1
|
Lammert FC, Pannhausen J, Noetzel E, Friedland F, Wirtz J, Herfs Y, Leypold S, Gan L, Weiskirchen R, Schnitzler T, Knüchel R, Maurer J, Jonigk DD, Rose M, Gaisa NT. Dual role of GRHL3 in bladder carcinogenesis depending on histological subtypes. Mol Oncol 2024; 18:1397-1416. [PMID: 38429970 PMCID: PMC11164254 DOI: 10.1002/1878-0261.13623] [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: 09/15/2023] [Revised: 01/23/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
The effect of grainyhead-like transcription factor 3 (GRHL3) on cancer development depends on the cancer subtypes as shown in tumor entities such as colorectal or oral squamous cell carcinomas. Here, we analyzed the subtype-specific role of GRHL3 in bladder carcinogenesis, comparing common urothelial carcinoma (UC) with squamous bladder cancer (sq-BLCA). We examined GRHL3 mRNA and protein expression in cohorts of patient samples, its prognostic role and its functional impact on tumorigeneses in different molecular and histopathological subtypes of bladder cancer. We showed for GRHL3 a reverse expression in squamous and urothelial bladder cancer subtypes. Stably GRHL3-overexpressing EJ28, J82, and SCaBER in vitro models revealed a tumor-suppressive function in squamous and an oncogenic role in the urothelial cancer cells affecting cell and colony growth, and migratory and invasive capacities. Transcriptomic profiling demonstrated highly subtype-specific GRHL3-regulated expression networks coined by the enrichment of genes involved in integrin-mediated pathways. In SCaBER, loss of ras homolog family member A (RHOA) GTPase activity was demonstrated to be associated with co-regulation of eukaryotic translation initiation factor 4E family member 3 (EIF4E3), a potential tumor suppressor gene. Thus, our data provide for the first time a detailed insight into the role of the transcription factor GRHL3 in different histopathological subtypes of bladder cancer.
Collapse
Affiliation(s)
- Franziska C. Lammert
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Julia Pannhausen
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Erik Noetzel
- Institute of Biological Information Processing 2 (IBI‐2), Mechanobiology, Forschungszentrum Jülich GmbHGermany
| | - Florian Friedland
- Institute of Biological Information Processing 2 (IBI‐2), Mechanobiology, Forschungszentrum Jülich GmbHGermany
| | - Julia Wirtz
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Yannick Herfs
- Institute of Biological Information Processing 2 (IBI‐2), Mechanobiology, Forschungszentrum Jülich GmbHGermany
| | - Sophie Leypold
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Lin Gan
- IZKF AachenMedical Faculty of the RWTH Aachen UniversityGermany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University HospitalRWTH Aachen UniversityGermany
| | - Tician Schnitzler
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
| | - Ruth Knüchel
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
| | - Jochen Maurer
- Department of Obstetrics and GynecologyUniversity Hospital AachenGermany
| | - Danny D. Jonigk
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- German Center for Lung Research, DZL, BREATHHanoverGermany
| | - Michael Rose
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- Institute of Pathology, University HospitalUniversity of UlmGermany
| | - Nadine T. Gaisa
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- Institute of Pathology, University HospitalUniversity of UlmGermany
| |
Collapse
|
2
|
Peng L, Chen JW, Chen YZ, Zhang C, Shen SH, Liu MZ, Fan Y, Yang SQ, Zhang XZ, Wang W, Gao XS, Di XP, Ma YC, Zeng X, Shen H, Jin X, Luo DY. UPK3A + umbrella cell damage mediated by TLR3-NR2F6 triggers programmed destruction of urothelium in Hunner-type interstitial cystitis/painful bladder syndrome. J Pathol 2024; 263:203-216. [PMID: 38551071 DOI: 10.1002/path.6275] [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: 11/21/2023] [Revised: 01/16/2024] [Accepted: 02/19/2024] [Indexed: 05/12/2024]
Abstract
Urothelial damage and barrier dysfunction emerge as the foremost mechanisms in Hunner-type interstitial cystitis/bladder pain syndrome (HIC). Although treatments aimed at urothelial regeneration and repair have been employed, their therapeutic effectiveness remains limited due to the inadequate understanding of specific cell types involved in damage and the lack of specific molecular targets within these mechanisms. Therefore, we harnessed single-cell RNA sequencing to elucidate the heterogeneity and developmental trajectory of urothelial cells within HIC bladders. Through reclustering, we identified eight distinct clusters of urothelial cells. There was a significant reduction in UPK3A+ umbrella cells and a simultaneous increase in progenitor-like pluripotent cells (PPCs) within the HIC bladder. Pseudotime analysis of the urothelial cells in the HIC bladder revealed that cells faced challenges in differentiating into UPK3A+ umbrella cells, while PPCs exhibited substantial proliferation to compensate for the loss of UPK3A+ umbrella cells. The urothelium in HIC remains unrepaired, despite the substantial proliferation of PPCs. Thus, we propose that inhibiting the pivotal signaling pathways responsible for the injury to UPK3A+ umbrella cells is paramount for restoring the urothelial barrier and alleviating lower urinary tract symptoms in HIC patients. Subsequently, we identified key molecular pathways (TLR3 and NR2F6) associated with the injury of UPK3A+ umbrella cells in HIC urothelium. Finally, we conducted in vitro and in vivo experiments to confirm the potential of the TLR3-NR2F6 axis as a promising therapeutic target for HIC. These findings hold the potential to inhibit urothelial injury, providing promising clues for early diagnosis and functional bladder self-repair strategies for HIC patients. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Liao Peng
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Jia-Wei Chen
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yuan-Zhuo Chen
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Chi Zhang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Si-Hong Shen
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Meng-Zhu Liu
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yang Fan
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Shi-Qin Yang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xiu-Zhen Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Department of Orthopedics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Wei Wang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xiao-Shuai Gao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xing-Peng Di
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yu-Cheng Ma
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xiao Zeng
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Hong Shen
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xi Jin
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - De-Yi Luo
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
- Pelvic Floor Diseases Center, West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Deuper L, Meuser M, Thiesler H, Jany UWH, Rudat C, Hildebrandt H, Trowe MO, Kispert A. Mesenchymal FGFR1 and FGFR2 control patterning of the ureteric mesenchyme by balancing SHH and BMP4 signaling. Development 2022; 149:276592. [DOI: 10.1242/dev.200767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 08/19/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The coordinated development of the mesenchymal and epithelial progenitors of the murine ureter depends on a complex interplay of diverse signaling activities. We have recently shown that epithelial FGFR2 signaling regulates stratification and differentiation of the epithelial compartment by enhancing epithelial Shh expression, and mesenchymal SHH and BMP4 activity. Here, we show that FGFR1 and FGFR2 expression in the mesenchymal primordium impinges on the SHH/BMP4 signaling axis to regulate mesenchymal patterning and differentiation. Mouse embryos with conditional loss of Fgfr1 and Fgfr2 in the ureteric mesenchyme exhibited reduced mesenchymal proliferation and prematurely activated lamina propria formation at the expense of the smooth muscle cell program. They also manifested hydroureter at birth. Molecular profiling detected increased SHH, WNT and retinoic acid signaling, whereas BMP4 signaling in the mesenchyme was reduced. Pharmacological activation of SHH signaling in combination with inhibition of BMP4 signaling recapitulated the cellular changes in explant cultures of wild-type ureters. Additional experiments suggest that mesenchymal FGFR1 and FGFR2 act as a sink for FGF ligands to dampen activation of Shh and BMP receptor gene expression by epithelial FGFR2 signaling.
Collapse
Affiliation(s)
- Lena Deuper
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Max Meuser
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover 2 , 30625 Hannover , Germany
| | - Ulrich W. H. Jany
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Carsten Rudat
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover 2 , 30625 Hannover , Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| |
Collapse
|
6
|
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: 2] [Impact Index Per Article: 1.0] [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
|
7
|
Kurz J, Weiss AC, Lüdtke THW, Deuper L, Trowe MO, Thiesler H, Hildebrandt H, Heineke J, Duncan SA, Kispert A. GATA6 is a crucial factor for Myocd expression in the visceral smooth muscle cell differentiation program of the murine ureter. Development 2022; 149:dev200522. [PMID: 35905011 PMCID: PMC10656427 DOI: 10.1242/dev.200522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2023]
Abstract
Smooth muscle cells (SMCs) are a crucial component of the mesenchymal wall of the ureter, as they account for the efficient removal of the urine from the renal pelvis to the bladder by means of their contractile activity. Here, we show that the zinc-finger transcription factor gene Gata6 is expressed in mesenchymal precursors of ureteric SMCs under the control of BMP4 signaling. Mice with a conditional loss of Gata6 in these precursors exhibit a delayed onset and reduced level of SMC differentiation and peristaltic activity, as well as dilatation of the ureter and renal pelvis (hydroureternephrosis) at birth and at postnatal stages. Molecular profiling revealed a delayed and reduced expression of the myogenic driver gene Myocd, but the activation of signaling pathways and transcription factors previously implicated in activation of the visceral SMC program in the ureter was unchanged. Additional gain-of-function experiments suggest that GATA6 cooperates with FOXF1 in Myocd activation and SMC differentiation, possibly as pioneer and lineage-determining factors, respectively.
Collapse
Affiliation(s)
- Jennifer Kurz
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Anna-Carina Weiss
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Timo H.-W. Lüdtke
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Lena Deuper
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Hauke Thiesler
- Institut für Klinische Biochemie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Herbert Hildebrandt
- Institut für Klinische Biochemie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Joerg Heineke
- Abteilung für Kardiovaskuläre Physiologie, European Center for Angioscience, Medizinische Fakultät Mannheim, Universität Heidelberg, D-68167 Mannheim, Germany
| | - Stephen A. Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| |
Collapse
|
8
|
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: 5] [Impact Index Per Article: 2.5] [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
|
9
|
Gasperoni JG, Fuller JN, Darido C, Wilanowski T, Dworkin S. Grainyhead-like (Grhl) Target Genes in Development and Cancer. Int J Mol Sci 2022; 23:ijms23052735. [PMID: 35269877 PMCID: PMC8911041 DOI: 10.3390/ijms23052735] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 12/12/2022] Open
Abstract
Grainyhead-like (GRHL) factors are essential, highly conserved transcription factors (TFs) that regulate processes common to both natural cellular behaviours during embryogenesis, and de-regulation of growth and survival pathways in cancer. Serving to drive the transcription, and therefore activation of multiple co-ordinating pathways, the three GRHL family members (GRHL1-3) are a critical conduit for modulating the molecular landscape that guides cellular decision-making processes during proliferation, epithelial-mesenchymal transition (EMT) and migration. Animal models and in vitro approaches harbouring GRHL loss or gain-of-function are key research tools to understanding gene function, which gives confidence that resultant phenotypes and cellular behaviours may be translatable to humans. Critically, identifying and characterising the target genes to which these factors bind is also essential, as they allow us to discover and understand novel genetic pathways that could ultimately be used as targets for disease diagnosis, drug discovery and therapeutic strategies. GRHL1-3 and their transcriptional targets have been shown to drive comparable cellular processes in Drosophila, C. elegans, zebrafish and mice, and have recently also been implicated in the aetiology and/or progression of a number of human congenital disorders and cancers of epithelial origin. In this review, we will summarise the state of knowledge pertaining to the role of the GRHL family target genes in both development and cancer, primarily through understanding the genetic pathways transcriptionally regulated by these factors across disparate disease contexts.
Collapse
Affiliation(s)
- Jemma G. Gasperoni
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia; (J.G.G.); (J.N.F.)
| | - Jarrad N. Fuller
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia; (J.G.G.); (J.N.F.)
| | - Charbel Darido
- The Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia;
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Tomasz Wilanowski
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland;
| | - Sebastian Dworkin
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia; (J.G.G.); (J.N.F.)
