Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Tissue Eng Part A ; 29(3-4): 102-111, 2023 02.
Article in English | MEDLINE | ID: mdl-36274231

ABSTRACT

Patient-oriented applications of cell culture include cell therapy of organ failure like chronic renal failure. Clinical deployment of a cell-based device for artificial renal replacement requires qualitative and quantitative fidelity of a cultured cell to its in vivo counterpart. Active specific apicobasal ion transport reabsorbs 90-99% of the filtered load of salt and water in the kidney. In a bioengineered kidney, tubular transport concentrates wastes and eliminates the need for hemodialysis, but renal tubule cells in culture transport little or no salt and water due to dedifferentiation that mammalian cells undergo in vitro thereby losing important cell-type specific functions. We previously identified transforming growth factor-ß (TGF-ß) as a signaling pathway necessary for in vitro differentiation of renal tubule cells. Inhibition of TGF-ß receptor-1 led to active and inhibitable electrolyte and water transport by primary human renal tubule epithelial cells in vitro. Addition of metformin increased transport, in the context of a transient effect on 5'-AMP-activated kinase phosphorylation. These data motivated us to examine whether increased transport was an idiosyncratic effect of SB431542, probe pathways downstream of TGF-ß receptors possibly responsible for the improved differentiation, evaluate whether TGF-ß inhibition induced a range of differentiated tubule functions, and to explore crosstalk between the effects of SB431542 and metformin. In this study, we use multiple small-molecule inhibitors of canonical and noncanonical pathways to confirm that inhibition of canonical TGF-ß signaling caused the increased apicobasal transport. Hallmarks of proximal tubule cell function, including sodium reabsorption, para-amino hippurate excretion, and glucose uptake increased with TGF-ß inhibition, and the specificity of the response was shown using inhibitors of each transport protein. We did not find any evidence of crosstalk between metformin and SB431542. These data suggest that the TGF-ß signaling pathway governs multiple features of differentiation in renal proximal tubule cells in vitro. Inhibition of TGF-ß by pharmacologic or genome engineering approaches may be a viable approach to enhancing differentiated function of tubule cells in vitro. Impact statement Cell therapy of renal failure requires qualitative and quantitative fidelity between in vitro and in vivo phenotypes, which has been elusive. We show that control of transforming growth factor-ß signaling can promote differentiation of renal tubule cells grown in artificial environments. This is a key enabling step for cell therapy of renal failure.


Subject(s)
Renal Insufficiency , Transforming Growth Factor beta , Animals , Humans , Cell Differentiation , Mammals/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factors/pharmacology
2.
Tissue Eng Part A ; 28(19-20): 845-854, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36074946

ABSTRACT

A functional renal tubule bioreactor needs to reproduce the reabsorption and barrier functions of the renal tubule. Our prior work has demonstrated that primary human renal tubule cells respond favorably when cultured on substrates with elasticity similar to healthy tissue and when subjected to fluid shear stress. Polyacrylamide (PA) is widely used in industrial processes such as water purification because it is electrically neutral and chemically inert. PA is a versatile tool as the concentration and mechanical properties of the gel are easily adjusted by varying the proportions of monomer and crosslinker. Control of mechanical properties is attractive for preparing cell culture substrates with tunable stiffness, but PA's inert chemical properties require additional steps to prepare PA for cell attachment, such as chemical reactions to bind extracellular matrix proteins. Methods based on protein functionalization for cell attachment work well in the short term but fail to provide sufficient attachment to withstand the mechanical traction of fluid shear stress. In our present work, we tested the effects of subjecting primary renal tubule cells to fluid shear stress on an elastic substrate by developing a simple method of incorporating N-(3-Aminopropyl) methacrylamide hydrochloride (APMA) into PA hydrogels. Integration of APMA into the PA hydrogel formed a nondegradable elastic substrate promoting excellent long-term cell attachment despite the forces of fluid shear stress. Impact statement Cell culture on artificial materials requires the presence of ligands on the surface to which extracellular matrix receptors on the cell can bind. Simple nonspecific adsorption or covalent linkage of plasma or extracellular matrix proteins only suffices for short-term static culture. Prolonged culture may result in degradation of the original protein such that linkage is severed but new proteins secreted by the cell are blocked from adsorbing to the artificial scaffold. This results in detachment and loss of cell mass, as well as defects in monolayers. We present a simple technique to integrate amine moeities into a polyacrylamide hydrogel that resist degradation and support long-term culture.


