy with cytoplasmic inclusions accounted for the majority (53%), followed by tubulopathy associated with interstitial inflammation reaction (26%), proximal tubulopathy without cytoplasmic inclusions (16%), and proximal tubulopathy with lysosomal indigestion/constipation (5%). One patient presented with acute kidney injury and 16 patients presented with chronic kidney disease. Regarding follow-up, patients received bortezomib-based or R-CHOP chemotherapy or supportive treatment only. The mean follow-up time was 22 ± 16 months, and the mean eGFR was 63.098 ± 27.439 ml/min/1.73 m2 at the end of follow-up. These patients showed improved or stable kidney function.Conclusions: This is the first case series report of LCPT in four different pathological types in northern China. Clone-targeted chemotherapy may help preserve the kidney function in these patients.


Subject(s)
Kidney Diseases , Nephrology , Renal Insufficiency, Chronic , Humans , Male , Female , Retrospective Studies , Kidney Tubules, Proximal/pathology , Kidney Diseases/pathology , Kidney/pathology , Renal Insufficiency, Chronic/complications
14.
Apoptosis ; 29(5-6): 620-634, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38281282

ABSTRACT

Maleic acid (MA) induces renal tubular cell dysfunction directed to acute kidney injury (AKI). AKI is an increasing global health burden due to its association with mortality and morbidity. However, targeted therapy for AKI is lacking. Previously, we determined mitochondrial-associated proteins are MA-induced AKI affinity proteins. We hypothesized that mitochondrial dysfunction in tubular epithelial cells plays a critical role in AKI. In vivo and in vitro systems have been used to test this hypothesis. For the in vivo model, C57BL/6 mice were intraperitoneally injected with 400 mg/kg body weight MA. For the in vitro model, HK-2 human proximal tubular epithelial cells were treated with 2 mM or 5 mM MA for 24 h. AKI can be induced by administration of MA. In the mice injected with MA, the levels of blood urea nitrogen (BUN) and creatinine in the sera were significantly increased (p < 0.005). From the pathological analysis, MA-induced AKI aggravated renal tubular injuries, increased kidney injury molecule-1 (KIM-1) expression and caused renal tubular cell apoptosis. At the cellular level, mitochondrial dysfunction was found with increasing mitochondrial reactive oxygen species (ROS) (p < 0.001), uncoupled mitochondrial respiration with decreasing electron transfer system activity (p < 0.001), and decreasing ATP production (p < 0.05). Under transmission electron microscope (TEM) examination, the cristae formation of mitochondria was defective in MA-induced AKI. To unveil the potential target in mitochondria, gene expression analysis revealed a significantly lower level of ATPase6 (p < 0.001). Renal mitochondrial protein levels of ATP subunits 5A1 and 5C1 (p < 0.05) were significantly decreased, as confirmed by protein analysis. Our study demonstrated that dysfunction of mitochondria resulting from altered expression of ATP synthase in renal tubular cells is associated with MA-induced AKI. This finding provides a potential novel target to develop new strategies for better prevention and treatment of MA-induced AKI.


Subject(s)
Acute Kidney Injury , Apoptosis , Maleates , Mice, Inbred C57BL , Mitochondria , Mitochondrial Proton-Translocating ATPases , Animals , Humans , Male , Mice , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Apoptosis/drug effects , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Proton-Translocating ATPases/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Reactive Oxygen Species/metabolism
15.
Kidney Int ; 105(2): 312-327, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37977366

ABSTRACT

Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part, this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Identifying the molecular and genetic regulators unique to nephron segments that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. Here, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI and found them upregulated after severe AKI and correlated with chronic injury. Surprisingly, proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to pre-conditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of proximal tubule cells in the S3 segment that displayed features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic pre-conditioning, and female sex. Thus, our results identified a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both the injury response and protection from ischemic AKI.


