1
|
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 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
|
2
|
Jasmine, Baraiya DH, Kavya TT, Mandal A, Chakraborty S, Sathish N, Francis CMR, Binoy Joseph D. Epithelial and mesenchymal compartments of the developing bladder and urethra display spatially distinct gene expression patterns. Dev Biol 2025; 520:155-170. [PMID: 39798644 DOI: 10.1016/j.ydbio.2025.01.005] [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: 03/22/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
The lower urinary tract is comprised of the bladder and urethra and develops from the cloaca, a transient endoderm-derived structure formed from the caudal hindgut. After cloacal septation to form the urogenital sinus and anorectal tract, the bladder gradually develops from the anterior portion of the urogenital sinus while the urethra elongates distally into the genital tubercle. The bladder is a target for regenerative and reconstructive therapies but engineering an impermeable bladder epithelial lining has proven challenging. Urethral epithelial function, including its role as an active immune barrier, is poorly studied and neglected in regenerative therapy. A deeper understanding of epithelial patterning of the urogenital sinus by the surrounding mesenchyme, also accounting for sex-specific differences, can inform regenerative therapies. In this study, we identified spatially distinct genes in the epithelial and mesenchymal compartments of the developing mouse bladder and urethra that could be potential drivers of patterning in the lower urinary tract. Our data revealed spatially restricted domains of transcription factor expression in the epithelium that corresponded with bladder or urethra-specific differentiation. Additionally, we identified the genes Wnt2, Klf4 and Pitx2 that localize to the mesenchyme of the developing bladder and could be potential drivers of bladder differentiation. Our data revealed an increase in the expression of several chemokine genes including Cx3cl1 and Cxcl14 in the developing urethral epithelium that correlated with an increase in epithelial-associated macrophages in the urethra. A survey of sex-specific differences in epithelial and mesenchymal compartments revealed several differentially expressed genes between the male and female urethra but few sex-specific differences in bladder. By comparing spatially distinct gene expression in the developing lower urinary tract, our study provides insights into the divergent differentiation trajectories of the fetal bladder and urethra that establish their adult functions.
Collapse
Affiliation(s)
- Jasmine
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Divyeksha H Baraiya
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - T T Kavya
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Aparna Mandal
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Shreya Chakraborty
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Neha Sathish
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Cynthia Marian Rebecca Francis
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India
| | - Diya Binoy Joseph
- Institute for Stem Cell Science and Regenerative Medicine (iBRIC-inStem), GKVK-Post, Bellary Road, Bengaluru, Karnataka, 560065, India.
| |
Collapse
|
3
|
Kispert A. Ureter development and associated congenital anomalies. Nat Rev Nephrol 2025:10.1038/s41581-025-00951-4. [PMID: 40164775 DOI: 10.1038/s41581-025-00951-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
4
|
Dragar B, Kranjc Brezar S, Čemažar M, Jesenko T, Romih R, Kreft ME, Kuret T, Zupančič D. Vitamin A-Enriched Diet Increases Urothelial Cell Proliferation by Upregulating Itga3 and Areg After Cyclophosphamide-Induced Injury in Mice. Mol Nutr Food Res 2025:e70045. [PMID: 40119798 DOI: 10.1002/mnfr.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/03/2025] [Accepted: 03/11/2025] [Indexed: 03/24/2025]
Abstract
Vitamin A (VitA) is an essential nutrient, affecting many cell functions, such as proliferation, apoptosis, and differentiation, all of which are important for the regeneration of various tissues. In this study, we investigated the effects of a VitA-enriched diet on the regeneration of the urothelium of the urinary bladder in mice after cyclophosphamide (CP)-induced injury. Female mice were fed VitA-enriched and normal diet for 1 week before receiving an intraperitoneal injection of CP (150 mg/kg). Urinary bladders were removed 1 and 3 days after CP. On Day 1, RNA sequencing showed that VitA upregulated two Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways: the cell cycle and the PI3K-Akt pathway. This was confirmed by qPCR, which showed significantly increased expression of the Itga3 and Areg genes. In addition, the effect of VitA on the proliferation of urothelial cells was analyzed by immunohistochemistry of Ki-67, which confirmed an increased proliferation rate. No significant effects of the VitA-enriched diet were observed on the expression of apoptosis-related genes and on differentiation-related markers of superficial urothelial cells. Our results suggest that a VitA-enriched diet improves early urothelial regeneration after CP-induced injury by promoting cell proliferation.
Collapse
Affiliation(s)
- Brina Dragar
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | | - Maja Čemažar
- Department of Experimental Oncology, Institute of Oncology, Ljubljana, Slovenia
| | - Tanja Jesenko
- Department of Experimental Oncology, Institute of Oncology, Ljubljana, Slovenia
| | - Rok Romih
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tadeja Kuret
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Daša Zupančič
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
5
|
Straube P, Beckers A, Jany UWH, Bergmann F, Lüdtke THW, Rudat C, Trowe MO, Peters I, Klopf MG, Mamo TM, Kispert A. Interplay of SHH, WNT and BMP4 signaling regulates the development of the lamina propria in the murine ureter. Development 2025; 152:DEV204214. [PMID: 39817691 PMCID: PMC11829765 DOI: 10.1242/dev.204214] [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: 06/26/2024] [Accepted: 12/31/2024] [Indexed: 01/18/2025]
Abstract
In mammalian ureters, the lamina propria presents as a prominent layer of connective tissue underneath the urothelium. Despite its important structural and signaling functions, little is known how the lamina propria develops. Here, we show that in the murine ureter the lamina propria arises at late fetal stages and massively increases by fibrocyte proliferation and collagen deposition after birth. WNT, SHH, BMP4 and retinoic acid signaling are all active in the common mesenchymal progenitor of smooth muscle cells and lamina propria fibrocytes. However, around birth, the lamina propria becomes a target for epithelial WNT and SHH signals and a source of BMP4 and retinoic acid. SHH and WNT signaling promote lamina propria and smooth muscle cell differentiation and proliferation at fetal and early postnatal stages, whereas BMP4 signaling is required for early smooth muscle cell differentiation but not for its later maintenance. Our findings suggest that, in the presence of SHH and WNT signaling, it is the modulation of BMP4 signaling which is the major determinant for the segregation of lamina propria and smooth muscle cells.
Collapse
Affiliation(s)
- Philipp Straube
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Anja Beckers
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Ulrich W. H. Jany
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Florian Bergmann
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Timo H.-W. Lüdtke
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Carsten Rudat
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Imke Peters
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Maximilian G. Klopf
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Tamrat M. Mamo
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
6
|
Fashemi BE, Rougeau AK, Salazar AM, Bark SJ, Chappidi R, Brown JW, Cho CJ, Mills JC, Mysorekar IU. IFRD1 is required for maintenance of bladder epithelial homeostasis. iScience 2024; 27:111282. [PMID: 39628564 PMCID: PMC11613175 DOI: 10.1016/j.isci.2024.111282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/21/2024] [Accepted: 10/25/2024] [Indexed: 12/06/2024] Open
Abstract
The maintenance of homeostasis and rapid regeneration of the urothelium following stress are critical for bladder function. Here, we identify a key role for IFRD1 in maintaining urothelial homeostasis in a mouse model. We demonstrate that the murine bladder expresses IFRD1 at homeostasis, particularly in the urothelium, and its loss alters the global transcriptome with significant accumulation of endolysosomes and dysregulated uroplakin expression pattern. We show that IFRD1 interacts with mRNA-translation-regulating factors in human urothelial cells. Loss of Ifrd1 leads to disrupted proteostasis, enhanced endoplasmic reticulum (ER stress) with activation of the PERK arm of the unfolded protein response pathway, and increased oxidative stress. Ifrd1-deficient bladders exhibit urothelial cell apoptosis/exfoliation, enhanced basal cell proliferation, reduced differentiation into superficial cells, increased urothelial permeability, and aberrant voiding behavior. These findings highlight a crucial role for IFRD1 in urothelial homeostasis, suggesting its potential as a therapeutic target for bladder dysfunction.
Collapse
Affiliation(s)
- Bisiayo E. Fashemi
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amala K. Rougeau
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Arnold M. Salazar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Steven J. Bark
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Rayvanth Chappidi
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey W. Brown
- Department of Medicine, Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Charles J. Cho
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Jason C. Mills
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Indira U. Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Huffington Center on Aging, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
7
|
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]
|
8
|
Plumber SA, Tate T, Al-Ahmadie H, Chen X, Choi W, Basar M, Lu C, Viny A, Batourina E, Li J, Gretarsson K, Alija B, Molotkov A, Wiessner G, Lee BHL, McKiernan J, McConkey DJ, Dinney C, Czerniak B, Mendelsohn CL. Rosiglitazone and trametinib exhibit potent anti-tumor activity in a mouse model of muscle invasive bladder cancer. Nat Commun 2024; 15:6538. [PMID: 39095358 PMCID: PMC11297265 DOI: 10.1038/s41467-024-50678-2] [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: 08/28/2023] [Accepted: 07/18/2024] [Indexed: 08/04/2024] Open
Abstract
Muscle invasive bladder cancers (BCs) can be divided into 2 major subgroups-basal/squamous (BASQ) tumors and luminal tumors. Since Pparg has low or undetectable expression in BASQ tumors, we tested the effects of rosiglitazone, Pparg agonist, in a mouse model of BASQ BC. We find that rosiglitazone reduces proliferation while treatment with rosiglitazone plus trametinib, a MEK inhibitor, induces apoptosis and reduces tumor volume by 91% after 1 month. Rosiglitazone and trametinib also induce a shift from BASQ to luminal differentiation in tumors, which our analysis suggests is mediated by retinoid signaling, a pathway known to drive the luminal differentiation program. Our data suggest that rosiglitazone, trametinib, and retinoids, which are all FDA approved, may be clinically active in BASQ tumors in patients.
