Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Genes Dis ; 9(1): 187-200, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35005118

ABSTRACT

TSC renal cystic disease is poorly understood and has no approved treatment. In a new principal cell-targeted murine model of Tsc cystic disease, the renal cystic epithelium is mostly composed of type A intercalated cells with an intact Tsc2 gene confirmed by sequencing, although these cells exhibit a Tsc-mutant disease phenotype. We used a newly derived targeted murine model in lineage tracing and extracellular vesicle (EV) characterization experiments and a cell culture model in EV characterization and cellular induction experiments to understand TSC cystogenesis. Using lineage tracing experiments, we found principal cells undergo clonal expansion but contribute very few cells to the cyst. We determined that cystic kidneys contain more interstitial EVs than noncystic kidneys, excrete fewer EVs in urine, and contain EVs in cyst fluid. Moreover, the loss of Tsc2 gene in EV-producing cells greatly changes the effect of EVs on renal tubular epithelium, such that the epithelium develops increased secretory and proliferative pathway activity. We demonstate that the mTORC1 pathway activity is independent form the EV production, and that the EV effects for a single cell line can vary significantly. TSC cystogenesis involves significant contribution from genetically intact cells conscripted to the mutant phenotype by mutant cell derived EVs.

2.
Front Physiol ; 12: 630933, 2021.
Article in English | MEDLINE | ID: mdl-34262466

ABSTRACT

In tuberous sclerosis complex (TSC), Tsc2 mutations are associated with more severe disease manifestations than Tsc1 mutations and the role of extracellular vesicles (EVs) in this context is not yet studied. We report a comparative analysis of EVs derived from isogenic renal cells except for Tsc1 or Tsc2 gene status and hypothesized that in spite of having similar physical characteristics, EVs modulate signaling pathways differently, thus leading to TSC heterogenicity. We used mouse inner medullary collecting duct (mIMCD3) cells with the Tsc1 (T1G cells) or Tsc2 (T2J cells) gene disrupted by CRISPR/CAS9. EVs were isolated from the cell culture media by size-exclusion column chromatography followed by detailed physical and chemical characterization. Physical characterization of EVs was accessed by tunable resistive pulse sensing and dynamic light scattering, revealing similar average sizes and zeta potentials (at pH 7.4) for EVs from mIMCD3 (123.5 ± 5.7 nm and -16.3 ± 2.1 mV), T1G cells (131.5 ± 8.3 nm and -19.8 ± 2.7 mV), and T2J cells (127.3 ± 4.9 nm and -20.2 ± 2.1 mV). EVs derived from parental mIMCD3 cells and both mutated cell lines were heterogeneous (>90% of EVs < 150 nm) in nature. Immunoblotting detected cilial Hedgehog signaling protein Arl13b; intercellular proteins TSG101 and Alix; and transmembrane proteins CD63, CD9, and CD81. Compared to Tsc2 deletion, Tsc1 deletion cells had reduced EV production and release rates. EVs from Tsc1 mutant cells altered mTORC1, autophagy, and ß-catenin pathways differently than EVs from Tsc2-mutated cells. Quantitative PCR analysis revealed the down regulation of miR-212a-3p and miR-99a-5p in EVs from Tsc2-mutated cells compared to EVs from Tsc1-mutant cells. Thus, EV-derived miR-212-3p and mIR-99a-5p axes may represent therapeutic targets or biomarkers for TSC disease.

3.
PLoS One ; 14(3): e0214053, 2019.
Article in English | MEDLINE | ID: mdl-30883612

ABSTRACT

Primary cilia of renal epithelial cells express several members of the transient receptor potential (TRP) class of cation-conducting channel, including TRPC1, TRPM3, TRPM4, TRPP2, and TRPV4. Some cases of autosomal dominant polycystic kidney disease (ADPKD) are caused by defects in TRPP2 (also called polycystin-2, PC2, or PKD2). A large-conductance, TRPP2-dependent channel in renal cilia has been well described, but it is not known whether this channel includes any other protein subunits. To study this question, we investigated the pharmacology of the TRPP2-dependent channel through electrical recordings from the cilia of mIMCD-3 cells, a murine cell line of renal epithelial origin. The pharmacology was found to match that of TRPM3 channels. The ciliary TRPP2-dependent channel is known to be activated by depolarization and by increasing cytoplasmic Ca2+. This activation was greatly enhanced by external pregnenolone sulfate, an agonist of TRPM3 channels. Pregnenolone sulfate did not change the single-channel current-voltage relation. The channels were effectively blocked by isosakuranetin, a specific inhibitor of TRPM3 channels. Both pregnenolone sulfate and isosakuranetin were effective at concentrations as low as 1 µM. Knocking out TRPM3 by CRISPR/Cas9 genome editing eliminated the ciliary channel. Thus the channel is both TRPM3-dependent and TRPP2-dependent, suggesting that it may include both types of subunit. Knocking out TRPM3 did not change the level of TRPP2 protein in the cilia, so it is unlikely that the absence of functional ciliary channels results from a failure of trafficking.