- Correspondence:
| |
Collapse
|
10
|
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: 4.0] [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
|
11
|
Kashgari G, Venkatesh S, Refuerzo S, Pham B, Bayat A, Klein RH, Ramos R, Ta AP, Plikus MV, Wang PH, Andersen B. GRHL3 activates FSCN1 to relax cell-cell adhesions between migrating keratinocytes during wound reepithelialization. JCI Insight 2021; 6:e142577. [PMID: 34494554 PMCID: PMC8492311 DOI: 10.1172/jci.insight.142577] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/21/2021] [Indexed: 01/23/2023] Open
Abstract
The migrating keratinocyte wound front is required for skin wound closure. Despite significant advances in wound healing research, we do not fully understand the molecular mechanisms that orchestrate collective keratinocyte migration. Here, we show that, in the wound front, the epidermal transcription factor Grainyhead like-3 (GRHL3) mediates decreased expression of the adherens junction protein E-cadherin; this results in relaxed adhesions between suprabasal keratinocytes, thus promoting collective cell migration and wound closure. Wound fronts from mice lacking GRHL3 in epithelial cells (Grhl3-cKO) have lower expression of Fascin-1 (FSCN1), a known negative regulator of E-cadherin. Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) on wounded keratinocytes shows decreased wound-induced chromatin accessibility near the Fscn1 gene in Grhl3-cKO mice, a region enriched for GRHL3 motifs. These data reveal a wound-induced GRHL3/FSCN1/E-cadherin pathway that regulates keratinocyte-keratinocyte adhesion during wound-front migration; this pathway is activated in acute human wounds and is altered in diabetic wounds in mice, suggesting translational relevance.
Collapse
Affiliation(s)
| | | | | | - Brandon Pham
- Department of Biological Chemistry, School of Medicine
| | - Anita Bayat
- Department of Biological Chemistry, School of Medicine
| | | | - Raul Ramos
- Department of Developmental & Cell Biology, School of Biological Sciences, and
| | - Albert Paul Ta
- Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine (UCI), California, USA
| | - Maksim V Plikus
- Department of Developmental & Cell Biology, School of Biological Sciences, and
| | - Ping H Wang
- Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine (UCI), California, USA
| | - Bogi Andersen
- Department of Biological Chemistry, School of Medicine.,Department of Medicine, Division of Endocrinology, School of Medicine, University of California, Irvine (UCI), California, USA
| |
Collapse
|
12
|
Phatak M, Kulkarni S, Miles LB, Anjum N, Dworkin S, Sonawane M. Grhl3 promotes retention of epidermal cells under endocytic stress to maintain epidermal architecture in zebrafish. PLoS Genet 2021; 17:e1009823. [PMID: 34570762 PMCID: PMC8496789 DOI: 10.1371/journal.pgen.1009823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 10/07/2021] [Accepted: 09/11/2021] [Indexed: 11/19/2022] Open
Abstract
Epithelia such as epidermis cover large surfaces and are crucial for survival. Maintenance of tissue homeostasis by balancing cell proliferation, cell size, and cell extrusion ensures epidermal integrity. Although the mechanisms of cell extrusion are better understood, how epithelial cells that round up under developmental or perturbed genetic conditions are reintegrated in the epithelium to maintain homeostasis remains unclear. Here, we performed live imaging in zebrafish embryos to show that epidermal cells that round up due to membrane homeostasis defects in the absence of goosepimples/myosinVb (myoVb) function, are reintegrated into the epithelium. Transcriptome analysis and genetic interaction studies suggest that the transcription factor Grainyhead-like 3 (Grhl3) induces the retention of rounded cells by regulating E-cadherin levels. Moreover, Grhl3 facilitates the survival of MyoVb deficient embryos by regulating cell adhesion, cell retention, and epidermal architecture. Our analyses have unraveled a mechanism of retention of rounded cells and its importance in epithelial homeostasis.
Collapse
Affiliation(s)
- Mandar Phatak
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shruti Kulkarni
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Lee B. Miles
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Australia
| | - Nazma Anjum
- Center for Biotechnology, A.C. College of Technology, Anna University, Chennai, India
| | - Sebastian Dworkin
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Australia
| | - Mahendra Sonawane
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
13
|
Deng Z, Cangkrama M, Butt T, Jane SM, Carpinelli MR. Grainyhead-like transcription factors: guardians of the skin barrier. Vet Dermatol 2021; 32:553-e152. [PMID: 33843098 DOI: 10.1111/vde.12956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/24/2020] [Accepted: 12/14/2020] [Indexed: 01/02/2023]
Abstract
There has been selective pressure to maintain a skin barrier since terrestrial animals evolved 360 million years ago. These animals acquired an unique integumentary system with a keratinized, stratified, squamous epithelium surface barrier. The barrier protects against dehydration and entry of microbes and toxins. The skin barrier centres on the stratum corneum layer of the epidermis and consists of cornified envelopes cemented by the intercorneocyte lipid matrix. Multiple components of the barrier undergo cross-linking by transglutaminase (TGM) enzymes, while keratins provide additional mechanical strength. Cellular tight junctions also are crucial for barrier integrity. The grainyhead-like (GRHL) transcription factors regulate the formation and maintenance of the integument in diverse species. GRHL3 is essential for formation of the skin barrier during embryonic development, whereas GRHL1 maintains the skin barrier postnatally. This is achieved by transactivation of Tgm1 and Tgm5, respectively. In addition to its barrier function, GRHL3 plays key roles in wound repair and as an epidermal tumour suppressor. In its former role, GRHL3 activates the planar cell polarity signalling pathway to mediate wound healing by providing directional migration cues. In squamous epithelium, GRHL3 regulates the balance between proliferation and differentiation, and its loss induces squamous cell carcinoma (SCC). In the skin, this is mediated through increased expression of MIR21, which reduces the expression levels of GRHL3 and its direct target, PTEN, leading to activation of the PI3K-AKT signalling pathway. These data position the GRHL family as master regulators of epidermal homeostasis across a vast gulf of evolutionary history.
Collapse
Affiliation(s)
- Zihao Deng
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Michael Cangkrama
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Tariq Butt
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Stephen M Jane
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Marina R Carpinelli
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
14
|
Wezel F, Lustig J, Azoitei A, Liu J, Meessen S, Najjar G, Zehe V, Faustmann P, Zengerling F, John A, Martini T, Bolenz C, Günes C. Grainyhead-Like 3 Influences Migration and Invasion of Urothelial Carcinoma Cells. Int J Mol Sci 2021; 22:ijms22062959. [PMID: 33803949 PMCID: PMC8000182 DOI: 10.3390/ijms22062959] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/25/2022] Open
Abstract
Invasive urothelial carcinomas of the bladder (UCB) characteristically show a loss of differentiation markers. The transcription factor Grainyhead-like 3 (GRHL3) plays an important role in the development and differentiation of normal urothelium. The contribution to UCB progression is still elusive. Differential expression of GRHL3 was assessed in normal human urothelium and in non-invasive and invasive bladder cancer cell lines. The contribution of GRHL3 to cell proliferation, viability and invasion in UCB cell lines was determined by gain- and loss-of-function assays in vitro and in an organ culture model using de-epithelialized porcine bladders. GRHL3 expression was detectable in normal human urothelial cells and showed significantly higher mRNA and protein levels in well-differentiated, non-invasive RT4 urothelial carcinoma cells compared to moderately differentiated RT112 cells. GRHL3 expression was absent in anaplastic and invasive T24 cells. Ectopic de novo expression of GRHL3 in T24 cells significantly impaired their migration and invasion properties in vitro and in organ culture. Its downregulation improved the invasive capacity of RT4 cells. The results indicate that GRHL3 may play a role in progression and metastasis in UCB. In addition, this work demonstrates that de-epithelialized porcine bladder organ culture can be a useful, standardized tool to assess the invasive capacity of cancer cells.
Collapse
|
15
|
Li Y, Liu Y, Gao Z, Zhang L, Chen L, Wu Z, Liu Q, Wang S, Zhou N, Chai TC, Shi B. Single-cell transcriptomes of mouse bladder urothelium uncover novel cell type markers and urothelial differentiation characteristics. Cell Prolif 2021; 54:e13007. [PMID: 33538002 PMCID: PMC8016651 DOI: 10.1111/cpr.13007] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/16/2020] [Accepted: 01/24/2021] [Indexed: 01/06/2023] Open
Abstract
Objectives Much of the information to date in terms of subtypes and function of bladder urothelial cells were derived from anatomical location or by the expression of a small number of marker genes. To have a comprehensive map of the cellular anatomy of bladder urothelial cells, we performed single‐cell RNA sequencing to thoroughly characterize mouse bladder urothelium. Materials and methods A total of 18,917 single cells from mouse bladder urothelium were analysed by unbiased single‐cell RNA sequencing. The expression of the novel cell marker was confirmed by immunofluorescence using urinary tract infection models. Results Unsupervised clustering analysis identified 8 transcriptionally distinct cell subpopulations from mouse bladder urothelial cells. We discovered a novel type of bladder urothelial cells marked by Plxna4 that may be involved with host response and wound healing. We also found a group of basal‐like cells labelled by ASPM that could be the progenitor cells of adult bladder urothelium. ASPM+ urothelial cells are significantly increased after injury by UPEC. In addition, specific transcription factors were found to be associated with urothelial cell differentiation. At the last, a number of interstitial cystitis/bladder pain syndrome–regulating genes were found differentially expressed among different urothelial cell subpopulations. Conclusions Our study provides a comprehensive characterization of bladder urothelial cells, which is fundamental to understanding the biology of bladder urothelium and associated bladder disease.
Collapse
Affiliation(s)
- Yan Li
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| | - Yaxiao Liu
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China.,Laboratory of Basic Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhengdong Gao
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| | - Lekai Zhang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| | - Lipeng Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| | - Zonglong Wu
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| | - Qinggang Liu
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| | - Shuai Wang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| | - Nan Zhou
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| | - Toby C Chai
- Department of Urology, Boston University/Boston Medical Center, Boston, MA, USA
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| |
Collapse
|
16
|
Liu Y, Huang W, Cai Z. Synthesizing AND gate minigene circuits based on CRISPReader for identification of bladder cancer cells. Nat Commun 2020; 11:5486. [PMID: 33127914 PMCID: PMC7599332 DOI: 10.1038/s41467-020-19314-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/07/2020] [Indexed: 11/09/2022] Open
Abstract
The logical AND gate gene circuit based on the CRISPR-Cas9 system can distinguish bladder cancer cells from normal bladder epithelial cells. However, the layered artificial gene circuits have the problems of high complexity, difficulty in accurately predicting the behavior, and excessive redundancy, which cannot be applied to clinical translation. Here, we construct minigene circuits based on the CRISPReader, a technology used to control promoter-less gene expression in a robust manner. The minigene circuits significantly induce robust gene expression output in bladder cancer cells, but have nearly undetectable gene expression in normal bladder epithelial cells. The minigene circuits show a higher capability for cancer identification and intervention when compared with traditional gene circuits, and could be used for in vivo cancer gene therapy using the all-in-one AAV vector. This approach expands the design ideas and concepts of gene circuits in medical synthetic biology.