Collapse
Affiliation(s)
- Sakina A Plumber
- Department of Urology, Columbia University Irving Medical Center, New York, NY, USA
| | - Tiffany Tate
- Department of Urology, Columbia University Irving Medical Center, New York, NY, USA
- Generation Bio, Cambridge, MA, USA
| | | | - Xiao Chen
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
- Marine College, Shandong University, Weihai, China
| | - Woonyoung Choi
- Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, MD, USA
| | - Merve Basar
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Chao Lu
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Aaron Viny
- Department of Medicine, Division of Hematology & Oncology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ekatherina Batourina
- Department of Urology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jiaqi Li
- Department of Urology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kristjan Gretarsson
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Besmira Alija
- Department of Medicine, Division of Hematology & Oncology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrei Molotkov
- Department of Urology, Columbia University Irving Medical Center, New York, NY, USA
| | - Gregory Wiessner
- Department of Urology, Columbia University Irving Medical Center, New York, NY, USA
| | - Byron Hing Lung Lee
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James McKiernan
- Department of Urology, Columbia University Irving Medical Center, New York, NY, USA
| | - David J McConkey
- Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, MD, USA
| | - Colin Dinney
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bogdan Czerniak
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cathy Lee Mendelsohn
- Department of Urology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
9
|
Becknell B, El-Harakeh M, Rodriguez-Tirado F, Grounds KM, Li B, Kercsmar M, Wang X, Jackson AR. Keratin 5 basal cells are temporally regulated developmental and tissue repair progenitors in bladder urothelium. Am J Physiol Renal Physiol 2024; 326:F1078-F1090. [PMID: 38634130 PMCID: PMC11386981 DOI: 10.1152/ajprenal.00378.2023] [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: 12/05/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
Urothelium forms a distensible yet impermeable barrier, senses and transduces stimuli, and defends the urinary tract from mechanical, chemical, and bacterial injuries. Biochemical and genetic labeling studies support the existence of one or more progenitor populations with the capacity to rapidly regenerate the urothelium following injury, but slow turnover, a low mitotic index, and inconsistent methodologies obscure progenitor identity. The progenitor properties of basal keratin 5 urothelial cells (K5-UCs) have been previously investigated, but those studies focused on embryonic or adult bladder urothelium. Urothelium undergoes desquamation and apoptosis after birth, which requires postnatal proliferation and restoration. Therefore, we mapped the fate of bladder K5-UCs across postnatal development/maturation and following administration of cyclophosphamide to measure homeostatic and reparative progenitor capacities, respectively. In vivo studies demonstrate that basal K5-UCs are age-restricted progenitors in neonates and juveniles, but not in adult mice. Neonatal K5-UCs retain a superior progenitor capacity in vitro, forming larger and more differentiated urothelial organoids than adult K5-UCs. Accordingly, K5-UC transcriptomes are temporally distinct, with enrichment of transcripts associated with cell proliferation and differentiation in neonates. Induction of urothelial proliferation is sufficient to restore adult K5-UC progenitor capacity. Our findings advance the understanding of urothelial progenitors and support a linear model of urothelial formation and regeneration, which may have significant impact on therapeutic development or tissue engineering strategies.NEW & NOTEWORTHY Fate mapping reveals an important linear relationship, whereby bladder basal urothelial cells give rise to intermediate and superficial cells in an age-restricted manner and contribute to tissue repair. Neonatal basal cells reprise their role as superior progenitors in vitro and display distinct transcriptional signatures, which suggest progenitor function is at least partially cell intrinsic. However, the urothelium progenitor niche cannot be overlooked, since FGF7 rescues adult basal cell progenitor function.
Collapse
Affiliation(s)
- Brian Becknell
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, Ohio, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States
| | - Mohammad El-Harakeh
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Felipe Rodriguez-Tirado
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Kelly M Grounds
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Birong Li
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Macie Kercsmar
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Xin Wang
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Ashley R Jackson
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, Ohio, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States
| |
Collapse
|
10
|
Qu N, Daoud A, Kechele DO, Múnera JO. Human Pluripotent Stem Cell Derived Organoids Reveal a Role for WNT Signaling in Dorsal-Ventral Patterning of the Hindgut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583343. [PMID: 38496665 PMCID: PMC10942392 DOI: 10.1101/2024.03.04.583343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The cloaca is a transient structure that forms in the terminal hindgut giving rise to the rectum dorsally and the urogenital sinus ventrally. Similarly, human hindgut cultures derived from human pluripotent stem cells generate human colonic organoids (HCOs) which also contain co-developing urothelial tissue. In this study, our goal was to identify pathways involved in cloacal patterning and apply this to human hindgut cultures. RNA-seq data comparing dorsal versus ventral cloaca in e10.5 mice revealed that WNT signaling was elevated in the ventral versus dorsal cloaca. Inhibition of WNT signaling in hindgut cultures biased their differentiation towards a colorectal fate. WNT activation biased differentiation towards a urothelial fate, giving rise to human urothelial organoids (HUOs). HUOs contained cell types present in human urothelial tissue. Based on our results, we propose a mechanism whereby WNT signaling patterns the ventral cloaca, prior to cloacal septation, to give rise to the urogenital sinus.
Collapse
|
11
|
Mann EA, Mogle MS, Park J, Reddy P. Transcription factor Tcf21 modulates urinary bladder size and differentiation. Dev Growth Differ 2024; 66:106-118. [PMID: 38197329 PMCID: PMC11457511 DOI: 10.1111/dgd.12906] [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: 09/12/2023] [Revised: 11/28/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024]
Abstract
Urinary bladder organogenesis requires coordinated cell growth, specification, and patterning of both mesenchymal and epithelial compartments. Tcf21, a gene that encodes a helix-loop-helix transcription factor, is specifically expressed in the mesenchyme of the bladder during development. Here we show that Tcf21 is required for normal development of the bladder. We found that the bladders of mice lacking Tcf21 were notably hypoplastic and that the Tcf21 mutant mesenchyme showed increased apoptosis. There was also a marked delay in the formation of visceral smooth muscle, accompanied by a defect in myocardin (Myocd) expression. Interestingly, there was also a marked delay in the formation of the basal cell layer of the urothelium, distinguished by diminished expression of Krt5 and Krt14. Our findings suggest that Tcf21 regulates the survival and differentiation of mesenchyme cell-autonomously and the maturation of the adjacent urothelium non-cell-autonomously during bladder development.
Collapse
Affiliation(s)
- Elizabeth A. Mann
- Division of Pediatric UrologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Melissa S. Mogle
- Division of Pediatric UrologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Joo‐Seop Park
- Division of Nephrology and HypertensionNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- The Feinberg Cardiovascular and Renal Research InstituteChicagoIllinoisUSA
| | - Pramod Reddy
- Division of Pediatric UrologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| |
Collapse
|
12
|
Fashemi BE, Rougeau AK, Salazar AM, Bark SJ, Chappidi R, Brown JW, Cho CJ, Mills JC, Mysorekar IU. A new role for IFRD1 in regulation of ER stress in bladder epithelial homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574887. [PMID: 38260387 PMCID: PMC10802459 DOI: 10.1101/2024.01.09.574887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
A healthy bladder requires the homeostatic maintenance of and rapid regeneration of urothelium upon stress/injury/infection. Several factors have been identified to play important roles in urothelial development, injury and disease response, however, little is known about urothelial regulation at homeostasis. Here, we identify a new role for IFRD1, a stress-induced gene that has recently been demonstrated to play a critical role in adult tissue proliferation and regeneration, in maintenance of urothelial function/ homeostasis in a mouse model. We show that the mouse bladder expresses IFRD1 at homeostasis and its loss alters the global transcriptome of the bladder with significant accumulation of cellular organelles including multivesicular bodies with undigested cargo, lysosomes and mitochondria. We demonstrate that IFRD1 interacts with several mRNA-translation-regulating factors in human urothelial cells and that the urothelium of Ifrd1-/- mice reveal decreased global translation and enhanced endoplasmic reticulum (ER) stress response. Ifrd1-/- bladders have activation of the unfolded protein response (UPR) pathway, specifically the PERK arm, with a concomitant increase in oxidative stress and spontaneous exfoliation of urothelial cells. Further, we show that such increase in cell shedding is associated with a compensatory proliferation of the basal cells but impaired regeneration of superficial cells. Finally, we show that upon loss of IFRD1, mice display aberrant voiding behavior. Thus, we propose that IFRD1 is at the center of many crucial cellular pathways that work together to maintain urothelial homeostasis, highlighting its importance as a target for diagnosis and/or therapy in bladder conditions.
Collapse
Affiliation(s)
- Bisiayo E. Fashemi
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Amala K. Rougeau
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - Arnold M. Salazar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX
| | - Steven J. Bark
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX
| | - Rayvanth Chappidi
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jeffrey W. Brown
- John T. Milliken Department of Medicine, Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO
| | - Charles J. Cho
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - Jason C. Mills
- Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Indira U. Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX
- Department of Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Huffington Center of Aging, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
13
|
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
|
14
|
Peng M, Chu X, Peng Y, Li D, Zhang Z, Wang W, Zhou X, Xiao D, Yang X. Targeted therapies in bladder cancer: signaling pathways, applications, and challenges. MedComm (Beijing) 2023; 4:e455. [PMID: 38107059 PMCID: PMC10724512 DOI: 10.1002/mco2.455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023] Open
Abstract
Bladder cancer (BC) is one of the most prevalent malignancies in men. Understanding molecular characteristics via studying signaling pathways has made tremendous breakthroughs in BC therapies. Thus, targeted therapies including immune checkpoint inhibitors (ICIs), antibody-drug conjugates (ADCs), and tyrosine kinase inhibitor (TKI) have markedly improved advanced BC outcomes over the last few years. However, the considerable patients still progress after a period of treatment with current therapeutic regimens. Therefore, it is crucial to guide future drug development to improve BC survival, based on the molecular characteristics of BC and clinical outcomes of existing drugs. In this perspective, we summarize the applications and benefits of these targeted drugs and highlight our understanding of mechanisms of low response rates and immune escape of ICIs, ADCs toxicity, and TKI resistance. We also discuss potential solutions to these problems. In addition, we underscore the future drug development of targeting metabolic reprogramming and cancer stem cells (CSCs) with a deep understanding of their signaling pathways features. We expect that finding biomarkers, developing novo drugs and designing clinical trials with precisely selected patients and rationalized drugs will dramatically improve the quality of life and survival of patients with advanced BC.
Collapse
Affiliation(s)
- Mei Peng
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Xuetong Chu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Yan Peng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Duo Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Zhirong Zhang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Weifan Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Xiaochen Zhou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Di Xiao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| |
Collapse
|
15
|
Luo P, Zheng L, Zou J, Chen T, Zou J, Li W, Chen Q, Qian B. Insights into vitamin A in bladder cancer, lack of attention to gut microbiota? Front Immunol 2023; 14:1252616. [PMID: 37711628 PMCID: PMC10497765 DOI: 10.3389/fimmu.2023.1252616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Vitamin A has long been associated with bladder cancer, and many exogenous vitamin A supplements, vitamin A derivatives, and synthetic drugs have been investigated over the years. However, the effectiveness of these strategies in clinical practice has not met expectations, and they have not been widely adopted. Recent medical research on intestinal flora has revealed that bladder cancer patients exhibit reduced serum vitamin A levels and an imbalance of gut microbiota. In light of the close relationship between gut microbiota and vitamin A, one can speculate that a complex regulatory mechanism exists between the two in the development and occurrence of bladder cancer. As such, further exploration of their interaction in bladder cancer may help guide the use of vitamin A for preventive purposes. During the course of this review, attention is paid to the influence of intestinal microbiota on the vitamin A metabolism and the RA signaling pathway, as well as the mutual promotion relationships between them in the prevention of bladder cancer, In addition, it emphasizes the importance of intestinal microbiota for bladder cancer prevention and treatment.
