Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Genet ; 52(8): 790-799, 2020 08.
Article in English | MEDLINE | ID: mdl-32690948

ABSTRACT

Epigenetic processes govern prostate cancer (PCa) biology, as evidenced by the dependency of PCa cells on the androgen receptor (AR), a prostate master transcription factor. We generated 268 epigenomic datasets spanning two state transitions-from normal prostate epithelium to localized PCa to metastases-in specimens derived from human tissue. We discovered that reprogrammed AR sites in metastatic PCa are not created de novo; rather, they are prepopulated by the transcription factors FOXA1 and HOXB13 in normal prostate epithelium. Reprogrammed regulatory elements commissioned in metastatic disease hijack latent developmental programs, accessing sites that are implicated in prostate organogenesis. Analysis of reactivated regulatory elements enabled the identification and functional validation of previously unknown metastasis-specific enhancers at HOXB13, FOXA1 and NKX3-1. Finally, we observed that prostate lineage-specific regulatory elements were strongly associated with PCa risk heritability and somatic mutation density. Examining prostate biology through an epigenomic lens is fundamental for understanding the mechanisms underlying tumor progression.


Subject(s)
Prostatic Neoplasms/genetics , Cell Line , Cell Line, Tumor , Disease Progression , Epigenomics/methods , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Male , Prostate/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Regulatory Sequences, Nucleic Acid/genetics
2.
Drug Metab Dispos ; 44(7): 1050-60, 2016 07.
Article in English | MEDLINE | ID: mdl-27044799

ABSTRACT

The hepatocyte nuclear factors, Hnf1a and Hnf4a, in addition to playing key roles in determining hepatocyte fate, have been implicated as candidate lineage-determining transcription factors in the kidney proximal tubule (PT) [Martovetsky et. al., (2012) Mol Pharmacol 84:808], implying an additional level of regulation that is potentially important in developmental and/or tissue-engineering contexts. Mouse embryonic fibroblasts (MEFs) transduced with Hnf1a and Hnf4a form tight junctions and express multiple PT drug transporters (e.g., Slc22a6/Oat1, Slc47a1/Mate1, Slc22a12/Urat1, Abcg2/Bcrp, Abcc2/Mrp2, Abcc4/Mrp4), nutrient transporters (e.g., Slc34a1/NaPi-2, Slco1a6), and tight junction proteins (occludin, claudin 6, ZO-1/Tjp1, ZO-2/Tjp2). In contrast, the coexpression (with Hnf1a and Hnf4a) of GATA binding protein 4 (Gata4), as well as the forkhead box transcription factors, Foxa2 and Foxa3, in MEFs not only downregulates PT markers but also leads to upregulation of several hepatocyte markers, including albumin, apolipoprotein, and transferrin. A similar result was obtained with primary mouse PT cells. Thus, the presence of Gata4 and Foxa2/Foxa3 appears to alter the effect of Hnf1a and Hnf4a by an as-yet unidentified mechanism, leading toward the generation of more hepatocyte-like cells as opposed to cells exhibiting PT characteristics. The different roles of Hnf4a in the kidney and liver was further supported by reanalysis of ChIP-seq data, which revealed Hnf4a colocalization in the kidney near PT-enriched genes compared with those genes enriched in the liver. These findings provide valuable insight, not only into the developmental, and perhaps organotypic, regulation of drug transporters, drug-metabolizing enzymes, and tight junctions, but also for regenerative medicine strategies aimed at restoring the function of the liver and/or kidney (acute kidney injury, AKI; chronic kidney disease, CKD).


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cell Differentiation , Cell Lineage , Hepatocytes/metabolism , Kidney Tubules, Proximal/metabolism , Liver/metabolism , Organic Cation Transport Proteins/metabolism , Tight Junction Proteins/metabolism , Transcription Factors/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Biomarkers/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Kidney Tubules, Proximal/cytology , Liver/cytology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Multidrug Resistance-Associated Protein 2 , Organic Cation Transport Proteins/genetics , Rats , Tight Junction Proteins/genetics , Transcription Factors/genetics , Transfection
3.
Physiol Rev ; 95(1): 83-123, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25540139

