1
|
Collins-McMillen D, De Oliveira Pessoa D, Zarrella K, Parkins CJ, Daily M, McKinzey DR, Moorman NJ, Kamil JP, Caposio P, Padi M, Goodrum FD. Viral and host network analysis of the human cytomegalovirus transcriptome in latency. Proc Natl Acad Sci U S A 2025; 122:e2416114122. [PMID: 40434638 DOI: 10.1073/pnas.2416114122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
The human cytomegalovirus (HCMV) UL135 and UL138 genes play opposing roles regulating latency and reactivation in CD34+ human progenitor cells. We designed an RNA sequencing study to compare the transcriptional profile of HCMV infection in the presence and absence of these genes using the Tohoku Hospital Pediatrics-1 (THP-1) monocytic cell line model for latency. Relative to primary cell models, THP-1 cells offer the strength of a homogenous population that uniformly silences gene expression and will synchronously reexpress viral genes following stimulation to differentiate, which models early phases of viral reactivation. The loss of UL138 resulted in elevated levels of viral gene expression and in spontaneous adhesion of distinct cell populations that support HCMV gene expression and genome synthesis. The loss of UL135 resulted in diminished viral gene expression during an initial burst that occurs as latency is established and in no expression of eleven viral genes from the ULb' region even following differentiation and reexpression of viral genes. Transcriptional network analysis revealed host transcription factors (TFs) with potential to regulate the ULb' genes in coordination with pUL135. We show that the cellular TF peroxisome proliferator-activated receptor gamma binds to the viral genome and influences the expression of UL133-UL138 locus genes. Our results define roles for UL135 and UL138 in regulation of patterns of viral gene expression for the establishment of latency and reexpression of viral genes for reactivation and reveal insights into differentiation-linked mechanisms of transcriptional control of the HCMV genome.
Collapse
Affiliation(s)
- Donna Collins-McMillen
- Bioscience Innovation Organization Five Disciplines (BIO5) Institute, University of Arizona, Tucson, AZ 85719
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719
| | | | - Kristen Zarrella
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719
| | - Christopher J Parkins
- Vaccine and Gene Therapy Institute, Oregon Health Science University, Beaverton, OR 97006
| | - Michael Daily
- Vaccine and Gene Therapy Institute, Oregon Health Science University, Beaverton, OR 97006
| | - David R McKinzey
- Bioscience Innovation Organization Five Disciplines (BIO5) Institute, University of Arizona, Tucson, AZ 85719
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719
| | - Nathaniel J Moorman
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jeremy P Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA 71103
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health Science University, Beaverton, OR 97006
| | - Megha Padi
- Bioinformatics Shared Resource, Arizona Cancer Center, University of Arizona, Tucson, AZ 85719
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85719
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721
| | - Felicia D Goodrum
- Bioscience Innovation Organization Five Disciplines (BIO5) Institute, University of Arizona, Tucson, AZ 85719
- Department of Immunobiology, University of Arizona, Tucson, AZ 85719
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85719
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721
| |
Collapse
|
2
|
Kispert A. Ureter development and associated congenital anomalies. Nat Rev Nephrol 2025; 21:366-382. [PMID: 40164775 DOI: 10.1038/s41581-025-00951-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2025] [Indexed: 04/02/2025]
Abstract
Malformations of the ureter are among the most common birth defects in humans. Although some of these anomalies are asymptomatic, others are clinically relevant, causing perinatal lethality or progressing to kidney failure in childhood. The genetic causes and developmental aetiology of ureteral anomalies are difficult to study in humans; however, embryological and genetic analyses in the mouse have provided insights into the complex developmental programmes that govern ureter formation from simple tissue primordia, and the pathological consequences that result from disruption of these programmes. Abnormalities in the formation of the nephric duct and ureteric bud lead to changes in the number of ureters (and kidneys), whereas the formation of ectopic ureteric buds, failure of the nephric duct to target the cloaca or failure of the distal ureter to mature underlie vesicoureteral reflux, ureter ectopia, ureterocoele and subsequent hydroureter. Alterations in ureter specification, early growth or cyto-differentiation programmes have now also been associated with various forms of perinatal hydroureter and hydronephrosis as a consequence of functional obstruction. The characterization of cellular processes and molecular drivers of ureterogenesis in the mouse may not only aid understanding of the aetiology of human ureteral anomalies, improve prognostication and benefit the development of therapeutic strategies, but may also prove important for efforts to generate a bioartificial organ.
Collapse
Affiliation(s)
- Andreas Kispert
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
3
|
El-Harakeh M, Rodriguez-Tirado F, Hardman AJ, Miehls A, Camara O, Grounds KM, Tocaj G, Kercsmar M, Li B, Wang X, Becknell B, Jackson AR. PPARγ promotes urothelial remodeling during urinary tract obstruction. Exp Mol Med 2025:10.1038/s12276-025-01441-0. [PMID: 40307567 DOI: 10.1038/s12276-025-01441-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 05/02/2025] Open
Abstract
Urinary tract obstruction (UTO) is a common cause of kidney injury that can result in chronic kidney disease and end-stage renal disease. Heterogeneity in the extent of obstructive renal damage in humans with UTO implies the existence of unknown mechanisms that protect against or accelerate kidney injury. Prior studies show that congenital and acquired UTO initiate a conserved, protective program of renal urothelium remodeling that culminates in expansion of uroplakin (UPK)+ cells to promote renal structural integrity. However, the cellular and molecular mechanisms that regulate UPK expression in the renal urothelium are unknown. Peroxisome proliferator-activated receptor γ (PPARγ) drives urothelial differentiation and UPK expression in other tissues but has not been investigated in the renal urothelium. Here we demonstrate that activation of PPARγ in UPK+ cells is critical for UTO-induced renal urothelium remodeling. Conditional deletion of Pparg perturbs UPK expression and accelerates parenchymal thinning during UTO, while conditional activation of PPARγ increases UPK expression and results in parenchymal preservation. This study underscores the significance of renal urothelium during UTO and shows that UTO-induced renal urothelial remodeling is achieved through activation of PPARγ. These findings form the foundation for future studies that will determine the therapeutic utility of PPARγ agonists during congenital and acquired UTO.
Collapse
Affiliation(s)
- Mohammad El-Harakeh
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Felipe Rodriguez-Tirado
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Andrew J Hardman
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Alexa Miehls
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Oumoulkhairy Camara
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Kelly M Grounds
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Glenis Tocaj
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Macie Kercsmar
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Birong Li
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Xin Wang
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Brian Becknell
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Ashley R Jackson
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH, USA.
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
4
|
Rudat C, Straube P, Hegermann J, Trowe MO, Thiesler H, Hildebrandt H, Witt L, Kispert A. PPARG contributes to urothelial integrity in the murine ureter by activating the expression of Shh and superficial cell-specific genes. Development 2025; 152:dev204324. [PMID: 40167323 PMCID: PMC12045629 DOI: 10.1242/dev.204324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
The urothelium is a stratified epithelium with an important barrier function in the urinary drainage system. The differentiation and maintenance of the three major urothelial cell types (basal, intermediate and superficial cells) is incompletely understood. Here, we show that mice with a conditional deletion of the transcription factor gene peroxisome proliferator activated receptor gamma (Pparg) in the ureteric epithelium have a dilated ureter at postnatal stages with a urothelium consisting of a layer of undifferentiated luminal cells and a layer of proliferating basal cells. Molecular analysis of fetal stages revealed that the expression of a large number of genes is not activated in superficial cells and that of a few genes, including Shh, is not activated in intermediate and basal cells. Pharmacological activation of SHH signaling in explant cultures of perinatal Pparg-deficient ureters reduced ureteral width and urothelial cell number to normal levels, increased the number of intermediate cells and slightly reduced basal cell proliferation. Our data suggest that PPARG independently activates the expression of structural genes in superficial cells and of Shh in basal and intermediate cells, and that both functions contribute to urothelial integrity.
Collapse
Affiliation(s)
- Carsten Rudat
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Philipp Straube
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, Hannover Medical School, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
- Center for Systems Neuroscience Hannover, Hannover, Germany
| | - Lisa Witt
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
5
|
Hoseini ZS, Zeinalilathori S, Fathi-karkan S, Zeinali S, Rahdar A, Siddiqui B, Kharaba Z, Pandey S. Cell-targeting nanomedicine for bladder cancer: A cellular bioengineering approach for precise drug delivery. J Drug Deliv Sci Technol 2024; 101:106220. [DOI: 10.1016/j.jddst.2024.106220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
6
|
Collins-McMillen D, De Oliveira Pessoa D, Zarrella K, Parkins CJ, Daily M, Moorman NJ, Kamil JP, Caposio P, Padi M, Goodrum FD. Viral and host network analysis of the human cytomegalovirus transcriptome in latency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.594597. [PMID: 38826434 PMCID: PMC11142044 DOI: 10.1101/2024.05.21.594597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
HCMV genes UL135 and UL138 play opposing roles regulating latency and reactivation in CD34+ human progenitor cells (HPCs). Using the THP-1 cell line model for latency and reactivation, we designed an RNA sequencing study to compare the transcriptional profile of HCMV infection in the presence and absence of these genes. The loss of UL138 results in elevated levels of viral gene expression and increased differentiation of cell populations that support HCMV gene expression and genome synthesis. The loss of UL135 results in diminished viral gene expression during an initial burst that occurs as latency is established and no expression of eleven viral genes from the ULb' region even following stimulation for differentiation and reactivation. Transcriptional network analysis revealed host transcription factors with potential to regulate the ULb' genes in coordination with pUL135. These results reveal roles for UL135 and UL138 in regulation of viral gene expression and potentially hematopoietic differentiation.
Collapse
Affiliation(s)
- Donna Collins-McMillen
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Diogo De Oliveira Pessoa
- Bioinformatics Shared Resource, Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
| | - Kristen Zarrella
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Christopher J. Parkins
- Vaccine and Gene Therapy Institute, Oregon Health Science University, Beaverton, Oregon, United States of America
| | - Michael Daily
- Vaccine and Gene Therapy Institute, Oregon Health Science University, Beaverton, Oregon, United States of America
| | - Nathaniel J. Moorman
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jeremy P. Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, United States of America
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health Science University, Beaverton, Oregon, United States of America
| | - Megha Padi
- Bioinformatics Shared Resource, Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Felicia D. Goodrum
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, United States of America
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
7
|
Gina NNT, Kuo JL, Wu ML, Chuang SM. Sesamin and sesamolin potentially inhibit adipogenesis through downregulating the peroxisome proliferator-activated receptor γ protein expression and activity in 3T3-L1 cells. Nutr Res 2024; 123:4-17. [PMID: 38228077 DOI: 10.1016/j.nutres.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024]
Abstract
Sesamin and sesamolin are major sesame lignans that have demonstrated anti-inflammatory, anticancer, and neuroprotective properties and potential benefits in the liver, cardiovascular diseases, and metabolic syndrome. However, despite previous research on their antiobesity effects and underlying mechanisms, a comprehensive investigation of these aspects is still lacking. In this study, we evaluated the regulatory effects of 20 to 80 µM sesamin and sesamolin on adipogenesis in vitro using 3T3-L1 cells as a model cell line. We hypothesized that the lignans would inhibit adipogenic differentiation in 3T3-L1 cells through the regulation of peroxisome proliferator-activated receptor γ (PPARγ). Our data indicate that sesamin and sesamolin inhibited the adipogenic differentiation of 3T3-L1 cells by dose-dependently decreasing lipid accumulation and triglyceride formation. Sesamin and sesamolin reduced the mRNA and protein expression of the adipogenesis-related transcription factors, PPARγ and CCAAT/enhancer-binding protein α, leading to the dose-dependent downregulations of their downstream targets, fatty acid binding protein 4, hormone-sensitive lipase, lipoprotein lipase, and glucose transporter 4. In addition, glucose uptake was dose-dependently attenuated by sesamin and sesamolin in both differentiated 3T3-L1 cells and HepG2 cells. Interestingly, our results suggested that sesamin and sesamolin might directly bind to PPARγ to inhibit its transcriptional activity. Finally, sesamin and sesamolin decreased the phosphorylation of 3 mitogen-activated protein kinase signaling components in differentiated 3T3-L1 cells. Taken together, our findings suggest that sesamin and sesamolin may exhibit antiobesity effects by potentially downregulating PPARγ and its downstream genes through the mitogen-activated protein kinase signaling pathway, offering important insights into the molecular mechanisms underlying the potential antiobesity effects of sesamin and sesamolin.
Collapse
Affiliation(s)
- Nelma Nyvonne Tiqu Gina
- Food Science Department, National Pingtung University of Science and Technology, Pingtung 91012, Taiwan
| | - Jui-Ling Kuo
- Food Science Department, National Pingtung University of Science and Technology, Pingtung 91012, Taiwan
| | - Mei-Li Wu
- Food Science Department, National Pingtung University of Science and Technology, Pingtung 91012, Taiwan.
| | - Show-Mei Chuang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan; Department of Law, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
8
|
Ramal M, Corral S, Kalisz M, Lapi E, Real FX. The urothelial gene regulatory network: understanding biology to improve bladder cancer management. Oncogene 2024; 43:1-21. [PMID: 37996699 DOI: 10.1038/s41388-023-02876-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
The urothelium is a stratified epithelium composed of basal cells, one or more layers of intermediate cells, and an upper layer of differentiated umbrella cells. Most bladder cancers (BLCA) are urothelial carcinomas. Loss of urothelial lineage fidelity results in altered differentiation, highlighted by the taxonomic classification into basal and luminal tumors. There is a need to better understand the urothelial transcriptional networks. To systematically identify transcription factors (TFs) relevant for urothelial identity, we defined highly expressed TFs in normal human bladder using RNA-Seq data and inferred their genomic binding using ATAC-Seq data. To focus on epithelial TFs, we analyzed RNA-Seq data from patient-derived organoids recapitulating features of basal/luminal tumors. We classified TFs as "luminal-enriched", "basal-enriched" or "common" according to expression in organoids. We validated our classification by differential gene expression analysis in Luminal Papillary vs. Basal/Squamous tumors. Genomic analyses revealed well-known TFs associated with luminal (e.g., PPARG, GATA3, FOXA1) and basal (e.g., TP63, TFAP2) phenotypes and novel candidates to play a role in urothelial differentiation or BLCA (e.g., MECOM, TBX3). We also identified TF families (e.g., KLFs, AP1, circadian clock, sex hormone receptors) for which there is suggestive evidence of their involvement in urothelial differentiation and/or BLCA. Genomic alterations in these TFs are associated with BLCA. We uncover a TF network involved in urothelial cell identity and BLCA. We identify novel candidate TFs involved in differentiation and cancer that provide opportunities for a better understanding of the underlying biology and therapeutic intervention.