Collapse
Affiliation(s)
- Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Liying Zheng
- Department of Graduate, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junrong Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Jun Zou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Wei Li
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Qi Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Biao Qian
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| |
Collapse
|
16
|
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
|
17
|
Uno W, Ofuji K, Wymeersch FJ, Takasato M. In vitro induction of prostate buds from murine urogenital epithelium in the absence of mesenchymal cells. Dev Biol 2023; 498:49-60. [PMID: 36963625 DOI: 10.1016/j.ydbio.2023.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/08/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
The prostate is a male reproductive gland which secretes prostatic fluid that enhances male fertility. During development and instigated by fetal testosterone, prostate cells arise caudal to the bladder at the urogenital sinus (UGS), when the urogenital mesenchyme (UGM) secretes signals to the urogenital epithelium (UGE). These initial mesenchymal signals induce prostate-specific gene expression in the UGE, after which epithelial progenitor cells form prostatic buds. Although many important factors for prostate development have been described using UGS organ cultures, those necessary and sufficient for prostate budding have not been clearly identified. This has been in part due to the difficulty to dissect the intricate signaling and feedback between epithelial and mesenchymal UGS cells. In this study, we separated the UGM from the UGE and tested candidate growth factors to show that when FGF10 is present, testosterone is not required for initiating prostate budding from the UGE. Moreover, in the presence of low levels of FGF10, canonical WNT signaling enhances the expression of several prostate progenitor markers in the UGE before budding of the prostate occurs. At the later budding stage, higher levels of FGF10 are required to increase budding and retinoic acid is indispensable for the upregulation of prostate-specific genes. Lastly, we show that under optimized conditions, female UGE can be instructed towards a prostatic fate, and in vitro generated prostate buds from male UGE can differentiate into a mature prostate epithelium after in vivo transplantation. Taken together, our results clarify the signals that can induce fetal prostate buds in the urogenital epithelium in the absence of the surrounding, instructive mesenchyme.
Collapse
Affiliation(s)
- Wataru Uno
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan; Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Kazuhiro Ofuji
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan
| | - Filip J Wymeersch
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan
| | - Minoru Takasato
- Laboratory for Human Organogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan; Laboratory of Molecular Cell Biology and Development, Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
18
|
Como CN, Cervantes C, Pawlikowski B, Siegenthaler J. Retinoic acid signaling in mouse retina endothelial cells is required for early angiogenic growth. Differentiation 2023; 130:16-27. [PMID: 36528974 PMCID: PMC10006372 DOI: 10.1016/j.diff.2022.12.002] [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: 05/09/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
The development of the retinal vasculature is essential to maintain health of the tissue, but the developmental mechanisms are not completely understood. The aim of this study was to investigate the cell-autonomous role of retinoic acid signaling in endothelial cells during retina vascular development. Using a temporal and cell-specific mouse model to disrupt retinoic acid signaling in endothelial cells in the postnatal retina (Pdgfbicre/+dnRAR403fl/fl mutants), we discovered that angiogenesis in the retina is significantly decreased with a reduction in retina vascularization, endothelial tip cell number and filipodia, and endothelial 'crowding' of stalk cells. Interestingly, by P15, the vasculature can overcome the early angiogenic defect and fully vascularized the retina. At P60, the vasculature is intact with no evidence of retina cell death or altered blood retinal barrier integrity. Further, we identified that the angiogenic defect seen in mutants at P6 correlates with decreased Vegfr3 expression in endothelial cells. Collectively, our work identified a previously unappreciated function for endothelial retinoic acid signaling in early retinal angiogenesis.
Collapse
Affiliation(s)
- Christina N Como
- University of Colorado, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Summer Research Training Program, Aurora, CO, 80045, USA
| | - Cesar Cervantes
- University of Colorado, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Summer Research Training Program, Aurora, CO, 80045, USA
| | - Brad Pawlikowski
- University of Colorado, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO, 80045, USA
| | - Julie Siegenthaler
- University of Colorado, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Neuroscience Graduate Program, Aurora, CO, 80045, USA; University of Colorado, Anschutz Medical Campus, Summer Research Training Program, Aurora, CO, 80045, USA.
| |
Collapse
|
19
|
Alì A, Leibowitz D, Bhatt N, Doubrovin M, Spina CS, Bates-Pappas GE, Taub RN, McKiernan JM, Mintz A, Molotkov A. Preliminary efficacy of [ 90Y]DOTA-biotin-avidin radiotherapy against non-muscle invasive bladder cancer. Eur J Nucl Med Mol Imaging 2023; 50:692-700. [PMID: 36350400 DOI: 10.1007/s00259-022-06027-w] [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: 04/14/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE Bladder cancer represents 3% of all new cancer diagnoses per year. We propose intravesical radionuclide therapy using the β-emitter 90Y linked to DOTA-biotin-avidin ([90Y]DBA) to deliver short-range radiation against non-muscle invasive bladder cancer (NMIBC). MATERIAL AND METHODS Image-guided biodistribution of intravesical DBA was investigated in an animal model by radiolabeling DBA with the 68Ga and dynamic microPET imaging following intravesical infusion of [68Ga]DBA for up to 4 h and post-necropsy γ-counting of organs. The antitumor activity of [90Y]DBA was investigated using an orthotopic MB49 murine bladder cancer model. Mice were injected with luciferase-expressing MB49 cells and treated via intravesical administration with 9.2 MBq of [90Y]DBA or unlabeled DBA 3 days after the tumor implantation. Bioluminescence imaging was conducted after tumor implantation to monitor the bladder tumor growth. In addition, we investigated the effects of [90Y]DBA radiation on urothelial histology with immunohistochemistry analysis of bladder morphology. RESULTS Our results demonstrated that DBA is contained in the bladder for up to 4 h after intravesical infusion. A single dose of [90Y]DBA radiation treatment significantly reduced growth of MB49 bladder carcinoma. Attaching 90Y-DOTA-biotin to avidin prevents its re-absorption into the blood and distribution throughout the rest of the body. Furthermore, immunohistochemistry demonstrated that [90Y]DBA radiation treatment did not cause short-term damage to urothelium at day 10, which appeared similar to the normal urothelium of healthy mice. CONCLUSION Our data demonstrates the potential of intravesical [90Y]DBA as a treatment for non-muscle invasive bladder cancer.
Collapse
Affiliation(s)
- Alessandra Alì
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Dev Leibowitz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Nikunj Bhatt
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Mikhail Doubrovin
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Catherine S Spina
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Gleneara E Bates-Pappas
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, 10021, USA
| | - Robert N Taub
- Department of Medicine (Retired), Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - James M McKiernan
- Department of Urology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Akiva Mintz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Andrei Molotkov
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| |
Collapse
|
20
|
Gundogdu G, Nguyen T, Hosseini Sharifi SH, Starek S, Costa K, Jones CE, Barham D, Gelman J, Clayman RV, Mauney JR. Evaluation of silk fibroin-based urinary conduits in a porcine model of urinary diversion. Front Bioeng Biotechnol 2023; 11:1100507. [PMID: 36726743 PMCID: PMC9885082 DOI: 10.3389/fbioe.2023.1100507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/03/2023] [Indexed: 01/17/2023] Open
Abstract
Background: The primary strategy for urinary diversion in radical cystectomy patients involves incorporation of autologous gastrointestinal conduits into the urinary tract which leads to deleterious consequences including chronic infections and metabolic abnormalities. This report investigates the efficacy of an acellular, tubular bi-layer silk fibroin (BLSF) graft to function as an alternative urinary conduit in a porcine model of urinary diversion. Materials and methods: Unilateral urinary diversion with stented BLSF conduits was executed in five adult female, Yucatan mini-swine over a 3 month period. Longitudinal imaging analyses including ultrasonography, retrograde ureteropyelography and video-endoscopy were carried out monthly. Histological, immunohistochemical (IHC), and histomorphometric assessments were performed on neoconduits at harvest. Results: All animals survived until scheduled euthanasia and displayed moderate hydronephrosis (Grades 1-3) in reconstructed collecting systems over the course of the study period. Stented BLSF constructs supported formation of vascularized, retroperitoneal tubes capable of facilitating external urinary drainage. By 3 months post-operative, neoconduits contained α-smooth muscle actin+ and SM22α+ smooth muscle as well as uroplakin 3A+ and pan-cytokeratin + urothelium. However, the degree of tissue regeneration in neotissues was significantly lower in comparison to ureteral controls as determined by histomorphometry. In addition, neoconduit stenting was necessary to prevent stomal occlusion. Conclusion: BLSF biomaterials represent emerging platforms for urinary conduit construction and may offer a functional replacement for conventional urinary diversion techniques following further optimization of mechanical properties and regenerative responses.
Collapse
Affiliation(s)
- Gokhan Gundogdu
- Department of Urology, University of California, Irvine, Orange, CA, United States
| | - Travis Nguyen
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | | | - Stephanie Starek
- Department of Urology, University of California, Irvine, Orange, CA, United States
| | - Kyle Costa
- Department of Urology, University of California, Irvine, Orange, CA, United States
| | - Clara E. Jones
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - David Barham
- Department of Urology, University of California, Irvine, Orange, CA, United States
| | - Joel Gelman
- Department of Urology, University of California, Irvine, Orange, CA, United States
| | - Ralph V. Clayman
- Department of Urology, University of California, Irvine, Orange, CA, United States
| | - Joshua R. Mauney
- Department of Urology, University of California, Irvine, Orange, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
21
|
Marchese PV, Mollica V, Tassinari E, De Biase D, Giunchi F, Marchetti A, Rosellini M, Fiorentino M, Massari F. Implications of TERT promoter mutations and telomerase activity in solid tumors with a focus on genitourinary cancers. Expert Rev Mol Diagn 2022; 22:997-1008. [PMID: 36503370 DOI: 10.1080/14737159.2022.2154148] [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] [Received: 10/06/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The reactivation of telomerase represents a key moment in the carcinogenesis process. Mutations in the central promoter region of the telomerase reverse transcriptase (TERT) gene cause telomerase reactivation in approximately 90% of solid tumors. In some of these, its prognostic and predictive role in response to treatments has already been demonstrated, in others (such as tumors of the genitourinary tract like urothelial carcinoma) data are controversial and the research is still ongoing. In the future, TERT promoter mutations and telomerase activity could have diagnostic, prognostic, and therapeutic applications in many types of cancer. AREAS COVERED We performed a review the literature with the aim of describing the current evidence on the prognostic and predictive role of TERT promoter mutations. In some tumor types, TERT promoter mutations have been associated with a worse prognosis and could have a potential value as biomarkers to guide therapeutic decisions. Mutations in TERT promoter seems to make the tumor particularly immunogenic and more responsive to immunotherapy, although data is controversial. EXPERT OPINION We described the role of TERT promoter mutations in solid tumors with a particular focus in genitourinary cancers, considering their frequency in this tract.
