Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.013
Filter
1.
Genes (Basel) ; 15(9)2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39336766

ABSTRACT

Dent disease type 1 is a rare X-linked recessive inherited renal disorder affecting mainly young males, generally leading to end-stage renal failure and for which there is no cure. It is caused by inactivating mutations in the gene encoding ClC-5, a 2Cl-/H+ exchanger found on endosomes in the renal proximal tubule. This transporter participates in reabsorbing all filtered plasma proteins, which justifies why proteinuria is commonly observed when ClC-5 is defective. In the context of Dent disease type 1, a proximal tubule dedifferentiation was shown to be accompanied by a dysfunctional cell metabolism. However, the exact mechanisms linking such alterations to chronic kidney disease are still unclear. In this review, we gather knowledge from several Dent disease type 1 models to summarize the current hypotheses generated to understand the progression of this disorder. We also highlight some urinary biomarkers for Dent disease type 1 suggested in different studies.


Subject(s)
Chloride Channels , Kidney Tubules, Proximal , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Chloride Channels/genetics , Chloride Channels/metabolism , Animals , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Genetic Diseases, X-Linked/physiopathology , Mutation , Biomarkers/urine , Nephrolithiasis
2.
Cell Mol Life Sci ; 81(1): 385, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39235496

ABSTRACT

Cisplatin-induced renal tubular injury largely restricts the wide-spread usage of cisplatin in the treatment of malignancies. Identifying the key signaling pathways that regulate cisplatin-induced renal tubular injury is thus clinically important. PARVB, a focal adhesion protein, plays a crucial role in tumorigenesis. However, the function of PARVB in kidney disease is largely unknown. To investigate whether and how PARVB contributes to cisplatin-induced renal tubular injury, a mouse model (PARVB cKO) was generated in which PARVB gene was specifically deleted from proximal tubular epithelial cells using the Cre-LoxP system. In this study, we found depletion of PARVB in proximal tubular epithelial cells significantly attenuates cisplatin-induced renal tubular injury, including tubular cell death and inflammation. Mechanistically, PARVB associates with transforming growth factor-ß-activated kinase 1 (TAK1), a central regulator of cell survival and inflammation that is critically involved in mediating cisplatin-induced renal tubular injury. Depletion of PARVB promotes cisplatin-induced TAK1 degradation, inhibits TAK1 downstream signaling, and ultimately alleviates cisplatin-induced tubular cell damage. Restoration of PARVB or TAK1 in PARVB-deficient cells aggravates cisplatin-induced tubular cell injury. Finally, we demonstrated that PARVB regulates TAK1 protein expression through an E3 ligase ITCH-dependent pathway. PARVB prevents ITCH association with TAK1 to block its ubiquitination. Our study reveals that PARVB deficiency protects against cisplatin-induced tubular injury through regulation of TAK1 signaling and indicates targeting this pathway may provide a novel therapeutic strategy to alleviate cisplatin-induced kidney damage.


Subject(s)
Cisplatin , MAP Kinase Kinase Kinases , Mice, Knockout , Signal Transduction , Cisplatin/adverse effects , Cisplatin/toxicity , Animals , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Signal Transduction/drug effects , Mice , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/drug effects , Humans , Mice, Inbred C57BL , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/adverse effects , Kidney Tubules/pathology , Kidney Tubules/metabolism , Kidney Tubules/drug effects , Adaptor Proteins, Signal Transducing
3.
Nat Commun ; 15(1): 7010, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237549

ABSTRACT

Kidney injury disrupts the intricate renal architecture and triggers limited regeneration, together with injury-invoked inflammation and fibrosis. Deciphering the molecular pathways and cellular interactions driving these processes is challenging due to the complex tissue structure. Here, we apply single cell spatial transcriptomics to examine ischemia-reperfusion injury in the mouse kidney. Spatial transcriptomics reveals injury-specific and spatially-dependent gene expression patterns in distinct cellular microenvironments within the kidney and predicts Clcf1-Crfl1 in a molecular interplay between persistently injured proximal tubule cells and their neighboring fibroblasts. Immune cell types play a critical role in organ repair. Spatial analysis identifies cellular microenvironments resembling early tertiary lymphoid structures and associated molecular pathways. Collectively, this study supports a focus on molecular interactions in cellular microenvironments to enhance understanding of injury, repair and disease.