Collapse
Affiliation(s)
- Yuchen Liu
- National and Local Joint Engineering Laboratory of Medical Synthetic Biology, Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, 518035, Shenzhen, China. .,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, 518035, Shenzhen, China. .,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, 518035, Shenzhen, China.
| | - Weiren Huang
- National and Local Joint Engineering Laboratory of Medical Synthetic Biology, Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, 518035, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, 518035, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, 518035, Shenzhen, China
| | - Zhiming Cai
- National and Local Joint Engineering Laboratory of Medical Synthetic Biology, Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, 518035, Shenzhen, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, 518035, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, 518035, Shenzhen, China
| |
Collapse
|
17
|
Sundararajan V, Pang QY, Choolani M, Huang RYJ. Spotlight on the Granules (Grainyhead-Like Proteins) - From an Evolutionary Conserved Controller of Epithelial Trait to Pioneering the Chromatin Landscape. Front Mol Biosci 2020; 7:213. [PMID: 32974388 PMCID: PMC7471608 DOI: 10.3389/fmolb.2020.00213] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Among the transcription factors that are conserved across phylogeny, the grainyhead family holds vital roles in driving the epithelial cell fate. In Drosophila, the function of grainyhead (grh) gene is essential during developmental processes such as epithelial differentiation, tracheal tube formation, maintenance of wing and hair polarity, and epidermal barrier wound repair. Three main mammalian orthologs of grh: Grainyhead-like 1-3 (GRHL1, GRHL2, and GRHL3) are highly conserved in terms of their gene structures and functions. GRHL proteins are essentially associated with the development and maintenance of the epithelial phenotype across diverse physiological conditions such as epidermal differentiation and craniofacial development as well as pathological functions including hearing impairment and neural tube defects. More importantly, through direct chromatin binding and induction of epigenetic alterations, GRHL factors function as potent suppressors of oncogenic cellular dedifferentiation program - epithelial-mesenchymal transition and its associated tumor-promoting phenotypes such as tumor cell migration and invasion. On the contrary, GRHL factors also induce pro-tumorigenic effects such as increased migration and anchorage-independent growth in certain tumor types. Furthermore, investigations focusing on the epithelial-specific activation of grh and GRHL factors have revealed that these factors potentially act as a pioneer factor in establishing a cell-type/cell-state specific accessible chromatin landscape that is exclusive for epithelial gene transcription. In this review, we highlight the essential roles of grh and GRHL factors during embryogenesis and pathogenesis, with a special focus on its emerging pioneering function.
Collapse
Affiliation(s)
- Vignesh Sundararajan
- Center for Translational Medicine, Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Qing You Pang
- Center for Translational Medicine, Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore
| | - Ruby Yun-Ju Huang
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore, Singapore
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
18
|
Boivin FJ, Schmidt-Ott KM. Functional roles of Grainyhead-like transcription factors in renal development and disease. Pediatr Nephrol 2020; 35:181-190. [PMID: 30554362 DOI: 10.1007/s00467-018-4171-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 11/07/2018] [Accepted: 12/06/2018] [Indexed: 12/11/2022]
Abstract
Proper renal function relies on the tightly regulated development of nephrons and collecting ducts. This process, known as tubulogenesis, involves dynamic cellular and molecular changes that instruct cells to form highly organized tubes of epithelial cells which compartmentalize the renal interstitium and tubular lumen via assembly of a selective barrier. The integrity and diversity of the various renal epithelia is achieved via formation of intercellular protein complexes along the apical-basal axis of the epithelial cells. In recent years, the evolutionarily conserved family of Grainyhead-like (GRHL) transcription factors which encompasses three mammalian family members (Grainyhead-like 1, 2, 3) has emerged as a group of critical regulators for organ development, epithelial differentiation, and barrier formation. Evidence from transgenic animal models supports the presence of Grainyhead-like-dependent transcriptional mechanisms that promote formation and maintenance of epithelial barriers in the kidney. In this review, we highlight different Grhl-dependent mechanisms that modulate epithelial differentiation in the kidney. Additionally, we discuss how disruptions in these mechanisms result in impaired renal function later in life.
Collapse
Affiliation(s)
- Felix J Boivin
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125, Berlin, Germany. .,Department of Nephrology, Charité Medical University, Berlin, Germany.
| |
Collapse
|
19
|
Meta-Analysis of Grainyhead-Like Dependent Transcriptional Networks: A Roadmap for Identifying Novel Conserved Genetic Pathways. Genes (Basel) 2019; 10:genes10110876. [PMID: 31683705 PMCID: PMC6896185 DOI: 10.3390/genes10110876] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/17/2022] Open
Abstract
The Drosophila grainyhead (grh) and vertebrate Grainyhead-like (Grhl) transcription factors are among the most critical genes for epithelial development, maintenance and homeostasis, and are remarkably well conserved from fungi to humans. Mutations affecting grh/Grhl function lead to a myriad of developmental and adult onset epithelial disease, such as aberrant skin barrier formation, facial/palatal clefting, impaired neural tube closure, age-related hearing loss, ectodermal dysplasia, and importantly, cancers of epithelial origin. Recently, mutations in the family member GRHL3 have been shown to lead to both syndromic and non-syndromic facial and palatal clefting in humans, particularly the genetic disorder Van Der Woude Syndrome (VWS), as well as spina bifida, whereas mutations in mammalian Grhl2 lead to exencephaly and facial clefting. As transcription factors, Grhl proteins bind to and activate (or repress) a substantial number of target genes that regulate and drive a cascade of transcriptional networks. A multitude of large-scale datasets have been generated to explore the grh/Grhl-dependent transcriptome, following ablation or mis-regulation of grh/Grhl-function. Here, we have performed a meta-analysis of all 41 currently published grh and Grhl RNA-SEQ, and microarray datasets, in order to identify and characterise the transcriptional networks controlled by grh/Grhl genes across disparate biological contexts. Moreover, we have also cross-referenced our results with published ChIP and ChIP-SEQ datasets, in order to determine which of the critical effector genes are likely to be direct grh/Grhl targets, based on genomic occupancy by grh/Grhl genes. Lastly, to interrogate the predictive strength of our approach, we experimentally validated the expression of the top 10 candidate grhl target genes in epithelial development, in a zebrafish model lacking grhl3, and found that orthologues of seven of these (cldn23, ppl, prom2, ocln, slc6a19, aldh1a3, and sod3) were significantly down-regulated at 48 hours post-fertilisation. Therefore, our study provides a strong predictive resource for the identification of putative grh/grhl effector target genes.
Collapse
|
20
|
Ming Q, Roske Y, Schuetz A, Walentin K, Ibraimi I, Schmidt-Ott KM, Heinemann U. Structural basis of gene regulation by the Grainyhead/CP2 transcription factor family. Nucleic Acids Res 2019; 46:2082-2095. [PMID: 29309642 PMCID: PMC5829564 DOI: 10.1093/nar/gkx1299] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/20/2017] [Indexed: 12/18/2022] Open
Abstract
Grainyhead (Grh)/CP2 transcription factors are highly conserved in multicellular organisms as key regulators of epithelial differentiation, organ development and skin barrier formation. In addition, they have been implicated as being tumor suppressors in a variety of human cancers. Despite their physiological importance, little is known about their structure and DNA binding mode. Here, we report the first structural study of mammalian Grh/CP2 factors. Crystal structures of the DNA-binding domains of grainyhead-like (Grhl) 1 and Grhl2 reveal a closely similar conformation with immunoglobulin-like core. Both share a common fold with the tumor suppressor p53, but differ in important structural features. The Grhl1 DNA-binding domain binds duplex DNA containing the consensus recognition element in a dimeric arrangement, supporting parsimonious target-sequence selection through two conserved arginine residues. We elucidate the molecular basis of a cancer-related mutation in Grhl1 involving one of these arginines, which completely abrogates DNA binding in biochemical assays and transcriptional activation of a reporter gene in a human cell line. Thus, our studies establish the structural basis of DNA target-site recognition by Grh transcription factors and reveal how tumor-associated mutations inactivate Grhl proteins. They may serve as points of departure for the structure-based development of Grh/CP2 inhibitors for therapeutic applications.
Collapse
Affiliation(s)
- Qianqian Ming
- Macromolecular Structure and Interaction, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany.,Chemistry and Biochemistry Institute, Freie Universität Berlin, Takustr. 6, 14195 Berlin, Germany
| | - Yvette Roske
- Macromolecular Structure and Interaction, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Anja Schuetz
- Macromolecular Structure and Interaction, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany.,Helmholtz Protein Sample Production Facility, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Katharina Walentin
- Molecular and Translational Kidney Research, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Ibraim Ibraimi
- Molecular and Translational Kidney Research, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Kai M Schmidt-Ott
- Molecular and Translational Kidney Research, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany.,Department of Nephrology, Charité Medical University, Charitéplatz 1, 10117 Berlin, Germany
| | - Udo Heinemann
- Macromolecular Structure and Interaction, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany.,Chemistry and Biochemistry Institute, Freie Universität Berlin, Takustr. 6, 14195 Berlin, Germany.,Helmholtz Protein Sample Production Facility, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| |
Collapse
|
21
|
Reese RM, Harrison MM, Alarid ET. Grainyhead-like Protein 2: The Emerging Role in Hormone-Dependent Cancers and Epigenetics. Endocrinology 2019; 160:1275-1288. [PMID: 30958537 DOI: 10.1210/en.2019-00213] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 01/16/2023]
Abstract
In mammals, the grainyhead-like transcription factor (GRHL) family is composed of three nuclear proteins that are responsible for driving epithelial cell fate: GRHL1, GRHL2, and GRHL3. GRHL2 is important in maintaining proper tubulogenesis during development and in suppressing the epithelial-to-mesenchymal transition. Within the last decade, evidence indicates both tumor-suppressive and oncogenic roles for GRHL2 in various types of cancers. Recent studies suggest that GRHL2 may be especially important in hormone-dependent cancers, as correlative relationships exist between GRHL2 and various steroid receptors, such as the androgen and estrogen receptors. Acting as a pioneer factor and coactivator, GRHL2 may directly affect steroid receptor transcriptional activity. This review will highlight recent discoveries of GRHL2 activity in cancer and in maintaining the epithelial state, while also exploring recent literature on the role of GRHL2 in hormone-dependent cancers and epigenetics.
Collapse
Affiliation(s)
- Rebecca M Reese
- Department of Oncology and Carbone Comprehensive Cancer Center, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin
| | - Melissa M Harrison
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Elaine T Alarid
- Department of Oncology and Carbone Comprehensive Cancer Center, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
22
|
De Castro SCP, Gustavsson P, Marshall AR, Gordon WM, Galea G, Nikolopoulou E, Savery D, Rolo A, Stanier P, Andersen B, Copp AJ, Greene NDE. Overexpression of Grainyhead-like 3 causes spina bifida and interacts genetically with mutant alleles of Grhl2 and Vangl2 in mice. Hum Mol Genet 2018; 27:4218-4230. [PMID: 30189017 PMCID: PMC6276835 DOI: 10.1093/hmg/ddy313] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/31/2018] [Accepted: 08/31/2018] [Indexed: 12/31/2022] Open
Abstract
The genetic basis of human neural tube defects (NTDs), such as anencephaly and spina bifida (SB), is complex and heterogeneous. Grainyhead-like genes represent candidates for involvement in NTDs based on the presence of SB and exencephaly in mice carrying loss-of-function alleles of Grhl2 or Grhl3. We found that reinstatement of Grhl3 expression, by bacterial artificial chromosome (BAC)-mediated transgenesis, prevents SB in Grhl3-null embryos, as in the Grhl3 hypomorphic curly tail strain. Notably, however, further increase in expression of Grhl3 causes highly penetrant SB. Grhl3 overexpression recapitulates the spinal NTD phenotype of loss-of-function embryos, although the underlying mechanism differs. However, it does not phenocopy other defects of Grhl3-null embryos such as abnormal axial curvature, cranial NTDs (exencephaly) or skin barrier defects, the latter being rescued by the Grhl3-transgene. Grhl2 and Grhl3 can form homodimers and heterodimers, suggesting a possible model in which defects arising from overexpression of Grhl3 result from sequestration of Grhl2 in heterodimers, mimicking Grhl2 loss of function. This hypothesis predicts that increased abundance of Grhl2 would have an ameliorating effect in Grhl3 overexpressing embryo. Instead, we observed a striking additive genetic interaction between Grhl2 and Grhl3 gain-of-function alleles. Severe SB arose in embryos in which both genes were expressed at moderately elevated levels that individually do not cause NTDs. Furthermore, moderate Grhl3 overexpression also interacted with the Vangl2Lp allele to cause SB, demonstrating genetic interaction with the planar cell polarity signalling pathway that is implicated in mouse and human NTDs.
