Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 432
Filter
1.
J Transl Med ; 22(1): 154, 2024 02 14.
Article in English | MEDLINE | ID: mdl-38355577

ABSTRACT

BACKGROUND: Tumor cells of diffuse-type gastric cancer (DGC) are discohesive and infiltrate into the stroma as single cells or small subgroups, so the stroma significantly impacts DGC progression. Cancer-associated fibroblasts (CAFs) are major components of the tumor stroma. Here, we identified CAF-specific secreted molecules and investigated the mechanism underlying CAF-induced DGC progression. METHODS: We conducted transcriptome analysis for paired normal fibroblast (NF)-CAF isolated from DGC patient tissues and proteomics for conditioned media (CM) of fibroblasts. The effects of fibroblasts on cancer cells were examined by transwell migration and soft agar assays, western blotting, and in vivo. We confirmed the effect of blocking tubulointerstitial nephritis antigen-like 1 (TINAGL1) in CAFs using siRNA or shRNA. We evaluated the expression of TINAGL1 protein in frozen tissues of DGC and paired normal stomach and mRNA in formalin-fixed, paraffin-embedded (FFPE) tissue using RNA in-situ hybridization (RNA-ISH). RESULTS: CAFs more highly expressed TINAGL1 than NFs. The co-culture of CAFs increased migration and tumorigenesis of DGC. Moreover, CAFs enhanced the phosphorylation of focal adhesion kinase (FAK) and mesenchymal marker expression in DGC cells. In an animal study, DGC tumors co-injected with CAFs showed aggressive phenotypes, including lymph node metastasis. However, increased phosphorylation of FAK and migration were reduced by blocking TINAGL1 in CAFs. In the tissues of DGC patients, TINAGL1 was higher in cancer than paired normal tissues and detected with collagen type I alpha 1 chain (COL1A1) in the same spot. Furthermore, high TINAGL1 expression was significantly correlated with poor prognosis in several public databases and our patient cohort diagnosed with DGC. CONCLUSIONS: These results indicate that TINAGL1 secreted by CAFs induces phosphorylation of FAK in DGC cells and promotes tumor progression. Thus, targeting TINAGL1 in CAFs can be a novel therapeutic strategy for DGC.


Subject(s)
Cancer-Associated Fibroblasts , Nephritis, Interstitial , Stomach Neoplasms , Animals , Humans , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Movement/genetics , Fibroblasts/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , RNA, Small Interfering/metabolism , Stomach Neoplasms/pathology , Tumor Microenvironment
3.
Lab Invest ; 103(7): 100131, 2023 07.
Article in English | MEDLINE | ID: mdl-36948295

ABSTRACT

Renal fibrosis is a hallmark of chronic and progressive renal diseases characterized by excessive fibroblast proliferation, extracellular matrix accumulation, and a loss of renal function, eventually leading to end-stage renal diseases. MicroRNA-26a-5p (miR-26a-5p) downregulation has been previously noted in the sera of unilateral ureteral occlusion (UUO)-injured mice, and exosome-mediated miR-26a-5p reportedly attenuated experimental pulmonary and cardiac fibrosis. This study evaluated the expression patterns of miR-26a in a human tissue microarray with kidney fibrosis and in tissues from a mouse model of UUO-induced renal fibrosis. Histologic analyses showed that miR-26a-5p was downregulated in human and mouse tissues with renal interstitial nephritis and fibrosis. Moreover, miR-26a-5p restoration by intravenous injection of a mimic agent prominently suppressed the expression of transforming growth factor ß1 (TGF-ß1) and its cognate receptors, the inflammatory transcription factor NF-κB, epithelial-mesenchymal transition, and inflammatory markers in UUO-injured kidney tissues. In vitro, miR-26a-5p mimic delivery significantly inhibited TGF-ß1-induced activation of cultured normal rat kidney NRK-49F cells, in terms of downregulation of TGF-ß1 receptors, restoration of the epithelial marker E-cadherin, and suppression of mesenchymal markers, including vimentin, fibronectin, and α-smooth muscle actin, as well as TGF-ß1/SMAD3 signaling activity. Our findings identified miR-26a-5p downregulation in kidney tissues with human interstitial nephritis and UUO-induced mouse kidney fibrosis. MiR-26a-5p restoration may exhibit an antifibrotic effect through the blockade of both TGF-ß and NF-κB signaling axes and is considered a novel therapeutic target for treating obstruction-induced renal fibrosis.