Collapse
Affiliation(s)
- Paola Valeria Marchese
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Via Albertoni - 15 40138, Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Via Albertoni - 15 40138, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Elisa Tassinari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Via Albertoni - 15 40138, Bologna, Italy
| | - Dario De Biase
- Department of Pharmacy and Biotechnology (Fabit), University of Bologna, 40138 Bologna, Italy
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Francesca Giunchi
- Pathology Unit, IRCCS Policlinico Sant'Orsola-Malpighi, University of Bologna, Bologna, Italy
| | - Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Via Albertoni - 15 40138, Bologna, Italy
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Via Albertoni - 15 40138, Bologna, Italy
| | | | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Via Albertoni - 15 40138, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
22
|
Toyoda T, Ogawa K. Early detection of urinary bladder carcinogens in rats by immunohistochemistry for γ-H2AX: a review from analyses of 100 chemicals. J Toxicol Pathol 2022; 35:283-298. [PMID: 36406171 PMCID: PMC9647216 DOI: 10.1293/tox.2022-0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/28/2022] [Indexed: 11/25/2022] Open
Abstract
In safety evaluations of chemicals, there is an urgent need to develop short-term methods to replace long-term carcinogenicity tests. We have reported that immunohistochemistry for γ-H2AX, a well-established biomarker of DNA damage, can detect bladder carcinogens at an early stage using histopathological specimens from 28-day repeated-dose oral toxicity studies in rats. Given the markedly low level of γ-H2AX formation in the bladder urothelium of untreated rats, an increase in γ-H2AX-positive cells following chemical exposure can be relatively easy to identify. Among the 100 compounds examined to date, bladder carcinogens can be detected with high sensitivity (33/39; 84.6%) and specificity (58/61; 95.1%). As expected, γ-H2AX formation levels tended to be high following exposure to genotoxic bladder carcinogens, whereas nongenotoxic bladder carcinogens also increased the number of γ-H2AX-positive cells, probably through secondary DNA damage associated with sustained proliferative stimulation. γ-H2AX formation in the bladder urothelium reflects species differences in susceptibility to bladder carcinogenesis between rats and mice and shows a clear dose-dependency associated with the intensity of tumor development as well as high reproducibility. Some of the bladder carcinogens that showed false-negative results in the evaluation of γ-H2AX alone could be detected by combined evaluation with immunostaining for bladder stem cell markers, including aldehyde dehydrogenase 1A1. This method may be useful for the early detection of bladder carcinogens, as it can be performed by simple addition of conventional immunostaining using formalin-fixed paraffin-embedded tissues from 28-day repeated-dose toxicity studies in rodents, which are commonly used in safety evaluations of chemical substances.
Collapse
Affiliation(s)
- Takeshi Toyoda
- Division of Pathology, National Institute of Health
Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan
| | - Kumiko Ogawa
- Division of Pathology, National Institute of Health
Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan
| |
Collapse
|
23
|
Ortega MA, Villiger RK, Harrison-Chau M, Lieu S, Tamashiro KK, Lee AJ, Fujimoto BA, Patwardhan GY, Kepler J, Fogelgren B. Exocyst inactivation in urothelial cells disrupts autophagy and activates non-canonical NF-κB signaling. Dis Model Mech 2022; 15:dmm049785. [PMID: 36004645 PMCID: PMC9586569 DOI: 10.1242/dmm.049785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 12/02/2022] Open
Abstract
Ureter obstruction is a highly prevalent event during embryonic development and is a major cause of pediatric kidney disease. We have previously reported that ureteric bud-specific ablation of the gene expressing the exocyst subunit EXOC5 in late murine gestation results in failure of urothelial stratification, cell death and complete ureter obstruction. However, the mechanistic connection between disrupted exocyst activity, urothelial cell death and subsequent ureter obstruction was unclear. Here, we report that inhibited urothelial stratification does not drive cell death during ureter development. Instead, we demonstrate that the exocyst plays a critical role in autophagy in urothelial cells, and that disruption of autophagy activates a urothelial NF-κB stress response. Impaired autophagy first provokes canonical NF-κB activity, which is progressively followed by increasing levels of non-canonical NF-κB activity and cell death if the stress remains unresolved. Furthermore, we demonstrate that ureter obstructions can be completely rescued in Exoc5 conditional knockout mice by administering a single dose of the pan-caspase inhibitor z-VAD-FMK at embryonic day 16.5 prior to urothelial cell death. Taken together, ablation of Exoc5 disrupts autophagic stress response and activates progressive NF-κB signaling, which promotes obstructive uropathy.
Collapse
Affiliation(s)
- Michael A. Ortega
- Center for Biomedical Research at The Queen's Medical Center, Honolulu, Hawaii 96813, USA
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
| | - Ross K. Villiger
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
| | - Malia Harrison-Chau
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
| | - Suzanna Lieu
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
| | - Kadee-Kalia Tamashiro
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
| | - Amanda J. Lee
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
- Math and Sciences Department, Kapiolani Community College, Honolulu, Hawaii 96816, USA
| | - Brent A. Fujimoto
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
| | - Geetika Y. Patwardhan
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
| | - Joshua Kepler
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
| | - Ben Fogelgren
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaiʿi at Manoa, Honolulu, Hawaii 96813, USA
| |
Collapse
|
24
|
In focus in HCB. Histochem Cell Biol 2022; 158:297-299. [PMID: 36114392 PMCID: PMC9483531 DOI: 10.1007/s00418-022-02149-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
25
|
Fink EE, Sona S, Tran U, Desprez PE, Bradley M, Qiu H, Eltemamy M, Wee A, Wolkov M, Nicolas M, Min B, Haber GP, Wessely O, Lee BH, Ting AH. Single-cell and spatial mapping Identify cell types and signaling Networks in the human ureter. Dev Cell 2022; 57:1899-1916.e6. [PMID: 35914526 PMCID: PMC9381170 DOI: 10.1016/j.devcel.2022.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/18/2022] [Accepted: 07/05/2022] [Indexed: 01/16/2023]
Abstract
Tissue engineering offers a promising treatment strategy for ureteral strictures, but its success requires an in-depth understanding of the architecture, cellular heterogeneity, and signaling pathways underlying tissue regeneration. Here, we define and spatially map cell populations within the human ureter using single-cell RNA sequencing, spatial gene expression, and immunofluorescence approaches. We focus on the stromal and urothelial cell populations to enumerate the distinct cell types composing the human ureter and infer potential cell-cell communication networks underpinning the bi-directional crosstalk between these compartments. Furthermore, we analyze and experimentally validate the importance of the sonic hedgehog (SHH) signaling pathway in adult progenitor cell maintenance. The SHH-expressing basal cells support organoid generation in vitro and accurately predict the differentiation trajectory from basal progenitor cells to terminally differentiated umbrella cells. Our results highlight the essential processes involved in adult ureter tissue homeostasis and provide a blueprint for guiding ureter tissue engineering.
Collapse
Affiliation(s)
- Emily E Fink
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Surbhi Sona
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Nutrition, Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Uyen Tran
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Pierre-Emmanuel Desprez
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Urology, CHU Lille, Claude Huriez Hospital, Université Lille, 59000 Lille, France
| | - Matthew Bradley
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Hong Qiu
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mohamed Eltemamy
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Alvin Wee
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Madison Wolkov
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Marlo Nicolas
- Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Georges-Pascal Haber
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Oliver Wessely
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Byron H Lee
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Angela H Ting
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
26
|
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: 4] [Impact Index Per Article: 1.3] [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
|
27
|
Fowler JC, Jones PH. Somatic Mutation: What Shapes the Mutational Landscape of Normal Epithelia? Cancer Discov 2022; 12:1642-1655. [PMID: 35397477 PMCID: PMC7613026 DOI: 10.1158/2159-8290.cd-22-0145] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022]
Abstract
Epithelial stem cells accumulate mutations throughout life. Some of these mutants increase competitive fitness and may form clones that colonize the stem cell niche and persist to acquire further genome alterations. After a transient expansion, mutant stem cells must revert to homeostatic behavior so normal tissue architecture is maintained. Some positively selected mutants may promote cancer development, whereas others inhibit carcinogenesis. Factors that shape the mutational landscape include wild-type and mutant stem cell dynamics, competition for the niche, and environmental exposures. Understanding these processes may give new insight into the basis of cancer risk and opportunities for cancer prevention. SIGNIFICANCE Recent advances in sequencing have found somatic mutations in all epithelial tissues studied to date. Here we review how the mutational landscape of normal epithelia is shaped by clonal competition within the stem cell niche combined with environmental exposures. Some of the selected mutant genes are oncogenic, whereas others may be inhibitory of transformation. Discoveries in this area leave many open questions, such as the definition of cancer driver genes, the mechanisms by which tissues constrain a high proportion of oncogenic mutant cells, and whether clonal fitness can be modulated to decrease cancer risk.
Collapse
Affiliation(s)
| | - Philip H Jones
- Wellcome Sanger Institute, Hinxton CB10 1SA, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| |
Collapse
|
28
|
Clayton DB, Tong CMC, Li B, Taylor AS, De S, Mason MD, Dudley AG, Davidoff O, Kobayashi H, Haase VH. Inhibition of hypoxia-inducible factor-prolyl hydroxylation protects from cyclophosphamide-induced bladder injury and urinary dysfunction. Am J Physiol Renal Physiol 2022; 323:F81-F91. [PMID: 35499237 PMCID: PMC9236868 DOI: 10.1152/ajprenal.00344.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Disruption of the blood-urine barrier can result in acute or chronic inflammatory bladder injury. Activation of the oxygen-regulated hypoxia-inducible factor (HIF) pathway has been shown to protect mucosal membranes by increasing the expression of cytoprotective genes and by suppressing inflammation. The activity of HIF is controlled by prolyl hydroxylase domain (PHD) dioxygenases, which have been exploited as therapeutic targets for the treatment of anemia of chronic kidney disease. Here, we established a mouse model of acute cyclophosphamide (CYP)-induced blood-urine barrier disruption associated with inflammation and severe urinary dysfunction to investigate the HIF-PHD axis in inflammatory bladder injury. We found that systemic administration of dimethyloxalylglycine or molidustat, two small-molecule inhibitors of HIF-prolyl hydroxylases, profoundly mitigated CYP-induced bladder injury and inflammation as assessed by morphological analysis of transmural edema and urothelial integrity and by measuring tissue cytokine expression. Void spot analysis to examine bladder function quantitatively demonstrated that HIF-prolyl hydroxylase inhibitor administration normalized micturition patterns and protected against CYP-induced alteration of urinary frequency and micturition patterns. Our study highlights the therapeutic potential of HIF-activating small-molecule compounds for the prevention or therapy of bladder injury and urinary dysfunction due to blood-urine barrier disruption.NEW & NOTEWORTHY Disruption of the blood-urine barrier can result in acute or chronic inflammatory bladder injury. Here, we demonstrate that pharmacological inhibition of hypoxia-inducible factor (HIF)-prolyl hydroxylation prevented bladder injury and protected from urinary dysfunction in a mouse model of cyclophosphamide-induced disruption of the blood-urine barrier. Our study highlights a potential role for HIF-activating small-molecule compounds in the prevention or therapy of bladder injury and urinary dysfunction and provides a rationale for future clinical studies.