Subject(s)
Cell Culture Techniques , Hydrogels , Humans , Hydrogels/chemistry , Ligands , Extracellular Matrix Proteins , Amines
3.
Tissue Eng Part A ; 26(19-20): 1091-1098, 2020 10.
Article in English | MEDLINE | ID: mdl-32312181

ABSTRACT

Patient-oriented applications of cell culture include cell therapy of organ failure like chronic renal failure. Clinical deployment of a cell-based device for artificial renal replacement requires qualitative and quantitative fidelity of a cultured cell to its in vivo counterpart. Active specific apicobasal ion transport reabsorbs 90-99% of the filtered load of salt and water in the kidney. In a bioengineered kidney, tubular transport concentrates wastes and eliminates the need for hemodialysis, but renal tubule cells in culture transport little or no salt and water. We previously identified transforming growth factor-beta as a signaling pathway necessary for in vitro differentiation of renal tubule cells. Inhibition of TGF-ß receptor-1 led to active inhabitable electrolyte and water transport by primary human renal tubule epithelial cells in vitro. Addition of metformin increased transport, in the context of a transient effect on 5' AMP-activated kinase phosphorylation. The signals that undermine in vitro differentiation are complex, but susceptible to pharmacologic intervention. This achievement overcomes a major hurdle limiting the development of a bioreactor of cultured cells for renal replacement therapy that encompasses not only endocrine and metabolic functions but also transport and excretion. Impact statement Clinical tissue engineering requires functional fidelity of the cultured cell to its in vivo counterpart, but this has been elusive in renal tissue engineering. Typically, renal tubule cells in culture have a flattened morphology and do not express key transporters essential to their function. In this study, we build on our prior work by using small molecules to modulate pathways affected by substrate elasticity. In doing so, we are able to enhance differentiation of these cells on conventional noncompliant substrates and show transport. These results are fundamentally enabling a new generation of cell-based renal therapies.


Subject(s)
Kidney Tubules/cytology , Metformin , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Transforming Growth Factor beta , Cells, Cultured , Humans , Metformin/pharmacology
4.
Tissue Eng Part A ; 25(13-14): 1013-1022, 2019 07.
Article in English | MEDLINE | ID: mdl-30484388

ABSTRACT

IMPACT STATEMENT: Successful clinical tissue engineering requires functional fidelity of the cultured cell to its in vivo counterpart, but this has been elusive in renal tissue engineering. Typically, renal proximal tubule cells in culture have a flattened morphology and do not express key transporters essential to their function. In this article, we show for the first time that in vitro substrate mechanical properties dictate differentiation of cultured renal proximal tubule cells. Remarkably, this effect was only discernable after 4 weeks in culture, longer than usually reported for this cell type. These results demonstrate a new tunable parameter to optimize cell differentiation in renal tissue engineering.


Subject(s)
Cell Differentiation , Elasticity , Hydrogels/pharmacology , Kidney Tubules/cytology , Animals , Aquaporins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Humans , Mice , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Sodium-Hydrogen Exchanger 3/metabolism , Transforming Growth Factor beta/metabolism
5.
Oncol Res ; 25(9): 1653-1664, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28695795

ABSTRACT

Wilms tumor (WT) is the most common renal malignancy in children and the fourth most common pediatric solid malignancy in the US. Although the mechanisms underlying the WT biology are complex, these tumors most often demonstrate activation of the canonical Wnt/ß-catenin pathway. We and others have shown that constitutive activation of ß-catenin restricted to the renal epithelium is sufficient to induce primitive renal epithelial tumors, which resemble human WT. Here we demonstrate that pharmacologic inhibition of ß-catenin gene transcription with pyrvinium inhibits tumor growth and metastatic progression in a murine model of WT. Cellular invasion is significantly inhibited in both murine WT-like and human WT cells and is accompanied by downregulation of the oncogenes Myc and Birc5 (survivin). Our studies provide proof of the concept that the canonical Wnt/ß-catenin pathway may be a novel therapeutic target in the management of WT.