Collapse
Affiliation(s)
- Maria Ramal
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sonia Corral
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Mark Kalisz
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Eleonora Lapi
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- CIBERONC, Madrid, Spain.
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
9
|
Tate T, Plumber SA, Al-Ahmadie H, Chen X, Choi W, Lu C, Viny A, Batourina E, Gartensson K, Alija B, Molotkov A, Wiessner G, McKiernan J, McConkey D, Dinney C, Czerniak B, Mendelsohn CL. Combined Mek inhibition and Pparg activation Eradicates Muscle Invasive Bladder cancer in a Mouse Model of BBN-induced Carcinogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.19.553961. [PMID: 37662238 PMCID: PMC10473651 DOI: 10.1101/2023.08.19.553961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Bladder cancers (BCs) can be divided into 2 major subgroups displaying distinct clinical behaviors and mutational profiles: basal/squamous (BASQ) tumors that tend to be muscle invasive, and luminal/papillary (LP) tumors that are exophytic and tend to be non-invasive. Pparg is a likely driver of LP BC and has been suggested to act as a tumor suppressor in BASQ tumors, where it is likely suppressed by MEK-dependent phosphorylation. Here we tested the effects of rosiglitazone, a Pparg agonist, in a mouse model of BBN-induced muscle invasive BC. Rosiglitazone activated Pparg signaling in suprabasal epithelial layers of tumors but not in basal-most layers containing highly proliferative invasive cells, reducing proliferation but not affecting tumor survival. Addition of trametinib, a MEK inhibitor, induced Pparg signaling throughout all tumor layers, and eradicated 91% of tumors within 7-days of treatment. The 2-drug combination also activated a luminal differentiation program, reversing squamous metaplasia in the urothelium of tumor-bearing mice. Paired ATAC-RNA-seq analysis revealed that tumor apoptosis was most likely linked to down-regulation of Bcl-2 and other pro-survival genes, while the shift from BASQ to luminal differentiation was associated with activation of the retinoic acid pathway and upregulation of Kdm6a, a lysine demethylase that facilitates retinoid-signaling. Our data suggest that rosiglitazone, trametinib, and retinoids, which are all FDA approved, may be clinically active in BASQ tumors in patients. That muscle invasive tumors are populated by basal and suprabasal cell types with different responsiveness to PPARG agonists will be an important consideration when designing new treatments.
Collapse
|
10
|
Pettitt GA, Hurst CD, Khan Z, McPherson HR, Dunning MC, Alder O, Platt FM, Black EVI, Burns JE, Knowles MA. Development of resistance to FGFR inhibition in urothelial carcinoma via multiple pathways in vitro. J Pathol 2023; 259:220-232. [PMID: 36385700 PMCID: PMC10107504 DOI: 10.1002/path.6034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/14/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
Alterations of fibroblast growth factor receptors (FGFRs) are common in bladder and other cancers and result in disrupted signalling via several pathways. Therapeutics that target FGFRs have now entered the clinic, but, in common with many cancer therapies, resistance develops in most cases. To model this, we derived resistant sublines of two FGFR-driven bladder cancer cell lines by long-term culture with the FGFR inhibitor PD173074 and explored mechanisms using expression profiling and whole-exome sequencing. We identified several resistance-associated molecular profiles. These included HRAS mutation in one case and reversible mechanisms resembling a drug-tolerant persister phenotype in others. Upregulated IGF1R expression in one resistant derivative was associated with sensitivity to linsitinib and a profile with upregulation of a YAP/TAZ signature to sensitivity to the YAP inhibitor CA3 in another. However, upregulation of other potential therapeutic targets was not indicative of sensitivity. Overall, the heterogeneity in resistance mechanisms and commonality of the persister state present a considerable challenge for personalised therapy. Nevertheless, the reversibility of resistance may indicate a benefit from treatment interruptions or retreatment following disease relapse in some patients. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Geoffrey A Pettitt
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| | - Carolyn D Hurst
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| | - Zubeda Khan
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| | - Helen R McPherson
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| | - Matthew C Dunning
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| | - Olivia Alder
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| | - Fiona M Platt
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| | - Emma VI Black
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| | - Julie E Burns
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| | - Margaret A Knowles
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James'sSt James's University HospitalLeedsUK
| |
Collapse
|
11
|
Jackson AR, Narla ST, Bates CM, Becknell B. Urothelial progenitors in development and repair. Pediatr Nephrol 2022; 37:1721-1731. [PMID: 34471946 PMCID: PMC8942604 DOI: 10.1007/s00467-021-05239-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
Urothelium is a specialized multilayer epithelium that lines the urinary tract from the proximal urethra to the kidney. In addition to proliferation and differentiation during development, urothelial injury postnatally triggers a robust regenerative capacity to restore the protective barrier between the urine and tissue. Mounting evidence supports the existence of dedicated progenitor cell populations that give rise to urothelium during development and in response to injury. Understanding the cellular and molecular basis for urothelial patterning and repair will inform tissue regeneration therapies designed to ameliorate a number of structural and functional defects of the urinary tract. Here, we review the current understanding of urothelial progenitors and the signaling pathways that govern urothelial development and repair. While most published studies have focused on bladder urothelium, we also discuss literature on upper tract urothelial progenitors. Furthermore, we discuss evidence supporting existence of context-specific progenitors. This knowledge is fundamental to the development of strategies to regenerate or engineer damaged or diseased urothelium.
Collapse
Affiliation(s)
- Ashley R Jackson
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute At Nationwide Children's Hospital, 700 Children's Drive, W308, Columbus, 43205, OH, USA
- Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH, USA
| | - Sridhar T Narla
- Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, Rangos Research Building, 4401 Penn Avenue, Pittsburgh, 15224, PA, USA
| | - Carlton M Bates
- Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, Rangos Research Building, 4401 Penn Avenue, Pittsburgh, 15224, PA, USA.
- Division of Nephrology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| | - Brian Becknell
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute At Nationwide Children's Hospital, 700 Children's Drive, W308, Columbus, 43205, OH, USA.
- Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA.
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
12
|
Reiswich V, Könemann S, Lennartz M, Höflmayer D, Menz A, Chirico V, Hube-Magg C, Fraune C, Bernreuther C, Simon R, Clauditz TS, Sauter G, Hinsch A, Kind S, Jacobsen F, Steurer S, Minner S, Büscheck F, Burandt E, Marx AH, Lebok P, Krech T. Large-scale human tissue analysis identifies Uroplakin 1a as a putative diagnostic marker for urothelial cancer. Pathol Res Pract 2022; 237:154028. [PMID: 35872365 DOI: 10.1016/j.prp.2022.154028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 11/27/2022]
Abstract
Uroplakin 1A (Upk1a) protein is relevant for stabilizing and strengthening urothelial cells and helps to prevent them from rupturing during bladder distension. Based on RNA expression data Upk1a is expressed in a limited number of normal tissues and tumors. To comprehensively evaluate the potential diagnostic and prognostic utility of Upk1a immunohistochemistry, a tissue microarray containing 6929 samples from 115 different tumor types and subtypes and 608 samples of 76 different normal tissue types was analyzed. Upk1a positivity was found in 34 (29.6 %) different tumor types including 9 (7.8 %) tumor types with at least one strongly positive case. The highest rates of Upk1a positivity were seen in various subtypes of urothelial neoplasms (42.6-98 %) including Brenner tumors of the ovary (64.9 %) followed by neoplasms of the thyroid (10.4-33.3 %). In urothelial tumors, Upk1a staining predominated at the cell membranes and staining intensity was often moderate to strong. In thyroidal neoplasms the staining was mostly purely cytoplasmic and of low to moderate intensity. Upk1a positivity was also seen in up to 15 % of cases in 25 additional tumor categories but the staining intensity was often cytoplasmic and the intensity was usually judged as weak and only rarely as moderate. Within non-invasive (pTa) tumors, the Upk1a positivity rate decreased from 94 % in pTa G2 (low grade) to 90.1 % in pTa G3 (p = 0.012) and was even lower in muscle-invasive carcinomas (41.5 %; p < 0.0001 vs pTaG3). Within muscle invasive carcinomas, Upk1a expression was unrelated to nodal metastasis (p > 0.05) and patient outcome (p > 0.05). In conclusion, Upk1a immunohistochemistry is a potentially useful and specific diagnostic marker for the distinction of urothelial carcinomas from other neoplasms. However, its sensitivity is less than 50 % in muscle-invasive cancers because Upk1a expression decreases during grade and stage progression.
Collapse
Affiliation(s)
- Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Steffi Könemann
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Doris Höflmayer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Viktoria Chirico
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon Kind
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Marx
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Pathology, Academic Hospital Fuerth, Fuerth, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| |
Collapse
|
13
|
Barrier-Forming Potential of Epithelial Cells from the Exstrophic Bladder. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:943-955. [PMID: 35358476 PMCID: PMC9194657 DOI: 10.1016/j.ajpath.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/04/2022] [Accepted: 03/03/2022] [Indexed: 11/20/2022]
Abstract
Bladder exstrophy (BEX) is a rare developmental abnormality resulting in an open, exposed bladder plate. Although normal bladder urothelium is a mitotically quiescent barrier epithelium, histologic studies of BEX epithelia report squamous and proliferative changes that can persist beyond surgical closure. The current study examined whether patient-derived BEX epithelial cells in vitro were capable of generating a barrier-forming epithelium under permissive conditions. Epithelial cells isolated from 11 BEX samples, classified histologically as transitional (n = 6) or squamous (n = 5), were propagated in vitro. In conditions conducive to differentiated tight barrier formation by normal human urothelial cell cultures, 8 of 11 BEX lines developed transepithelial electrical resistances of more than 1000 Ω.cm2, with 3 squamous lines failing to generate tight barriers. An inverse relationship was found between expression of squamous KRT14 transcript and barrier development. Transcriptional drivers of urothelial differentiation PPARG, GATA3, and FOXA1 showed reduced expression in squamous BEX cultures. These findings implicate developmental interruption of urothelial transcriptional programming in the spectrum of transitional to squamous epithelial phenotypes found in BEX. Assessment of BEX epithelial phenotype may inform management and treatment strategies, for which distinction between reversible versus intractably squamous epithelium could identify patients at risk of medical complications or those who are most appropriate for reconstructive tissue engineering strategies.
Collapse
|
14
|
Reiswich V, Akdeniz G, Lennartz M, Menz A, Chirico V, Hube-Magg C, Fraune C, Bernreuther C, Simon R, Clauditz TS, Sauter G, Uhlig R, Hinsch A, Kind S, Jacobsen F, Möller K, Steurer S, Minner S, Burandt E, Marx AH, Lebok P, Krech T, Dum D. Large-scale human tissue analysis identifies Uroplakin 1b as a putative diagnostic marker in surgical pathology. Hum Pathol 2022; 126:108-120. [PMID: 35550834 DOI: 10.1016/j.humpath.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/03/2022] [Indexed: 11/28/2022]
Abstract
Uroplakin 1B (Upk1b) stabilizes epithelial cells lining the bladder lumen to prevent rupturing during bladder distension. Little is known about Upk1b expression in other normal and malignant tissues. To comprehensively evaluate the potential diagnostic and prognostic utility of Upk1b expression analysis, a tissue microarray containing 14,061 samples from 127 different tumor types and subtypes and 608 samples of 76 different normal tissue types was analyzed by immunohistochemistry. Upk1b immunostaining was found in 61 (48%) different tumor types including 50 (39%) with at least one moderately positive and 39 tumor types (31%) with at least one strongly positive tumor. Highest positivity rates were found in urothelial neoplasms (58-95%), Brenner tumors of the ovary (92%), epitheloid mesothelioma (87%), serous carcinoma of the ovary (58%) and the endometrium (53%) as well as in squamous cell carcinoma of the head and neck (18-37%), lung (39%) and esophagus (26%). In urothelial carcinoma, low Upk1b expression was linked to high grade and invasive tumor growth (p<0.0001 each) and nodal metastasis (p=0.0006). Our data suggest diagnostic applications of Upk1b immunohistochemistry in panels for the distinction of malignant mesothelioma from adenocarcinoma of the lung, urothelial carcinoma from prostatic adenocarcinoma in the bladder, or pancreatico-biliary and gastro-esophageal from colorectal adenocarcinoma.
Collapse
Affiliation(s)
- Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gonca Akdeniz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Viktoria Chirico
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ria Uhlig
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon Kind
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Möller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Marx
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Pathology, Academic Hospital Fuerth, Fuerth Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Pathology, Clinical Center Osnabrueck, Osnabrueck, Germany
| | - David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
15
|
Wiessner GB, Plumber SA, Xiang T, Mendelsohn CL. Development, regeneration and tumorigenesis of the urothelium. Development 2022; 149:dev198184. [PMID: 35521701 PMCID: PMC10656457 DOI: 10.1242/dev.198184] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The urothelium of the bladder functions as a waterproof barrier between tissue and outflowing urine. Largely quiescent during homeostasis, this unique epithelium rapidly regenerates in response to bacterial or chemical injury. The specification of the proper cell types during development and injury repair is crucial for tissue function. This Review surveys the current understanding of urothelial progenitor populations in the contexts of organogenesis, regeneration and tumorigenesis. Furthermore, we discuss pathways and signaling mechanisms involved in urothelial differentiation, and consider the relevance of this knowledge to stem cell biology and tissue regeneration.