Subject(s)
MicroRNAs , Nephritis, Interstitial , Ureteral Obstruction , Animals , Humans , Mice , Rats , Fibrosis , Kidney/metabolism , MicroRNAs/metabolism , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , NF-kappa B/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/complications , Ureteral Obstruction/metabolism
4.
Cells ; 11(13)2022 06 21.
Article in English | MEDLINE | ID: mdl-35805070

ABSTRACT

Canonical transient receptor potential-6 (TRPC6) channels have been implicated in familial and acquired forms of focal and segmental glomerulosclerosis (FSGS), and in renal fibrosis following ureteral obstruction in mice. TRPC6 channels also appear to play a role in driving glomerular disease in aging and in autoimmune glomerulonephritis. In the present study, we examine the role of TRPC6 in the proteinuric state caused by prolonged albumin overload (AO) in Sprague Dawley rats induced by daily injections of exogenous albumin. This was assessed in rats with a global and constitutive inactivation of TRPC6 channels (Trpc6del/del rats) and in wild-type littermates (Trpc6wt/wt rats). AO for 14 and 28 days caused increased urine albumin excretion that was significantly attenuated in Trpc6del/del rats compared to Trpc6wt/wt controls. AO overload did not induce significant glomerulosclerosis or azotemia in either genotype. AO induced mild tubulointerstitial disease characterized by fibrosis, hypercellularity and increased expression of markers of fibrosis and inflammation. Those changes were equally severe in Trpc6wt/wt and Trpc6del/del rats. Immunoblot analysis of renal cortex indicated that AO increased the abundances of TRPC3 and TRPC6, and caused a nearly complete loss of TRPC5 in Trpc6wt/wt rats. The increase in TRPC3 and the loss of TRPC5 occurred to the same extent in Trpc6del/del rats. These data also suggest that TRPC6 plays a role in the normal function of the glomerular filtration barrier. However, whether TRPC6 inactivation protects the tubulointerstitial compartments in Sprague Dawley rats depends on the disease model examined.


Subject(s)
Albuminuria , TRPC Cation Channels/metabolism , Albumins/toxicity , Albuminuria/chemically induced , Albuminuria/metabolism , Albuminuria/physiopathology , Animals , Disease Models, Animal , Glomerular Filtration Barrier , Nephritis, Interstitial/chemically induced , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Rats, Sprague-Dawley , TRPC Cation Channels/genetics
5.
Poult Sci ; 101(9): 102011, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35901645

ABSTRACT

Bird death is often caused by renal lesions induced by chemicals. The avian kidney has a renal portal system with significant blood flow that is sensitive to many chemicals. However, early avian biomarkers for kidney injury are yet to be identified. This study aimed to identify novel renal biomarkers. Acute kidney injury (AKI) can be divided into acute interstitial nephritis (AIN) and acute tubular necrosis (ATN). A chicken model of kidney damage was created by an injection of diclofenac or cisplatin, which caused either AIN or ATN, respectively. Microarray analysis was performed to profile the gene expression patterns in the chickens with nephropathy. A gene enrichment analysis suggested that the genes related to responses to external stimuli showed expression changes in both AIN and ATN. However, hierarchical clustering analyses suggested that gene expression patterns differed between AIN and ATN, and the number of biomarkers relating to renal damage was low. To identify early biomarkers for nephropathy, we focused on genes that were induced at various levels of renal damage. The gene, vanin-1 (VNN1) was highly induced in the early stages of renal damage. A quantitative real-time PCR analysis supported this finding. These results suggest VNN1 could be a useful early biomarker of kidney injury in avian species.


Subject(s)
Kidney Tubular Necrosis, Acute , Nephritis, Interstitial , Animals , Biomarkers/metabolism , Chickens/genetics , Chickens/metabolism , Gene Expression Profiling/veterinary , Kidney/metabolism , Kidney Tubular Necrosis, Acute/metabolism , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubular Necrosis, Acute/veterinary , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Nephritis, Interstitial/veterinary
6.
Biochim Biophys Acta Mol Cell Res ; 1869(4): 119200, 2022 04.
Article in English | MEDLINE | ID: mdl-34968577

ABSTRACT

Tubulointerstitial fibrosis is a common pathway of chronic kidney disease (CKD) and is closely related to the progression of CKD. LMCD1, acting as an intermediary, has been reported to play a role in cardiac fibrosis. However, its role in renal fibrosis is yet to be deciphered. Based on the GEO database, we found the expression of LMCD1 is increased in kidney tissues of CKD patients and in human proximal tubular epithelial (HK-2) cells treated with transforming growth factor-ß1 (TGF-ß1), suggesting that LMCD1 may be involved in tubulointerstitial fibrosis. Herein, we investigated the role of LMCD1 in mice with unilateral ureteral obstruction (UUO) and in TGF-ß1-stimulated HK-2 cells. In the UUO model, the expression of LMCD1 was upregulated. UUO-induced renal histopathological changes were mitigated by knockdown of LMCD1. LMCD1 silence alleviated renal interstitial fibrosis in UUO mice by decreasing the expression of TGF-ß1, fibronectin, collagen I, and collagen III. LMCD1 deficiency suppressed cell apoptosis in kidney to prevent UUO-triggered renal injury. Furthermore, LMCD1 deficiency blocked the activation of ERK signaling in UUO mice. In vitro, LMCD1 was upregulated in HK-2 cells after TGF-ß1 stimulation. LMCD1 silence abrogated TGF-ß1-mediated upregulation of fibrotic genes. Treatment of HK-2 cells with ERK-specific inhibitor SCH772984 and agonist TPA validated LMCD1 exerted its function via activating ERK signaling. Together, our findings suggest that inhibition of LMCD1 protects against renal interstitial fibrosis by impeding ERK activation.


