Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Am J Physiol Endocrinol Metab ; 321(1): E90-E104, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34029162

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a spectrum of disorders ranging from hepatic steatosis [excessive accumulation of triglycerides (TG)] to nonalcoholic steatohepatitis, which can progress to cirrhosis and hepatocellular carcinoma. The molecular pathogenesis of steatosis and progression to more severe NAFLD remains unclear. Obesity and aging, two principal risk factors for NAFLD, are associated with a hyperadrenergic state. ß-Adrenergic responsiveness in liver increases in animal models of obesity and aging, and in both is linked to increased hepatic expression of ß2-adrenergic receptors (ß2-ARs). We previously showed that in aging rodents intracellular signaling from elevated hepatic levels of ß2-ARs may contribute to liver steatosis. In this study we demonstrate that injection of formoterol, a highly selective ß2-AR agonist, to mice acutely results in hepatic TG accumulation. Further, we have sought to define the intrahepatic mechanisms underlying ß2-AR mediated steatosis by investigating changes in hepatic expression and cellular localization of enzymes, transcription factors, and coactivators involved in processes of lipid accrual and disposition-and also functional aspects thereof-in livers of formoterol-treated animals. Our results suggest that ß2-AR activation by formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, increased but incomplete ß-oxidation of fatty acids with accumulation of potentially toxic long-chain acylcarnitine intermediates, and reduced TG secretion-all previously invoked as contributors to fatty liver disease. Experiments are ongoing to determine whether sustained activation of hepatic ß2-AR signaling by formoterol might be utilized to model fatty liver changes occurring in hyperadrenergic states of obesity and aging, and thereby identify novel molecular targets for the prevention or treatment of NAFLD.NEW & NOTEWORTHY Results of our study suggest that ß2-adrenergic receptor (ß2-AR) activation by agonist formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, incomplete ß-oxidation of fatty acids with accumulation of long-chain acylcarnitine intermediates, and reduced TG secretion. These findings may, for the first time, implicate a role for ß2-AR responsive dysregulation of hepatic lipid metabolism in the pathogenetic processes underlying NAFLD in hyperadrenergic states such as obesity and aging.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Fatty Liver/chemically induced , Non-alcoholic Fatty Liver Disease/physiopathology , Receptors, Adrenergic, beta-2/physiology , Animals , Carnitine/analogs & derivatives , Carnitine/analysis , Formoterol Fumarate/pharmacology , Gene Expression/drug effects , Hepatic Stellate Cells , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipogenesis/genetics , Liver/chemistry , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/chemically induced , Phosphatidate Phosphatase/analysis , Triglycerides/biosynthesis
3.
Geroscience ; 40(2): 163-176, 2018 04.
Article in English | MEDLINE | ID: mdl-29717417

ABSTRACT

Aging is associated with replacement of normal kidney parenchyma by fibrosis. Because hydrogen sulfide (H2S) ameliorates kidney fibrosis in disease models, we examined its status in the aging kidney. In the first study, we examined kidney cortical H2S metabolism and signaling pathways related to synthesis of proteins including matrix proteins in young and old male C57BL/6 mice. In old mice, increase in renal cortical content of matrix protein involved in fibrosis was associated with decreased H2S generation and AMPK activity, and activation of insulin receptor (IR)/IRS-2-Akt-mTORC1-mRNA translation signaling axis that can lead to increase in protein synthesis. In the second study, we randomized 18-19 month-old male C57BL/6 mice to receive 30 µmol/L sodium hydrosulfide (NaHS) in drinking water vs. water alone (control) for 5 months. Administration of NaHS increased plasma free sulfide levels. NaHS inhibited the increase in kidney cortical content of matrix proteins involved in fibrosis and ameliorated glomerulosclerosis. NaHS restored AMPK activity and inhibited activation of IR/IRS-2-Akt-mTORC1-mRNA translation axis. NaHS inhibited age-related increase in kidney cortical content of p21, IL-1ß, and IL-6, components of the senescence-associated secretory phenotype. NaHS abolished increase in urinary albumin excretion seen in control mice and reduced serum cystatin C levels suggesting improved glomerular clearance function. We conclude that aging-induced changes in the kidney are associated with H2S deficiency. Administration of H2S ameliorates aging-induced kidney changes probably by inhibiting signaling pathways leading to matrix protein synthesis.