Subject(s)
Kidney/metabolism , TRPM Cation Channels/metabolism , TRPP Cation Channels/metabolism , Animals , Calcium Signaling , Cell Line , Cilia/drug effects , Cilia/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Flavonoids/pharmacology , Gene Knockout Techniques , Humans , Kidney/cytology , Mice , Pregnenolone/pharmacology , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics , TRPP Cation Channels/antagonists & inhibitors , TRPP Cation Channels/genetics
4.
Physiol Rep ; 7(2): e13983, 2019 01.
Article in English | MEDLINE | ID: mdl-30675765

ABSTRACT

Tuberous sclerosis complex (TSC) is a tumor predisposition syndrome with significant renal cystic and solid tumor disease. While the most common renal tumor in TSC, the angiomyolipoma, exhibits a loss of heterozygosity associated with disease, we have discovered that the renal cystic epithelium is composed of type A intercalated cells that have an intact Tsc gene that have been induced to exhibit Tsc-mutant disease phenotype. This mechanism appears to be different than that for ADPKD. The murine models described here closely resemble the human disease and both appear to be mTORC1 inhibitor responsive. The induction signaling driving cystogenesis may be mediated by extracellular vesicle trafficking.


Subject(s)
Kidney Diseases, Cystic/pathology , Tuberous Sclerosis/pathology , Animals , Disease Models, Animal , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Female , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism
5.
Sci Rep ; 8(1): 14161, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242175

ABSTRACT

Tuberous Sclerosis Complex (TSC), a rare genetic disorder with mechanistic target of rapamycin complex 1 (mTORC1) hyperactivation, is characterized by multi-organ hamartomatous benign tumors including brain, skin, kidney, and lung (Lymphangioleiomyomatosis). mTORC1 hyperactivation drives metabolic reprogramming including glucose and glutamine utilization, protein, nucleic acid and lipid synthesis. To investigate the mechanisms of exogenous nutrients uptake in Tsc2-deficient cells, we measured dextran uptake, a polysaccharide internalized via macropinocytosis. Tsc2-deficient cells showed a striking increase in dextran uptake (3-fold, p < 0.0001) relative to Tsc2-expressing cells, which was decreased (3-fold, p < 0.0001) with mTOR inhibitor, Torin1. Pharmacologic and genetic inhibition of the lipid kinase Vps34 markedly abrogated uptake of Dextran in Tsc2-deficient cells. Macropinocytosis was further increased in Tsc2-deficient cells that lack autophagic mechanisms, suggesting that autophagy inhibition leads to dependence on exogenous nutrient uptake in Tsc2-deficient cells. Treatment with a macropinocytosis inhibitor, ethylisopropylamiloride (EIPA), resulted in selective growth inhibition of Atg5-deficient, Tsc2-deficient cells (50%, p < 0.0001). Genetic inhibition of autophagy (Atg5-/- MEFs) sensitized cells with Tsc2 downregulation to the Vps34 inhibitor, SAR405, resulting in growth inhibition (75%, p < 0.0001). Finally, genetic downregulation of Vps34 inhibited tumor growth and increased tumor latency in an in vivo xenograft model of TSC. Our findings show that macropinocytosis is upregulated with Tsc2-deficiency via a Vps34-dependent mechanism to support their anabolic state. The dependence of Tsc2-deficient cells on exogenous nutrients may provide novel approaches for the treatment of TSC.


Subject(s)
Carcinogenesis/metabolism , Class III Phosphatidylinositol 3-Kinases/metabolism , Pinocytosis/physiology , Tuberous Sclerosis/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Autophagy/drug effects , Autophagy/physiology , Autophagy-Related Protein 5/metabolism , Carcinogenesis/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dextrans/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Pinocytosis/drug effects , Protein Kinase Inhibitors/pharmacology , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis Complex 2 Protein/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
6.
Nat Rev Nephrol ; 14(11): 704-716, 2018 11.
Article in English | MEDLINE | ID: mdl-30232410

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant disease characterized by hamartomatous tumours of the brain, heart, skin, lung and kidney. Patients with TSC show a diverse range of neurological features (including seizures, cognitive disability and autism) and renal manifestations (including angiomyolipomas, epithelial cysts and renal cell carcinoma (RCC)). TSC is caused by inactivating mutations in TSC1 and TSC2, which encode hamartin and tuberin, respectively. These two proteins form a complex that negatively regulates mechanistic target of rapamycin complex 1 (mTORC1), a master regulator of cellular growth and metabolism. In clinical trials, allosteric inhibitors of mTORC1 decrease angiomyolipoma size, but the tumours regrow after treatment cessation. Therefore, the development of strategies to eliminate rather than suppress angiomyolipomas remains a high priority. This Review describes important advances in the TSC field and highlights several remaining critical knowledge gaps: the factors that promote aggressive behaviour by a subset of TSC-associated RCCs; the molecular mechanisms underlying early-onset cystogenesis in TSC2-PKD1 contiguous gene deletion syndrome; the effect of early, long-term mTORC1 inhibition on the development of TSC renal disease; and the identification of the cell or cells of origin of angiomyolipomas.