Subject(s)
Acute Kidney Injury , Kidney Tubules, Proximal , PAX2 Transcription Factor , PAX8 Transcription Factor , Renal Insufficiency, Chronic , Animals , Female , Mice , Acute Kidney Injury/complications , Acute Kidney Injury/genetics , Ischemia/complications , Kidney Tubules, Proximal/pathology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/genetics , Reperfusion Injury/genetics , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism
16.
Nat Commun ; 14(1): 8159, 2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38071212

ABSTRACT

Autosomal Recessive Renal Tubular Dysgenesis (AR-RTD) is a fatal genetic disorder characterized by complete absence or severe depletion of proximal tubules (PT) in patients harboring pathogenic variants in genes involved in the Renin-Angiotensin-Aldosterone System. To uncover the pathomechanism of AR-RTD, differentiation of ACE-/- and AGTR1-/- induced pluripotent stem cells (iPSCs) and AR-RTD patient-derived iPSCs into kidney organoids is leveraged. Comprehensive marker analyses show that both mutant and control organoids generate indistinguishable PT in vitro under normoxic (21% O2) or hypoxic (2% O2) conditions. Fully differentiated (d24) AGTR1-/- and control organoids transplanted under the kidney capsule of immunodeficient mice engraft and mature well, as do renal vesicle stage (d14) control organoids. By contrast, d14 AGTR1-/- organoids fail to engraft due to insufficient pro-angiogenic VEGF-A expression. Notably, growth under hypoxic conditions induces VEGF-A expression and rescues engraftment of AGTR1-/- organoids at d14, as does ectopic expression of VEGF-A. We propose that PT dysgenesis in AR-RTD is primarily a non-autonomous consequence of delayed angiogenesis, starving PT at a critical time in their development.


Subject(s)
Angiogenesis , Renin-Angiotensin System , Humans , Animals , Mice , Renin-Angiotensin System/genetics , Vascular Endothelial Growth Factor A , Kidney Tubules, Proximal/pathology , Organoids
17.
Tissue Cell ; 84: 102188, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37567074

ABSTRACT

Diclofenac, a non-steroidal anti-inflammatory drug, reportedly targets mitochondria and induces nephrotoxicity via reactive oxygen species. However, there are few detailed reports of pathological analyses of mitochondria and the factors that cause acute kidney injury (AKI) as a result of nephrotoxicity. In this study, we investigated mitochondrial damage in the proximal tubule in AKI mice at 6, 12, and 24 h after administration of diclofenac. Statistical analysis of immunohistochemistry results confirmed that expression of p62 and LC3, which is associated with autophagy, reached a maximum level in the degenerated proximal renal tubule 12 h after diclofenac treatment, with high autophagy activity. Electron microscopy images provided clear evidence that confirmed mitochondrial degeneration and injury as well as autophagy (mitophagy) in mitochondria treated with diclofenac. The purpose of this study was to pathologically characterize both mitochondrial damage in the proximal renal tubules induced by diclofenac and the course of mitophagy to remove the damaged mitochondria. This report provides important information regarding mitochondrial damage in the proximal tubules in diclofenac-induced nephropathy.


Subject(s)
Acute Kidney Injury , Kidney Tubules, Proximal , Mice , Animals , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Diclofenac/toxicity , Diclofenac/metabolism , Acute Kidney Injury/chemically induced , Mitochondria/metabolism , Autophagy
18.
Kidney Int ; 104(4): 754-768, 2023 10.
Article in English | MEDLINE | ID: mdl-37406929

ABSTRACT

Proteinuria is a prominent feature of chronic kidney disease. Interventions that reduce proteinuria slow the progression of chronic kidney disease and the associated risk of cardiovascular disease. Here, we propose a mechanistic coupling between proteinuria and proprotein convertase subtilisin/kexin type 9 (PCSK9), a regulator of cholesterol and a therapeutic target in cardiovascular disease. PCSK9 undergoes glomerular filtration and is captured by megalin, the receptor responsible for driving protein reabsorption in the proximal tubule. Accordingly, megalin-deficient mice and patients carrying megalin pathogenic variants (Donnai Barrow syndrome) were characterized by elevated urinary PCSK9 excretion. Interestingly, PCSK9 knockout mice displayed increased kidney megalin while PCSK9 overexpression resulted in its reduction. Furthermore, PCSK9 promoted trafficking of megalin to lysosomes in cultured proximal tubule cells, suggesting that PCSK9 is a negative regulator of megalin. This effect can be accelerated under disease conditions since either genetic destruction of the glomerular filtration barrier in podocin knockout mice or minimal change disease (a common cause of nephrotic syndrome) in patients resulted in enhanced tubular PCSK9 uptake and urinary PCSK9 excretion. Pharmacological PCSK9 inhibition increased kidney megalin while reducing urinary albumin excretion in nephrotic mice. Thus, glomerular damage increases filtration of PCSK9 and concomitantly megalin degradation, resulting in escalated proteinuria.