Subject(s)
Anthelmintics/therapeutic use , Pyrvinium Compounds/pharmacology , Wilms Tumor/drug therapy , beta Catenin/antagonists & inhibitors , Animals , Anthelmintics/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Mice , Signal Transduction , Transcription, Genetic/drug effects , Wilms Tumor/genetics , Wilms Tumor/pathology , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
6.
Mol Carcinog ; 56(9): 2112-2126, 2017 09.
Article in English | MEDLINE | ID: mdl-28470764

ABSTRACT

Glyoxalase 2 (Glo2), a metabolic enzyme, is overexpressed in some human cancers which suggests this enzyme may play a role in human tumorigenesis. In prostate cancer (PCa), the role of Glo2 has been scarcely investigated and there are no studies addressing a causative involvement of this protein in this neoplasia. Here, we examined the immunohistochemical profile of Glo2 in human PCa and benign adjacent tissues and investigated Glo2 involvement in PCa development in human prostate cell lines. PCa and matched adjacent normal tissues were obtained from paraffin sections of primary PCa from 20 patients who had undergone radical prostatectomy. Histopathological diagnosis was confirmed for each sample. Glo2 expression analysis was performed by immunohistochemistry in prostate tissues, and by qRT-PCR and immunoblotting in prostate cell lines. The causative and mechanistic role of Glo2 in prostate tumorigenesis was demonstrated by Glo2 ectopic expression/silencing and employing specific activators/inhibitors. Our results showed that Glo2 was selectively expressed in PCa but not in the luminal compartment of the adjacent benign epithelium consistently in all the examined 20 cases. Glo2 expression in PCa was dependent on androgen receptor (AR) and was aimed at stimulating cell proliferation and eluding apoptosis through a mechanism involving the p53-p21 axis. Glo2 was intensely expressed in the basal cells of benign glands but was not involved in PCa genesis. Our results demonstrate for the first time that Glo2 drives prostate tumorigenesis and suggest that it may represent a novel adjuvant marker in the pathological diagnosis of early PCa.


Subject(s)
Carcinogenesis , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Thiolester Hydrolases/metabolism , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Gene Knockdown Techniques , Humans , Lactoylglutathione Lyase/metabolism , Male , Prostate/metabolism , Signal Transduction , Thiolester Hydrolases/genetics
7.
Mol Oncol ; 11(4): 405-421, 2017 04.
Article in English | MEDLINE | ID: mdl-28188683

ABSTRACT

Wilms tumor (WT) is the most common renal neoplasm of childhood and affects 1 in 10 000 children aged less than 15 years. These embryonal tumors are thought to arise from primitive nephrogenic rests that derive from the metanephric mesenchyme during kidney development and are characterized partly by increased Wnt/ß-catenin signaling. We previously showed that coordinate activation of Ras and ß-catenin accelerates the growth and metastatic progression of a murine WT model. Here, we show that activating KRAS mutations can be found in human WT. In addition, high levels of phosphorylated AKT are present in the majority of WT. We further show in a mouse model and in renal epithelial cells that Ras cooperates with ß-catenin to drive metastatic disease progression and promotes in vitro tumor cell growth, migration, and colony formation in soft agar. Cellular transformation and metastatic disease progression of WT cells are in part dependent on PI3K/AKT activation and are inhibited via pharmacological inhibition of this pathway. Our studies suggest both KRAS mutations and AKT activation are present in WT and may represent novel therapeutic targets for this disease.


Subject(s)
Mutation/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Wilms Tumor/genetics , Animals , Base Sequence , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Disease Progression , Enzyme Activation , Humans , Immunohistochemistry , Kidney Neoplasms/pathology , Mice, Inbred C57BL , Neoplasm Metastasis , Wilms Tumor/metabolism , Wilms Tumor/pathology , beta Catenin/metabolism
8.
Urol Oncol ; 35(5): 286-293, 2017 05.
Article in English | MEDLINE | ID: mdl-28108243

ABSTRACT

OBJECTIVES: Our aims were to determine if targeting protein for Xklp2 (TPX2) is correlated with clear cell renal cell carcinoma (ccRCC) histology and oncologic outcomes using The Cancer Genome Atlas (TCGA) and an institutional tissue microarray (TMA). METHODS: Clinicopathological data obtained from the TCGA consisted of 415 samples diagnosed with ccRCC. A TMA was constructed from tumors of 207 patients who underwent radical nephrectomy for ccRCC. TPX2 expression by immunohistochemistry on TMA was assessed by a genitourinary pathologist. Clinical data were extracted and linked to TMA cores. TPX2 and Aurora-A mRNA coexpression were evaluated in the TCGA cohort. Overall survival (OS), cancer-specific survival, and recurrence-free survival (RFS) were analyzed using the Kaplan-Meier method and log-rank statistics. Univariate and multivariate analyses were conducted using Cox proportional hazard models. RESULTS: Median follow-up time for the TCGA cohort was 3.07 years. Aurora-A and TPX2 mRNA coexpression were significantly correlated (Pearson correlation = 0.918). High TPX2 mRNA expression was associated with advanced stage, metastasis, poor OS, and RFS. Median follow-up time for the TMA cohort was 5.3 years. Elevated TPX2 protein expression, defined as greater than 75th percentile staining intensity, was identified in 47/207 (22.7%) patients. Increased TPX2 immunostaining was associated with poor OS (P = 0.0327, 53% 5-year mortality), cancer-specific survival (P<0.01, 47.8% 5-year cancer-specific mortality), RFS (P = 0.0313, 73.6%, 5-year recurrence rate), grade, T stage, and metastasis. Multivariate analysis demonstrated elevated expression served as an independent predictor of RFS (hazard ratio = 3.62 (1.13-11.55), P = 0.029). CONCLUSIONS: We show TPX2, a regulator of Aurora-A, is associated with high grade and stage of ccRCC, and is an independent predictor of recurrence. Future studies are warranted testing its role in ccRCC biology, and its potential as a therapeutic target.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Cycle Proteins/metabolism , Kidney Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasm Recurrence, Local/metabolism , Nuclear Proteins/metabolism , RNA, Messenger/metabolism , Adult , Aged , Aged, 80 and over , Aurora Kinase A/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/secondary , Cell Cycle Proteins/genetics , DNA Methylation , Disease-Free Survival , Female , Follow-Up Studies , Humans , Kidney/metabolism , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Microtubule-Associated Proteins/genetics , Middle Aged , Neoplasm Grading , Neoplasm Staging , Nuclear Proteins/genetics , Survival Rate
9.
Prostate ; 77(2): 196-210, 2017 02.
Article in English | MEDLINE | ID: mdl-27696457

ABSTRACT

BACKGROUND: Glyoxalase 2 (Glo2), together with glyoxalase 1 (Glo1), forms the main scavenging system of methylglyoxal, a potent pro-apoptotic agent mainly generated by glycolysis. An increased rate of glycolysis is a well known signature of cancer cells. As a survival strategy, Glo1 is overexpressed in many human malignant cells, including prostate cancer (PCa), where it plays a crucial role in progression. No information is available on the role of Glo2 in the same ambit. PCa is the most common malignancy affecting men in the western world. Progression to a lethal hormone-refractory PCa represents the major concern in this pathology. Therefore, a deeper understanding of the molecular mechanisms underlying PCa invasiveness and metastasis is urgently needed in order to develop novel therapeutic targets for this incurable state of the malignancy. METHODS: Glo2 and Glo1 expression was examined in clinical samples of PCa by immunohistochemistry and in different PCa cell models by western blotting and quantitative real-time polymerase chain reaction. Gene silencing/overexpression and scavenging/inhibitory agents were used for functional analyses. RESULTS: We demonstrated that Glo2, together with Glo1, represents a novel mechanism in PCa progression as part of a pathway driven by PTEN/PI3K/AKT/mTOR signaling with involvement of PKM2 and ERα. Importantly, Glo1/Glo2 silencing did not alter the behavior of benign cells. CONCLUSIONS: Targeting glyoxalases metabolic pathway may represent a strategy to selectively inhibit advanced PCa. Prostate 77:196-210, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Carrier Proteins/metabolism , Estrogen Receptor alpha/metabolism , Lactoylglutathione Lyase/metabolism , Membrane Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Thiolester Hydrolases/metabolism , Thyroid Hormones/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Transformed , Disease Progression , Humans , Male , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Thyroid Hormone-Binding Proteins
10.
Prostate ; 76(11): 1004-18, 2016 08.
Article in English | MEDLINE | ID: mdl-27197599

ABSTRACT

BACKGROUND: Benign prostatic hyperplasia (BPH) is treated with 5α-reductase inhibitors (5ARI). These drugs inhibit the conversion of testosterone to dihydrotestosterone resulting in apoptosis and prostate shrinkage. Most patients initially respond to 5ARIs; however, failure is common especially in inflamed prostates, and often results in surgery. This communication examines a link between activation of NF-κB and increased expression of SRD5A2 as a potential mechanism by which patients fail 5ARI therapy. METHODS: Tissue was collected from "Surgical" patients, treated specifically for lower urinary tract symptoms secondary to advanced BPH; and, cancer free transition zone from "Incidental" patients treated for low grade, localized peripheral zone prostate cancer. Clinical, molecular and histopathological profiles were analyzed. Human prostatic stromal and epithelial cell lines were genetically modified to regulate NF-κB activity, androgen receptor (AR) full length (AR-FL), and AR variant 7 (AR-V7) expression. RESULTS: SRD5A2 is upregulated in advanced BPH. SRD5A2 was significantly associated with prostate volume determined by Transrectal Ultrasound (TRUS), and with more severe lower urinary tract symptoms (LUTS) determined by American Urological Association Symptom Score (AUASS). Synthesis of androgens was seen in cells in which NF-κB was activated. AR-FL and AR-V7 expression increased SRD5A2 expression while forced activation of NF-κB increased all three SRD5A isoforms. Knockdown of SRD5A2 in the epithelial cells resulted in significant reduction in proliferation, AR target gene expression, and response to testosterone (T). In tissue recombinants, canonical NF-κB activation in prostatic epithelium elevated all three SRD5A isoforms and resulted in in vivo growth under castrated conditions. CONCLUSION: Increased BPH severity in patients correlates with SRD5A2 expression. We demonstrate that NF-κB and AR-V7 upregulate SRD5A expression providing a mechanism to explain failure of 5ARI therapy in BPH patients. Prostate 76:1004-1018, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , 5-alpha Reductase Inhibitors/therapeutic use , Drug Resistance , NF-kappa B/physiology , Prostatic Hyperplasia/drug therapy , Receptors, Androgen/physiology , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/physiology , Animals , Apoptosis , Gene Expression , Gene Knockdown Techniques , Humans , Isoenzymes/genetics , Isoenzymes/physiology , Lower Urinary Tract Symptoms/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Nude , NF-kappa B/antagonists & inhibitors , Orchiectomy , Prostate/pathology , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/surgery , Prostatic Neoplasms, Castration-Resistant , Testosterone/biosynthesis , Treatment Failure , Up-Regulation
11.
Prostate ; 76(5): 491-511, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26709083

ABSTRACT

BACKGROUND: Benign prostatic hyperplasia (BPH) is a common, chronic progressive disease. Inflammation is associated with prostatic enlargement and resistance to 5α-reductase inhibitor (5ARI) therapy. Activation of the nuclear factor-kappa B (NF-κB) pathway is linked to both inflammation and ligand-independent prostate cancer progression. METHODS: NF-κB activation and androgen receptor variant (AR-V) expression were quantified in transition zone tissue samples from patients with a wide range of AUASS from incidental BPH in patients treated for low grade, localized peripheral zone prostate cancer to advanced disease requiring surgical intervention. To further investigate these pathways, human prostatic stromal and epithelial cell lines were transduced with constitutively active or kinase dead forms of IKK2 to regulate canonical NF-κB activity. The effects on AR full length (AR-FL) and androgen-independent AR-V expression as well as cellular growth and differentiation were assessed. RESULTS: Canonical NF-κB signaling was found to be upregulated in late versus early stage BPH, and to be strongly associated with non-insulin dependent diabetes mellitus. Elevated expression of AR-variant 7 (AR-V7), but not other AR variants, was found in advanced BPH samples. Expression of AR-V7 significantly correlated with the patient AUASS and TRUS volume. Forced activation of canonical NF-κB in human prostatic epithelial and stromal cells resulted in elevated expression of both AR-FL and AR-V7, with concomitant ligand-independent activation of AR reporters. Activation of NF-κB and over expression of AR-V7 in human prostatic epithelial cells maintained cell viability in the face of 5ARI treatment. CONCLUSION: Activation of NF-κB and AR-V7 in the prostate is associated with increased disease severity. AR-V7 expression is inducible in human prostate cells by forced activation of NF-κB resulting in resistance to 5ARI treatment, suggesting a potential mechanism by which patients may become resistant to 5ARI therapy.


Subject(s)
NF-kappa B/metabolism , Prostate/metabolism , Prostatic Hyperplasia/metabolism , Receptors, Androgen/metabolism , Aged , Cell Line, Tumor , Cell Survival/genetics , Disease Progression , Humans , Male , Middle Aged , Prostate/pathology , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Receptors, Androgen/genetics , Signal Transduction/genetics
12.
J Urol ; 194(6): 1762-70, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25934441

ABSTRACT

PURPOSE: Wilms tumor is the most common renal neoplasm of childhood. We previously found that restricted activation of the WNT/ß-catenin pathway in renal epithelium late in kidney development is sufficient to induce small primitive neoplasms with features of epithelial Wilms tumor. Metastatic disease progression required simultaneous addition of an activating mutation of the oncogene K-RAS. We sought to define the molecular pathways activated in this process and their relationship to human renal malignancies. MATERIALS AND METHODS: Affymetrix® expression microarray data from murine kidneys with activation of K-ras and/or Ctnnb1 (ß-catenin) restricted to renal epithelium were analyzed and compared to publicly available expression data on normal and neoplastic human renal tissue. Target genes were verified by immunoblot and immunohistochemistry. RESULTS: Mouse kidney tumors with activation of K-ras and Ctnnb1, and human renal malignancies had similar mRNA expression signatures and were associated with activation of networks centered on ß-catenin and TP53. Up-regulation of WNT/ß-catenin targets (MYC, Survivin, FOXA2, Axin2 and Cyclin D1) was confirmed by immunoblot. K-RAS/ß-catenin murine kidney tumors were more similar to human Wilms tumor than to other renal malignancies and demonstrated activation of a TP53 dependent network of genes, including the transcription factor E2F1. Up-regulation of E2F1 was confirmed in murine and human Wilms tumor samples. CONCLUSIONS: Simultaneous activation of K-RAS and ß-catenin in embryonic renal epithelium leads to neoplasms similar to human Wilms tumor and associated with activation of TP53 and up-regulation of E2F1. Further studies are warranted to evaluate the role of TP53 and E2F1 in human Wilms tumor.


Subject(s)
Disease Models, Animal , E2F1 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic/genetics , Kidney Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics , Wilms Tumor/genetics , beta Catenin/genetics , Animals , Genotype , Kidney/metabolism , Mice , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Transcriptional Activation/genetics , Transcriptome/genetics , Up-Regulation/genetics
13.
J Am Soc Nephrol ; 26(3): 597-610, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25071086

ABSTRACT

In the kidney, 20-hydroxyeicosatetraenoic acid (20-HETE) is a primary cytochrome P450 4 (Cyp4)-derived eicosanoid that enhances vasoconstriction of renal vessels and induces hypertension, renal tubular cell hypertrophy, and podocyte apoptosis. Hypertension and podocyte injury contribute to diabetic nephropathy and are strong predictors of disease progression. In this study, we defined the mechanisms whereby 20-HETE affects the progression of diabetic nephropathy. We used Cyp4a14KO male mice that exhibit androgen-sensitive hypertension due to increased Cyp4a12-mediated 20-HETE production. We show that, upon induction of diabetes type 1 via streptozotocin injection, Cyp4a14KO male mice developed worse renal disease than streptozotocin-treated wild-type mice, characterized by increased albuminuria, mesangial expansion, glomerular matrix deposition, and thickness of the glomerular basement membranes. Castration blunted androgen-mediated Cyp4a12 synthesis and 20-HETE production, normalized BP, and ameliorated renal damage in diabetic Cyp4a14KO mice. Notably, treatment with a 20-HETE antagonist or agents that normalized BP without affecting Cyp4a12 expression and 20-HETE biosynthesis also ameliorated diabetes-mediated renal damage and albuminuria in Cyp4a14KO male mice. Taken together, these results suggest that hypertension is the major contributor to 20-HETE-driven diabetes-mediated kidney injury.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Diabetic Nephropathies/etiology , Hydroxyeicosatetraenoic Acids/metabolism , Hypertension/complications , Animals , Collagen/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 4 , Diabetic Nephropathies/pathology , Glomerular Basement Membrane/pathology , Hydralazine , Hydrochlorothiazide , Hydroxyeicosatetraenoic Acids/antagonists & inhibitors , Male , Mice, Knockout , Orchiectomy , Renin-Angiotensin System , Reserpine , Sodium/metabolism
14.
Am J Pathol ; 182(2): 449-59, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23219716

ABSTRACT

Hindgut-derived endoderm can differentiate into rectal, prostatic, and bladder phenotypes. Stromal-epithelial interactions are crucial for this development; however, the precise mechanisms by which epithelium responds to stromal cues remain unknown. We have previously reported ectopic expression of peroxisome proliferator-activated receptor-γ2 (PPARγ2) increased androgen receptor expression and promoted differentiation of mouse prostate epithelium. PPARγ is also implicated in urothelial differentiation. Herein we demonstrate that knockdown of PPARγ2 in benign human prostate epithelial cells (BHPrEs) promotes urothelial transdifferentiation. Furthermore, in vitro and in vivo heterotypic tissue regeneration models with embryonic bladder mesenchyme promoted urothelial differentiation of PPARγ2-deficient BHPrE cells, and deficiency of both PPARγ isoforms 1 and 2 arrested differentiation. Because PTEN deficiency is cooperative in urothelial pathogenesis, we engineered BHPrE cells with combined knockdown of PPARγ and PTEN and performed heterotypic recombination experiments using embryonic bladder mesenchyme. Whereas PTEN deficiency alone induced latent squamous differentiation in BHPrE cells, combined PPARγ and PTEN deficiency accelerated the development of keratinizing squamous metaplasia (KSM). We further confirmed via immunohistochemistry that gene expression changes in metaplastic recombinants reflected human urothelium undergoing KSM. In summary, these data suggest that PPARγ isoform expression provides a molecular basis for observations that adult human epithelium can be transdifferentiated on the basis of heterotypic mesenchymal induction. These data also implicate PPARγ and PTEN inactivation in the development of KSM.


Subject(s)
Models, Biological , PPAR gamma/deficiency , PTEN Phosphohydrolase/deficiency , Regeneration , Urothelium/metabolism , Urothelium/pathology , Adult , Animals , Base Sequence , Cell Line , Cell Transdifferentiation , Coculture Techniques , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Hyperplasia , Mesoderm/metabolism , Mesoderm/pathology , Metaplasia , Mice , Molecular Sequence Data , PPAR gamma/metabolism , PTEN Phosphohydrolase/metabolism , Urothelium/physiopathology
15.
Am J Pathol ; 179(6): 3045-55, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21983638

ABSTRACT

Wilms' tumor (WT) is the most common childhood renal cancer. Although mutations in known tumor-associated genes (WT1, WTX, and CATNB) occur only in a third of tumors, many tumors show evidence of activated ß-catenin-dependent Wnt signaling, but the molecular mechanism by which this occurs is unknown. A key obstacle to understanding the pathogenesis of WT is the paucity of mouse models that recapitulate its features in humans. Herein, we describe a transgenic mouse model of primitive renal epithelial neoplasms that have high penetrance and mimic the epithelial component of human WT. Introduction of a stabilizing ß-catenin mutation restricted to the kidney is sufficient to induce primitive renal epithelial tumors; however, when compounded with activation of K-RAS, the mice develop large, bilateral, metastatic, multifocal primitive renal epithelial tumors that have the histologic and staining characteristics of the epithelial component of human WT. These highly malignant tumors have increased activation of the phosphatidylinositol 3-kinase-AKT and extracellular signal-regulated kinase pathways, increased expression of total and nuclear ß-catenin, and increased downstream targets of this pathway, such as c-Myc and survivin. Thus, we developed a novel mouse model in which activated K-RAS synergizes with canonical Wnt/ß-catenin signaling to form metastatic primitive renal epithelial tumors that mimic the epithelial component of human WT.


Subject(s)
Genes, ras/genetics , Kidney Neoplasms/genetics , Wilms Tumor/genetics , beta Catenin/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Cell Transformation, Neoplastic , Exons/genetics , Humans , Inhibitor of Apoptosis Proteins/metabolism , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Kidney Tubules, Proximal/pathology , MAP Kinase Signaling System/physiology , Mice , Mice, Transgenic , Mutation/genetics , Nuclear Proteins/genetics , PAX2 Transcription Factor/metabolism , PAX8 Transcription Factor , Paired Box Transcription Factors/metabolism , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Survivin , Wilms Tumor/metabolism , Wilms Tumor/pathology
16.
PLoS One ; 4(12): e8384, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-20027305

ABSTRACT

Benign prostatic hyperplasia (BPH) and prostate carcinoma (CaP) are linked to aging and the presence of androgens, suggesting that androgen regulated genes play a major role in these common diseases. Androgen regulation of prostate growth and development depends on the presence of intact epithelial-stromal interactions. Further, the prostatic stroma is implicated in BPH. This suggests that epithelial cell lines are inadequate to identify androgen regulated genes that could contribute to BPH and CaP and which could serve as potential clinical biomarkers. In this study, we used a human prostate xenograft model to define a profile of genes regulated in vivo by androgens, with an emphasis on identifying candidate biomarkers. Benign transition zone (TZ) human prostate tissue from radical prostatectomies was grafted to the sub-renal capsule site of intact or castrated male immunodeficient mice, followed by the removal or addition of androgens, respectively. Microarray analysis of RNA from these tissues was used to identify genes that were; 1) highly expressed in prostate, 2) had significant expression changes in response to androgens, and, 3) encode extracellular proteins. A total of 95 genes meeting these criteria were selected for analysis and validation of expression in patient prostate tissues using quantitative real-time PCR. Expression levels of these genes were measured in pooled RNAs from human prostate tissues with varying severity of BPH pathologic changes and CaP of varying Gleason score. A number of androgen regulated genes were identified. Additionally, a subset of these genes were over-expressed in RNA from clinical BPH tissues, and the levels of many were found to correlate with disease status. Our results demonstrate the feasibility, and some of the problems, of using a mouse xenograft model to characterize the androgen regulated expression profiles of intact human prostate tissues.


Subject(s)
Androgens/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Prostate/drug effects , Prostate/pathology , Prostatic Hyperplasia/genetics , Prostatic Neoplasms/genetics , Xenograft Model Antitumor Assays , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Mice , Prostate/metabolism , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/pathology , RNA/genetics , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects
17.
Cancer Res ; 67(17): 8188-97, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804732

ABSTRACT

The cyclin D1 oncogene encodes the regulatory subunit of a holoenzyme that phosphorylates and inactivates the Rb protein and promotes progression through G(1) to S phase of the cell cycle. Several prostate cancer cell lines and a subset of primary prostate cancer samples have increased cyclin D1 protein expression. However, the relationship between cyclin D1 expression and prostate tumor progression has yet to be clearly characterized. This study examined the effects of manipulating cyclin D1 expression in either human prostatic epithelial or stromal cells using a tissue recombination model. The data showed that overexpression of cyclin D1 in the initiated BPH-1 cell line increased cell proliferation rate but did not elicit tumorigenicity in vivo. However, overexpression of cyclin D1 in normal prostate fibroblasts (NPF) that were subsequently recombined with BPH-1 did induce malignant transformation of the epithelial cells. The present study also showed that recombination of BPH-1 + cyclin D1-overexpressing fibroblasts (NPF(cyclin D1)) resulted in permanent malignant transformation of epithelial cells (BPH-1(NPF-cyclin D1) cells) similar to that seen with carcinoma-associated fibroblasts (CAF). Microarray analysis showed that the expression profiles between CAFs and NPF(cyclin D1) cells were highly concordant including cyclin D1 up-regulation. These data indicated that the tumor-promoting activity of cyclin D1 may be tissue specific.


Subject(s)
Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Line , Disease Progression , Epithelial Cells/metabolism , Humans , Male , Mice , Mice, SCID , Neoplasm Invasiveness , Organ Specificity/genetics , Rats , Stromal Cells/metabolism , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
18.
Prostate ; 63(4): 369-84, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15937962

ABSTRACT

BACKGROUND: We have sought to develop a new in vivo model of prostate carcinogenesis using human prostatic epithelial cell cultures. Human prostate cancers frequently display DNA amplification in the 8q24 amplicon, which leads to an increase in the copy number of the c-MYC gene, a finding that suggests a role for c-MYC in human prostate carcinogenesis. In addition overexpression of c-MYC in transgenic mouse models results in prostatic carcinogenesis. METHODS: We took advantage of the ability of retroviruses to integrate foreign DNA into human prostatic epithelium (huPrE) to generate cell lines that overexpress the c-MYC protooncogene. These cells were recombined with inductive rat urogenital sinus mesenchyme and grafted beneath the renal capsule of immunocompromised rodent hosts. RESULTS: The resultant tissue displayed a phenotype consistent with a poorly differentiated human prostatic adenocarcinoma. The tumors were rapidly growing with a high proliferative index. The neoplastic cells in the tumor expressed both androgen receptors (AR) and prostate-specific antigen (PSA), both characteristic markers of human prostate cancers. Microarray analysis of human prostatic epithelial cells overexpression c-MYC identified a large number of differentially expressed genes some of which have been suggested to characterize a subset of human cancers that have myc overexpression. Specific examples were confirmed by Western blot analysis and include upregulation of c-Myb and decreased expression of PTEN. Control grafts using either uninfected huPrE or using huPrE cells infected using an empty vector expressing a green fluorescent protein tag gave rise to well differentiated benign prostatic glandular ducts. CONCLUSIONS: By using a retroviral infection strategy followed by tissue recombination we have created a model of human prostate cancer that demonstrates that the c-MYC gene is sufficient to induce carcinogenesis.


Subject(s)
Adenocarcinoma/genetics , Prostate/cytology , Prostate/physiology , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Animals , Biomarkers, Tumor , Cells, Cultured , Disease Models, Animal , Epithelium/physiology , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Humans , Male , Mice , Mice, SCID , Oligonucleotide Array Sequence Analysis , Phenotype , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , Retroviridae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...