ABSTRACT

The organic anion transporter (OAT) subfamily, which constitutes roughly half of the SLC22 (solute carrier 22) transporter family, has received a great deal of attention because of its role in handling of common drugs (antibiotics, antivirals, diuretics, nonsteroidal anti-inflammatory drugs), toxins (mercury, aristolochic acid), and nutrients (vitamins, flavonoids). Oats are expressed in many tissues, including kidney, liver, choroid plexus, olfactory mucosa, brain, retina, and placenta. Recent metabolomics and microarray data from Oat1 [Slc22a6, originally identified as NKT (novel kidney transporter)] and Oat3 (Slc22a8) knockouts, as well as systems biology studies, indicate that this pathway plays a central role in the metabolism and handling of gut microbiome metabolites as well as putative uremic toxins of kidney disease. Nuclear receptors and other transcription factors, such as Hnf4α and Hnf1α, appear to regulate the expression of certain Oats in conjunction with phase I and phase II drug metabolizing enzymes. Some Oats have a strong selectivity for particular signaling molecules, including cyclic nucleotides, conjugated sex steroids, odorants, uric acid, and prostaglandins and/or their metabolites. According to the "Remote Sensing and Signaling Hypothesis," which is elaborated in detail here, Oats may function in remote interorgan communication by regulating levels of signaling molecules and key metabolites in tissues and body fluids. Oats may also play a major role in interorganismal communication (via movement of small molecules across the intestine, placental barrier, into breast milk, and volatile odorants into the urine). The role of various Oat isoforms in systems physiology appears quite complex, and their ramifications are discussed in the context of remote sensing and signaling.


Subject(s)
Gene Expression Regulation/physiology , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Humans , Organic Anion Transporters/chemistry , Tissue Distribution
4.
Nephron Exp Nephrol ; 126(2): 101, 2014.
Article in English | MEDLINE | ID: mdl-24854650

ABSTRACT

BACKGROUND: With the rate of kidney disease on the rise, and a serious imbalance between the number of patients requiring a kidney transplant and the number of available donor kidneys, it is becoming increasingly important to develop alternative strategies to restore organ function to diminish the need for human donors. SUMMARY: We review the current progress and future directions of a subset of these strategies which are ultimately aimed towards bioengineering a functional, implantable, kidney-like tissue construct or organoid that might be genetically matched to the patient. KEY MESSAGES: By combining the knowledge about normal kidney development with the rapidly growing knowledge in the field of cell differentiation and transdifferentiation, there is hope that partial or complete kidney function can be restored in patients with kidney disease - including genetic disorders, acute kidney injury, or chronic kidney disease - with tissue-engineered construct(s).


Subject(s)
Kidney Diseases/therapy , Kidney/cytology , Tissue Engineering/methods , Animals , Cell Transdifferentiation , Humans , Kidney/embryology , Morphogenesis
5.
Curr Opin Organ Transplant ; 19(2): 153-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24503495

ABSTRACT

PURPOSE OF REVIEW: Chronic kidney disease is expected to continue to be a major health problem. There remains a huge shortage of donor tissues. A potential solution is to engineer a kidney-like tissue capable of performing differentiated renal functions. These functions are strikingly dependent upon appropriate three-dimensional relationships established during development, including those arising from branching morphogenesis of the ureteric bud. RECENT FINDINGS: The ureteric bud, an 'iterative tip-stalk generator' (ITSG) forming the scaffold around which the kidney is built, can be cultured and propagated ex vivo while retaining the capacity to induce and appropriately interact with nascent nephrons. Progress has been made toward construction of a ureteric bud from cells. SUMMARY: The myriad functions of the kidney are critically dependent upon its three-dimensional spatial architecture established by branching of the ureteric bud. Ureteric bud branching morphogenesis can be recapitulated ex vivo; we discuss how this intrinsic property of the ureteric bud might be exploited for engineering of kidney-like tissues potentially useful for the treatment of chronic kidney disease, acute kidney injury, and/or other renal diseases.


Subject(s)
Acute Kidney Injury/therapy , Kidney/physiology , Regeneration/physiology , Renal Insufficiency, Chronic/therapy , Tissue Engineering/methods , Ureter/embryology , Acute Kidney Injury/physiopathology , Animals , Cell Differentiation/physiology , Humans , Morphogenesis , Renal Insufficiency, Chronic/physiopathology , Ureter/cytology
6.
Mol Pharmacol ; 84(6): 808-23, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24038112

ABSTRACT

The transcriptional regulation of drug-metabolizing enzymes and transporters (here collectively referred to as DMEs) in the developing proximal tubule (PT) is not well understood. As in the liver, DME regulation in the PT may be mediated through nuclear receptors, which are thought to "sense" deviations from homeostasis by being activated by ligands, some of which are handled by DMEs, including drug transporters. Systems analysis of transcriptomic data during kidney development predicted a set of upstream transcription factors, including hepatocyte nuclear factor 4α (Hnf4a) and Hnf1a, as well as Nr3c1 (Gr), Nfe2l2 (Nrf2), peroxisome proliferator-activated receptor α (Pparα), and Tp53. Motif analysis of cis-regulatory enhancers further suggested that Hnf4a and Hnf1a are the main transcriptional regulators of DMEs in the PT. Available expression data from tissue-specific Hnf4a knockout tissues revealed that distinct subsets of DMEs were regulated by Hnf4a in a tissue-specific manner. Chromatin immunoprecipitation combined with massively parallel DNA sequencing was performed to characterize the PT-specific binding sites of Hnf4a in rat kidneys at three developmental stages (prenatal, immature, adult), which further supported a major role for Hnf4a in regulating PT gene expression, including DMEs. In ex vivo kidney organ culture, an antagonist of Hnf4a (but not a similar inactive compound) led to predicted changes in DME expression, including among others Fmo1, Cyp2d2, Cyp2d4, Nqo2, as well as organic cation transporters and organic anion transporters Slc22a1 (Oct1), Slc22a2 (Oct2), Slc22a6 (Oat1), Slc22a8 (Oat3), and Slc47a1 (Mate1). Conversely, overexpression of Hnf1a and Hnf4a in primary mouse embryonic fibroblasts, sometimes considered a surrogate for mesenchymal stem cells, induced expression of several of these proximal tubule DMEs, as well as epithelial markers and a PT-enriched brush border marker Ggt1. These cells had organic anion transporter function. Taken together, the data strongly supports a critical role for HNF4a and Hnf1a in the tissue-specific regulation of drug handling and differentiation toward a PT-like cellular identity. We discuss our data in the context of the "remote sensing and signaling hypothesis" (Ahn and Nigam, 2009; Wu et al., 2011).


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Inactivation, Metabolic/genetics , Kidney/metabolism , Animals , Cells, Cultured , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/genetics , Kidney/embryology , Kidney/growth & development , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/growth & development , Kidney Tubules, Proximal/metabolism , Lentivirus/genetics , Metabolic Detoxication, Phase I/genetics , Metabolic Detoxication, Phase II/genetics , Mice , Mice, Knockout , Protein Binding , Rats , Tissue Culture Techniques
7.
PLoS One ; 7(7): e40796, 2012.
Article in English | MEDLINE | ID: mdl-22808265

ABSTRACT

Proper physiological function in the pre- and post-natal proximal tubule of the kidney depends upon the acquisition of selective permeability, apical-basolateral epithelial polarity and the expression of key transporters, including those involved in metabolite, toxin and drug handling. Particularly important are the SLC22 family of transporters, including the organic anion transporters Oat1 (originally identified as NKT) and Oat3 as well as the organic cation transporter Oct1. In ex vivo cultures of metanephric mesenchyme (MM; the embryonic progenitor tissue of the nephron) Oat function was evident before completion of nephron segmentation and corresponded with the maturation of tight junctions as measured biochemically by detergent extractability of the tight junction protein, ZO-1. Examination of available time series microarray data sets in the context of development and differentiation of the proximal tubule (derived from both in vivo and in vitro/ex vivo developing nephrons) allowed for correlation of gene expression data to biochemically and functionally defined states of development. This bioinformatic analysis yielded a network of genes with connectivity biased toward Hnf4α (but including Hnf1α, hyaluronic acid-CD44, and notch pathways). Intriguingly, the Oat1 and Oat3 genes were found to have strong temporal co-expression with Hnf4α in the cultured MM supporting the notion of some connection between the transporters and this transcription factor. Taken together with the ChIP-qPCR finding that Hnf4α occupies Oat1, Oat3, and Oct1 proximal promoters in the in vivo differentiating rat kidney, the data suggest a network of genes with Hnf4α at its center plays a role in regulating the terminal differentiation and capacity for drug and toxin handling by the nascent proximal tubule of the kidney.


Subject(s)
Gene Expression Regulation, Developmental , Kidney Tubules, Proximal/embryology , Kidney Tubules, Proximal/metabolism , Organic Anion Transporters/genetics , Animals , Anions , Biomarkers/metabolism , Cations , Cell Differentiation , Chromatin Immunoprecipitation , Computational Biology , Epithelium/embryology , Epithelium/metabolism , Genome/genetics , Hepatocyte Nuclear Factor 4/metabolism , Kidney Tubules, Proximal/cytology , Nephrons/embryology , Nephrons/metabolism , Oligonucleotide Array Sequence Analysis , Organic Anion Transporters/metabolism , Organogenesis/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Transport , Rats , Reproducibility of Results , Signal Transduction/genetics , Tight Junctions/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...