Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Biochem Biophys Res Commun ; 620: 15-20, 2022 09 10.
Article in English | MEDLINE | ID: mdl-35772212

ABSTRACT

Lipoprotein lipase (LPL) is an enzyme that catalyzes the hydrolysis of circulating triglyceride and the transport of fatty acids into cells. Its activity is positively regulated by insulin, and insulin resistance is associated with low LPL activity and subsequent hypertriglyceridemia. The involvement of hypertriglyceridemia in chronic kidney disease (CKD) is still under the debate in a clinical setting. Therefore, we aimed to study the role of hypertriglyceridemia in the disease using mice with systemic or renal-specific LPL deficiency. Systemic LPL deficiency was characterized by hypertriglyceridemia, but not renal injury or dyslipidemia-related conditions, such as fatty liver. Furthermore, the LPL deficiency-induced hypertriglyceridemia was not associated with a worsening of the CKD phenotype or atherosclerosis, even when CKD was induced by 5/6 nephrectomy. Next, because LPL-mediated fatty acid uptake may be important for energy metabolism in proximal tubular epithelial cells (PTECs), the role of renal LPL in renal physiology was studied by generating mice lacking LPL specifically in PTECs. These mice showed no abnormalities in their histology or renal reabsorption of micro molecules. These findings suggest that systemic and renal lipid abnormalities caused by LPL deficiency do not cause or worsen the development of renal injury, and provide novel insight regarding the potential role of lipotoxicity in the pathogenesis of obesity-related kidney injury.


Subject(s)
Hyperlipoproteinemia Type I , Hypertriglyceridemia , Renal Insufficiency, Chronic , Animals , Kidney/metabolism , Lipoprotein Lipase/metabolism , Mice , Renal Insufficiency, Chronic/etiology , Triglycerides/metabolism
2.
Biochem Biophys Res Commun ; 525(2): 319-325, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32089264

ABSTRACT

To examine the cell-protective role of podocyte autophagy against glomerular endothelial dysfunction in diabetes, we analyzed the renal phenotype of tamoxifen (TM)-inducible podocyte-specific Atg5-deficient (iPodo-Atg5-/-) mice with experimental endothelial dysfunction. In both control and iPodo-Atg5-/- mice, high fat diet (HFD) feeding induced glomerular endothelial damage characterized by decreased urinary nitric oxide (NO) excretion, collapsed endothelial fenestrae, and reduced endothelial glycocalyx. HFD-fed control mice showed slight albuminuria and nearly normal podocyte morphology. In contrast, HFD-fed iPodo-Atg5-/- mice developed massive albuminuria accompanied by severe podocyte injury that was observed predominantly in podocytes adjacent to damaged endothelial cells by scanning electron microscopy. Although podocyte-specific autophagy deficiency did not affect endothelial NO synthase deficiency-associated albuminuria, it markedly exacerbated albuminuria and severe podocyte morphological damage when the damage was induced by intravenous neuraminidase injection to remove glycocalyx from the endothelial surface. Furthermore, endoplasmic reticulum stress was accelerated in podocytes of iPodo-Atg5-/- mice stimulated with neuraminidase, and treatment with molecular chaperone tauroursodeoxycholic acid improved neuraminidase-induced severe albuminuria and podocyte injury. In conclusion, podocyte autophagy plays a renoprotective role against diabetes-related structural endothelial damage, providing an additional insight into the pathogenesis of massive proteinuria in diabetic nephropathy.


Subject(s)
Autophagy/physiology , Diabetes Mellitus, Experimental/pathology , Endothelial Cells/pathology , Kidney Glomerulus/pathology , Podocytes/pathology , Albuminuria/etiology , Animals , Autophagy-Related Protein 5/deficiency , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/complications , Diet, High-Fat , Mice , Proteinuria/etiology
3.
J Am Soc Nephrol ; 30(6): 962-978, 2019 06.
Article in English | MEDLINE | ID: mdl-31043434

