1
|
Cai S, Yin N. Single-cell transcriptome and chromatin accessibility mapping of upper lip and primary palate fusion. J Cell Mol Med 2024; 28:e70128. [PMID: 39392189 PMCID: PMC11467802 DOI: 10.1111/jcmm.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/17/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
Cleft lip and/or primary palate (CL/P) represent a prevalent congenital malformation, the aetiology of which is highly intricate. Although it is generally accepted that the condition arises from failed fusion between the upper lip and primary palate, the precise mechanism underlying this fusion process remains enigmatic. In this study, we utilized transposase-accessible chromatin sequencing (scATAC-seq) and single-cell RNA sequencing (scRNA-seq) to interrogate lambdoidal junction tissue derived from C57BL/6J mouse embryos at critical stages of embryogenesis (10.5, 11.5 and 12.5 embryonic days). We successfully identified distinct subgroups of mesenchymal and ectodermal cells involved in the fusion process and characterized their unique transcriptional profiles. Furthermore, we conducted cell differentiation trajectory analysis, revealing a dynamic repertoire of genes that are sequentially activated or repressed during pseudotime, facilitating the transition of relevant cell types. Additionally, we employed scATAC data to identify key genes associated with the fusion process and demonstrated differential chromatin accessibility across major cell types. Finally, we constructed a dynamic intercellular communication network and predicted upstream transcriptional regulators of critical genes involved in important signalling pathways. Our findings provide a valuable resource for future studies on upper lip and primary palate development, as well as congenital defects.
Collapse
Affiliation(s)
- Sini Cai
- The Department of Cleft Lip and Palate of Plastic Surgery HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Medical Cosmetic Center of Dermatology Hospital of Southern Medical UniversityGuangdong Provincial Dermatology HospitalGuangzhouChina
| | - Ningbei Yin
- The Department of Cleft Lip and Palate of Plastic Surgery HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Deng Z, Carpinelli MR, Butt T, Magor GW, Zhao P, Gillinder KR, Perkins AC, Jane SM. Single-cell transcriptomics reveals the cellular identity of a novel progenitor population crucial for murine neural tube closure. Heliyon 2024; 10:e37259. [PMID: 39296075 PMCID: PMC11408003 DOI: 10.1016/j.heliyon.2024.e37259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Neural tube closure in vertebrates is achieved through a highly dynamic and coordinated series of morphogenic events involving neuroepithelium, surface ectoderm, and neural plate border. Failure of this process in the caudal region causes spina bifida. Grainyhead-like 3 (GRHL3) is an indispensable transcription factor for neural tube closure as constitutive inactivation of the Grhl3 gene in mice leads to fully penetrant spina bifida. Here, through single-cell transcriptomics we show that at E8.5, the time-point preceding mouse neural tube closure, co-expression of Grhl3, Tfap2a, and Tfap2c defines a previously unrecognised progenitor population of surface ectoderm integral for neural tube closure. Deletion of Grhl3 expression in this cell population using a Tfap2a-Cre transgene recapitulates the spina bifida observed in Grhl3-null animals. Moreover, conditional inactivation of Tfap2c expression in Grhl3-expressing neural plate border cells also induces spina bifida. These findings indicate that a specific neural plate border cellular cohort is required for the early-stage neurulation.
Collapse
Affiliation(s)
- Zihao Deng
- Department of Medicine (Alfred Hospital), School of Translational Medicine, Monash University, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
| | - Marina R Carpinelli
- Department of Medicine (Alfred Hospital), School of Translational Medicine, Monash University, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
| | - Tariq Butt
- Department of Medicine (Alfred Hospital), School of Translational Medicine, Monash University, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
| | - Graham W Magor
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, QLD, 4006, Australia
| | - Peinan Zhao
- Department of Medicine (Alfred Hospital), School of Translational Medicine, Monash University, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
| | - Kevin R Gillinder
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
- Institute of Genetic Medicine and North-East England Stem Cell Institute, Centre for Life, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, United Kingdom
| | - Andrew C Perkins
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
| | - Stephen M Jane
- Department of Medicine (Alfred Hospital), School of Translational Medicine, Monash University, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
| |
Collapse
|
3
|
Fukagawa A, Hama N, Totoki Y, Nakamura H, Arai Y, Saito-Adachi M, Maeshima A, Matsui Y, Yachida S, Ushiku T, Shibata T. Genomic and epigenomic integrative subtypes of renal cell carcinoma in a Japanese cohort. Nat Commun 2023; 14:8383. [PMID: 38104198 PMCID: PMC10725467 DOI: 10.1038/s41467-023-44159-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023] Open
Abstract
Renal cell carcinoma (RCC) comprises several histological types characterised by different genomic and epigenomic aberrations; however, the molecular pathogenesis of each type still requires further exploration. We perform whole-genome sequencing of 128 Japanese RCC cases of different histology to elucidate the significant somatic alterations and mutagenesis processes. We also perform transcriptomic and epigenomic sequencing to identify distinguishing features, including assay for transposase-accessible chromatin sequencing (ATAC-seq) and methyl sequencing. Genomic analysis reveals that the mutational signature differs among the histological types, suggesting that different carcinogenic factors drive each histology. From the ATAC-seq results, master transcription factors are identified for each histology. Furthermore, clear cell RCC is classified into three epi-subtypes, one of which expresses highly immune checkpoint molecules with frequent loss of chromosome 14q. These genomic and epigenomic features may lead to the development of effective therapeutic strategies for RCC.
Collapse
Affiliation(s)
- Akihiko Fukagawa
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Natsuko Hama
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasushi Totoki
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
- Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiromi Nakamura
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasuhito Arai
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Mihoko Saito-Adachi
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Akiko Maeshima
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshiyuki Matsui
- Department of Urology, National Cancer Center Hospital, Tokyo, Japan
| | - Shinichi Yachida
- Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan.
- Laboratory of Molecular Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
4
|
Proft S, Leiz J, Heinemann U, Seelow D, Schmidt-Ott KM, Rutkiewicz M. Discovery of a non-canonical GRHL1 binding site using deep convolutional and recurrent neural networks. BMC Genomics 2023; 24:736. [PMID: 38049725 PMCID: PMC10696883 DOI: 10.1186/s12864-023-09830-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/22/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Transcription factors regulate gene expression by binding to transcription factor binding sites (TFBSs). Most models for predicting TFBSs are based on position weight matrices (PWMs), which require a specific motif to be present in the DNA sequence and do not consider interdependencies of nucleotides. Novel approaches such as Transcription Factor Flexible Models or recurrent neural networks consequently provide higher accuracies. However, it is unclear whether such approaches can uncover novel non-canonical, hitherto unexpected TFBSs relevant to human transcriptional regulation. RESULTS In this study, we trained a convolutional recurrent neural network with HT-SELEX data for GRHL1 binding and applied it to a set of GRHL1 binding sites obtained from ChIP-Seq experiments from human cells. We identified 46 non-canonical GRHL1 binding sites, which were not found by a conventional PWM approach. Unexpectedly, some of the newly predicted binding sequences lacked the CNNG core motif, so far considered obligatory for GRHL1 binding. Using isothermal titration calorimetry, we experimentally confirmed binding between the GRHL1-DNA binding domain and predicted GRHL1 binding sites, including a non-canonical GRHL1 binding site. Mutagenesis of individual nucleotides revealed a correlation between predicted binding strength and experimentally validated binding affinity across representative sequences. This correlation was neither observed with a PWM-based nor another deep learning approach. CONCLUSIONS Our results show that convolutional recurrent neural networks may uncover unanticipated binding sites and facilitate quantitative transcription factor binding predictions.
Collapse
Affiliation(s)
- Sebastian Proft
- Exploratory Diagnostic Sciences, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany
| | - Janna Leiz
- Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany
- Department of Nephrology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203, Berlin, Germany
- Molecular and Translational Kidney Research, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
| | - Udo Heinemann
- Macromolecular Structure and Interaction, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany.
| | - Dominik Seelow
- Exploratory Diagnostic Sciences, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany.
- Institute of Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353, Berlin, Germany.
| | - Kai M Schmidt-Ott
- Department of Nephrology and Hypertension, Hannover Medical School, 30625, Hannover, Germany.
- Department of Nephrology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203, Berlin, Germany.
- Molecular and Translational Kidney Research, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany.
| | - Maria Rutkiewicz
- Macromolecular Structure and Interaction, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125, Berlin, Germany
- Department of Structural Biology of Eukaryotes, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznań, 61-704, Poland
| |
Collapse
|
5
|
Forrester-Gauntlett B, Peters L, Oback B. Grainyhead-like 2 is required for morphological integrity of mouse embryonic stem cells and orderly formation of inner ear-like organoids. Front Cell Dev Biol 2023; 11:1112069. [PMID: 37745294 PMCID: PMC10513505 DOI: 10.3389/fcell.2023.1112069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Mutations in the transcription factor gene grainyhead-like 2 (GRHL2) are associated with progressive non-syndromic sensorineural deafness autosomal dominant type 28 (DFNA28) in humans. Since complete loss of Grhl2 is lethal in mouse embryos, we studied its role during inner ear pathology and hearing loss in vitro. To this end, we generated different homozygous deletions to knockout Grhl2 in mouse embryonic stem cells (Grhl2-KO ESCs), including some mimicking naturally occurring truncations in the dimerisation domain related to human DFNA28. Under naïve culture conditions, Grhl2-KO cells in suspension were more heterogenous in size and larger than wild-type controls. Adherent Grhl2-KO cells were also larger, with a less uniform shape, flattened, less circular morphology, forming loose monolayer colonies with poorly defined edges. These changes correlated with lower expression of epithelial cadherin Cdh1 but no changes in tight junction markers (Ocln, Tjp2) or other Grhl isoforms (Grhl1, Grhl3). Clonogenicity from single cells, proliferation rates of cell populations and proliferation markers were reduced in Grhl2-KO ESCs. We next induced stepwise directed differentiation of Grhl2-KO ESCs along an otic pathway, giving rise to three-dimensional inner ear-like organoids (IELOs). Quantitative morphometry revealed that Grhl2-KO cells initially formed larger IELOs with a less compacted structure, more eccentric shape and increased surface area. These morphological changes persisted for up to one week. They were partially rescued by forced cell aggregation and fully restored by stably overexpressing exogenous Grhl2 in Grhl2-KO ESCs, indicating that Grhl2 alters cell-cell interactions. On day 8, aggregates were transferred into minimal maturation medium to allow self-guided organogenesis for another two weeks. During this period, Grhl2-KO cells and wild-type controls developed similarly, expressing neural, neuronal and sensory hair cell markers, while maintaining their initial differences in size and shape. In summary, Grhl2 is required for morphological maintenance of ESCs and orderly formation of IELOs, consistent with an essential role in organising epithelial integrity during inner ear development. Our findings validate quantitative morphometry as a useful, non-invasive screening method for molecular phenotyping of candidate mutations during organoid development.
Collapse
Affiliation(s)
- Blaise Forrester-Gauntlett
- Animal Biotech, AgResearch, Hamilton, New Zealand
- School of Science, University of Waikato, Hamilton, New Zealand
| | - Linda Peters
- School of Science, University of Waikato, Hamilton, New Zealand
| | - Björn Oback
- Animal Biotech, AgResearch, Hamilton, New Zealand
- School of Science, University of Waikato, Hamilton, New Zealand
- School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
6
|
Crane-Smith Z, De Castro SCP, Nikolopoulou E, Wolujewicz P, Smedley D, Lei Y, Mather E, Santos C, Hopkinson M, Pitsillides AA, Finnell RH, Ross ME, Copp AJ, Greene NDE. A non-coding insertional mutation of Grhl2 causes gene over-expression and multiple structural anomalies including cleft palate, spina bifida and encephalocele. Hum Mol Genet 2023; 32:2681-2692. [PMID: 37364051 PMCID: PMC10460492 DOI: 10.1093/hmg/ddad094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/19/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
Orofacial clefts, including cleft lip and palate (CL/P) and neural tube defects (NTDs) are among the most common congenital anomalies, but knowledge of the genetic basis of these conditions remains incomplete. The extent to which genetic risk factors are shared between CL/P, NTDs and related anomalies is also unclear. While identification of causative genes has largely focused on coding and loss of function mutations, it is hypothesized that regulatory mutations account for a portion of the unidentified heritability. We found that excess expression of Grainyhead-like 2 (Grhl2) causes not only spinal NTDs in Axial defects (Axd) mice but also multiple additional defects affecting the cranial region. These include orofacial clefts comprising midline cleft lip and palate and abnormalities of the craniofacial bones and frontal and/or basal encephalocele, in which brain tissue herniates through the cranium or into the nasal cavity. To investigate the causative mutation in the Grhl2Axd strain, whole genome sequencing identified an approximately 4 kb LTR retrotransposon insertion that disrupts the non-coding regulatory region, lying approximately 300 base pairs upstream of the 5' UTR. This insertion also lies within a predicted long non-coding RNA, oriented on the reverse strand, which like Grhl2 is over-expressed in Axd (Grhl2Axd) homozygous mutant embryos. Initial analysis of the GRHL2 upstream region in individuals with NTDs or cleft palate revealed rare or novel variants in a small number of cases. We hypothesize that mutations affecting the regulation of GRHL2 may contribute to craniofacial anomalies and NTDs in humans.
Collapse
Affiliation(s)
- Zoe Crane-Smith
- Developmental Biology and Cancer Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sandra C P De Castro
- Developmental Biology and Cancer Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Evanthia Nikolopoulou
- Developmental Biology and Cancer Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Paul Wolujewicz
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - Damian Smedley
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Yunping Lei
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Emma Mather
- Developmental Biology and Cancer Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Chloe Santos
- Developmental Biology and Cancer Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Mark Hopkinson
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Andrew A Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | | | - Richard H Finnell
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - M Elisabeth Ross
- Center for Neurogenetics, Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065, USA
| | - Andrew J Copp
- Developmental Biology and Cancer Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Nicholas D E Greene
- Developmental Biology and Cancer Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| |
Collapse
|
7
|
Deng Z, Butt T, Arhatari BD, Darido C, Auden A, Swaroop D, Partridge DD, Haigh K, Nguyen T, Haigh JJ, Carpinelli MR, Jane SM. Dysregulation of Grainyhead-like 3 expression causes widespread developmental defects. Dev Dyn 2023; 252:647-667. [PMID: 36606449 PMCID: PMC10952483 DOI: 10.1002/dvdy.565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The gene encoding the transcription factor, Grainyhead-like 3 (Grhl3), plays critical roles in mammalian development and homeostasis. Grhl3-null embryos exhibit thoraco-lumbo-sacral spina bifida and soft-tissue syndactyly. Additional studies reveal that these embryos also exhibit an epidermal proliferation/differentiation imbalance. This manifests as skin barrier defects resulting in peri-natal lethality and defective wound repair. Despite these extensive analyses of Grhl3 loss-of-function models, the consequences of gain-of-function of this gene have been difficult to achieve. RESULTS In this study, we generated a novel mouse model that expresses Grhl3 from a transgene integrated in the Rosa26 locus on an endogenous Grhl3-null background. Expression of the transgene rescues both the neurulation and skin barrier defects of the knockout mice, allowing survival into adulthood. Despite this, the mice are not normal, exhibiting a range of phenotypes attributable to dysregulated Grhl3 expression. In mice homozygous for the transgene, we observe a severe Shaker-Waltzer phenotype associated with hearing impairment. Micro-CT scanning of the inner ear revealed profound structural alterations underlying these phenotypes. In addition, these mice exhibit other developmental anomalies including hair loss, digit defects, and epidermal dysmorphogenesis. CONCLUSION Taken together, these findings indicate that diverse developmental processes display low tolerance to dysregulation of Grhl3.
Collapse
Affiliation(s)
- Zihao Deng
- Department of Medicine (Alfred Hospital), Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Tariq Butt
- Department of Medicine (Alfred Hospital), Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Benedicta D. Arhatari
- ARC Centre of Excellence in Advanced Molecular Imaging, Department of Chemistry and PhysicsLa Trobe UniversityBundooraVictoriaAustralia
- Australian Synchrotron, ANSTOClaytonVictoriaAustralia
| | - Charbel Darido
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Alana Auden
- Department of Medicine (Alfred Hospital), Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Dijina Swaroop
- Department of Medicine (Alfred Hospital), Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Darren D. Partridge
- Department of Medicine (Alfred Hospital), Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Katharina Haigh
- Department of Pharmacology and Therapeutics, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- Research Institute in Oncology and HematologyCancerCare ManitobaWinnipegManitobaCanada
| | - Thao Nguyen
- Australian Centre for Blood Diseases, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Jody J. Haigh
- Department of Pharmacology and Therapeutics, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- Research Institute in Oncology and HematologyCancerCare ManitobaWinnipegManitobaCanada
| | - Marina R. Carpinelli
- Department of Medicine (Alfred Hospital), Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Stephen M. Jane
- Department of Medicine (Alfred Hospital), Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
8
|
Georgy SR, Rudiatmoko DR, Auden A, Partridge D, Butt T, Srivastava S, Wong N, Swaroop D, Carpinelli MR, Bogeski M, Jane SM. Identification of a Novel GRHL3/HOPX/Wnt/β-Catenin Proto-oncogenic Axis in Squamous Cell Carcinoma of the Esophagus. Cell Mol Gastroenterol Hepatol 2022; 15:1051-1069. [PMID: 36442813 PMCID: PMC10036738 DOI: 10.1016/j.jcmgh.2022.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND & AIMS Esophageal squamous cell carcinoma (ESCC) is an aggressive malignancy with a poor long-term prognosis. The molecular mechanisms underlying the initiation and progression of this tumor are largely unknown. The transcription factor GRHL3 functions as a potent tumor suppressor in SCC of skin, head, and neck. This study aims to determine whether GRHL3 also plays a role in the homeostasis of the esophageal epithelium and in the development of ESCC. METHODS The effects of Grhl3 deletion on squamous epithelial homeostasis in embryos and adult mice were examined using immunohistochemistry, transmission electron microscopy, and real-time polymerase chain reaction. The conditionally deleted mice were subsequently used to determine susceptibility to ESCC. Whole-transcriptome sequencing (RNA-seq) was performed on ESCC in wild-type and Grhl3 deleted animals. To decipher the signaling pathways, real-time polymerase chain reaction, immunohistochemistry, analysis of chromatin immunoprecipitation sequencing, chromatin immunoprecipitation-polymerase chain reaction, and RNA seq datasets were used. Primary human samples were used to validate the findings in the mouse model. RESULTS Loss of Grhl3 perturbs the proliferation-differentiation balance in the esophageal epithelium, thereby increasing the susceptibility to esophageal carcinogenesis in adult mice. Grhl3 imparts its tumor suppressor function by regulating the expression of HOPX. We have identified the Wnt/β-catenin pathway as the downstream effectors of GRHL3 and HOPX through our integrated approach using patient-derived ESCC samples and mouse models. CONCLUSIONS GRHL3 conveys its tumor suppressor function in ESCC through regulating its target gene HOPX, which limits Wnt/β-catenin signaling. Targeted therapies to inhibit this pathway could be a potential treatment strategy for ESCC patients with reduced GRHL3 expression.
Collapse
Affiliation(s)
- Smitha Rose Georgy
- Department of Anatomic Pathology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Werribee, Australia.
| | | | - Alana Auden
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Darren Partridge
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Tariq Butt
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Seema Srivastava
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Nick Wong
- Monash Bioinformatics Platform, Central Clinical School, Melbourne, Australia
| | - Dijina Swaroop
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Marina Rose Carpinelli
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Mirjana Bogeski
- Department of Anatomic Pathology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Werribee, Australia
| | - Stephen M Jane
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
9
|
GRHL2 Enhances Phosphorylated Estrogen Receptor (ER) Chromatin Binding and Regulates ER-Mediated Transcriptional Activation and Repression. Mol Cell Biol 2022; 42:e0019122. [PMID: 36036613 PMCID: PMC9584124 DOI: 10.1128/mcb.00191-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Phosphorylation of estrogen receptor α (ER) at serine 118 (pS118-ER) is induced by estrogen and is the most abundant posttranslational mark associated with a transcriptionally active receptor. Cistromic analysis of pS118-ER from our group revealed enrichment of the GRHL2 motif near pS118-ER binding sites. In this study, we used cistromic and transcriptomic analyses to interrogate the relationship between GRHL2 and pS118-ER. We found that GRHL2 is bound to chromatin at pS118-ER/GRHL2 co-occupancy sites prior to ligand treatment, and GRHL2 binding is required for maximal pS118-ER recruitment. pS118-ER/GRHL2 co-occupancy sites were enriched at active enhancers marked by H3K27ac and H3K4me1, along with FOXA1 and p300, compared to sites where each factor binds independently. Transcriptomic analysis yielded four subsets of ER/GRHL2-coregulated genes revealing that GRHL2 can both enhance and antagonize E2-mediated ER transcriptional activity. Gene ontology analysis indicated that coregulated genes are involved in cell migration. Accordingly, knockdown of GRHL2, combined with estrogen treatment, resulted in increased cell migration but no change in proliferation. These results support a model in which GRHL2 binds to selected enhancers and facilitates pS118-ER recruitment to chromatin, which then results in differential activation and repression of genes that control estrogen-regulated ER-positive breast cancer cell migration.
Collapse
|
10
|
Du X, He X, Liu Q, Liu Q, Di R, Chu M. Identification of photoperiod-induced specific miRNAs in the adrenal glands of Sunite sheep (Ovis aries). Front Vet Sci 2022; 9:888207. [PMID: 35937294 PMCID: PMC9354845 DOI: 10.3389/fvets.2022.888207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022] Open
Abstract
In seasonal estrus, it is well known that melatonin-regulated biorhythm plays a key role. Some studies indicate that the adrenal gland plays an important role in reproduction in mammals, but the molecular mechanism is not clear. This study used an artificially controlled light photoperiod model, combined with RNA-seq technology and bioinformatics analysis, to analyze the messenger RNA (mRNA) and microRNA (miRNA) of ewe (Sunite) adrenal glands under different photoperiod treatments. After identification, the key candidate genes GRHL2, CENPF, FGF16 and SLC25A30 that photoperiod affects reproduction were confirmed. The miRNAs (oar-miR-544-3p, oar-miR-411b-5p, oar-miR-376e-3p, oar-miR-376d, oar-miR-376b-3p, oar-miR-376a-3p) were specifically expressed in the adrenal gland. The candidate mRNA-miRNA pairs (e.g., SLC25A30 coagulated by novel miRNA554, novel miRNA555 and novel miRNA559) may affect seasonal estrus. In summary, we constructed relation network of the mRNAs and miRNAs of sheep adrenal glands using RNA sequencing and bioinformatics analysis, thereby, providing a valuable genetic variation resource for sheep genome research, which will contribute to the study of complex traits in sheep.
Collapse
Affiliation(s)
- Xiaolong Du
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoyun He
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qingqing Liu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Qiuyue Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ran Di
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mingxing Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Mingxing Chu
| |
Collapse
|
11
|
Hua J, Ma C, Wang CH, Wang Y, Feng S, Xiao T, Zhu C. Abnormal GRHL2 Methylation Confers Malignant Progression to Acute Leukemia. Appl Bionics Biomech 2022; 2022:9708829. [PMID: 35855840 PMCID: PMC9288345 DOI: 10.1155/2022/9708829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/16/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose Abnormal methylation of Grainyhead-like 2 (GRHL2) is associated with a substantial role in the malignant phenotype of tumor patients. Our present research is aimed at studying the abnormal expression of GRHL2 and the association of methylation in patients with acute leukemia and its relationship with prognosis. Materials and Methods We used quantitative real-time polymerase chain reaction (qRT-PCR) for detecting the aberrant expression level of GRHL2 in 60 patients with acute leukemia and 60 normal controls. We analyzed the significant correlation between the expression level of GRHL2 with clinicopathological features and patients' prognosis in acute leukemia using the corresponding statistical methods. Secondly, we employed qRT-PCR and Western blotting to detect the mRNA and protein levels of GRHL2 in leukemia cell lines. Next, we used methylation-specific polymerase chain reaction (MSP) technology for detecting the methylation of GRHL2 in clinical samples with acute leukemia and cell lines. Then we investigated the demethylating effect of arsenic trioxide and 5-azacitidine on the mRNA and protein expression levels of GRHL2 in cell lines of acute leukemia. Finally, we studied the effects of arsenide trioxide and 5-azacitidine on the proliferation of leukemia cells and the TGF-β signaling pathway. Results We found a lower level of GRHL2 expression not only in acute leukemia patients but also in cell lines when compared with normal controls. At the same time, the expression level of GRHL2 in patients with acute leukemia was significantly correlated with leukocyte count, platelet count, and cytogenetic risk grouping. In addition, the lower GRHL2 expression group showed a significantly lower overall survival rate in acute leukemia patients than that of patients with a higher GRHL2 expression group. Univariate and multivariate analyses revealed that the expression of GRHL2 is an independent risk factor in acute leukemia patients. The methylation level of the GRHL2 promoter region in acute leukemia patients and cell lines was significantly higher than the normal control group, and we found the elevated mRNA and protein levels of GRHL2 in acute leukemia cell lines after the use of the demethylation drug arsenic trioxide and 5-azacitidine. At the same time, arsenide trioxide and 5-azacitidine are associated with the inhibition of cellular proliferation of acute leukemia cells and also promote the elevated expression of TGF-β signaling pathway-linked proteins, including TGF-β, Smad2, Smad3, and Smad4. Conclusion Increased expression and methylation level of GRHL2 are closely associated with the prognosis and malignant phenotype of acute leukemia patients and play an irreplaceable role in the occurrence and development of patients with acute leukemia.
Collapse
Affiliation(s)
- Jing Hua
- Department of Hematology, Shandong Provincial Qianfoshan Hospital, Shandong University, China
| | - Congcong Ma
- Department of Hematology, Liaocheng People's Hospital, Shandong University, China
| | - Chao Hui Wang
- Department of Hematology, Qingdao Haici Medical Group, China
| | - Yan Wang
- Department of Hematology, Shandong Provincial Qianfoshan Hospital, Shandong University, China
| | - Saran Feng
- Department of Hematology, Shandong Provincial Qianfoshan Hospital, Shandong University, China
| | - Taiwu Xiao
- Department of Hematology, Liaocheng People's Hospital, Shandong University, China
| | - ChuanSheng Zhu
- Department of Hematology, Shandong Provincial Qianfoshan Hospital, Shandong University, China
| |
Collapse
|
12
|
Smith RJ, Zhang H, Hu SS, Yung T, Francis R, Lee L, Onaitis MW, Dirks PB, Zang C, Kim TH. Single-cell chromatin profiling of the primitive gut tube reveals regulatory dynamics underlying lineage fate decisions. Nat Commun 2022; 13:2965. [PMID: 35618699 PMCID: PMC9135761 DOI: 10.1038/s41467-022-30624-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/06/2022] [Indexed: 01/07/2023] Open
Abstract
Development of the gastrointestinal system occurs after gut tube closure, guided by spatial and temporal control of gene expression. However, it remains unclear what forces regulate these spatiotemporal gene expression patterns. Here we perform single-cell chromatin profiling of the primitive gut tube to reveal organ-specific chromatin patterns that reflect the anatomical patterns of distinct organs. We generate a comprehensive map of epigenomic changes throughout gut development, demonstrating that dynamic chromatin accessibility patterns associate with lineage-specific transcription factor binding events to regulate organ-specific gene expression. Additionally, we show that loss of Sox2 and Cdx2, foregut and hindgut lineage-specific transcription factors, respectively, leads to fate shifts in epigenomic patterns, linking transcription factor binding, chromatin accessibility, and lineage fate decisions in gut development. Notably, abnormal expression of Sox2 in the pancreas and intestine impairs lineage fate decisions in both development and adult homeostasis. Together, our findings define the chromatin and transcriptional mechanisms of organ identity and lineage plasticity in development and adult homeostasis.
Collapse
Affiliation(s)
- Ryan J Smith
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Hongpan Zhang
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Shengen Shawn Hu
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Theodora Yung
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Roshane Francis
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Lilian Lee
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Mark W Onaitis
- Division of Cardiovascular and Thoracic Surgery, University of California San Diego Medical Center, San Diego, CA, USA
| | - Peter B Dirks
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Chongzhi Zang
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA.
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA.
| | - Tae-Hee Kim
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
13
|
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: 16] [Impact Index Per Article: 5.3] [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
|
14
|
Huang W, He Q, Li M, Ding Y, Liang W, Li W, Lin J, Zhao H, Chen F. Two rare variants reveal the significance of Grainyhead‐like 3 Arginine 391 underlying non‐syndromic cleft palate only. Oral Dis 2022; 29:1632-1643. [PMID: 35189007 DOI: 10.1111/odi.14164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Non-syndromic cleft palate only (NSCPO) is one of the most common craniofacial birth defects with largely undetermined genetic etiology. It has been established that Grainyhead-like 3 (GRHL3) plays an essential role in the pathogenesis of NSCPO. This study aimed to identify and verify the first-reported GRHL3 variant underlying NSCPO among the Chinese cohort. METHODS We performed whole-exome sequencing (WES) on a Chinese NSCPO patient and identified a rare variant of GRHL3 (p.Arg391His). A validated deleterious variant p.Arg391Cys was introduced as a positive control. Zebrafish embryos injection, reporter assays, live-cell imaging, and RNA sequencing were conducted to test the pathogenicity of the variants. RESULTS Zebrafish embryos microinjection demonstrated that overexpression of the variants could disrupt the normal development of zebrafish embryos. Reporter assays showed that Arg391His disturbed transcriptional activity of GRHL3 and exerted a dominant-negative effect. Interestingly, Arg391His and Arg391Cys displayed distinct nuclear localization patterns from that of wild-type GRHL3 in live-cell imaging. Bulk RNA sequencing suggested that the two variants changed the pattern of gene expression. CONCLUSIONS In aggregate, this study identified and characterized a rare GRHL3 variant in NSCPO, revealing the critical role of Arginine 391 in GRHL3. Our findings will help facilitate understanding and genetic counseling of NSCPO.
Collapse
Affiliation(s)
- Wenbin Huang
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Qing He
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi’an Jiaotong University Health Science Center 710061 Xi’an, Shaanxi China
| | - Mingzhao Li
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Yi Ding
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi’an Jiaotong University Health Science Center 710061 Xi’an, Shaanxi China
| | - Wei Liang
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Weiran Li
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Jiuxiang Lin
- Department of Orthodontics Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| | - Huaxiang Zhao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research College of Stomatology Xi’an Jiaotong University 710004 Xi'an, Shaanxi China
- Department of Orthodontics College of Stomatology Xi’an Jiaotong University 710004 Xi’an, Shaanxi China
| | - Feng Chen
- Central laboratory Peking University School and Hospital of Stomatology 100081 Beijing China
- National Center of Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials 100081 Beijing China
| |
Collapse
|
15
|
Grigolon G, Araldi E, Erni R, Wu JY, Thomas C, La Fortezza M, Laube B, Pöhlmann D, Stoffel M, Zarse K, Carreira EM, Ristow M, Fischer F. Grainyhead 1 acts as a drug-inducible conserved transcriptional regulator linked to insulin signaling and lifespan. Nat Commun 2022; 13:107. [PMID: 35013237 PMCID: PMC8748497 DOI: 10.1038/s41467-021-27732-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is impacted by interventions across species, often converging on metabolic pathways. Transcription factors regulate longevity yet approaches for their pharmacological modulation to exert geroprotection remain sparse. We show that increased expression of the transcription factor Grainyhead 1 (GRH-1) promotes lifespan and pathogen resistance in Caenorhabditis elegans. A compound screen identifies FDA-approved drugs able to activate human GRHL1 and promote nematodal GRH-1-dependent longevity. GRHL1 activity is regulated by post-translational lysine methylation and the phosphoinositide (PI) 3-kinase C2A. Consistently, nematodal longevity following impairment of the PI 3-kinase or insulin/IGF-1 receptor requires grh-1. In BXD mice, Grhl1 expression is positively correlated with lifespan and insulin sensitivity. In humans, GRHL1 expression positively correlates with insulin receptor signaling and also with lifespan. Fasting blood glucose levels, including in individuals with type 2 diabetes, are negatively correlated with GRHL1 expression. Thereby, GRH-1/GRHL1 is identified as a pharmacologically malleable transcription factor impacting insulin signaling and lifespan. Life- and healthspan of organisms can be modulated by dietary, genetic, or pharmacological interventions, which often affect metabolic pathways. Here the authors report that Grainyhead 1 is an evolutionarily conserved, drug-inducible transcription factor that promotes longevity in C. elegans, and thus a potential target for the development of geroprotective drugs.
Collapse
Affiliation(s)
- Giovanna Grigolon
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Elisa Araldi
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland.,Metabolism and Metabolic Disease Laboratory, Institute for Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8093, Switzerland
| | - Reto Erni
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8093, Switzerland
| | - Jia Yee Wu
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Carolin Thomas
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Marco La Fortezza
- Evolutionary Biology Laboratory, Department of Environmental Systems Science, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8092, Switzerland
| | - Beate Laube
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Doris Pöhlmann
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Markus Stoffel
- Metabolism and Metabolic Disease Laboratory, Institute for Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8093, Switzerland
| | - Kim Zarse
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| | - Erick M Carreira
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, CH-8093, Switzerland
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland.
| | - Fabian Fischer
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, CH-8603, Switzerland
| |
Collapse
|
16
|
Kotarba G, Taracha-Wisniewska A, Miller M, Dabrowski M, Wilanowski T. Transcription factors Krüppel-like factor 4 and paired box 5 regulate the expression of the Grainyhead-like genes. PLoS One 2021; 16:e0257977. [PMID: 34570823 PMCID: PMC8476022 DOI: 10.1371/journal.pone.0257977] [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: 04/20/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
Genes from the Grainyhead-like (GRHL) family code for transcription factors necessary for the development and maintenance of various epithelia. These genes are also very important in the development of many types of cancer. However, little is known about the regulation of expression of GRHL genes. Previously, there were no systematic analyses of the promoters of GRHL genes or transcription factors that bind to these promoters. Here we report that the Krüppel-like factor 4 (KLF4) and the paired box 5 factor (PAX5) bind to the regulatory regions of the GRHL genes and regulate their expression. Ectopic expression of KLF4 or PAX5 alters the expression of GRHL genes. In KLF4-overexpressing HEK293 cells, the expression of GRHL1 and GRHL3 genes was upregulated by 32% and 60%, respectively, whereas the mRNA level of GRHL2 gene was lowered by 28% when compared to the respective controls. The levels of GRHL1 and GRHL3 expression were decreased by 30% or 33% in PAX5-overexpressing HEK293 cells. The presence of minor frequency allele of single nucleotide polymorphism rs115898376 in the promoter of the GRHL1 gene affected the binding of KLF4 to this site. The evidence presented here suggests an important role of KLF4 and PAX5 in the regulation of expression of GRHL1-3 genes.
Collapse
Affiliation(s)
- Grzegorz Kotarba
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Warsaw, Poland
| | | | - Michal Miller
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Michal Dabrowski
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Tomasz Wilanowski
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Warsaw, Poland
- * E-mail:
| |
Collapse
|
17
|
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: 2.5] [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
|
18
|
Głowacka A, Bieganowski P, Jurewicz E, Leśniak W, Wilanowski T, Filipek A. Regulation of S100A10 Gene Expression. Biomolecules 2021; 11:biom11070974. [PMID: 34356598 PMCID: PMC8301800 DOI: 10.3390/biom11070974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 01/18/2023] Open
Abstract
S100A10, a member of the S100 family of Ca2+-binding proteins, is a widely distributed protein involved in many cellular and extracellular processes. The best recognized role of S100A10 is the regulation, via interaction with annexin A2, of plasminogen conversion to plasmin. Plasmin, together with other proteases, induces degradation of the extracellular matrix (ECM), which is an important step in tumor progression. Additionally, S100A10 interacts with 5-hydroxytryptamine 1B (5-HT1B) receptor, which influences neurotransmitter binding and, through that, depressive symptoms. Taking this into account, it is evident that S100A10 expression in the cell should be under strict control. In this work, we summarize available literature data concerning the physiological stimuli and transcription factors that influence S100A10 expression. We also present our original results showing for the first time regulation of S100A10 expression by grainyhead-like 2 transcription factor (GRHL2). By applying in silico analysis, we have found two highly conserved GRHL2 binding sites in the 1st intron of the gene encoding S100A10 protein. Using chromatin immunoprecipitation (ChIP) and luciferase assays, we have shown that GRHL2 directly binds to these sites and that this DNA region can affect transcription of S100A10.
Collapse
Affiliation(s)
- Aleksandra Głowacka
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., 02-093 Warsaw, Poland; (A.G.); (E.J.); (W.L.)
| | - Paweł Bieganowski
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland;
| | - Ewelina Jurewicz
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., 02-093 Warsaw, Poland; (A.G.); (E.J.); (W.L.)
| | - Wiesława Leśniak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., 02-093 Warsaw, Poland; (A.G.); (E.J.); (W.L.)
| | - Tomasz Wilanowski
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, 1 Miecznikowa Str., 02-096 Warsaw, Poland
- Correspondence: (T.W.); (A.F.); Tel.: +48-22-589-23-32 (A.F.); Fax: +48-22-822-53-42 (A.F.)
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., 02-093 Warsaw, Poland; (A.G.); (E.J.); (W.L.)
- Correspondence: (T.W.); (A.F.); Tel.: +48-22-589-23-32 (A.F.); Fax: +48-22-822-53-42 (A.F.)
| |
Collapse
|
19
|
Jaffe E, Niswander L. Loss of Grhl3 is correlated with altered cellular protrusions in the non-neural ectoderm during neural tube closure. Dev Dyn 2021; 250:732-744. [PMID: 33378081 DOI: 10.1002/dvdy.292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The transcription factor Grainyhead-like 3 (GRHL3) has multiple roles in a variety of tissues during development including epithelial patterning and actin cytoskeletal regulation. During neural tube closure (NTC) in the mouse embryo, GRHL3 is expressed and functions in the non-neural ectoderm (NNE). Two important functions of GRHL3 are regulating the actin cytoskeleton during NTC and regulating the boundary between the NNE and neural ectoderm. However, an open question that remains is whether these functions explain the caudally restricted neural tube defect (NTD) of spina bifida observed in Grhl3 mutants. RESULTS Using scanning electron microscopy and immunofluorescence based imaging on Grhl3 mutants and wildtype controls, we show that GRHL3 is dispensable for NNE identity or epithelial maintenance in the caudal NNE but is needed for regulation of cellular protrusions during NTC. Grhl3 mutants have decreased lamellipodia relative to wildtype embryos during caudal NTC, first observed at the onset of delays when lamellipodia become prominent in wildtype embryos. At the axial level of NTD, half of the mutants show increased and disorganized filopodia and half lack cellular protrusions. CONCLUSION These data suggest that altered cellular protrusions during NTC contribute to the etiology of NTD in Grhl3 mutants.
Collapse
Affiliation(s)
- Eric Jaffe
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Lee Niswander
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
20
|
He Y, Gan M, Wang Y, Huang T, Wang J, Han T, Yu B. EGFR-ERK induced activation of GRHL1 promotes cell cycle progression by up-regulating cell cycle related genes in lung cancer. Cell Death Dis 2021; 12:430. [PMID: 33931584 PMCID: PMC8087693 DOI: 10.1038/s41419-021-03721-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 11/09/2022]
Abstract
Grainyhead-like 1 (GRHL1) is a transcription factor involved in embryonic development. However, little is known about the biological functions of GRHL1 in cancer. In this study, we found that GRHL1 was upregulated in non-small cell lung cancer (NSCLC) and correlated with poor survival of patients. GRHL1 overexpression promoted the proliferation of NSCLC cells and knocking down GRHL1 inhibited the proliferation. RNA sequencing showed that a series of cell cycle-related genes were altered when knocking down GRHL1. We further demonstrated that GRHL1 could regulate the expression of cell cycle-related genes by binding to the promoter regions and increasing the transcription of the target genes. Besides, we also found that EGF stimulation could activate GRHL1 and promoted its nuclear translocation. We identified the key phosphorylation site at Ser76 on GRHL1 that is regulated by the EGFR-ERK axis. Taken together, these findings elucidate a new function of GRHL1 on regulating the cell cycle progression and point out the potential role of GRHL1 as a drug target in NSCLC.
Collapse
Affiliation(s)
- Yiming He
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Mingxi Gan
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330031, P. R. China
| | - Yanan Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330031, P. R. China
| | - Tong Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China
| | - Jianbin Wang
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330031, P. R. China.
| | - Tianyu Han
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China.
| | - Bentong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, P. R. China.
| |
Collapse
|
21
|
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: 7] [Impact Index Per Article: 1.8] [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
|
22
|
de Vries M, Owens HG, Carpinelli MR, Partridge D, Kersbergen A, Sutherland KD, Auden A, Anderson PJ, Jane SM, Dworkin S. Delineating the roles of Grhl2 in craniofacial development through tissue-specific conditional deletion and epistasis approaches in mouse. Dev Dyn 2021; 250:1191-1209. [PMID: 33638290 DOI: 10.1002/dvdy.322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/31/2021] [Accepted: 02/20/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The highly conserved Grainyhead-like (Grhl) family of transcription factors play critical roles in the development of the neural tube and craniofacial skeleton. In particular, deletion of family member Grainyhead-like 2 (Grhl2) leads to mid-gestational embryonic lethality, maxillary clefting, abdominoschisis, and both cranial and caudal neural tube closure defects. These highly pleiotropic and systemic defects suggest that Grhl2 plays numerous critical developmental roles to ensure correct morphogenesis and patterning. RESULTS Here, using four separate Cre-lox conditional deletion models, as well as one genetic epistasis approach (Grhl2+/- ;Edn1+/- double heterozygous mice) we have investigated tissue-specific roles of Grhl2 in embryonic development, with a particular focus on the craniofacial skeleton. We find that loss of Grhl2 in the pharyngeal epithelium (using the ShhCre driver) leads to low-penetrance micrognathia, whereas deletion of Grhl2 within the ectoderm of the pharynx (NestinCre ) leads to small, albeit significant, differences in the proximal-distal elongation of both the maxilla and mandible. Loss of Grhl2 in endoderm (Sox17-2aiCre ) resulted in noticeable lung defects and a single instance of secondary palatal clefting, although formation of other endoderm-derived organs such as the stomach, bladder and intestines was not affected. Lastly, deletion of Grhl2 in cells of the neural crest (Wnt1Cre ) did not lead to any discernible defects in craniofacial development, and similarly, our epistasis approach did not detect any phenotypic consequences of loss of a single allele of both Grhl2 and Edn1. CONCLUSION Taken together, our study identifies a pharyngeal-epithelium intrinsic, non-cell-autonomous role for Grhl2 in the patterning and formation of the craniofacial skeleton, as well as an endoderm-specific role for Grhl2 in the formation and establishment of the mammalian lung.
Collapse
Affiliation(s)
- Michael de Vries
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Harley G Owens
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, Australia
| | - Marina R Carpinelli
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, Australia
| | - Darren Partridge
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, Australia
| | - Ariena Kersbergen
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kate D Sutherland
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Alana Auden
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, Australia
| | - Peter J Anderson
- Australian Craniofacial Unit, Women and Children's Hospital, Adelaide, South Australia, Australia.,Faculty of Health Sciences, University of Adelaide, South Australia, Australia.,Nanjing Medical University, Nanjing, China
| | - Stephen M Jane
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, Australia
| | - Sebastian Dworkin
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
23
|
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: 3.0] [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
|
24
|
Abstract
During embryonic development, the central nervous system forms as the neural plate and then rolls into a tube in a complex morphogenetic process known as neurulation. Neural tube defects (NTDs) occur when neurulation fails and are among the most common structural birth defects in humans. The frequency of NTDs varies greatly anywhere from 0.5 to 10 in 1000 live births, depending on the genetic background of the population, as well as a variety of environmental factors. The prognosis varies depending on the size and placement of the lesion and ranges from death to severe or moderate disability, and some NTDs are asymptomatic. This chapter reviews how mouse models have contributed to the elucidation of the genetic, molecular, and cellular basis of neural tube closure, as well as to our understanding of the causes and prevention of this devastating birth defect.
Collapse
Affiliation(s)
- Irene E Zohn
- Center for Genetic Medicine, Children's Research Institute, Children's National Medical Center, Washington, DC, USA.
| |
Collapse
|
25
|
Carpinelli MR, de Vries ME, Auden A, Butt T, Deng Z, Partridge DD, Miles LB, Georgy SR, Haigh JJ, Darido C, Brabletz S, Brabletz T, Stemmler MP, Dworkin S, Jane SM. Inactivation of Zeb1 in GRHL2-deficient mouse embryos rescues mid-gestation viability and secondary palate closure. Dis Model Mech 2020; 13:dmm.042218. [PMID: 32005677 PMCID: PMC7104862 DOI: 10.1242/dmm.042218] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022] Open
Abstract
Cleft lip and palate are common birth defects resulting from failure of the facial processes to fuse during development. The mammalian grainyhead-like (Grhl1-3) genes play key roles in a number of tissue fusion processes including neurulation, epidermal wound healing and eyelid fusion. One family member, Grhl2, is expressed in the epithelial lining of the first pharyngeal arch in mice at embryonic day (E)10.5, prompting analysis of the role of this factor in palatogenesis. Grhl2-null mice die at E11.5 with neural tube defects and a cleft face phenotype, precluding analysis of palatal fusion at a later stage of development. However, in the first pharyngeal arch of Grhl2-null embryos, dysregulation of transcription factors that drive epithelial-mesenchymal transition (EMT) occurs. The aberrant expression of these genes is associated with a shift in RNA-splicing patterns that favours the generation of mesenchymal isoforms of numerous regulators. Driving the EMT perturbation is loss of expression of the EMT-suppressing transcription factors Ovol1 and Ovol2, which are direct GRHL2 targets. The expression of the miR-200 family of microRNAs, also GRHL2 targets, is similarly reduced, resulting in a 56-fold upregulation of Zeb1 expression, a major driver of mesenchymal cellular identity. The critical role of GRHL2 in mediating cleft palate in Zeb1−/− mice is evident, with rescue of both palatal and facial fusion seen in Grhl2−/−;Zeb1−/− embryos. These findings highlight the delicate balance between GRHL2/ZEB1 and epithelial/mesenchymal cellular identity that is essential for normal closure of the palate and face. Perturbation of this pathway may underlie cleft palate in some patients. Summary: Epithelial transcription factor GRHL2 is required for face closure while mesenchymal transcription factor ZEB1 is required for palate closure. Surprisingly, animals lacking both factors close their face and secondary palate.
Collapse
Affiliation(s)
- Marina R Carpinelli
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Michael E de Vries
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Alana Auden
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Tariq Butt
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Zihao Deng
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Darren D Partridge
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Lee B Miles
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Smitha R Georgy
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Jody J Haigh
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Charbel Darido
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Simone Brabletz
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Sebastian Dworkin
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| | - Stephen M Jane
- Department of Medicine, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
| |
Collapse
|
26
|
de Vries M, Carpinelli M, Rutland E, Hatzipantelis A, Partridge D, Auden A, Anderson PJ, De Groef B, Wu H, Osterwalder M, Visel A, Jane SM, Dworkin S. Interrogating the Grainyhead-like 2 (Grhl2) genomic locus identifies an enhancer element that regulates palatogenesis in mouse. Dev Biol 2020; 459:194-203. [PMID: 31782997 DOI: 10.1016/j.ydbio.2019.11.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 10/25/2022]
Abstract
The highly-conserved Grainyhead-like (Grhl) transcription factors are critical regulators of embryogenesis that regulate cellular survival, proliferation, migration and epithelial integrity, especially during the formation of the craniofacial skeleton. Family member Grhl2 is expressed throughout epithelial tissues during development, and loss of Grhl2 function leads to significant defects in neurulation, abdominal wall closure, formation of the face and fusion of the maxilla/palate. Whereas numerous downstream target genes of Grhl2 have been identified, very little is known about how this crucial developmental transcription factor itself is regulated. Here, using in silico and in utero expression analyses and functional deletion in mice, we have identified a novel 2.4 kb enhancer element (mm1286) that drives reporter gene expression in a pattern that strongly recapitulates endogenous Grhl2 in the craniofacial primordia, modulates Grhl2 expression in these tissues, and augments Grhl2-mediated closure of the secondary palate. Deletion of this genomic element, in the context of inactivation of one allele of Grhl2 (through generation of double heterozygous Grhl2+/-;mm1286+/- mice), results in a significant predisposition to palatal clefting at birth. Moreover, we found that a highly conserved 325 bp region of mm1286 is both necessary and sufficient for mediating the craniofacial-specific enhancer activity of this region, and that an extremely well-conserved 12-bp sequence within this element (CTGTCAAACAGGT) substantially determines full enhancer function. Together, these data provide valuable new insights into the upstream genomic regulatory landscape responsible for transcriptional control of Grhl2 during palatal closure.
Collapse
Affiliation(s)
- Michael de Vries
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3004, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Marina Carpinelli
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3004, Australia
| | - Emilie Rutland
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3004, Australia
| | - Aaron Hatzipantelis
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3004, Australia
| | - Darren Partridge
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3004, Australia
| | - Alana Auden
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3004, Australia
| | - Peter J Anderson
- Australian Craniofacial Unit, Women and Children's Hospital, Adelaide, SA, 5005, Australia; Faculty of Health Sciences, University of Adelaide, SA, 5005, Australia; Nanjing Medical University, Nanjing, PR China
| | - Bert De Groef
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Han Wu
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA; U.S. Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA; School of Natural Sciences, University of California, Merced, CA, 95343, USA
| | - Stephen M Jane
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3004, Australia
| | - Sebastian Dworkin
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, 3086, Australia.
| |
Collapse
|
27
|
Kashgari G, Meinecke L, Gordon W, Ruiz B, Yang J, Ma AL, Xie Y, Ho H, Plikus MV, Nie Q, Jester JV, Andersen B. Epithelial Migration and Non-adhesive Periderm Are Required for Digit Separation during Mammalian Development. Dev Cell 2020; 52:764-778.e4. [PMID: 32109382 DOI: 10.1016/j.devcel.2020.01.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/26/2019] [Accepted: 01/28/2020] [Indexed: 01/04/2023]
Abstract
The fusion of digits or toes, syndactyly, can be part of complex syndromes, including van der Woude syndrome. A subset of van der Woude cases is caused by dominant-negative mutations in the epithelial transcription factor Grainyhead like-3 (GRHL3), and Grhl3-/-mice have soft-tissue syndactyly. Although impaired interdigital cell death of mesenchymal cells causes syndactyly in multiple genetic mutants, Grhl3-/- embryos had normal interdigital cell death, suggesting alternative mechanisms for syndactyly. We found that in digit separation, the overlying epidermis forms a migrating interdigital epithelial tongue (IET) when the epithelium invaginates to separate the digits. Normally, the non-adhesive surface periderm allows the IET to bifurcate as the digits separate. In contrast, in Grhl3-/- embryos, the IET moves normally between the digits but fails to bifurcate because of abnormal adhesion of the periderm. Our study identifies epidermal developmental processes required for digit separation.
Collapse
Affiliation(s)
- Ghaidaa Kashgari
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Lina Meinecke
- Department of Mathematics, School of Physical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Developmental & Cell Biology, School of the Biological Sciences, University of California, Irvine, Irvine, CA, USA
| | - William Gordon
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Bryan Ruiz
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Jady Yang
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Amy Lan Ma
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Yilu Xie
- The Gavin Herbert Eye Institute, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Hsiang Ho
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Maksim V Plikus
- Department of Developmental & Cell Biology, School of the Biological Sciences, University of California, Irvine, Irvine, CA, USA
| | - Qing Nie
- Department of Mathematics, School of Physical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Developmental & Cell Biology, School of the Biological Sciences, University of California, Irvine, Irvine, CA, USA
| | - James V Jester
- The Gavin Herbert Eye Institute, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Bogi Andersen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA; Department of Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
28
|
Kotarba G, Taracha-Wisniewska A, Wilanowski T. Grainyhead-like transcription factors in cancer - Focus on recent developments. Exp Biol Med (Maywood) 2020; 245:402-410. [PMID: 32008358 DOI: 10.1177/1535370220903009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The role of grainyhead-like transcription factors in cancer has been widely investigated by the scientific community. However, some of its aspects do not seem to be adequately appreciated, and these are the topic of our article. In addition to their well-documented role as tumor suppressors, in many cases the grainyhead-like proteins perform tumor-promoting functions, which make them potential drug targets. However, it is difficult to directly target transcription factors, which is why we recommend an alternative approach. The transcriptional transactivation activity of grainyhead-like transcription factors is regulated by phosphorylation, and protein kinases are much more feasible drug targets. Studying the phosphorylation of grainyhead-like proteins may thus allow to identify protein kinases regulating the activity of these factors, and design inhibitors of these kinases to indirectly regulate the activity of grainyhead-like transcription factors. There are many somatic mutations in the GRHL genes that occur during cancer development. These mutations are widely distributed across the GRHL loci, and these mutations are very rare. For this reason, they are unlikely to become targets of future therapies, nevertheless some of them may be driver mutations and studying them may provide important novel information about the regulation of functioning of the GRHL genes and proteins. Analogous information may be obtained by studying single nucleotide polymorphisms in GRHL genes that are associated with disease risk. Such polymorphisms may also prove useful in identifying individuals with an increased risk of a particular disease. Impact statement In the present article, we focus on relatively little appreciated aspects of involvement of the grainyhead-like (GRHL) transcription factors in cancer. These aspects are nevertheless very important for the functioning of GRHL proteins, as well as for cancer development. Some of the GRHL factors perform tumor-promoting functions in certain types of cancer, which makes them potential drug targets. Much information is available about somatic cancer mutations in the GRHL genes, yet there are very few analyses of these mutations in the scientific literature. The activity of GRHL transcription factors is controlled by phosphorylation, and we suggest that regulating their phosphorylation with specific protein kinases provides an alternative approach to modify the activity of GRHL proteins. Some single nucleotide polymorphisms (SNPs) in the GRHL genes are associated with disease risk. Studying such SNPs may yield new information about the functioning of GRHL genes and proteins, and may also allow to identify people with an increased risk of a particular disease.
Collapse
Affiliation(s)
- Grzegorz Kotarba
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw 02-096, Poland
| | | | - Tomasz Wilanowski
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw 02-096, Poland
| |
Collapse
|
29
|
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: 0.6] [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
|
30
|
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: 33] [Impact Index Per Article: 5.5] [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
|
31
|
Li H, Jones KL, Hooper JE, Williams T. The molecular anatomy of mammalian upper lip and primary palate fusion at single cell resolution. Development 2019; 146:dev.174888. [PMID: 31118233 DOI: 10.1242/dev.174888] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Abstract
The mammalian lip and primary palate form when coordinated growth and morphogenesis bring the nasal and maxillary processes into contact, and the epithelia co-mingle, remodel and clear from the fusion site to allow mesenchyme continuity. Although several genes required for fusion have been identified, an integrated molecular and cellular description of the overall process is lacking. Here, we employ single cell RNA sequencing of the developing mouse face to identify ectodermal, mesenchymal and endothelial populations associated with patterning and fusion of the facial prominences. This analysis indicates that key cell populations at the fusion site exist within the periderm, basal epithelial cells and adjacent mesenchyme. We describe the expression profiles that make each population unique, and the signals that potentially integrate their behaviour. Overall, these data provide a comprehensive high-resolution description of the various cell populations participating in fusion of the lip and primary palate, as well as formation of the nasolacrimal groove, and they furnish a powerful resource for those investigating the molecular genetics of facial development and facial clefting that can be mined for crucial mechanistic information concerning this prevalent human birth defect.
Collapse
Affiliation(s)
- Hong Li
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Kenneth L Jones
- Department of Pediatrics, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Joan E Hooper
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
32
|
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: 2.7] [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
|
33
|
Kersbergen A, Best SA, Dworkin S, Ah-Cann C, de Vries ME, Asselin-Labat ML, Ritchie ME, Jane SM, Sutherland KD. Lung morphogenesis is orchestrated through Grainyhead-like 2 (Grhl2) transcriptional programs. Dev Biol 2018; 443:1-9. [DOI: 10.1016/j.ydbio.2018.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/17/2018] [Accepted: 09/02/2018] [Indexed: 01/04/2023]
|
34
|
Bardill J, Williams SM, Shabeka U, Niswander L, Park D, Marwan AI. An Injectable Reverse Thermal Gel for Minimally Invasive Coverage of Mouse Myelomeningocele. J Surg Res 2018; 235:227-236. [PMID: 30691800 DOI: 10.1016/j.jss.2018.09.078] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 11/17/2022]
Abstract
BACKGROUND Myelomeningocele (MMC) results in lifelong neurologic and functional deficits. Currently, prenatal repair of MMC closes the defect, resulting in a 50% reduction in postnatal ventriculoperitoneal shunting. However, this invasive fetal surgery is associated with significant morbidities to mother and baby. We have pioneered a novel reverse thermal gel (RTG) to cover MMC defects in a minimally invasive manner. Here, we test in-vitro RTG long-term stability in amniotic fluid and in vivo application in the Grainy head-like 3 (Grhl3) mouse MMC model. MATERIALS AND METHODS RTG stability in amniotic fluid (in-vitro) was monitored for 6 mo and measured using gel permeation chromatography and solution-gel transition temperature (lower critical solution temperature). E16.5 Grhl3 mouse fetuses were injected with the RTG or saline and harvested on E19.5. Tissue was assessed for RTG coverage of the gross defect and inflammatory response by immunohistochemistry for macrophages. RESULTS Polymer backbone molecular weight and lower critical solution temperature remain stable in amniotic fluid after 6 mo. Needle injection over the MMC of Grhl3 fetuses successfully forms a stable gel that covers the entire defect. On harvest, some animals demonstrate >50% RTG coverage. RTG injection is not associated with inflammation. CONCLUSIONS Our results demonstrate that the RTG is a promising candidate for a minimally invasive approach to patch MMC. We are now poised to test our RTG patch in the large preclinical ovine model used to evaluate prenatal repair of MMC.
Collapse
Affiliation(s)
- James Bardill
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado; Division of Pediatric Surgery, Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Sarah M Williams
- Division of Pediatric Surgery, Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Uladzimir Shabeka
- Division of Pediatric Surgery, Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado
| | - Lee Niswander
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, Colorado
| | - Daewon Park
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado.
| | - Ahmed I Marwan
- Department of Bioengineering, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado; Division of Pediatric Surgery, Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
35
|
Wang XK, Zhou FF, Tao HR, Wang X, Zhang C, Su F, Wang SP, Xu LH, Pan XK, Feng MH, Xie W. Knockdown of GRHL3 inhibits activities and induces cell cycle arrest and apoptosis of human colorectal cancer cells. Curr Med Sci 2017; 37:880-885. [PMID: 29270747 DOI: 10.1007/s11596-017-1821-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/01/2017] [Indexed: 12/14/2022]
Abstract
The Grainyhead-like 3 (GRHL3) is involved in epidermal barrier formation, neural tube closure and wound repair. Previous studies have suggested that GRHL3 has been linked to many different types of cancers. However, to date, its effects on human colorectal cancer (CRC) has not been clarified yet. Our microarray analysis has indicated predominant GRHL3 expression in CRC. The purpose of this study was to investigate the expression and significance of GRHL3 in CRC tumorigenesis using CRC tissues and paired paracancerous tissues, as well as using distinct CRC cell lines (HT29 and DLD1). We observed increased GRHL3 expression at both mRNA and protein levels in CRC tissues and CRC cell lines using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. Moreover, silencing GRHL3 with siRNA could suppress CRC cell proliferation, viability and migration in vitro. We also found that knockdown of GRHL3 could promote cell cycle arrest at G0/G1 phase in HT29 cells and DLD1 cells, and induce cell apoptosis in HT29 cells. Together, our study revealed the down-regulation of GRHL3 in vitro could inhibit CRC cell activity and trigger cell cycle arrest at G0/G1 phase and apoptosis.
Collapse
Affiliation(s)
- Xiao-Kang Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fen-Fang Zhou
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hao-Ran Tao
- School of Clinical Medicine, Fenyang Medical College, Shanxi Medical University, Fenyang, 032200, China
| | - Xin Wang
- Department of General Surgery, Tongshan Renmin Hospital, Tongshan, 437400, China
| | - Chi Zhang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fei Su
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shi-Pei Wang
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Li-Hua Xu
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xue-Kai Pan
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Mao-Hui Feng
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Wei Xie
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
36
|
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: 2.6] [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
|
37
|
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: 2.4] [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
|
38
|
Nikolopoulou E, Galea GL, Rolo A, Greene NDE, Copp AJ. Neural tube closure: cellular, molecular and biomechanical mechanisms. Development 2017; 144:552-566. [PMID: 28196803 DOI: 10.1242/dev.145904] [Citation(s) in RCA: 320] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neural tube closure has been studied for many decades, across a range of vertebrates, as a paradigm of embryonic morphogenesis. Neurulation is of particular interest in view of the severe congenital malformations - 'neural tube defects' - that result when closure fails. The process of neural tube closure is complex and involves cellular events such as convergent extension, apical constriction and interkinetic nuclear migration, as well as precise molecular control via the non-canonical Wnt/planar cell polarity pathway, Shh/BMP signalling, and the transcription factors Grhl2/3, Pax3, Cdx2 and Zic2. More recently, biomechanical inputs into neural tube morphogenesis have also been identified. Here, we review these cellular, molecular and biomechanical mechanisms involved in neural tube closure, based on studies of various vertebrate species, focusing on the most recent advances in the field.
Collapse
Affiliation(s)
- Evanthia Nikolopoulou
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Gabriel L Galea
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Ana Rolo
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Nicholas D E Greene
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Andrew J Copp
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
39
|
Boivin FJ, Schmidt-Ott KM. Transcriptional mechanisms coordinating tight junction assembly during epithelial differentiation. Ann N Y Acad Sci 2017. [PMID: 28636799 DOI: 10.1111/nyas.13367] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epithelial tissues form a selective barrier via direct cell-cell interactions to separate and establish concentration gradients between the different compartments of the body. Proper function and formation of this barrier rely on the establishment of distinct intercellular junction complexes. These complexes include tight junctions, adherens junctions, desmosomes, and gap junctions. The tight junction is by far the most diverse junctional complex in the epithelial barrier. Its composition varies greatly across different epithelial tissues to confer various barrier properties. Thus, epithelial cells rely on tightly regulated transcriptional mechanisms to ensure proper formation of the epithelial barrier and to achieve tight junction diversity. Here, we review different transcriptional mechanisms utilized during embryogenesis and disease development to promote tight junction assembly and maintenance of intercellular barrier integrity. We focus particularly on the Grainyhead-like transcription factors and ligand-activated nuclear hormone receptors, two central families of proteins in epithelialization.
Collapse
Affiliation(s)
- Felix J Boivin
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology, Charité Medical University, Berlin, Germany
| |
Collapse
|
40
|
Tanimizu N, Mitaka T. Epithelial Morphogenesis during Liver Development. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027862. [PMID: 28213465 DOI: 10.1101/cshperspect.a027862] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue stem/progenitor cells supply multiple types of epithelial cells that eventually acquire specialized functions during organ development. In addition, three-dimensional (3D) tissue structures need to be established for organs to perform their physiological functions. The liver contains two types of epithelial cells, namely, hepatocytes and cholangiocytes, which are derived from hepatoblasts, fetal liver stem/progenitor cells (LPCs), in mid-gestation. Hepatocytes performing many metabolic reactions form cord-like structures, whereas cholangiocytes, biliary epithelial cells, form tubular structures called intrahepatic bile ducts. Analyses for human genetic diseases and mutant mice have identified crucial molecules for liver organogenesis. Functions of those molecules can be examined in in vitro culture systems where LPCs are induced to differentiate into hepatocytes or cholangiocytes. Recent technical advances have revealed 3D epithelial morphogenesis during liver organogenesis. Therefore, the liver is a good model to understand how tissue stem/progenitor cells differentiate and establish 3D tissue architectures during organ development.
Collapse
Affiliation(s)
- Naoki Tanimizu
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| | - Toshihiro Mitaka
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Chuo-ku, Sapporo 060-8556, Japan
| |
Collapse
|
41
|
Yao L, Wang S, Westholm JO, Dai Q, Matsuda R, Hosono C, Bray S, Lai EC, Samakovlis C. Genome-wide identification of Grainy head targets in Drosophila reveals regulatory interactions with the POU domain transcription factor Vvl. Development 2017; 144:3145-3155. [PMID: 28760809 DOI: 10.1242/dev.143297] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 07/21/2017] [Indexed: 12/17/2022]
Abstract
Grainy head (Grh) is a conserved transcription factor (TF) controlling epithelial differentiation and regeneration. To elucidate Grh functions we identified embryonic Grh targets by ChIP-seq and gene expression analysis. We show that Grh controls hundreds of target genes. Repression or activation correlates with the distance of Grh-binding sites to the transcription start sites of its targets. Analysis of 54 Grh-responsive enhancers during development and upon wounding suggests cooperation with distinct TFs in different contexts. In the airways, Grh-repressed genes encode key TFs involved in branching and cell differentiation. Reduction of the POU domain TF Ventral veins lacking (Vvl) largely ameliorates the airway morphogenesis defects of grh mutants. Vvl and Grh proteins additionally interact with each other and regulate a set of common enhancers during epithelial morphogenesis. We conclude that Grh and Vvl participate in a regulatory network controlling epithelial maturation.
Collapse
Affiliation(s)
- Liqun Yao
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, S10691, Stockholm, Sweden
| | - Shenqiu Wang
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, S10691, Stockholm, Sweden.,Cancer Biology & Genetics Program, Sloan-Kettering Institute, 1275 York Ave, Box 252, New York, NY 10065, USA
| | - Jakub O Westholm
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Ave, Box 252, New York, NY 10065, USA.,Science for Life Laboratory, Tomtebodavägen 232, 171 21 Solna, Sweden
| | - Qi Dai
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, S10691, Stockholm, Sweden.,Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Ave, Box 252, New York, NY 10065, USA
| | - Ryo Matsuda
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, S10691, Stockholm, Sweden
| | - Chie Hosono
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, S10691, Stockholm, Sweden
| | - Sarah Bray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Eric C Lai
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Ave, Box 252, New York, NY 10065, USA
| | - Christos Samakovlis
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, S10691, Stockholm, Sweden .,Science for Life Laboratory, Tomtebodavägen 232, 171 21 Solna, Sweden.,Molecular Pneumology, UGMLC, Aulweg 130, 35392 Giessen, Germany
| |
Collapse
|
42
|
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: 6.1] [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
|
43
|
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: 2.5] [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
|
44
|
Janssen R, Muller A, Simonides WS. Cardiac Thyroid Hormone Metabolism and Heart Failure. Eur Thyroid J 2017; 6:130-137. [PMID: 28785539 PMCID: PMC5527173 DOI: 10.1159/000469708] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 03/07/2017] [Indexed: 12/18/2022] Open
Abstract
The heart is a principal target of thyroid hormone, and a reduction of cardiac thyroid hormone signaling is thought to play a role in pathological ventricular remodeling and the development of heart failure. Studies in various rodent models of heart disease have identified increased activity of cardiac type III deiodinase as a possible cause of diminished levels and action of thyroid hormone. Recent data indicate novel mechanisms underlying the induction of this thyroid hormone-degrading enzyme in the heart as well as post-transcriptional regulation of its expression by microRNAs. In addition, the relevance of diminished thyroid hormone signaling for cardiac remodeling is suggested to include miRNA-mediated effects on pathological signaling pathways. These and other recent studies are reviewed and discussed in the context of other processes and factors that have been implicated in the reduction of cardiac thyroid hormone signaling in heart failure.
Collapse
Affiliation(s)
| | | | - Warner S. Simonides
- *Warner S. Simonides, PhD, Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, De Boelelaan 1118, NL–1081 HV Amsterdam (The Netherlands), E-Mail
| |
Collapse
|
45
|
Abstract
The two main mechanisms that expand the proteomic output of eukaryotic genes are alternative splicing and alternative translation initiation signals. Despite being essential to generate isoforms of gene products that create functional diversity during development, the impact of these mechanisms on fine-tuning regulatory gene networks is still underappreciated. In this review, we use the Grainyhead-like (Grhl) family as a case study to illustrate the importance of isoforms when investigating transcription factor family function during development and disease, and highlight the potential for differential modulation of downstream target genes. We provide insights into the importance of considering alternative gene products when designing, undertaking, and analysing primary research, and the effect that isoforms may have on development. This review also covers known mutations in Grhl family members, and postulates how genetic changes may dictate transcriptional specificity between the Grhl family members. It also contrasts and compares the available literature on the function and importance of the Grhl isoforms, and highlights current gaps in our understanding of their regulatory gene networks in development and disease.
Collapse
Affiliation(s)
- Lee B Miles
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia
| | - Sebastian Dworkin
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia
| | - Charbel Darido
- Division of Cancer Research, Peter MacCallum Cancer Centre, Grattan Street, Parkville, VIC 3052, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, VIC 3052, Australia.
| |
Collapse
|
46
|
Pawlak M, Kikulska A, Wrzesinski T, Rausch T, Kwias Z, Wilczynski B, Benes V, Wesoly J, Wilanowski T. Potential protective role of Grainyhead-like genes in the development of clear cell renal cell carcinoma. Mol Carcinog 2017; 56:2414-2423. [DOI: 10.1002/mc.22682] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/18/2017] [Accepted: 05/19/2017] [Indexed: 01/20/2023]
Affiliation(s)
- Magdalena Pawlak
- Laboratory of Signal Transduction; Department of Cell Biology; Nencki Institute of Experimental Biology of Polish Academy of Sciences; Warsaw Poland
| | - Agnieszka Kikulska
- Laboratory of Signal Transduction; Department of Cell Biology; Nencki Institute of Experimental Biology of Polish Academy of Sciences; Warsaw Poland
| | - Tomasz Wrzesinski
- Faculty of Biology; Laboratory of High Throughput Technologies; Institute of Molecular Biology and Biotechnology; Adam Mickiewicz University; Poznan Poland
| | - Tobias Rausch
- Genomics Core Facility; European Molecular Biology Laboratory; Heidelberg Germany
| | - Zbigniew Kwias
- Department of Urology and Urological Oncology; Poznan University of Medical Sciences; Poznan Poland
| | - Bartek Wilczynski
- Faculty of Mathematics, Informatics and Mechanics; Institute of Informatics; University of Warsaw; Warsaw Poland
| | - Vladimir Benes
- Genomics Core Facility; European Molecular Biology Laboratory; Heidelberg Germany
| | - Joanna Wesoly
- Faculty of Biology; Laboratory of High Throughput Technologies; Institute of Molecular Biology and Biotechnology; Adam Mickiewicz University; Poznan Poland
| | - Tomasz Wilanowski
- Laboratory of Signal Transduction; Department of Cell Biology; Nencki Institute of Experimental Biology of Polish Academy of Sciences; Warsaw Poland
| |
Collapse
|
47
|
Mohd-Zin SW, Marwan AI, Abou Chaar MK, Ahmad-Annuar A, Abdul-Aziz NM. Spina Bifida: Pathogenesis, Mechanisms, and Genes in Mice and Humans. SCIENTIFICA 2017; 2017:5364827. [PMID: 28286691 PMCID: PMC5327787 DOI: 10.1155/2017/5364827] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/14/2016] [Accepted: 12/01/2016] [Indexed: 05/26/2023]
Abstract
Spina bifida is among the phenotypes of the larger condition known as neural tube defects (NTDs). It is the most common central nervous system malformation compatible with life and the second leading cause of birth defects after congenital heart defects. In this review paper, we define spina bifida and discuss the phenotypes seen in humans as described by both surgeons and embryologists in order to compare and ultimately contrast it to the leading animal model, the mouse. Our understanding of spina bifida is currently limited to the observations we make in mouse models, which reflect complete or targeted knockouts of genes, which perturb the whole gene(s) without taking into account the issue of haploinsufficiency, which is most prominent in the human spina bifida condition. We thus conclude that the need to study spina bifida in all its forms, both aperta and occulta, is more indicative of the spina bifida in surviving humans and that the measure of deterioration arising from caudal neural tube defects, more commonly known as spina bifida, must be determined by the level of the lesion both in mouse and in man.
Collapse
Affiliation(s)
- Siti W. Mohd-Zin
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ahmed I. Marwan
- Laboratory for Fetal and Regenerative Biology, Colorado Fetal Care Center, Division of Pediatric Surgery, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, 12700 E 17th Ave, Aurora, CO 80045, USA
| | | | - Azlina Ahmad-Annuar
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Noraishah M. Abdul-Aziz
- Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
48
|
Dworkin S, Auden A, Partridge DD, Daglas M, Medcalf RL, Mantamadiotis T, Georgy SR, Darido C, Jane SM, Ting SB. Grainyhead-like 3 (Grhl3) deficiency in brain leads to altered locomotor activity and decreased anxiety-like behaviors in aged mice. Dev Neurobiol 2017; 77:775-788. [PMID: 27907249 DOI: 10.1002/dneu.22469] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/17/2016] [Accepted: 11/15/2016] [Indexed: 01/24/2023]
Abstract
The highly conserved Grainyhead-like (Grhl) family of transcription factors, comprising three members in vertebrates (Grhl1-3), play critical regulatory roles during embryonic development, cellular proliferation, and apoptosis. Although loss of Grhl function leads to multiple neural abnormalities in numerous animal models, a comprehensive analysis of Grhl expression and function in the mammalian brain has not been reported. Here they show that only Grhl3 expression is detectable in the embryonic mouse brain; particularly within the habenula, an organ known to modulate repressive behaviors. Using both Grhl3-knockout mice (Grhl3-/- ), and brain-specific conditional deletion of Grhl3 in adult mice (Nestin-Cre/Grhl3flox/flox ), they performed histological expression analyses and behavioral tests to assess long-term effects of Grhl3 loss on motor co-ordination, spatial memory, anxiety, and stress. They found that complete deletion of Grhl3 did not lead to noticeable structural or cell-intrinsic defects in the embryonic brain; however, aged Grhl3 conditional knockout (cKO) mice showed enlarged lateral ventricles and displayed marked changes in motor function and behaviors suggestive of decreased fear and anxiety. They conclude that loss of Grhl3 in the brain leads to significant alterations in locomotor activity and decreased self-inhibition, and as such, these mice may serve as a novel model of human conditions of impulsive behavior or hyperactivity. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 775-788, 2017.
Collapse
Affiliation(s)
- Sebastian Dworkin
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3181, Australia
| | - Alana Auden
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3181, Australia
| | - Darren D Partridge
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3181, Australia
| | - Maria Daglas
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University Central Clinical School, Prahran, Victoria, 3181, Australia
| | - Robert L Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University Central Clinical School, Prahran, Victoria, 3181, Australia
| | - Theo Mantamadiotis
- Department of Pathology, University of Melbourne, Parkville, Victoria, 3050, Australia
| | - Smitha R Georgy
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3181, Australia
| | - Charbel Darido
- Peter MacCallum Cancer Centre, The Victorian Comprehensive Cancer Centre, Parkville, Victoria, 3050, Australia
| | - Stephen M Jane
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3181, Australia.,Department of Hematology, Alfred Hospital, Prahran, Victoria, 3181, Australia
| | - Stephen B Ting
- Department of Medicine, Monash University Central Clinical School, Prahran, Victoria, 3181, Australia.,Department of Hematology, Alfred Hospital, Prahran, Victoria, 3181, Australia
| |
Collapse
|
49
|
Goldie SJ, Arhatari BD, Anderson P, Auden A, Partridge DD, Jane SM, Dworkin S. Mice lacking the conserved transcription factor Grainyhead-like 3 (Grhl3) display increased apposition of the frontal and parietal bones during embryonic development. BMC DEVELOPMENTAL BIOLOGY 2016; 16:37. [PMID: 27756203 PMCID: PMC5070091 DOI: 10.1186/s12861-016-0136-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Increased apposition of the frontal and parietal bones of the skull during embryogenesis may be a risk factor for the subsequent development of premature skull fusion, or craniosynostosis. Human craniosynostosis is a prevalent, and often serious embryological and neonatal pathology. Other than known mutations in a small number of contributing genes, the aetiology of craniosynostosis is largely unknown. Therefore, the identification of novel genes which contribute to normal skull patterning, morphology and premature suture apposition is imperative, in order to fully understand the genetic regulation of cranial development. RESULTS Using advanced imaging techniques and quantitative measurement, we show that genetic deletion of the highly-conserved transcription factor Grainyhead-like 3 (Grhl3) in mice (Grhl3 -/- ) leads to decreased skull size, aberrant skull morphology and premature apposition of the coronal sutures during embryogenesis. Furthermore, Grhl3 -/- mice also present with premature collagen deposition and osteoblast alignment at the sutures, and the physical interaction between the developing skull, and outermost covering of the brain (the dura mater), as well as the overlying dermis and subcutaneous tissue, appears compromised in embryos lacking Grhl3. Although Grhl3 -/- mice die at birth, we investigated skull morphology and size in adult animals lacking one Grhl3 allele (heterozygous; Grhl3 +/- ), which are viable and fertile. We found that these adult mice also present with a smaller cranial cavity, suggestive of post-natal haploinsufficiency in the context of cranial development. CONCLUSIONS Our findings show that our Grhl3 mice present with increased apposition of the frontal and parietal bones, suggesting that Grhl3 may be involved in the developmental pathogenesis of craniosynostosis.
Collapse
Affiliation(s)
- Stephen J Goldie
- Department of Medicine, Monash University Central Clinical School, Prahran, VIC, 3004, Australia
| | - Benedicta D Arhatari
- ARC Centre of Excellence for Advanced Molecular Imaging, Department of Chemistry and Physics, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Peter Anderson
- Australian Craniofacial Unit, Women and Children's Hospital, Adelaide, SA, 5005, Australia.,Faculty of Health Sciences, University of Adelaide, Adelaide, SA, 5005, Australia.,Nanjing Medical University, Nanjing, People's Republic of China
| | - Alana Auden
- Department of Medicine, Monash University Central Clinical School, Prahran, VIC, 3004, Australia
| | - Darren D Partridge
- Department of Medicine, Monash University Central Clinical School, Prahran, VIC, 3004, Australia
| | - Stephen M Jane
- Department of Medicine, Monash University Central Clinical School, Prahran, VIC, 3004, Australia
| | - Sebastian Dworkin
- Department of Medicine, Monash University Central Clinical School, Prahran, VIC, 3004, Australia. .,Present address: Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
50
|
Tanimizu N, Mitaka T. Morphogenesis of liver epithelial cells. Hepatol Res 2016; 46:964-76. [PMID: 26785307 DOI: 10.1111/hepr.12654] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 12/17/2022]
Abstract
The mammalian liver is a physiologically important organ performing various types of metabolism, producing serum proteins, detoxifying bilirubin and ammonia, and protecting the body from infection. Those physiological functions are achieved with the 3D tissue architecture of liver epithelial cells. The liver contains two types of epithelial cells, namely, hepatocytes and cholangiocytes. They split from hepatoblasts (embryonic liver stem cells) in mid-gestation and differentiate into structurally and functionally mature cells. Analyses of mutant mice showing abnormal liver organogenesis have identified genes involved in liver development. In vitro culture systems have been used to examine the mechanism in which each molecule or signaling pathway regulates the morphogenesis and functional differentiation of hepatocytes and cholangiocytes. In addition, liver epithelial cells as well as mesenchymal, sinusoidal endothelial and hematopoietic cells can be purified from developing livers, which enables us to perform genome-wide screening to identify novel genes regulating epithelial morphogenesis in the liver. By combining these in vivo and in vitro systems, the liver could be a unique and suitable model for revealing a principle, governing epithelial morphogenesis. In this review, we summarize recent progress in the understanding of the development of liver epithelial tissue structures.
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
|