Subject(s)
Angiomyolipoma/drug therapy , Carcinoma, Renal Cell/genetics , Kidney Diseases, Cystic/genetics , Kidney Neoplasms/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Tuberous Sclerosis/drug therapy , Tuberous Sclerosis/metabolism , Angiomyolipoma/genetics , Animals , Autophagy/drug effects , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Disease Models, Animal , Humans , In Vitro Techniques , Kidney Diseases, Cystic/drug therapy , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Lipid Metabolism/drug effects , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Signal Transduction , Stress, Physiological/drug effects , Tuberous Sclerosis/complications , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tumor Microenvironment/drug effects
7.
Mol Cancer Res ; 15(10): 1318-1330, 2017 10.
Article in English | MEDLINE | ID: mdl-28710231

ABSTRACT

Tuberous sclerosis complex (TSC) is a tumor-suppressor syndrome affecting multiple organs, including the brain, skin, kidneys, heart, and lungs. TSC is associated with mutations in TSC1 or TSC2, resulting in hyperactivation of mTOR complex 1 (mTORC1). Clinical trials demonstrate that mTORC1 inhibitors decrease tumor volume and stabilize lung function in TSC patients; however, mTOR inhibitors are cytostatic not cytocidal, and long-term benefits and toxicities are uncertain. Previously, we identified rapamycin-insensitive upregulation of cyclooxygenase 2 (PTGS2/COX2) and prostaglandin E2 (PGE2) production in TSC2-deficient cells and postulated that the action of excess PGE2 and its cognate receptors (EP) contributes to cell survival. In this study, we identify upregulation of EP3 (PTGER3) expression in TSC2-deficient cells, TSC renal angiomyolipomas, lymphangioleiomyomatosis lung nodules, and epileptic brain tubers. TSC2 negatively regulated EP3 expression via Rheb in a rapamycin-insensitive manner. The EP3 antagonist, L-798106, selectively suppressed the viability of TSC2-deficient cells in vitro and decreased the lung colonization of TSC2-deficient cells. Collectively, these data reveal a novel function of TSC2 and Rheb in the regulation of EP3 expression and cell viability.Implications: Therapeutic targeting of an aberrant PGE2-EP3 signaling axis may have therapeutic benefit for TSC patients and for other mTOR-hyperactive neoplasms. Mol Cancer Res; 15(10); 1318-30. ©2017 AACR.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Tumor Suppressor Proteins/metabolism , Angiomyolipoma/genetics , Angiomyolipoma/metabolism , Animals , Brain/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Child , Child, Preschool , Epilepsy/genetics , Epilepsy/metabolism , Female , Humans , Infant , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/metabolism , Male , Mice , Mutation , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency , Up-Regulation
8.
Am J Physiol Renal Physiol ; 313(3): F699-F705, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28679593

ABSTRACT

The early events that signal renal dysfunction in presymptomatic heart failure are unclear. We tested the hypothesis that functional and mechanistic changes occur in the kidney that precede the development of symptomatic heart failure. We employed a transgenic mouse model with cardiomyocyte-specific overexpression of mutant α-B-crystallin that develops slowly progressive cardiomyopathy. Presymptomatic transgenic mice displayed an increase in serum creatinine (1.17 ± 0.34 vs. wild type 0.65 ± 0.16 mg/dl, P < 0.05) and in urinary neutrophil gelatinase-associated lipocalin (NGAL; 278.92 ± 176.24 vs. wild type 49.11 ± 22.79 ng/ml, P < 0.05) but no renal fibrosis. Presymptomatic transgenic mouse kidneys exhibited a twofold upregulation of the Ren1 gene, marked overexpression of renin protein in the tubules, and a worsened response to ischemia-reperfusion injury based on serum creatinine (2.77 ± 0.66 in transgenic mice vs. 2.01 ± 0.58 mg/dl in wild type, P < 0.05), urine NGAL (9,198.79 ± 3,799.52 in transgenic mice vs. 3,252.94 ± 2,420.36 ng/ml in wild type, P < 0.05), tubule dilation score (3.4 ± 0.5 in transgenic mice vs. 2.6 ± 0.5 in wild type, P < 0.05), tubule cast score (3.2 ± 0.4 in transgenic mice vs. 2.5 ± 0.5 in wild type, P < 0.05), and TdT-mediated dUTP nick-end labeling (TUNEL)-positive nuclei (10.1 ± 2.1 in the transgenic group vs. 5.7 ± 1.6 per 100 cells counted in wild type, P < 0.01). Our findings indicate functional renal impairment, urinary biomarker elevations, and induction of renin gene and protein expression in the kidney that occur in early presymptomatic heart failure, which increase the susceptibility to subsequent acute kidney injury.