Subject(s)
Cell Communication , Cellular Microenvironment , Kidney , Regeneration , Reperfusion Injury , Transcriptome , Animals , Mice , Regeneration/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Fibroblasts/metabolism , Gene Expression Profiling , Single-Cell Analysis , Fibrosis
4.
Biol Pharm Bull ; 47(9): 1557-1564, 2024.
Article in English | MEDLINE | ID: mdl-39313392

ABSTRACT

Oxidative stress plays a crucial role in the development and progression of various kidney diseases. Nuclear factor erythroid 2-related factor 2 (NRF2) is the primary transcription factor that protects cells from oxidative stress by regulating cytoprotective genes including those involved in the antioxidant glutathione (GSH) pathway. GSH maintains cellular redox status and affects redox signaling, cell proliferation, and cell death. Antimycin A, an inhibitor of complex III of the electron transport chain, causes oxidative stress and reduces GSH levels. In this study, we induced mitochondrial damage in rat renal proximal tubular cells using antimycin A and investigated cellular viability and levels of NRF2 and GSH. Treatment with antimycin A altered the expression of antioxidant genes, including reduction in the transcription of glutathione-cysteine ligase subunits (Gclc and Gclm) and glutathione reductase (Gsr1), followed by a reduction in total GSH content with a concomitant decrease in NRF2 protein expression. AR-20007, previously described as an NRF2 activator, stabilizes and increases NRF2 protein expression in cells. By stimulating NRF2, AR-20007 increased the expression of antioxidant and detoxifying enzymes, thereby enhancing protection against oxidative stress induced by antimycin A. These data suggest that NRF2 activation effectively inhibits antimycin A-induced oxidative stress and that NRF2 may be a promising therapeutic target for preventing cell death during acute kidney injury.


Subject(s)
Antimycin A , Epithelial Cells , Glutathione , NF-E2-Related Factor 2 , Oxidative Stress , Antimycin A/pharmacology , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Glutathione/metabolism , Rats , Oxidative Stress/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Cell Death/drug effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Cell Line , Cell Survival/drug effects , Antioxidants/pharmacology , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/metabolism
5.
Kidney Int ; 106(4): 560-562, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39304269

ABSTRACT

Acute kidney injury is a devasting clinical syndrome resulting from multiple causes, characterized by an abrupt deterioration of kidney function for which there is no pharmacologic treatment. Cilastatin has demonstrated direct nephroprotective effects in acute kidney injury and now is shown to be effective to specifically target therapeutically loaded nanoparticles to the proximal tubule to treat acute kidney injury.


Subject(s)
Acute Kidney Injury , Cilastatin , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Humans , Cilastatin/therapeutic use , Cilastatin/administration & dosage , Animals , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/metabolism , Nanoparticles
6.
J Biochem Mol Toxicol ; 38(10): e23854, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39287333

ABSTRACT

Ischemia-reperfusion (I-R) injury is the most common cause of acute kidney injury. In experiments involving primary human renal proximal tubular epithelial cells (RPTECs) exposed to anoxia-reoxygenation, we explored the hypothesis that mitochondrial malate dehydrogenase-2 (MDH-2) inhibition redirects malate metabolism from the mitochondria to the cytoplasm, towards the malate-pyruvate cycle and reversed malate-aspartate shuttle. Colorimetry, fluorometry, and western blotting showed that MDH2 inhibition accelerates the malate-pyruvate cycle enhancing cytoplasmic NADPH, thereby regenerating the potent antioxidant reduced glutathione. It also reversed the malate-aspartate shuttle and potentially diminished mitochondrial reactive oxygen species (ROS) production by transferring electrons, in the form of NADH, from the mitochondria to the cytoplasm. The excessive ROS production induced by anoxia-reoxygenation led to DNA damage and protein modification, triggering DNA damage and unfolded protein response, ultimately resulting in apoptosis and senescence. Additionally, ROS induced lipid peroxidation, which may contribute to the process of ferroptosis. Inhibiting MDH-2 proved effective in mitigating ROS overproduction during anoxia-reoxygenation, thereby rescuing RPTECs from death or senescence. Thus, targeting MDH-2 holds promise as a pharmaceutical strategy against I-R injury.