Collapse
Affiliation(s)
- Gregory B. Wiessner
- Departments of Urology, Genetics and Development, Pathology and Cell Biology, Columbia Stem Cell Initiative and Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
- Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| | - Sakina A. Plumber
- Departments of Urology, Genetics and Development, Pathology and Cell Biology, Columbia Stem Cell Initiative and Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| | - Tina Xiang
- Departments of Urology, Genetics and Development, Pathology and Cell Biology, Columbia Stem Cell Initiative and Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| | - Cathy L. Mendelsohn
- Departments of Urology, Genetics and Development, Pathology and Cell Biology, Columbia Stem Cell Initiative and Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
- Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| |
Collapse
|
16
|
Lee J, Lee J, Sim W, Kim JH. Soluble TGFBI aggravates the malignancy of cholangiocarcinoma through activation of the ITGB1 dependent PPARγ signalling pathway. Cell Oncol (Dordr) 2022; 45:275-291. [PMID: 35357655 DOI: 10.1007/s13402-022-00668-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Cholangiocarcinoma is a devastating cancer with a poor prognosis. Previous reports have presented conflicting results on the role of transforming growth factor-β-induced protein (TGFBI) in malignant cancers. Currently, our understanding of the role of TGFBI in cholangiocarcinoma is ambiguous. The aim of the present study was to investigate the role of TGFBI in human cholangiocarcinoma. METHODS Iterative patient partitioning (IPP) scoring and consecutive elimination methods were used to select prognostic biomarkers. mRNA and protein expression levels were determined using Gene Expression Omnibus (GEO), Western blot and ELISA analyses. Biological activities of selected biomarkers were examined using both in vitro and in vivo assays. Prognostic values were assessed using Kaplan-Meier and Liptak's z score analyses. RESULTS TGFBI was selected as a candidate cholangiocarcinoma biomarker. GEO database analysis revealed significantly higher TGFBI mRNA expression levels in cholangiocarcinoma tissues compared to matched normal tissues. TGFBI protein was specifically detected in a soluble form in vitro and in vivo. TGFBI silencing evoked significant anti-cancer effects in vitro. Soluble TGFBI treatment aggravated the malignancy of cholangiocarcinoma cells both in vitro and in vivo through activation of the integrin beta-1 (ITGB1) dependent PPARγ signalling pathway. High TGFBI expression was associated with a poor prognosis in patients with cholangiocarcinoma. CONCLUSIONS Our data suggest that TGFBI may serve as a promising prognostic biomarker and therapeutic target for cholangiocarcinoma.
Collapse
Affiliation(s)
- Jungwhoi Lee
- Department of Biotechnology, College of Applied Life Science, Jeju National University, 102 Jejudaehak-ro, Jeju-si, Jeju-do, 63243, Republic of Korea.
| | - Jungsul Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Woogwang Sim
- Department of Anatomy, University of California,, San Francisco, CA, 94143, USA
| | - Jae-Hoon Kim
- Department of Biotechnology, College of Applied Life Science, Jeju National University, 102 Jejudaehak-ro, Jeju-si, Jeju-do, 63243, Republic of Korea.
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju-si, Jeju-do, 690-756, Republic of Korea.
| |
Collapse
|
17
|
Papaetis GS. Pioglitazone, Bladder Cancer and the Presumption of Innocence. Curr Drug Saf 2022; 17:294-318. [PMID: 35249505 DOI: 10.2174/1574886317666220304124756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/01/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Thiazolidinediones are potent exogenous agonists of PPAR-γ, which augment the effects of insulin to its cellular targets and mainly at the level of adipose tissue. Pioglitazone, the main thiazolidinedione in clinical practice, has shown cardiovascular and renal benefits in patients with type 2 diabetes, durable reduction of glycated hemoglobulin levels, important improvements of several components of the metabolic syndrome and beneficial effects of non-alcoholic fatty liver disease. OBJECTIVE Despite all of its established advantages, the controversy for an increased risk of developing bladder cancer, combined with the advent of newer drug classes that achieved major cardiorenal effects have significantly limited its use spreading a persistent shadow of doubt for its future role. METHODS Pubmed, Google and Scope databases have been thoroughly searched and relevant studies were selected. RESULTS This paper explores thoroughly both in vitro and in vivo (animal models and humans) studies that investigated the possible association of pioglitazone with bladder cancer. CONCLUSION Currently the association of pioglitazone with bladder cancer cannot be based on solid evidence. This evidence cannot justify its low clinical administration, especially in the present era of individualised treatment strategies. Definite clarification of this issue is imperative and urgently anticipated from future high quality and rigorous pharmacoepidemiologic research, keeping in mind its unique mechanism of action and its significant pleiotropic effects.
Collapse
Affiliation(s)
- Georgios S Papaetis
- Internal Medicine and Diabetes Clinic, Eleftherios Venizelos Avenue 62, Paphos, Cyprus.
- CDA College, 73 Democratias Avenue, Paphos, Cyprus
| |
Collapse
|
18
|
Understanding Molecular Mechanisms and Identifying Key Processes in Chronic Radiation Cystitis. Int J Mol Sci 2022; 23:ijms23031836. [PMID: 35163758 PMCID: PMC8836784 DOI: 10.3390/ijms23031836] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic radiation cystitis (CRC) is a consequence of pelvic radiotherapy and affects 5–10% of patients. The pathology of CRC is without curative treatment and is characterized by incontinence, pelvic pain and hematuria, which severely degrades patients’ quality of life. Current management strategies rely primarily on symptomatic measures and have certain limitations. Thanks to a better understanding of the pathophysiology of radiation cystitis, studies targeting key manifestations such as inflammation, neovascularization and cell atrophy have emerged and are promising avenues for future treatment. However, the mechanisms of CRC are still better described in animal models than in human models. Preclinical studies conducted to elucidate the pathophysiology of CRC use distinct models and are most often limited to specific processes, such as fibrosis, vascular damage and inflammation. This review presents a synthesis of experimental studies aimed at improving our understanding of the molecular mechanisms at play and identifying key processes in CRC.
Collapse
|
19
|
Molecular pathology of the non-luminal Ba/Sq-like and Sc/NE-like classes of urothelial tumours: an integrated immunohistochemical analysis. Hum Pathol 2022; 122:11-24. [DOI: 10.1016/j.humpath.2022.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/12/2022]
|
20
|
Hurst CD, Cheng G, Platt FM, Castro MAA, Marzouka NADS, Eriksson P, Black EVI, Alder O, Lawson ARJ, Lindskrog SV, Burns JE, Jain S, Roulson JA, Brown JC, Koster J, Robertson AG, Martincorena I, Dyrskjøt L, Höglund M, Knowles MA. Stage-stratified molecular profiling of non-muscle-invasive bladder cancer enhances biological, clinical, and therapeutic insight. Cell Rep Med 2021; 2:100472. [PMID: 35028613 PMCID: PMC8714941 DOI: 10.1016/j.xcrm.2021.100472] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 08/09/2021] [Accepted: 11/18/2021] [Indexed: 12/26/2022]
Abstract
Understanding the molecular determinants that underpin the clinical heterogeneity of non-muscle-invasive bladder cancer (NMIBC) is essential for prognostication and therapy development. Stage T1 disease in particular presents a high risk of progression and requires improved understanding. We present a detailed multi-omics study containing gene expression, copy number, and mutational profiles that show relationships to immune infiltration, disease recurrence, and progression to muscle invasion. We compare expression and genomic subtypes derived from all NMIBCs with those derived from the individual disease stages Ta and T1. We show that sufficient molecular heterogeneity exists within the separate stages to allow subclassification and that this is more clinically meaningful for stage T1 disease than that derived from all NMIBCs. This provides improved biological understanding and identifies subtypes of T1 tumors that may benefit from chemo- or immunotherapy.
Collapse
Affiliation(s)
- Carolyn D Hurst
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Guo Cheng
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Fiona M Platt
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Mauro A A Castro
- Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba, Brazil
| | | | - Pontus Eriksson
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Emma V I Black
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Olivia Alder
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Andrew R J Lawson
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton CB10 1SA, UK
| | - Sia V Lindskrog
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Julie E Burns
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Sunjay Jain
- Pyrah Department of Urology, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Jo-An Roulson
- Department of Histopathology, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Joanne C Brown
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Jan Koster
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Center, BC Cancer, Vancouver, BC V5Z 4S6, Canada
| | - Inigo Martincorena
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton CB10 1SA, UK
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mattias Höglund
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Margaret A Knowles
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James's, St James's University Hospital, Beckett Street, Leeds LS9 7TF, UK
| |
Collapse
|
21
|
Evaluating the effect of Luffa cylindrica stem sap on dermal fibroblasts; An invitro study. Biochem Biophys Res Commun 2021; 580:41-47. [PMID: 34619551 DOI: 10.1016/j.bbrc.2021.09.079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 01/02/2023]
Abstract
Luffa cylindrica stem sap (LuCS) has been traditionally used as a facial cosmetic supplement to enhance the skin condition of Asians. However, LuCS has yet to be described and there is no solid scientific evidence regarding the use of LuCS as an anti-wrinkle agent. In the present study, we have evaluated the functional effect of LuCS and its underlying mechanisms based on scientific evidence. Treatment with LuCS stimulated the growth and migration of human skin fibroblasts. LuCS treatment activated EGFR signaling via the enhanced expression of EGFR and down-regulation of PPARγ in human skin fibroblasts. Exposure to LuCS induced the synthesis of cellular type I procollagen and elastin in consort with the down-regulation of various proteinases including MMP-1, -2 and -9 in human skin fibroblasts. LuCS treatment also reversed the skin damage induced by UV-A irradiation in human skin fibroblasts. 3-bromo-3-methylisoxazol-5-amine was identified as the functional component using UPLC-MS-MS analysis and increased production of cellular type I procollagen. Collectively, these results suggest the efficacy of LuCS supplementation in improving the skin condition via anti-wrinkle effect.
Collapse
|
22
|
Bekele RT, Samant AS, Nassar AH, So J, Garcia EP, Curran CR, Hwang JH, Mayhew DL, Nag A, Thorner AR, Börcsök J, Sztupinszki Z, Pan CX, Bellmunt J, Kwiatkowski DJ, Sonpavde GP, Van Allen EM, Mouw KW. RAF1 amplification drives a subset of bladder tumors and confers sensitivity to MAPK-directed therapeutics. J Clin Invest 2021; 131:147849. [PMID: 34554931 DOI: 10.1172/jci147849] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 09/16/2021] [Indexed: 11/17/2022] Open
Abstract
Bladder cancer is a genetically heterogeneous disease, and novel therapeutic strategies are needed to expand treatment options and improve clinical outcomes. Here, we identified a unique subset of urothelial tumors with focal amplification of the RAF1 (CRAF) kinase gene. RAF1-amplified tumors had activation of the RAF/MEK/ERK signaling pathway and exhibited a luminal gene expression pattern. Genetic studies demonstrated that RAF1-amplified tumors were dependent upon RAF1 activity for survival, and RAF1-activated cell lines and patient-derived models were sensitive to available and emerging RAF inhibitors as well as combined RAF plus MEK inhibition. Furthermore, we found that bladder tumors with HRAS- or NRAS-activating mutations were dependent on RAF1-mediated signaling and were sensitive to RAF1-targeted therapy. Together, these data identified RAF1 activation as a dependency in a subset making up nearly 20% of urothelial tumors and suggested that targeting RAF1-mediated signaling represents a rational therapeutic strategy.
Collapse
Affiliation(s)
- Raie T Bekele
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Amruta S Samant
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Amin H Nassar
- Brigham and Women's Hospital, Boston, Massachusetts, USA.,Department of Medical Oncology and
| | | | | | | | - Justin H Hwang
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medical Oncology and
| | - David L Mayhew
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medical Oncology and
| | - Anwesha Nag
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Aaron R Thorner
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Judit Börcsök
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Chong-Xian Pan
- VA Boston Healthcare System, Harvard Medical School, West Roxbury, Massachusetts, USA
| | - Joaquim Bellmunt
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - David J Kwiatkowski
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Eliezer M Van Allen
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Department of Medical Oncology and
| | - Kent W Mouw
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Ho SR, Lee YC, Ittmann MM, Lin FT, Chan KS, Lin WC. RNF144A deficiency promotes PD-L1 protein stabilization and carcinogen-induced bladder tumorigenesis. Cancer Lett 2021; 520:344-360. [PMID: 34400221 PMCID: PMC9420248 DOI: 10.1016/j.canlet.2021.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/21/2021] [Accepted: 08/10/2021] [Indexed: 12/14/2022]
Abstract
RNF144A is a DNA damage-induced E3 ubiquitin ligase that targets proteins involved in genome instability for degradation, e.g., DNA-PKcs and BMI1. RNF144A is frequently mutated or epigenetically silenced in cancer, providing the rationale to evaluate RNF144A loss of function in tumorigenesis. Here we report that RNF144A-deficient mice are more prone to the development of bladder tumors upon carcinogen exposure. In addition to DNA-PKcs and BMI1, we identify the immune checkpoint protein PD-L1 as a novel degradation target of RNF144A, since these proteins are expressed at higher levels in Rnf144a KO tumors. RNF144A interacts with PD-L1 in the plasma membrane and intracellular vesicles and promotes poly-ubiquitination and degradation of PD-L1. Therefore, Rnf144a KO stabilizes PD-L1 and leads to a reduction of tumor-infiltrating CD8+ T cell populations in the BBN-induced bladder tumors. The bladder tumors developed in WT and Rnf144a KO mice primarily express CK5 and CK14, markers of basal cancer subtype, as expected in BBN-induced bladder tumors. Intriguingly, the Rnf144a KO tumors also express GATA3, a marker for the luminal subtype, suggesting that RNF144A loss of function promotes features of cellular differentiation. Such differentiation features in Rnf144a KO tumors likely result from a decrease of EGFR expression, consistent with the reported role of RNF144A in maintaining EGFR expression. In summary, for the first time our study demonstrates the in vivo tumor suppressor activity of RNF144A upon carcinogenic insult. Loss of RNF144A promotes the expression of DNA-PKcs, BMI1 and PD-L1, likely contributing to the carcinogen-induced bladder tumorigenesis.