Subject(s)
Aging/drug effects , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Kidney/pathology , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Aging/metabolism , Animals , Biomarkers/metabolism , Biopsy, Needle , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Fibrosis/drug therapy , Fibrosis/pathology , Immunohistochemistry , Kidney/drug effects , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Random Allocation , Reference Values , Risk Factors , Signal Transduction/drug effects
4.
Am J Pathol ; 188(3): 768-784, 2018 03.
Article in English | MEDLINE | ID: mdl-29269120

ABSTRACT

In vitro models for the investigation of renal vascular development are limited. We previously showed that isolated metanephric mesenchymal (MM) and ureteric bud (UB) cells grown in three-dimensional (3D) matrices formed organoids that consisted of primitive vascular structures surrounding a polarized epithelium. Here, we examined the potential of two principal effectors of vasculogenesis, vascular endothelial growth factor A (VEGF-A), and platelet-derived growth factor B chain (PDGF-BB), to stimulate MM cell differentiation. The results showed that MM cells possess angioblast characteristics by expressing phenotypic markers for endothelial and mesenchymal cells. UB cells synthesize VEGF-A and PDGF-BB proteins and RNA, whereas the MM cells express the respective cognate receptors, supporting their role in directional induction of vasculogenesis. VEGF-A stimulated proliferation of MM cells in monolayer and in 3D sponges but did not affect MM cell migration, organization, or vasculogenesis. However, PDGF-BB stimulated MM cell proliferation, migration, and vasculogenesis in monolayer and organization of the cells into primitive capillary-like assemblies in 3D sea sponge scaffolds in vitro. A role for PDGF-BB in vasculogenesis in the 3D MM/UB co-culture system was validated by direct interference with PDGF-BB or PDGF receptor-ß cell interactions to implicate PDGF-BB as a primary effector of MM cell vasculogenesis. Thus, MM cells resemble early renal angioblasts that may provide an ideal platform for the investigation of renal vasculogenesis in vitro.


Subject(s)
Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic/physiology , Animals , Becaplermin/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Mesenchymal Stem Cells/drug effects , Mice , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology
5.
Am J Physiol Regul Integr Comp Physiol ; 314(4): R574-R583, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29212811

ABSTRACT

Increased ß-adrenergic receptor (ß-AR)-mediated activation of adenylyl cyclase (AC) in rat liver during aging has been linked to age-related increases in hepatic glucose output and hepatosteatosis. In this study, we investigated the expression of ß-ARs, individual receptor subtypes, and G protein-coupled receptor (GPCR) regulatory proteins in livers from aging rats. Radioligand-binding studies demonstrated that ß-AR density increased by greater than threefold in hepatocyte membranes from senescent (24-mo-old) compared with young adult (7-mo-old) rats and that this phenomenon was blocked by food restriction, which is known to retard aging processes in rodents. Competition-binding studies revealed a mixed population of ß1- and ß2-AR subtypes in liver membranes over the adult life span, with a trend for greater ß2-AR density with age. Expression of both ß-AR subtype mRNAs in rat liver increased with age, whereas ß2- but not ß1-AR protein levels declined in livers of old animals. Immunoreactive ß2- but not ß1-ARs were preferentially distributed in pericentral hepatic regions. Levels of GRK2/3 and ß-arrestin 2 proteins, which are involved in downregulation of agonist-activated GPCRs, including ß-ARs, increased during aging. Insofar as sympathetic tone increases with age, our findings suggest that, despite enhanced agonist-mediated downregulation of hepatic ß-ARs preferentially affecting the ß2-AR subtype, increased generation of both receptor subtypes during aging augments the pool of plasma membrane-bound ß-ARs coupled to AC in hepatocytes. This study thus identifies one or both ß-AR subtypes as possible therapeutic targets involved in aberrant hepatic processes of glucose and lipid metabolism during aging.


Subject(s)
Aging/metabolism , Cell Membrane/metabolism , Energy Metabolism , Hepatocytes/metabolism , Liver/metabolism , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Age Factors , Aging/genetics , Animals , Caloric Restriction , Energy Metabolism/genetics , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/genetics , G-Protein-Coupled Receptor Kinase 3/metabolism , Gene Expression Regulation , Glucose/metabolism , Ligands , Lipid Metabolism , Liver/physiopathology , Male , Rats, Inbred F344 , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , beta-Arrestin 2/genetics , beta-Arrestin 2/metabolism
6.
FASEB J ; 30(9): 3026-38, 2016 09.
Article in English | MEDLINE | ID: mdl-27221979

ABSTRACT

Systemic fibrosis can be induced in humans with gadolinium-based contrast, and cumulative doses correlate with severity. Bone marrow-derived fibrocytes accumulate in the dermis. Whether target organs liberate chemokines to recruit these fibrocytes or whether fibrocytes are stimulated to home to the affected tissue is unknown. Transgenic (tagged) donor rats were treated with gadolinium-based contrast. Bone marrow was obtained from diseased animals and age-matched controls. Rats with subtotal nephrectomies were lethally irradiated and underwent salvage transplantation with either the contrast-naïve or contrast-exposed bone marrow. Groups were randomly assigned to control or contrast treatment. Contrast treatment led to dermal fibrosis, and this was exacerbated in recipients of contrast-exposed marrow. Fibronectin, C-C chemokine receptors (CCRs)2 and 7, and oxidative stress were all increased in skin from contrast-treated animals-all parameters more severe in recipients of contrast-treated animals. The respective ligands, monocyte chemoattractant protein and C-C motif ligand 19, were both elevated in skin from contrast-treated animals. Coadministration of gadolinium-based contrast and a CCR2 inhibitor reduced the severity of skin disease as well as dermal cellularity. The functional role of chemokines in the effects of gadolinium-based contrast was further confirmed in in situ coculture studies using neutralizing CCR2 antibodies. These data implicate dermal liberation of specific chemokines in the recruitment of circulating bone marrow-derived cells. The disease is augmented by bone marrow exposure to contrast, which explains why multiple exposures correlate with severity.-Drel, V. R., Tan, C., Barnes, J. L., Gorin, Y., Lee, D.-Y., Wagner, B. Centrality of bone marrow in the severity of gadolinium-based contrast-induced systemic fibrosis.


Subject(s)
Bone Marrow/drug effects , Contrast Media/adverse effects , Gadolinium DTPA/adverse effects , Nephrogenic Fibrosing Dermopathy/chemically induced , Animals , Animals, Genetically Modified , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Bone Marrow Transplantation , Contrast Media/administration & dosage , Contrast Media/pharmacology , Female , Gadolinium DTPA/metabolism , Gene Expression Regulation , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Male , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Nephrogenic Fibrosing Dermopathy/pathology , Random Allocation , Rats , Reactive Oxygen Species , Receptors, CCR2/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Skin/drug effects , Skin/metabolism
7.
J Gerontol A Biol Sci Med Sci ; 71(7): 850-7, 2016 07.
Article in English | MEDLINE | ID: mdl-26442901

ABSTRACT

We examined the effect of rapamycin on the life span of a mouse model of type 2 diabetes, db/db mice. At 4 months of age, male and female C57BLKSJ-lepr (db/db) mice (db/db) were placed on either a control diet, lacking rapamycin or a diet containing rapamycin and maintained on these diets over their life span. Rapamycin was found to reduce the life span of the db/db mice. The median survival of male db/db mice fed the control and rapamycin diets was 349 and 302 days, respectively, and the median survival of female db/db mice fed the control and rapamycin diets was 487 and 411 days, respectively. Adjusting for gender differences, rapamycin increased the mortality risk 1.7-fold in both male and female db/db mice. End-of-life pathological data showed that suppurative inflammation was the main cause of death in the db/db mice, which is enhanced slightly by rapamycin treatment.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Inflammation/pathology , Longevity , Sirolimus , Animals , Cause of Death , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/mortality , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/mortality , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Female , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Longevity/drug effects , Longevity/physiology , Male , Mice , Mice, Inbred C57BL , Mortality , Sex Factors , Sirolimus/metabolism , Sirolimus/pharmacology , Treatment Outcome
8.
J Cell Commun Signal ; 9(4): 327-39, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26698861

ABSTRACT

The CCN family of matricellular signaling proteins is emerging as a unique common link across multiple diseases and organs related to injury and repair. They are now being shown to play a central role in regulating the pathways to the initiation and resolution of normal wound healing and fibrosis in response to multiple forms of injury. Similarly, it is also emerging that they play a key role in regulating the establishment, growth, metastases and tissue regeneration in many forms of cancer via the interaction of cancer cells with the tumor stroma. Evidence has been recently provided that these proteins do not act independently but are co-regulated working in a yin/yang manner to alter the outcome of both normal physiological processes as well as pathology. The purpose of this review is to twofold. First, it will summarize work to date supporting CCN2 as a therapeutic target in the formation and progression of renal, skin, and other organ fibrosis, as well as cancer stroma formation. Second, it will highlight recent evidence for CCN3 as a counter-regulator and a potential therapeutic agent in these diseases with an exciting, novel potential to both treat and then restore tissue homeostasis in those afflicted by these devastating disorders.

9.
Am J Pathol ; 185(8): 2168-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26071397

ABSTRACT

Transforming growth factor (TGF)-ß contributes to tubulointerstitial fibrosis. We investigated the mechanism by which TGF-ß exerts its profibrotic effects and specifically the role of AMP-activated protein kinase (AMPK) in kidney tubular epithelial cells and interstitial fibroblasts. In proximal tubular epithelial cells, TGF-ß1 treatment causes a decrease in AMPK phosphorylation and activation together with increased fibronectin and α-smooth muscle actin expression and decreased in E-cadherin. TGF-ß1 causes similar changes in interstitial fibroblasts. Activation of AMPK with 5-aminoimidazole-4-carboxamide 1-ß-d-ribofuranoside, metformin, or overexpression of constitutively active AMPK markedly attenuated TGF-ß1 functions. Conversely, inhibition of AMPK with adenine 9-ß-d-arabinofuranoside or siRNA-mediated knockdown of AMPK (official name PRKAA1) mimicked the effect of TGF-ß1 and enhanced basal and TGF-ß1-induced phenotypic changes. Importantly, we found that tuberin contributed to the protective effects of AMPK and that TGF-ß1 promoted cell injury by blocking AMPK-mediated tuberin phosphorylation and activation. In the kidney cortex of TGF-ß transgenic mice, the significant decrease in AMPK phosphorylation and tuberin phosphorylation on its AMPK-dependent activating site was associated with an increase in mesenchymal markers and a decrease in E-cadherin. Collectively, the data indicate that TGF-ß exerts its profibrotic action in vitro and in vivo via inactivation of AMPK. AMPK and tuberin activation prevent tubulointerstitial injury induced by TGF-ß. Activators of AMPK provide potential therapeutic strategy to prevent kidney fibrosis and progressive kidney disease.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Epithelial-Mesenchymal Transition/drug effects , Kidney Tubules, Proximal/metabolism , Myofibroblasts/metabolism , Transforming Growth Factor beta1/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cadherins/metabolism , Fibronectins/metabolism , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Mice , Mice, Knockout , Myofibroblasts/drug effects , Phosphorylation/drug effects , Ribonucleosides/pharmacology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
10.
Am J Physiol Renal Physiol ; 308(11): F1276-87, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25656366

ABSTRACT

Reactive oxygen species (ROS) generated by Nox NADPH oxidases may play a critical role in the pathogenesis of diabetic nephropathy (DN). The efficacy of the Nox1/Nox4 inhibitor GKT137831 on the manifestations of DN was studied in OVE26 mice, a model of type 1 diabetes. Starting at 4-5 mo of age, OVE26 mice were treated with GKT137831 at 10 or 40 mg/kg, once-a-day for 4 wk. At both doses, GKT137831 inhibited NADPH oxidase activity, superoxide generation, and hydrogen peroxide production in the renal cortex from diabetic mice without affecting Nox1 or Nox4 protein expression. The increased expression of fibronectin and type IV collagen was reduced in the renal cortex, including glomeruli, of diabetic mice treated with GKT137831. GKT137831 significantly reduced glomerular hypertrophy, mesangial matrix expansion, urinary albumin excretion, and podocyte loss in OVE26 mice. GKT137831 also attenuated macrophage infiltration in glomeruli and tubulointerstitium. Collectively, our data indicate that pharmacological inhibition of Nox1/4 affords broad renoprotection in mice with preexisting diabetes and established kidney disease. This study validates the relevance of targeting Nox4 and identifies GKT137831 as a promising compound for the treatment of DN in type 1 diabetes.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Enzyme Inhibitors/pharmacology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADPH Oxidases/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Kidney/metabolism , Kidney/pathology , Mice , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , Podocytes/drug effects , Podocytes/metabolism , Pyrazolones , Pyridones , Reactive Oxygen Species/metabolism
11.
J Biol Chem ; 289(51): 35363-75, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25339176

ABSTRACT

Increase in protein synthesis contributes to kidney hypertrophy and matrix protein accumulation in diabetes. We have previously shown that high glucose-induced matrix protein synthesis is associated with inactivation of glycogen synthase kinase 3ß (GSK3ß) in renal cells and in the kidneys of diabetic mice. We tested whether activation of GSK3ß by sodium nitroprusside (SNP) mitigates kidney injury in diabetes. Studies in kidney-proximal tubular epithelial cells showed that SNP abrogated high glucose-induced laminin increment by stimulating GSK3ß and inhibiting Akt, mTORC1, and events in mRNA translation regulated by mTORC1 and ERK. NONOate, an NO donor, also activated GSK3ß, indicating that NO may mediate SNP stimulation of GSK3ß. SNP administered for 3 weeks to mice with streptozotocin-induced type 1 diabetes ameliorated kidney hypertrophy, accumulation of matrix proteins, and albuminuria without changing blood glucose levels. Signaling studies showed that diabetes caused inactivation of GSK3ß by activation of Src, Pyk2, Akt, and ERK; GSK3ß inhibition activated mTORC1 and downstream events in mRNA translation in the kidney cortex. These reactions were abrogated by SNP. We conclude that activation of GSK3ß by SNP ameliorates kidney injury induced by diabetes.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Glycogen Synthase Kinase 3/metabolism , Kidney/drug effects , Nitroprusside/pharmacology , Albuminuria/prevention & control , Animals , Cell Line, Transformed , Diabetes Mellitus, Experimental/enzymology , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycogen Synthase Kinase 3 beta , Hypertrophy/prevention & control , Immunoblotting , Kidney/enzymology , Kidney/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Laminin/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Multiprotein Complexes/metabolism , Nitric Oxide Donors/pharmacology , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
12.
Am J Pathol ; 184(11): 2908-21, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25193594

ABSTRACT

Fibrosis is at the core of the high morbidity and mortality rates associated with the complications of diabetes and obesity, including diabetic nephropathy (DN), without any US Food and Drug Administration-approved drugs with this specific target. We recently provided the first evidence that the matricellular protein CCN3 (official symbol NOV) functions in a reciprocal manner, acting on the profibrotic family member CCN2 to inhibit fibrosis in a mesangial cell model of DN. Herein, we used the BT/BR ob/ob mouse as a best model of human obesity and DN progression to determine whether recombinant human CCN3 could be used therapeutically, and the mechanisms involved. Eight weeks of thrice-weekly i.p. injections (0.604 and 6.04 µg/kg of recombinant human CCN3) beginning in early-stage DN completely blocked and/or reversed the up-regulation of mRNA expression of kidney cortex fibrosis genes (CCN2, Col1a2, TGF-ß1, and PAI-1) seen in placebo-treated diabetic mice. The treatment completely blocked glomerular fibrosis, as determined by altered mesangial expansion and deposition of laminin. Furthermore, it protected against, or reversed, podocyte loss and kidney function reduction (rise in plasma creatinine concentration); albuminuria was also greatly reduced. This study demonstrates the potential efficacy of recombinant human CCN3 treatment in DN and points to mechanisms operating at multiple levels or pathways, upstream (eg, protecting against cell injury) and downstream (eg, regulating CCN2 activity and extracellular matrix metabolism).


Subject(s)
Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Fibrosis/drug therapy , Kidney/drug effects , Nephroblastoma Overexpressed Protein/therapeutic use , Animals , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/complications , Fibrosis/pathology , Fibrosis/prevention & control , Kidney/pathology , Male , Mice , Nephroblastoma Overexpressed Protein/pharmacology , Obesity/complications , Obesity/pathology , Treatment Outcome
13.
Am J Physiol Renal Physiol ; 307(7): F844-55, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25100280

ABSTRACT

It has been presupposed that the thermodynamic stability constant (K(therm)) of gadolinium-based MRI chelates relate to the risk of precipitating nephrogenic systemic fibrosis. The present study compared low-K(therm) gadodiamide with high-K(therm) gadoteridol in cultured fibroblasts and rats with uninephrectomies. Gadolinium content was assessed using scanning electron microscopy equipped with energy-dispersive X-ray spectroscopy in paraffin-embedded tissues. In vitro, fibroblasts demonstrated dose-dependent fibronectin generation, transforming growth factor-ß production, and expression of activated myofibroblast stress fiber protein α-smooth muscle actin. There were negligible differences with respect to toxicity or proliferation between the two contrast agents. In the rodent model, gadodiamide treatment led to greater skin fibrosis and dermal cellularity than gadoteridol. In the kidney, both contrast agents led to proximal tubule vacuolization and increased fibronectin accumulation. Despite large detectable gadolinium signals in the spleen, skin, muscle, and liver from the gadodiamide-treated group, contrast-induced fibrosis appeared to be limited to the skin and kidney. These findings support the hypothesis that low-K(therm) chelates have a greater propensity to elicit nephrogenic systemic fibrosis and demonstrate that certain tissues are resistant to these effects.


Subject(s)
Contrast Media/adverse effects , Gadolinium DTPA/adverse effects , Heterocyclic Compounds/adverse effects , Nephrogenic Fibrosing Dermopathy/chemically induced , Organometallic Compounds/adverse effects , Animals , Cells, Cultured , Contrast Media/chemistry , Female , Fibroblasts/metabolism , Fibronectins/metabolism , Gadolinium/adverse effects , Gadolinium/chemistry , Gadolinium DTPA/chemistry , Heterocyclic Compounds/chemistry , Humans , Magnetic Resonance Imaging , Nephrogenic Fibrosing Dermopathy/metabolism , Organometallic Compounds/chemistry , Rats, Inbred F344 , Renal Insufficiency/complications , Thermodynamics
14.
Am J Physiol Renal Physiol ; 307(2): F159-71, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24872317

ABSTRACT

The small G proteins Rac1 and RhoA regulate actin cytoskeleton, cell shape, adhesion, migration, and proliferation. Recent studies in our laboratory have shown that NADPH oxidase Nox4-derived ROS are involved in transforming growth factor (TGF)-ß1-induced rat kidney myofibroblast differentiation assessed by the acquisition of an α-smooth muscle actin (α-SMA) phenotype and expression of an alternatively spliced fibronectin variant (Fn-EIIIA). Rac1 and RhoA are essential in signaling by some Nox homologs, but their role as effectors of Nox4 in kidney myofibroblast differentiation is not known. In the present study, we explored a link among Rac1 and RhoA and Nox4-dependent ROS generation in TGF-ß1-induced kidney myofibroblast activation. TGF-ß1 stimulated an increase in Nox4 protein expression, NADPH oxidase activity, and abundant α-SMA and Fn-EIIIA expression. RhoA but not Rac1 was involved in TGF-ß1 induction of Nox4 signaling of kidney myofibroblast activation. TGF-ß1 stimulated active RhoA-GTP and increased Rho kinase (ROCK). Inhibition of RhoA with small interfering RNA and ROCK using Y-27632 significantly reduced TGF-ß1-induced stimulation of Nox4 protein, NADPH oxidase activity, and α-SMA and Fn-EIIIA expression. Treatment with diphenyleneiodonium, an inhibitor of NADPH oxidase, did not decrease RhoA activation but inhibited TGF-ß1-induced α-SMA and Fn-EIIIA expression, indicating that RhoA is upstream of ROS generation. RhoA/ROCK also regulated polymerase (DNA-directed) δ-interacting protein 2 (Poldip2), a newly discovered Nox4 enhancer protein. Collectively, these data indicate that RhoA/ROCK is upstream of Poldip2-dependent Nox4 regulation and ROS production and induces redox signaling of kidney myofibroblast activation and may broader implications in the pathophysiology of renal fibrosis.


Subject(s)
Cell Cycle Proteins/metabolism , Kidney/enzymology , Myofibroblasts/enzymology , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Line , Cell Movement , Enzyme Activation , Fibronectins/metabolism , Fibrosis , Kidney/pathology , Myofibroblasts/pathology , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Oxidation-Reduction , Phenotype , Protein Kinase Inhibitors/pharmacology , RNA Interference , Rats , Signal Transduction , Time Factors , Transfection , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics
15.
Infect Immun ; 82(8): 3098-112, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24818662

ABSTRACT

Malnutrition is thought to contribute to more than one-third of all childhood deaths via increased susceptibility to infection. Malnutrition is a significant risk factor for the development of visceral leishmaniasis, which results from skin inoculation of the intracellular protozoan Leishmania donovani. We previously established a murine model of childhood malnutrition and found that malnutrition decreased the lymph node barrier function and increased the early dissemination of L. donovani. In the present study, we found reduced numbers of resident dendritic cells (conventional and monocyte derived) but not migratory dermal dendritic cells in the skin-draining lymph nodes of L. donovani-infected malnourished mice. Expression of chemokines and their receptors involved in trafficking of dendritic cells and their progenitors to the lymph nodes was dysregulated. C-C chemokine receptor type 2 (CCR2) and its ligands (CCL2 and CCL7) were reduced in the lymph nodes of infected malnourished mice, as were CCR2-bearing monocytes/macrophages and monocyte-derived dendritic cells. However, CCR7 and its ligands (CCL19 and CCL21) were increased in the lymph node and CCR7 was increased in lymph node macrophages and dendritic cells. CCR2-deficient mice recapitulated the profound reduction in the number of resident (but not migratory dermal) dendritic cells in the lymph node but showed no alteration in the expression of CCL19 and CCL21. Collectively, these results suggest that the malnutrition-related reduction in the lymph node barrier to dissemination of L. donovani is related to insufficient numbers of lymph node-resident but not migratory dermal dendritic cells. This is likely driven by the altered activity of the CCR2 and CCR7 chemoattractant pathways.


Subject(s)
Chemokines/metabolism , Dendritic Cells/immunology , Leishmania donovani/immunology , Leishmaniasis, Visceral/complications , Leishmaniasis, Visceral/immunology , Lymph Nodes/cytology , Malnutrition/immunology , Animals , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred BALB C , Receptors, Chemokine/biosynthesis
16.
PLoS Negl Trop Dis ; 7(8): e2329, 2013.
Article in English | MEDLINE | ID: mdl-23967356

ABSTRACT

In a murine model of moderate childhood malnutrition we found that polynutrient deficiency led to a 4-5-fold increase in early visceralization of L. donovani (3 days post-infection) following cutaneous infection and a 16-fold decrease in lymph node barrier function (p<0.04 for all). To begin to understand the mechanistic basis for this malnutrition-related parasite dissemination we analyzed the cellularity, architecture, and function of the skin-draining lymph node. There was no difference in the localization of multiple cell populations in the lymph node of polynutrient deficient (PND) mice, but there was reduced cellularity with fewer CD11c(+)dendritic cells (DCs), fibroblastic reticular cells (FRCs), MOMA-2(+) macrophages, and CD169(+) subcapsular sinus macrophage (p<0.05 for all) compared to the well-nourished (WN) mice. The parasites were equally co-localized with DCs associated with the lymph node conduit network in the WN and PND mice, and were found in the high endothelial venule into which the conduits drain. When a fluorescent low molecular weight (10 kD) dextran was delivered in the skin, there was greater efflux of the marker from the lymph node conduit system to the spleens of PND mice (p<0.04), indicating that flow through the conduit system was altered. There was no evidence of disruption of the conduit or subcapsular sinus architecture, indicating that the movement of parasites into the subcortical conduit region was due to an active process and not from passive movement through a leaking barrier. These results indicate that the impaired capacity of the lymph node to act as a barrier to dissemination of L. donovani infection is associated with a reduced number of lymph node phagocytes, which most likely leads to reduced capture of parasites as they transit through the sinuses and conduit system.


Subject(s)
Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/pathology , Lymph Nodes/immunology , Malnutrition/complications , Phagocytes/immunology , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Time Factors
17.
Exp Biol Med (Maywood) ; 238(6): 610-22, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23918873

ABSTRACT

A growing number of reports indicate that anti-inflammatory actions of fish oil (FO) are beneficial against systemic lupus erythematosus (SLE). However, the majority of pre-clinical studies were performed using 5-20% FO, which is higher than the clinically relevant dose for lupus patients. The present study was performed in order to determine the effective low dose of FDA-approved concentrated FO (Lovaza®) compared to the commonly used FO-18/12 (18-Eicosapentaenoic acid [EPA]/12-Docosahexaenoic acid [DHA]). We examined the dose-dependent response of Lovaza® (1% and 4%) on an SLE mouse strain (NZBxNZW)F1 and compared the same with 1% and 4% placebo, as well as 4% FO-18/12, maintaining standard chow as the control. Results show for the first time that 1% Lovaza® extends maximal lifespan (517 d) and 4% Lovaza® significantly extends both the median (502 d) and maximal (600 d) life span of (NZBxNZW)F1 mice. In contrast, FO-18/12 extends only median lifespan (410 d) compared to standard chow diet (301 d). Additionally, 4% Lovaza® significantly decreased anti-dsDNA antibodies, reduced glomerulonephritis and attenuated lipopolysaccharide-induced pro-inflammatory cytokines (IL-1ß, IL-6, TNF-α) in splenocytes compared to placebo. 4% Lovaza® was also shown to reduce the expression of inflammatory cytokines, including IL-1ß, IL-6 and TNF-α, while increasing renal anti-oxidant enzymes in comparison to placebo. Notably, NFκB activation and p65 nuclear translocation were lowered by 4% Lovaza® compared to placebo. These data indicate that 1% Lovaza® is beneficial, but 4% Lovaza® is more effective in suppressing glomerulonephritis and extending life span of SLE-prone short-lived mice, possibly via reducing inflammation signaling and modulating oxidative stress.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Fish Oils/pharmacology , Kidney Diseases/drug therapy , Lupus Erythematosus, Systemic/drug therapy , Animals , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Female , Fish Oils/administration & dosage , Inflammation/drug therapy , Inflammation/immunology , Kidney Diseases/immunology , Kidney Diseases/pathology , Lupus Erythematosus, Systemic/immunology , Male , Mice
18.
Am J Physiol Renal Physiol ; 305(3): F323-32, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23678045

ABSTRACT

Matrix protein accumulation is a prominent feature of diabetic nephropathy that contributes to renal fibrosis and decline in renal function. The pathogenic mechanisms of matrix accumulation are incompletely characterized. We investigated if the matrix metalloprotease a disintegrin and metalloprotease1 7 (ADAM17), known to cleave growth factors and cytokines, is activated in the kidney cortex of OVE26 type 1 diabetic mice and the potential mechanisms by which ADAM17 mediates extracellular matrix accumulation. Protein expression and activity of ADAM17 were increased in OVE26 kidney cortex. Using a pharmacological inhibitor to ADAM17, TMI-005, we determined that ADAM17 activation results in increased type IV collagen, Nox4, and NADPH oxidase activity in the kidney cortex of diabetic mice. In cultured mouse proximal tubular epithelial cells (MCTs), high glucose increases ADAM17 activity, Nox4 and fibronectin expression, cellular collagen content, and NADPH oxidase activity. These effects of glucose were inhibited when cells were pretreated with TMI-005 and/or transfected with small interfering ADAM17. Collectively, these data indicate a novel mechanism whereby hyperglycemia in diabetes increases extracellular matrix protein expression in the kidney cortex through activation of ADAM17 and enhanced oxidative stress through Nox enzyme activation. Additionally, our study is the first to provide evidence that Nox4 is downstream of ADAM17.


Subject(s)
ADAM Proteins/physiology , Diabetes Mellitus, Type 1/metabolism , Kidney Cortex/metabolism , NADPH Oxidases/metabolism , ADAM17 Protein , Animals , Blotting, Western , Body Weight/physiology , Collagen/metabolism , Collagen Type IV/metabolism , Fibronectins/metabolism , Fluorescent Antibody Technique , Glucose/metabolism , Glucose/pharmacology , Immunoenzyme Techniques , Kidney Cortex/enzymology , Male , Mice , NADPH Oxidase 4 , NADPH Oxidases/biosynthesis , NADPH Oxidases/genetics , Organ Size/physiology , RNA, Small Interfering/pharmacology
19.
Diabetes ; 62(8): 2935-47, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23557706

ABSTRACT

Podocyte apoptosis is a critical mechanism for excessive loss of urinary albumin that eventuates in kidney fibrosis. Pharmacological doses of the mammalian target of rapamycin (mTOR) inhibitor rapamycin reduce albuminuria in diabetes. We explored the hypothesis that mTOR mediates podocyte injury in diabetes. High glucose (HG) induces apoptosis of podocytes, inhibits AMP-activated protein kinase (AMPK) activation, inactivates tuberin, and activates mTOR. HG also increases the levels of Nox4 and Nox1 and NADPH oxidase activity. Inhibition of mTOR by low-dose rapamycin decreases HG-induced Nox4 and Nox1, NADPH oxidase activity, and podocyte apoptosis. Inhibition of mTOR had no effect on AMPK or tuberin phosphorylation, indicating that mTOR is downstream of these signaling molecules. In isolated glomeruli of OVE26 mice, there is a similar decrease in the activation of AMPK and tuberin and activation of mTOR with increase in Nox4 and NADPH oxidase activity. Inhibition of mTOR by a small dose of rapamycin reduces podocyte apoptosis and attenuates glomerular injury and albuminuria. Our data provide evidence for a novel function of mTOR in Nox4-derived reactive oxygen species generation and podocyte apoptosis that contributes to urinary albumin excretion in type 1 diabetes. Thus, mTOR and/or NADPH oxidase inhibition may represent a therapeutic modality of diabetic kidney disease.


Subject(s)
Diabetic Nephropathies/metabolism , Glucose/pharmacology , NADPH Oxidases/metabolism , Podocytes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adenylate Kinase/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Line , Hypoglycemic Agents/pharmacology , Mice , NADPH Oxidase 4 , Phosphorylation/drug effects , Podocytes/drug effects , Reactive Oxygen Species/metabolism , Ribonucleotides/pharmacology , Sirolimus/pharmacology
20.
Aging Cell ; 11(6): 1065-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23020145

ABSTRACT

We explored molecular events associated with aging-induced matrix changes in the kidney. C57BL6 mice were studied in youth, middle age, and old age. Albuminuria and serum cystatin C level (an index of glomerular filtration) increased with aging. Renal hypertrophy was evident in middle-aged and old mice and was associated with glomerulomegaly and increase in mesangial fraction occupied by extracellular matrix. Content of collagen types I and III and fibronectin was increased with aging; increment in their mRNA varied with the phase of aging. The content of ZEB1 and ZEB2, collagen type I transcription inhibitors, and their binding to the collagen type Iα2 promoter by ChIP assay also showed age-phase-specific changes. Lack of increase in mRNA and data from polysome assay suggested decreased degradation as a potential mechanism for kidney collagen type I accumulation in the middle-aged mice. These changes occurred with increment in TGFß mRNA and protein and activation of its SMAD3 pathway; SMAD3 binding to the collagen type Iα2 promoter was also increased. TGFß-regulated microRNAs (miRs) exhibited selective regulation. The renal cortical content of miR-21 and miR-200c, but not miR-192, miR-200a, or miR-200b, was increased with aging. Increased miR-21 and miR-200c contents were associated with reduced expression of their targets, Sprouty-1 and ZEB2, respectively. These data show that aging is associated with complex molecular events in the kidney that are already evident in the middle age and progress to old age. Age-phase-specific regulation of matrix protein synthesis occurs and involves matrix protein-specific transcriptional and post-transcriptional mechanisms.


Subject(s)
Aging/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental , Glomerular Mesangium/metabolism , Kidney Cortex/metabolism , Adaptor Proteins, Signal Transducing , Aging/metabolism , Aging/pathology , Animals , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Cystatin C/blood , Extracellular Matrix/pathology , Female , Glomerular Filtration Rate , Glomerular Mesangium/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kidney Cortex/pathology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic , Protein Binding , Proteolysis , RNA, Messenger/biosynthesis , Repressor Proteins/genetics , Repressor Proteins/metabolism , Serum Albumin/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
SELECTION OF CITATIONS
SEARCH DETAIL
...