Subject(s)
Acute Kidney Injury/etiology , Cardio-Renal Syndrome/etiology , Cardiomyopathies/etiology , Heart Failure/etiology , Kidney/pathology , Reperfusion Injury/etiology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Asymptomatic Diseases , Biomarkers/urine , Cardio-Renal Syndrome/genetics , Cardio-Renal Syndrome/pathology , Cardio-Renal Syndrome/physiopathology , Cardiomyopathies/genetics , Creatinine/urine , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Heart Failure/genetics , Kidney/metabolism , Kidney/physiopathology , Lipocalin-2/urine , Mice, Transgenic , Mutation , Phenotype , Renin/genetics , Renin/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Time Factors , Up-Regulation , alpha-Crystallin B Chain/genetics
9.
J Pediatr ; 187: 318-322.e2, 2017 08.
Article in English | MEDLINE | ID: mdl-28600153

ABSTRACT

Renal cysts occur in approximately 50% of patients with tuberous sclerosis complex, but their clinical significance and response to treatment are unknown. Abdominal imaging of 15 patients with tuberous sclerosis complex-associated renal cystic disease who had received mammalian target of rapamycin inhibitor therapy for other tuberous sclerosis complex-related indications was evaluated. Reductions in cyst number, sum diameter, and volume were observed.


Subject(s)
Immunosuppressive Agents/therapeutic use , Kidney Diseases, Cystic/drug therapy , Sirolimus/therapeutic use , Tuberous Sclerosis/complications , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Kidney/pathology , Kidney Diseases, Cystic/etiology , Magnetic Resonance Imaging , Male , Treatment Outcome , Tuberous Sclerosis/drug therapy , Young Adult
10.
Am J Physiol Renal Physiol ; 312(4): F791-F805, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28122715

ABSTRACT

Primary cilia sense environmental conditions, including osmolality, but whether cilia participate in the osmotic response in renal epithelial cells is not known. The transient receptor potential (TRP) channels TRPV4 and TRPM3 are osmoresponsive. TRPV4 localizes to cilia in certain cell types, while renal subcellular localization of TRPM3 is not known. We hypothesized that primary cilia are required for maximal activation of the osmotic response of renal epithelial cells and that ciliary TRPM3 and TRPV4 mediate that response. Ciliated [murine epithelial cells from the renal inner medullary collecting duct (mIMCD-3) and 176-5] and nonciliated (176-5Δ) renal cells expressed Trpv4 and Trpm3 Ciliary expression of TRPM3 was observed in mIMCD-3 and 176-5 cells and in wild-type mouse kidney tissue. TRPV4 was identified in cilia and apical membrane of mIMCD-3 cells by electrophysiology and in the cell body by immunofluorescence. Hyperosmolal stress at 500 mOsm/kg (via NaCl addition) induced the osmotic response genes betaine/GABA transporter (Bgt1) and aldose reductase (Akr1b3) in all ciliated cell lines. This induction was attenuated in nonciliated cells. A TRPV4 agonist abrogated Bgt1 and Akr1b3 induction in ciliated and nonciliated cells. A TRPM3 agonist attenuated Bgt1 and Akr1b3 induction in ciliated cells only. TRPM3 knockout attenuated Akr1b3 induction. Viability under osmotic stress was greater in ciliated than nonciliated cells. Akr1b3 induction was also less in nonciliated than ciliated cells when mannitol was used to induce hyperosmolal stress. These findings suggest that primary cilia are required for the maximal osmotic response in renal epithelial cells and that TRPM3 is involved in this mechanism. TRPV4 appears to modulate the osmotic response independent of cilia.