Subject(s)
Co-Repressor Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , LIM Domain Proteins/metabolism , Nephritis, Interstitial/pathology , Animals , Apoptosis , Cell Line , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Indazoles/pharmacology , Kidney/metabolism , Kidney/pathology , LIM Domain Proteins/antagonists & inhibitors , LIM Domain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Nephritis, Interstitial/etiology , Nephritis, Interstitial/metabolism , Piperazines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects , Ureteral Obstruction/complications
7.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830314

ABSTRACT

The kidney is one of the most susceptible organs to age-related impairments. Generally, renal aging is accompanied by renal fibrosis, which is the final common pathway of chronic kidney diseases. Aristolochic acid (AA), a nephrotoxic agent, causes AA nephropathy (AAN), which is characterized by progressive renal fibrosis and functional decline. Although renal fibrosis is associated with renal aging, whether AA induces renal aging remains unclear. The aim of the present study is to investigate the potential use of AAN as a model of renal aging. Here, we examined senescence-related factors in AAN models by chronically administering AA to C57BL/6 mice. Compared with controls, the AA group demonstrated aging kidney phenotypes, such as renal atrophy, renal functional decline, and tubulointerstitial fibrosis. Additionally, AA promoted cellular senescence specifically in the kidneys, and increased renal p16 mRNA expression and senescence-associated ß-galactosidase activity. Furthermore, AA-treated mice exhibited proximal tubular mitochondrial abnormalities, as well as reactive oxygen species accumulation. Klotho, an antiaging gene, was also significantly decreased in the kidneys of AA-treated mice. Collectively, the results of the present study indicate that AA alters senescence-related factors, and that renal fibrosis is closely related to renal aging.


Subject(s)
Aging/drug effects , Aristolochic Acids/pharmacology , Collagen/genetics , Kidney/drug effects , Nephritis, Interstitial/chemically induced , Renal Insufficiency, Chronic/chemically induced , Aging/genetics , Animals , Collagen/agonists , Collagen/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Humans , Kidney/metabolism , Kidney/pathology , Klotho Proteins/genetics , Klotho Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Nephritis, Interstitial/genetics , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Signal Transduction , Transforming Growth Factor beta/agonists , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
8.
Cells ; 10(11)2021 11 06.
Article in English | MEDLINE | ID: mdl-34831283

ABSTRACT

Renal tubulointerstitial lesions (TILs), a common pathologic hallmark of chronic kidney disease that evolves to end-stage renal disease, is characterized by progressive inflammation and pronounced fibrosis of the kidney. However, current therapeutic approaches to treat these lesions remain largely ineffectual. Previously, we demonstrated that elevated IL-36α levels in human renal tissue and urine are implicated in impaired renal function, and IL-36 signaling enhances activation of NLRP3 inflammasome in a mouse model of TILs. Recently, we synthesized NSC828779, a salicylanilide derivative (protected by U.S. patents with US 8975255 B2 and US 9162993 B2), which inhibits activation of NF-κB signaling with high immunomodulatory potency and low IC50, and we hypothesized that it would be a potential drug candidate for renal TILs. The current study validated the therapeutic effects of NSC828779 on TILs using a mouse model of unilateral ureteral obstruction (UUO) and relevant cell models, including renal tubular epithelial cells under mechanically induced constant pressure. Treatment with NSC828779 improved renal lesions, as demonstrated by dramatically reduced severity of renal inflammation and fibrosis and decreased urinary cytokine levels in UUO mice. This small molecule specifically inhibits the IL-36α/NLRP3 inflammasome pathway. Based on these results, the beneficial outcome represents synergistic suppression of both the IL-36α-activated MAPK/NLRP3 inflammasome and STAT3- and Smad2/3-dependent fibrogenic signaling. NSC828779 appears justified as a new drug candidate to treat renal progressive inflammation and fibrosis.


Subject(s)
Interleukin-1/metabolism , Nephritis, Interstitial/metabolism , Salicylanilides/pharmacology , Signal Transduction , Animals , Cell Line , Cytokines/urine , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Hydrogen Peroxide , Inflammasomes/metabolism , Lipopolysaccharides , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Molecular Docking Simulation , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephritis, Interstitial/complications , Nephritis, Interstitial/pathology , Nephritis, Interstitial/urine , STAT3 Transcription Factor/metabolism , Ureteral Obstruction/complications
9.
Ren Fail ; 43(1): 658-663, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33820492

ABSTRACT

All-trans retinoic acid (ATRA) is one of essentially active metabolite of vitamin A, and plays an important role in diverse physiological processes, such as cellular growth and function. Renal interstitial fibrosis (RIF) is a common pathological characteristic of chronic renal disease causing end-stage renal disease currently lacking effective treatment. Low level of Angiopoietins-1 (Angpt-1) is associated with extracellular matrix accumulation and fibrosis diseases. This study was performed to assess the association of ATRA with Angpt-1 in RIF disease. Rats were divided into three groups: group of sham (SHO group), group of unilateral ureteral obstruction group (UUO group), UUO mice administrated daily at the dose of ATRA (ATRA group). Masson-staining was used to detect the histologic lesion. Immunohistochemistry and Western-blot were applied to determine the targeted proteins. RIF score was significantly increased in UUO rats when compared with that of SHO group, and the fibrosis score was notably reduced in ATRA group. Transforming growth factor-ß1 (TGF-ß1), collagen IV (Col-IV) and fibronectin (FN) expressions in UUO group were significantly up-regulated, whereas Angpt-1 expression was significantly down-regulated compared with the SHO group. ATRA treatment reduced TGF-ß1, Col-IV and FN expressions and improved Angpt-1 expression compared with the UUO group. The protein expression of Angpt-1 in kidney tissue of UUO group was negatively correlated with RIF index and protein expressions of Col-IV, FN and TGF-ß1. In conclusion, low expression of Angpt-1 was associated with the RIF disease and ATRA treatment can increase the Angpt-1 and alleviate the RIF lesion in UUO rats.


Subject(s)
Angiopoietin-1/metabolism , Extracellular Matrix/metabolism , Nephritis, Interstitial/drug therapy , Nephritis, Interstitial/pathology , Tretinoin/pharmacology , Angiopoietin-1/genetics , Animals , Collagen Type IV/metabolism , Disease Models, Animal , Disease Progression , Down-Regulation , Extracellular Matrix/drug effects , Fibronectins/metabolism , Fibrosis/pathology , Male , Nephritis, Interstitial/genetics , Nephritis, Interstitial/metabolism , Rats , Transforming Growth Factor beta1/metabolism
10.
Immunotherapy ; 13(4): 283-288, 2021 03.
Article in English | MEDLINE | ID: mdl-33397120

ABSTRACT

Renal toxicity from immune checkpoint inhibitors (ICIs) is an increasingly recognized cause of acute kidney injury among patients with cancer. ICI-associated acute kidney injuries typically present as acute interstitial nephritis and the timing of onset is highly variable. Herein, we present a case of a patient with relapsed metastatic melanoma previously treated with pembrolizumab who developed grade 3 immune-related renal toxicity after reintroduction of the same ICI, secondary to acute interstitial nephritis with accompanying high PR3-antineutrophil cytoplasmic antibody titer. The patient improved after steroid treatment and discontinuation of pembrolizumab. This case highlights the importance of not excluding ICI-related nephrotoxicity as a possible cause of renal failure, including in those who previously tolerated ICI treatment, since it is a treatable entity.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/drug effects , Antibodies, Monoclonal, Humanized/adverse effects , Immune Checkpoint Inhibitors/adverse effects , Nephritis, Interstitial/chemically induced , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Aged , Antibodies, Antineutrophil Cytoplasmic/blood , B7-H1 Antigen/metabolism , Female , Humans , Immunotherapy/adverse effects , Melanoma/drug therapy , Melanoma/pathology , Nephritis, Interstitial/drug therapy , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Recurrence , Steroids/therapeutic use , Treatment Outcome
11.
Life Sci ; 264: 118686, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33129879

ABSTRACT

AIM: Diabetic nephropathy (DN) is the dominant cause of end-stage renal disease which is characterized by extracellular matrix accumulation. The purpose of this study was to investigate the role of activating transcription factor 4 (ATF4) in regulating renal fibrosis and autophagy in DN. MAIN METHOD: Streptozotocin (STZ) was administered to heterozygous ATF4 knockout (KO) and wild-type (WT) mice via an intraperitoneal injection to induce DN. NRK-52E cells were cultured in high glucose to mimic diabetic pathological. qRT-PCR, western blot, immunofluorescence, histology and electron microscopic analysis were performed. The autophagy flux was observed by tandem mRFP-GFP-LC3 fluorescence microscopy. KEY FINDINGS: DN mice experienced severe renal injury and fibrosis and showed increased expression of ATF4 and inhibition of autophagy in kidney tissues. We found that STZ-induced ATF4 KO mice showed significant improvement in urinary albumin, serum creatinine and blood urea nitrogen and the pathological changes of renal tubulointerstitial fibrosis compared with STZ-induced WT mice. Furthermore, inhibition of ATF4 could restore autophagy in DN mice. Similar results were shown in vitro. Overexpression of ATF4 in NRK-52E cells cultured in high glucose condition suppressed autophagy and upregulated Collagen type 4 (Col-IV) expression, while inhibition of ATF4 could increase the number of the autophagosomes, improve autophagic flux and decrease Col-IV level. SIGNIFICANCE: Our study provided the evidence of a crucial role for ATF4 in inhibiting autophagy against diabetic kidney damage. Suppression of ATF4 may be an effective therapy in restraining renal tubulointerstitial fibrosis in DN.


Subject(s)
Activating Transcription Factor 4/biosynthesis , Autophagy/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Nephritis, Interstitial/metabolism , Activating Transcription Factor 4/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Fibrosis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nephritis, Interstitial/genetics , Nephritis, Interstitial/pathology
12.
Biomed Pharmacother ; 133: 110949, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33227703

ABSTRACT

Autophagy has been implicated in the pathogenesis of chronic kidney disease (CKD). Transcription factor EB (TFEB) is a master controller of autophagy. However, the pathophysiological roles of TFEB in modulating autophagy and tubulointerstitial injury in CKD are unknown. This study aimed to determine whether TFEB-mediated autophagy contributed to the tubulointerstitial injury in mice with CKD. After the mice were treated with an adenine diet (0.2 % adenine) for 8 weeks, the development of CKD was observed to be characterised by increased levels of plasma blood urea nitrogen (BUN), creatinine (Cre), tubulointerstitial inflammation and fibrosis. Immunohistochemical and Western blot analysis further revealed that TFEB and autophagy genes were significantly up-regulated in the kidney of the mice with adenine-induced CKD, and this increase was mostly found in the tubular epithelial cells. Interestingly, a similar expression pattern of TFEB-autophagy genes was observed in tubular epithelial cells in the kidney tissue of patients with immunoglobulin A (IgA) nephropathy. Moreover, a pathogenic role of TFEB in adenine-induced CKD was speculated because the pharmacological activation of TFEB by trehalose failed to protect mice from tubulointerstitial injuries. In the epithelioid clone of normal rat kidney cells (NRK-52E), the activation of TFEB by trehalose increased autophagy induction, cell death and inflammatory cytokine (Interleukin-6, IL-6) release. Collectively, these results suggested that the activation of TFEB-mediated autophagy might cause autophagic cell death and inflammation in tubular epithelial cells, contributing to renal fibrosis in adenine-induced CKD. This study provided novel insights into the pathogenic role of TFEB in CKD associated with a high purine diet.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Epithelial Cells/metabolism , Kidney Tubules/metabolism , Nephritis, Interstitial/metabolism , Renal Insufficiency, Chronic/metabolism , Adenine , Animals , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/agonists , Cell Line , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/ultrastructure , Fibrosis , Humans , Inflammation Mediators/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Kidney Tubules/drug effects , Kidney Tubules/ultrastructure , Male , Mice, Inbred C57BL , Nephritis, Interstitial/chemically induced , Nephritis, Interstitial/pathology , Rats , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Signal Transduction , Trehalose/pharmacology
13.
Adv Chronic Kidney Dis ; 27(5): 365-376, 2020 09.
Article in English | MEDLINE | ID: mdl-33308501

ABSTRACT

Acute kidney injury (AKI) is common among hospitalized patients with Coronavirus Infectious Disease 2019 (COVID-19), with the occurrence of AKI ranging from 0.5% to 80%. The variability in the occurrence of AKI has been attributed to the difference in geographic locations, race/ethnicity, and severity of illness. AKI among hospitalized patients is associated with increased length of stay and in-hospital deaths. Even patients with AKI who survive to hospital discharge are at risk of developing chronic kidney disease or end-stage kidney disease. An improved knowledge of the pathophysiology of AKI in COVID-19 is crucial to mitigate and manage AKI and to improve the survival of patients who developed AKI during COVID-19. The goal of this article is to provide our current understanding of the etiology and the pathophysiology of AKI in the setting of COVID-19.


Subject(s)
Acute Kidney Injury/metabolism , COVID-19/metabolism , Cytokines/metabolism , Glomerulonephritis/metabolism , Thrombotic Microangiopathies/metabolism , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Anti-Bacterial Agents/adverse effects , Antiviral Agents/adverse effects , Apolipoprotein L1/genetics , Ascorbic Acid/adverse effects , Azotemia/metabolism , Azotemia/pathology , Azotemia/physiopathology , COVID-19/pathology , COVID-19/physiopathology , Disease Progression , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Glomerulonephritis, Membranous/metabolism , Glomerulonephritis, Membranous/pathology , Glomerulonephritis, Membranous/physiopathology , Hospital Mortality , Humans , Kidney Tubules, Proximal/injuries , Length of Stay , Myoglobin/metabolism , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Nephritis, Interstitial/physiopathology , Nephrosis, Lipoid/metabolism , Nephrosis, Lipoid/pathology , Nephrosis, Lipoid/physiopathology , Renal Insufficiency, Chronic , Rhabdomyolysis/metabolism , SARS-CoV-2 , Severity of Illness Index , Thrombotic Microangiopathies/pathology , Thrombotic Microangiopathies/physiopathology , Vitamins/adverse effects , COVID-19 Drug Treatment
14.
Acta Histochem ; 122(8): 151655, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33171392

ABSTRACT

BACKGROUND: It is believed that tubulo-interstitial fibrosis and atrophy in diabetic patients are directly associated with the progression of chronic kidney disease, CKD. AIF is one of the crucial factors responsible for mitochondrial apoptosis, however, it can also promote cell survival independently from its role in apoptosis, and therefore can be potentially used as a tool in prevention of the onset of CKD in diabetic patients. Our aim was to investigate the significance of AIF expression in the development of CKD by observing the expression of AIF in 2 weeks' and 2 months' kidneys of diabetic rats compared to their controls. METHODS: Male Sprague-Dawley rats were treated with 55 mg/kg streptozotocin (model of type 1 diabetes mellitus; DM group) or citrate buffer (control). After 2 weeks and 2 months kidney samples were collected and analysed in different renal areas. RESULTS: Characteristic morphologic changes were found between the 2 months' control and 2 months' diabetic groups. Those changes, including fibrosis and possible replacement of podocytes with connective tissue were mainly present in the glomeruli. AIF expression was seen in the both cortex, and in the collecting ducts of the medulla. Strong intensity of AIF expression was seen in proximal and distal convoluted tubules in both diabetic groups. In the control groups the glomeruli showed no AIF staining but moderate staining was seen in both diabetic groups. Overall, the percentage of AIF positive cells in the glomeruli was the lowest. The greatest rise in cell positivity was displayed from the 2 weeks' control group to 2 weeks' diabetes group (38 %) in glomeruli. The cell positivity of the 2 weeks' diabetic group is significantly reduced to 18 % in the 2 months' diabetic group in glomeruli. A similar pattern was seen in the proximal tubular cells (92 % positivity 2 weeks diabetic groups; 89 % positivity 2 months diabetic groups), as well as in the distal tubules. The highest percentage of AIF positive cells was seen in the collecting ducts, more than 80 % in all groups. CONCLUSIONS: Our study provides insight into AIF expression pattern during short term diabetes model, confirming possible dual role of AIF, not only in apoptosis but also in cell function and homeostasis, and proving AIF as potential therapeutic target and marker of advancement of CKD.


Subject(s)
Apoptosis Inducing Factor/genetics , Diabetes Mellitus, Experimental/genetics , Diabetic Nephropathies/genetics , Nephritis, Interstitial/genetics , Animals , Apoptosis/drug effects , Apoptosis Inducing Factor/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Fibrosis , Gene Expression Regulation , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/pathology , Kidney Tubules, Distal/metabolism , Kidney Tubules, Distal/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Podocytes/metabolism , Podocytes/pathology , Rats , Rats, Sprague-Dawley , Streptozocin/administration & dosage
15.
BMC Nephrol ; 21(1): 315, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32727411

ABSTRACT

BACKGROUND: IgG4-related disease (IgG4-RD) is a newly classified but poorly understood immune-medicated systemic disease. It causes potential fibroinflammation in one or more organs, characterized by tumescent organs and marked IgG4-positive plasma cells infiltration in the affected tissues. There have been a few cases revealing close relationship between IgG4-RD and formation of B cell lymphoma. Diffuse large B cell lymphoma (DLBCL) and extranodal marginal zone lymphoma (EMZL) of mucosa-associated lymphoid tissue are the most common sub-types ever described, whereas the exact mechanism remain unclear. CASE PRESENTATION: We report a 64-year old Chinese male who presented chronic kidney disease and was initially diagnosed typical IgG4-RD. Pathological findings revealed there was restricted expression of lambda light chain in the kidney. There was also elevated uptake abnormality observed in 18F-FDG-PET/CT. Prednisone combined with oral cyclophosphamide helped the patient to get a partial remission of renal function and an obvious decrease of IgG4 level. However, he developed DLBCL 16 months after IgG4-RD diagnosis. The DLBCL is speculated to transform from a pre-existing but possible missed diagnosed EMZL. CONCLUSIONS: Concurrent IgG4-RD with kidney-origin EMZL developing DLBCL has never been reported in the literature. Clinicians should keep in mind that lymphoma may occur in IgG4-RD. The mechanism of lymphomagenesis potential in IgG4-RD needs further study.


Subject(s)
Immunoglobulin G4-Related Disease/diagnosis , Immunoglobulin lambda-Chains/metabolism , Lymphoma, Large B-Cell, Diffuse/diagnosis , Renal Insufficiency, Chronic/diagnosis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/therapeutic use , Disease Progression , Fluorodeoxyglucose F18 , Glucocorticoids/therapeutic use , Humans , Immunoglobulin G4-Related Disease/drug therapy , Immunoglobulin G4-Related Disease/metabolism , Immunoglobulin G4-Related Disease/pathology , Immunosuppressive Agents/therapeutic use , Lymphoma, B-Cell, Marginal Zone/diagnosis , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Missed Diagnosis , Nephritis, Interstitial/drug therapy , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Positron Emission Tomography Computed Tomography , Prednisone/therapeutic use , Radiopharmaceuticals , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
16.
FASEB J ; 34(9): 12599-12614, 2020 09.
Article in English | MEDLINE | ID: mdl-32706145

ABSTRACT

Renal tubulointerstitial fibrosis (TIF) is a common pathological feature of aristolochic acid (AA) nephropathy (AAN). G2/M arrest of proximal tubular cells (PTCs) is implicated in renal fibrosis of AAN, but the upstream regulatory molecule remains unknown. Hypoxia inducible factor-1α (HIF-1α) promotes renal fibrosis in kidney disease, but the role of HIF-1α in AAN is unclear. Evidence shows that HIF-1α and p21, a known inducer of cellular G2/M arrest, are closely related to each other. To investigate the role of HIF-1α in renal fibrosis of AAN and its effects on p21 expression and PTCs G2/M arrest, mice with HIF-1α gene knockout PTCs (PT-HIF-1α-KO) were generated, and AAN was induced by AA. In vitro tests were conducted on the human PTCs line HK-2 and primary mouse PTCs. HIF-1α and p21 expression, fibrogenesis, and G2/M arrest of PTCs were determined. Results showed that HIF-1α was upregulated in the kidneys of wild-type (WT) AAN mice, accompanied by p21 upregulation, PTCs G2/M arrest and renal fibrosis, and these alterations were reversed in PT-HIF-1α-KO AAN mice. Similar results were observed in HK-2 cells and were further confirmed in primary PTCs from PT-HIF-1α-KO and WT mice. Inhibiting p21 in HK-2 cells and primary PTCs did not change the expression of HIF-1α, but G2/M arrest and fibrogenesis were reduced. These data indicate that HIF-1α plays a key role in renal fibrosis in AAN by inducing PTCs G2/M arrest modulated through p21. HIF-1α may serve as a potential therapeutic target for AAN.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/cytology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Kidney Tubules, Proximal , Nephritis, Interstitial/metabolism , Animals , Aristolochic Acids , Cell Line , Fibrosis/chemically induced , G2 Phase Cell Cycle Checkpoints , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/pathology , Mice , Mice, Knockout
17.
Biochem Pharmacol ; 180: 114079, 2020 10.
Article in English | MEDLINE | ID: mdl-32511988

ABSTRACT

Renal interstitial fibrosis (RIF) is a major pathological feature of chronic kidney disease at middle and end stages. Chrysophanol (CP), 1,8-dihydroxy-3-methyl-9,10-anthraquinone, is an anthraquinone isolated from Rheum palmatum L. with a variety of pharmacological activities including the suppression of RIF. However, the effect of CP on renal fibrosis and its potential mechanism have not been elucidated. We conducted a comprehensive study by determining the expression levels of fibrotic markers and proteins including TGF-ß1, α-SMA, and Smad3 related to transforming growth factor-beta/Smad (TGF-ß/Smad) pathway in unilateral ureteral obstruction (UUO) mice and TGF-ß1-stimulated HK-2 cells with the treatment of CP using western blotting and RT-qPCR analyses. Using small interfering RNA and co-immunoprecipitation, we evaluated the influences of CP on the interactions between Smad3 and Smad7 proteins and also on TGF-ß RI and TGF-ßR II. We found that CP administration significantly ameliorated UUO-induced kidney damage by reversing abnormal serum and urine biochemical parameters and decreasing the production of fibrotic markers including collagen I, collagen III, fibronectin, and α-SMA. Our results showed that TGF-ß1 and phospho-Smad3 (p-Smad3) expression was significantly down-regulated and Smad7 expression was up-regulated by CP in UUO mice compared to the model group; however, the expression of Smad2, Smad4, and TGF-ß receptors was not affected. Furthermore, CP modulated these fibrotic markers as well as p-Smad3 and Smad7 in TGF-ß1-induced HK-2 cells. The inhibitory effect of CP was markedly reduced in TGF-ß1-treated HK-2 cells transfected with Smad3 siRNA. Additionally, co-immunoprecipitation analysis indicated that CP blocked the interaction between Smad3 and TGF-ß receptor I to suppress p-Smad3 expression. These findings demonstrated that CP alleviated RIF by inhibiting Smad3 phosphorylation, which provides a molecular basis for a new drug candidate for the treatment of RIF.


Subject(s)
Anthraquinones/therapeutic use , Kidney , Nephritis, Interstitial/prevention & control , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Anthraquinones/administration & dosage , Cell Line , Cell Survival/drug effects , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Humans , Kidney/drug effects , Kidney/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Inbred C57BL , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Signal Transduction
18.
Theranostics ; 10(6): 2803-2816, 2020.
Article in English | MEDLINE | ID: mdl-32194836

ABSTRACT

Background: Our previous study demonstrated that the disruption of cholesterol homeostasis promotes tubulointerstitial injury in diabetic nephropathy (DN). This study aimed to further investigate the effects of gut microbiota dysbiosis on this process and explored its potential mechanism. Methods: Diabetic rats treated with broad-spectrum oral antibiotics or faecal microbiota transplantation (FMT) from the healthy donor group and human kidney 2 (HK-2) cells stimulated with sodium acetate were used to observe the effects of gut microbiota on cholesterol homeostasis. The gut microbiota distribution was measured by 16S rDNA sequencing with faeces. Serum acetate level was examined by gas chromatographic analysis. Protein expression of G protein coupled receptor 43 (GPR43) and molecules involved in cholesterol homeostasis were assessed by immunohistochemical staining, immunofluorescence staining, and Western Blotting. Results: Depletion of gut microbiota significantly attenuated albuminuria and tubulointerstitial injury. Interestingly, serum acetate levels were also markedly decreased in antibiotics-treated diabetic rats and positively correlated with the cholesterol contents in kidneys. An in vitro study demonstrated that acetate significantly increased cholesterol accumulation in HK-2 cells, which was caused by increased expression of proteins mainly modulating cholesterol synthesis and uptake. As expected, FMT effectively decreased serum acetate levels and alleviated tubulointerstitial injury in diabetic rats through overriding the disruption of cholesterol homeostasis. Furthermore, GPR43 siRNA treatment blocked acetate-mediated cholesterol homeostasis dysregulation in HK-2 cells through decreasing the expression of proteins governed cholesterol synthesis and uptake. Conclusion: Our studies for the first time demonstrated that the acetate produced from gut microbiota mediated the dysregulation of cholesterol homeostasis through the activation of GPR43, thereby contributing to the tubulointerstitial injury of DN, suggesting that gut microbiota reprogramming might be a new strategy for DN prevention and therapy.


Subject(s)
Cholesterol/metabolism , Diabetic Nephropathies , Dysbiosis , Gastrointestinal Microbiome , Nephritis, Interstitial , Acetates/blood , Animals , Cell Line , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/microbiology , Dysbiosis/metabolism , Dysbiosis/microbiology , Homeostasis , Humans , Male , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/microbiology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism
19.
Inflammation ; 43(3): 878-891, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31940108

ABSTRACT

Norcantharidin (NCTD) is a potential anti-renal interstitial fibrotic drug. However, the underlying molecular mechanism of how NCTD works remains unclear. In this study, using both in vivo and in vitro models, we report that the level of ß-catenin is positively correlated to the degree of renal interstitial fibrosis (RIF). Protein phosphatase 2A (PP2A) binds to ß-catenin and suppresses its phosphorylation, thereby increasing the total ß-catenin expression. Additionally, NCTD dramatically elevates the level of phosphorylated ß-catenin and decreases total ß-catenin expression in a dose-dependent manner, consequently leading to the reduction of RIF. Mechanistically, PP2A-mediated suppression of ß-catenin phosphorylation is an essential target for the anti-renal interstitial fibrotic effect of NCTD. Therefore, we report a potential theoretical basis for clinical application of NCTD in treating RIF.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Nephritis, Interstitial/drug therapy , Nephritis, Interstitial/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , beta Catenin/metabolism , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Fibrosis/drug therapy , Fibrosis/metabolism , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Male , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Treatment Outcome
20.
Nephrol Dial Transplant ; 35(4): 576-586, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31495893

ABSTRACT

BACKGROUND: Tubulointerstitial fibrosis is a hallmark of chronic kidney disease (CKD), and is initiated by tubular epithelial cell (TEC) injury. Hypoxia promotes tubular cell death, fibrosis and CKD progression. Munc18-1-interacting protein 3 (Mint3) is a molecule that activates hypoxia-inducible factors (HIFs) by binding and suppressing factor inhibiting HIF-1 (FIH). However, the role of Mint3 in tubulointerstitial fibrosis remains unknown. METHODS: We induced fibrosis of the kidney after unilateral ischemia-reperfusion injury (uIRI) in Mint3-knockout and littermate wild-type mice. The duration of ischemia was 23 min and the kidneys were harvested at 24 h and 7 days after ischemia-reperfusion. The function of Mint3 was further investigated by using mouse cortical tubular (MCT) cells, which were treated with Mint3 and/or FIH small interfering RNA and exposed to normoxia or hypoxia. RESULTS: Knockout of Mint3 did not affect the acute injury induced by uIRI, but exacerbated the tubulointerstitial fibrosis, accompanied by an increase in TEC apoptosis. Consistently, hypoxia-induced apoptosis of MCT cells was aggravated by Mint3 knockdown. Unexpectedly, the additional knockdown of FIH did not suppress the increase in apoptosis by Mint3 knockdown, demonstrating the irrelevance of the FIH/HIF pathway. Therefore, we next focused on nuclear factor (NF)-κB, which has an anti-apoptotic role. Indeed, not only the expression of the inhibitory NF-κB p50 but also the DNA-binding activity of p50/p50 homodimer was increased by knockdown of Mint3 in the TECs, along with the decreased expressions of the NF-κB-targeted anti-apoptotic genes. An increase in NF-κB p50 was also confirmed in Mint3-knockout kidneys. CONCLUSIONS: Mint3 in epithelial cells protects the cells from apoptosis by up-regulating anti-apoptotic effects of NF-κB, leading to fibrosis suppression. This new pathophysiology of tubulointerstitial fibrosis could be a target of future therapy for CKD.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis , Epithelial Cells/pathology , Fibrosis/prevention & control , NF-kappa B/metabolism , Nephritis, Interstitial/prevention & control , Reperfusion Injury/complications , Animals , Epithelial Cells/metabolism , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Mice , Mice, Knockout , NF-kappa B/genetics , Nephritis, Interstitial/etiology , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology
SELECTION OF CITATIONS
SEARCH DETAIL