Collapse
Affiliation(s)
- Shiuh-Rong Ho
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yu-Cheng Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Michael M Ittmann
- Department of Pathology and Immunology, Baylor College of Medicine, Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Fang-Tsyr Lin
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Keith Syson Chan
- Department of Pathology and Laboratory Medicine, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Weei-Chin Lin
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
24
|
Efremenko A, Balbuena P, Clewell RA, Black M, Pluta L, Andersen ME, Gentry PR, Yager JW, Clewell HJ. Time-dependent genomic response in primary human uroepithelial cells exposed to arsenite for up to 60 days. Toxicology 2021; 461:152893. [PMID: 34425169 DOI: 10.1016/j.tox.2021.152893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/02/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
Evidence from both in vivo and in vitro studies suggests that gene expression changes from long-term exposure to arsenite evolve markedly over time, including reversals in the direction of expression change in key regulatory genes. In this study, human uroepithelial cells from the ureter segments of 4 kidney-donors were continuously treated in culture with arsenite at concentrations of 0.1 or 1 μM for 60 days. Gene expression at 10, 20, 30, 40, and 60 days was determined using Affymetrix human genome microarrays and signal pathway analysis was performed using GeneGo Metacore. Arsenic treated cells continued to proliferate for the full 60-day period, whereas untreated cells ceased proliferating after approximately 30 days. A peak in the number of gene changes in the treated cells compared to untreated controls was observed between 30 and 40 days of exposure, with substantially fewer changes at 10 and 60 days, suggesting remodeling of the cells over time. Consistent with this possibility, the direction of expression change for a number of key genes was reversed between 20 and 30 days, including CFOS and MDM2. While the progression of gene changes was different for each subject, a common pattern was observed in arsenic treated cells over time, with early upregulation of oxidative stress responses (HMOX1, NQ01, TXN, TXNRD1) and down-regulation of immune/inflammatory responses (IKKα). At around 30 days, there was a transition to increased inflammatory and proliferative signaling (AKT, CFOS), evidence of epithelial-to-mesenchymal transition (EMT), and alterations in DNA damage responses (MDM2, ATM). A common element in the changing response of cells to arsenite over time appears to involve up-regulation of MDM2 by inflammatory signaling (through AP-1 and NF-κB), leading to inhibition of P53 function.
Collapse
Affiliation(s)
- Alina Efremenko
- The Hamner Institutes for Health Sciences, RTP, NC, United States
| | | | | | - Michael Black
- The Hamner Institutes for Health Sciences, RTP, NC, United States
| | - Linda Pluta
- The Hamner Institutes for Health Sciences, RTP, NC, United States
| | | | | | - Janice W Yager
- Ramboll US Corporation, Emeryville, CA, United States(1)
| | - Harvey J Clewell
- The Hamner Institutes for Health Sciences, RTP, NC, United States.
| |
Collapse
|
25
|
Mukha A, Kalkhoven E, van Mil SWC. Splice variants of metabolic nuclear receptors: Relevance for metabolic disease and therapeutic targeting. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166183. [PMID: 34058349 DOI: 10.1016/j.bbadis.2021.166183] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022]
Abstract
Metabolic nuclear receptors are ligand-activated transcription factors which control a wide range of metabolic processes and signaling pathways in response to nutrients and xenobiotics. Targeting these NRs is at the forefront of our endeavours to generate novel treatment options for diabetes, metabolic syndrome and fatty liver disease. Numerous splice variants have been described for these metabolic receptors. Structural changes, as a result of alternative splicing, lead to functional differences among NR isoforms, resulting in the regulation of different metabolic pathways by these NR splice variants. In this review, we describe known splice variants of FXR, LXRs, PXR, RXR, LRH-1, CAR and PPARs. We discuss their structure and functions, and elaborate on the regulation of splice variant abundance by nutritional signals. We conclude that NR splice variants pose an intriguing new layer of complexity in metabolic signaling, which needs to be taken into account in the development of treatment strategies for metabolic diseases.
Collapse
Affiliation(s)
- Anna Mukha
- Center for Molecular Medicine, UMC Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, UMC Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Saskia W C van Mil
- Center for Molecular Medicine, UMC Utrecht and Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
26
|
Ozgun G, Senturk S, Erkek-Ozhan S. Retinoic acid signaling and bladder cancer: Epigenetic deregulation, therapy and beyond. Int J Cancer 2021; 148:2364-2374. [PMID: 33128775 DOI: 10.1002/ijc.33374] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/10/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022]
Abstract
Retinoic acid (RA) signaling is a crucial developmental pathway involved in urothelium development, differentiation and regeneration. Deregulation of the RA signaling is highly implicated in several cancers, including bladder cancer, underlying the need to unravel the complete regulatory aspects of the retinoids in bladder tumorigenesis. Given the fact that RA receptors are transcription factors functioning at the chromatin level and act in close cooperation with chromatin modifiers, it is known that retinoids show their efficacy by changing the epigenome. Bladder cancer can be defined as a "disease of chromatin" with mutations identified in the genes involved in chromatin regulation in 80% of the patients. Therefore, a careful examination of the epigenetic backgrounds and the breakdown of the emerging and highly underexplored field of RA dependent regulation of the epigenome is essential to fully understand the retinoid-dependent effects on bladder cancer. With this motivation, in this review, we evaluate the role of RA signaling in bladder cancer with a focus on the regulatory and mutational aspects, emphasizing the deregulatory characteristics in bladder cancer and highlighting the potential treatment opportunities with the RA and derivatives alone or in combination with epigenetic drugs.
Collapse
Affiliation(s)
- Gizem Ozgun
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Dokuz Eylül University Izmir International Biomedicine and Genome Institute, Izmir, Turkey
| | - Serif Senturk
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Dokuz Eylül University Izmir International Biomedicine and Genome Institute, Izmir, Turkey
| | | |
Collapse
|
27
|
Kurdi BA, Ababneh NA, Abuharfeil N, Al Demour S, Awidi AS. Use of conditioned media (CM) and xeno-free serum substitute on human adipose-derived stem cells (ADSCs) differentiation into urothelial-like cells. PeerJ 2021; 9:e10890. [PMID: 33850639 PMCID: PMC8019311 DOI: 10.7717/peerj.10890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/12/2021] [Indexed: 01/01/2023] Open
Abstract
Background Congenital abnormalities, cancers as well as injuries can cause irreversible damage to the urinary tract, which eventually requires tissue reconstruction. Smooth muscle cells, endothelial cells, and urothelial cells are the major cell types required for the reconstruction of lower urinary tract. Adult stem cells represent an accessible source of unlimited repertoire of untransformed cells. Aim Fetal bovine serum (FBS) is the most vital supplement in the culture media used for cellular proliferation and differentiation. However, due to the increasing interest in manufacturing xeno-free stem cell-based cellular products, optimizing the composition of the culture media and the serum-type used is of paramount importance. In this study, the effects of FBS and pooled human platelet (pHPL) lysate were assessed on the capacity of human adipose-derived stem cells (ADSCs) to differentiate into urothelial-like cells. Also, we aimed to compare the ability of both conditioned media (CM) and unconditioned urothelial cell media (UCM) to induce urothelial differentiation of ADCS in vitro. Methods ADSCs were isolated from human lipoaspirates and characterized by flow cytometry for their ability to express the most common mesenchymal stem cell (MSCs) markers. The differentiation potential was also assessed by differentiating them into osteogenic and adipogenic cell lineages. To evaluate the capacity of ADSCs to differentiate towards the urothelial-like lineage, cells were cultured with either CM or UCM, supplemented with either 5% pHPL, 2.5% pHPL or 10% FBS. After 14 days of induction, cells were utilized for gene expression and immunofluorescence analysis. Results ADSCs cultured in CM and supplemented with FBS exhibited the highest upregulation levels of the urothelial cell markers; cytokeratin-18 (CK-18), cytokeratin-19 (CK-19), and Uroplakin-2 (UPK-2), with a 6.7, 4.2- and a 2-folds increase in gene expression, respectively. Meanwhile, the use of CM supplemented with either 5% pHPL or 2.5% pHPL, and UCM supplemented with either 5% pHPL or 2.5% pHPL showed low expression levels of CK-18 and CK-19 and no upregulation of UPK-2 level was observed. In contrast, the use of UCM with FBS has increased the levels of CK-18 and CK-19, however to a lesser extent compared to CM. At the cellular level, CK-18 and UPK-2 were only detected in CM/FBS supplemented group. Growth factor analysis revealed an increase in the expression levels of EGF, VEGF and PDGF in all of the differentiated groups. Conclusion Efficient ADSCs urothelial differentiation is dependent on the use of conditioned media. The presence of high concentrations of proliferation-inducing growth factors present in the pHPL reduces the efficiency of ADSCs differentiation towards the urothelial lineage. Additionally, the increase in EGF, VEGF and PDGF during the differentiation implicates them in the mechanism of urothelial cell differentiation.
Collapse
Affiliation(s)
- Ban Al- Kurdi
- Cell Therapy Center, University of Jordan, Amman, Jordan.,Department of Hematology and Oncology, Jordan University Hospital, Amman, Jordan
| | | | - Nizar Abuharfeil
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Saddam Al Demour
- Department of Urology, School of medicine, University of Jordan, Amman, Jordan, University of Jordan, Amman, Jordan
| | - Abdalla S Awidi
- Cell Therapy Center, University of Jordan, Amman, Jordan.,Department of Hematology and Oncology, Jordan University Hospital, Amman, Jordan
| |
Collapse
|
28
|
Hernandez-Quiles M, Broekema MF, Kalkhoven E. PPARgamma in Metabolism, Immunity, and Cancer: Unified and Diverse Mechanisms of Action. Front Endocrinol (Lausanne) 2021; 12:624112. [PMID: 33716977 PMCID: PMC7953066 DOI: 10.3389/fendo.2021.624112] [Citation(s) in RCA: 247] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022] Open
Abstract
The proliferator-activated receptor γ (PPARγ), a member of the nuclear receptor superfamily, is one of the most extensively studied ligand-inducible transcription factors. Since its identification in the early 1990s, PPARγ is best known for its critical role in adipocyte differentiation, maintenance, and function. Emerging evidence indicates that PPARγ is also important for the maturation and function of various immune system-related cell types, such as monocytes/macrophages, dendritic cells, and lymphocytes. Furthermore, PPARγ controls cell proliferation in various other tissues and organs, including colon, breast, prostate, and bladder, and dysregulation of PPARγ signaling is linked to tumor development in these organs. Recent studies have shed new light on PPARγ (dys)function in these three biological settings, showing unified and diverse mechanisms of action. Classical transactivation-where PPARγ activates genes upon binding to PPAR response elements as a heterodimer with RXRα-is important in all three settings, as underscored by natural loss-of-function mutations in FPLD3 and loss- and gain-of-function mutations in tumors. Transrepression-where PPARγ alters gene expression independent of DNA binding-is particularly relevant in immune cells. Interestingly, gene translocations resulting in fusion of PPARγ with other gene products, which are unique to specific carcinomas, present a third mode of action, as they potentially alter PPARγ's target gene profile. Improved understanding of the molecular mechanism underlying PPARγ activity in the complex regulatory networks in metabolism, cancer, and inflammation may help to define novel potential therapeutic strategies for prevention and treatment of obesity, diabetes, or cancer.
Collapse
Affiliation(s)
- Miguel Hernandez-Quiles
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marjoleine F. Broekema
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- *Correspondence: Eric Kalkhoven,
| |
Collapse
|
29
|
Silina L, Maksut F, Bernard-Pierrot I, Radvanyi F, Créhange G, Mégnin-Chanet F, Verrelle P. Review of Experimental Studies to Improve Radiotherapy Response in Bladder Cancer: Comments and Perspectives. Cancers (Basel) 2020; 13:E87. [PMID: 33396795 PMCID: PMC7795454 DOI: 10.3390/cancers13010087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 12/22/2020] [Indexed: 01/04/2023] Open
Abstract
Bladder cancer is among the top ten most common cancer types in the world. Around 25% of all cases are muscle-invasive bladder cancer, for which the gold standard treatment in the absence of metastasis is the cystectomy. In recent years, trimodality treatment associating maximal transurethral resection and radiotherapy combined with concurrent chemotherapy is increasingly used as an organ-preserving alternative. However, the use of this treatment is still limited by the lack of biomarkers predicting tumour response and by a lack of targeted radiosensitising drugs that can improve the therapeutic index, especially by limiting side effects such as bladder fibrosis. In order to improve the bladder-preserving treatment, experimental studies addressing these main issues ought to be considered (both in vitro and in vivo studies). Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for systematic reviews, we conducted a literature search in PubMed on experimental studies investigating how to improve bladder cancer radiotherapy with different radiosensitising agents using a comprehensive search string. We made comments on experimental model selection, experimental design and results, formulating the gaps of knowledge still existing: such as the lack of reliable predictive biomarkers of tumour response to chemoradiation according to the molecular tumour subtype and lack of efficient radiosensitising agents specifically targeting bladder tumour cells. We provided guidance to improve forthcoming studies, such as taking into account molecular characteristics of the preclinical models and highlighted the value of using patient-derived xenografts as well as syngeneic models. Finally, this review could be a useful tool to set up new radiation-based combined treatments with an improved therapeutic index that is needed for bladder preservation.
Collapse
Affiliation(s)
- Linda Silina
- French League Against Cancer Team, CNRS UMR144, Curie Institute and PSL Research University, 75005 Paris, France; (I.B.-P.); (F.R.)
- CNRS UMR 9187, INSERM U1196, Curie Institute, PSL Research University and Paris-Saclay University, Rue H. Becquerel, 91405 Orsay, France; (F.M.); (F.M.-C.)
| | - Fatlinda Maksut
- CNRS UMR 9187, INSERM U1196, Curie Institute, PSL Research University and Paris-Saclay University, Rue H. Becquerel, 91405 Orsay, France; (F.M.); (F.M.-C.)
| | - Isabelle Bernard-Pierrot
- French League Against Cancer Team, CNRS UMR144, Curie Institute and PSL Research University, 75005 Paris, France; (I.B.-P.); (F.R.)
| | - François Radvanyi
- French League Against Cancer Team, CNRS UMR144, Curie Institute and PSL Research University, 75005 Paris, France; (I.B.-P.); (F.R.)
| | - Gilles Créhange
- Radiation Oncology Department, Curie Institute, 75005 Paris, France;
| | - Frédérique Mégnin-Chanet
- CNRS UMR 9187, INSERM U1196, Curie Institute, PSL Research University and Paris-Saclay University, Rue H. Becquerel, 91405 Orsay, France; (F.M.); (F.M.-C.)
| | - Pierre Verrelle
- CNRS UMR 9187, INSERM U1196, Curie Institute, PSL Research University and Paris-Saclay University, Rue H. Becquerel, 91405 Orsay, France; (F.M.); (F.M.-C.)