Subject(s)
Epithelial Cells/metabolism , Kidney Tubules, Collecting/metabolism , Osmoregulation , Osmotic Pressure , TRPM Cation Channels/metabolism , Animals , CRISPR-Cas Systems , Cell Line , Cilia/metabolism , Epithelial Cells/drug effects , GABA Plasma Membrane Transport Proteins/genetics , GABA Plasma Membrane Transport Proteins/metabolism , Gene Editing , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Kidney Tubules, Collecting/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osmoregulation/drug effects , Osmotic Pressure/drug effects , Saline Solution, Hypertonic/pharmacology , Signal Transduction , TRPM Cation Channels/genetics , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transfection
11.
Am J Physiol Renal Physiol ; 309(1): F79-87, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25904703

ABSTRACT

In polycystic kidney disease (PKD), the rate of cyst formation and disease progression is highly variable. The lack of predictability in disease progression may be due to additional environmental factors or pathophysiological processes called "third hits." Diabetes is a growing epidemic, and recent studies suggest that PKD patients may be at an increased risk for this disease. We sought to determine if hyperglycemia enhances the initiation and rate of cystogenesis. Tamoxifen was administered to adult Ift88 conditional floxed allele mice to induce cilia loss in the presence of Cre. Subsequent administration of streptozotocin resulted in equivalent hyperglycemia in cilia(+) and cilia(-) mice. Hyperglycemia with loss of cilia increased the rate of cyst formation and cell proliferation. Structural and functional alterations in the kidney, including focal glomerular foot process effacement, interstitial inflammation, formation of primitive renal tubules, polyuria, and increased proteinuria, were also observed in hyperglycemic cilia(-) mice. Gene array analysis indicated enhanced Wnt and epithelial-to-mesenchymal transition signaling in the kidney of hyperglycemic cilia(-) mice. These data show that hyperglycemia, in the absence of cilia, results in renal structural and functional damage and accelerates cystogenesis, suggesting that diabetes is a risk factor in the progression of PKD.


Subject(s)
Hyperglycemia/complications , Kidney/pathology , Polycystic Kidney Diseases/etiology , Animals , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Hemodynamics , Hyperglycemia/pathology , Hyperglycemia/physiopathology , Kidney Function Tests , Male , Mice, Knockout , Polycystic Kidney Diseases/pathology , Random Allocation , Wnt Proteins/metabolism
12.
Ren Fail ; 37(3): 469-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25578815

ABSTRACT

Acute kidney injury (AKI) leads to chronic kidney disease. The mechanisms involved with recovery from AKI are poorly understood and molecular mediators responsible for healing and restoration of kidney function are understudied. We previously discovered differential expression of matrix metalloproteinase-8 (MMP-8) mRNA and protein in patients with severe sepsis associated AKI versus sepsis without AKI. Here, we demonstrate the involvement of MMP-8 in purely ischemic AKI. Mice subjected to 30 min of bilateral renal ischemia developed increased plasma creatinine and MMP-8 expression within 24 h versus sham controls. After an initial surge and subsequent return toward baseline, both kidney MMP-8 expression and activity exhibited a late increase (Days 5-7 post-ischemia reperfusion) in mice subjected to AKI. Neutrophil infiltration of the kidney was significantly higher after AKI in wild-type mice than in MMP-8 null mice, starting at 4 days. Additionally, MMP-8 null mice subjected to AKI demonstrated a persistent histopathologic and functional injury and worsened health (greater overall weight loss) versus wild-type cohorts after seven days. Taken together, our findings suggest that MMP-8 is involved with restoration of baseline kidney health after ischemic kidney injury and that a potential mechanism involves the interaction of MMP-8 and neutrophil recruitment to the site of injury.


Subject(s)
Acute Kidney Injury/complications , Kidney , Matrix Metalloproteinase 8/metabolism , Renal Insufficiency , Reperfusion Injury/complications , Animals , Apoptosis , Creatinine/blood , Disease Models, Animal , Inflammation/metabolism , Inflammation/pathology , Kidney/metabolism , Kidney/pathology , Kidney Function Tests , Mice , Recovery of Function , Renal Insufficiency/etiology , Renal Insufficiency/metabolism , Renal Insufficiency/pathology , Renal Insufficiency/physiopathology , Time Factors
13.
Am J Physiol Renal Physiol ; 307(5): F560-70, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24920756

ABSTRACT

Nearly all patients with tuberous sclerosis complex (TSC) develop renal angiomyolipomas, although the tumor cell of origin is unknown. We observed decreased renal angiomyolipoma development in patients with TSC2- polycystic kidney disease 1 deletion syndrome and hypertension that were treated from an early age with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers compared with patients who did not receive this therapy. TSC-associated renal angiomyolipomas expressed ANG II type 1 receptors, platelet-derived growth factor receptor-ß, desmin, α-smooth muscle actin, and VEGF receptor 2 but did not express the adipocyte marker S100 or the endothelial marker CD31. Sera of TSC patients exhibited increased vascular mural cell-secreted peptides, such as VEGF-A, VEGF-D, soluble VEGF receptor 2, and collagen type IV. These findings suggest that angiomyolipomas may arise from renal pericytes. ANG II treatment of angiomyolipoma cells in vitro resulted in an exaggerated intracellular Ca(2+) response and increased proliferation, which were blocked by the ANG II type 2 receptor antagonist valsartan. Blockade of ANG II signaling may have preventative therapeutic potential for angiomyolipomas.