Subject(s)
Cardiovascular Diseases , Nephrotic Syndrome , Renal Insufficiency, Chronic , Humans , Mice , Animals , Nephrotic Syndrome/pathology , Proprotein Convertase 9/metabolism , Low Density Lipoprotein Receptor-Related Protein-2 , Cardiovascular Diseases/metabolism , Proteinuria/genetics , Kidney Tubules, Proximal/pathology , Renal Insufficiency, Chronic/pathology , Mice, Knockout , Subtilisins/metabolism
19.
BMC Nephrol ; 24(1): 180, 2023 06 19.
Article in English | MEDLINE | ID: mdl-37337149

ABSTRACT

Diabetic kidney disease (DKD) is the most common complication of diabetes mellitus and a leading cause of kidney failure worldwide. Despite its prevalence, the mechanisms underlying early kidney damage in DKD remain poorly understood. In this study, we used single nucleus RNA-seq to construct gene regulatory networks (GRNs) in the kidney cortex of patients with early DKD. By comparing these networks with those of healthy controls, we identify cell type-specific changes in genetic regulation associated with diabetic status. The regulon activities of FXR (NR1H4) and CREB5 were found to be upregulated in kidney proximal convoluted tubule epithelial cells (PCTs), which were validated using immunofluorescence staining in kidney biopsies from DKD patients. In vitro experiments using cultured HK2 cells showed that FXR and CREB5 protected cells from apoptosis and epithelial-mesenchymal transition. Our findings suggest that FXR and CREB5 may be promising targets for early intervention in patients with DKD.


Subject(s)
Cyclic AMP Response Element-Binding Protein A , Diabetes Mellitus , Diabetic Nephropathies , Receptors, Cytoplasmic and Nuclear , Humans , Cyclic AMP Response Element-Binding Protein A/genetics , Diabetes Mellitus/genetics , Diabetic Nephropathies/pathology , Gene Expression Regulation , Kidney/pathology , Kidney Tubules, Proximal/pathology , Regulon , Receptors, Cytoplasmic and Nuclear/genetics
20.
Sci Rep ; 13(1): 4025, 2023 03 10.
Article in English | MEDLINE | ID: mdl-36899130

ABSTRACT

Acute kidney injury (AKI) relates to an abrupt reduction in renal function resulting from numerous conditions. Morbidity, mortality, and treatment costs related to AKI are relatively high. This condition is strongly associated with damage to proximal tubule cells (PTCs), generating distinct patterns of transcriptional and epigenetic alterations that result in structural changes in the nuclei of this epithelium. To this date, AKI-related nuclear chromatin redistribution in PTCs is poorly understood, and it is unclear whether changes in PTC chromatin patterns can be detected using conventional microscopy during mild AKI, which can progress to more debilitating forms of injury. In recent years, gray level co-occurrence matrix (GLCM) analysis and discrete wavelet transform (DWT) have emerged as potentially valuable methods for identifying discrete structural changes in nuclear chromatin architecture that are not visible during the conventional histopathological exam. Here we present findings indicating that GLCM and DWT methods can be successfully used in nephrology to detect subtle nuclear morphological alterations associated with mild tissue injury demonstrated in rodents by inducing a mild form of AKI through ischemia-reperfusion injury. Our results show that mild ischemic AKI is associated with the reduction of local textural homogeneity of PTC nuclei quantified by GLCM and the increase of nuclear structural heterogeneity indirectly assessed with DWT energy coefficients. This rodent model allowed us to show that mild ischemic AKI is associated with the significant reduction of textural homogeneity of PTC nuclei, indirectly assessed by GLCM indicators and DWT energy coefficients.


Subject(s)
Acute Kidney Injury , Cell Nucleus , Kidney Tubules, Proximal , Acute Kidney Injury/pathology , Male , Animals , Rats , Rats, Sprague-Dawley , Wavelet Analysis , Kidney Tubules, Proximal/pathology , Cell Nucleus/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...