ABSTRACT

BACKGROUND: Energy metabolism in proximal tubular epithelial cells (PTECs) is unique, because ATP production largely depends on lipolysis in both the fed and fasting states. Furthermore, disruption of renal lipolysis is involved in the pathogenesis of diabetic tubulopathy. Emerging evidence suggests that protein O-GlcNAcylation, an intracellular nutrient-sensing system, may regulate a number of metabolic pathways according to changes in nutritional status. Although O-GlcNAcylation in PTECs has been demonstrated experimentally, its precise role in lipolysis in PTECs is unclear. METHODS: To investigate the mechanism of renal lipolysis in PTECs-specifically, the role played by protein O-GlcNAcylation-we generated mice with PTECs deficient in O-GlcNAc transferase (Ogt). We analyzed their renal phenotypes during ad libitum feeding, after prolonged fasting, and after mice were fed a high-fat diet for 16 weeks to induce obesity and diabetes. RESULTS: Although PTEC-specific Ogt-deficient mice lacked a marked renal phenotype during ad libitum feeding, after fasting 48 hours, they developed Fanconi syndrome-like abnormalities, PTEC apoptosis, and lower rates of renal lipolysis and ATP production. Proteomic analysis suggested that farnesoid X receptor-dependent upregulation of carboxylesterase-1 is involved in O-GlcNAcylation's regulation of lipolysis in fasted PTECs. PTEC-specific Ogt-deficient mice with diabetes induced by a high-fat diet developed severe tubular cell damage and enhanced lipotoxicity. CONCLUSIONS: Protein O-GlcNAcylation is essential for renal lipolysis during prolonged fasting and offers PTECs significant protection against lipotoxicity in diabetes.


Subject(s)
Gene Expression Regulation , Kidney Tubules, Proximal/metabolism , Lipid Metabolism/genetics , Lipolysis/genetics , N-Acetylglucosaminyltransferases/genetics , Animals , Apoptosis/genetics , Cells, Cultured , Diabetes Mellitus, Experimental , Diet, High-Fat , Disease Models, Animal , Energy Metabolism/genetics , Fasting , Homeostasis/genetics , Kidney Tubules, Proximal/cytology , Male , Mice , Mice, Knockout , N-Acetylglucosaminyltransferases/metabolism , Proteomics , Random Allocation , Reference Values
4.
Biochem Biophys Res Commun ; 495(1): 1115-1121, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29175208

ABSTRACT

A single nucleotide polymorphism (SNP) within the acetyl CoA carboxylase (ACC) ß gene (ACACB), rs2268388, has been shown to be associated with susceptibility to development of proteinuria in patients with type 2 diabetes. To investigate the biological roles of ACCß in the pathogenesis of diabetic nephropathy, we examined the effects of overexpression of ACACB using podocyte-specific ACACB-transgenic mice or ACACB-overexpressing murine podocytes. Podocyte-specific ACACB-transgenic mice or littermate mice were treated with streptozotocin (STZ) to induce diabetes, and 12 weeks after induction of diabetes, we examined the expression of podocyte markers to evaluate the degree of podocyte injury in these mice. We also examined the effects of ACCß on podocyte injury in ACACB- or LacZ-overexpressing murine podocytes. Podocyte-specific ACACB overexpression did not cause visible podocyte injury in non-diabetic mice. In STZ-induced diabetic mice, ACACB-transgenic mice showed a significant increase in urinary albumin excretion, accompanied by decreased synaptopodin expression and podocin mislocalization in podocytes, compared with wild-type mice. In cultured murine podocytes, overexpression of ACACB significantly decreased synaptopodin expression and reorganized stress fibers under high glucose conditions, but not in normal glucose conditions. The decrease of synaptopodin expression and reorganized stress fibers observed in ACACB overexpressing cells cultured under high glucose conditions was reversed by a treatment of 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside (AICAR), activator of AMP-activated protein kinase (AMPK). The excess of ACCß might contribute to exacerbation of podocyte injury in the kidney of an animal model for diabetes mellitus, and the AMPK/ACCß pathway may be a novel therapeutic target for the prevention of diabetes-related podocyte injury.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Diabetic Nephropathies/enzymology , Diabetic Nephropathies/pathology , Podocytes/enzymology , Podocytes/pathology , Animals , Cells, Cultured , Gene Expression Regulation, Enzymologic , Mice , Mice, Inbred C57BL , Mice, Transgenic , Up-Regulation
5.
Nephrol Dial Transplant ; 32(9): 1477-1487, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28339907