- Radiation Oncology Department, Curie Institute, 75005 Paris, France;
- Clermont Auvergne University, 63000 Clermont-Ferrand, France
| |
Collapse
|
30
|
Loss of UTX/KDM6A and the activation of FGFR3 converge to regulate differentiation gene-expression programs in bladder cancer. Proc Natl Acad Sci U S A 2020; 117:25732-25741. [PMID: 32989154 DOI: 10.1073/pnas.2008017117] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Bladder cancer prognosis is closely linked to the underlying differentiation state of the tumor, ranging from the less aggressive and most-differentiated luminal tumors to the more aggressive and least-differentiated basal tumors. Sequencing of bladder cancer has revealed that loss-of-function mutations in chromatin regulators and mutations that activate receptor tyrosine kinase (RTK) signaling frequently occur in bladder cancer. However, little is known as to whether and how these two types of mutations functionally interact or cooperate to regulate tumor growth and differentiation state. Here, we focus on loss of the histone demethylase UTX (also known as KDM6A) and activation of the RTK FGFR3, two events that commonly cooccur in muscle invasive bladder tumors. We show that UTX loss and FGFR3 activation cooperate to disrupt the balance of luminal and basal gene expression in bladder cells. UTX localized to enhancers surrounding many genes that are important for luminal cell fate, and supported the transcription of these genes in a catalytic-independent manner. In contrast to UTX, FGFR3 activation was associated with lower expression of luminal genes in tumors and FGFR inhibition increased transcription of these same genes in cell culture models. This suggests an antagonistic relationship between UTX and FGFR3. In support of this model, UTX loss-of-function potentiated FGFR3-dependent transcriptional effects and the presence of UTX blocked an FGFR3-mediated increase in the colony formation of bladder cells. Taken together, our study reveals how mutations in UTX and FGFR3 converge to disrupt bladder differentiation programs that could serve as a therapeutic target.
Collapse
|
31
|
Peng T, Wang G, Cheng S, Xiong Y, Cao R, Qian K, Ju L, Wang X, Xiao Y. The role and function of PPARγ in bladder cancer. J Cancer 2020; 11:3965-3975. [PMID: 32328200 PMCID: PMC7171493 DOI: 10.7150/jca.42663] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/08/2020] [Indexed: 12/15/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ), a member of the nuclear receptor superfamily, participates in multiple physiological and pathological processes. Extensive studies have revealed the relationship between PPARγ and various tumors. However, the expression and function of PPARγ in bladder cancer seem to be controversial. It has been demonstrated that PPARγ affects the occurrence and progression of bladder cancer by regulating proliferation, apoptosis, metastasis, and reactive oxygen species (ROS) and lipid metabolism, probably through PPARγ-SIRT1 feedback loops, the PI3K-Akt signaling pathway, and the WNT/β-catenin signaling pathway. Considering the frequent relapses after chemotherapy, some researchers have focused on the relationship between PPARγ and chemotherapy sensitivity in bladder cancer. Moreover, the feasibility of PPARγ ligands as potential therapeutic targets for bladder cancer has been uncovered. Taken together, this review summarizes the relevant literature and our findings to explore the complicated role and function of PPARγ in bladder cancer.
Collapse
Affiliation(s)
- Tianchen Peng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, China
| | - Gang Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Songtao Cheng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, China
| | - Yaoyi Xiong
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, China
| | - Rui Cao
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Kaiyu Qian
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China.,Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Wuhan, China
| |
Collapse
|
32
|
Knockdown of UTX/KDM6A Enriches Precursor Cell Populations in Urothelial Cell Cultures and Cell Lines. Cancers (Basel) 2020; 12:cancers12041023. [PMID: 32326336 PMCID: PMC7226239 DOI: 10.3390/cancers12041023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 11/29/2022] Open
Abstract
The histone demethylase UTX (gene: KDM6A) directs cell and tissue differentiation during development. Deleterious mutations in KDM6A occur in many human cancers, most frequently in urothelial carcinoma. The consequences of these mutations are poorly understood; plausibly, they may disturb urothelial differentiation. We therefore investigated the effects of UTX siRNA-mediated knockdown in two in vitro models of urothelial differentiation; namely, primary cultures of urothelial epithelial cells treated with troglitazone and PD153035 and the immortalized urothelial cell line HBLAK treated with high calcium and serum. In both models, efficient UTX knockdown did not block morphological and biochemical differentiation. An apparent delay was due to a cytotoxic effect on the cell cultures before the initiation of differentiation, which induced apoptosis partly in a p53-dependent manner. As a consequence, slowly cycling, smaller, KRT14high precursor cells in the HBLAK cell line were enriched at the expense of more differentiated, larger, proliferating KRT14low cells. UTX knockdown induced apoptosis and enriched KRT14high cells in the BFTC-905 papillary urothelial carcinoma cell line as well. Our findings suggest an explanation for the frequent occurrence of KDM6A mutations across all stages and molecular subtypes of urothelial carcinoma, whereby loss of UTX function does not primarily impede later stages of urothelial differentiation, but favors the expansion of precursor populations to provide a reservoir of potential tumor-initiating cells.
Collapse
|
33
|
Hua TNM, Oh J, Kim S, Antonio JM, Vo VTA, Om J, Choi JW, Kim JY, Jung CW, Park MJ, Jeong Y. Peroxisome proliferator-activated receptor gamma as a theragnostic target for mesenchymal-type glioblastoma patients. Exp Mol Med 2020; 52:629-642. [PMID: 32280134 PMCID: PMC7210935 DOI: 10.1038/s12276-020-0413-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 01/18/2023] Open
Abstract
Glioblastomas (GBMs) are characterized by four subtypes, proneural (PN), neural, classical, and mesenchymal (MES) GBMs, and they all have distinct activated signaling pathways. Among the subtypes, PN and MES GBMs show mutually exclusive genetic signatures, and the MES phenotype is, in general, believed to be associated with more aggressive features of GBM: tumor recurrence and drug resistance. Therefore, targeting MES GBMs would improve the overall prognosis of patients with fatal tumors. In this study, we propose peroxisome proliferator-activated receptor gamma (PPARγ) as a potential diagnostic and prognostic biomarker as well as therapeutic target for MES GBM; we used multiple approaches to assess PPARγ, including biostatistics analysis and assessment of preclinical studies. First, we found that PPARγ was exclusively expressed in MES glioblastoma stem cells (GSCs), and ligand activation of endogenous PPARγ suppressed cell growth and stemness in MES GSCs. Further in vivo studies involving orthotopic and heterotopic xenograft mouse models confirmed the therapeutic efficacy of targeting PPARγ; compared to control mice, those that received ligand treatment exhibited longer survival as well as decreased tumor burden. Mechanistically, PPARγ activation suppressed proneural-mesenchymal transition (PMT) by inhibiting the STAT3 signaling pathway. Biostatistical analysis using The Cancer Genomics Atlas (TCGA, n = 206) and REMBRANDT (n = 329) revealed that PPARγ upregulation is linked to poor overall survival and disease-free survival of GBM patients. Analysis was performed on prospective (n = 2) and retrospective (n = 6) GBM patient tissues, and we finally confirmed that PPARγ expression was distinctly upregulated in MES GBM. Collectively, this study provides insight into PPARγ as a potential therapeutic target for patients with MES GBM.
Collapse
Affiliation(s)
- Tuyen N M Hua
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
- Departments of Global Medical Science, Yonsei University, Wonju, Republic of Korea
- Departments of Mitohormesis Research Center, Yonsei University, Wonju, Republic of Korea
| | - Jiwoong Oh
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sohyun Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jayson M Antonio
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
- Departments of Global Medical Science, Yonsei University, Wonju, Republic of Korea
- Departments of Mitohormesis Research Center, Yonsei University, Wonju, Republic of Korea
| | - Vu T A Vo
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
- Departments of Global Medical Science, Yonsei University, Wonju, Republic of Korea
- Departments of Mitohormesis Research Center, Yonsei University, Wonju, Republic of Korea
| | - Jiyeon Om
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
| | - Jong-Whan Choi
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
| | - Jeong-Yub Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Chan-Woong Jung
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Myung-Jin Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea.
| | - Yangsik Jeong
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea.
- Departments of Global Medical Science, Yonsei University, Wonju, Republic of Korea.
- Departments of Mitohormesis Research Center, Yonsei University, Wonju, Republic of Korea.
- Institutes of Lifestyle Medicine, Yonsei University, Wonju, Republic of Korea.
- Departments of Mitochondrial Medicine, Yonsei University, Wonju, Republic of Korea.
- Departments of Nuclear Receptor Research Consortium, Wonju College of Medicine, Yonsei University, Wonju, Gangwon-Do, 26426, Republic of Korea.
| |
Collapse
|
34
|
Abdal Dayem A, Kim K, Lee SB, Kim A, Cho SG. Application of Adult and Pluripotent Stem Cells in Interstitial Cystitis/Bladder Pain Syndrome Therapy: Methods and Perspectives. J Clin Med 2020; 9:jcm9030766. [PMID: 32178321 PMCID: PMC7141265 DOI: 10.3390/jcm9030766] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/04/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022] Open
Abstract
Interstitial cystitis/bladder pain syndrome (IC/BPS) is a multifactorial, chronic disease without definite etiology characterized by bladder-related pelvic pain. IC/BPS is associated with pain that negatively affects the quality of life. There are various therapeutic approaches against IC/BPS. However, no efficient therapeutic agent against IC/BPS has been discovered yet. Urothelium dysfunction is one of the key factors of IC/BPS-related pathogenicity. Stem cells, including adult stem cells (ASCs) and pluripotent stem cells (PSCs), such as embryonic stem cells (ESCs) and induced PSCs (iPSCs), possess the abilities of self-renewal, proliferation, and differentiation into various cell types, including urothelial and other bladder cells. Therefore, stem cells are considered robust candidates for bladder regeneration. This review provides a brief overview of the etiology, pathophysiology, diagnosis, and treatment of IC/BPS as well as a summary of ASCs and PSCs. The potential of ASCs and PSCs in bladder regeneration via differentiation into bladder cells or direct transplantation into the bladder and the possible applications in IC/BPS therapy are described in detail. A better understanding of current studies on stem cells and bladder regeneration will allow further improvement in the approaches of stem cell applications for highly efficient IC/BPS therapy.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (A.A.D.); (K.K.); (S.B.L.)
| | - Kyeongseok Kim
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (A.A.D.); (K.K.); (S.B.L.)
| | - Soo Bin Lee
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (A.A.D.); (K.K.); (S.B.L.)
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Korea
- Correspondence: (A.K.); (S.-G.C.); Tel.: +82-2-2030-7675 (A.K.); +82-2-450-4207 (S.-G.C.); Fax: +82-2-2030-7748 (A.K.); +82-2-450-4207 (S.-G.C.)
| | - Ssang-Goo Cho
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (A.A.D.); (K.K.); (S.B.L.)
- Correspondence: (A.K.); (S.-G.C.); Tel.: +82-2-2030-7675 (A.K.); +82-2-450-4207 (S.-G.C.); Fax: +82-2-2030-7748 (A.K.); +82-2-450-4207 (S.-G.C.)
| |
Collapse
|
35
|
Majka J, Wierdak M, Szlachcic A, Magierowski M, Targosz A, Urbanczyk K, Krzysiek-Maczka G, Ptak-Belowska A, Bakalarz D, Magierowska K, Chmura A, Brzozowski T. Interaction of epidermal growth factor with COX-2 products and peroxisome proliferator-activated receptor-γ system in experimental rat Barrett's esophagus. Am J Physiol Gastrointest Liver Physiol 2020; 318:G375-G389. [PMID: 31928220 DOI: 10.1152/ajpgi.00410.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mixed acidic-alkaline refluxate is a major pathogenic factor in chronic esophagitis progressing to Barrett's esophagus (BE). We hypothesized that epidermal growth factor (EGF) can interact with COX-2 and peroxisome proliferator-activated receptor-γ (PPARγ) in rats surgically prepared with esophagogastroduodenal anastomosis (EGDA) with healthy or removed salivary glands to deplete salivary EGF. EGDA rats were treated with 1) vehicle, 2) EGF or PPARγ agonist pioglitazone with or without EGFR kinase inhibitor tyrphostin A46, EGF or PPARγ antagonist GW9662 respectively, 3) ranitidine or pantoprazole, and 4) the selective COX-2 inhibitor celecoxib combined with pioglitazone. At 3 mo, the esophageal damage and the esophageal blood flow (EBF) were determined, the mucosal expression of EGF, EGFR, COX-2, TNFα, and PPARγ mRNA and phospho-EGFR/EGFR protein was analyzed. All EGDA rats developed chronic esophagitis, esophageal ulcerations, and intestinal metaplasia followed by a fall in the EBF, an increase in the plasma of IL-1β, TNFα, and mucosal PGE2 content, the overexpression of COX-2-, and EGF-EGFR mRNAs, and proteins, and these effects were aggravated by EGF and attenuated by pioglitazone. The rise in EGF and COX-2 mRNA was inhibited by pioglitazone but reversed by pioglitazone cotreated with GW9662. We conclude that 1) EGF can interact with PG/COX-2 and the PPARγ system in the mechanism of chronic esophagitis; 2) the deleterious effect of EGF involves an impairment of EBF and the overexpression of COX-2 and EGFR, and 3) agonists of PPARγ and inhibitors of EGFR may be useful in the treatment of chronic esophagitis progressing to BE.NEW & NOTEWORTHY Rats with EGDA exhibited chronic esophagitis accompanied by a fall in EBF and an increase in mucosal expression of mRNAs for EGF, COX-2, and TNFα, and these effects were exacerbated by exogenous EGF and reduced by removal of a major source of endogenous EGF with salivectomy or concurrent treatment with tyrphostin A46 or pioglitazone combined with EGF. Beneficial effects of salivectomy in an experimental model of BE were counteracted by PPARγ antagonist, whereas selective COX-2 inhibitor celecoxib synergistically with pioglitazone reduced severity of esophageal damage and protected esophageal mucosa from reflux. We propose the cross talk among EGF/EGFR, PG/COX-2, and proinflammatory cytokines with PPARγ pathway in the mechanism of pathogenesis of chronic esophagitis progressing to BE and EAC.
Collapse
Affiliation(s)
- Jolanta Majka
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Mateusz Wierdak
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Aleksandra Szlachcic
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Marcin Magierowski
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Aneta Targosz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Katarzyna Urbanczyk
- Department of Pathomorphology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Gracjana Krzysiek-Maczka
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Agata Ptak-Belowska
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Dominik Bakalarz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Katarzyna Magierowska
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Anna Chmura
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Tomasz Brzozowski
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
36
|
Bernardo C, Eriksson P, Marzouka N, Liedberg F, Sjödahl G, Höglund M. Molecular pathology of the luminal class of urothelial tumors. J Pathol 2019; 249:308-318. [PMID: 31232464 PMCID: PMC6851980 DOI: 10.1002/path.5318] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/16/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022]
Abstract
Molecular subtypes of urothelial carcinoma may be divided into luminal and nonluminal tumors. Nonluminal tumors are composed of cases with basal/squamous-like or small cell/neuroendocrine features, with a consensus on the molecular characteristics of the respective subtype. In contrast, luminal tumors are more disparate with three to five suggested subtypes and with definitions that do not always cohere. To resolve some of these disparities we assembled a cohort of 344 luminal tumors classified as urothelial-like (Uro), with the subtypes UroA, UroAp, UroB, and UroC, or genomically unstable (GU) according to the LundTax system. Cases were systematically analyzed by immunohistochemistry using antibodies for proteins representing important biological processes or cellular states: KRT5, EGFR, and CDH3 for the integrity of a basal cell layer; CCNB1, Ki67, and FOXM1 for proliferation; FGFR3 and ERBB2 for receptor tyrosine kinase status; CCND1, CDKN2A(p16), RB1, and E2F3 for cell cycle regulation; PPARG, GATA3, and TP63 for the differentiation regulatory system; and KRT20 and UPK3 for the differentiation readout. We show that Uro tumors form one, albeit heterogenous, group characterized by FGFR3, CCND1, and RB1 expression, but low or absence of CDKN2A(p16) and ERBB2 expression. The opposite expression pattern is observed in GU cases. Furthermore, Uro tumors are distinguished from GU tumors by showing a high RB1/p16 expression ratio. Class defining characteristics were independent of pathological stage and growth pattern, and thus intrinsic. In Uro tumors, proliferation was limited to a well-defined single layer of basal-like cells in UroA tumors but occurred throughout the tumor parenchyma, independent of the basal layer, in the more progressed UroAp and UroC tumors. A similar change in proliferation topology was not observed in GU. We conclude that luminal urothelial carcinomas consist, at the molecular pathology level, of two major subtypes, the larger heterogenous Uro and the biologically distinct GU subtype. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Carina Bernardo
- Division of Oncology and Pathology, Department of Clinical SciencesLund UniversityLundSweden
| | - Pontus Eriksson
- Division of Oncology and Pathology, Department of Clinical SciencesLund UniversityLundSweden
| | - Nour‐al‐dain Marzouka
- Division of Oncology and Pathology, Department of Clinical SciencesLund UniversityLundSweden
| | - Fredrik Liedberg
- Division of Urological Research, Department of Translational MedicineLund University, Skåne University HospitalMalmöSweden
| | - Gottfrid Sjödahl
- Division of Urological Research, Department of Translational MedicineLund University, Skåne University HospitalMalmöSweden
| | - Mattias Höglund
- Division of Oncology and Pathology, Department of Clinical SciencesLund UniversityLundSweden
| |
Collapse
|
37
|
Liu C, Tate T, Batourina E, Truschel ST, Potter S, Adam M, Xiang T, Picard M, Reiley M, Schneider K, Tamargo M, Lu C, Chen X, He J, Kim H, Mendelsohn CL. Pparg promotes differentiation and regulates mitochondrial gene expression in bladder epithelial cells. Nat Commun 2019; 10:4589. [PMID: 31597917 PMCID: PMC6785552 DOI: 10.1038/s41467-019-12332-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 08/28/2019] [Indexed: 12/22/2022] Open
Abstract
The urothelium is an epithelial barrier lining the bladder that protects against infection, fluid exchange and damage from toxins. The nuclear receptor Pparg promotes urothelial differentiation in vitro, and Pparg mutations are associated with bladder cancer. However, the function of Pparg in the healthy urothelium is unknown. Here we show that Pparg is critical in urothelial cells for mitochondrial biogenesis, cellular differentiation and regulation of inflammation in response to urinary tract infection (UTI). Superficial cells, which are critical for maintaining the urothelial barrier, fail to mature in Pparg mutants and basal cells undergo squamous-like differentiation. Pparg mutants display persistent inflammation after UTI, and Nf-KB, which is transiently activated in response to infection in the wild type urothelium, persists for months. Our observations suggest that in addition to its known roles in adipogegnesis and macrophage differentiation, that Pparg-dependent transcription plays a role in the urothelium controlling mitochondrial function development and regeneration.
Collapse
Affiliation(s)
- Chang Liu
- Department of Urology, Genetics, and Devlopment, Pathology and Cell Biology and CSCI, Columbia University, New York, NY, 10032, USA
| | - Tiffany Tate
- Department of Urology, Genetics, and Devlopment, Pathology and Cell Biology and CSCI, Columbia University, New York, NY, 10032, USA
| | - Ekatherina Batourina
- Department of Urology, Genetics, and Devlopment, Pathology and Cell Biology and CSCI, Columbia University, New York, NY, 10032, USA
| | - Steven T Truschel
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Steven Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, OH, USA
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, OH, USA
| | - Tina Xiang
- Department of Urology, Genetics, and Devlopment, Pathology and Cell Biology and CSCI, Columbia University, New York, NY, 10032, USA
| | - Martin Picard
- Department of Psychiatry and Neurology, Columbia University, New York, NY, 10032, USA
| | - Maia Reiley
- Department of Urology, Genetics, and Devlopment, Pathology and Cell Biology and CSCI, Columbia University, New York, NY, 10032, USA
- Department of Surgery, Ascension/St. John Providence, 16001 West Nine Mile Road, Southfield, MI, 48075, USA
| | - Kerry Schneider
- Department of Urology, Genetics, and Devlopment, Pathology and Cell Biology and CSCI, Columbia University, New York, NY, 10032, USA
- College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Manuel Tamargo
- Department of Urology, Genetics, and Devlopment, Pathology and Cell Biology and CSCI, Columbia University, New York, NY, 10032, USA
| | - Chao Lu
- Department of Genetics and Development, Columbia University, New York, NY, 10032, USA
| | - Xiao Chen
- Department of Genetics and Development, Columbia University, New York, NY, 10032, USA
| | - Jing He
- Department of Systems Biology, Columbia University, New York, NY, 10032, USA
| | - Hyunwoo Kim
- Department of Urology, Genetics, and Devlopment, Pathology and Cell Biology and CSCI, Columbia University, New York, NY, 10032, USA
| | - Cathy Lee Mendelsohn
- Department of Urology, Genetics, and Devlopment, Pathology and Cell Biology and CSCI, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
38
|
Santos CP, Lapi E, Martínez de Villarreal J, Álvaro-Espinosa L, Fernández-Barral A, Barbáchano A, Domínguez O, Laughney AM, Megías D, Muñoz A, Real FX. Urothelial organoids originating from Cd49f high mouse stem cells display Notch-dependent differentiation capacity. Nat Commun 2019; 10:4407. [PMID: 31562298 PMCID: PMC6764959 DOI: 10.1038/s41467-019-12307-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 09/02/2019] [Indexed: 12/29/2022] Open
Abstract
Understanding urothelial stem cell biology and differentiation has been limited by the lack of methods for their unlimited propagation. Here, we establish mouse urothelial organoids that can be maintained uninterruptedly for >1 year. Organoid growth is dependent on EGF and Wnt activators. High CD49f/ITGA6 expression features a subpopulation of organoid-forming cells expressing basal markers. Upon differentiation, multilayered organoids undergo reduced proliferation, decreased cell layer number, urothelial program activation, and acquisition of barrier function. Pharmacological modulation of PPARγ and EGFR promotes differentiation. RNA sequencing highlighted genesets enriched in proliferative organoids (i.e. ribosome) and transcriptional networks involved in differentiation, including expression of Wnt ligands and Notch components. Single-cell RNA sequencing (scRNA-Seq) analysis of the organoids revealed five clusters with distinct gene expression profiles. Together, with the use of γ-secretase inhibitors and scRNA-Seq, confirms that Notch signaling is required for differentiation. Urothelial organoids provide a powerful tool to study cell regeneration and differentiation.
Collapse
Affiliation(s)
- Catarina P Santos
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Eleonora Lapi
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Jaime Martínez de Villarreal
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Laura Álvaro-Espinosa
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Asunción Fernández-Barral
- CIBERONC, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM and IdiPAZ, 28029, Madrid, Spain
| | - Antonio Barbáchano
- CIBERONC, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM and IdiPAZ, 28029, Madrid, Spain
| | - Orlando Domínguez
- Genomics Unit, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | | | - Diego Megías
- Confocal Microscopy Unit, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Alberto Muñoz
- CIBERONC, Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols, CSIC-UAM and IdiPAZ, 28029, Madrid, Spain
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain.
- CIBERONC, Madrid, Spain.
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
39
|
Daza-Cajigal V, Albuquerque AS, Pearson J, Hinley J, Mason AS, Stahlschmidt J, Thrasher AJ, Mishra V, Southgate J, Burns SO. Loss of Janus Associated Kinase 1 Alters Urothelial Cell Function and Facilitates the Development of Bladder Cancer. Front Immunol 2019; 10:2065. [PMID: 31552026 PMCID: PMC6746825 DOI: 10.3389/fimmu.2019.02065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 08/15/2019] [Indexed: 11/13/2022] Open
Abstract
Inherited Primary Immunodeficiency (PID) disorders are associated with increased risk of malignancy that may relate to impaired antitumor immune responses or a direct role for PID germline mutations in tumorigenesis. We recently identified germline loss of function mutations in Janus Associated Kinase 1 (JAK1) causing primary immunodeficiency characterized by infections and associated with early onset, fatal high-grade bladder carcinoma. Somatic mutations in JAK1, required for immune cell signaling in response to interferon gamma (IFNγ), have been associated with several non-hematopoietic and hematopoietic cancer cell types but pathogenic mechanisms remain largely unexplored. Here we demonstrate that JAK1 is required for the intrinsic IFNγ response of urothelial cells impacting immunogenicity and cell survival. Specifically, JAK1-deficient urothelial cells showed reduced surface expression of major histocompatibility complex class II (MHC II), intercellular adhesion molecule-1 (ICAM-1) and programmed death-ligand-1 (PD-L1) after IFNγ stimulation and were resistant to IFNγ-induced apoptosis and lymphocyte-mediated killing. In addition, we identify a previously unknown role for IFNγ signaling in modulating urothelial differentiation. Together, our findings support a role for urothelial cell JAK1 in immune surveillance and development of bladder cancer. Our results have implications for patients with rare JAK1 PID and, more broadly, inform development of biomarker and targeted therapies for urothelial carcinoma.
Collapse
Affiliation(s)
- Vanessa Daza-Cajigal
- Institute of Immunity and Transplantation, University College London, London, United Kingdom.,Department of Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom.,School of Medicine, Universidad Complutense, Madrid, Spain.,Department of Immunology, Hospital Universitario Son Espases, Palma, Spain.,Human Immunopathology Research Laboratory, Institut d'Investigació Sanitaria de Palma (IdISPa), Palma, Spain
| | - Adriana S Albuquerque
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Joanna Pearson
- Jack Birch Unit, Department of Biology, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Jennifer Hinley
- Jack Birch Unit, Department of Biology, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Andrew S Mason
- Jack Birch Unit, Department of Biology, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Jens Stahlschmidt
- Jack Birch Unit, Department of Biology, York Biomedical Research Institute, University of York, York, United Kingdom.,Department of Histopathology, St James's University Hospital, Leeds, United Kingdom
| | - Adrian J Thrasher
- Great Ormond Hospital for Children NHS Foundation Trust, London, United Kingdom.,Section of Molecular and Cellular Immunology, Institute of Child Health, University College London, London, United Kingdom
| | - Vibhash Mishra
- Department of Urology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Jennifer Southgate
- Jack Birch Unit, Department of Biology, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Siobhan O Burns
- Institute of Immunity and Transplantation, University College London, London, United Kingdom.,Department of Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
40
|
Inoue Y, Kishida T, Kotani SI, Akiyoshi M, Taga H, Seki M, Ukimura O, Mazda O. Direct conversion of fibroblasts into urothelial cells that may be recruited to regenerating mucosa of injured urinary bladder. Sci Rep 2019; 9:13850. [PMID: 31554870 PMCID: PMC6761134 DOI: 10.1038/s41598-019-50388-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 09/11/2019] [Indexed: 02/08/2023] Open
Abstract
Urothelial cells play essential roles in protection of urine exudation and bacterial invasion at the urothelial mucosa, so that defect or damage of urothelial cells associated with urinary tract diseases may cause serious problems. If a sufficient number of functional urothelial cells are prepared in culture and transplanted into the damaged urothelial lesions, such technology may provide beneficial effects to patients with diseases of the urinary tract. Here we found that human adult dermal fibroblasts were converted into urothelial cells by transducing genes for four transcription factors, FOXA1, TP63, MYCL and KLF4 (FTLK). The directly converted urothelial cells (dUCs) formed cobblestone-like colonies and expressed urothelium-specific markers. dUCs were successfully expanded and enriched after serial passages using a specific medium that we optimized for the cells. The passaged dUCs showed similar genome-wide gene expression profiles to normal urothelial cells and had a barrier function. The FTLK-transduced fibroblasts were also converted into urothelial cells in vivo and recruited to the regenerating urothelial tissue after they were transplanted into the bladder of mice with interstitial cystitis. Our technology may provide a promising solution for a number of patients with urinary tract disorders.
Collapse
Affiliation(s)
- Yuta Inoue
- Department of Immunology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan.,Department of Urology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tsunao Kishida
- Department of Immunology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shin-Ichiro Kotani
- Department of Immunology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Mika Akiyoshi
- Department of Immunology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan.,CellAxia Inc. 1-10-9-6F Nihonbashi Horidome-cho, Chuo-ku, Tokyo, 103-0012, Japan
| | - Hideto Taga
- Department of Immunology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan.,Department of Urology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Makoto Seki
- Department of Immunology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan.,CellAxia Inc. 1-10-9-6F Nihonbashi Horidome-cho, Chuo-ku, Tokyo, 103-0012, Japan
| | - Osamu Ukimura
- Department of Urology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefecture University of Medicine, Kamigyo-ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
41
|
Directed differentiation of human induced pluripotent stem cells into mature stratified bladder urothelium. Sci Rep 2019; 9:10506. [PMID: 31324820 PMCID: PMC6642190 DOI: 10.1038/s41598-019-46848-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 07/05/2019] [Indexed: 02/06/2023] Open
Abstract
For augmentation or reconstruction of urinary bladder after cystectomy, bladder urothelium derived from human induced pluripotent stem cells (hiPSCs) has recently received focus. However, previous studies have only shown the emergence of cells expressing some urothelial markers among derivatives of hiPSCs, and no report has demonstrated the stratified structure, which is a particularly important attribute of the barrier function of mature bladder urothelium. In present study, we developed a method for the directed differentiation of hiPSCs into mature stratified bladder urothelium. The caudal hindgut, from which the bladder urothelium develops, was predominantly induced via the high-dose administration of CHIR99021 during definitive endoderm induction, and this treatment subsequently increased the expressions of uroplakins. Terminal differentiation, characterized by the increased expression of uroplakins, CK13, and CK20, was induced with the combination of Troglitazone + PD153035. FGF10 enhanced the expression of uroplakins and the stratification of the epithelium, and the transwell culture system further enhanced such stratification. Furthermore, the barrier function of our urothelium was demonstrated by a permeability assay using FITC-dextran. According to an immunohistological analysis, the stratified uroplakin II-positive epithelium was observed in the transwells. This method might be useful in the field of regenerative medicine of the bladder.
Collapse
|
42
|
Ruan JL, Hsu JW, Browning RJ, Stride E, Yildiz YO, Vojnovic B, Kiltie AE. Mouse Models of Muscle-invasive Bladder Cancer: Key Considerations for Clinical Translation Based on Molecular Subtypes. Eur Urol Oncol 2019; 2:239-247. [PMID: 31200837 DOI: 10.1016/j.euo.2018.08.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/22/2018] [Accepted: 08/08/2018] [Indexed: 02/07/2023]
Abstract
CONTEXT In the past few years, research has suggested that molecular subtypes in muscle-invasive bladder cancer (MIBC) may be exploited to accelerate developments in clinical disease management and novel therapeutics. OBJECTIVE To review MIBC mouse models from a molecular subtype perspective, their advantages and limitations, and their applications in translational medicine, based on a PubMed search for publications from January 2000 to February 2018. EVIDENCE ACQUISITION Publications relevant to MIBC mouse models and their molecular subtypes were identified in a literature review. EVIDENCE SYNTHESIS We classified the models according to the technique used for their establishment. For xenotransplant and allograft models, the inoculated cells and inoculated locations are the major determinants of molecular subtypes. Although the cell lines used in xenotransplant models can cover most of the basal-squamous and luminal subtypes, allograft models offer a more realistic environment in which to reconstruct aspects of the associated stromal and immune features. Autochthonous models, using genetic and/or chemical stimuli to induce disease progression, can also generate models with basal-squamous and luminal subtypes, but further molecular characterisation is needed since other mutational variants may be introduced in these models. CONCLUSIONS We identified preclinical MIBC models with different subtype specifications and assessed their promise and current limitations. These models are versatile tools that can reproduce the molecular complexity of MIBC and support novel therapeutic development. PATIENT SUMMARY Understanding which models of muscle-invasive bladder cancer most accurately represent the clinical situation is important for the development of novel drugs and disease management strategies. We review the different models currently available and their relevance to different clinical subtypes.
Collapse
Affiliation(s)
- Jia-Ling Ruan
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Jong-Wei Hsu
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | | | - Eleanor Stride
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Yesna O Yildiz
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Borivoj Vojnovic
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Anne E Kiltie
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
43
|
Rochel N, Krucker C, Coutos-Thévenot L, Osz J, Zhang R, Guyon E, Zita W, Vanthong S, Hernandez OA, Bourguet M, Badawy KA, Dufour F, Peluso-Iltis C, Heckler-Beji S, Dejaegere A, Kamoun A, de Reyniès A, Neuzillet Y, Rebouissou S, Béraud C, Lang H, Massfelder T, Allory Y, Cianférani S, Stote RH, Radvanyi F, Bernard-Pierrot I. Recurrent activating mutations of PPARγ associated with luminal bladder tumors. Nat Commun 2019; 10:253. [PMID: 30651555 PMCID: PMC6335423 DOI: 10.1038/s41467-018-08157-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 12/18/2018] [Indexed: 11/21/2022] Open
Abstract
The upregulation of PPARγ/RXRα transcriptional activity has emerged as a key event in luminal bladder tumors. It renders tumor cell growth PPARγ-dependent and modulates the tumor microenvironment to favor escape from immuno-surveillance. The activation of the pathway has been linked to PPARG gains/amplifications resulting in PPARγ overexpression and to recurrent activating point mutations of RXRα. Here, we report recurrent mutations of PPARγ that also activate the PPARγ/RXRα pathway, conferring PPARγ-dependency and supporting a crucial role of PPARγ in luminal bladder cancer. These mutations are found throughout the protein—including N-terminal, DNA-binding and ligand-binding domains—and most of them enhance protein activity. Structure-function studies of PPARγ variants with mutations in the ligand-binding domain allow identifying structural elements that underpin their gain-of-function. Our study reveals genomic alterations of PPARG that lead to pro-tumorigenic PPARγ/RXRα pathway activation in luminal bladder tumors and may open the way towards alternative options for treatment. Activation of the PPARγ/RXRα pathway in luminal bladder cancers has mainly been linked to PPARG gene amplifications and activating point mutations in RXRα. Here, the authors identify recurrent PPARγ mutations with similar effects and elucidate the structural basis for this mutational PPARγ activation.
Collapse
Affiliation(s)
- Natacha Rochel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, 67404 Illkirch, France.
| | - Clémentine Krucker
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France
| | - Laure Coutos-Thévenot
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France
| | - Judit Osz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, 67404 Illkirch, France
| | - Ruiyun Zhang
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France.,Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Elodie Guyon
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France
| | - Wayne Zita
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, 67404 Illkirch, France
| | - Séverin Vanthong
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, 67404 Illkirch, France
| | - Oscar Alba Hernandez
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000, Strasbourg, France
| | - Maxime Bourguet
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000, Strasbourg, France
| | - Kays Al Badawy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, 67404 Illkirch, France
| | - Florent Dufour
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France
| | - Carole Peluso-Iltis
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, 67404 Illkirch, France
| | - Syrine Heckler-Beji
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, 67404 Illkirch, France
| | - Annick Dejaegere
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, 67404 Illkirch, France
| | - Aurélie Kamoun
- Ligue Nationale Contre le Cancer, Programme Cartes d'Identité des Tumeurs (CIT), 75013 Paris, France
| | - Aurélien de Reyniès
- Ligue Nationale Contre le Cancer, Programme Cartes d'Identité des Tumeurs (CIT), 75013 Paris, France
| | - Yann Neuzillet
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France
| | - Sandra Rebouissou
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France.,INSERM, UMR-1162 "Génomique Fonctionnelle des tumeurs solides", 75010, Paris, France
| | - Claire Béraud
- UROLEAD SAS, School of Medicine, 67085, Strasbourg, France
| | - Hervé Lang
- Department of Urology, Nouvel Hôpital Civil Hôpitaux Universitaires de Strasbourg, Hôpitaux Universitaires de Strasbourg, 67091, Strasbourg, France
| | - Thierry Massfelder
- INSERM UMRS1113, Section of Cell Signalization and Communication in Kidney and Prostate Cancer, INSERM and University of Strasbourg, School of Medicine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67085, Strasbourg, France
| | - Yves Allory
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000, Strasbourg, France
| | - Roland H Stote
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, 67404 Illkirch, France
| | - François Radvanyi
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France
| | - Isabelle Bernard-Pierrot
- Institut Curie, PSL Research University, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, 75005, Paris, France. .,Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005, Paris, France.
| |
Collapse
|
44
|
SIU-ICUD consultation on bladder cancer: basic science. World J Urol 2018; 37:15-29. [PMID: 30547196 DOI: 10.1007/s00345-018-2594-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/29/2018] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To provide a condensed summary of the Basic Science chapter that was included in the Third International Consultation on Bladder Cancer. METHODS World bladder cancer basic science experts used the published literature to create summaries of recent progress in their areas of expertise. RESULTS The completion of several large-scale genomics projects coupled with a strong collaborative culture within the research community and the exciting clinical activity of immune checkpoint blockade have combined to transform the bladder cancer research landscape. Bladder cancer molecular subtypes and the presence of specific DNA alterations provide important information about disease heterogeneity that has direct implications for clinical management, and some can be targeted by compounds that are already clinically available. Tests are being developed that can measure many of these alterations non-invasively in peripheral blood or urine, raising confidence that they could be used as biomarkers for surveillance and monitoring the effects of local and systemic therapies. CONCLUSIONS Although the bulk of the mechanistic work lies ahead, the genomics results have created a hypothesis-generating description of bladder cancer heterogeneity that has set the stage for deeper mechanistic studies, and they have already provided us with extremely attractive candidate biomarkers to guide clinical practice. Here, we will summarize the recent progress in basic bladder cancer research and highlight near-term opportunities for the future.
Collapse
|
45
|
Hustler A, Eardley I, Hinley J, Pearson J, Wezel F, Radvanyi F, Baker SC, Southgate J. Differential transcription factor expression by human epithelial cells of buccal and urothelial derivation. Exp Cell Res 2018; 369:284-294. [PMID: 29842880 PMCID: PMC6092173 DOI: 10.1016/j.yexcr.2018.05.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022]
Abstract
Identification of transcription factors expressed by differentiated cells is informative not only of tissue-specific pathways, but to help identify master regulators for cellular reprogramming. If applied, such an approach could generate healthy autologous tissue-specific cells for clinical use where cells from the homologous tissue are unavailable due to disease. Normal human epithelial cells of buccal and urothelial derivation maintained in identical culture conditions that lacked significant instructive or permissive signaling cues were found to display inherent similarities and differences of phenotype. Investigation of transcription factors implicated in driving urothelial-type differentiation revealed buccal epithelial cells to have minimal or absent expression of PPARG, GATA3 and FOXA1 genes. Retroviral overexpression of protein coding sequences for GATA3 or PPARy1 in buccal epithelial cells resulted in nuclear immunolocalisation of the respective proteins, with both transductions also inducing expression of the urothelial differentiation-associated claudin 3 tight junction protein. PPARG1 overexpression alone entrained expression of nuclear FOXA1 and GATA3 proteins, providing objective evidence of its upstream positioning in a transcription factor network and identifying it as a candidate factor for urothelial-type transdifferentiation or reprogramming.
Collapse
Affiliation(s)
- Arianna Hustler
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Ian Eardley
- Pyrah Department of Urology, St. James's University Hospital, Leeds LS9 7TF, United Kingdom
| | - Jennifer Hinley
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Joanna Pearson
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Felix Wezel
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Francois Radvanyi
- Oncologie Moléculaire, Institut Curie, Centre de Recherche, 75248 Paris cedex 05, France
| | - Simon C Baker
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Jennifer Southgate
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom.
| |
Collapse
|
46
|
Mahal A, Young-Lin N, Dobberfuhl A, Estes J, Comiter CV. Peroxisome proliferator-activated receptor gamma agonist as a novel treatment for interstitial cystitis: A rat model. Investig Clin Urol 2018; 59:257-262. [PMID: 29984341 PMCID: PMC6028466 DOI: 10.4111/icu.2018.59.4.257] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/24/2018] [Indexed: 01/02/2023] Open
Abstract
Purpose To understand the therapeutic potential of pioglitazone, a peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist with a propensity to cause bladder mucosal proliferation, on interstitial cystitis (IC) in a rat model. Materials and Methods Using a previously described animal model for IC, Sprague-Dawley rats were treated with biweekly cyclophosphamide injections (35 mg/kg) to induce cystitis. Animals were divided into 4 groups (n=6 for each group): IC plus daily sham saline gavage (IC+Pio−), IC plus daily pioglitazone gavage (15 mg/kg) (IC+Pio+), normal rats with daily pioglitazone (IC−Pio+), and normal rats with neither IC nor pioglitazone (IC−Pio− or Control). At the end of four weeks, urinary frequency and bladder capacity were measured. Histologic examination of urothelial integrity was also performed. Results Average voids per hour were significantly lower in IC+Pio+ (4.0±1.9) vs. IC+Pio− (10.0±2.4) rats (p<0.01) and were similar to IC−Pio+ (6.0±1.4) and IC−Pio− (6.0±1.5) controls. Cystometric capacity was significantly higher in IC+Pio+ (0.945±0.122 mL) vs. IC+Pio− rats (0.588±0.165 mL, p=0.01) and was comparable to IC−Pio− capacity (0.817±0.196 mL) and IC−Pio+ capacity (0.941±0.188 mL). Urothelial structural integrity was improved in IC+Pio+ rats versus IC+Pio− rats upon histologic observation. Conclusions Pioglitazone, a PPAR-γ agonist, improved bladder function in cyclophosphamide-induced cystitis by both observed urinary frequency and measured cystometric capacity. Urothelial structural integrity was also improved. Pioglitazone, due to a propensity to cause bladder mucosal proliferation, may prove useful for treating IC, and deserves further investigation.
Collapse
Affiliation(s)
- Amandeep Mahal
- Division of Urogynecology and Pelvic Reconstructive Surgery, Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nichole Young-Lin
- Division of Urogynecology and Pelvic Reconstructive Surgery, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Amy Dobberfuhl
- Division of Urogynecology and Pelvic Reconstructive Surgery, Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jaclyn Estes
- Division of Urogynecology and Pelvic Reconstructive Surgery, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Craig Vance Comiter
- Division of Urogynecology and Pelvic Reconstructive Surgery, Department of Urology, Stanford University School of Medicine, Stanford, CA, USA.,Division of Urogynecology and Pelvic Reconstructive Surgery, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
47
|
Weber L, Schulz WA, Philippou S, Eckardt J, Ubrig B, Hoffmann MJ, Tannapfel A, Kalbe B, Gisselmann G, Hatt H. Characterization of the Olfactory Receptor OR10H1 in Human Urinary Bladder Cancer. Front Physiol 2018; 9:456. [PMID: 29867524 PMCID: PMC5964926 DOI: 10.3389/fphys.2018.00456] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
Olfactory receptors (ORs) are a large group of G-protein coupled receptors predominantly found in the olfactory epithelium. Many ORs are, however, ectopically expressed in other tissues and involved in several diseases including cancer. In this study, we describe that one OR, OR10H1, is predominantly expressed in the human urinary bladder with a notably higher expression at mRNA and protein level in bladder cancer tissues. Interestingly, also significantly higher amounts of OR10H1 transcripts were detectable in the urine of bladder cancer patients than in the urine of control persons. We identified the sandalwood-related compound Sandranol as a specific agonist of OR10H1. This deorphanization allowed the functional characterization of OR10H1 in BFTC905 bladder cancer cells. The effect of receptor activation was morphologically apparent in cell rounding, accompanied by changes in the cytoskeleton detected by β-actin, T-cadherin and β-Catenin staining. In addition, Sandranol treatment significantly diminished cell viability, cell proliferation and migration and induced a limited degree of apoptosis. Cell cycle analysis revealed an increased G1 fraction. In a concentration-dependent manner, Sandranol application elevated cAMP levels, which was reduced by inhibition of adenylyl cyclase, and elicited intracellular Ca2+ concentration increase. Furthermore, activation of OR10H1 enhanced secretion of ATP and serotonin. Our results suggest OR10H1 as a potential biomarker and therapeutic target for bladder cancer.
Collapse
Affiliation(s)
- Lea Weber
- Department of Cellular Physiology, Ruhr University Bochum, Bochum, Germany.,Department of Translational Wound Research, Witten/Herdecke University, Witten, Germany
| | - Wolfgang A Schulz
- Department of Urology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stathis Philippou
- Department of Pathology and Cytology, Augusta Kliniken Bochum Hattingen, Bochum, Germany
| | - Josephine Eckardt
- Department of Cellular Physiology, Ruhr University Bochum, Bochum, Germany.,Institute for Physiology, Ruhr University Bochum, Bochum, Germany
| | - Burkhard Ubrig
- Clinic for Urology, Augusta Kliniken Bochum Hattingen, Bochum, Germany
| | - Michéle J Hoffmann
- Department of Urology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andrea Tannapfel
- Institute for Pathology, Ruhr University Bochum, Bochum, Germany
| | - Benjamin Kalbe
- Department of Cellular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Günter Gisselmann
- Department of Cellular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Hanns Hatt
- Department of Cellular Physiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
48
|
Baker SC, Arlt VM, Indra R, Joel M, Stiborová M, Eardley I, Ahmad N, Otto W, Burger M, Rubenwolf P, Phillips DH, Southgate J. Differentiation-associated urothelial cytochrome P450 oxidoreductase predicates the xenobiotic-metabolizing activity of "luminal" muscle-invasive bladder cancers. Mol Carcinog 2018; 57:606-618. [PMID: 29323757 PMCID: PMC5900743 DOI: 10.1002/mc.22784] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/21/2017] [Accepted: 01/09/2018] [Indexed: 11/12/2022]
Abstract
Extra-hepatic metabolism of xenobiotics by epithelial tissues has evolved as a self-defence mechanism but has potential to contribute to the local activation of carcinogens. Bladder epithelium (urothelium) is bathed in excreted urinary toxicants and pro-carcinogens. This study reveals how differentiation affects cytochrome P450 (CYP) activity and the role of NADPH:P450 oxidoreductase (POR). CYP1A1 and CYP1B1 transcripts were inducible in normal human urothelial (NHU) cells maintained in both undifferentiated and functional barrier-forming differentiated states in vitro. However, ethoxyresorufin O-deethylation (EROD) activity, the generation of reactive BaP metabolites and BaP-DNA adducts, were predominantly detected in differentiated NHU cell cultures. This gain-of-function was attributable to the expression of POR, an essential electron donor for all CYPs, which was significantly upregulated as part of urothelial differentiation. Immunohistology of muscle-invasive bladder cancer (MIBC) revealed significant overall suppression of POR expression. Stratification of MIBC biopsies into "luminal" and "basal" groups, based on GATA3 and cytokeratin 5/6 labeling, showed POR over-expression by a subgroup of the differentiated luminal tumors. In bladder cancer cell lines, CYP1-activity was undetectable/low in basal PORlo T24 and SCaBER cells and higher in the luminal POR over-expressing RT4 and RT112 cells than in differentiated NHU cells, indicating that CYP-function is related to differentiation status in bladder cancers. This study establishes POR as a predictive biomarker of metabolic potential. This has implications in bladder carcinogenesis for the hepatic versus local activation of carcinogens and as a functional predictor of the potential for MIBC to respond to prodrug therapies.
Collapse
Affiliation(s)
- Simon C. Baker
- Jack Birch Unit of Molecular CarcinogenesisDepartment of BiologyUniversity of YorkHeslingtonYorkUK
| | - Volker M. Arlt
- Department of Analytical, Environmental and Forensic SciencesMRC‐PHE Centre for Environment and HealthKing's College LondonFranklin‐Wilkins BuildingLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King's College London in Partnership with Public Health EnglandFranklin‐Wilkins BuildingLondonUK
| | - Radek Indra
- Faculty of ScienceDepartment of BiochemistryCharles UniversityAlbertovPragueCzech Republic
| | - Madeleine Joel
- Department of Analytical, Environmental and Forensic SciencesMRC‐PHE Centre for Environment and HealthKing's College LondonFranklin‐Wilkins BuildingLondonUK
| | - Marie Stiborová
- Faculty of ScienceDepartment of BiochemistryCharles UniversityAlbertovPragueCzech Republic
| | | | | | - Wolfgang Otto
- Department of UrologyRegensburg University Medical CentreRegensburgGermany
| | - Maximilian Burger
- Department of UrologyRegensburg University Medical CentreRegensburgGermany
- Department of UrologyFrankfurt University Medical Center, Johann Wolfgang Goethe‐UniversityFrankfurt am MainGermany
| | - Peter Rubenwolf
- Department of UrologyRegensburg University Medical CentreRegensburgGermany
| | - David H. Phillips
- Department of Analytical, Environmental and Forensic SciencesMRC‐PHE Centre for Environment and HealthKing's College LondonFranklin‐Wilkins BuildingLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King's College London in Partnership with Public Health EnglandFranklin‐Wilkins BuildingLondonUK
| | - Jennifer Southgate
- Jack Birch Unit of Molecular CarcinogenesisDepartment of BiologyUniversity of YorkHeslingtonYorkUK
| |
Collapse
|
49
|
Mo Q, Nikolos F, Chen F, Tramel Z, Lee YC, Hayashi K, Xiao J, Shen J, Chan KS. Prognostic Power of a Tumor Differentiation Gene Signature for Bladder Urothelial Carcinomas. J Natl Cancer Inst 2018; 110:448-459. [PMID: 29342309 PMCID: PMC6279371 DOI: 10.1093/jnci/djx243] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/13/2017] [Accepted: 10/20/2017] [Indexed: 01/05/2023] Open
Abstract
Background Muscle-invasive bladder cancers (MIBCs) cause approximately 150 000 deaths per year worldwide. Survival for MIBC patients is heterogeneous, with no clinically validated molecular markers that predict clinical outcome. Non-MIBCs (NMIBCs) generally have favorable outcome; however, a portion progress to MIBC. Hence, development of a prognostic tool that can guide decision-making is crucial for improving clinical management of bladder urothelial carcinomas. Methods Tumor grade is defined by pathologic evaluation of tumor cell differentiation, and it often associates with clinical outcome. The current study extrapolates this conventional wisdom and combines it with molecular profiling. We developed an 18-gene signature that molecularly defines urothelial cellular differentiation, thus classifying MIBCs and NMIBCs into two subgroups: basal and differentiated. We evaluated the prognostic capability of this "tumor differentiation signature" and three other existing gene signatures including the The Cancer Genome Atlas (TCGA; 2707 genes), MD Anderson Cancer Center (MDA; 2252 genes/2697 probes), and University of North Carolina at Chapel Hill (UNC; 47 genes) using five gene expression data sets derived from MIBC and NMIBC patients. All statistical tests were two-sided. Results The tumor differentiation signature demonstrated consistency and statistical robustness toward stratifying MIBC patients into different overall survival outcomes (TCGA cohort 1, P = .03; MDA discovery, P = .009; MDA validation, P = .01), while the other signatures were not as consistent. In addition, we analyzed the progression (Ta/T1 progressing to ≥T2) probability of NMIBCs. NMIBC patients with a basal tumor differentiation signature associated with worse progression outcome (P = .008). Gene functional term enrichment and gene set enrichment analyses revealed that genes involved in the biologic process of immune response and inflammatory response are among the most elevated within basal bladder cancers, implicating them as candidates for immune checkpoint therapies. Conclusions These results provide definitive evidence that a biology-prioritizing clustering methodology generates meaningful insights into patient stratification and reveals targetable molecular pathways to impact future therapeutic approach.
Collapse
Affiliation(s)
- Qianxing Mo
- Department of Medicine, University of Texas MD Anderson Cancer Center, Smithville, TX
- Dan L. Duncan Comprehensive Cancer Center, University of Texas MD Anderson Cancer Center, Smithville, TX
| | - Fotis Nikolos
- Department of Molecular and Cellular Biology, University of Texas MD Anderson Cancer Center, Smithville, TX
| | - Fengju Chen
- Dan L. Duncan Comprehensive Cancer Center, University of Texas MD Anderson Cancer Center, Smithville, TX
| | - Zoe Tramel
- Department of Molecular and Cellular Biology, University of Texas MD Anderson Cancer Center, Smithville, TX
| | - Yu-Cheng Lee
- Department of Molecular and Cellular Biology, University of Texas MD Anderson Cancer Center, Smithville, TX
| | - Kazukuni Hayashi
- Department of Molecular and Cellular Biology, University of Texas MD Anderson Cancer Center, Smithville, TX
| | - Jing Xiao
- Department of Molecular and Cellular Biology, University of Texas MD Anderson Cancer Center, Smithville, TX
| | - Jianjun Shen
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX
| | - Keith Syson Chan
- Department of Molecular and Cellular Biology, University of Texas MD Anderson Cancer Center, Smithville, TX
- Stem Cells and Regenerative Medicine (STaR) Center, Center for Drug Discovery, and Scott Department of Urology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
50
|
Choi W, Ochoa A, McConkey DJ, Aine M, Höglund M, Kim WY, Real FX, Kiltie AE, Milsom I, Dyrskjøt L, Lerner SP. Genetic Alterations in the Molecular Subtypes of Bladder Cancer: Illustration in the Cancer Genome Atlas Dataset. Eur Urol 2017; 72:354-365. [PMID: 28365159 PMCID: PMC5764190 DOI: 10.1016/j.eururo.2017.03.010] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/06/2017] [Indexed: 12/14/2022]
Abstract
CONTEXT Recent whole genome mRNA expression profiling studies revealed that bladder cancers can be grouped into molecular subtypes, some of which share clinical properties and gene expression patterns with the intrinsic subtypes of breast cancer and the molecular subtypes found in other solid tumors. The molecular subtypes in other solid tumors are enriched with specific mutations and copy number aberrations that are thought to underlie their distinct progression patterns, and biological and clinical properties. OBJECTIVE The availability of comprehensive genomic data from The Cancer Genome Atlas (TCGA) and other large projects made it possible to correlate the presence of DNA alterations with tumor molecular subtype membership. Our overall goal was to determine whether specific DNA mutations and/or copy number variations are enriched in specific molecular subtypes. EVIDENCE We used the complete TCGA RNA-seq dataset and three different published classifiers developed by our groups to assign TCGA's bladder cancers to molecular subtypes, and examined the prevalence of the most common DNA alterations within them. We interpreted the results against the background of what was known from the published literature about the prevalence of these alterations in nonmuscle-invasive and muscle-invasive bladder cancers. EVIDENCE SYNTHESIS The results confirmed that alterations involving RB1 and NFE2L2 were enriched in basal cancers, whereas alterations involving FGFR3 and KDM6A were enriched in luminal tumors. CONCLUSIONS The results further reinforce the conclusion that the molecular subtypes of bladder cancer are distinct disease entities with specific genetic alterations. PATIENT SUMMARY Our observation showed that some of subtype-enriched mutations and copy number aberrations are clinically actionable, which has direct implications for the clinical management of patients with bladder cancer.
Collapse
Affiliation(s)
- Woonyoung Choi
- Department of Urology, U.T. M.D. Anderson Cancer Center, Houston, Texas, USA; Department of Cancer Biology, U.T. M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Andrea Ochoa
- Department of Urology, U.T. M.D. Anderson Cancer Center, Houston, Texas, USA; Department of Cancer Biology, U.T. M.D. Anderson Cancer Center, Houston, Texas, USA
| | - David J McConkey
- Department of Urology, U.T. M.D. Anderson Cancer Center, Houston, Texas, USA; Department of Cancer Biology, U.T. M.D. Anderson Cancer Center, Houston, Texas, USA.
| | - Mattias Aine
- Division of Oncology and Pathology, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Mattias Höglund
- Division of Oncology and Pathology, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - William Y Kim
- University of North Carolina, Chapel Hill, North Carolina, USA
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain; Departament de Ciències Experimentalsi de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Anne E Kiltie
- Department of Oncology, University of Oxford, Oxford, UK
| | - Ian Milsom
- Institute of Clinical Sciences, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Seth P Lerner
- TCGA Analysis Working Group, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|