Subject(s)
Epithelial Cells , Malate Dehydrogenase , Reactive Oxygen Species , Reactive Oxygen Species/metabolism , Humans , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/drug effects , Malate Dehydrogenase/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/cytology , Cell Hypoxia/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , DNA Damage , Apoptosis/drug effects
7.
Mol Biol Rep ; 51(1): 974, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259342

ABSTRACT

BACKGROUND: One of the causes of tubulointerstitial nephritis is viral infection, with innate immune responses affecting its pathogenesis. Toll-like receptor 3 (TLR3) recognizes viral infections and acts antivirally by activating signaling to produce inflammatory cytokines/chemokines, including C-C motif chemokine ligand 5 (CCL5) and interferon-ß (IFN-ß). Although cylindromatosis lysine 63 deubiquitinase (CYLD) is known to be associated with tubulointerstitial nephritis and renal function, its role in the antiviral innate immune response in tubular epithelial cells remains unknown. In this study, we investigated the association between CYLD and TLR3-mediated CCL5 production in cultured human renal proximal tubular epithelial cells (hRPTECs). METHODS AND RESULTS: Polyinosinic-polycytidylic acid (poly IC), a synthetic TLR3 ligand, was used to stimulate hRPTECs. mRNA expression was measured using reverse transcription-quantitative polymerase chain reaction. Protein expression was assayed using western blotting or an enzyme-linked immunosorbent assay. Knockdown of IFN-ß, nuclear factor-kappa B (NF-κB) p65, and CYLD was performed by transfecting cells with specific small interfering RNAs. The intracellular localization of CYLD in hRPTECs was analyzed using immunofluorescence. Poly IC induced CCL5 expression in a time- and concentration-dependent manner, and knockdown of either IFN-ß or p65 reduced poly IC-induced CCL5 expression. CYLD knockdown increased the poly IC-induced CCL5, phosphorylated IκB kinase α/ß (IKK complex), and phosphorylated p65 expression. The CYLD protein was localized in the cytoplasm, and poly IC did not alter its expression. CONCLUSION: CYLD may prevent excessive inflammation due to an antiviral innate immune response by suppressing IKK complex and NF-κB activation downstream of TLR3 in hRPTECs.


Subject(s)
Chemokine CCL5 , Deubiquitinating Enzyme CYLD , Epithelial Cells , Kidney Tubules, Proximal , Poly I-C , Toll-Like Receptor 3 , Humans , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/genetics , Deubiquitinating Enzyme CYLD/metabolism , Deubiquitinating Enzyme CYLD/genetics , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , Kidney Tubules, Proximal/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Poly I-C/pharmacology , Interferon-beta/metabolism , Interferon-beta/genetics , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Immunity, Innate , NF-kappa B/metabolism , Cell Line
8.
Nat Commun ; 15(1): 8038, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271683

ABSTRACT

Diabetic kidney disease (DKD) is the main cause of chronic kidney disease worldwide. While injury to the podocytes, visceral epithelial cells that comprise the glomerular filtration barrier, drives albuminuria, proximal tubule (PT) dysfunction is the critical mediator of DKD progression. Here, we report that the podocyte-specific induction of human KLF6, a zinc-finger binding transcription factor, attenuates podocyte loss, PT dysfunction, and eventual interstitial fibrosis in a male murine model of DKD. Utilizing combination of snRNA-seq, snATAC-seq, and tandem mass spectrometry, we demonstrate that podocyte-specific KLF6 triggers the release of secretory ApoJ to activate calcium/calmodulin dependent protein kinase 1D (CaMK1D) signaling in neighboring PT cells. CaMK1D is enriched in the first segment of the PT, proximal to the podocytes, and is critical to attenuating mitochondrial fission and restoring mitochondrial function under diabetic conditions. Targeting podocyte-PT signaling by enhancing ApoJ-CaMK1D might be a key therapeutic strategy in attenuating the progression of DKD.


Subject(s)
Diabetic Nephropathies , Kidney Tubules, Proximal , Kruppel-Like Factor 6 , Podocytes , Signal Transduction , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/genetics , Podocytes/metabolism , Podocytes/pathology , Animals , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Humans , Mice , Kruppel-Like Factor 6/metabolism , Kruppel-Like Factor 6/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Mice, Inbred C57BL , Disease Models, Animal
9.
Exp Cell Res ; 442(1): 114186, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39098465