Collapse
Affiliation(s)
- Douglass B Clayton
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ching Man Carmen Tong
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Belinda Li
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Abby S Taylor
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shuvro De
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Matthew D Mason
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anne G Dudley
- Division of Pediatric Urology, Department of Urology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Olena Davidoff
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Hanako Kobayashi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Volker H Haase
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
29
|
Jafari NV, Rohn JL. The urothelium: a multi-faceted barrier against a harsh environment. Mucosal Immunol 2022; 15:1127-1142. [PMID: 36180582 PMCID: PMC9705259 DOI: 10.1038/s41385-022-00565-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/18/2022] [Accepted: 08/28/2022] [Indexed: 02/04/2023]
Abstract
All mucosal surfaces must deal with the challenge of exposure to the outside world. The urothelium is a highly specialized layer of stratified epithelial cells lining the inner surface of the urinary bladder, a gruelling environment involving significant stretch forces, osmotic and hydrostatic pressures, toxic substances, and microbial invasion. The urinary bladder plays an important barrier role and allows the accommodation and expulsion of large volumes of urine without permitting urine components to diffuse across. The urothelium is made up of three cell types, basal, intermediate, and umbrella cells, whose specialized functions aid in the bladder's mission. In this review, we summarize the recent insights into urothelial structure, function, development, regeneration, and in particular the role of umbrella cells in barrier formation and maintenance. We briefly review diseases which involve the bladder and discuss current human urothelial in vitro models as a complement to traditional animal studies.
Collapse
Affiliation(s)
- Nazila V Jafari
- Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, London, UK
| | - Jennifer L Rohn
- Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, London, UK.
| |
Collapse
|
30
|
S100A1 expression characterizes terminally differentiated superficial cells in the urothelium of the murine bladder and ureter. Histochem Cell Biol 2022; 158:389-399. [PMID: 35648290 PMCID: PMC9512885 DOI: 10.1007/s00418-022-02120-1] [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] [Accepted: 05/18/2022] [Indexed: 11/04/2022]
Abstract
The urothelium is a stratified epithelium that lines the inner surface of the components of the urinary drainage system. It is composed of a layer of basal cells, one or several layers of intermediate cells, and a layer of large luminal superficial or umbrella cells. In the mouse, only a small set of markers is available that allows easy molecular distinction of these urothelial cell types. Here, we analyzed expression of S100A1, a member of the S100 family of calcium-binding proteins, in the urothelium of the two major organs of the murine urinary tract, the ureter and the bladder. Using RNA in situ hybridization analysis, we found exclusive expression of S100a1 mRNA in luminal cells of the ureter from embryonic day (E)17.5 onwards and of the bladder from E15.5 to adulthood. Immunofluorescence analysis showed that expression of S100A1 protein is confined to terminally differentiated superficial cells of both the ureter and bladder where it localized to the nucleus and cytoplasm. We conclude that S100A1 is a suitable marker for mature superficial cells in the urothelial lining of the drainage system of the developing and mature mouse.
Collapse
|
31
|
Lokeshwar SD, Lopez M, Sarcan S, Aguilar K, Morera DS, Shaheen DM, Lokeshwar BL, Lokeshwar VB. Molecular Oncology of Bladder Cancer from Inception to Modern Perspective. Cancers (Basel) 2022; 14:cancers14112578. [PMID: 35681556 PMCID: PMC9179261 DOI: 10.3390/cancers14112578] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/05/2023] Open
Abstract
Within the last forty years, seminal contributions have been made in the areas of bladder cancer (BC) biology, driver genes, molecular profiling, biomarkers, and therapeutic targets for improving personalized patient care. This overview includes seminal discoveries and advances in the molecular oncology of BC. Starting with the concept of divergent molecular pathways for the development of low- and high-grade bladder tumors, field cancerization versus clonality of bladder tumors, cancer driver genes/mutations, genetic polymorphisms, and bacillus Calmette-Guérin (BCG) as an early form of immunotherapy are some of the conceptual contributions towards improving patient care. Although beginning with a promise of predicting prognosis and individualizing treatments, "-omic" approaches and molecular subtypes have revealed the importance of BC stem cells, lineage plasticity, and intra-tumor heterogeneity as the next frontiers for realizing individualized patient care. Along with urine as the optimal non-invasive liquid biopsy, BC is at the forefront of the biomarker field. If the goal is to reduce the number of cystoscopies but not to replace them for monitoring recurrence and asymptomatic microscopic hematuria, a BC marker may reach clinical acceptance. As advances in the molecular oncology of BC continue, the next twenty-five years should significantly advance personalized care for BC patients.
Collapse
Affiliation(s)
- Soum D. Lokeshwar
- Department of Urology, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Maite Lopez
- Departments of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd., Augusta, GA 30912, USA; (M.L.); (S.S.); (K.A.); (D.S.M.)
| | - Semih Sarcan
- Departments of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd., Augusta, GA 30912, USA; (M.L.); (S.S.); (K.A.); (D.S.M.)
- Department of Urology, University Hospital Schleswig-Holstein, Campus Lübeck, 23562 Lübeck, Germany
| | - Karina Aguilar
- Departments of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd., Augusta, GA 30912, USA; (M.L.); (S.S.); (K.A.); (D.S.M.)
| | - Daley S. Morera
- Departments of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd., Augusta, GA 30912, USA; (M.L.); (S.S.); (K.A.); (D.S.M.)
| | - Devin M. Shaheen
- Yale School of Nursing, Yale University, New Haven, CT 06520, USA;
| | - Bal L. Lokeshwar
- Georgia Cancer Center, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd., Augusta, GA 30912, USA
- Research Service, Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Correspondence: (B.L.L.); (V.B.L.)
| | - Vinata B. Lokeshwar
- Departments of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1410 Laney Walker Blvd., Augusta, GA 30912, USA; (M.L.); (S.S.); (K.A.); (D.S.M.)
- Correspondence: (B.L.L.); (V.B.L.)
| |
Collapse
|
32
|
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: 8] [Impact Index Per Article: 2.7] [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
|
33
|
Guo C, Zhang Y, Tan R, Tang Z, Lam CM, Ye X, Wang Z, Li X. Arid1a regulates bladder urothelium formation and maintenance. Dev Biol 2022; 485:61-69. [PMID: 35283102 PMCID: PMC10276770 DOI: 10.1016/j.ydbio.2022.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/22/2021] [Accepted: 02/19/2022] [Indexed: 12/12/2022]
Abstract
Epigenetic regulation of gene expression plays a central role in bladder urothelium development and maintenance. ATPase-dependent chromatin remodeling is a major epigenetic regulatory mechanism, but its role in the bladder has not been explored. Here, we show the functions of Arid1a, the largest subunit of the SWI/SNF or BAF chromatin remodeling ATPase complex, in embryonic and adult bladder urothelium. Knockout of Arid1a in urothelial progenitor cells significantly increases cell proliferation during bladder development. Deletion of Arid1a causes ectopic cell proliferation in the terminally differentiated superficial cells in adult mice. Consistently, gene-set enrichment analysis of differentially expressed genes demonstrates that the cell cycle-related pathways are significantly enriched in Arid1a knockouts. Gene-set of the polycomb repression complex 2 (PRC2) pathway is also enriched, suggesting that Arid1a antagonizes the PRC2-dependent epigenetic gene silencing program in the bladder. During acute cyclophosphamide-induced bladder injury, Arid1a knockouts develop hyperproliferative and hyperinflammatory phenotypes and exhibit a severe loss of urothelial cells. A Hallmark gene-set of the oxidative phosphorylation pathway is significantly reduced in Aria1a mutants before injury and is unexpectedly enriched during injury response. Together, this study uncovers functions of Arid1a in both bladder progenitor cells and the mature urothelium, suggesting its critical roles in urothelial development and regeneration.
Collapse
Affiliation(s)
- Chunming Guo
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA, 90048, USA
| | - Yingsheng Zhang
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA, 90048, USA
| | - Ruirong Tan
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA, 90048, USA
| | - Zonghao Tang
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA, 90048, USA
| | - Christa M Lam
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA, 90048, USA
| | - Xing Ye
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA, 90048, USA
| | - Zhong Wang
- Department of Cardiac Surgery Cardiovascular Research Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xue Li
- Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Davis 3089, Los Angeles, CA, 90048, USA.
| |
Collapse
|
34
|
Kamasako T, Kaga K, Inoue KI, Hariyama M, Yamanishi T. Supervised machine learning algorithm identified KRT20, BATF and TP63 as biologically relevant biomarkers for bladder biopsy specimens from interstitial cystitis/bladder pain syndrome patients. Int J Urol 2022; 29:406-412. [PMID: 35102612 DOI: 10.1111/iju.14795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVES This study was carried out to identify biomarkers that distinguish Hunner-type interstitial cystitis from non-Hunner-type interstitial cystitis patients. METHODS Total ribonucleic acid was purified from 212 punch biopsy specimens of 89 individuals who were diagnosed as interstitial cystitis/bladder pain syndrome. To examine the expression profile of patients' bladder specimens, 68 urothelial master transcription factors and nine known markers (E-cadherin, cytokeratins, uroplakins and sonic hedgehog) were selected. To classify the biopsy samples, principal component analysis was carried out. A decision tree algorithm was adopted to identify critical determinants, in which 102 and 116 bladder specimens were used for learning and validation, respectively. RESULTS Principal component analysis segregated tissues from Hunner-type and non-Hunner-type interstitial cystitis specimens in principal component axes 2 and 4. Principal components 2 and 4 contained urothelial stem/progenitor transcription factors and cytokeratins, respectively. A decision tree identified KRT20, BATF and TP63 to classify non-Hunner-type and Hunner-type interstitial cystitis specimens. KRT20 was lower in tissues from Hunner-type compared with non-Hunner-type interstitial cystitis specimens (P < 0.001). TP63 was lower in Hunner's lesions compared with adjacent mucosa from Hunner-type interstitial cystitis patients (P < 0.001). Blinded validation using additional biopsy specimens verified that the decision tree showed fairly precise concordance with cystoscopic diagnosis. CONCLUSION KRT20, BATF and TP63 were identified as biologically relevant biomarkers to classify tissues from interstitial cystitis/bladder pain syndrome specimens. The biologically explainable determinants could contribute to defining the elusive interstitial cystitis/bladder pain syndrome pathogenesis.
Collapse
Affiliation(s)
- Tomohiko Kamasako
- Department of Urology, Continence Center, Dokkyo Medical University, Tochigi, Japan
| | - Kanya Kaga
- Department of Urology, Continence Center, Dokkyo Medical University, Tochigi, Japan
| | - Ken-Ichi Inoue
- Comprehensive Research Facilities for Advanced Medical Science, Research Center for Advanced Medical Science, Dokkyo Medical University, Tochigi, Japan
| | - Masanori Hariyama
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Tomonori Yamanishi
- Department of Urology, Continence Center, Dokkyo Medical University, Tochigi, Japan
- Department of Urology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
35
|
Male Lower Urinary Tract Dysfunction: An Underrepresented Endpoint in Toxicology Research. TOXICS 2022; 10:toxics10020089. [PMID: 35202275 PMCID: PMC8880407 DOI: 10.3390/toxics10020089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023]
Abstract
Lower urinary tract dysfunction (LUTD) is nearly ubiquitous in men of advancing age and exerts substantial physical, mental, social, and financial costs to society. While a large body of research is focused on the molecular, genetic, and epigenetic underpinnings of the disease, little research has been dedicated to the influence of environmental chemicals on disease initiation, progression, or severity. Despite a few recent studies indicating a potential developmental origin of male LUTD linked to chemical exposures in the womb, it remains a grossly understudied endpoint in toxicology research. Therefore, we direct this review to toxicologists who are considering male LUTD as a new aspect of chemical toxicity studies. We focus on the LUTD disease process in men, as well as in the male mouse as a leading research model. To introduce the disease process, we describe the physiology of the male lower urinary tract and the cellular composition of lower urinary tract tissues. We discuss known and suspected mechanisms of male LUTD and examples of environmental chemicals acting through these mechanisms to contribute to LUTD. We also describe mouse models of LUTD and endpoints to diagnose, characterize, and quantify LUTD in men and mice.
Collapse
|
36
|
Meuser M, Deuper L, Rudat C, Aydoğdu N, Thiesler H, Zarnovican P, Hildebrandt H, Trowe MO, Kispert A. FGFR2 signaling enhances the SHH-BMP4 signaling axis in early ureter development. Development 2022; 149:273983. [DOI: 10.1242/dev.200021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The patterned array of basal, intermediate and superficial cells in the urothelium of the mature ureter arises from uncommitted epithelial progenitors of the distal ureteric bud. Urothelial development requires signaling input from surrounding mesenchymal cells, which, in turn, depend on cues from the epithelial primordium to form a layered fibro-muscular wall. Here, we have identified FGFR2 as a crucial component in this reciprocal signaling crosstalk in the murine ureter. Loss of Fgfr2 in the ureteric epithelium led to reduced proliferation, stratification, intermediate and basal cell differentiation in this tissue, and affected cell survival and smooth muscle cell differentiation in the surrounding mesenchyme. Loss of Fgfr2 impacted negatively on epithelial expression of Shh and its mesenchymal effector gene Bmp4. Activation of SHH or BMP4 signaling largely rescued the cellular defects of mutant ureters in explant cultures. Conversely, inhibition of SHH or BMP signaling in wild-type ureters recapitulated the mutant phenotype in a dose-dependent manner. Our study suggests that FGF signals from the mesenchyme enhance, via epithelial FGFR2, the SHH-BMP4 signaling axis to drive urothelial and mesenchymal development in the early ureter.
Collapse
Affiliation(s)
- Max Meuser
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Lena Deuper
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Carsten Rudat
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Nurullah Aydoğdu
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Patricia Zarnovican
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| |
Collapse
|
37
|
Chen Z, Liu L, Chen Y, Liu M, Peng Xiang A, Deng C, Jiang MH. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:659-673. [PMID: 35648087 PMCID: PMC9216508 DOI: 10.1093/stcltm/szac025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/27/2022] [Indexed: 11/14/2022] Open
Abstract
Interstitial cystitis (IC) is a bladder syndrome of unclear etiology with no generally accepted treatment. Growing evidence suggest that periostin (POSTN) is an important homeostatic component in the tissue repair and regeneration in adulthood, but its function in urinary bladder regeneration is still unknown. Here we investigate whether POSTN is involved in bladder tissue repair in a cyclophosphamide (CYP)-induced interstitial cystitis model. POSTN is primarily expressed in bladder stroma (detrusor smooth muscle and lamina propria) and upregulated in response to CYP-induced injury. POSTN deficiency resulted in more severe hematuria, aggravated edema of the bladder, and delayed umbrella cell recovery. Besides, less proliferative urothelial cells (labeled by pHH3, Ki67, and EdU) and lower expression of Krt14 (a urothelial stem cell marker) were detected in POSTN−/− mice post CYP exposure, indicating a limited urothelial regeneration. Further investigations revealed that POSTN could induce Wnt4 upregulation and activate AKT signaling, which together activates β-catenin signaling to drive urothelial stem cell proliferation. In addition, POSTN can promote resident macrophage proliferation and polarization to a pro-regenerative (M2) phenotype, which favors urothelial regeneration. Furthermore, we generated injectable P-GelMA granular hydrogel as a biomaterial carrier to deliver recombinant POSTN into the bladder, which could increase urothelial stem cells number, decrease umbrella cells exfoliation, and hence alleviate hematuria in a CYP-induced interstitial cystitis model. In summary, our findings identify a pivotal role of POSTN in bladder urothelial regeneration and suggest that intravesical biomaterials-assisted POSTN delivery may be an efficacious treatment for interstitial cystitis.
Collapse
Affiliation(s)
| | | | | | - Minjie Liu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Andy Peng Xiang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children’s Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Chunhua Deng
- Chunhua Deng, Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, People’s Republic of China. Tel: +86 20 87335633; Fax: +86 20 87332200;
| | - Mei Hua Jiang
- Corresponding authors: Mei Hua Jiang, Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, People’s Republic of China. Tel: +86 20 87330639; Fax: +86 20 87330709;
| |
Collapse
|
38
|
Alterations of Chromatin Regulators in the Pathogenesis of Urinary Bladder Urothelial Carcinoma. Cancers (Basel) 2021; 13:cancers13236040. [PMID: 34885146 PMCID: PMC8656749 DOI: 10.3390/cancers13236040] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Urinary bladder cancer is one of the ten major cancers worldwide, with higher incidences in males, in smokers, and in highly industrialized countries. New therapies beyond cytotoxic chemotherapy are urgently needed to improve treatment of these tumors. A better understanding of the mechanisms underlying their development may help in this regard. Recently, it was discovered that a group of proteins regulating the state of chromatin and thus gene expression is exceptionally and frequently affected by gene mutations in bladder cancers. Altered function of these mutated chromatin regulators must therefore be fundamental in their development, but how and why is poorly understood. Here we review the current knowledge on changes in chromatin regulators and discuss their possible consequences for bladder cancer development and options for new therapies. Abstract Urothelial carcinoma (UC) is the most frequent histological type of cancer in the urinary bladder. Genomic changes in UC activate MAPK and PI3K/AKT signal transduction pathways, which increase cell proliferation and survival, interfere with cell cycle and checkpoint control, and prevent senescence. A more recently discovered additional category of genetic changes in UC affects chromatin regulators, including histone-modifying enzymes (KMT2C, KMT2D, KDM6A, EZH2), transcription cofactors (CREBBP, EP300), and components of the chromatin remodeling complex SWI/SNF (ARID1A, SMARCA4). It is not yet well understood how these changes contribute to the development and progression of UC. Therefore, we review here the emerging knowledge on genomic and gene expression alterations of chromatin regulators and their consequences for cell differentiation, cellular plasticity, and clonal expansion during UC pathogenesis. Our analysis identifies additional relevant chromatin regulators and suggests a model for urothelial carcinogenesis as a basis for further mechanistic studies and targeted therapy development.
Collapse
|
39
|
Galdadas I, Bonis V, Vgenopoulou P, Papadourakis M, Kakoulidis P, Stergiou G, Cournia Z, Klinakis A. The effect of S427F mutation on RXRα activity depends on its dimeric partner. Chem Sci 2021; 12:14700-14710. [PMID: 34820085 PMCID: PMC8597827 DOI: 10.1039/d1sc04465f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/18/2021] [Indexed: 12/24/2022] Open
Abstract
RXRs are nuclear receptors acting as transcription regulators that control key cellular processes in all tissues. All type II nuclear receptors require RXRs for transcriptional activity by forming heterodimeric complexes. Recent whole-exome sequencing studies have identified the RXRα S427F hotspot mutation in 5% of the bladder cancer patients, which is always located at the interface of RXRα with its obligatory dimerization partners. Here, we show that mutation of S427 deregulates transcriptional activity of RXRα dimers, albeit with diverse allosteric mechanisms of action depending on its dimeric partner. S427F acts by allosteric mechanisms, which range from inducing the collapse of the binding pocket to allosteric stabilization of active co-activator competent RXRα states. Unexpectedly, RXR S427F heterodimerization leads to either loss- or gain-of-function complexes, in both cases likely compromising its tumor suppressor activity. This is the first report of a cancer-associated single amino acid substitution that affects the function of the mutant protein variably depending on its dimerization partner.
Collapse
Affiliation(s)
| | - Vangelis Bonis
- Biomedical Research Foundation Academy of Athens Athens Greece
| | | | | | - Panos Kakoulidis
- Biomedical Research Foundation Academy of Athens Athens Greece
- Data Science and Information Technologies, Department of Informatics and Telecommunication, National and Kapodistrian University of Athens Athens Greece
| | - Georgia Stergiou
- Biomedical Research Foundation Academy of Athens Athens Greece
- Data Science and Information Technologies, Department of Informatics and Telecommunication, National and Kapodistrian University of Athens Athens Greece
| | - Zoe Cournia
- Biomedical Research Foundation Academy of Athens Athens Greece
- Data Science and Information Technologies, Department of Informatics and Telecommunication, National and Kapodistrian University of Athens Athens Greece
| | | |
Collapse
|
40
|
Cwiek A, Suzuki M, deRonde K, Conaway M, Bennett KM, El Dahr S, Reidy KJ, Charlton JR. Premature differentiation of nephron progenitor cell and dysregulation of gene pathways critical to kidney development in a model of preterm birth. Sci Rep 2021; 11:21667. [PMID: 34737344 PMCID: PMC8569166 DOI: 10.1038/s41598-021-00489-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 10/05/2021] [Indexed: 12/31/2022] Open
Abstract
Preterm birth is a leading cause of neonatal morbidity. Survivors have a greater risk for kidney dysfunction and hypertension. Little is known about the molecular changes that occur in the kidney of individuals born preterm. Here, we demonstrate that mice delivered two days prior to full term gestation undergo premature cessation of nephrogenesis, resulting in a lower glomerular density. Kidneys from preterm and term groups exhibited differences in gene expression profiles at 20- and 27-days post-conception, including significant differences in the expression of fat-soluble vitamin-related genes. Kidneys of the preterm mice exhibited decreased proportions of endothelial cells and a lower expression of genes promoting angiogenesis compared to the term group. Kidneys from the preterm mice also had altered nephron progenitor subpopulations, early Six2 depletion, and altered Jag1 expression in the nephrogenic zone, consistent with premature differentiation of nephron progenitor cells. In conclusion, preterm birth alone was sufficient to shorten the duration of nephrogenesis and cause premature differentiation of nephron progenitor cells. These candidate genes and pathways may provide targets to improve kidney health in preterm infants.
Collapse
Affiliation(s)
- Aleksandra Cwiek
- Division of Nephrology, Department of Pediatrics, University of Virginia, Box 800386, Charlottesville, VA, 22903, USA
- Cell & Developmental Biology Graduate Program, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Masako Suzuki
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Kimberly deRonde
- Division of Nephrology, Department of Pediatrics, University of Virginia, Box 800386, Charlottesville, VA, 22903, USA
| | - Mark Conaway
- University of Virginia Health System, Charlottesville, VA, USA
- Division of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Kevin M Bennett
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Samir El Dahr
- Department of Pediatrics, Tulane University School of Medicine and Children's Hospital of New Orleans, New Orleans, LA, USA
| | - Kimberly J Reidy
- Division of Nephrology, Department of Pediatrics, Children's Hospital at Montefiore, New York, NY, USA
| | - Jennifer R Charlton
- Division of Nephrology, Department of Pediatrics, University of Virginia, Box 800386, Charlottesville, VA, 22903, USA.
| |
Collapse
|
41
|
Tate T, Xiang T, Wobker SE, Zhou M, Chen X, Kim H, Batourina E, Lin CS, Kim WY, Lu C, Mckiernan JM, Mendelsohn CL. Pparg signaling controls bladder cancer subtype and immune exclusion. Nat Commun 2021; 12:6160. [PMID: 34697317 PMCID: PMC8545976 DOI: 10.1038/s41467-021-26421-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 09/27/2021] [Indexed: 12/15/2022] Open
Abstract
Pparg, a nuclear receptor, is downregulated in basal subtype bladder cancers that tend to be muscle invasive and amplified in luminal subtype bladder cancers that tend to be non-muscle invasive. Bladder cancers derive from the urothelium, one of the most quiescent epithelia in the body, which is composed of basal, intermediate, and superficial cells. We find that expression of an activated form of Pparg (VP16;Pparg) in basal progenitors induces formation of superficial cells in situ, that exit the cell cycle, and do not form tumors. Expression in basal progenitors that have been activated by mild injury however, results in luminal tumor formation. We find that these tumors are immune deserted, which may be linked to down-regulation of Nf-kb, a Pparg target. Interestingly, some luminal tumors begin to shift to basal subtype tumors with time, down-regulating Pparg and other luminal markers. Our findings have important implications for treatment and diagnosis of bladder cancer. PPARg is differentially expressed in bladder cancer subtypes. Here, the authors show in mice that when an activated form of PPARg is expressed in basal bladder cells tumours do not form, however in the presence of injury the basal cells differentiate into luminal cells.
Collapse
Affiliation(s)
- Tiffany Tate
- Department of Urology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Tina Xiang
- Department of Urology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sarah E Wobker
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mi Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiao Chen
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Hyunwoo Kim
- Department of Urology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Ekatherina Batourina
- Department of Urology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Chyuan-Sheng Lin
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Transgenic Mouse Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chao Lu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - James M Mckiernan
- Department of Urology, Columbia University Irving Medical Center, New York, NY, 10032, USA.,Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.,New York-Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Cathy Lee Mendelsohn
- Department of Urology, Columbia University Irving Medical Center, New York, NY, 10032, USA. .,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA. .,Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, 10032, USA. .,Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY, 10032, USA. .,Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
42
|
Park S, Rong L, Owczarek TB, Bernardo MD, Shoulson RL, Chua CW, Kim JY, Lankarani A, Chakrapani P, Syed T, McKiernan JM, Solit DB, Shen MM, Al-Ahmadie HA, Abate-Shen C. Novel Mouse Models of Bladder Cancer Identify a Prognostic Signature Associated with Risk of Disease Progression. Cancer Res 2021; 81:5161-5175. [PMID: 34470779 PMCID: PMC8609963 DOI: 10.1158/0008-5472.can-21-1254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/11/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022]
Abstract
To study the progression of bladder cancer from non-muscle-invasive to muscle-invasive disease, we have developed a novel toolkit that uses complementary approaches to achieve gene recombination in specific cell populations in the bladder urothelium in vivo, thereby allowing us to generate a new series of genetically engineered mouse models (GEMM) of bladder cancer. One method is based on the delivery of adenoviruses that express Cre recombinase in selected cell types in the urothelium, and a second uses transgenic drivers in which activation of inducible Cre alleles can be limited to the bladder urothelium by intravesicular delivery of tamoxifen. Using both approaches, targeted deletion of the Pten and p53 tumor suppressor genes specifically in basal urothelial cells gave rise to muscle-invasive bladder tumors. Furthermore, preinvasive lesions arising in basal cells displayed upregulation of molecular pathways related to bladder tumorigenesis, including proinflammatory pathways. Cross-species analyses comparing a mouse gene signature of early bladder cancer with a human signature of bladder cancer progression identified a conserved 28-gene signature of early bladder cancer that is associated with poor prognosis for human bladder cancer and that outperforms comparable gene signatures. These findings demonstrate the relevance of these GEMMs for studying the biology of human bladder cancer and introduce a prognostic gene signature that may help to stratify patients at risk for progression to potentially lethal muscle-invasive disease. SIGNIFICANCE: Analyses of bladder cancer progression in a new series of genetically engineered mouse models has identified a gene signature of poor prognosis in human bladder cancer.
Collapse
Affiliation(s)
- Soonbum Park
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York
| | - Lijie Rong
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York
| | - Tomasz B Owczarek
- Department of Urology, Columbia University Irving Medical Center, New York, New York
| | - Matteo Di Bernardo
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York
| | - Rivka L Shoulson
- Institute of Comparative Medicine, Columbia University, New York, New York
| | - Chee-Wai Chua
- Department of Urology, Columbia University Irving Medical Center, New York, New York
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, New York
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
| | - Jaime Y Kim
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York
| | - Amir Lankarani
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York
| | - Prithi Chakrapani
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York
| | - Talal Syed
- Department of Urology, Columbia University Irving Medical Center, New York, New York
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, New York
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
- Department of Biological Sciences, Columbia University, New York, New York
| | - James M McKiernan
- Department of Urology, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - David B Solit
- Departments of Human Oncology and Pathogenesis and Medicine, Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York
- Weill Medical College, Cornell University, New York, New York
| | - Michael M Shen
- Department of Urology, Columbia University Irving Medical Center, New York, New York
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, New York
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Hikmat A Al-Ahmadie
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Cory Abate-Shen
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York.
- Department of Urology, Columbia University Irving Medical Center, New York, New York
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
43
|
Little MH, Howden SE, Lawlor KT, Vanslambrouck JM. Determining lineage relationships in kidney development and disease. Nat Rev Nephrol 2021; 18:8-21. [PMID: 34594045 DOI: 10.1038/s41581-021-00485-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 12/17/2022]
Abstract
The lineage relationships of cells provide information about the origins of component cell types during development and repair as well as the source of aberrant cells during disease. Genetic approaches to lineage tracing applied in the mouse have revealed much about how the mammalian kidney forms, including the identification of key progenitors for the nephrons and stromal compartments. Inducible Cre systems have also facilitated lineage tracing studies in the postnatal animal that illustrate the changes in cellular fate that can occur during kidney injury. With the advent of single-cell transcriptional profiling and trajectory analyses, predictions of cellular relationships across development are now being made in model systems, such as the mouse, as well as in human fetal kidney. Importantly, these approaches provide predictions of lineage relationships rather than definitive evidence. Although genetic approaches to the study of lineage have not previously been possible in a human setting, the application of CRISPR-Cas9 gene editing of pluripotent stem cells is beginning to teach us about human lineage relationships.
Collapse
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Parkville, VIC, Australia. .,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia. .,Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC, Australia.
| | - Sara E Howden
- Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Kynan T Lawlor
- Murdoch Children's Research Institute, Parkville, VIC, Australia
| | | |
Collapse
|
44
|
Yin Y, Haller ME, Chadchan SB, Kommagani R, Ma L. Signaling through retinoic acid receptors is essential for mammalian uterine receptivity and decidualization. JCI Insight 2021; 6:e150254. [PMID: 34292881 PMCID: PMC8492326 DOI: 10.1172/jci.insight.150254] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
Retinoic acid (RA) signaling has long been speculated to regulate embryo implantation, because many enzymes and proteins responsible for maintaining RA homeostasis and transducing RA signals are tightly regulated in the endometrium during this critical period. However, due to a lack of genetic data, it was unclear whether RA signaling is truly required for implantation and which specific RA signaling cascades are at play. Herein we utilize a genetic murine model that expresses a dominant-negative form of RA receptor (RAR) specifically in female reproductive organs to show that functional RA signaling is fundamental to female fertility, particularly implantation and decidualization. Reduction in RA signaling activity severely affects the ability of the uterus to achieve receptive status and decidualize, partially through dampening follistatin expression and downstream activin B/bone morphogenetic protein 2 signaling. To confirm translational relevance of these findings to humans, human endometrial stromal cells (hESCs) were treated with a pan-RAR antagonist to show that in vitro decidualization is impaired. RNA interference perturbation of individual RAR transcripts in hESCs revealed that RARα in particular was essential for proper decidualization. These data provide direct functional evidence that uterine RAR-mediated RA signaling was crucial for mammalian embryo implantation, and its disruption led to failure of uterine receptivity and decidualization, resulting in severely compromised fertility.