Subject(s)
Angiomyolipoma/drug therapy , Angiomyolipoma/pathology , Angiotensin Receptor Antagonists/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Pericytes/pathology , Tuberous Sclerosis/complications , Angiomyolipoma/physiopathology , Angiotensin II/physiology , Angiotensin Receptor Antagonists/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Humans , In Vitro Techniques , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/physiopathology , Receptor, Angiotensin, Type 1/physiology , Renin-Angiotensin System/physiology , Signal Transduction/physiology , Tetrazoles/pharmacology , Tetrazoles/therapeutic use , Tuberous Sclerosis/pathology , Tuberous Sclerosis/physiopathology , Valine/analogs & derivatives , Valine/pharmacology , Valine/therapeutic use , Valsartan
14.
J Am Soc Nephrol ; 25(6): 1187-97, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24511141

ABSTRACT

Domoic acid (DA), an excitatory amino acid produced by diatoms belonging to the genus Pseudo-nitzschia, is a glutamate analog responsible for the neurologic condition referred to as amnesic shellfish poisoning. To date, the renal effects of DA have been underappreciated, although renal filtration is the primary route of systemic elimination and the kidney expresses ionotropic glutamate receptors. To characterize the renal effects of DA, we administered either a neurotoxic dose of DA or doses below the recognized limit of toxicity to adult Sv128/Black Swiss mice. DA preferentially accumulated in the kidney and elicited marked renal vascular and tubular damage consistent with acute tubular necrosis, apoptosis, and renal tubular cell desquamation, with toxic vacuolization and mitochondrial swelling as hallmarks of the cellular damage. Doses≥0.1 mg/kg DA elevated the renal injury biomarkers kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin, and doses≥0.005 mg/kg induced the early response genes c-fos and junb. Coadministration of DA with the broad spectrum excitatory amino acid antagonist kynurenic acid inhibited induction of c-fos, junb, and neutrophil gelatinase-associated lipocalin. These findings suggest that the kidney may be susceptible to excitotoxic agonists, and renal effects should be considered when examining glutamate receptor activation. Additionally, these results indicate that DA is a potent nephrotoxicant, and potential renal toxicity may require consideration when determining safe levels for human exposure.


Subject(s)
Kainic Acid/analogs & derivatives , Marine Toxins/toxicity , Neuromuscular Depolarizing Agents/toxicity , Neuromuscular Junction/drug effects , Animals , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Kainic Acid/pharmacokinetics , Kainic Acid/toxicity , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Marine Toxins/pharmacokinetics , Mice, Inbred Strains , Microscopy, Electron, Transmission , Mitochondrial Swelling/drug effects , Myocardium/metabolism , Myocardium/pathology , Neuromuscular Depolarizing Agents/pharmacokinetics , Neuromuscular Junction/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Vacuoles/pathology , Vacuoles/ultrastructure , GluK2 Kainate Receptor
15.
Am J Physiol Renal Physiol ; 306(3): F279-83, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24226526

ABSTRACT

Remarkable basic and translational advances have elucidated the role of the mammalian target of rapamycin (mTOR) signaling network in the pathogenesis of renal disease. Many of these advances originated from studies of the genetic disease tuberous sclerosis complex (TSC), leading to one of the clearest therapeutic opportunities to target mTOR with rapamycin and its analogs ("rapalogs"), which effectively inhibit mTOR complex 1 (mTORC1) by an allosteric mechanism. Clinical trials based on these discoveries have provided strongly positive therapeutic results in TSC (Bissler JJ, McCormack FX, Young LR, Elwing JM, Chuck G, Leonard JM, Schmithorst VJ, Laor T, Brody AS, Bean J, Salisbury S, Franz DN. N Engl J Med 358: 140-151, 2008; Krueger DA, Care MM, Holland K, Agricola K, Tudor C, Mangeshkar P, Wilson KA, Byars A, Sahmoud T, Franz DN. N Engl J Med 363: 1801-1811, 2010; McCormack FX, Inoue Y, Moss J, Singer LG, Strange C, Nakata K, Barker AF, Chapman JT, Brantly ML, Stocks JM, Brown KK, Lynch JP 3rd, Goldberg HJ, Young LR, Kinder BW, Downey GP, Sullivan EJ, Colby TV, McKay RT, Cohen MM, Korbee L, Taveira-DaSilva AM, Lee HS, Krischer JP, Trapnell BC. N Engl J Med 364: 1595-1606, 2011). In June 2013, the National Institute of Diabetes and Digestive and Kidney Diseases convened a small panel of physicians and scientists working in the field to identify key unknowns and define possible "next steps" in advancing understanding of TSC- and mTOR-dependent renal phenotypes. TSC-associated renal disease, which affects >85% of TSC patients, and was a major topic of discussion, focused on angiomyolipomas and epithelial cysts. The third major topic was the role of mTOR and mTOR inhibition in the pathogenesis and therapy of chronic renal disease. Renal cell carcinoma, while recognized as a manifestation of TSC that occurs in a small fraction of patients, was not the primary focus of this workshop and thus was omitted from panel discussions and from this report.


Subject(s)
TOR Serine-Threonine Kinases/antagonists & inhibitors , Tuberous Sclerosis/complications , Angiomyolipoma/drug therapy , Angiomyolipoma/etiology , Angiomyolipoma/genetics , Angiomyolipoma/prevention & control , Biomedical Research/trends , Humans , Kidney Diseases, Cystic/etiology , Renal Insufficiency, Chronic/etiology , Sirolimus/analogs & derivatives , Sirolimus/therapeutic use , Translational Research, Biomedical/trends , Tuberous Sclerosis/drug therapy , Tuberous Sclerosis/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
16.
Am J Physiol Renal Physiol ; 303(6): F831-44, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22791333

ABSTRACT

Tuberous sclerosis complex (TSC), an inherited tumor predisposition syndrome associated with mutations in TSC1 or TSC2, affects ∼1 in 6,000 individuals. Eighty percent of TSC patients develop renal angiomyolipomas, and renal involvement is a major contributor to patient morbidity and mortality. Recent work has shown that mammalian target of rapamycin complex 1 (mTORC1) inhibition caused angiomyolipoma shrinkage but that this treatment may cause cytostatic not a cytotoxic effect. Endoplasmic reticulum (ER) stress can develop in TSC-associated cells due to mTORC1-driven protein translation. We hypothesized that renal angiomyolipoma cells experience ER stress that can be leveraged to result in targeted cytotoxicity. We used immortalized human angiomyolipoma cells stably transfected with empty vector or TSC2 (encoding tuberin). Using cell number quantification and cell death assays, we found that mTORC1 inhibition with RAD001 suppressed angiomyolipoma cell proliferation in a cytostatic manner. Angiomyolipoma cells exhibited enhanced sensitivity to proteasome inhibitor-induced ER stress compared with TSC2-rescued cells. After proteasome inhibition with MG-132, Western blot analyses showed greater induction of C/EBP-homologous protein (CHOP) and more poly (ADP-ribose) polymerase (PARP) and caspase-3 cleavage, supporting ER stress-induced apoptosis. Live cell numbers also were decreased and cell death increased by MG-132 in angiomyolipoma cells compared with TSC2 rescued. Intriguingly, while pretreatment of angiomyolipoma cells with RAD001 attenuated CHOP and BiP induction, apoptotic markers cleaved PARP and caspase-3 and eukaryotic translation initiation factor 2α phosphorylation were increased, along with evidence of increased autophagy. These results suggest that human angiomyolipoma cells are uniquely susceptible to agents that exacerbate ER stress and that additional synergy may be achievable with targeted combination therapy.


Subject(s)
Angiomyolipoma/metabolism , Endoplasmic Reticulum Stress , Kidney Neoplasms/metabolism , Tuberous Sclerosis/complications , Tumor Suppressor Proteins/metabolism , Angiomyolipoma/etiology , Angiomyolipoma/genetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/metabolism , Cell Line, Tumor , Eukaryotic Initiation Factor-2/metabolism , Everolimus , Humans , Immunosuppressive Agents/pharmacology , Kidney Neoplasms/etiology , Kidney Neoplasms/genetics , Leupeptins/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Proteasome Inhibitors/pharmacology , Proteins/metabolism , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transcription Factor CHOP/metabolism , Transfection , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
17.
Am J Physiol Renal Physiol ; 302(7): F801-8, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22205228

ABSTRACT

Polycystic kidney disease (PKD) is a ciliopathy characterized by renal cysts and hypertension. These changes are presumably due to altered fluid and electrolyte transport in the collecting duct (CD). This is the site where vasopressin (AVP) stimulates vasopressin-2 receptor (V2R)-mediated aquaporin-2 (AQP2) insertion into the apical membrane. Since cysts frequently occur in the CD, we studied V2R and AQP2 trafficking and function in CD cell lines with stunted and normal cilia [cilia (-), cilia (+)] derived from the orpk mouse (hypomorph of the Tg737/Ift88 gene). Interestingly, only cilia (-) cells grown on culture dishes formed domes after apical AVP treatment. This observation led to our hypothesis that V2R mislocalizes to the apical membrane in the absence of a full-length cilium. Immunofluorescence indicated that AQP2 localizes to cilia and in a subapical compartment in cilia (+) cells, but AQP2 levels were elevated in both apical and basolateral membranes in cilia (-) cells after apical AVP treatment. Western blot analysis revealed V2R and glycosylated AQP2 in biotinylated apical membranes of cilia (-) but not in cilia (+) cells. In addition, apical V2R was functional upon apical desmopressin (DDAVP) treatment by demonstrating increased cAMP, water transport, and benzamil-sensitive equivalent short-circuit current (I(sc)) in cilia (-) cells but not in cilia (+) cells. Moreover, pretreatment with a PKA inhibitor abolished DDAVP stimulation of I(sc) in cilia (-) cells. Thus we propose that structural or functional loss of cilia leads to abnormal trafficking of AQP2/V2R leading to enhanced salt and water absorption. Whether such apical localization contributes to enhanced fluid retention and hypertension in PKD remains to be determined.


Subject(s)
Aquaporin 2/metabolism , Cilia/pathology , Kidney Tubules, Collecting/metabolism , Polycystic Kidney Diseases/metabolism , Receptors, Vasopressin/metabolism , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Isoquinolines , Kidney Tubules, Collecting/pathology , Mice , Polycystic Kidney Diseases/pathology , Sulfonamides , Vasopressins
19.
J Am Soc Nephrol ; 22(5): 839-48, 2011 May.
Article in English | MEDLINE | ID: mdl-21493775

ABSTRACT

Primary cilia dysfunction alters renal tubular cell proliferation and differentiation and associates with accelerated cyst formation in polycystic kidney disease. However, the mechanism leading from primary ciliary dysfunction to renal cyst formation is unknown. We hypothesize that primary cilia prevent renal cyst formation by suppressing pathologic tubular cell hypertrophy and proliferation. Unilateral nephrectomy initiates tubular cell hypertrophy and proliferation in the contralateral kidney and provides a tool to examine primary cilia regulation of renal hypertrophy. Conditional knockout of the primary cilia ift88 gene leads to delayed, adult-onset renal cystic disease, which provides a window of opportunity to conduct unilateral nephrectomy and examine downstream kinetics of renal hypertrophy and cyst formation. In wild-type animals, unilateral nephrectomy activated the mTOR pathway and produced appropriate structural and functional hypertrophy without renal cyst formation. However, in ift88 conditional knockout animals, unilateral nephrectomy triggered increased renal hypertrophy and accelerated renal cyst formation, leading to renal dysfunction. mTOR signaling also increased compared with wild-type animals, suggesting a mechanistic cascade starting with primary ciliary dysfunction, leading to excessive mTOR signaling and renal hypertrophic signaling and culminating in cyst formation. These data suggest that events initiating hypertrophic signaling, such as structural or functional loss of renal mass, may accelerate progression of adult polycystic kidney disease toward end-stage renal disease.


Subject(s)
Cilia/physiology , Kidney Diseases, Cystic/etiology , Kidney/pathology , Signal Transduction/physiology , Animals , Cell Proliferation , Female , Glomerular Filtration Rate , Hypertrophy , Male , Mice , TOR Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Up-Regulation
20.
PLoS One ; 6(1): e16595, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21305048

ABSTRACT

BACKGROUND: Children with complex urogenital anomalies often require bladder reconstruction. Gastrointestinal tissues used in bladder augmentations exhibit a greatly increased risk of malignancy, and the bladder microenvironment may play a role in this carcinogenesis. Investigating the influences of the bladder microenvironment on gastrointestinal and urothelial cell cycle checkpoint activation and DNA damage response has been limited by the lack of an appropriate well-differentiated urothelial cell line system. METHODOLOGY/PRINCIPAL FINDINGS: To meet this need, we have developed a well-differentiated conditionally immortalized urothelial cell line by isolating it from the H-2K(b)-tsA58 transgenic mouse. These cells express a thermosensitive SV40 large T antigen that can be deactivated by adjustment of cell culture conditions, allowing the cell line to regain normal control of the cell cycle. The isolated urothelial cell line demonstrates a polygonal, dome-shaped morphology, expresses cytokeratin 18, and exhibits well-developed tight junctions. Adaptation of the urothelial cell line to hyperosmolal culture conditions induces expression of both cytokeratin 20 and uroplakin II, markers of a superficial urothelial cell or "umbrella cell." This cell line can be maintained indefinitely in culture under permissive conditions but when cultured under non-permissive conditions, large T antigen expression is reduced substantially, leading to increased p53 activity and reduced cellular proliferation. CONCLUSIONS/SIGNIFICANCE: This new model of urothelial cells, along with gastrointestinal cell lines previously derived from the H-2K(b)-tsA58 transgenic mouse, will be useful for studying the potential mechanisms of carcinogenesis of the augmented bladder.


Subject(s)
Cell Cycle , Cell Line , DNA Damage , Urothelium/pathology , Animals , Antigens, Polyomavirus Transforming , Mice , Mice, Transgenic , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...