Collapse
Affiliation(s)
- Sandra C P De Castro
- Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Peter Gustavsson
- Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Abigail R Marshall
- Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - William M Gordon
- Department of Biological Chemistry, University of California Irvine, Irvine, California, USA
| | - Gabriel Galea
- Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Evanthia Nikolopoulou
- Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Dawn Savery
- Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Ana Rolo
- Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Philip Stanier
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Bogi Andersen
- Department of Biological Chemistry, University of California Irvine, Irvine, California, USA
- Department of Medicine, School of Medicine, University of California Irvine, Irvine, California, USA
| | - Andrew J Copp
- Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Nicholas D E Greene
- Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
23
|
Hustler A, Eardley I, Hinley J, Pearson J, Wezel F, Radvanyi F, Baker SC, Southgate J. Differential transcription factor expression by human epithelial cells of buccal and urothelial derivation. Exp Cell Res 2018; 369:284-294. [PMID: 29842880 PMCID: PMC6092173 DOI: 10.1016/j.yexcr.2018.05.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022]
Abstract
Identification of transcription factors expressed by differentiated cells is informative not only of tissue-specific pathways, but to help identify master regulators for cellular reprogramming. If applied, such an approach could generate healthy autologous tissue-specific cells for clinical use where cells from the homologous tissue are unavailable due to disease. Normal human epithelial cells of buccal and urothelial derivation maintained in identical culture conditions that lacked significant instructive or permissive signaling cues were found to display inherent similarities and differences of phenotype. Investigation of transcription factors implicated in driving urothelial-type differentiation revealed buccal epithelial cells to have minimal or absent expression of PPARG, GATA3 and FOXA1 genes. Retroviral overexpression of protein coding sequences for GATA3 or PPARy1 in buccal epithelial cells resulted in nuclear immunolocalisation of the respective proteins, with both transductions also inducing expression of the urothelial differentiation-associated claudin 3 tight junction protein. PPARG1 overexpression alone entrained expression of nuclear FOXA1 and GATA3 proteins, providing objective evidence of its upstream positioning in a transcription factor network and identifying it as a candidate factor for urothelial-type transdifferentiation or reprogramming.
Collapse
Affiliation(s)
- Arianna Hustler
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Ian Eardley
- Pyrah Department of Urology, St. James's University Hospital, Leeds LS9 7TF, United Kingdom
| | - Jennifer Hinley
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Joanna Pearson
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Felix Wezel
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Francois Radvanyi
- Oncologie Moléculaire, Institut Curie, Centre de Recherche, 75248 Paris cedex 05, France
| | - Simon C Baker
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Jennifer Southgate
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, United Kingdom.
| |
Collapse
|
24
|
Miles LB, Darido C, Kaslin J, Heath JK, Jane SM, Dworkin S. Mis-expression of grainyhead-like transcription factors in zebrafish leads to defects in enveloping layer (EVL) integrity, cellular morphogenesis and axial extension. Sci Rep 2017; 7:17607. [PMID: 29242584 PMCID: PMC5730563 DOI: 10.1038/s41598-017-17898-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/01/2017] [Indexed: 02/07/2023] Open
Abstract
The grainyhead-like (grhl) transcription factors play crucial roles in craniofacial development, epithelial morphogenesis, neural tube closure, and dorso-ventral patterning. By utilising the zebrafish to differentially regulate expression of family members grhl2b and grhl3, we show that both genes regulate epithelial migration, particularly convergence-extension (CE) type movements, during embryogenesis. Genetic deletion of grhl3 via CRISPR/Cas9 results in failure to complete epiboly and pre-gastrulation embryonic rupture, whereas morpholino (MO)-mediated knockdown of grhl3 signalling leads to aberrant neural tube morphogenesis at the midbrain-hindbrain boundary (MHB), a phenotype likely due to a compromised overlying enveloping layer (EVL). Further disruptions of grhl3-dependent pathways (through co-knockdown of grhl3 with target genes spec1 and arhgef19) confirm significant MHB morphogenesis and neural tube closure defects. Concomitant MO-mediated disruption of both grhl2b and grhl3 results in further extensive CE-like defects in body patterning, notochord and somite morphogenesis. Interestingly, over-expression of either grhl2b or grhl3 also leads to numerous phenotypes consistent with disrupted cellular migration during gastrulation, including embryo dorsalisation, axial duplication and impaired neural tube migration leading to cyclopia. Taken together, our study ascribes novel roles to the Grhl family in the context of embryonic development and morphogenesis.
Collapse
Affiliation(s)
- Lee B Miles
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Charbel Darido
- The Victorian Comprehensive Cancer Centre, Peter MacCallum Cancer Centre, Parkville, VIC, 3050, Australia
| | - Jan Kaslin
- The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3168, Australia
| | - Joan K Heath
- Department of Chemical Biology, The Walter and Eliza Hall Institute, Parkville, VIC, 3050, Australia
| | - Stephen M Jane
- Department of Medicine, Monash University Central Clinical School, Prahran, VIC 3181, Australia.,Department of Hematology, Alfred Hospital, Prahran, VIC 3181, Australia
| | - Sebastian Dworkin
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
25
|
Hinze C, Ruffert J, Walentin K, Himmerkus N, Nikpey E, Tenstad O, Wiig H, Mutig K, Yurtdas ZY, Klein JD, Sands JM, Branchi F, Schumann M, Bachmann S, Bleich M, Schmidt-Ott KM. GRHL2 Is Required for Collecting Duct Epithelial Barrier Function and Renal Osmoregulation. J Am Soc Nephrol 2017; 29:857-868. [PMID: 29237740 DOI: 10.1681/asn.2017030353] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 11/09/2017] [Indexed: 12/31/2022] Open
Abstract
Collecting ducts make up the distal-most tubular segments of the kidney, extending from the cortex, where they connect to the nephron proper, into the medulla, where they release urine into the renal pelvis. During water deprivation, body water preservation is ensured by the selective transepithelial reabsorption of water into the hypertonic medullary interstitium mediated by collecting ducts. The collecting duct epithelium forms tight junctions composed of barrier-enforcing claudins and exhibits a higher transepithelial resistance than other segments of the renal tubule exhibit. However, the functional relevance of this strong collecting duct epithelial barrier is unresolved. Here, we report that collecting duct-specific deletion of an epithelial transcription factor, grainyhead-like 2 (GRHL2), in mice led to reduced expression of tight junction-associated barrier components, reduced collecting duct transepithelial resistance, and defective renal medullary accumulation of sodium and other osmolytes. In vitro, Grhl2-deficient collecting duct cells displayed increased paracellular flux of sodium, chloride, and urea. Consistent with these effects, Grhl2-deficient mice had diabetes insipidus, produced dilute urine, and failed to adequately concentrate their urine after water restriction, resulting in susceptibility to prerenal azotemia. These data indicate a direct functional link between collecting duct epithelial barrier characteristics, which appear to prevent leakage of interstitial osmolytes into urine, and body water homeostasis.
Collapse
Affiliation(s)
- Christian Hinze
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany.,Departments of Nephrology and Medical Intensive Care
| | - Janett Ruffert
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Berlin Institute of Urologic Research, Berlin, Germany
| | - Katharina Walentin
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Nina Himmerkus
- Institute of Physiology, Christian Albrechts University Kiel, Kiel, Germany
| | - Elham Nikpey
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway; and
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Zeliha Yesim Yurtdas
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Berlin Institute of Urologic Research, Berlin, Germany
| | - Janet D Klein
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Jeff M Sands
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Federica Branchi
- Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin, Berlin, Germany
| | - Michael Schumann
- Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin, Berlin, Germany
| | | | - Markus Bleich
- Institute of Physiology, Christian Albrechts University Kiel, Kiel, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; .,Departments of Nephrology and Medical Intensive Care
| |
Collapse
|
26
|
Klein RH, Hu W, Kashgari G, Lin Z, Nguyen T, Doan M, Andersen B. Characterization of enhancers and the role of the transcription factor KLF7 in regulating corneal epithelial differentiation. J Biol Chem 2017; 292:18937-18950. [PMID: 28916725 DOI: 10.1074/jbc.m117.793117] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 09/11/2017] [Indexed: 02/01/2023] Open
Abstract
During tissue development, transcription factors bind regulatory DNA regions called enhancers, often located at great distances from the genes they regulate, to control gene expression. The enhancer landscape during embryonic stem cell differentiation has been well characterized. By contrast, little is known about the shared and unique enhancer regulatory mechanisms in different ectodermally derived epithelial cells. Here we use ChIP sequencing (ChIP-seq) to identify domains enriched for the histone marks histone H3 lysine 4 trimethylation, histone H3 lysine 4 monomethylation, and histone H3 lysine 27 acetylation (H3K4me3, H3K4me1, and H3K27ac) and define, for the first time, the super enhancers and typical enhancers active in primary human corneal epithelial cells. We show that regulatory regions are often shared between cell types of the ectodermal lineage and that corneal epithelial super enhancers are already marked as potential regulatory domains in embryonic stem cells. Kruppel-like factor (KLF) motifs were enriched in corneal epithelial enhancers, consistent with the important roles of KLF4 and KLF5 in promoting corneal epithelial differentiation. We now show that the Kruppel family member KLF7 promotes the corneal progenitor cell state; on many genes, KLF7 antagonized the corneal differentiation-promoting KLF4. Furthermore, we found that two SNPs linked previously to corneal diseases, astigmatism, and Stevens-Johnson syndrome fall within corneal epithelial enhancers and alter their activity by disrupting transcription factor motifs that overlap these SNPs. Taken together, our work defines regulatory enhancers in corneal epithelial cells, highlights global gene-regulatory relationships shared among different epithelial cells, identifies a role for KLF7 as a KLF4 antagonist in corneal epithelial cell differentiation, and explains how two SNPs may contribute to corneal diseases.
Collapse
Affiliation(s)
- Rachel Herndon Klein
- From the Departments of Biological Chemistry and.,Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697
| | - William Hu
- From the Departments of Biological Chemistry and
| | | | - Ziguang Lin
- From the Departments of Biological Chemistry and
| | - Tuyen Nguyen
- From the Departments of Biological Chemistry and.,Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697
| | - Michael Doan
- From the Departments of Biological Chemistry and
| | - Bogi Andersen
- From the Departments of Biological Chemistry and .,Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697.,Medicine, Division of Endocrinology, and
| |
Collapse
|
27
|
Frisch SM, Farris JC, Pifer PM. Roles of Grainyhead-like transcription factors in cancer. Oncogene 2017; 36:6067-6073. [PMID: 28714958 DOI: 10.1038/onc.2017.178] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/12/2017] [Accepted: 05/04/2017] [Indexed: 12/18/2022]
Abstract
The mammalian homologs of the D. melanogaster Grainyhead gene, Grainyhead-like 1-3 (GRHL1, GRHL2 and GRHL3), are transcription factors implicated in wound healing, tubulogenesis and cancer. Their induced target genes encode diverse epithelial cell adhesion molecules, while mesenchymal genes involved in cell migration and invasion are repressed. Moreover, GRHL2 suppresses the oncogenic epithelial-mesencyhmal transition, thereby acting as a tumor suppressor. Mechanisms, some involving established cancer-related signaling/transcription factor pathways (for example, Wnt, TGF-β, mir200, ZEB1, OVOL2, p63 and p300) and translational implications of the Grainyhead proteins in cancer are discussed in this review article.
Collapse
Affiliation(s)
- S M Frisch
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - J C Farris
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| | - P M Pifer
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
28
|
Carpinelli MR, de Vries ME, Jane SM, Dworkin S. Grainyhead-like Transcription Factors in Craniofacial Development. J Dent Res 2017; 96:1200-1209. [PMID: 28697314 DOI: 10.1177/0022034517719264] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Craniofacial development in vertebrates involves the coordinated growth, migration, and fusion of several facial prominences during embryogenesis, processes governed by strict genetic and molecular controls. A failure in any of the precise spatiotemporal sequences of events leading to prominence fusion often leads to anomalous facial, skull, and jaw formation-conditions termed craniofacial defects (CFDs). Affecting approximately 0.1% to 0.3% of live births, CFDs are a highly heterogeneous class of developmental anomalies, which are often underpinned by genetic mutations. Therefore, identifying novel disease-causing mutations in genes that regulate craniofacial development is a critical prerequisite to develop new preventive or therapeutic measures. The Grainyhead-like ( GRHL) transcription factors are one such gene family, performing evolutionarily conserved roles in craniofacial patterning. The antecedent member of this family, Drosophila grainyhead ( grh), is required for head skeleton development in fruit flies, loss or mutation of Grhl family members in mouse and zebrafish models leads to defects of both maxilla and mandible, and recently, mutations in human GRHL3 have been shown to cause or contribute to both syndromic (Van Der Woude syndrome) and nonsyndromic palatal clefts. In this review, we summarize the current knowledge regarding the craniofacial-specific function of the Grainyhead-like family in multiple model species, identify some of the major target genes regulated by the Grhl transcription factors in craniofacial patterning, and, by examining animal models, draw inferences as to how these data will inform the likely roles of GRHL factors in human CFDs comprising palatal clefting. By understanding the molecular networks regulated by Grhl2 and Grhl3 target genes in other systems, we can propose likely pathways that mediate the effects of these transcription factors in human palatogenesis.
Collapse
Affiliation(s)
- M R Carpinelli
- 1 Central Clinical School, Monash University, Prahran, VIC, Australia
| | - M E de Vries
- 2 Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - S M Jane
- 1 Central Clinical School, Monash University, Prahran, VIC, Australia
| | - S Dworkin
- 2 Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
29
|
Klein RH, Lin Z, Hopkin AS, Gordon W, Tsoi LC, Liang Y, Gudjonsson JE, Andersen B. GRHL3 binding and enhancers rearrange as epidermal keratinocytes transition between functional states. PLoS Genet 2017; 13:e1006745. [PMID: 28445475 PMCID: PMC5425218 DOI: 10.1371/journal.pgen.1006745] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 05/10/2017] [Accepted: 04/05/2017] [Indexed: 11/19/2022] Open
Abstract
Transcription factor binding, chromatin modifications and large scale chromatin re-organization underlie progressive, irreversible cell lineage commitments and differentiation. We know little, however, about chromatin changes as cells enter transient, reversible states such as migration. Here we demonstrate that when human progenitor keratinocytes either differentiate or migrate they form complements of typical enhancers and super-enhancers that are unique for each state. Unique super-enhancers for each cellular state link to gene expression that confers functions associated with the respective cell state. These super-enhancers are also enriched for skin disease sequence variants. GRHL3, a transcription factor that promotes both differentiation and migration, binds preferentially to super-enhancers in differentiating keratinocytes, while during migration, it binds preferentially to promoters along with REST, repressing the expression of migration inhibitors. Key epidermal differentiation transcription factor genes, including GRHL3, are located within super-enhancers, and many of these transcription factors in turn bind to and regulate super-enhancers. Furthermore, GRHL3 represses the formation of a number of progenitor and non-keratinocyte super-enhancers in differentiating keratinocytes. Hence, chromatin relocates GRHL3 binding and enhancers to regulate both the irreversible commitment of progenitor keratinocytes to differentiation and their reversible transition to migration.
Collapse
Affiliation(s)
- Rachel Herndon Klein
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Ziguang Lin
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Amelia Soto Hopkin
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - William Gordon
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yun Liang
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Johann E. Gudjonsson
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Bogi Andersen
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California Irvine, Irvine, California, United States of America
- Department of Medicine, School of Medicine, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
30
|
Fishwick C, Higgins J, Percival-Alwyn L, Hustler A, Pearson J, Bastkowski S, Moxon S, Swarbreck D, Greenman CD, Southgate J. Heterarchy of transcription factors driving basal and luminal cell phenotypes in human urothelium. Cell Death Differ 2017; 24:809-818. [PMID: 28282036 PMCID: PMC5423105 DOI: 10.1038/cdd.2017.10] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 02/06/2023] Open
Abstract
Cell differentiation is affected by complex networks of transcription factors that co-ordinate re-organisation of the chromatin landscape. The hierarchies of these relationships can be difficult to dissect. During in vitro differentiation of normal human uro-epithelial cells, formaldehyde-assisted isolation of regulatory elements (FAIRE-seq) and RNA-seq was used to identify alterations in chromatin accessibility and gene expression changes following activation of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) as a differentiation-initiating event. Regions of chromatin identified by FAIRE-seq, as having altered accessibility during differentiation, were found to be enriched with sequence-specific binding motifs for transcription factors predicted to be involved in driving basal and differentiated urothelial cell phenotypes, including forkhead box A1 (FOXA1), P63, GRHL2, CTCF and GATA-binding protein 3 (GATA3). In addition, co-occurrence of GATA3 motifs was observed within subsets of differentiation-specific peaks containing P63 or FOXA1. Changes in abundance of GRHL2, GATA3 and P63 were observed in immunoblots of chromatin-enriched extracts. Transient siRNA knockdown of P63 revealed that P63 favoured a basal-like phenotype by inhibiting differentiation and promoting expression of basal marker genes. GATA3 siRNA prevented differentiation-associated downregulation of P63 protein and transcript, and demonstrated positive feedback of GATA3 on PPARG transcript, but showed no effect on FOXA1 transcript or protein expression. This approach indicates that as a transcriptionally regulated programme, urothelial differentiation operates as a heterarchy, wherein GATA3 is able to co-operate with FOXA1 to drive expression of luminal marker genes, but that P63 has potential to transrepress expression of the same genes.
Collapse
Affiliation(s)
- Carl Fishwick
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, UK
| | - Janet Higgins
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
| | | | - Arianna Hustler
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, UK
| | - Joanna Pearson
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, UK
| | | | - Simon Moxon
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
| | - David Swarbreck
- Earlham Institute, Norwich Research Park, Norwich NR4 7UZ, UK
| | - Chris D Greenman
- School of Computing Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Jennifer Southgate
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York YO10 5DD, UK
| |
Collapse
|
31
|
Al-Kurdi B. Hierarchical transcriptional profile of urothelial cells development and differentiation. Differentiation 2017; 95:10-20. [PMID: 28135607 DOI: 10.1016/j.diff.2016.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 10/09/2016] [Accepted: 10/14/2016] [Indexed: 11/27/2022]
Abstract
The urothelial lining of the lower urinary tract is the most efficient permeability barrier in animals, exhibiting a highly differentiated phenotype and a remarkable regenerative capacity upon wounding. During development and possibly during repair, cells undergo a sequence of hierarchical transcriptional events that mark the transition of these cells from the least differentiated urothelial phenotype characteristic of the basal cell layer, to the most differentiated cellular phenotype characteristic of the superficial cell layer. Unraveling normal urothelial differentiation program is essential to uncover the underlying causes of many congenital abnormalities and for the development of an appropriate differentiation niche for stem cells, for future use in urinary tract tissue engineering and organ reconstruction. Kruppel like factor-5 appears to be at the top of the hierarchy activating several downstream transcription factors, the most prominent of which is peroxisome proliferator activator receptor-γ. Eventually those lead to the activation of transcription factors that directly regulate the expression of uroplakin proteins along with other proteins that mediate the permeability function of the urothelium. In this review, we discuss the most recent findings in the area of urothelial cellular differentiation and transcriptional regulation, aiming for a comprehensive overview that aids in a refined understanding of this process.
Collapse
Affiliation(s)
- Ban Al-Kurdi
- Cell Therapy Center, The University of Jordan, Amman, Jordan.
| |
Collapse
|
32
|
Warrick JI, Walter V, Yamashita H, Chung E, Shuman L, Amponsa VO, Zheng Z, Chan W, Whitcomb TL, Yue F, Iyyanki T, Kawasawa YI, Kaag M, Guo W, Raman JD, Park JS, DeGraff DJ. FOXA1, GATA3 and PPARɣ Cooperate to Drive Luminal Subtype in Bladder Cancer: A Molecular Analysis of Established Human Cell Lines. Sci Rep 2016; 6:38531. [PMID: 27924948 PMCID: PMC5141480 DOI: 10.1038/srep38531] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/10/2016] [Indexed: 12/22/2022] Open
Abstract
Discrete bladder cancer molecular subtypes exhibit differential clinical aggressiveness and therapeutic response, which may have significant implications for identifying novel treatments for this common malignancy. However, research is hindered by the lack of suitable models to study each subtype. To address this limitation, we classified bladder cancer cell lines into molecular subtypes using publically available data in the Cancer Cell Line Encyclopedia (CCLE), guided by genomic characterization of bladder cancer by The Cancer Genome Atlas (TCGA). This identified a panel of bladder cancer cell lines which exhibit genetic alterations and gene expression patterns consistent with luminal and basal molecular subtypes of human disease. A subset of bladder cancer cell lines exhibit in vivo histomorphologic patterns consistent with luminal and basal subtypes, including papillary architecture and squamous differentiation. Using the molecular subtype assignments, and our own RNA-seq analysis, we found overexpression of GATA3 and FOXA1 cooperate with PPARɣ activation to drive transdifferentiation of a basal bladder cancer cells to a luminial phenotype. In summary, our analysis identified a set of human cell lines suitable for the study of molecular subtypes in bladder cancer, and furthermore indicates a cooperative regulatory network consisting of GATA3, FOXA1, and PPARɣ drive luminal cell fate.
Collapse
Affiliation(s)
- Joshua I Warrick
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA
| | - Vonn Walter
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA.,Department of Public Health Sciences, Pennsylvania State University College of Medicine, PA, USA
| | - Hironobu Yamashita
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Eunah Chung
- Division of Pediatric Urology and Developmental Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Lauren Shuman
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA
| | - Vasty Osei Amponsa
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Zongyu Zheng
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Wilson Chan
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA
| | - Tiffany L Whitcomb
- Department of Comparative Medicine, Pennsylvania State University College of Medicine, PA, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA.,Institute for Personalized Medicine, Pennsylvania State University College of Medicine, PA, USA
| | - Tejaswi Iyyanki
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA
| | - Yuka I Kawasawa
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA.,Institute for Personalized Medicine, Pennsylvania State University College of Medicine, PA, USA
| | - Matthew Kaag
- Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA
| | - Wansong Guo
- Department of Surgery, Division of Urology, Changchun Central Hospital, Changchun, China
| | - Jay D Raman
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA
| | - Joo-Seop Park
- Division of Pediatric Urology and Developmental Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - David J DeGraff
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA.,Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA
| |
Collapse
|
33
|
Abstract
Genomic and transcriptional studies have identified discrete molecular subtypes of bladder cancer. These observations could be the starting point to identify new treatments. Several members of the forkhead box (FOX) superfamily of transcription factors have been found to be differentially expressed in the different bladder cancer subtypes. In addition, the FOXA protein family are key regulators of embryonic bladder development and patterning. Both experimental and clinical data support a role for FOXA1 and FOXA2 in urothelial carcinoma. FOXA1 is expressed in embryonic and adult urothelium and its expression is altered in urothelial carcinomas and across disparate molecular bladder cancer subtypes. FOXA2 is normally absent from the adult urothelium, but developmental studies identified FOXA2 as a marker of a transient urothelial progenitor cell population during bladder development. Studies also implicate FOXA2 in bladder cancer and several other FOX proteins might be involved in development and/or progression of this disease; for example, FOXA1 and FOXO3A have been associated with clinical patient outcomes. Future studies should investigate to what extent and by which mechanisms FOX proteins might be directly involved in bladder cancer pathogenesis and treatment responses.
Collapse
|
34
|
Klein RH, Stephens DN, Ho H, Chen JK, Salmans ML, Wang W, Yu Z, Andersen B. Cofactors of LIM Domains Associate with Estrogen Receptor α to Regulate the Expression of Noncoding RNA H19 and Corneal Epithelial Progenitor Cell Function. J Biol Chem 2016; 291:13271-85. [PMID: 27129775 DOI: 10.1074/jbc.m115.709386] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Indexed: 11/06/2022] Open
Abstract
Cofactors of LIM domain proteins, CLIM1 and CLIM2, are widely expressed transcriptional cofactors that are recruited to gene regulatory regions by DNA-binding proteins, including LIM domain transcription factors. In the cornea, epithelium-specific expression of a dominant negative (DN) CLIM under the keratin 14 (K14) promoter causes blistering, wounding, inflammation, epithelial hyperplasia, and neovascularization followed by epithelial thinning and subsequent epidermal-like differentiation of the corneal epithelium. The defects in corneal epithelial differentiation and cell fate determination suggest that CLIM may regulate corneal progenitor cells and the transition to differentiation. Consistent with this notion, the K14-DN-Clim corneal epithelium first exhibits increased proliferation followed by fewer progenitor cells with decreased proliferative potential. In vivo ChIP-sequencing experiments with corneal epithelium show that CLIM binds to and regulates numerous genes involved in cell adhesion and proliferation, including limbally enriched genes. Intriguingly, CLIM associates primarily with non-LIM homeodomain motifs in corneal epithelial cells, including that of estrogen receptor α. Among CLIM targets is the noncoding RNA H19 whose deregulation is associated with Silver-Russell and Beckwith-Wiedemann syndromes. We demonstrate here that H19 negatively regulates corneal epithelial proliferation. In addition to cell cycle regulators, H19 affects the expression of multiple cell adhesion genes. CLIM interacts with estrogen receptor α at the H19 locus, potentially explaining the higher expression of H19 in female than male corneas. Together, our results demonstrate an important role for CLIM in regulating the proliferative potential of corneal epithelial progenitors and identify CLIM downstream target H19 as a regulator of corneal epithelial proliferation and adhesion.
Collapse
Affiliation(s)
- Rachel Herndon Klein
- From the Departments of Biological Chemistry and Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697 and
| | | | | | | | - Michael L Salmans
- From the Departments of Biological Chemistry and Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697 and
| | - Winnie Wang
- From the Departments of Biological Chemistry and
| | - Zhengquan Yu
- From the Departments of Biological Chemistry and State Key Laboratories for AgroBiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Bogi Andersen
- From the Departments of Biological Chemistry and Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697 and Medicine and
| |
Collapse
|
35
|
Abstract
As bladder reconstruction strategies evolve, a feasible and safe source of transplantable urothelium becomes a major consideration for patients with advanced bladder disease, particularly cancer. Pluripotent stem cells, such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are attractive candidates from which to derive urothelium as they renew and proliferate indefinitely in vitro and fulfill the non-autologous and/or non-urologic criteria, respectively, that is required for many patients. This review presents the latest advancements in differentiating urothelium from pluripotent stem cells in vitro in the context of current bladder tissue engineering strategies.
Collapse
|
36
|
Mlacki M, Kikulska A, Krzywinska E, Pawlak M, Wilanowski T. Recent discoveries concerning the involvement of transcription factors from the Grainyhead-like family in cancer. Exp Biol Med (Maywood) 2015; 240:1396-401. [PMID: 26069269 DOI: 10.1177/1535370215588924] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/01/2015] [Indexed: 12/17/2022] Open
Abstract
The Grainyhead-like (GRHL) family of transcription factors has three mammalian members, which are currently termed Grainyhead-like 1 (GRHL1), Grainyhead-like 2 (GRHL2), and Grainyhead-like 3 (GRHL3). These factors adopt a DNA-binding immunoglobulin fold homologous to the DNA-binding domain of key tumor suppressor p53. Their patterns of expression are tissue and developmentally specific. Earlier studies of the GRHL proteins focused on their functions in mammalian development. In recent years, these factors have been linked to many different types of cancer: squamous cell carcinoma of the skin, breast cancer, gastric cancer, hepatocellular carcinoma, colorectal cancer, clear cell renal cell carcinoma, neuroblastoma, prostate cancer, and cervical cancer. The roles of GRHL proteins in these various types of cancer are complex, and in some cases appear to be contradictory: they can serve to promote cancer development, or they may act as tumor suppressors, depending on the particular GRHL protein involved and on the cancer type. The reasons for obvious discrepancies in results from different studies remain unclear. At the molecular level, the GRHL transcription factors regulate the expression of genes whose products are involved in cellular proliferation, differentiation, adhesion, and polarity. We herein review the roles of GRHL proteins in cancer development, and we critically examine relevant molecular mechanisms, which were proposed by different authors. We also discuss the significance of recent discoveries implicating the involvement of GRHL transcription factors in cancer and highlight potential future applications of this knowledge in cancer treatment.
Collapse
Affiliation(s)
- Michal Mlacki
- Laboratory of Signal Transduction, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Agnieszka Kikulska
- Laboratory of Signal Transduction, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Ewa Krzywinska
- Laboratory of Signal Transduction, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Magdalena Pawlak
- Laboratory of Signal Transduction, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Tomasz Wilanowski
- Laboratory of Signal Transduction, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| |
Collapse
|
37
|
Bouhout S, Chabaud S, Bolduc S. Organ-specific matrix self-assembled by mesenchymal cells improves the normal urothelial differentiation in vitro. World J Urol 2015; 34:121-30. [PMID: 26008115 DOI: 10.1007/s00345-015-1596-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/16/2015] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Enterocystoplasty is the gold standard to perform bladder reconstruction. Since this technique has a high morbidity rate, several matrix scaffolds have been proposed to support the urothelial maturation. Unfortunately, epithelial cells failed to fully integrate the cell-matrix interactions and therefore appropriate signalling pathways of normal differentiation. Based on these observations, we proposed to culture bladder urothelial cells (BUC) onto a matrix self-assembled by bladder mesenchymal cells (BMC), to form a vesical model (VM). METHODS Different serum proportions were assessed to obtain a manipulable matrix deposited by BMC. The BUC were then seeded onto the BMC's matrix to evolve in a three-dimensional culture. Haematoxylin-eosin staining, immunolabeling, scanning electron microscopy, western blot and matrix metalloproteinases analysis were performed for the VM characterization. RESULTS We were able to obtain an original matrix made of collagen-I and presenting specific organization. Matrix remodelling was observed and led to a cellular compartmentalization. The reconstructed urothelium developed in a pseudostratified arrangement, displaying an adequate cellular polarity and apical membrane remodelling of superficial cells. Like native bladder, cytokeratin 14 immunolabeling was not observed in our VM, which indicate the conformity of the development sequence taken by BUC under the influence of the BMC's matrix. CONCLUSION Thus, it was possible to elaborate a VM without the use of exogenous matrices. The particular characteristics of the BMC's matrix permitted the development of an urothelium that shared the phenotype of native tissue. The autologous character of our VM, and its appropriate urothelial maturation, could potentially promote a better integration after grafting.
Collapse
Affiliation(s)
- S Bouhout
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX Faculté de médecine, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada.
| | - S Chabaud
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX Faculté de médecine, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada
| | - S Bolduc
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX Faculté de médecine, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada
| |
Collapse
|
38
|
Walentin K, Hinze C, Werth M, Haase N, Varma S, Morell R, Aue A, Pötschke E, Warburton D, Qiu A, Barasch J, Purfürst B, Dieterich C, Popova E, Bader M, Dechend R, Staff AC, Yurtdas ZY, Kilic E, Schmidt-Ott KM. A Grhl2-dependent gene network controls trophoblast branching morphogenesis. Development 2015; 142:1125-36. [PMID: 25758223 DOI: 10.1242/dev.113829] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Healthy placental development is essential for reproductive success; failure of the feto-maternal interface results in pre-eclampsia and intrauterine growth retardation. We found that grainyhead-like 2 (GRHL2), a CP2-type transcription factor, is highly expressed in chorionic trophoblast cells, including basal chorionic trophoblast (BCT) cells located at the chorioallantoic interface in murine placentas. Placentas from Grhl2-deficient mouse embryos displayed defects in BCT cell polarity and basement membrane integrity at the chorioallantoic interface, as well as a severe disruption of labyrinth branching morphogenesis. Selective Grhl2 inactivation only in epiblast-derived cells rescued all placental defects but phenocopied intraembryonic defects observed in global Grhl2 deficiency, implying the importance of Grhl2 activity in trophectoderm-derived cells. ChIP-seq identified 5282 GRHL2 binding sites in placental tissue. By integrating these data with placental gene expression profiles, we identified direct and indirect Grhl2 targets and found a marked enrichment of GRHL2 binding adjacent to genes downregulated in Grhl2(-/-) placentas, which encoded known regulators of placental development and epithelial morphogenesis. These genes included that encoding the serine protease inhibitor Kunitz type 1 (Spint1), which regulates BCT cell integrity and labyrinth formation. In human placenta, we found that human orthologs of murine GRHL2 and its targets displayed co-regulation and were expressed in trophoblast cells in a similar domain as in mouse placenta. Our data indicate that a conserved Grhl2-coordinated gene network controls trophoblast branching morphogenesis, thereby facilitating development of the site of feto-maternal exchange. This might have implications for syndromes related to placental dysfunction.
Collapse
Affiliation(s)
- Katharina Walentin
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany Experimental and Clinical Research Center, a collaboration between the Max Delbrück Center and the Medical Faculty of the Charité, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Christian Hinze
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany Experimental and Clinical Research Center, a collaboration between the Max Delbrück Center and the Medical Faculty of the Charité, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Max Werth
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany Experimental and Clinical Research Center, a collaboration between the Max Delbrück Center and the Medical Faculty of the Charité, Robert-Rössle-Str. 10, Berlin 13125, Germany Department of Medicine, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Nadine Haase
- Experimental and Clinical Research Center, a collaboration between the Max Delbrück Center and the Medical Faculty of the Charité, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Saaket Varma
- Department of Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Children's Hospital Los Angeles, 4650 Sunset Blvd., Los Angeles, CA 90027, USA
| | - Robert Morell
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders (NIDCD)/National Institutes of Health (NIH), 5 Research Court, Rockville, MD 20850, USA
| | - Annekatrin Aue
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany Experimental and Clinical Research Center, a collaboration between the Max Delbrück Center and the Medical Faculty of the Charité, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Elisabeth Pötschke
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - David Warburton
- Department of Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Children's Hospital Los Angeles, 4650 Sunset Blvd., Los Angeles, CA 90027, USA
| | - Andong Qiu
- Department of Medicine, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Jonathan Barasch
- Department of Medicine, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | - Bettina Purfürst
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Christoph Dieterich
- Bioinformatics, Max Planck Institute for Biology of Ageing, Robert-Koch-Str. 21, Cologne 50931, Germany
| | - Elena Popova
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Michael Bader
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, a collaboration between the Max Delbrück Center and the Medical Faculty of the Charité, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Anne Cathrine Staff
- Department of Gynecology and Obstetrics, Institute of Clinical Medicine, Oslo University Hospital and University of Oslo, Kirkeveien 166, Oslo 0450, Norway
| | - Zeliha Yesim Yurtdas
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany Department of Urology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany Berlin Institute of Urologic Research, Berlin 10117, Germany
| | - Ergin Kilic
- Department of Pathology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany Experimental and Clinical Research Center, a collaboration between the Max Delbrück Center and the Medical Faculty of the Charité, Robert-Rössle-Str. 10, Berlin 13125, Germany Department of Nephrology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, Germany
| |
Collapse
|
39
|
Farrokh S, Brillen AL, Haendeler J, Altschmied J, Schaal H. Critical regulators of endothelial cell functions: for a change being alternative. Antioxid Redox Signal 2015; 22:1212-29. [PMID: 25203279 DOI: 10.1089/ars.2014.6023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE The endothelium regulates vessel dilation and constriction, balances hemostasis, and inhibits thrombosis. In addition, pro- and anti-angiogenic molecules orchestrate proliferation, survival, and migration of endothelial cells. Regulation of all these processes requires fine-tuning of signaling pathways, which can easily be tricked into running the opposite direction when exogenous or endogenous signals get out of hand. Surprisingly, some critical regulators of physiological endothelial functions can turn malicious by mere alternative splicing, leading to the expression of protein isoforms with opposite functions. RECENT ADVANCES While reviewing the evidence of alternative splicing on cellular physiology, it became evident that expression of splice factors and their activities are regulated by externally triggered signaling cascades. Furthermore, genome-wide identification of RNA-binding sites of splicing regulatory proteins now offer a glimpse into the splicing code responsible for alternative splicing of molecules regulating endothelial functions. CRITICAL ISSUES Due to the constantly growing number of transcript and protein isoforms, it will become more and more important to identify and characterize all transcripts and proteins regulating endothelial cell functions. One critical issue will be a non-ambiguous nomenclature to keep consistency throughout different laboratories. FUTURE DIRECTIONS RNA-deep sequencing focusing on exon-exon junction needs to more reliably identify alternative splicing events combined with functional analyses that will uncover more splice variants contributing to or inhibiting proper endothelial functions. In addition, understanding the signals mediating alternative splicing and its regulation might allow us to derive new strategies to preserve endothelial function by suppressing or upregulating specific protein isoforms. Antioxid. Redox Signal. 22, 1212-1229.
Collapse
Affiliation(s)
- Sabrina Farrokh
- 1 Heisenberg-Group-Environmentally-Induced Cardiovascular Degeneration, IUF-Leibniz Research Institute for Environmental Medicine , Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
40
|
Aue A, Hinze C, Walentin K, Ruffert J, Yurtdas Y, Werth M, Chen W, Rabien A, Kilic E, Schulzke JD, Schumann M, Schmidt-Ott KM. A Grainyhead-Like 2/Ovo-Like 2 Pathway Regulates Renal Epithelial Barrier Function and Lumen Expansion. J Am Soc Nephrol 2015; 26:2704-15. [PMID: 25788534 DOI: 10.1681/asn.2014080759] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 12/30/2014] [Indexed: 12/20/2022] Open
Abstract
Grainyhead transcription factors control epithelial barriers, tissue morphogenesis, and differentiation, but their role in the kidney is poorly understood. Here, we report that nephric duct, ureteric bud, and collecting duct epithelia express high levels of grainyhead-like homolog 2 (Grhl2) and that nephric duct lumen expansion is defective in Grhl2-deficient mice. In collecting duct epithelial cells, Grhl2 inactivation impaired epithelial barrier formation and inhibited lumen expansion. Molecular analyses showed that GRHL2 acts as a transcriptional activator and strongly associates with histone H3 lysine 4 trimethylation. Integrating genome-wide GRHL2 binding as well as H3 lysine 4 trimethylation chromatin immunoprecipitation sequencing and gene expression data allowed us to derive a high-confidence GRHL2 target set. GRHL2 transactivated a group of genes including Ovol2, encoding the ovo-like 2 zinc finger transcription factor, as well as E-cadherin, claudin 4 (Cldn4), and the small GTPase Rab25. Ovol2 induction alone was sufficient to bypass the requirement of Grhl2 for E-cadherin, Cldn4, and Rab25 expression. Re-expression of either Ovol2 or a combination of Cldn4 and Rab25 was sufficient to rescue lumen expansion and barrier formation in Grhl2-deficient collecting duct cells. Hence, we identified a Grhl2/Ovol2 network controlling Cldn4 and Rab25 expression that facilitates lumen expansion and barrier formation in subtypes of renal epithelia.
Collapse
Affiliation(s)
- Annekatrin Aue
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; Experimental and Clinical Research Center, and
| | - Christian Hinze
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; Departments of Nephrology
| | | | - Janett Ruffert
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Yesim Yurtdas
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; Urology, Berlin Institute of Urologic Research, Berlin, Germany
| | - Max Werth
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Wei Chen
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Anja Rabien
- Urology, Berlin Institute of Urologic Research, Berlin, Germany
| | | | | | - Michael Schumann
- Gastroenterology, Charité Medical University, Berlin, Germany; and
| | - Kai M Schmidt-Ott
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; Experimental and Clinical Research Center, and Departments of Nephrology,
| |
Collapse
|
41
|
Epidermal cell junctions and their regulation by p63 in health and disease. Cell Tissue Res 2015; 360:513-28. [PMID: 25645146 DOI: 10.1007/s00441-014-2108-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/17/2014] [Indexed: 12/17/2022]
Abstract
As the outermost tissue of the body, the epidermis is the first physical barrier for any pressure, stress or trauma. Several specialized cell-matrix and cell-cell adhesion structures, together with an intracellular network of dedicated intermediate filaments, are required to confer critical resilience to mechanical stress. The transcription factor p63 is a master regulator of gene expression in the epidermis and in other stratified epithelia. It has been extensively demonstrated that p63 positively controls a large number of tissue-specific genes, including those encoding a large fraction of tissue-restricted cell adhesion molecules. Consistent with p63 functions in cell adhesion and in epidermal differentiation, heterozygous mutations clustered mainly in the p63 C-terminus are causative of AEC syndrome, an autosomal dominant disorder characterized by cleft palate, ankyloblepharon and ectodermal dysplasia associated with severe skin erosions, bleeding and infections. The molecular basis of skin erosions in AEC patients is not fully understood, although defects in desmosomes and in other cell junctions are likely to be involved. Here, we provide an extensive review of the different epidermal cell junctions that cooperate to withstand mechanical stress and on the mechanisms by which p63 regulates gene expression of their components in healthy skin and in AEC syndrome. Collectively, advancement in understanding the molecular mechanisms by which epidermal cell junctions precisely exert their functions and how p63 orchestrates their coordinated expression, will ultimately lead to insight into developing future strategies for the treatment of AEC syndrome and more in generally for diseases that share an overlapping phenotype.
Collapse
|
42
|
SNX31: a novel sorting nexin associated with the uroplakin-degrading multivesicular bodies in terminally differentiated urothelial cells. PLoS One 2014; 9:e99644. [PMID: 24914955 PMCID: PMC4051706 DOI: 10.1371/journal.pone.0099644] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/16/2014] [Indexed: 11/19/2022] Open
Abstract
Uroplakins (UP), a group of integral membrane proteins, are major urothelial differentiation products that form 2D crystals of 16-nm particles (urothelial plaques) covering the apical surface of mammalian bladder urothelium. They contribute to the urothelial barrier function and, one of them, UPIa, serves as the receptor for uropathogenic Escherichia coli. It is therefore important to understand the mechanism by which these surface-associated uroplakins are degraded. While it is known that endocytosed uroplakin plaques are targeted to and line the multivesicular bodies (MVBs), it is unclear how these rigid-looking plaques can go to the highly curved membranes of intraluminal vesicles (ILVs). From a cDNA subtraction library, we identified a highly urothelium-specific sorting nexin, SNX31. SNX31 is expressed, like uroplakins, in terminally differentiated urothelial umbrella cells where it is predominantly associated with MVBs. Apical membrane proteins including uroplakins that are surface biotin-tagged are endocytosed and targeted to the SNX31-positive MVBs. EM localization demonstrated that SNX31 and uroplakins are both associated not only with the limiting membranes of MVBs containing uroplakin plaques, but also with ILVs. SNX31 can bind, on one hand, the PtdIns3P-enriched lipids via its N-terminal PX-domain, and, on the other hand, it binds uroplakins as demonstrated by co-immunoprecipitation and proximity ligation assay, and by its reduced membrane association in uroplakin II-deficient urothelium. The fact that in urothelial umbrella cells MVBs are the only major intracellular organelles enriched in both PtdIns3P and uroplakins may explain SNX31's MVB-specificity in these cells. However, in MDCK and other cultured cells transfected SNX31 can bind to early endosomes possibly via lipids. These data support a model in which SNX31 mediates the endocytic degradation of uroplakins by disassembling/collapsing the MVB-associated uroplakin plaques, thus enabling the uroplakin-containing (but ‘softened’) membranes to bud and form the ILVs for lysosomal degradation and/or exosome formation.
Collapse
|
43
|
Tanimizu N, Mitaka T. Role of grainyhead-like 2 in the formation of functional tight junctions. Tissue Barriers 2014; 1:e23495. [PMID: 24665375 PMCID: PMC3875637 DOI: 10.4161/tisb.23495] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 12/24/2012] [Accepted: 01/04/2013] [Indexed: 02/02/2023] Open
Abstract
Epithelial cells develop intercellular junctions, including tight junctions (TJs) and adherens junctions (AJs). In epithelial tissues, TJs act as barriers that protect bodies from dehydration, infection and toxic substances. However, the molecular mechanisms regulating the establishment of functional TJs during organogenesis remain largely unknown. Recently, we identified grainyhead-like 2 (Grhl2) as a transcription factor that is specifically expressed in cholangiocytes, which are epithelial cells lining the bile duct tubules in the liver. Using our three-dimensional (3D) culture system of hepatic progenitor cells, we demonstrated that Grhl2 enhanced barrier functions of hepatic progenitor cells by upregulating claudin (Cldn) 3 and Cldn4, thereby promoting epithelial morphogenesis. In addition, we identified Rab25 as another target of Grhl2, which promotes the localization of Cldn4 at TJs. Our results indicate that a transcription factor promotes epithelial morphogenesis by establishing functional TJs by not only regulating the transcription of Cldns but also affecting their localization at TJs.
Collapse
Affiliation(s)
- Naoki Tanimizu
- Department of Tissue Development and Regeneration; Research Institute for Frontier Medicine; Sapporo Medical University School of Medicine; Sapporo, Japan
| | - Toshihiro Mitaka
- Department of Tissue Development and Regeneration; Research Institute for Frontier Medicine; Sapporo Medical University School of Medicine; Sapporo, Japan
| |
Collapse
|
44
|
Osborn SL, Thangappan R, Luria A, Lee JH, Nolta J, Kurzrock EA. Induction of human embryonic and induced pluripotent stem cells into urothelium. Stem Cells Transl Med 2014; 3:610-9. [PMID: 24657961 DOI: 10.5966/sctm.2013-0131] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
In vitro generation of human urothelium from stem cells would be a major advancement in the regenerative medicine field, providing alternate nonurologic and/or nonautologous tissue sources for bladder grafts. Such a model would also help decipher the mechanisms of urothelial differentiation and would facilitate investigation of deviated differentiation of normal progenitors into urothelial cancer stem cells, perhaps elucidating areas of intervention for improved treatments. Thus far, in vitro derivation of urothelium from human embryonic stem cells (hESCs) or human induced pluripotent stem (hiPS) cells has not been reported. The goal of this work was to develop an efficient in vitro protocol for the induction of hESCs into urothelium through an intermediary definitive endoderm step and free of matrices and cell contact. During directed differentiation in a urothelial-specific medium ("Uromedium"), hESCs produced up to 60% urothelium, as determined by uroplakin expression; subsequent propagation selected for 90% urothelium. Alteration of the epithelial and mesenchymal cell signaling contribution through noncell contact coculture or conditioned media did not enhance the production of urothelium. Temporospatial evaluation of transcription factors known to be involved in urothelial specification showed association of IRF1, GET1, and GATA4 with uroplakin expression. Additional hESC and hiPS cell lines could also be induced into urothelium using this in vitro system. These results demonstrate that derivation and propagation of urothelium from hESCs and hiPS cells can be efficiently accomplished in vitro in the absence of matrices, cell contact, or adult cell signaling and that the induction process appears to mimic normal differentiation.
Collapse
Affiliation(s)
- Stephanie L Osborn
- Departments of Urology and Internal Medicine, Davis School of Medicine, and Stem Cell Program, Institute for Regenerative Cures, Davis Medical Center, University of California, Sacramento, California, USA
| | | | | | | | | | | |
Collapse
|
45
|
Identification of ELF3 as an early transcriptional regulator of human urothelium. Dev Biol 2013; 386:321-30. [PMID: 24374157 DOI: 10.1016/j.ydbio.2013.12.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 12/13/2013] [Accepted: 12/18/2013] [Indexed: 01/01/2023]
Abstract
Despite major advances in high-throughput and computational modelling techniques, understanding of the mechanisms regulating tissue specification and differentiation in higher eukaryotes, particularly man, remains limited. Microarray technology has been explored exhaustively in recent years and several standard approaches have been established to analyse the resultant datasets on a genome-wide scale. Gene expression time series offer a valuable opportunity to define temporal hierarchies and gain insight into the regulatory relationships of biological processes. However, unless datasets are exactly synchronous, time points cannot be compared directly. Here we present a data-driven analysis of regulatory elements from a microarray time series that tracked the differentiation of non-immortalised normal human urothelial (NHU) cells grown in culture. The datasets were obtained by harvesting differentiating and control cultures from finite bladder- and ureter-derived NHU cell lines at different time points using two previously validated, independent differentiation-inducing protocols. Due to the asynchronous nature of the data, a novel ranking analysis approach was adopted whereby we compared changes in the amplitude of experiment and control time series to identify common regulatory elements. Our approach offers a simple, fast and effective ranking method for genes that can be applied to other time series. The analysis identified ELF3 as a candidate transcriptional regulator involved in human urothelial cytodifferentiation. Differentiation-associated expression of ELF3 was confirmed in cell culture experiments and by immunohistochemical demonstration in situ. The importance of ELF3 in urothelial differentiation was verified by knockdown in NHU cells, which led to reduced expression of FOXA1 and GRHL3 transcription factors in response to PPARγ activation. The consequences of this were seen in the repressed expression of late/terminal differentiation-associated uroplakin 3a gene expression and in the compromised development and regeneration of urothelial barrier function.
Collapse
|
46
|
Stephens DN, Klein RH, Salmans ML, Gordon W, Ho H, Andersen B. The Ets transcription factor EHF as a regulator of cornea epithelial cell identity. J Biol Chem 2013; 288:34304-24. [PMID: 24142692 DOI: 10.1074/jbc.m113.504399] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The cornea is the clear, outermost portion of the eye composed of three layers: an epithelium that provides a protective barrier while allowing transmission of light into the eye, a collagen-rich stroma, and an endothelium monolayer. How cornea development and aging is controlled is poorly understood. Here we characterize the mouse cornea transcriptome from early embryogenesis through aging and compare it with transcriptomes of other epithelial tissues, identifying cornea-enriched genes, pathways, and transcriptional regulators. Additionally, we profiled cornea epithelium and stroma, defining genes enriched in these layers. Over 10,000 genes are differentially regulated in the mouse cornea across the time course, showing dynamic expression during development and modest expression changes in fewer genes during aging. A striking transition time point for gene expression between postnatal days 14 and 28 corresponds with completion of cornea development at the transcriptional level. Clustering classifies co-expressed, and potentially co-regulated, genes into biologically informative categories, including groups that exhibit epithelial or stromal enriched expression. Based on these findings, and through loss of function studies and ChIP-seq, we show that the Ets transcription factor EHF promotes cornea epithelial fate through complementary gene activating and repressing activities. Furthermore, we identify potential interactions between EHF, KLF4, and KLF5 in promoting cornea epithelial differentiation. These data provide insights into the mechanisms underlying epithelial development and aging, identifying EHF as a regulator of cornea epithelial identity and pointing to interactions between Ets and KLF factors in promoting epithelial fate. Furthermore, this comprehensive gene expression data set for the cornea is a powerful tool for discovery of novel cornea regulators and pathways.
Collapse
|
47
|
LI X, HUO X, LIU K, LI X, WANG M, CHU H, HU F, SHENG H, ZHANG Z, ZHU B. Association between genetic variations in GRHL2 and noise-induced hearing loss in Chinese high intensity noise exposed workers: a case-control analysis. INDUSTRIAL HEALTH 2013; 51:612-621. [PMID: 24131873 PMCID: PMC4202744 DOI: 10.2486/indhealth.2012-0084] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 10/02/2013] [Indexed: 06/01/2023]
Abstract
The grainyhead like 2 (GRHL2) is a transcription factor, and the role among noise exposed workers is not well established. We tested whether GRHL2 polymorphisms are associated with the risk of noise-induced hearing loss (NIHL) in Chinese high intensity noise exposed workers. We genotyped six polymorphisms of GRHL2 gene (i.e., rs611419, rs3779617, rs3735713, rs3735714, rs3735715, and rs6989650) of 340 NIHL cases and 356 control subjects who exposed to noise higher than 85 dB (A) [Lex, 8 h=time-weighted average of levels of noise exposure (Lex) for a nominal 8 h working day] in a Chinese population. Compared with rs611419 AA genotype, the AT/TT genotypes conferred protection against NIHL [adjusted odds ratio (OR)=0.71, 95% confidence interval (CI)=0.52-0.98]. No altered NIHL risk was associated with the other five polymorphisms. In the combined analyses, we found that the combined genotypes with three to eight variant alleles were associated with an decrease risk of NIHL compared with those with zero to two variant alleles, and the decrease risk was more pronounced among subgroups of exposure time>20 yr (0.31, 0.16-0.62) and drinkers (0.51, 0.29-0.90). Polymorphisms of GRHL2 may positively contribute to the etiology of NIHL.
Collapse
Affiliation(s)
- Xin LI
- Department of Environmental Genomics, Jiangsu Key Lab of
Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University,
China
- Department of Genetic Toxicology, The Key Laboratory of
Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical
University, China
| | - Xinying HUO
- Department of Environmental Genomics, Jiangsu Key Lab of
Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University,
China
- Department of Genetic Toxicology, The Key Laboratory of
Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical
University, China
| | - Kai LIU
- Department of Disease Prevention, The Third Affiliated
Hospital of Nanjing Medical University, China
| | - Xiuting LI
- Department of Environmental Genomics, Jiangsu Key Lab of
Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University,
China
- Institute of Occupational Disease Prevention, Jiangsu
Provincial Center for Disease Prevention and Control, China
| | - Meilin WANG
- Department of Environmental Genomics, Jiangsu Key Lab of
Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University,
China
- Department of Genetic Toxicology, The Key Laboratory of
Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical
University, China
| | - Haiyan CHU
- Department of Environmental Genomics, Jiangsu Key Lab of
Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University,
China
- Department of Genetic Toxicology, The Key Laboratory of
Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical
University, China
| | - Feifei HU
- Department of Environmental Genomics, Jiangsu Key Lab of
Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University,
China
- Department of Genetic Toxicology, The Key Laboratory of
Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical
University, China
| | - Huanxi SHENG
- Department of Environmental Genomics, Jiangsu Key Lab of
Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University,
China
- Institute of Occupational Disease Prevention, Jiangsu
Provincial Center for Disease Prevention and Control, China
| | - Zhengdong ZHANG
- Department of Environmental Genomics, Jiangsu Key Lab of
Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University,
China
- Department of Genetic Toxicology, The Key Laboratory of
Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical
University, China
| | - Baoli ZHU
- Department of Environmental Genomics, Jiangsu Key Lab of
Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University,
China
- Institute of Occupational Disease Prevention, Jiangsu
Provincial Center for Disease Prevention and Control, China
| |
Collapse
|
48
|
Molecular basis of renal adaptation in a murine model of congenital obstructive nephropathy. PLoS One 2013; 8:e72762. [PMID: 24023768 PMCID: PMC3762787 DOI: 10.1371/journal.pone.0072762] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 07/15/2013] [Indexed: 12/02/2022] Open
Abstract
Congenital obstructive nephropathy is a common cause of chronic kidney disease and a leading indication for renal transplant in children. The cellular and molecular responses of the kidney to congenital obstruction are incompletely characterized. In this study, we evaluated global transcription in kidneys with graded hydronephrosis in the megabladder (mgb−/−) mouse to better understand the pathophysiology of congenital obstructive nephropathy. Three primary pathways associated with kidney remodeling/repair were induced in mgb−/− kidneys independent of the degree of hydronephrosis. These pathways included retinoid signaling, steroid hormone metabolism, and renal response to injury. Urothelial proliferation and the expression of genes with roles in the integrity and maintenance of the renal urothelium were selectively increased in mgb−/− kidneys. Ngal/Lcn2, a marker of acute kidney injury, was elevated in 36% of kidneys with higher grades of hydronephrosis. Evaluation of Ngalhigh versus Ngallow kidneys identified the expression of several novel candidate markers of renal injury. This study indicates that the development of progressive hydronephrosis in mgb−/− mice results in renal adaptation that includes significant changes in the morphology and potential functionality of the renal urothelium. These observations will permit the development of novel biomarkers and therapeutic approaches to progressive renal injury in the context of congenital obstruction.
Collapse
|
49
|
Zhidkova OV, Petrov NS, Popov BV. Preparation and characteristics of growth and marker properties of urinary bladder mesenchymal stem cells. J EVOL BIOCHEM PHYS+ 2013. [DOI: 10.1134/s0022093013010131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
50
|
Bell SM, Zhang L, Xu Y, Besnard V, Wert SE, Shroyer N, Whitsett JA. Kruppel-like factor 5 controls villus formation and initiation of cytodifferentiation in the embryonic intestinal epithelium. Dev Biol 2012; 375:128-39. [PMID: 23266329 DOI: 10.1016/j.ydbio.2012.12.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 11/29/2012] [Accepted: 12/14/2012] [Indexed: 12/16/2022]
Abstract
Kruppel-like factor 5 (Klf5) is a transcription factor expressed by embryonic endodermal progenitors that form the lining of the gastrointestinal tract. A Klf5 floxed allele was efficiently deleted from the intestinal epithelium by a Cre transgene under control of the Shh promoter resulting in the inhibition of villus morphogenesis and epithelial differentiation. Although proliferation of the intestinal epithelium was maintained, the expression of Elf3, Pparγ, Atoh1, Ascl2, Neurog3, Hnf4α, Cdx1, and other genes associated with epithelial cell differentiation was inhibited in the Klf5-deficient intestines. At E18.5, Klf5(Δ/Δ) fetuses lacked the apical brush border characteristic of enterocytes, and a loss of goblet and enteroendocrine cells was observed. The failure to form villi was not attributable to the absence of HH or PDGF signaling, known mediators of this developmental process. Klf5-deletion blocked the decrease in FoxA1 and Sox9 expression that accompanies normal villus morphogenesis. KLF5 directly inhibited activity of the FoxA1 promoter, and in turn FOXA1 inhibited Elf3 gene expression in vitro, linking the observed loss of Elf3 with the persistent expression of FoxA1 observed in Klf5-deficient mice. Genetic network analysis identified KLF5 as a key transcription factor regulating intestinal cell differentiation and cell adhesion. These studies indicate a novel requirement for KLF5 to initiate morphogenesis of the early endoderm into a compartmentalized intestinal epithelium comprised of villi and terminally differentiated cells.
Collapse
Affiliation(s)
- Sheila M Bell
- Perinatal Institute, Divisions of Neonatology-Perinatal-Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | |
Collapse
|