ABSTRACT

TGFß1 is a powerful regulator of fibrosis; secreted in a latent form, it becomes active after release from the latent complex. During tissue fibrosis, the EDA + isoform of cellular fibronectin is overexpressed. In pulmonary fibrosis it has been proposed that the fibronectin splice variant including an EDA domain (FN EDA+) activates latent TGFß. Our work investigates the potential of blocking the 'splicing in' of EDA with antisense oligonucleotides to inhibit TGFß1-induced EDA + fibronectin and to prevent the cascade of events initiated by TGFß1 in human renal proximal tubule cells (PTEC). Human primary PTEC were treated with TGFß1 for 48 h, medium removed and the cells transfected with RNase H-independent antisense oligonucleotides (ASO) designed to block EDA exon inclusion (ASO5). The efficacy of ASO to block EDA exon inclusion was assessed by EDA + fibronectin RNA and protein expression; the expression of TGFß, αSMA (α smooth muscle actin), MMP2 (matrix metalloproteinse-2), MMP9 (matrix metalloproteinse-9), Collagen I, K Cadherin and connexin 43 was analysed. Targeting antisense oligonucleotides designed to block EDA exon inclusion in fibronectin pre mRNA were effective in reducing the amount of TGFß1 -induced cellular EDA + fibronectin RNA and secreted EDA + fibronectin protein (assessed by western immunoblotting and immunocytochemistry) in human proximal tubule cells in an in vitro cell culture model. The effect was selective for EDA + exon with no effect on EDB + fibronectin RNA and total fibronectin mRNA. Exogenous TGFß1 induced endogenous TGFß, αSMA, MMP2, MMP9 and Col I mRNA. TGFß1 treatment for 48h reduced the expression of K-Cadherin and increased the expression of connexin-43. These TGFß1-induced pro-fibrotic changes were attenuated by ASO5 treatment. 48 h after the removal of exogenous TGFß, further increases in αSMA, MMP2, MMP9 was observed; ASO5 significantly inhibited this subsequent increase. ASO5 treatment also significantly inhibited ability of the cell culture medium harvested at the end of the experiment (96h) to stimulate SMAD3 reporter cells. The role of endogenous TGFß1 was confirmed by the use of a TGFß receptor inhibitor. Our results demonstrate a critical role of FN EDA+ in a cycle of TGFß driven pro-fibrotic responses in human PTEC and blocking its production with ASO technology offers a potential therapy to interrupt this vicious circle and hence limit the progression of renal fibrosis.


Subject(s)
Alternative Splicing , Epithelial Cells , Fibronectins , Fibrosis , Kidney Tubules, Proximal , Oligonucleotides, Antisense , Transforming Growth Factor beta1 , Humans , Fibronectins/metabolism , Fibronectins/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/cytology , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/genetics , Fibrosis/metabolism , Alternative Splicing/genetics , Transforming Growth Factor beta1/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/drug effects , Cells, Cultured , Autocrine Communication , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics
11.
Eur J Pharmacol ; 982: 176931, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39182553

ABSTRACT

Renal fibrosis is among the major factors contributing to the development of chronic kidney disease. In this regard, although N6-methyladenosine (m6A) modification and micro-RNAs (miRNAs) have been established to play key roles in diverse physiological processes and disease/disorder development, further research is required to identify the probable mechanisms and processes associated with their involvement in renal fibrosis. In this study, we show that transforming growth factor ß1 (TGF-ß1)-induced human proximal tubule epithelial cells (HK2) are characterized by dose-dependently higher methyltransferase-like 3 (METTL3) expression. Furthermore, METTL3 was found to enhance pri-miR-199a-3p maturation and miR-199a-3p expression in an m6A-dependent manner, whereas miR-199a-3p sponges prostate apoptotic response 4 (Par4), thereby regulating its expression. Collectively, our findings in this study indicate that the METTL3/miR-199a-3p/Par4 axis plays a key role in the development of obstructive nephrogenic fibrosis.


Subject(s)
Fibrosis , Methyltransferases , MicroRNAs , Transforming Growth Factor beta1 , Animals , Humans , Adenosine/analogs & derivatives , Adenosine/metabolism , Cell Line , Gene Expression Regulation , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/metabolism , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Male , Mice
12.
FASEB J ; 38(16): e23884, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39135512

ABSTRACT

The inhibition of the autophagolysosomal pathway mediated by transcription factor EB (TFEB) inactivation in proximal tubular epithelial cells (TECs) is a key mechanism of TEC injury in diabetic kidney disease (DKD). Acetylation is a novel mechanism that regulates TFEB activity. However, there are currently no studies on whether the adjustment of the acetylation level of TFEB can reduce the damage of diabetic TECs. In this study, we investigated the effect of Trichostatin A (TSA), a typical deacetylase inhibitor, on TFEB activity and damage to TECs in both in vivo and in vitro models of DKD. Here, we show that TSA treatment can alleviate the pathological damage of glomeruli and renal tubules and delay the DKD progression in db/db mice, which is associated with the increased expression of TFEB and its downstream genes. In vitro studies further confirmed that TSA treatment can upregulate the acetylation level of TFEB, promote its nuclear translocation, and activate the expression of its downstream genes, thereby reducing the apoptosis level of TECs. TFEB deletion or HDAC6 knockdown in TECs can counteract the activation effect of TSA on autophagolysosomal pathway. We also found that TFEB enhances the transcription of Tfeb through binding to its promoter and promotes its own expression. Our results, thus, provide a novel therapeutic mechanism for DKD that the alleviation of TEC damage by activating the autophagic lysosomal pathway through upregulating TFEB acetylation can, thus, delay DKD progression.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Diabetic Nephropathies , Epithelial Cells , Histone Deacetylase Inhibitors , Hydroxamic Acids , Kidney Tubules, Proximal , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Diabetic Nephropathies/metabolism , Mice , Acetylation , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Hydroxamic Acids/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Male , Mice, Inbred C57BL , Autophagy/drug effects , Apoptosis/drug effects
13.
J Biochem Mol Toxicol ; 38(9): e23777, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39165170