Collapse
Affiliation(s)
- Yan Yin
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, United States of America
| | - Meade E Haller
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, United States of America
| | - Sangappa B Chadchan
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, United States of America
| | - Ramakrishna Kommagani
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, United States of America
| | - Liang Ma
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, United States of America
| |
Collapse
|
45
|
Armfield BA, Cohn MJ. Single cell transcriptomic analysis of external genitalia reveals complex and sexually dimorphic cell populations in the early genital tubercle. Dev Biol 2021; 477:145-154. [PMID: 34033822 DOI: 10.1016/j.ydbio.2021.05.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 11/27/2022]
Abstract
External genital organs are among the most recognizable sexually dimorphic characters. The penis and clitoris develop from the embryonic genital tubercle, an outgrowth at the anterior margin of the cloaca that undergoes an extensive period of development in male and female embryos prior to the onset of sexual differentiation. In mice, differentiation into the penis and clitoris begins around embryonic day (E)15.5. Current knowledge of cell types that comprise the genital tubercle is limited to a few studies that have fate mapped derivatives of endoderm, mesoderm, and ectoderm. Here we use single cell transcriptomics to characterize the cell populations in the genital tubercles of male and female mouse embryos at E14.5, approximately 24 h before the onset of sexual differentiation, and we present the first comprehensive atlas of single-cell gene expression during external genital development. Clustering analyses and annotation using marker genes shows 19 distinct cell populations in E14.5 genital tubercles. Mapping of cell clusters to anatomical locations using in situ gene expression patterns revealed granularity of cellular specializations and positional identities. Although E14.5 precedes sexually dimorphic morphogenesis of the genital tubercle, comparative analysis of males and females identified sexual dimorphisms at the single cell level, including male-specific cell clusters with transcriptional signatures of smooth muscle and bone progenitors, both of which are known to be sexually dimorphic in adult genitalia, as well as immune cells. These results provide a new resource for classification of external genital cell types based on gene expression profiles and reveal sex-specific cellular specializations in the early genital tubercle.
Collapse
Affiliation(s)
- Brooke A Armfield
- Department of Molecular Genetics and Microbiology, UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
| | - Martin J Cohn
- Department of Molecular Genetics and Microbiology, UF Genetics Institute, University of Florida, Gainesville, FL, 32610, USA; Department of Biology, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
46
|
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
|
47
|
Cheng X, Lai H, Luo W, Zhang M, Miao J, Song W, Xing S, Wang J, Gao WQ. Single-cell analysis reveals urothelial cell heterogeneity and regenerative cues following cyclophosphamide-induced bladder injury. Cell Death Dis 2021; 12:446. [PMID: 33953164 PMCID: PMC8099875 DOI: 10.1038/s41419-021-03740-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023]
Abstract
Cyclophosphamide is a commonly used chemotherapeutic drug to treat cancer with side effects that trigger bladder injury and hemorrhagic cystitis. Although previous studies have demonstrated that certain cell subsets and communications are activated to drive the repair and regeneration of bladder, it is not well understood how distinct bladder cell subsets function synergistically in this process. Here, we used droplet-based single-cell RNA sequencing (scRNA-seq) to profile the cell types within the murine bladder mucous layer under normal and injured conditions. Our analysis showed that superficial cells are directly repaired by cycling intermediate cells. We further identified two resident mesenchymal lineages (Acta2+ myofibroblasts and Cd34+ fibroblasts). The delineation of cell-cell communications revealed that Acta2+ myofibroblasts upregulated Fgf7 expression during acute injury, which activated Fgfr signaling in progenitor cells within the basal/intermediate layers to promote urothelial cell growth and repair. Overall, our study contributes to a more comprehensive understanding of the cellular dynamics during cyclophosphamide-induced bladder injury and may help identify important niche factors contributing to the regeneration of injured bladders.
Collapse
Affiliation(s)
- Xiaomu Cheng
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Huadong Lai
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wenqin Luo
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Man Zhang
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Juju Miao
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Weichen Song
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunpeng Xing
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jia Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China. .,Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
48
|
Iyyanki T, Zhang B, Wang Q, Hou Y, Jin Q, Xu J, Yang H, Liu T, Wang X, Song F, Luan Y, Yamashita H, Chien R, Lyu H, Zhang L, Wang L, Warrick J, Raman JD, Meeks JJ, DeGraff DJ, Yue F. Subtype-associated epigenomic landscape and 3D genome structure in bladder cancer. Genome Biol 2021; 22:105. [PMID: 33858483 PMCID: PMC8048365 DOI: 10.1186/s13059-021-02325-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 03/25/2021] [Indexed: 12/24/2022] Open
Abstract
Muscle-invasive bladder cancers are characterized by their distinct expression of luminal and basal genes, which could be used to predict key clinical features such as disease progression and overall survival. Transcriptionally, FOXA1, GATA3, and PPARG are shown to be essential for luminal subtype-specific gene regulation and subtype switching, while TP63, STAT3, and TFAP2 family members are critical for regulation of basal subtype-specific genes. Despite these advances, the underlying epigenetic mechanisms and 3D chromatin architecture responsible for subtype-specific regulation in bladder cancer remain unknown. RESULT: We determine the genome-wide transcriptome, enhancer landscape, and transcription factor binding profiles of FOXA1 and GATA3 in luminal and basal subtypes of bladder cancer. Furthermore, we report the first-ever mapping of genome-wide chromatin interactions by Hi-C in both bladder cancer cell lines and primary patient tumors. We show that subtype-specific transcription is accompanied by specific open chromatin and epigenomic marks, at least partially driven by distinct transcription factor binding at distal enhancers of luminal and basal bladder cancers. Finally, we identify a novel clinically relevant transcription factor, Neuronal PAS Domain Protein 2 (NPAS2), in luminal bladder cancers that regulates other subtype-specific genes and influences cancer cell proliferation and migration. CONCLUSION: In summary, our work identifies unique epigenomic signatures and 3D genome structures in luminal and basal urinary bladder cancers and suggests a novel link between the circadian transcription factor NPAS2 and a clinical bladder cancer subtype.
Collapse
Affiliation(s)
- Tejaswi Iyyanki
- Department of Biochemistry and Molecular Biology, Penn State School of Medicine, Hershey, PA, USA
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Baozhen Zhang
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
- Present address: Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Qixuan Wang
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Ye Hou
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Qiushi Jin
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Jie Xu
- Department of Biochemistry and Molecular Biology, Penn State School of Medicine, Hershey, PA, USA
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Hongbo Yang
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Tingting Liu
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Xiaotao Wang
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Fan Song
- Department of Biochemistry and Molecular Biology, Penn State School of Medicine, Hershey, PA, USA
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Yu Luan
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Hironobu Yamashita
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
- Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Ruby Chien
- University of Illinois College of Medicine, Chicago, IL, USA
| | - Huijue Lyu
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Lijun Zhang
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Lu Wang
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA
| | - Joshua Warrick
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
- Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Jay D Raman
- Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Joshua J Meeks
- Department of Urology, Feinberg School of Medicine and The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - David J DeGraff
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.
- Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine Northwestern University, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
| |
Collapse
|
49
|
Maurizi E, Adamo D, Magrelli FM, Galaverni G, Attico E, Merra A, Maffezzoni MBR, Losi L, Genna VG, Sceberras V, Pellegrini G. Regenerative Medicine of Epithelia: Lessons From the Past and Future Goals. Front Bioeng Biotechnol 2021; 9:652214. [PMID: 33842447 PMCID: PMC8026866 DOI: 10.3389/fbioe.2021.652214] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
This article explores examples of successful and unsuccessful regenerative medicine on human epithelia. To evaluate the applications of the first regenerated tissues, the analysis of the past successes and failures addresses some pending issues and lay the groundwork for developing new therapies. Research should still be encouraged to fill the gap between pathologies, clinical applications and what regenerative medicine can attain with current knowledge.
Collapse
Affiliation(s)
| | - Davide Adamo
- Interdepartmental Centre for Regenerative Medicine “Stefano Ferrari”, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Giulia Galaverni
- Interdepartmental Centre for Regenerative Medicine “Stefano Ferrari”, University of Modena and Reggio Emilia, Modena, Italy
| | - Eustachio Attico
- Interdepartmental Centre for Regenerative Medicine “Stefano Ferrari”, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Lorena Losi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Graziella Pellegrini
- Holostem Terapie Avanzate S.r.l., Modena, Italy
- Interdepartmental Centre for Regenerative Medicine “Stefano Ferrari”, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
50
|
Chen B, Chen X, Wang W, Shen J, Song Z, Ji H, Zhang F, Wu J, Na J, Li S. Tissue-engineered autologous peritoneal grafts for bladder reconstruction in a porcine model. J Tissue Eng 2021; 12:2041731420986796. [PMID: 33613958 PMCID: PMC7874343 DOI: 10.1177/2041731420986796] [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: 08/19/2020] [Accepted: 12/18/2020] [Indexed: 01/08/2023] Open
Abstract
Ileal neobladder construction is a common treatment for patients with bladder cancer after radical cystectomy. However, metabolic disorders caused by transposed bowel segments occur frequently. Bladder tissue engineering is a promising alternative approach. Although numerous studies have reported bladder reconstruction using acellular and cellular scaffolds, there are also disadvantages associated with these methods, such as immunogenicity of synthetic grafts and incompatible mechanical properties of the biomaterials. Here, we engineered an autologous peritoneal graft consisting of a peritoneal sheet and the seromuscular layer from the ileum. Three months after the surgery, compared with the neobladder made from the ileum, the reconstructed neobladder using our new method showed normal function and better gross morphological characteristics. Moreover, histopathological and transcriptomic analysis revealed urothelium-like cells expressing urothelial biomarkers appeared in the neobladder, while no such changes were observed in the control group. Overall, our study provides a new strategy for bladder tissue engineering and informs a variety of future research prospects.
Collapse
Affiliation(s)
- Biao Chen
- School of Clinical Medicine, Tsinghua University, Beijing, China.,Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Xia Chen
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Wenjia Wang
- Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Jun Shen
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhiqiang Song
- Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Haoyu Ji
- School of Clinical Medicine, Tsinghua University, Beijing, China.,Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Fangyuan Zhang
- School of Clinical Medicine, Tsinghua University, Beijing, China.,Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Jianchen Wu
- Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| | - Jie Na
- Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Shengwen Li
- School of Clinical Medicine, Tsinghua University, Beijing, China.,Department of Urology, The First Hospital of Tsinghua University, Beijing, China
| |
Collapse
|