ABSTRACT

BACKGROUND: O-linked ß- N -acetylglucosamine modification O-GlcNAcylation) is a post-translational modification of intracellular proteins, serving as a nutrient sensor. Growing evidence has demonstrated its physiological and pathological importance in various mammalian tissues. This study examined the physiological role of O-GlcNAcylation in podocyte function and development. METHODS: O-GlcNAc transferase (Ogt) is a critical enzyme for O-GlcNAcylation and resides on the X chromosome. To abrogate O-GlcNAcylation in podocytes, we generated congenital and tamoxifen (TM)-inducible podocyte-specific Ogt knockout mice (Podo-Ogt y/- and TM-Podo-Ogt y/- , respectively) and analyzed their renal phenotypes. RESULTS: Podo-Ogt y/- mice showed normal podocyte morphology at birth. However, they developed albuminuria at 8 weeks of age, increasing progressively until age 32 weeks. Glomerular sclerosis, proteinuria-related tubulointerstitial lesions and markedly altered podocyte foot processes, with decreased podocin expression, were observed histologically in 32-week-old Podo-Ogt y/- mice. Next, we induced adult-onset deletion of the Ogt gene in podocytes by TM injection in 8-week-old TM-Podo-Ogt y/- mice. In contrast to Podo-Ogt y/- mice, the induced TM-Podo-Ogt y/- mice did not develop albuminuria or podocyte damage, suggesting a need for O-GlcNAcylation to form mature foot processes after birth. To test this possibility, 3-week-old Podo-Ogt y/- mice were treated with Bis-T-23, which stimulates actin-dependent dynamin oligomerization, actin polymerization and subsequent foot process elongation in podocytes. Albuminuria and podocyte damage in 16-week-old Podo-Ogt y/- mice were prevented by Bis-T-23 treatment. CONCLUSIONS: O-GlcNAcylation is necessary for maturation of podocyte foot processes, particularly after birth. Our study provided new insights into podocyte biology and O-GlcNAcylation.


Subject(s)
Acetylglucosamine/chemistry , Foot/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , N-Acetylglucosaminyltransferases/physiology , Podocytes/metabolism , Protein Processing, Post-Translational , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Kidney Int ; 90(6): 1211-1225, 2016 12.
Article in English | MEDLINE | ID: mdl-27591086

ABSTRACT

Hypoxia causes proximal tubular cell damage in diabetes, even though proximal tubular cells have an adaptive system to combat hypoxia involving induction of hypoxia factor-1 (HIF-1) and inhibition of mechanistic target of rapamycin complex 1 (mTORC1). Here, we examined the interference effect of altered glucose and lipid metabolism on the hypoxia responses in proximal tubular cells. In culture, hypoxia alone induced HIF-1 and inhibited mTORC1, preventing death in proximal tubular cells. However, hypoxia with high glucose and palmitate increased mTORC1 activity and promoted apoptosis in proximal tubular cells, which was inhibited by pharmacological and genetic inactivation of mTORC1. Since inhibition of all mTORC1's physiological functions regulated by growth factors including insulin causes various adverse effects, we screened for a microRNA that can inhibit only pro-apoptotic effects of mTORC1 to discover a safe therapeutic target. This screen found microRNA-148b-3p was able to specifically inhibit mTORC1-dependent apoptosis in hypoxic proximal tubular cells exposed to high glucose and palmitate, without affecting insulin-dependent mTORC1 activation. Furthermore, tumor necrosis factor receptor (TNFR) 2 was the target of microRNA-148b-3p and its suppression inhibited apoptosis. Finally, enhanced apoptosis with TNFR2 overexpression was found in hypoxic and mTORC1-activated proximal tubular cells in diabetic rats. Thus, diabetes activated mTORC1 even in hypoxic proximal tubular cells, leading to apoptosis by reducing microRNA-148b-3p expression. Modulating this pathogenic pathway may be a novel therapy for proximal tubular cell damage in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Kidney Tubules, Proximal/metabolism , MicroRNAs/metabolism , Multiprotein Complexes/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Cells, Cultured , Glucose , Hypoxia/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipid Metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Palmitic Acid , Rats , Signal Transduction
7.
Biochem Biophys Res Commun ; 470(3): 539-545, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26802469

ABSTRACT

Dipeptidyl peptidase (DPP)-4 inhibitors, a new class of antidiabetic agent, have recently been suggested to exert pleiotropic effects beyond glucose lowering. Renal prognosis in patients with diabetic nephropathy depends on the severity of tubulointerstitial injury induced by massive proteinuria. We thus examined the renoprotective effect of DPP-4 inhibitors on inflammation in cultured mouse proximal tubular cells stimulated with free fatty acid (FFA)-bound albumin. Linagliptin and higher concentrations of sitagliptin, vildagliptin, and alogliptin all inhibited FFA-bound albumin-induced increases in mRNA expression of MCP-1 in cultured mouse proximal tubular cells. Furthermore, linagliptin significantly inhibited tubulointerstitial injury induced by peritoneal injection of FFA-bound albumin, such as inflammation, fibrosis, and apoptosis, in mice without altering systemic characteristics including body weight, fasting blood glucose, and food intake. These results indicate that DPP-4 inhibitors pleiotropically exert a direct renoprotective effect, and may serve as an additional therapeutic strategy to protect proximal tubular cells against proteinuria in patients with diabetic nephropathy.


Subject(s)
Apoptosis/drug effects , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Kidney Tubules, Proximal/injuries , Kidney Tubules, Proximal/metabolism , Albumins , Animals , Cells, Cultured , Cytoprotection/drug effects , Dipeptidyl Peptidase 4 , Dose-Response Relationship, Drug , Fatty Acids, Nonesterified , Kidney Tubules, Proximal/drug effects , Male , Mice , Mice, Inbred C57BL , Treatment Outcome
8.
Int J Mol Sci ; 17(11)2016 Nov 09.
Article in English | MEDLINE | ID: mdl-27834856

ABSTRACT

Saturated fatty acid (SFA)-related lipotoxicity is a pathogenesis of diabetes-related renal proximal tubular epithelial cell (PTEC) damage, closely associated with a progressive decline in renal function. This study was designed to identify a free fatty acid (FFA) metabolism-related enzyme that can protect PTECs from SFA-related lipotoxicity. Among several enzymes involved in FFA metabolism, we identified stearoyl-CoA desaturase-1 (SCD1), whose expression level significantly decreased in the kidneys of high-fat diet (HFD)-induced diabetic mice, compared with non-diabetic mice. SCD1 is an enzyme that desaturates SFAs, converting them to monounsaturated fatty acids (MUFAs), leading to the formation of neutral lipid droplets. In culture, retrovirus-mediated overexpression of SCD1 or MUFA treatment significantly ameliorated SFA-induced apoptosis in PTECs by enhancing intracellular lipid droplet formation. In contrast, siRNA against SCD1 exacerbated the apoptosis. Both overexpression of SCD1 and MUFA treatment reduced SFA-induced apoptosis via reducing endoplasmic reticulum stress in cultured PTECs. Thus, HFD-induced decrease in renal SCD1 expression may play a pathogenic role in lipotoxicity-induced renal injury, and enhancing SCD1-mediated desaturation of SFA and subsequent formation of neutral lipid droplets may become a promising therapeutic target to reduce SFA-induced lipotoxicity. The present study provides a novel insight into lipotoxicity in the pathogenesis of diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/enzymology , Diet, High-Fat , Epithelial Cells/enzymology , Fatty Acids, Monounsaturated/metabolism , Fatty Acids/metabolism , Kidney Tubules, Proximal/enzymology , Stearoyl-CoA Desaturase/metabolism , Animals , Apoptosis/genetics , Blood Glucose/metabolism , Cell Line , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Epithelial Cells/pathology , Fasting , Fatty Acids/toxicity , Gene Expression , HEK293 Cells , Humans , Kidney Tubules, Proximal/pathology , Lipid Droplets/metabolism , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL , Perilipin-2/genetics , Perilipin-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stearoyl-CoA Desaturase/antagonists & inhibitors , Stearoyl-CoA Desaturase/genetics
9.
Biochim Biophys Acta ; 1842(7): 1097-108, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24726883

ABSTRACT

Podocyte apoptosis is a potent mechanism of proteinuria in diabetic nephropathy. More detailed mechanistic insight into podocyte apoptosis is needed to better understand the pathogenesis of diabetic nephropathy. An elevated level of serum free fatty acid (FFA), as well as hyperglycemia, is a clinical characteristic in diabetes, although its causal role in podocyte apoptosis remains unclear. This study examined the effect of three types of FFAs, saturated, monounsaturated and polyunsaturated FFAs, on podocyte apoptosis. Palmitate, a saturated FFA, induced endoplasmic reticulum (ER) stress-dependent apoptosis in podocytes. Oleate, a monounsaturated FFA, and eicosapentaenoic acid (EPA), an ω-3 polyunsaturated FFA did not induce apoptosis; rather, they antagonized palmitate-induced apoptosis. Palmitate activated mammalian target of rapamycin (mTOR) complex 1 (mTORC1), a nutrient-sensing kinase regulating a wide range of cell biology. Furthermore, inhibition of mTORC1 activity by rapamycin or siRNA for Raptor, a component of mTORC1, ameliorated palmitate-induced ER stress and apoptosis in podocytes. Activity of mTORC1 is regulated by upstream kinases and Rag/Ragulator-dependent recruitment of mTOR onto lysosomal membranes. Palmitate activated mTORC1 by enhancing recruitment of mTOR onto lysosomal membranes, which was inhibited by co-incubation with oleate or EPA. Inhibition of mTOR translocation onto lysosomes by transfection with dominant-negative forms of Rag ameliorated palmitate-induced apoptosis. This study suggests that saturated and unsaturated FFAs have opposite effects on podocyte apoptosis by regulating mTORC1 activity via its translocation onto lysosomal membranes, and the results provide a better understanding of the pathogenesis in diabetic nephropathy and a novel role of mTORC1 in cell apoptosis.


Subject(s)
Apoptosis/physiology , Fatty Acids/metabolism , Lysosomes/metabolism , Multiprotein Complexes/metabolism , Podocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Diabetic Nephropathies/metabolism , Eicosapentaenoic Acid/metabolism , Endoplasmic Reticulum Stress/physiology , Food , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Oleic Acid/metabolism , Palmitates/metabolism
10.
Biochem Biophys Res Commun ; 465(2): 249-55, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26271596

ABSTRACT

Autophagy process is essential for maintaining intracellular homeostasis and consists of autophagosome formation and subsequent fusion with lysosome for degradation. Although the role of autophagosome formation in the pathogenesis of diabetes has been recently documented, the role of the latter process remains unclear. This study analyzed high-fat diet (HFD)-fed mice lacking lysosome-associated membrane protein-2 (lamp-2), which is essential for the fusion with lysosome and subsequent degradation of autophagosomes. Although lamp-2 deficient mice showed little alteration in glucose metabolism under normal diet feeding, they showed a resistance against high-fat diet (HFD)-induced obesity, hyperinsulinemic hyperglycemia and tissues lipid accumulation, accompanied with higher energy expenditure. The expression levels of thermogenic genes in brown adipose tissue were significantly increased in HFD-fed lamp-2-deficient mice. Of some serum factors related to energy expenditure, the serum level of fibroblast growth factor (FGF) 21 and its mRNA expression level in the liver were significantly higher in HFD-fed lamp-2-deficient mice in an ER stress-, but not PPARα-, dependent manner. In conclusion, a lamp-2-depenedent fusion and degradation process of autophagosomes is involved in the pathogenesis of obese diabetes, providing a novel insight into autophagy and diabetes.


Subject(s)
Adipose Tissue, Brown/metabolism , Diabetes Mellitus, Experimental/prevention & control , Energy Metabolism/genetics , Glycogen Storage Disease Type IIb/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomes/metabolism , Adipose Tissue, Brown/pathology , Animals , Autophagy/genetics , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Dietary Fats/adverse effects , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression , Glycogen Storage Disease Type IIb/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/genetics , PPAR alpha/metabolism , Phagosomes/metabolism , Phagosomes/pathology , Protective Factors , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Am J Pathol ; 183(3): 774-85, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23871833

ABSTRACT

Aging is a dominant risk factor for end-stage renal disease. We analyzed the mechanism involved in age-related exacerbation of proteinuria-induced proximal tubular cell (PTC) damage by focusing on endoplasmic reticulum-related unfolded protein response (UPR). After equal-degree induction of proteinuria in 24-month-old (aged) and 3-month-old (young) mice by intraperitoneal free fatty acid-bound albumin overload, tubulointerstitial lesions were more severe in aged than in young mice. In aged PTCs, proteinuria-induced cell-adaptive UPR resulting from induction of the molecular chaperone BiP was significantly suppressed, whereas proapoptotic UPR with CHOP overexpression was enhanced. Treatment with the exogenous molecular chaperone tauroursodeoxycholic acid (TUDCA) ameliorated proteinuria-induced tubulointerstitial lesions and PTC apoptosis in aged mice. Among the three UPR branches, alterations in the inositol-requiring 1α (IRE1α) pathway, but not the activating transcription factor 6 or PERK pathway, were associated with impaired BiP induction in aged kidneys. Moreover, siRNA-mediated suppression of BiP and IRE1α exacerbated free fatty acid-bound albumin-induced apoptosis in cultured PTCs, whereas siRNA-mediated CHOP suppression ameliorated apoptosis. Finally, proteinuria-induced BiP induction in PTCs was diminished in kidney specimens from elderly patients. These results indicate that maladaptive UPRs are involved in proteinuria-induced tubulointerstitial lesions exacerbation in aged kidneys, and that supplementation of chaperones may be used to treat elderly patients with persistent proteinuria. These results should improve understanding of cell vulnerability in aged kidneys.


Subject(s)
Aging/pathology , Disease Progression , Kidney Tubules, Proximal/pathology , Proteinuria/complications , Proteinuria/pathology , Unfolded Protein Response , Adult , Aged , Albumins/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/metabolism , Heat-Shock Proteins/metabolism , Humans , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , Molecular Chaperones/metabolism , Palmitates/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Taurochenodeoxycholic Acid/pharmacology , Transcription Factor CHOP/metabolism , Unfolded Protein Response/drug effects
12.
J Am Soc Nephrol ; 24(11): 1769-81, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24092929

ABSTRACT

Obesity is an independent risk factor for renal dysfunction in patients with CKDs, including diabetic nephropathy, but the mechanism underlying this connection remains unclear. Autophagy is an intracellular degradation system that maintains intracellular homeostasis by removing damaged proteins and organelles, and autophagy insufficiency is associated with the pathogenesis of obesity-related diseases. We therefore examined the role of autophagy in obesity-mediated exacerbation of proteinuria-induced proximal tubular epithelial cell damage in mice and in human renal biopsy specimens. In nonobese mice, overt proteinuria, induced by intraperitoneal free fatty acid-albumin overload, led to mild tubular damage and apoptosis, and activated autophagy in proximal tubules reabsorbing urinary albumin. In contrast, diet-induced obesity suppressed proteinuria-induced autophagy and exacerbated proteinuria-induced tubular cell damage. Proximal tubule-specific autophagy-deficient mice, resulting from an Atg5 gene deletion, subjected to intraperitoneal free fatty acid-albumin overload developed severe proteinuria-induced tubular damage, suggesting that proteinuria-induced autophagy is renoprotective. Mammalian target of rapamycin (mTOR), a potent suppressor of autophagy, was activated in proximal tubules of obese mice, and treatment with an mTOR inhibitor ameliorated obesity-mediated autophagy insufficiency. Furthermore, both mTOR hyperactivation and autophagy suppression were observed in tubular cells of specimens obtained from obese patients with proteinuria. Thus, in addition to enhancing the understanding of obesity-related cell vulnerability in the kidneys, these results suggest that restoring the renoprotective action of autophagy in proximal tubules may improve renal outcomes in obese patients.


Subject(s)
Autophagy/physiology , Kidney Tubules, Proximal/pathology , Obesity/complications , Proteinuria/complications , Animals , Cells, Cultured , Diet, High-Fat , Endoplasmic Reticulum Stress , Epithelial Cells/pathology , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Transcription Factor TFIIH , Transcription Factors/metabolism
13.
Kidney360 ; 5(2): 320-326, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38227425

ABSTRACT

Ketone bodies have a negative image because of ketoacidosis, one of the acute and serious complications in diabetes. The negative image persists despite the fact that ketone bodies are physiologically produced in the liver and serve as an indispensable energy source in extrahepatic organs, particularly during long-term fasting. However, accumulating experimental evidence suggests that ketone bodies exert various health benefits. Particularly in the field of aging research, there is growing interest in the potential organoprotective effects of ketone bodies. In addition, ketone bodies have a potential role in preventing kidney diseases, including diabetic kidney disease (DKD), a diabetic complication caused by prolonged hyperglycemia that leads to a decline in kidney function. Ketone bodies may help alleviate the renal burden from hyperglycemia by being used as an alternative energy source in patients with diabetes. Furthermore, ketone body production may reduce inflammation and delay the progression of several kidney diseases in addition to DKD. Although there is still insufficient research on the use of ketone bodies as a treatment and their effects, their renoprotective effects are being gradually proven. This review outlines the ketone body-mediated renoprotective effects in DKD and other kidney diseases.


Subject(s)
Diabetes Complications , Diabetes Mellitus , Diabetic Nephropathies , Hyperglycemia , Ketosis , Humans , Ketone Bodies/metabolism , Ketosis/metabolism
14.
Biochem Biophys Rep ; 37: 101607, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38178924

ABSTRACT

Renal artery stenosis-induced chronic renal ischemia is an important cause of renal dysfunction, especially in older adults, and its incidence is currently increasing. To elucidate the mechanisms underlying chronic renal hypoperfusion-induced kidney damage, we developed a novel mouse model of renal artery coiling-based chronic hypoperfusion-related kidney injury. This model exhibits decreased renal blood flow and function, atrophy, and parenchymal injury in the coiled kidney, along with compensatory hypertrophy in the non-coiled kidney, without chronic hypertension. The availability of this mouse model, which can develop renal ischemia without genetic modification, will enhance kidney disease research by serving as a new tool to investigate the effects of acquired factors (e.g., obesity and aging) and genetic factors on renal artery stenosis-related renal parenchymal damage.

15.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167074, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38354758

ABSTRACT

For the advancement of DKD treatment, identifying unrecognized residual risk factors is essential. We explored the impact of obesity diversity derived from different carbohydrate qualities, with an emphasis on the increasing trend of excessive fructose consumption and its effect on DKD progression. In this study, we utilized db/db mice to establish a novel diabetic model characterized by fructose overconsumption, aiming to uncover the underlying mechanisms of renal damage. Compared to the control diet group, the fructose-fed db/db mice exhibited more pronounced obesity yet demonstrated milder glucose intolerance. Plasma cystatin C levels were elevated in the fructose model compared to the control, and this elevation was accompanied by enhanced glomerular sclerosis, even though albuminuria levels and tubular lesions were comparable. Single-cell RNA sequencing of the whole kidney highlighted an increase in Lrg1 in glomerular endothelial cells (GECs) in the fructose model, which appeared to drive mesangial fibrosis through enhanced TGF-ß1 signaling. Our findings suggest that excessive fructose intake exacerbates diabetic kidney disease progression, mediated by aberrant Lrg1-driven crosstalk between GECs and mesangial cells.


Subject(s)
Diabetic Nephropathies , Mesangial Cells , Mice , Animals , Endothelial Cells/pathology , Fructose/adverse effects , Diabetic Nephropathies/pathology , Mice, Inbred Strains , Obesity/complications , Cell Communication
16.
Aging Cell ; 22(6): e13833, 2023 06.
Article in English | MEDLINE | ID: mdl-37060184

ABSTRACT

Accumulating evidence suggests health benefits of ketone bodies, and especially for longevity. However, the precise role of endogenous ketogenesis in mammalian life span, and the safety and efficacy of the long-term exogenous supplementation of ketone bodies remain unclear. In the present study, we show that a deficiency in endogenous ketogenesis, induced by whole-body Hmgcs2 deletion, shortens life span in mice, and that this is prevented by daily ketone body supplementation using a diet containing 1,3-butanediol, a precursor of ß-hydroxybutyrate. Furthermore, feeding the 1,3-butanediol-containing diet from early in life increases midlife mortality in normal mice, but in aged mice it extends life span and prevents the high mortality associated with atherosclerosis in ApoE-deficient mice. By contrast, an ad libitum low-carbohydrate ketogenic diet markedly increases mortality. In conclusion, endogenous ketogenesis affects mammalian survival, and ketone body supplementation may represent a double-edged sword with respect to survival, depending on the method of administration and health status.


Subject(s)
Ketone Bodies , Longevity , Mice , Animals , Butylene Glycols , 3-Hydroxybutyric Acid , Mammals
17.
Biochem Biophys Res Commun ; 419(2): 244-9, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22342673

ABSTRACT

Fructose induces several kinds of human metabolic disorders; however, information regarding fructose-induced kidney injury is still limited. This study examined fructose-induced kidney injury in mice and clarified the differential susceptibility of three mouse strains: C57Bl/6J, CBA/JN and DBA/2N. In this study all mice were fed with an equal calorie count for sixteen weeks to remove the influence of total energy intake from metabolic effects by fructose-feeding. Only DBA/2N mice, but not C57Bl/6J and CBA/JN mice, fed with fructose displayed tubulointerstitial fibrosis localized on the outer cortex of the kidney together with the increase of mRNA expression of Kim1 and Ngal in the absence of distinct glomerular lesions and albuminuria - decidedly different from diabetic nephropathy. In time-course study of DBA/2N mice fed with fructose diet, the inflammation and fibrosis in the outer cortex of the kidney were enhancing after eight weeks, in parallel with the accumulation of oxidative stress. This progression of renal damage in DBA/2N mice was accompanied with increasing mRNA expression of GLUT5. These results suggest that the responsiveness of GLUT5 expression to fructose at the kidney is one of pivotal roles for the progression of fructose-induced kidney injury.


Subject(s)
Dietary Carbohydrates/adverse effects , Fructose/adverse effects , Kidney Diseases/chemically induced , Kidney Tubules/drug effects , Acute-Phase Proteins/genetics , Animals , Collagen Type I/genetics , Dietary Carbohydrates/administration & dosage , Fibronectins/genetics , Fructose/administration & dosage , Fructose/metabolism , Gene Expression/drug effects , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 2/genetics , Glucose Transporter Type 5 , Hepatitis A Virus Cellular Receptor 1 , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Lipocalin-2 , Lipocalins/genetics , Liver/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , Receptors, CCR2/genetics
18.
Biochem Biophys Res Commun ; 418(3): 559-64, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22293193

ABSTRACT

Salt-sensitive hypertension is a characteristic of the metabolic syndrome. Given the links to cardiovascular events, the mechanisms underlying sodium metabolism may represent an important therapeutic target for this disorder. Angiotensin II (AII) is a key peptide underlying sodium retention. However, 5'AMP-activated protein kinase (AMPK) has also been reported to participate in the regulation of ion transport. In this study we examined the relationship between AII and AMPK on the development of hypertension in two salt-sensitive mouse models. In the first model, the mice were maintained on a high-fat diet (HFD) for 12 weeks, in order to develop features similar to the metabolic syndrome, including salt-sensitive hypertension. HFD-induced obese mice showed elevated systolic blood pressure and lower sodium excretion in response to salt loading, along with an increase in AII contents and inactivation of AMPK in the kidney, which were significantly improved by the treatment of an angiotensin II antagonist, losartan, for 2 weeks. To clarify the effects of AII, a second group of mice was infused with AII via an osmotic pump, which led to higher systolic blood pressure, and decreases in urinary sodium excretion and the expression of AMPK, in a manner similar to those observed in the HFD mice. However, treatment with an AMPK activator, metformin, improved the changes induced by the AII, suggesting that AII induced sodium retention works by acting on AMPK activity. Finally, we evaluated the changes in salt-sensitivity by performing 2-week salt loading experiments with or without metformin. AII infusion elevated blood pressure by salt loading but metformin prevented it. These findings indicate that AII suppresses AMPK activity in the kidney, leading to sodium retention and enhanced salt-sensitivity, and that AMPK activation may represent a new therapeutic target for obesity-related salt-sensitive hypertension.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Angiotensin II/metabolism , Hypertension/etiology , Kidney/metabolism , Obesity/complications , Sodium/metabolism , Angiotensin II/administration & dosage , Angiotensin II/antagonists & inhibitors , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Blood Pressure/drug effects , Diet, High-Fat/adverse effects , Disease Models, Animal , Hypertension/drug therapy , Hypertension/metabolism , Kidney/drug effects , Losartan/pharmacology , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Metformin/therapeutic use , Mice , Mice, Inbred C57BL , Obesity/metabolism , Phosphorylation/drug effects , Sodium, Dietary/adverse effects
19.
Biochim Biophys Acta Mol Basis Dis ; 1868(5): 166368, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35202791

ABSTRACT

AIMS: Identifying the mechanisms that underlie progression from endothelial damage to podocyte damage, which leads to massive proteinuria, is an urgent issue that must be clarified to improve renal outcome in diabetic kidney disease (DKD). We aimed to examine the role of dynamin-related protein 1 (Drp1)-mediated regulation of mitochondrial fission in podocytes in the pathogenesis of massive proteinuria in DKD. METHODS: Diabetes- or albuminuria-associated changes in mitochondrial morphology in podocytes were examined by electron microscopy. The effects of albumin and other diabetes-related stimuli, including high glucose (HG), on mitochondrial morphology were examined in cultured podocytes. The role of Drp1 in podocyte damage was examined using diabetic podocyte-specific Drp1-deficient mice treated with neuraminidase, which removes endothelial glycocalyx. RESULTS: Neuraminidase-induced removal of glomerular endothelial glycocalyx in nondiabetic mice led to microalbuminuria without podocyte damage, accompanied by reduced Drp1 expression and mitochondrial elongation in podocytes. In contrast, streptozotocin-induced diabetes significantly exacerbated neuraminidase-induced podocyte damage and albuminuria, and was accompanied by increased Drp1 expression and enhanced mitochondrial fission in podocytes. Cell culture experiments showed that albumin stimulation decreased Drp1 expression and elongated mitochondria, although HG inhibited albumin-associated changes in mitochondrial dynamics, resulting in apoptosis. Podocyte-specific Drp1-deficiency in mice prevented diabetes-related exacerbation of podocyte damage and neuraminidase-induced development of albuminuria. Endothelial dysfunction-induced albumin exposure is cytotoxic to podocytes. Inhibition of mitochondrial fission in podocytes is a cytoprotective mechanism against albumin stimulation, which is impaired under diabetic condition. Inhibition of mitochondrial fission in podocytes may represent a new therapeutic strategy for massive proteinuria in DKD.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Podocytes , Albumins/metabolism , Albumins/pharmacology , Albuminuria/genetics , Albuminuria/metabolism , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/pathology , Female , Humans , Male , Mice , Mitochondrial Dynamics , Neuraminidase/metabolism , Podocytes/metabolism , Proteinuria/metabolism , Proteinuria/pathology
20.
Intern Med ; 61(20): 3077-3081, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-35283375

ABSTRACT

Fabry disease is an inherited lysosomal disorder caused by mutations in the alpha-galactosidase A gene. We herein report a Fabry disease patient with enzyme replacement therapy (ERT)-resistant proteinuria who showed improvement in the estimated glomerular filtration rate (eGFR) decline rate after uric acid (UA)-lowering therapy. The patient was diagnosed with Fabry disease at 36 years old. After that, even under ERT, proteinuria and eGFR decline persisted. During the clinical course, serum UA levels were elevated with increases in renal tubular damage markers. Febuxostat administration immediately improved tubular damage and prevented further eGFR decline. UA-mediated tubulopathy may become an additional therapeutic target for eGFR decline in Fabry disease.


Subject(s)
Fabry Disease , Hyperuricemia , Adult , Enzyme Replacement Therapy , Fabry Disease/complications , Fabry Disease/drug therapy , Febuxostat/therapeutic use , Glomerular Filtration Rate , Humans , Hyperuricemia/drug therapy , Proteinuria/drug therapy , Proteinuria/etiology , Treatment Outcome , Uric Acid , alpha-Galactosidase/genetics , alpha-Galactosidase/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL