Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Kidney360 ; 3(8): 1394-1410, 2022 08 25.
Article En | MEDLINE | ID: mdl-36176646

Background: PCSK9 modulates the uptake of circulating lipids through a range of receptors, including the low-density lipoprotein receptor (LDLR) and CD36. In the kidney, CD36 is known to contribute to renal injury through pro-inflammatory and -fibrotic pathways. In this study, we sought to investigate the role of PCSK9 in modulating renal lipid accumulation and injury through CD36 using a high fat diet (HFD)-induced murine model. Methods: The effect of PCSK9 on the expression of CD36 and intracellular accumulation of lipid was examined in cultured renal cells and in the kidneys of male C57BL/6J mice. The effect of these findings was subsequently explored in a model of HFD-induced renal injury in Pcsk9 -/- and Pcsk9 +/+ littermate control mice on a C57BL/6J background. Results: In the absence of PCSK9, we observed heightened CD36 expression levels, which increased free fatty acid (FFA) uptake in cultured renal tubular cells. As a result, PCSK9 deficiency was associated with an increase in long-chain saturated FFA-induced ER stress. Consistent with these observations, Pcsk9-/- mice fed a HFD displayed elevated ER stress, inflammation, fibrosis, and renal injury relative to HFD-fed control mice. In contrast to Pcsk9-/- mice, pretreatment of WT C57BL/6J mice with evolocumab, an anti-PCSK9 monoclonal antibody (mAb) that binds to and inhibits the function of circulating PCSK9, protected against HFD-induced renal injury in association with reducing cell surface CD36 expression on renal epithelia. Conclusions: We report that circulating PCSK9 modulates renal lipid uptake in a manner dependent on renal CD36. In the context of increased dietary fat consumption, the absence of circulating PCSK9 may promote renal lipid accumulation and subsequent renal injury. However, although the administration of evolocumab blocks the interaction of PCSK9 with the LDLR, this evolocumab/PCSK9 complex can still bind CD36, thereby protecting against HFD-induced renal lipotoxicity.


CD36 Antigens , Fatty Acids, Nonesterified , Animals , Antibodies, Monoclonal/pharmacology , Diet, High-Fat/adverse effects , Dietary Fats , Fibrosis , Kidney/metabolism , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Proprotein Convertase 9/genetics
2.
PLoS One ; 16(11): e0260519, 2021.
Article En | MEDLINE | ID: mdl-34847196

Endoplasmic reticulum (ER) stress is associated with acute kidney injury (AKI) caused by various mechanisms, including antibiotics, non-steroidal anti-inflammatory drugs, cisplatin, and radiocontrast. Tunicamycin (TM) is a nucleoside antibiotic that induces ER stress and is a commonly used model of AKI. 4-phenylbutyrate (4-PBA) is a chemical chaperone and histone deacetylase (HDAC) inhibitor and has been shown to protect the kidney from ER stress, apoptosis, and structural damage in a tunicamycin model of AKI. The renal protection provided by 4-PBA is attributed to its ability to prevent misfolded protein aggregation and inhibit ER stress; however, the HDAC inhibitor effects of 4-PBA have not been examined in the TM-induced model of AKI. As such, the main objective of this study was to determine if histone hyperacetylation provides any protective effects against TM-mediated AKI. The FDA-approved HDAC inhibitor vorinostat was used, as it has no ER stress inhibitory effects and therefore the histone hyperacetylation properties alone could be investigated. In vitro work demonstrated that vorinostat inhibited histone deacetylation in cultured proximal tubular cells but did not prevent ER stress or protein aggregation induced by TM. Vorinostat induced a significant increase in cell death, and exacerbated TM-mediated total cell death and apoptotic cell death. Wild type male mice were treated with TM (0.5 mg/kg, intraperitoneal injection), with or without vorinostat (50 mg/kg/day) or 4-PBA (1 g/kg/day). Mice treated with 4-PBA or vorinostat exhibited similar levels of histone hyperacetylation. Expression of the pro-apoptotic protein CHOP was induced with TM, and not inhibited by vorinostat. Further, vorinostat did not prevent any renal damage or decline in renal function caused by tunicamycin. These data suggest that the protective mechanisms found by 4-PBA are primarily due to its molecular chaperone properties, and the HDAC inhibitors used did not provide any protection against renal injury caused by ER stress.


Acute Kidney Injury , Endoplasmic Reticulum Stress/drug effects , Histone Deacetylase Inhibitors/pharmacology , Tunicamycin/adverse effects , Vorinostat/pharmacology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Animals , Cell Line , Disease Models, Animal , Male , Mice , Protein Aggregation, Pathological/chemically induced , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/prevention & control , Tunicamycin/pharmacology
3.
Cell Death Dis ; 12(10): 921, 2021 10 08.
Article En | MEDLINE | ID: mdl-34625532

Chronic kidney disease (CKD) is characterized by the gradual loss of renal function and is a major public health concern. Risk factors for CKD include hypertension and proteinuria, both of which are associated with endoplasmic reticulum (ER) stress. ER stress-induced TDAG51 protein expression is increased at an early time point in mice with CKD. Based on these findings, wild-type and TDAG51 knock-out (TDKO) mice were used in an angiotensin II/deoxycorticosterone acetate/salt model of CKD. Both wild-type and TDKO mice developed hypertension, increased proteinuria and albuminuria, glomerular injury, and tubular damage. However, TDKO mice were protected from apoptosis and renal interstitial fibrosis. Human proximal tubular cells were used to demonstrate that TDAG51 expression induces apoptosis through a CHOP-dependent mechanism. Further, a mouse model of intrinsic acute kidney injury demonstrated that CHOP is required for ER stress-mediated apoptosis. Renal fibroblasts were used to demonstrate that TGF-ß induces collagen production through an IRE1-dependent mechanism; cells treated with a TGF-ß receptor 1 inhibitor prevented XBP1 splicing, a downstream consequence of IRE1 activation. Interestingly, TDKO mice express significantly less TGF-ß receptor 1, thus, preventing TGF-ß-mediated XBP1 splicing. In conclusion, TDAG51 induces apoptosis in the kidney through a CHOP-dependent mechanism, while contributing to renal interstitial fibrosis through a TGF-ß-IRE1-XBP1 pathway.


Kidney/pathology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Blood Pressure/drug effects , Cell Line , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Humans , Hymecromone/analogs & derivatives , Hymecromone/pharmacology , Kidney/drug effects , Kidney/physiopathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Rats , Renal Insufficiency, Chronic/physiopathology , Risk Factors , Transcription Factor CHOP/metabolism , Tunicamycin/pharmacology , X-Box Binding Protein 1/metabolism
4.
Am J Physiol Heart Circ Physiol ; 316(5): H1214-H1223, 2019 05 01.
Article En | MEDLINE | ID: mdl-30848678

Essential hypertension is the leading cause of premature death worldwide. However, hypertension's cause remains uncertain. endoplasmic reticulum (ER) stress has recently been associated with hypertension, but it is unclear whether ER stress causes hypertension. To clarify this question, we examined if ER stress occurs in blood vessels before the development of hypertension and if ER stress inhibition would prevent hypertension development. We used the spontaneously hypertensive rat (SHR) as a model of human essential hypertension and the Wistar-Kyoto (WKY) rat as its normotensive control. Resistance arteries collected from young rats determined that ER stress was present in SHR vessels before the onset of hypertension. To assess the effect of ER stress inhibition on hypertension development, another subset of rats were treated with 4-phenylbutyric acid (4-PBA; 1 g·kg-1·day-1) for 8 wk from 5 wk of age. Blood pressure was measured via radiotelemetry and compared with untreated SHR and WKY rats. Mesenteric resistance arteries were collected and assessed for structural and functional changes associated with hypertension. Systolic and diastolic blood pressures were significantly lower in the 4-PBA-treated SHR groups than in untreated SHRs. Additionally, 4-PBA significantly decreased the media-to-lumen ratio and ER stress marker expression, improved vasodilatory response, and reduced contractile responses in resistance arteries from SHRs. Overall, ER stress inhibition blunted the development of hypertension in the SHR. These data add evidence to the hypothesis that a component of hypertension in the SHR is caused by ER stress. NEW & NOTEWORTHY In this study, 4-phenylbutyric acid's (4-PBA's) molecular chaperone capability was used to inhibit endoplasmic reticulum (ER) stress in the small arteries of young spontaneously hypertensive rats (SHRs) and reduce their hypertension. These effects are likely mediated through 4-PBA's effects to reduce resistant artery contractility and increase nitric oxide-mediated endothelial vasodilation through a process preventing endothelial dysfunction. Overall, ER stress inhibition blunted the development of hypertension in this young SHR model. This suggests that a component of the increase in blood pressure found in SHRs is due to ER stress. However, it is important to note that inhibition of ER stress was not able to fully restore the blood pressure to normal, suggesting that a component of hypertension may not be due to ER stress. This study points to the inhibition of ER stress as an important new physiological pathway to lower blood pressure, where other known approaches may not achieve blood pressure-lowering targets.


Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Endoplasmic Reticulum Stress/drug effects , Essential Hypertension/prevention & control , Mesenteric Arteries/drug effects , Phenylbutyrates/pharmacology , Animals , Biomarkers/blood , Disease Models, Animal , Essential Hypertension/metabolism , Essential Hypertension/physiopathology , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Rats, Inbred SHR , Rats, Inbred WKY , Vascular Resistance/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects
5.
Biochem Biophys Res Commun ; 486(1): 163-170, 2017 04 22.
Article En | MEDLINE | ID: mdl-28285140

Newly translated proteins must undergo proper folding to ensure their function. To enter a low energy state, misfolded proteins form aggregates, which are associated with many degenerative diseases, such as Huntington's disease and chronic kidney disease (CKD). Recent studies have shown the use of low molecular weight chemical chaperones to be an effective method of reducing protein aggregation in various cell types. This study demonstrates a novel non-biased assay to assess the molecular efficacy of these compounds at preventing protein misfolding and/or aggregation. This assay utilizes a thioflavin T fluorescent stain to provide a qualitative and quantitative measure of protein misfolding within cells. The functionality of this method was first assessed in renal proximal tubule epithelial cells treated with various endoplasmic reticulum (ER) stress inducers. Once established in the renal model system, we analyzed the ability of some known chemical chaperones to reduce ER stress. A total of five different compounds were selected: 4-phenylbutyrate (4-PBA), docosahexaenoic acid (DHA), tauroursodeoxycholic acid, trehalose, and glycerol. The dose-dependent effects of these compounds at reducing thapsigargin-induced ER stress was then analyzed, and used to determine their EC50 values. Of the chaperones, 4-PBA and DHA provided the greatest reduction of ER stress and did so at relatively low concentrations. Upon analyzing the efficiency of these compounds and their corresponding structures, it was determined that chaperones with a localized hydrophilic, polar end followed by a long hydrophobic chain, such as 4-PBA and DHA, were most effective at reducing ER stress. This study provides some insight into the use of low molecular weight chemical chaperones and may serve as the first step towards developing new chaperones of greater potency thereby providing potential treatments for diseases caused by protein aggregation.


Epithelial Cells/drug effects , Protein Aggregates/drug effects , Protein Aggregation, Pathological/prevention & control , Xenobiotics/pharmacology , Benzothiazoles , Cell Line , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/pharmacology , Endoplasmic Reticulum Stress/drug effects , Epithelial Cells/metabolism , Glycerol/chemistry , Glycerol/pharmacology , Humans , Kidney Tubules, Proximal/cytology , Molecular Weight , Phenylbutyrates/chemistry , Phenylbutyrates/pharmacology , Protein Folding/drug effects , Staining and Labeling/methods , Taurochenodeoxycholic Acid/chemistry , Taurochenodeoxycholic Acid/pharmacology , Thapsigargin/pharmacology , Thiazoles/chemistry , Trehalose/chemistry , Trehalose/pharmacology , Unfolded Protein Response/drug effects , Xenobiotics/chemistry
6.
Sci Rep ; 7: 41572, 2017 02 02.
Article En | MEDLINE | ID: mdl-28148966

Endoplasmic reticulum (ER) stress is implicated in chronic kidney disease (CKD) development in patients and in animal models. Here we show that ER stress inhibition through 4-phenylbutyric acid (4-PBA) administration decreases blood pressure, albuminuria, and tubular casts in an angiotensin II/deoxycorticosterone acetate/salt murine model of CKD. Lower albuminuria in 4-PBA-treated mice was associated with higher levels of cubilin protein in renal tissue membrane fractions. 4-PBA decreased renal interstitial fibrosis, renal CD3+ T-cell and macrophage infiltration, mRNA expression of TGFß1, Wnt signaling molecules, and ER stress-induced pro-inflammatory genes. CHOP deficient mice that underwent this model of CKD developed hypertension comparable to wild type mice, but had less albuminuria and tubular casts. CHOP deficiency resulted in higher nephrin levels and decreased glomerulosclerosis compared to wild type mice; this effect was accompanied by lower macrophage infiltration and fibrosis. Our findings portray ER stress inhibition as a means to alleviate hypertensive CKD by preserving glomerular barrier integrity and tubular function. These results demonstrate ER stress modulation as a novel target for preserving renal function in hypertensive CKD.


Endoplasmic Reticulum Stress/drug effects , Hypertension/etiology , Hypertension/metabolism , Proteinuria/etiology , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Angiotensin II/metabolism , Animals , Apoptosis/genetics , Biopsy , Blood Pressure/drug effects , Disease Models, Animal , Fibrosis , Gene Expression Profiling , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Nephrosclerosis/etiology , Nephrosclerosis/metabolism , Nephrosclerosis/pathology , Phenylbutyrates/pharmacology , Proteinuria/drug therapy , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Transcription Factor CHOP/deficiency , Transcriptome , Urinalysis
7.
Am J Physiol Renal Physiol ; 312(1): F230-F244, 2017 01 01.
Article En | MEDLINE | ID: mdl-28069662

Proteinuria is one of the primary risk factors for the progression of chronic kidney disease (CKD) and has been implicated in the induction of endoplasmic reticulum (ER) stress. We hypothesized that the suppression of ER stress with a low molecular weight chemical chaperone, 4-phenylbutyric acid (4-PBA), would reduce the severity of CKD and proteinuria in the Dahl salt-sensitive (SS) hypertensive rat. To induce hypertension and CKD, 12-wk-old male rats were placed on a high-salt (HS) diet for 4 wk with or without 4-PBA treatment. We assessed blood pressure and markers of CKD, including proteinuria, albuminuria, and renal pathology. Furthermore, we determined if HS feeding resulted in an impaired myogenic response, subsequent to ER stress. 4-PBA treatment reduced salt-induced hypertension, proteinuria, and albuminuria and preserved myogenic constriction. Furthermore, renal pathology was reduced with 4-PBA treatment, as indicated by lowered expression of profibrotic markers and fewer intratubular protein casts. In addition, ER stress in the glomerulus was reduced, and the integrity of the glomerular filtration barrier was preserved. These results suggest that 4-PBA treatment protects against proteinuria in the SS rat by preserving the myogenic response and by preventing ER stress, which led to a breakdown in the glomerular filtration barrier. As such, alleviating ER stress serves as a viable therapeutic strategy to preserve kidney function and to delay the progression of CKD in the animal model under study.


Endoplasmic Reticulum Stress/drug effects , Phenylbutyrates/pharmacology , Renal Insufficiency, Chronic/drug therapy , Sodium Chloride, Dietary/pharmacology , Sodium Chloride/metabolism , Animals , Blood Pressure/drug effects , Disease Models, Animal , Disease Progression , Humans , Hypertension/drug therapy , Hypertension/physiopathology , Kidney/drug effects , Kidney/metabolism , Proteinuria/metabolism , Rats , Renal Insufficiency, Chronic/pathology
8.
J Hypertens ; 34(8): 1556-69, 2016 08.
Article En | MEDLINE | ID: mdl-27115336

OBJECTIVE: Our purpose was to determine if endoplasmic reticulum stress inhibition lowers blood pressure (BP) in hypertension by correcting vascular dysfunction. METHODS: The spontaneously hypertensive rat (SHR) was used as a model of human essential hypertension with its normotensive control, the Wistar Kyoto rat. Animals were subjected to endoplasmic reticulum stress inhibition with 4-phenylbutyric acid (4-PBA; 1 g/kg per day, orally) for 5 weeks from 12 weeks of age. BP was measured weekly noninvasively and at endpoint with carotid arterial cannulation. Small mesenteric arteries were removed for vascular studies. Function was assessed with a Mulvany-Halpern style myograph, and structure was assessed by measurement of medial-to-lumen ratio in perfusion fixed vessels as well as three-dimensional confocal reconstruction of vessel wall components. Endoplasmic reticulum stress was assessed by quantitative real time-PCR and western blotting; oxidative stress was assessed by 3-nitrotyrosine and dihydroethidium staining. RESULTS: 4-PBA significantly lowered BP in SHR (vehicle 206.1 ±â€Š4.3 vs. 4-PBA 178.9 ±â€Š3.1, systolic) but not Wistar Kyoto. 4-PBA diminished contractility and augmented endothelial-dependent vasodilation in SHR small mesenteric arteries, as well as reducing media-to-lumen ratio. 4-PBA significantly reduced endoplasmic reticulum stress in SHR resistance vessels. Normotensive resistance vessels, treated with the endoplasmic reticulum stress-inducing agent, tunicamycin, show decreased endothelial-dependent vasodilation; this was improved with 4-PBA treatment. 3-Nitrotyrosine and dihydroethidium staining indicated that endoplasmic reticulum stress leads to reactive oxygen species generation resolvable by 4-PBA treatment. CONCLUSION: Endoplasmic reticulum stress caused endothelial-mediated vascular dysfunction contributing to elevated BP in the SHR model of human essential hypertension.


Blood Pressure/drug effects , Endoplasmic Reticulum Stress/drug effects , Hypertension/physiopathology , Phenylbutyrates/pharmacology , Animals , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/pathology , Mesenteric Arteries/physiopathology , Muscle Contraction , Muscle, Smooth, Vascular/drug effects , Oxidative Stress/drug effects , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Reactive Oxygen Species/metabolism , Tunicamycin/pharmacology , Vascular Resistance , Vasodilation/drug effects
9.
Biomed Res Int ; 2015: 172302, 2015.
Article En | MEDLINE | ID: mdl-26064882

Chronic kidney disease (CKD) is a major healthcare problem with increasing prevalence in the population. CKD leads to end stage renal disease and increases the risk of cardiovascular disease. As such, it is important to study the mechanisms underlying CKD progression. To this end, an animal model was developed to allow the testing of new treatment strategies or molecular targets for CKD prevention. Many underlying risk factors result in CKD but the disease itself has common features, including renal interstitial fibrosis, tubular epithelial cell loss through apoptosis, glomerular damage, and renal inflammation. Further, CKD shows differences in prevalence between the genders with premenopausal women being relatively resistant to CKD. We sought to develop and characterize an animal model with these common features of human CKD in the C57BL/6 mouse. Mice of this genetic background have been used to produce transgenic strains that are commercially available. Thus, a CKD model in this strain would allow the testing of the effects of numerous genes on the severity or progression of CKD with minimal cost. This paper describes such a mouse model of CKD utilizing angiotensin II and deoxycorticosterone acetate as inducers.


Disease Models, Animal , Disease Progression , Renal Insufficiency, Chronic/physiopathology , Angiotensin II/administration & dosage , Animals , Desoxycorticosterone Acetate/administration & dosage , Female , Humans , Male , Mice , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/genetics
10.
PLoS One ; 9(1): e84663, 2014.
Article En | MEDLINE | ID: mdl-24416259

Different forms of acute kidney injury (AKI) have been associated with endoplasmic reticulum (ER) stress; these include AKI caused by acetaminophen, antibiotics, cisplatin, and radiocontrast. Tunicamycin (TM) is a nucleoside antibiotic known to induce ER stress and is a commonly used inducer of AKI. 4-phenylbutyrate (4-PBA) is an FDA approved substance used in children who suffer from urea cycle disorders. 4-PBA acts as an ER stress inhibitor by aiding in protein folding at the molecular level and preventing misfolded protein aggregation. The main objective of this study was to determine if 4-PBA could protect from AKI induced by ER stress, as typified by the TM-model, and what mechanism(s) of 4-PBA's action were responsible for protection. C57BL/6 mice were treated with saline, TM or TM plus 4-PBA. 4-PBA partially protected the anatomic segment most susceptible to damage, the outer medullary stripe, from TM-induced AKI. In vitro work showed that 4-PBA protected human proximal tubular cells from apoptosis and TM-induced CHOP expression, an ER stress inducible proapoptotic gene. Further, immunofluorescent staining in the animal model found similar protection by 4-PBA from CHOP nuclear translocation in the tubular epithelium of the medulla. This was accompanied by a reduction in apoptosis and GRP78 expression. CHOP(-/-) mice were protected from TM-induced AKI. The protective effects of 4-PBA extended to the ultrastructural integrity of proximal tubule cells in the outer medulla. When taken together, these results indicate that 4-PBA acts as an ER stress inhibitor, to partially protect the kidney from TM-induced AKI through the repression of ER stress-induced CHOP expression.


Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Cytoprotection/drug effects , Down-Regulation/drug effects , Phenylbutyrates/pharmacology , Transcription Factor CHOP/metabolism , Tunicamycin/adverse effects , Acute Kidney Injury/chemically induced , Animals , Cell Line , Endoplasmic Reticulum Chaperone BiP , Humans , Mice , Mice, Inbred C57BL
11.
Am J Physiol Renal Physiol ; 303(3): F467-81, 2012 Aug 01.
Article En | MEDLINE | ID: mdl-22592641

Epithelial-to-mesenchymal transition (EMT) contributes to renal fibrosis in chronic kidney disease. Endoplasmic reticulum (ER) stress, a feature of many forms of kidney disease, results from the accumulation of misfolded proteins in the ER and leads to the unfolded protein response (UPR). We hypothesized that ER stress mediates EMT in human renal proximal tubules. ER stress is induced by a variety of stressors differing in their mechanism of action, including tunicamycin, thapsigargin, and the calcineurin inhibitor cyclosporine A. These ER stressors increased the UPR markers GRP78, GRP94, and phospho-eIF2α in human proximal tubular cells. Thapsigargin and cyclosporine A also increased cytosolic Ca(2+) concentration and T cell death-associated gene 51 (TDAG51) expression, whereas tunicamycin did not. Thapsigargin was also shown to increase levels of active transforming growth factor (TGF)-ß1 in the media of cultured human proximal tubular cells. Thapsigargin induced cytoskeletal rearrangement, ß-catenin nuclear translocation, and α-smooth muscle actin and vinculin expression in proximal tubular cells, indicating an EMT response. Subconfluent primary human proximal tubular cells were induced to undergo EMT by TGF-ß1 treatment. In contrast, tunicamycin treatment did not produce an EMT response. Plasmid-mediated overexpression of TDAG51 resulted in cell shape change and ß-catenin nuclear translocation. These results allowed us to develop a two-hit model of ER stress-induced EMT, where Ca(2+) dysregulation-mediated TDAG51 upregulation primes the cell for mesenchymal transformation via Wnt signaling and then TGF-ß1 activation leads to a complete EMT response. Thus the release of Ca(2+) from ER stores mediates EMT in human proximal tubular epithelium via the induction of TDAG51.


Epithelium/metabolism , Kidney Tubules, Proximal/metabolism , Mesoderm/metabolism , Transcription Factors/physiology , Animals , Calcium/metabolism , Cell Line , Cell Shape , Cells, Cultured , Chelating Agents/pharmacology , Cytosol/drug effects , Cytosol/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Enzyme-Linked Immunosorbent Assay , Fibrosis , Humans , Indicators and Reagents , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Plasmids/genetics , Spectrometry, Fluorescence , Transcription Factors/genetics , Transfection , Transforming Growth Factor beta1/biosynthesis , Up-Regulation/drug effects , Up-Regulation/physiology , beta Catenin/metabolism
12.
Am J Physiol Renal Physiol ; 303(2): F266-78, 2012 Jul 15.
Article En | MEDLINE | ID: mdl-22573382

Renal proximal tubule injury is induced by agents/conditions known to cause endoplasmic reticulum (ER) stress, including cyclosporine A (CsA), an immunosuppressant drug with nephrotoxic effects. However, the underlying mechanism by which ER stress contributes to proximal tubule cell injury is not well understood. In this study, we report lipid accumulation, sterol regulatory element-binding protein-2 (SREBP-2) expression, and ER stress in proximal tubules of kidneys from mice treated with the classic ER stressor tunicamycin (Tm) or in human renal biopsy specimens showing CsA-induced nephrotoxicity. Colocalization of ER stress markers [78-kDa glucose regulated protein (GRP78), CHOP] with SREBP-2 expression and lipid accumulation was prominent within the proximal tubule cells exposed to Tm or CsA. Prolonged ER stress resulted in increased apoptotic cell death of lipid-enriched proximal tubule cells with colocalization of GRP78, SREBP-2, and Ca(2+)-independent phospholipase A(2) (iPLA(2)ß), an SREBP-2 inducible gene with proapoptotic characteristics. In cultured HK-2 human proximal tubule cells, CsA- and Tm-induced ER stress caused lipid accumulation and SREBP-2 activation. Furthermore, overexpression of SREBP-2 or activation of endogenous SREBP-2 in HK-2 cells stimulated apoptosis. Inhibition of SREBP-2 activation with the site-1-serine protease inhibitor AEBSF prevented ER stress-induced lipid accumulation and apoptosis. Overexpression of the ER-resident chaperone GRP78 attenuated ER stress and inhibited CsA-induced SREBP-2 expression and lipid accumulation. In summary, our findings suggest that ER stress-induced SREBP-2 activation contributes to renal proximal tubule cell injury by dysregulating lipid homeostasis.


Apoptosis/physiology , Endoplasmic Reticulum/physiology , Kidney Tubules, Proximal/physiopathology , Lipid Metabolism/physiology , Sterol Regulatory Element Binding Protein 2/metabolism , Stress, Physiological/physiology , Animals , Apoptosis/drug effects , Biopsy , Cells, Cultured , Cyclosporine/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Homeostasis/physiology , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Lipid Metabolism/drug effects , Mice , Mice, Inbred C57BL , Sterol Regulatory Element Binding Protein 2/drug effects , Stress, Physiological/drug effects , Tunicamycin/pharmacology
13.
J Biol Chem ; 287(10): 7603-14, 2012 Mar 02.
Article En | MEDLINE | ID: mdl-22215680

The integrated stress response mediated by eukaryotic translation initiation factor 2α (eIF2α) phosphorylation maintains cellular homeostasis under endoplasmic reticulum (ER) stress. eIF2α phosphorylation induces activating transcription factor 4 (ATF4), a basic leucine zipper transcription factor that regulates the expression of genes responsible for amino acid metabolism, cellular redox state, and anti-stress responses. Cystathionine γ-lyase (CSE) and cystathionine ß-synthase are critical enzymes in the transsulfuration pathway, which also regulate cellular redox status by modulating glutathione (GSH) levels. To determine the link between the integrated stress response and the transsulfuration pathway, we used homocysteine (Hcy) as an inducer of eIF2α phosphorylation and ATF4 gene induction. Mouse embryonic fibroblasts (MEFs) lacking ATF4 (ATF4(-/-)) had reduced GSH levels and increased reactive oxygen species and were susceptible to apoptotic cell death under normal culture conditions. Further, ATF4(-/-) MEFs were more sensitive to Hcy-induced cytotoxicity and showed significantly reduced intracellular GSH levels associated with apoptosis. ATF4(-/-) MEFs could be rescued from l-Hcy-induced apoptosis by ß-mercaptoethanol medium supplementation that increases cysteine levels and restores GSH synthesis. ATF4(-/-) MEFs showed little or no CSE protein but did express cystathionine ß-synthase. Further, ER stress-inducing agents, including tunicamycin and thapsigargin, induced the expression of CSE in ATF4(+/+) MEFs. Consistent with ATF4(-/-) MEFs, CSE(-/-) MEFs showed significantly greater apoptosis when treated with tunicamycin, thapsigargin, and l-Hcy, compared with CSE(+/+) MEFs. Liver and kidney GSH levels were also reduced in CSE(-/-) mice, suggesting that CSE is a critical factor in GSH synthesis and may act to protect the liver and kidney from a variety of conditions that cause ER stress.


Endoplasmic Reticulum Stress/physiology , Fibroblasts/metabolism , Glutathione/metabolism , Homocysteine/metabolism , Lyases/metabolism , Sulfhydryl Compounds/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Apoptosis/physiology , Cells, Cultured , Cystathionine gamma-Lyase , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Glutathione/genetics , Homocysteine/genetics , Kidney/metabolism , Liver/metabolism , Lyases/genetics , Mice , Mice, Knockout , Oxidation-Reduction
14.
Circ Res ; 108(5): 629-42, 2011 Mar 04.
Article En | MEDLINE | ID: mdl-21372294

Synthesis of transmembrane and secretory proteins occurs within the endoplasmic reticulum (ER) and is extremely important in the normal functioning of both the heart and kidney. The dysregulation of protein synthesis/processing within the ER causes the accumulation of unfolded proteins, thereby leading to ER stress and the activation of the unfolded protein response. Sarcoplasmic reticulum/ER Ca2+ disequilibrium can lead to cardiac hypertrophy via cytosolic Ca2+ elevation and stimulation of the Ca2+/calmodulin, calcineurin, NF-AT3 pathway. Although cardiac hypertrophy may be initially adaptive, prolonged or severe ER stress resulting from the increased protein synthesis associated with cardiac hypertrophy can lead to apoptosis of cardiac myocytes and result in reduced cardiac output and chronic heart failure. The failing heart has a dramatic effect on renal function because of inadequate perfusion and stimulates the release of many neurohumoral factors that may lead to further ER stress within the heart, including angiotensin II and arginine-vasopressin. Renal failure attributable to proteinuria and uremia also induces ER stress within the kidney, which contributes to the transformation of tubular epithelial cells to a fibroblast-like phenotype, fibrosis, and tubular cell apoptosis, further diminishing renal function. As a consequence, cardiorenal syndrome may develop into a vicious circle with poor prognosis. New therapeutic modalities to alleviate ER stress through stimulation of the cytoprotective components of the unfolded protein response, including GRP78 upregulation and eukaryotic initiation factor 2α phosphorylation, may hold promise to reduce the high morbidity and mortality associated with cardiorenal syndrome.


Cardiomegaly/physiopathology , Endoplasmic Reticulum/physiology , Heart Failure/physiopathology , Kidney Diseases/physiopathology , Apoptosis , Cardiomegaly/etiology , Cardiomegaly/pathology , Chronic Disease , Endoplasmic Reticulum Chaperone BiP , Heart Failure/etiology , Heart Failure/pathology , Humans , Kidney Diseases/etiology , Kidney Diseases/pathology , Stress, Physiological/physiology
15.
FASEB J ; 25(2): 576-89, 2011 Feb.
Article En | MEDLINE | ID: mdl-20966213

Endoplasmic reticulum (ER) stress causes macrophage cell death within advanced atherosclerotic lesions, thereby contributing to necrotic core formation and increasing the risk of atherothrombotic disease. However, unlike in advanced lesions, the appearance of dead/apoptotic macrophages in early lesions is less prominent. Given that activation of the unfolded protein response (UPR) is detected in early lesion-resident macrophages and can enhance cell survival against ER stress, we investigated whether UPR activation occurs after monocyte to macrophage differentiation and confers a cytoprotective advantage to the macrophage. Human peripheral blood monocytes were treated with monocyte colony-stimulating factor to induce macrophage differentiation, as assessed by changes in ultrastructure and scavenger receptor expression. UPR markers, including GRP78, GRP94, and spliced XBP-1, were induced after macrophage differentiation and occurred after a significant increase in de novo protein synthesis. UPR activation after differentiation reduced macrophage cell death by ER stress-inducing agents. Further, GRP78 overexpression in macrophages was sufficient to reduce ER stress-induced cell death. Consistent with these in vitro findings, UPR activation was observed in viable lesion-resident macrophages from human carotid arteries and from the aortas of apoE(-/-) mice. However, no evidence of apoptosis was observed in early lesion-resident macrophages from the aortas of apoE(-/-) mice. Thus, our findings that UPR activation occurs during macrophage differentiation and is cytoprotective against ER stress-inducing agents suggest an important cellular mechanism for macrophage survival within early atherosclerotic lesions.


Atherosclerosis/metabolism , Cell Differentiation/physiology , Macrophages/metabolism , Monocytes/physiology , Unfolded Protein Response/physiology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cell Survival , Endoplasmic Reticulum Chaperone BiP , Female , Gene Expression Regulation , Humans , Macrophages/ultrastructure , Mice , Mice, Knockout , Monocytes/cytology
...