ABSTRACT

Tramadol (TR), a commonly prescribed pain reliever for moderate to severe pain, has been associated with kidney damage. This study investigates TR-induced nephrotoxicity mechanisms, focusing on its effects on renal proximal tubular cells (PTCs). The study findings demonstrate that TR disrupts PTC bioenergetic processes, leading to oxidative stress and inflammation. Significant toxicity to PTCs was observed with estimated effective concentration 50 values of 9.8 and 11.5 µM based on 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase assays, respectively. TR also interferes with the function of PTC transporters, including organic cation uptake transporter 1, organic cation transporter 2, P-glycoprotein, and multidrug resistance protein 2. Furthermore, bioenergetics assays showed that TR reduced the activities of mitochondrial complexes I and III, adenosine triphosphate production, mitochondrial membrane potential, and oxygen consumption rate while increasing lactate release. TR also increased the production of reactive oxygen species, lipid peroxidation thiobarbituric acid reactive substances end products, and the expression of the NRf2 gene while decreasing reduced glutathione (GSH-R) stores and catalase and superoxide dismutase antioxidant activities. Additionally, TR increased the production of inflammatory cytokines (TNF-α and IL-6) and their coding genes expression. Interestingly, the study found that antioxidants like GSH-R, inhibitors of IL-6 and TNF-α, and mitochondrial activating Co-Q10 could protect cells against TR-induced cytotoxicity. The study suggests that TR causes nephrotoxicity by disrupting bioenergetic processes, causing oxidative stress and inflammation, but antioxidants and agents targeting mitochondria may have protective and curative potential.


Subject(s)
Energy Metabolism , Inflammation , Oxidative Stress , Tramadol , Oxidative Stress/drug effects , Inflammation/chemically induced , Inflammation/metabolism , Tramadol/adverse effects , Tramadol/pharmacology , Energy Metabolism/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Humans , Cell Line , Animals , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology
14.
Anat Histol Embryol ; 53(5): e13103, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39155839

ABSTRACT

Dromedary camels can survive and reproduce in desert areas. The unique anatomical structure of the kidney enables the camel to prevent water loss. The present study aimed to investigate the ultrastructure of the peroxisomes in the normal kidney of the adult dromedary camel. Tissue samples were taken from the cortex and outer medulla of the kidney of eight camels. The samples were then processed for histological and ultrastructural investigations. The epithelial cells of the proximal tubules displayed peroxisomes with varying sizes and shapes. The peroxisomes were observed in either dispersed or clustered arrangement. Each peroxisome exhibited a homogenous matrix enveloped by a single membrane. Several peroxisomes exhibited one or more dark marginal plates that were always strongly associated with the smooth endoplasmic reticulum. The intensity of the peroxisomal matrix differed significantly, either within the same cell or across different cells. The intensity was light or dark, with a few peroxisomes presenting a similar intensity to that of the mitochondria. Some peroxisomes contained nucleoids within their matrix. The peroxisomes in the first and second sections of proximal convoluted tubules were scattered and primarily located in the region between the microvilli and the underlying mitochondria. The peroxisomes in the third region were abundant and frequently aggregated in clusters throughout the cytoplasm. In the fourth region, the number of peroxisomes was low. The proximal straight tubule had a limited quantity of peroxisomes. In conclusion, peroxisomes in the proximal tubule in kidney of normal dromedary camel were similar in shape and size to other mammals; however, heterogeneity exists as a result of differences in species-specific peroxisomal proteins. Peroxisomes are suggested to be a major source of metabolic energy and act as hydrogen peroxide (H2O2) scavengers, resulting in the release of water and oxygen.


Subject(s)
Camelus , Kidney , Peroxisomes , Animals , Camelus/anatomy & histology , Camelus/physiology , Peroxisomes/ultrastructure , Peroxisomes/metabolism , Kidney/ultrastructure , Kidney/anatomy & histology , Microscopy, Electron, Transmission/veterinary , Male , Kidney Tubules, Proximal/ultrastructure , Kidney Tubules, Proximal/anatomy & histology
15.
Sci Rep ; 14(1): 19443, 2024 08 21.
Article in English | MEDLINE | ID: mdl-39169052

ABSTRACT

Cisplatin-induced nephrotoxicity restricts its clinical use against solid tumors. The present study elucidated the pharmacological effects of Renogrit, a plant-derived prescription medicine, using cisplatin-induced human renal proximal tubular (HK-2) cells and Caenorhabditis elegans. Quantification of phytochemicals in Renogrit was performed on HPTLC and UHPLC platforms. Renogrit was assessed in vitro in HK-2 cells post-exposure to clinically relevant concentration of cisplatin. It was observed that renoprotective properties of Renogrit against cisplatin-induced injury stem from its ability to regulate renal injury markers (KIM-1, NAG levels; NGAL mRNA expression), redox imbalance (ROS generation; GST levels), and mitochondrial dysfunction (mitochondrial membrane potential; SKN-1, HSP-60 expression). Renogrit was also found to modulate apoptosis (EGL-1 mRNA expression; protein levels of p-ERK, p-JNK, p-p38, c-PARP1), necroptosis (intracellular calcium accumulation; RIPK1, RIPK3, MLKL mRNA expression), mitophagy (lysosome population; mRNA expression of PINK1, PDR1; protein levels of p-PINK1, LC3B), and inflammation (IL-1ß activity; protein levels of LXR-α). More importantly, Renogrit treatment did not hamper normal anti-proliferative effects of cisplatin as observed from cytotoxicity analysis on MCF-7, A549, SiHa, and T24 human cancer cells. Taken together, Renogrit could be a potential clinical candidate to mitigate cisplatin-induced nephrotoxicity without compromising the anti-neoplastic properties of cisplatin.


Subject(s)
Apoptosis , Caenorhabditis elegans , Cisplatin , Mitophagy , Cisplatin/adverse effects , Cisplatin/toxicity , Animals , Humans , Mitophagy/drug effects , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/metabolism , Apoptosis/drug effects , Cell Line , Plant Extracts/pharmacology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Antineoplastic Agents/adverse effects , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology
16.
Sci Total Environ ; 949: 175159, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39094650

ABSTRACT

Bisphenol P (BPP) has been detected in human biological samples; however studies on its nephrotoxicity are scarce. Given the susceptibility of kidneys to endocrine-disrupting chemicals, there is an urgent need to investigate the renal toxicity of BPP. This study aimed to evaluate the effects of different concentrations of BPPs on the kidneys of C57BL/6 mice and elucidate the underlying mechanisms of renal damage using a combination of mouse renal transcriptomic data and human renal proximal tubular epithelial cells (HK-2). Mice were exposed to BPP (0, 0.3, 30, 3000 µg/kg bw/d) via gavage for 5 weeks. Renal injury was assessed based on changes in body and kidney weights, serum renal function indices, and histopathological examination. Transcriptomic analysis identified differentially expressed genes and pathways, whereas cellular assays were used to measure cell viability, reactive oxygen species (ROS), apoptosis, and the expression of key genes and proteins. The results show that BPP exposure induces renal injury, as evidenced by increased body weight, abnormal renal function indices, and renal tissue damage. Transcriptomic analysis revealed alterations in genes and pathways related to oxidative stress, p53 signaling, autophagy, and apoptosis. Cellular experiments confirmed that BPP induces oxidative stress and apoptosis. Furthermore, BPP exposure significantly inhibits autophagy, potentially exacerbating apoptosis and contributing to kidney injury. Treatment with a ROS inhibitor (N-Acetylcysteine, NAC) mitigated BPP-induced autophagy inhibition and apoptosis, implicating oxidative stress as a key factor. BPP exposure may lead to renal injury through excessive ROS accumulation, oxidative stress, inflammatory responses, autophagy inhibition, and increased apoptosis. The effects of NAC highlight the role of oxidative stress in BPP-induced nephrotoxicity. These findings enhance our understanding of BPP-induced nephrotoxicity and underscore the need to control BPP exposure to prevent renal disease. This study emphasized the importance of evaluating the safety of new Bisphenol A analogs, including BPP, in environmental toxicology.


Subject(s)
Epithelial Cells , Mice, Inbred C57BL , Oxidative Stress , Phenols , Animals , Humans , Mice , Apoptosis/drug effects , Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Epithelial Cells/drug effects , Kidney/cytology , Kidney/drug effects , Kidney/pathology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Oxidative Stress/drug effects , Phenols/toxicity , Reactive Oxygen Species/metabolism
17.
Medicine (Baltimore) ; 103(33): e39174, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39151531

ABSTRACT

RATIONALE: Light chain proximal tubulopathy (LCPT) is a rare form of renal impairment associated with multiple myeloma (MM). LCPT is caused by inclusions formed of free light chains that are typically crystalline, but can also be noncrystalline structures. PATIENT CONCERNS: A 62-year-old man was hospitalized for the investigation of abnormal urine test results lasting for 1 year and kidney-function abnormalities persisting for more than 1 month. DIAGNOSES: Noncrystalline LCPT and MM. INTERVENTIONS: The patient was treated with the lenalidomide, bortezomib, and dexamethasone and pomalidomide, bortezomib, and dexamethasone chemotherapy regimens. OUTCOMES: Complete remission of MM was achieved, and the patient's renal function returned to normal. LESSONS: This case report highlights the importance of renal pathology in the diagnosis of patients with unexplained chronic kidney disease and proteinuria.


Subject(s)
Multiple Myeloma , Humans , Male , Middle Aged , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Multiple Myeloma/diagnosis , Multiple Myeloma/pathology , Immunoglobulin Light Chains/urine , Kidney Tubules, Proximal/pathology , Dexamethasone/therapeutic use , Inclusion Bodies/pathology , Thalidomide/therapeutic use , Thalidomide/analogs & derivatives , Lenalidomide/therapeutic use , Lenalidomide/administration & dosage , Bortezomib/therapeutic use
18.
In Vivo ; 38(5): 2107-2114, 2024.
Article in English | MEDLINE | ID: mdl-39187331

ABSTRACT

BACKGROUND/AIM: Angiotensinogen (AGT), a precursor of angiotensin II (AngII), contributes to regulating (patho)physiological conditions, including blood pressure changes, inflammation, and kidney fibrosis. However, the precise role of tissue-specific AGT in kidney fibrosis independent of blood pressure remains to be fully understood. This study investigated the source of intrarenal AGT and its role in kidney injury and fibrosis during obstructive nephropathy. MATERIALS AND METHODS: Proximal tubule- (PT, major source secreting AGT in the kidney; PKO) or liver- (major source of circulating AGT; LKO) AGT knockout (KO) mice were subjected to unilateral ureteral obstruction (UUO), a blood pressure-independent fibrosis model. RESULTS: UUO increased AGT mRNA and protein levels in the kidneys. PKO decreased AGT mRNA, but LKO enhanced it in UUO kidneys compared with the control. In contrast, the intrarenal protein levels of AGT increased in PKO, but not in LKO in UUO kidneys, indicating that the liver is a major source of intrarenal AGT protein. Expression of megalin, a PT receptor involved in the uptake of circulating AGT, was down-regulated in UUO kidneys and was independent of PKO or LKO. However, none of these changes prevented UUO-induced tubular injury and kidney fibrosis. CONCLUSION: Hepatic and proximal tubule AGT play distinct roles in contributing to intrarenal AGT levels during UUO, and their genetic inhibitions fail to prevent kidney injury and fibrosis, suggesting a highly complicated signaling pathway of the renin-angiotensin system and an associated compensatory mechanism in obstructive nephropathy.


Subject(s)
Angiotensinogen , Disease Models, Animal , Fibrosis , Kidney , Mice, Knockout , Ureteral Obstruction , Animals , Mice , Angiotensinogen/metabolism , Angiotensinogen/genetics , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/etiology , Kidney Diseases/pathology , Kidney Diseases/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Liver/metabolism , Liver/pathology , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/complications , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology
19.
Mol Pharm ; 21(9): 4603-4617, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39166754

ABSTRACT

Modulation of the transport-mediated active uptake by human serum albumin (HSA) for highly protein-bound substrates has been reported and improved the in vitro-to-in vivo extrapolation (IVIVE) of hepatic clearance. However, evidence for the relevance of such a phenomenon in the case of renal transporters is sparse. In this study, transport of renal organic anion transporter 1 or 3 (OAT1/3) substrates into conditionally immortalized proximal tubular epithelial cells transduced with OAT1/3 was measured in the presence and absence of 1 and 4% HSA while keeping the unbound substrate concentration constant (based on measured fraction unbound, fu,inc). In the presence of 4% HSA, the unbound intrinsic active uptake clearance (CLint,u,active) of six highly protein-bound substrates increased substantially relative to the HSA-free control (3.5- to 122-fold for the OAT1 CLint,u,active, and up to 28-fold for the OAT3 CLint,u,active). The albumin-mediated uptake effect (fold increase in CLint,u,active) was more pronounced with highly bound substrates compared to no effect seen for weakly protein-bound substrates adefovir (OAT1-specific) and oseltamivir carboxylate (OAT3-specific). The relationship between OAT1/3 CLint,u,active and fu,inc agreed with the facilitated-dissociation model; a relationship was established between the albumin-mediated fold change in CLint,u,active and fu,inc for both the OAT1 and OAT3, with implications for IVIVE modeling. The relative activity factor and the relative expression factor based on global proteomic quantification of in vitro OAT1/3 expression were applied for IVIVE of renal clearance. The inclusion of HSA improved the bottom-up prediction of the level of OAT1/3-mediated secretion and renal clearance (CLsec and CLr), in contrast to the underprediction observed with the control (HSA-free) scenario. For the first time, this study confirmed the presence of the albumin-mediated uptake effect with renal OAT1/3 transporters; the extent of the effect was more pronounced for highly protein-bound substrates. We recommend the inclusion of HSA in routine in vitro OAT1/3 assays due to considerable improvements in the IVIVE of CLsec and CLr.


Subject(s)
Organic Anion Transport Protein 1 , Organic Anion Transporters, Sodium-Independent , Organic Anion Transport Protein 1/metabolism , Humans , Organic Anion Transporters, Sodium-Independent/metabolism , Biological Transport/physiology , Kidney/metabolism , Animals , Kidney Tubules, Proximal/metabolism , Serum Albumin/metabolism , Serum Albumin, Human/metabolism , Cell Line
20.
Am J Physiol Renal Physiol ; 327(4): F610-F622, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39116349

ABSTRACT

Studies in animal models have suggested a linkage between the inflammatory response to injury and subsequent nephron loss during the acute kidney injury (AKI) to chronic kidney disease (CKD) transition. Failure of normal repair during the CKD transition correlates with de novo expression of vascular cell adhesion protein-1 (VCAM-1) by a subset of injured proximal tubule cells. This study identified the role of VCAM-1 expression in promoting the failed repair state. Single-cell transcriptome analysis of patients with AKI and CKD and whole kidney RNA and protein analyses of mouse models of CKD confirmed a marked increase of VCAM-1 expression in the proximal tubules of injured kidneys. In immortalized mouse proximal tubular cells and primary cultured renal cells (PCRCs), VCAM-1 expression was induced by proinflammatory cytokines including tumor necrosis factor (TNF)-α and interleukin (IL)-1ß. Analyses of bulk RNA sequencing of TNF-α-treated primary cultured renal cells or pseudo-bulk RNA sequencing of biopsies from Kidney Precision Medicine Project datasets indicated activation of NF-κB and an enrichment of inflammatory response and cell adhesion pathways in VCAM-1-positive cells. Pharmacological inhibition of NF-κB signaling or genetic deletion of myeloid differentiation factor 88 and TIR domain-containing adapter-inducing interferon-ß suppressed TNF-α- and IL-1ß-induced VCAM-1 expression in vitro. TNF-α stimulation or overexpression of VCAM-1 significantly increased splenocyte adhesion to the mouse proximal tubular monolayer in culture. These results demonstrate that persistence of proinflammatory cytokines after AKI can induce NF-κB-dependent VCAM-1 expression by proximal tubule cells, mediating increased immune cell adhesion to the tubule and thus promoting further tubule injury and greater risk of progression from AKI to CKD.NEW & NOTEWORTHY We demonstrated the induction of VCAM-1 and its biological function in proximal tubules. We found that proinflammatory cytokines (TNF-α and IL-1ß) significantly induced VCAM-1 expression via NF-κB signaling pathway. TNF-α treatment or overexpression of VCAM-1 in immortalized MPT cells increased CD45+ splenocyte adhesion. Pharmacological inhibition of NF-κB or genetic deletion of Vcam1 suppressed TNF-α-induced splenocyte adhesion in vitro, suggesting that VCAM-1 mediates proximal tubular-immune cell cross talk in failed tubule recovery during AKI-to-CKD transition.


Subject(s)
Acute Kidney Injury , Kidney Tubules, Proximal , Renal Insufficiency, Chronic , Vascular Cell Adhesion Molecule-1 , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Animals , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/immunology , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/immunology , Acute Kidney Injury/genetics , Humans , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Disease Models, Animal , Male , Signal Transduction , Mice, Inbred C57BL , NF-kappa B/metabolism , Mice , Disease Progression , Cell Adhesion/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL