1
|
Ou S, Jeyalatha MV, Mao Y, Wang J, Chen C, Zhang M, Liu X, Liang M, Lin S, Wu Y, Li Y, Li W. The Role of Ectodysplasin A on the Ocular Surface Homeostasis. Int J Mol Sci 2022; 23:ijms232415700. [PMID: 36555342 PMCID: PMC9779463 DOI: 10.3390/ijms232415700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/12/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
Ectodysplasin A (EDA), a ligand of the TNF family, plays an important role in maintaining the homeostasis of the ocular surface. EDA is necessary for the development of the meibomian gland, the lacrimal gland, as well as the proliferation and barrier function of the corneal epithelium. The mutation of EDA can induce the destruction of the ocular surface resulting in keratopathy, abnormality of the meibomian gland and maturation of the lacrimal gland. Experimental animal studies showed that a prenatal ultrasound-guided intra-amniotic injection or postnatal intravenous administration of soluble recombinant EDA protein can efficiently prevent the development of ocular surface abnormalities in EDA mutant animals. Furthermore, local application of EDA could restore the damaged ocular surface to some extent. Hence, a recombinant EDA-based therapy may serve as a novel paradigm to treat ocular surface disorders, such as meibomian gland dysfunction and corneal epithelium abnormalities.
Collapse
Affiliation(s)
- Shangkun Ou
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen 361000, China
| | - Mani Vimalin Jeyalatha
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Yi Mao
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen 361000, China
| | - Junqi Wang
- Department of Ophthalmology, Graduate School of Medicine, Osaka 5650871, Japan
| | - Chao Chen
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen 361000, China
| | - Minjie Zhang
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen 361000, China
| | - Xiaodong Liu
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen 361000, China
| | - Minghui Liang
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen 361000, China
| | - Sijie Lin
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen 361000, China
| | - Yiming Wu
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen 361000, China
| | - Yixuan Li
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Wei Li
- Eye Institute of Xiamen University and Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361000, China
- Fujian Provincial Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen 361000, China
- Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, China
- Correspondence: ; Tel./Fax: +86-592-2183761
| |
Collapse
|
2
|
Moradi MH, Mahmodi R, Farahani AHK, Karimi MO. Genome-wide evaluation of copy gain and loss variations in three Afghan sheep breeds. Sci Rep 2022; 12:14286. [PMID: 35996004 PMCID: PMC9395407 DOI: 10.1038/s41598-022-18571-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Copy number variation (CNV) is one of the main sources of variation between different individuals that has recently attracted much researcher interest as a major source for heritable variation in complex traits. The aim of this study was to identify CNVs in Afghan indigenous sheep consisting of three Arab, Baluchi, and Gadik breeds using genomic arrays containing 53,862 single nucleotide polymorphism (SNP) markers. Data were analyzed using the Hidden Markov Model (HMM) of PennCNV software. In this study, out of 45 sheep studied, 97.8% (44 animals) have shown CNVs. In total, 411 CNVs were observed for autosomal chromosomes and the entire sequence length of around 144 Mb was identified across the genome. The average number of CNVs per each sheep was 9.13. The identified CNVs for Arab, Baluchi, and Gadik breeds were 306, 62, and 43, respectively. After merging overlapped regions, a total of 376 copy number variation regions (CNVR) were identified, which are 286, 50, and 40 for Arab, Baluchi, and Gadik breeds, respectively. Bioinformatics analysis was performed to identify the genes and QTLs reported in these regions and the biochemical pathways involved by these genes. The results showed that many of these CNVRs overlapped with the genes or QTLs that are associated with various pathways such as immune system development, growth, reproduction, and environmental adaptions. Furthermore, to determine a genome-wide pattern of selection signatures in Afghan sheep breeds, the unbiased estimates of FST was calculated and the results indicated that 37 of the 376 CNVRs (~ 10%) have been also under selection signature, most of those overlapped with the genes influencing production, reproduction and immune system. Finally, the statistical methods used in this study was applied in an external dataset including 96 individuals of the Iranian sheep breed. The results indicated that 20 of the 114 CNVRs (18%) identified in Iranian sheep breed were also identified in our study, most of those overlapped with the genes influencing production, reproduction and immune system. Overall, this is the first attempts to develop the genomic map of loss and gain variation in the genome of Afghan indigenous sheep breeds, and may be important to shed some light on the genomic regions associated with some economically important traits in these breeds.
Collapse
Affiliation(s)
- Mohammad Hossein Moradi
- Department of Animal Science, Faculty of Agriculture and Natural Resources, Arak University, Arak, 38156-8-8349, Iran.
| | - Roqiah Mahmodi
- Department of Animal Science, Faculty of Agriculture and Natural Resources, Arak University, Arak, 38156-8-8349, Iran
| | | | - Mohammad Osman Karimi
- Department of Animal Science, Faculty of Agriculture and Natural Resources, Herat University, Herat, Afghanistan
| |
Collapse
|
3
|
Lee MY. Embryonic Programs in Cancer and Metastasis—Insights From the Mammary Gland. Front Cell Dev Biol 2022; 10:938625. [PMID: 35846378 PMCID: PMC9277484 DOI: 10.3389/fcell.2022.938625] [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: 05/07/2022] [Accepted: 06/07/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer is characterized as a reversion of a differentiated cell to a primitive cell state that recapitulates, in many aspects, features of embryonic cells. This review explores the current knowledge of developmental mechanisms that are essential for embryonic mouse mammary gland development, with a particular focus on genes and signaling pathway components that are essential for the induction, morphogenesis, and lineage specification of the mammary gland. The roles of these same genes and signaling pathways in mammary gland or breast tumorigenesis and metastasis are then summarized. Strikingly, key embryonic developmental pathways are often reactivated or dysregulated during tumorigenesis and metastasis in processes such as aberrant proliferation, epithelial-to-mesenchymal transition (EMT), and stem cell potency which affects cellular lineage hierarchy. These observations are in line with findings from recent studies using lineage tracing as well as bulk- and single-cell transcriptomics that have uncovered features of embryonic cells in cancer and metastasis through the identification of cell types, cell states and characterisation of their dynamic changes. Given the many overlapping features and similarities of the molecular signatures of normal development and cancer, embryonic molecular signatures could be useful prognostic markers for cancer. In this way, the study of embryonic development will continue to complement the understanding of the mechanisms of cancer and aid in the discovery of novel therapeutic targets and strategies.
Collapse
|
4
|
Abstract
The development and repair of dentin are strictly regulated by hundreds of genes. Abnormal dentin development is directly caused by gene mutations and dysregulation. Understanding and mastering this signal network is of great significance to the study of tooth development, tissue regeneration, aging, and repair and the treatment of dental diseases. It is necessary to understand the formation and repair mechanism of dentin in order to better treat the dentin lesions caused by various abnormal properties, whether it is to explore the reasons for the formation of dentin defects or to develop clinical drugs to strengthen the method of repairing dentin. Molecular biology of genes related to dentin development and repair are the most important basis for future research.
Collapse
Affiliation(s)
- Shuang Chen
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital, Fudan University, Shanghai, P. R. China.,Department of Prosthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, P. R. China
| | - Han Xie
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Shouliang Zhao
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Shuai Wang
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital, Fudan University, Shanghai, P. R. China
| | - Xiaoling Wei
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital, Fudan University, Shanghai, P. R. China.,Department of Endodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai, P. R. China
| | - Shangfeng Liu
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|
5
|
Spina E, Cowin P. Embryonic mammary gland development. Semin Cell Dev Biol 2021; 114:83-92. [DOI: 10.1016/j.semcdb.2020.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 12/03/2020] [Accepted: 12/29/2020] [Indexed: 12/27/2022]
|
6
|
Sadier A, Santana SE, Sears KE. The role of core and variable Gene Regulatory Network modules in tooth development and evolution. Integr Comp Biol 2020; 63:icaa116. [PMID: 32761089 DOI: 10.1093/icb/icaa116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 02/28/2024] Open
Abstract
Among the developmental processes that have been proposed to influence the direction of evolution, the modular organization of developmental gene regulatory networks (GRNs) has shown particular promise. In theory, GRNs have core modules comprised of essential, conserved circuits of genes, and sub-modules of downstream, secondary circuits of genes that are more susceptible to variation. While this idea has received considerable interest as of late, the field of evo-devo lacks the experimental systems needed to rigorously evaluate this hypothesis. Here, we introduce an experimental system, the vertebrate tooth, that has great potential as a model for testing this hypothesis. Tooth development and its associated GRN have been well studied and modeled in both model and non-model organisms. We propose that the existence of modules within the tooth GRN explains both the conservation of developmental mechanisms and the extraordinary diversity of teeth among vertebrates. Based on experimental data, we hypothesize that there is a conserved core module of genes that is absolutely necessary to ensure tooth or cusp initiation and development. In regard to tooth shape variation between species, we suggest that more relaxed sub-modules activated at later steps of tooth development, e.g., during the morphogenesis of the tooth and its cusps, control the different axes of tooth morphological variation.
Collapse
Affiliation(s)
- Alexa Sadier
- Department of Ecology and Evolutionary Biology, University of California at Los Angeles, Los Angeles, California
| | - Sharlene E Santana
- Department of Biology and Burke Museum of Natural History and Culture, University of Washington, Seattle, Washington
| | - Karen E Sears
- Department of Ecology and Evolutionary Biology, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
7
|
Ding Z, Kloss JM, Tuncali S, Tran NL, Loftus JC. TROY signals through JAK1-STAT3 to promote glioblastoma cell migration and resistance. Neoplasia 2020; 22:352-364. [PMID: 32629176 PMCID: PMC7338993 DOI: 10.1016/j.neo.2020.06.005] [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: 04/08/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 11/26/2022]
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor in adults and carries a discouraging prognosis. Its aggressive and highly infiltrative nature renders the current standard treatment of maximal surgical resection, radiation, and chemotherapy relatively ineffective. Identifying the signaling pathways that regulate GBM migration/invasion and resistance is required to develop more effective therapeutic regimens to treat GBM. Expression of TROY, an orphan receptor of the TNF receptor superfamily, increases with glial tumor grade, inversely correlates with patient overall survival, stimulates GBM cell invasion in vitro and in vivo, and increases resistance to temozolomide and radiation therapy. Conversely, silencing TROY expression inhibits GBM cell invasion, increases sensitivity to temozolomide, and prolongs survival in a preclinical intracranial xenograft model. Here, we have identified for the first time that TROY interacts with JAK1. Increased TROY expression increases JAK1 phosphorylation. In addition, increased TROY expression promotes STAT3 phosphorylation and STAT3 transcriptional activity that is dependent upon JAK1. TROY-mediated activation of STAT3 is independent of its ability to stimulate activity of NF-κB. Inhibition of JAK1 activity by ruxolitinib or knockdown of JAK1 expression by siRNA significantly inhibits TROY-induced STAT3 activation, GBM cell migration, and decreases resistance to temozolomide. Taken together, our data indicate that the TROY signaling complex may represent a potential therapeutic target with the distinctive capacity to exert effects on multiple pathways mediating GBM cell invasion and resistance.
Collapse
Affiliation(s)
- Zonghui Ding
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States
| | - Jean M Kloss
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States
| | - Serdar Tuncali
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States
| | - Nhan L Tran
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States; Department of Neurosurgery, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States.
| | - Joseph C Loftus
- Department of Cancer Biology, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States.
| |
Collapse
|
8
|
Enny A, Flaherty K, Mori S, Turner N, Nakamura T. Developmental constraints on fin diversity. Dev Growth Differ 2020; 62:311-325. [PMID: 32396685 PMCID: PMC7383993 DOI: 10.1111/dgd.12670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/17/2020] [Accepted: 04/06/2020] [Indexed: 12/31/2022]
Abstract
The fish fin is a breathtaking repository full of evolutionary diversity, novelty, and convergence. Over 500 million years, the adaptation to novel habitats has provided landscapes of fin diversity. Although comparative anatomy of evolutionarily divergent patterns over centuries has highlighted the fundamental architectures and evolutionary trends of fins, including convergent evolution, the developmental constraints on fin evolution, which bias the evolutionary trajectories of fin morphology, largely remain elusive. Here, we review the evolutionary history, developmental mechanisms, and evolutionary underpinnings of paired fins, illuminating possible developmental constraints on fin evolution. Our compilation of anatomical and genetic knowledge of fin development sheds light on the canalized and the unpredictable aspects of fin shape in evolution. Leveraged by an arsenal of genomic and genetic tools within the working arena of spectacular fin diversity, evolutionary developmental biology embarks on the establishment of conceptual framework for developmental constraints, previously enigmatic properties of evolution.
Collapse
Affiliation(s)
- Alyssa Enny
- Department of GeneticsRutgers the State University of New JerseyPiscatawayNJUSA
| | - Kathleen Flaherty
- Rutgers Animal CareRutgers the State University of New JerseyPiscatawayNJUSA
| | - Shunsuke Mori
- Department of GeneticsRutgers the State University of New JerseyPiscatawayNJUSA
| | - Natalie Turner
- Department of GeneticsRutgers the State University of New JerseyPiscatawayNJUSA
| | - Tetsuya Nakamura
- Department of GeneticsRutgers the State University of New JerseyPiscatawayNJUSA
| |
Collapse
|
9
|
Kuony A, Ikkala K, Kalha S, Magalhães AC, Pirttiniemi A, Michon F. Ectodysplasin-A signaling is a key integrator in the lacrimal gland-cornea feedback loop. Development 2019; 146:dev.176693. [PMID: 31221639 DOI: 10.1242/dev.176693] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 06/17/2019] [Indexed: 01/26/2023]
Abstract
A lack of ectodysplasin-A (Eda) signaling leads to dry eye symptoms, which have so far only been associated with altered Meibomian glands. Here, we used loss-of-function (Eda -/-) mutant mice to unravel the impact of Eda signaling on lacrimal gland formation, maturation and subsequent physiological function. Our study demonstrates that Eda activity is dispensable during lacrimal gland embryonic development. However, using a transcriptomic approach, we show that the Eda pathway is necessary for proper cell terminal differentiation in lacrimal gland epithelium and correlated with modified expression of secreted factors commonly found in the tear film. Finally, we discovered that lacrimal glands present a bilateral reduction of Eda signaling activity in response to unilateral corneal injury. This observation hints towards a role for the Eda pathway in controlling the switch from basal to reflex tears, to support corneal wound healing. Collectively, our data suggest a crucial implication of Eda signaling in the cornea-lacrimal gland feedback loop, both in physiological and pathophysiological conditions. Our findings demonstrate that Eda downstream targets could help alleviate dry eye symptoms.
Collapse
Affiliation(s)
- Alison Kuony
- Institute of Biotechnology, Helsinki Institute of Life Science, Developmental Biology Program, University of Helsinki, 00790 Helsinki, Finland.,Institut Jacques Monod, Université Denis Diderot - Paris 7, CNRS UMR 7592, Buffon building, 15 rue Hélène Brion, 75205 Paris Cedex 13, France
| | - Kaisa Ikkala
- Institute of Biotechnology, Helsinki Institute of Life Science, Developmental Biology Program, University of Helsinki, 00790 Helsinki, Finland
| | - Solja Kalha
- Institute of Biotechnology, Helsinki Institute of Life Science, Developmental Biology Program, University of Helsinki, 00790 Helsinki, Finland
| | - Ana Cathia Magalhães
- Institute of Biotechnology, Helsinki Institute of Life Science, Developmental Biology Program, University of Helsinki, 00790 Helsinki, Finland.,Institute for Neurosciences of Montpellier, INSERM UMR1051, University of Montpellier, 34295 Montpellier, France
| | - Anniina Pirttiniemi
- Institute of Biotechnology, Helsinki Institute of Life Science, Developmental Biology Program, University of Helsinki, 00790 Helsinki, Finland
| | - Frederic Michon
- Institute of Biotechnology, Helsinki Institute of Life Science, Developmental Biology Program, University of Helsinki, 00790 Helsinki, Finland .,Institute for Neurosciences of Montpellier, INSERM UMR1051, University of Montpellier, 34295 Montpellier, France
| |
Collapse
|
10
|
Molecular regulation of ocular gland development. Semin Cell Dev Biol 2018; 91:66-74. [PMID: 30266427 DOI: 10.1016/j.semcdb.2018.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 06/01/2018] [Accepted: 07/24/2018] [Indexed: 01/19/2023]
Abstract
The tear film is produced by two ocular glands, the lacrimal glands, which produce the aqueous component of this film, and the meibomian glands, which secrete the lipidic component that is key to reduce evaporation of the watery film at the surface of the eye. Embryonic development of these exocrine glands has been mostly studied in mice, which also develop Harderian glands, a third type of ocular gland whose role is still not well understood. This review provides an update on the signalling pathways, transcription factors andextracellular matrix components that have been shown to play a role in ocular gland development.
Collapse
|
11
|
PDZ-RhoGEF Is a Signaling Effector for TROY-Induced Glioblastoma Cell Invasion and Survival. Neoplasia 2018; 20:1045-1058. [PMID: 30219706 PMCID: PMC6140379 DOI: 10.1016/j.neo.2018.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of malignant brain tumors in adults and has a dismal prognosis. The highly aggressive invasion of malignant cells into the normal brain parenchyma renders complete surgical resection of GBM tumors impossible, increases resistance to therapeutic treatment, and leads to near-universal tumor recurrence. We have previously demonstrated that TROY (TNFRSF19) plays an important role in glioblastoma cell invasion and therapeutic resistance. However, the potential downstream effectors of TROY signaling have not been fully characterized. Here, we identified PDZ-RhoGEF as a binding partner for TROY that potentiated TROY-induced nuclear factor kappa B activation which is necessary for both cell invasion and survival. In addition, PDZ-RhoGEF also interacts with Pyk2, indicating that PDZ-RhoGEF is a component of a signalsome that includes TROY and Pyk2. PDZ-RhoGEF is overexpressed in glioblastoma tumors and stimulates glioma cell invasion via Rho activation. Increased PDZ-RhoGEF expression enhanced TROY-induced glioma cell migration. Conversely, silencing PDZ-RhoGEF expression inhibited TROY-induced glioma cell migration, increased sensitivity to temozolomide treatment, and extended survival of orthotopic xenograft mice. Furthermore, depletion of RhoC or RhoA inhibited TROY- and PDZ-RhoGEF-induced cell migration. Mechanistically, increased TROY expression stimulated Rho activation, and depletion of PDZ-RhoGEF expression reduced this activation. Taken together, these data suggest that PDZ-RhoGEF plays an important role in TROY signaling and provides insights into a potential node of vulnerability to limit GBM cell invasion and decrease therapeutic resistance.
Collapse
|
12
|
Species composition and environmental adaptation of indigenous Chinese cattle. Sci Rep 2017; 7:16196. [PMID: 29170422 PMCID: PMC5700937 DOI: 10.1038/s41598-017-16438-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022] Open
Abstract
Indigenous Chinese cattle combine taurine and indicine origins and occupy a broad range of different environments. By 50 K SNP genotyping we found a discontinuous distribution of taurine and indicine cattle ancestries with extremes of less than 10% indicine cattle in the north and more than 90% in the far south and southwest China. Model-based clustering and f4-statistics indicate introgression of both banteng and gayal into southern Chinese cattle while the sporadic yak influence in cattle in or near Tibetan area validate earlier findings of mitochondrial DNA analysis. Geographic patterns of taurine and indicine mitochondrial and Y-chromosomal DNA diversity largely agree with the autosomal cline. The geographic distribution of the genomic admixture of different bovine species is proposed to be the combined effect of prehistoric immigrations, gene flow, major rivers acting as genetic barriers, local breeding objectives and environmental adaptation. Whole-genome scan for genetic differentiation and association analyses with both environmental and morphological covariables are remarkably consistent with previous studies and identify a number of genes implicated in adaptation, which include TNFRSF19, RFX4, SP4 and several coat color genes. We propose indigenous Chinese cattle as a unique and informative resource for gene-level studies of climate adaptation in mammals.
Collapse
|
13
|
Ding Z, Roos A, Kloss J, Dhruv H, Peng S, Pirrotte P, Eschbacher JM, Tran NL, Loftus JC. A Novel Signaling Complex between TROY and EGFR Mediates Glioblastoma Cell Invasion. Mol Cancer Res 2017; 16:322-332. [PMID: 29117939 DOI: 10.1158/1541-7786.mcr-17-0454] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/13/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022]
Abstract
Glioblastoma is the most frequent primary brain tumor in adults and a highly lethal malignancy with a median survival of about 15 months. The aggressive invasion of the surrounding normal brain makes complete surgical resection impossible, increases the resistance to radiation and chemotherapy, and assures tumor recurrence. Thus, there is an urgent need to develop innovative therapeutics to target the invasive tumor cells for improved treatment outcomes of this disease. Expression of TROY (TNFRSF19), a member of the tumor necrosis factor (TNF) receptor family, increases with increasing glial tumor grade and inversely correlates with patient survival. Increased expression of TROY stimulates glioblastoma cell invasion in vitro and in vivo and increases resistance to temozolomide and radiation therapy. Conversely, silencing TROY expression inhibits glioblastoma cell invasion, increases temozolomide sensitivity, and prolongs survival in an intracranial xenograft model. Here, a novel complex is identified between TROY and EGFR, which is mediated predominantly by the cysteine-rich CRD3 domain of TROY. Glioblastoma tumors with elevated TROY expression have a statistically positive correlation with increased EGFR expression. TROY expression significantly increases the capacity of EGF to stimulate glioblastoma cell invasion, whereas depletion of TROY expression blocks EGF stimulation of glioblastoma cell invasion. Mechanistically, TROY expression modulates EGFR signaling by facilitating EGFR activation and delaying EGFR receptor internalization. Moreover, the association of EGFR with TROY increases TROY-induced NF-κB activation. These findings substantiate a critical role for the TROY-EGFR complex in regulation of glioblastoma cell invasion.Implications: The TROY-EGFR signaling complex emerges as a potential therapeutic target to inhibit glioblastoma cell invasion. Mol Cancer Res; 16(2); 322-32. ©2017 AACR.
Collapse
Affiliation(s)
- Zonghui Ding
- Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Alison Roos
- Departments of Cancer Biology and Neurosurgery, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Jean Kloss
- Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Harshil Dhruv
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Sen Peng
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona
| | - Patrick Pirrotte
- Cancer and Cell Biology Division, Translational Genomics Research Institute, Phoenix, Arizona.,Center for Proteomics, Translational Genomics Research Institute, Phoenix, Arizona
| | - Jennifer M Eschbacher
- Department of Neuropathology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Nhan L Tran
- Departments of Cancer Biology and Neurosurgery, Mayo Clinic Arizona, Scottsdale, Arizona
| | - Joseph C Loftus
- Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Scottsdale, Arizona.
| |
Collapse
|
14
|
Wahlbuhl-Becker M, Faschingbauer F, Beckmann MW, Schneider H. Hypohidrotic Ectodermal Dysplasia: Breastfeeding Complications Due to Impaired Breast Development. Geburtshilfe Frauenheilkd 2017; 77:377-382. [PMID: 28553001 PMCID: PMC5406230 DOI: 10.1055/s-0043-100106] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/15/2016] [Accepted: 01/01/2017] [Indexed: 01/23/2023] Open
Abstract
Background X-linked hypohidrotic ectodermal dysplasia (XLHED), the most common form of ectodermal dysplasia, is caused by mutations in the gene EDA. While only affected men develop the full-blown clinical picture, females who are heterozygous for an EDA mutation often also show symptoms such as hypodontia, hypotrichosis and hypohidrosis. These women may also suffer from malformations of the mammary gland which represent not just a cosmetic problem but can limit their breastfeeding capability. This paper summarizes the findings of the first systematic study on the impact of hypohidrotic ectodermal dysplasia on breastfeeding. Patients Thirty-eight adult female members of the German-Swiss-Austrian ectodermal dysplasia patient support group participated in a structured interview; most of them also agreed to a photodocumentation of their mammary region. Thirty-one women carried mutations in EDA (Group A) and seven were affected by other forms of hypohidrotic ectodermal dysplasia (Group B). Results 39 % of the women of Group A reported that their breasts were of different size or entirely absent on one side. In Group B, 86 % of the women reported differently sized or even absent breasts; two of these women lacked both breasts entirely. Most women described their nipples as exceptionally flat. 10 % of the women of Group A had more than two nipples. The high percentage of deviations from the norm was confirmed in the photodocumentation. Both groups had few or no sebaceous glands of Montgomery in the areolar region. Around 80 % of interviewed women had children and had attempted to breastfeed their first child. 67 % of the mothers in Group A had had difficulty in breastfeeding their infants and generally attributed this difficulty to their flat nipples. All of the mothers in Group B reported difficulties in breastfeeding; 60 % had not been able to breastfeed their first child. Conclusion Mothers with hypohidrotic ectodermal dysplasia very often have difficulty in breastfeeding because of their impaired breast development. This causal relationship needs to be taken into account in lactation counseling.
Collapse
Affiliation(s)
- Mandy Wahlbuhl-Becker
- Universitätsklinikum Erlangen, Kinder- und Jugendklinik, Kompetenzzentrum für Ektodermale Dysplasien, Erlangen, Germany
| | | | | | - Holm Schneider
- Universitätsklinikum Erlangen, Kinder- und Jugendklinik, Kompetenzzentrum für Ektodermale Dysplasien, Erlangen, Germany
| |
Collapse
|
15
|
|
16
|
Lefebvre S, Mikkola ML. Ectodysplasin research—Where to next? Semin Immunol 2014; 26:220-8. [DOI: 10.1016/j.smim.2014.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 05/08/2014] [Indexed: 01/29/2023]
|
17
|
Glazer AM, Cleves PA, Erickson PA, Lam AY, Miller CT. Parallel developmental genetic features underlie stickleback gill raker evolution. EvoDevo 2014; 5:19. [PMID: 24851181 PMCID: PMC4029907 DOI: 10.1186/2041-9139-5-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 04/23/2014] [Indexed: 01/08/2023] Open
Abstract
Background Convergent evolution, the repeated evolution of similar phenotypes in independent lineages, provides natural replicates to study mechanisms of evolution. Cases of convergent evolution might have the same underlying developmental and genetic bases, implying that some evolutionary trajectories might be predictable. In a classic example of convergent evolution, most freshwater populations of threespine stickleback fish have independently evolved a reduction of gill raker number to adapt to novel diets. Gill rakers are a segmentally reiterated set of dermal bones important for fish feeding. A previous large quantitative trait locus (QTL) mapping study using a marine × freshwater F2 cross identified QTL on chromosomes 4 and 20 with large effects on evolved gill raker reduction. Results By examining skeletal morphology in adult and developing sticklebacks, we find heritable marine/freshwater differences in gill raker number and spacing that are specified early in development. Using the expression of the Ectodysplasin receptor (Edar) gene as a marker of raker primordia, we find that the differences are present before the budding of gill rakers occurs, suggesting an early change to a lateral inhibition process controlling raker primordia spacing. Through linkage mapping in F2 fish from crosses with three independently derived freshwater populations, we find in all three crosses QTL overlapping both previously identified QTL on chromosomes 4 and 20 that control raker number. These two QTL affect the early spacing of gill raker buds. Conclusions Collectively, these data demonstrate that parallel developmental genetic features underlie the convergent evolution of gill raker reduction in freshwater sticklebacks, suggesting that even highly polygenic adaptive traits can have a predictable developmental genetic basis.
Collapse
Affiliation(s)
- Andrew M Glazer
- Molecular and Cell Biology Department, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Phillip A Cleves
- Molecular and Cell Biology Department, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Priscilla A Erickson
- Molecular and Cell Biology Department, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Angela Y Lam
- Molecular and Cell Biology Department, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Craig T Miller
- Molecular and Cell Biology Department, University of California-Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
18
|
Henningsen E, Svendsen MT, Lildballe DL, Jensen PKA. A novel mutation in theEDARgene causes severe autosomal recessive hypohidrotic ectodermal dysplasia. Am J Med Genet A 2014; 164A:2059-61. [DOI: 10.1002/ajmg.a.36582] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 03/16/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Emil Henningsen
- Department of Dermatology and Allergy Centre; Odense University Hospital; Odense Denmark
| | | | | | | |
Collapse
|
19
|
Schön S, Flierman I, Ofner A, Stahringer A, Holdt LM, Kolligs FT, Herbst A. β-catenin regulates NF-κB activityviaTNFRSF19 in colorectal cancer cells. Int J Cancer 2014; 135:1800-11. [DOI: 10.1002/ijc.28839] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 02/26/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Simon Schön
- Department of Medicine II; University of Munich; Munich Germany
| | | | - Andrea Ofner
- Department of Medicine II; University of Munich; Munich Germany
| | - Anika Stahringer
- Institute of Laboratory Medicine; University of Munich; Munich Germany
| | | | | | - Andreas Herbst
- Department of Medicine II; University of Munich; Munich Germany
| |
Collapse
|
20
|
Laugel-Haushalter V, Paschaki M, Marangoni P, Pilgram C, Langer A, Kuntz T, Demassue J, Morkmued S, Choquet P, Constantinesco A, Bornert F, Schmittbuhl M, Pannetier S, Viriot L, Hanauer A, Dollé P, Bloch-Zupan A. RSK2 is a modulator of craniofacial development. PLoS One 2014; 9:e84343. [PMID: 24416220 PMCID: PMC3885557 DOI: 10.1371/journal.pone.0084343] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 11/21/2013] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The RSK2 gene is responsible for Coffin-Lowry syndrome, an X-linked dominant genetic disorder causing mental retardation, skeletal growth delays, with craniofacial and digital abnormalities typically associated with this syndrome. Craniofacial and dental anomalies encountered in this rare disease have been poorly characterized. METHODOLOGY/PRINCIPAL FINDINGS We examined, using X-Ray microtomographic analysis, the variable craniofacial dysmorphism and dental anomalies present in Rsk2 knockout mice, a model of Coffin-Lowry syndrome, as well as in triple Rsk1,2,3 knockout mutants. We report Rsk mutation produces surpernumerary teeth midline/mesial to the first molar. This highly penetrant phenotype recapitulates more ancestral tooth structures lost with evolution. Most likely this leads to a reduction of the maxillary diastema. Abnormalities of molar shape were generally restricted to the mesial part of both upper and lower first molars (M1). Expression analysis of the four Rsk genes (Rsk1, 2, 3 and 4) was performed at various stages of odontogenesis in wild-type (WT) mice. Rsk2 is expressed in the mesenchymal, neural crest-derived compartment, correlating with proliferative areas of the developing teeth. This is consistent with RSK2 functioning in cell cycle control and growth regulation, functions potentially responsible for severe dental phenotypes. To uncover molecular pathways involved in the etiology of these defects, we performed a comparative transcriptomic (DNA microarray) analysis of mandibular wild-type versus Rsk2-/Y molars. We further demonstrated a misregulation of several critical genes, using a Rsk2 shRNA knock-down strategy in molar tooth germs cultured in vitro. CONCLUSIONS This study reveals RSK2 regulates craniofacial development including tooth development and patterning via novel transcriptional targets.
Collapse
Affiliation(s)
- Virginie Laugel-Haushalter
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR 7104), Institut National de la Santé et de la Recherche Médicale (U 964), University of Strasbourg, Illkirch, France
| | - Marie Paschaki
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR 7104), Institut National de la Santé et de la Recherche Médicale (U 964), University of Strasbourg, Illkirch, France
| | - Pauline Marangoni
- Team «Evo-Devo of Vertebrate Dentition», Institut de Génomique Fonctionnelle de Lyon, Unité Mixte de Recherche 5242 Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | - Coralie Pilgram
- Faculty of Dentistry, University of Strasbourg, Strasbourg France
| | - Arnaud Langer
- Faculty of Dentistry, University of Strasbourg, Strasbourg France
| | - Thibaut Kuntz
- Faculty of Dentistry, University of Strasbourg, Strasbourg France
| | - Julie Demassue
- Faculty of Dentistry, University of Strasbourg, Strasbourg France
| | - Supawich Morkmued
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR 7104), Institut National de la Santé et de la Recherche Médicale (U 964), University of Strasbourg, Illkirch, France
- Faculty of Dentistry, University of Strasbourg, Strasbourg France
- Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Philippe Choquet
- UF6237 Preclinical Imaging Lab, Biophysics and Nuclear Medicine, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg, France; ICube, CNRS, University of Strasbourg, Strasbourg, France
| | - André Constantinesco
- UF6237 Preclinical Imaging Lab, Biophysics and Nuclear Medicine, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg, France; ICube, CNRS, University of Strasbourg, Strasbourg, France
| | - Fabien Bornert
- Faculty of Dentistry, University of Strasbourg, Strasbourg France
- INSERM U1121, "Biomaterials and Bioengineering", University of Strasbourg, Strasbourg, France
| | - Matthieu Schmittbuhl
- Faculty of Dentistry, University of Strasbourg, Strasbourg France
- Reference Centre for Orodental Manifestations of Rare Diseases, Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg, France
- INSERM U1121, "Biomaterials and Bioengineering", University of Strasbourg, Strasbourg, France
| | - Solange Pannetier
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR 7104), Institut National de la Santé et de la Recherche Médicale (U 964), University of Strasbourg, Illkirch, France
| | - Laurent Viriot
- Team «Evo-Devo of Vertebrate Dentition», Institut de Génomique Fonctionnelle de Lyon, Unité Mixte de Recherche 5242 Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon, Claude Bernard Lyon 1 University, Lyon, France
| | - André Hanauer
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR 7104), Institut National de la Santé et de la Recherche Médicale (U 964), University of Strasbourg, Illkirch, France
| | - Pascal Dollé
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR 7104), Institut National de la Santé et de la Recherche Médicale (U 964), University of Strasbourg, Illkirch, France
| | - Agnès Bloch-Zupan
- Institute of Genetics and Molecular and Cellular Biology (IGBMC), Centre National de la Recherche Scientifique (UMR 7104), Institut National de la Santé et de la Recherche Médicale (U 964), University of Strasbourg, Illkirch, France
- Faculty of Dentistry, University of Strasbourg, Strasbourg France
- Reference Centre for Orodental Manifestations of Rare Diseases, Pôle de Médecine et Chirurgie Bucco-dentaires, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg, France
- * E-mail:
| |
Collapse
|
21
|
Howard BA, Lu P. Stromal regulation of embryonic and postnatal mammary epithelial development and differentiation. Semin Cell Dev Biol 2014; 25-26:43-51. [DOI: 10.1016/j.semcdb.2014.01.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/19/2013] [Accepted: 01/09/2014] [Indexed: 01/06/2023]
|
22
|
Lindfors PH, Voutilainen M, Mikkola ML. Ectodysplasin/NF-κB signaling in embryonic mammary gland development. J Mammary Gland Biol Neoplasia 2013; 18:165-9. [PMID: 23591968 DOI: 10.1007/s10911-013-9277-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 03/27/2013] [Indexed: 12/13/2022] Open
Abstract
The ectodysplasin (Eda) signaling pathway consists of a TNF-like ligand Eda, its receptor Edar, and an adaptor protein Edaradd and its activation leads to NF-κB mediated transcription. In humans, mutations in the EDA pathway genes cause hypohidrotic ectodermal dysplasia, a disorder characterized by defective formation of hair follicles, teeth, and several exocrine glands including the breast. Embryonic mammary gland development proceeds via placode, bud, bulb and sprout stages before the onset of branching morphogenesis. Studies on mouse models have linked Eda with two aspects of embryonic mammary gland morphogenesis: placode induction and ductal growth and branching. Here we summarize the current knowledge on the role of Eda/NF-κB in mammary gland development.
Collapse
Affiliation(s)
- Päivi H Lindfors
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, P.O. Box 56, 00014, Helsinki, Finland
| | | | | |
Collapse
|
23
|
Loftus JC, Dhruv H, Tuncali S, Kloss J, Yang Z, Schumacher CA, Cao B, Williams BO, Eschbacher JM, Ross JTD, Tran NL. TROY (TNFRSF19) promotes glioblastoma survival signaling and therapeutic resistance. Mol Cancer Res 2013; 11:865-74. [PMID: 23699535 DOI: 10.1158/1541-7786.mcr-13-0008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
UNLABELLED Of the features that characterize glioblastoma, arguably none is more clinically relevant than the propensity of malignant glioma cells to aggressively invade into the surrounding normal brain tissue. These invasive cells render complete resection impossible, confer significant resistance to chemo- and radiation-therapy, and virtually assure tumor recurrence. Expression of TROY (TNFRSF19), a member of the TNF receptor superfamily, inversely correlates with patient survival and stimulates glioblastoma cell migration and invasion in vitro. In this study, we report that TROY is overexpressed in glioblastoma tumor specimens and TROY mRNA expression is increased in the invasive cell population in vivo. In addition, inappropriate expression of TROY in mouse astrocytes in vivo using glial-specific gene transfer in transgenic mice induces astrocyte migration within the brain, validating the importance of the TROY signaling cascade in glioblastoma cell migration and invasion. Knockdown of TROY expression in primary glioblastoma xenografts significantly prolonged survival in vivo. Moreover, TROY expression significantly increased resistance of glioblastoma cells to both IR- and TMZ-induced apoptosis via activation of Akt and NF-κB. Inhibition of either Akt or NF-κB activity suppressed the survival benefits of TROY signaling in response to TMZ treatment. These findings position aberrant expression and/or signaling by TROY as a contributor to the dispersion of glioblastoma cells and therapeutic resistance. IMPLICATIONS Targeting of TROY may increase tumor vulnerability and improve therapeutic response in glioblastoma. Mol Cancer Res; 11(8); 865-74. ©2013 AACR.
Collapse
Affiliation(s)
- Joseph C Loftus
- The Translational Genomics Research Institute, 445 North Fifth Street, Suite 400, Phoenix, AZ 85004, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kamberov YG, Wang S, Tan J, Gerbault P, Wark A, Tan L, Yang Y, Li S, Tang K, Chen H, Powell A, Itan Y, Fuller D, Lohmueller J, Mao J, Schachar A, Paymer M, Hostetter E, Byrne E, Burnett M, McMahon AP, Thomas MG, Lieberman DE, Jin L, Tabin CJ, Morgan BA, Sabeti PC. Modeling recent human evolution in mice by expression of a selected EDAR variant. Cell 2013; 152:691-702. [PMID: 23415220 DOI: 10.1016/j.cell.2013.01.016] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 11/22/2012] [Accepted: 01/04/2013] [Indexed: 11/27/2022]
Abstract
An adaptive variant of the human Ectodysplasin receptor, EDARV370A, is one of the strongest candidates of recent positive selection from genome-wide scans. We have modeled EDAR370A in mice and characterized its phenotype and evolutionary origins in humans. Our computational analysis suggests the allele arose in central China approximately 30,000 years ago. Although EDAR370A has been associated with increased scalp hair thickness and changed tooth morphology in humans, its direct biological significance and potential adaptive role remain unclear. We generated a knockin mouse model and find that, as in humans, hair thickness is increased in EDAR370A mice. We identify new biological targets affected by the mutation, including mammary and eccrine glands. Building on these results, we find that EDAR370A is associated with an increased number of active eccrine glands in the Han Chinese. This interdisciplinary approach yields unique insight into the generation of adaptive variation among modern humans.
Collapse
Affiliation(s)
- Yana G Kamberov
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Laugel-Haushalter V, Paschaki M, Thibault-Carpentier C, Dembelé D, Dollé P, Bloch-Zupan A. Molars and incisors: show your microarray IDs. BMC Res Notes 2013; 6:113. [PMID: 23531410 PMCID: PMC3658942 DOI: 10.1186/1756-0500-6-113] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 03/08/2013] [Indexed: 01/31/2023] Open
Abstract
Background One of the key questions in developmental biology is how, from a relatively small number of conserved signaling pathways, is it possible to generate organs displaying a wide range of shapes, tissue organization, and function. The dentition and its distinct specific tooth types represent a valuable system to address the issues of differential molecular signatures. To identify such signatures, we performed a comparative transcriptomic analysis of developing murine lower incisors, mandibular molars and maxillary molars at the developmental cap stage (E14.5). Results 231 genes were identified as being differentially expressed between mandibular incisors and molars, with a fold change higher than 2 and a false discovery rate lower than 0.1, whereas only 96 genes were discovered as being differentially expressed between mandibular and maxillary molars. Numerous genes belonging to specific signaling pathways (the Hedgehog, Notch, Wnt, FGF, TGFβ/BMP, and retinoic acid pathways), and/or to the homeobox gene superfamily, were also uncovered when a less stringent fold change threshold was used. Differential expressions for 10 out of 12 (mandibular incisors versus molars) and 9 out of 10 selected genes were confirmed by quantitative reverse transcription-PCR (qRT-PCR). A bioinformatics tool (Ingenuity Pathway Analysis) used to analyze biological functions and pathways on the group of incisor versus molar differentially expressed genes revealed that 143 genes belonged to 9 networks with intermolecular connections. Networks with the highest significance scores were centered on the TNF/NFκB complex and the ERK1/2 kinases. Two networks ERK1/2 kinases and tretinoin were involved in differential molar morphogenesis. Conclusion These data allowed us to build several regulatory networks that may distinguish incisor versus molar identity, and may be useful for further investigations of these tooth-specific ontogenetic programs. These programs may be dysregulated in transgenic animal models and related human diseases leading to dental anomalies.
Collapse
Affiliation(s)
- Virginie Laugel-Haushalter
- Developmental Biology and Stem Cells Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, BP 10142, 1 rue Laurent Fries, Illkirch Cedex, 67404, France
| | | | | | | | | | | |
Collapse
|
26
|
Ookuma YF, Kiyoshima T, Kobayashi I, Nagata K, Wada H, Fujiwara H, Yamaza H, Nonaka K, Sakai H. Multiple functional involvement of Thymosin beta-4 in tooth germ development. Histochem Cell Biol 2012; 139:355-70. [DOI: 10.1007/s00418-012-1033-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2012] [Indexed: 12/17/2022]
|
27
|
Ectodysplasin regulates hormone-independent mammary ductal morphogenesis via NF-κB. Proc Natl Acad Sci U S A 2012; 109:5744-9. [PMID: 22451941 DOI: 10.1073/pnas.1110627109] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ductal growth of the mammary gland occurs in two distinct stages. The first round of branching morphogenesis occurs during embryogenesis, and the second round commences at the onset of puberty. Currently, relatively little is known about the genetic networks that control the initial phases of ductal expansion, which, unlike pubertal development, proceeds independent of hormonal input in female mice. Here we identify NF-κB downstream of the TNF-like ligand ectodysplasin (Eda) as a unique regulator of embryonic and prepubertal ductal morphogenesis. Loss of Eda, or inhibition of NF-κB, led to smaller ductal trees with fewer branches. On the other hand, overexpression of Eda caused a dramatic NF-κB-dependent phenotype in both female and male mice characterized by precocious and highly increased ductal growth and branching that correlated with enhanced cell proliferation. We have identified several putative transcriptional target genes of Eda/NF-κB, including PTHrP, Wnt10a, and Wnt10b, as well as Egf family ligands amphiregulin and epigen. We developed a mammary bud culture system that allowed us to manipulate mammary development ex vivo and found that recombinant PTHrP, Wnt3A, and Egf family ligands stimulate embryonic branching morphogenesis, suggesting that these pathways may cooperatively mediate the effects of Eda.
Collapse
|
28
|
Hasenpusch-Theil K, Magnani D, Amaniti EM, Han L, Armstrong D, Theil T. Transcriptional analysis of Gli3 mutants identifies Wnt target genes in the developing hippocampus. ACTA ACUST UNITED AC 2012; 22:2878-93. [PMID: 22235033 DOI: 10.1093/cercor/bhr365] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Early development of the hippocampus, which is essential for spatial memory and learning, is controlled by secreted signaling molecules of the Wnt gene family and by Wnt/β-catenin signaling. Despite its importance, little is known, however, about Wnt-regulated genes during hippocampal development. Here, we used the Gli3 mutant mouse extra-toes (Xt(J)), in which Wnt gene expression in the forebrain is severely affected, as a tool in a microarray analyses to identify potential Wnt target genes. This approach revealed 53 candidate genes with restricted or graded expression patterns in the dorsomedial telencephalon. We identified conserved Tcf/Lef-binding sites in telencephalon-specific enhancers of several of these genes, including Dmrt3, Gli3, Nfia, and Wnt8b. Binding of Lef1 to these sites was confirmed using electrophoretic mobility shift assays. Mutations in these Tcf/Lef-binding sites disrupted or reduced enhancer activity in vivo. Moreover, ectopic activation of Wnt/β-catenin signaling in an ex vivo explant system led to increased telencephalic expression of these genes. Finally, conditional inactivation of Gli3 results in defective hippocampal growth. Collectively, these data strongly suggest that we have identified a set of direct Wnt target genes in the developing hippocampus and provide inside into the genetic hierarchy underlying Wnt-regulated hippocampal development.
Collapse
|
29
|
Twohig JP, Cuff SM, Yong AA, Wang ECY. The role of tumor necrosis factor receptor superfamily members in mammalian brain development, function and homeostasis. Rev Neurosci 2011; 22:509-33. [PMID: 21861782 DOI: 10.1515/rns.2011.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tumor necrosis factor receptor superfamily (TNFRSF) members were initially identified as immunological mediators, and are still commonly perceived as immunological molecules. However, our understanding of the diversity of TNFRSF members' roles in mammalian physiology has grown significantly since the first discovery of TNFRp55 (TNFRSF1) in 1975. In particular, the last decade has provided evidence for important roles in brain development, function and the emergent field of neuronal homeostasis. Recent evidence suggests that TNFRSF members are expressed in an overlapping regulated pattern during neuronal development, participating in the regulation of neuronal expansion, growth, differentiation and regional pattern development. This review examines evidence for non-immunological roles of TNFRSF members in brain development, function and maintenance under normal physiological conditions. In addition, several aspects of brain function during inflammation will also be described, when illuminating and relevant to the non-immunological role of TNFRSF members. Finally, key questions in the field will be outlined.
Collapse
Affiliation(s)
- Jason P Twohig
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, Wales, UK
| | | | | | | |
Collapse
|
30
|
Häärä O, Fujimori S, Schmidt-Ullrich R, Hartmann C, Thesleff I, Mikkola ML. Ectodysplasin and Wnt pathways are required for salivary gland branching morphogenesis. Development 2011; 138:2681-91. [PMID: 21652647 DOI: 10.1242/dev.057711] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The developing submandibular salivary gland (SMG) is a well-studied model for tissue interactions and branching morphogenesis. Its development shares similar features with other ectodermal appendages such as hair and tooth. The ectodysplasin (Eda) pathway is essential for the formation and function of several ectodermal organs. Mutations in the signaling components of the Eda pathway lead to a human syndrome known as hypohidrotic ectodermal dysplasia (HED), which is characterized by missing and malformed teeth, sparse hair and reduced sweating. Individuals with HED suffer also from dry mouth because of reduced saliva flow. In order to understand the underlying mechanism, we analyzed salivary gland development in mouse models with altered Eda pathway activities. We have found that Eda regulates growth and branching of the SMG via transcription factor NF-κB in the epithelium, and that the hedgehog pathway is an important mediator of Eda/NF-κB. We also sought to determine whether a similar reciprocal interplay between the Eda and Wnt/β-catenin pathways, which are known to operate in other skin appendages, functions in developing SMG. Surprisingly and unlike in developing hair follicles and teeth, canonical Wnt signaling activity did not colocalize with Edar/NF-κB in salivary gland epithelium. Instead, we observed high mesenchymal Wnt activity and show that ablation of mesenchymal Wnt signaling either in vitro or in vivo compromised branching morphogenesis. We also provide evidence suggesting that the effects of mesenchymal Wnt/β-catenin signaling are mediated, at least in part, through regulation of Eda expression.
Collapse
Affiliation(s)
- Otso Häärä
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, P.O.B. 56, 00014 Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
31
|
Porntaveetus T, Ohazama A, Choi HY, Herz J, Sharpe PT. Wnt signaling in the murine diastema. Eur J Orthod 2011; 34:518-24. [PMID: 21531785 DOI: 10.1093/ejo/cjr049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The correct number and shape of teeth are critical factors for an aesthetic and functional dentition. Understanding the molecular mechanisms regulating tooth number and shape are therefore important in orthodontics. Mice have only one incisor and three molars in each jaw quadrant that are divided by a tooth-less region, the diastema. Although mice lost teeth in the diastema during evolution, the remnants of the evolutionary lost teeth are observed as transient epithelial buds in the wild-type diastema during early stages of development. Shh and Fgf signaling pathways that are essential for tooth development have been shown to be repressed in the diastema. It remains unclear however how Wnt signaling, that is also required for tooth development, is regulated in the diastema. In this study we found that in the embryonic diastema, Wnt5a expression was observed in mesenchyme, whereas Wnt4 and Wnt10b were expressed in epithelium. The expression of Wnt6 and Wnt11 was found in both tissues. The Wnt co-receptor, Lrp6, was weakly expressed in the diastema overlapping with weak Lrp4 expression, a co-receptor that inhibits Wnt signaling. Secreted Wnt inihibitors Dkk1, Dkk2, and Dkk3 were also expressed in the diastema. Lrp4 mutant mice develop supernumerary teeth in the diastema that is accompanied by upregulation of Wnt signaling and Lrp6 expression. Wnt signaling is thus usually attenuated in the diastema by these secreted and membrane bound Wnt inhibitors.
Collapse
Affiliation(s)
- Thantrira Porntaveetus
- Department of Craniofacial Development, Dental Institute, King's College London, Guy's Hospital, London, UK
| | | | | | | | | |
Collapse
|
32
|
Wells KL, Mou C, Headon DJ, Tucker AS. Recombinant EDA or Sonic Hedgehog rescue the branching defect in Ectodysplasin A pathway mutant salivary glands in vitro. Dev Dyn 2011; 239:2674-84. [PMID: 20803597 DOI: 10.1002/dvdy.22406] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Hypohidrotic ectodermal dysplasia (HED) is characterized by defective ectodermal organ development. This includes the salivary glands (SGs), which have an important role in lubricating the oral cavity. In humans and mice, HED is caused by mutations in Ectodysplasin A (Eda) pathway genes. Various phenotypes of the mutant mouse Eda(Ta/Ta), which lacks the ligand Eda, can be rescued by maternal injection or in vitro culture supplementation with recombinant EDA. However, the response of the SGs to this treatment has not been investigated. Here, we show that the submandibular glands (SMGs) of Eda(Ta/Ta) mice exhibit impaired branching morphogenesis, and that supplementation of Eda(Ta/Ta) SMG explants with recombinant EDA rescues the defect. Supplementation of Edar(dlJ/dlJ) SMGs with recombinant Sonic hedgehog (Shh) also rescues the defect, whereas treatment with recombinant Fgf8 does not. This work is the first to test the ability of putative Eda target molecules to rescue Eda pathway mutant SMGs.
Collapse
Affiliation(s)
- K L Wells
- Department of Craniofacial Development, Dental Institute, King's College London, London, United Kingdom
| | | | | | | |
Collapse
|
33
|
Li XY, Swaggerty CL, Kogut MH, Chiang HI, Wang Y, Genovese KJ, He H, Pevzner IY, Zhou HJ. Caecal transcriptome analysis of colonized and non-colonized chickens within two genetic lines that differ in caecal colonization by Campylobacter jejuni. Anim Genet 2011; 42:491-500. [PMID: 21906100 DOI: 10.1111/j.1365-2052.2010.02168.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Campylobacter jejuni is one of the most common causes of human bacterial enteritis worldwide. The molecular mechanisms of the host responses of chickens to C. jejuni colonization are not well understood. We have previously found differences in C. jejuni colonization at 7-days post-inoculation (pi) between two genetic broiler lines. However, within each line, not all birds were colonized by C. jejuni (27.5% colonized in line A, and 70% in line B). Therefore, the objective of the present experiments was to further define the differences in host gene expression between colonized and non-colonized chickens within each genetic line. RNA isolated from ceca of colonized and non-colonized birds within each line was applied to a chicken 44K Agilent microarray for the pair comparison. There were differences in the mechanisms of host resistant to C. jejuni colonization between line A and line B. Ten times more differentially expressed genes were observed between colonized and non-colonized chickens within line B than those within line A. Our study supports the fact that the MAPK pathway is important in host response to C. jejuni colonization in line B, but not in line A. The data indicate that inhibition of small GTPase-mediated signal transduction could enhance the resistance of chickens to C. jejuni colonization and that the tumour necrosis factor receptor superfamily genes play important roles in determining C. jejuni non-colonization in broilers.
Collapse
Affiliation(s)
- X Y Li
- Department of Poultry Science, Texas A&M University, College Station, TX 77843, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Larsen M, Yamada KM, Musselmann K. Systems analysis of salivary gland development and disease. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2010; 2:670-82. [PMID: 20890964 PMCID: PMC3398465 DOI: 10.1002/wsbm.94] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Branching morphogenesis is a crucial developmental process in which vertebrate organs generate extensive epithelial surface area while retaining a compact size. In the vertebrate submandibular salivary gland, branching morphogenesis is crucial for the generation of the large surface area necessary to produce sufficient saliva. However, in many salivary gland diseases, saliva-producing acinar cells are destroyed, resulting in dry mouth and secondary health conditions. Systems-based approaches can provide insights into understanding salivary gland development, function, and disease. The traditional approach to understanding these processes is the identification of molecular signals using reductionist approaches; we review current progress with such methods in understanding salivary gland development. Taking a more global approach, multiple groups are currently profiling the transcriptome, the proteome, and other 'omes' in both developing mouse tissues and in human patient samples. Computational methods have been successful in deciphering large data sets, and mathematical models are starting to make predictions regarding the contribution of molecules to the physical processes of morphogenesis and cellular function. A challenge for the future will be to establish comprehensive, publicly accessible salivary gland databases spanning the full range of genes and proteins; plans are underway to provide these resources to researchers in centralized repositories. The greatest challenge for the future will be to develop realistic models that integrate multiple types of data to both describe and predict embryonic development and disease pathogenesis.
Collapse
Affiliation(s)
- Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York
| | - Kenneth M. Yamada
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health
| | - Kurt Musselmann
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health
| |
Collapse
|
35
|
Paulino VM, Yang Z, Kloss J, Ennis MJ, Armstrong BA, Loftus JC, Tran NL. TROY (TNFRSF19) is overexpressed in advanced glial tumors and promotes glioblastoma cell invasion via Pyk2-Rac1 signaling. Mol Cancer Res 2010; 8:1558-67. [PMID: 20881009 DOI: 10.1158/1541-7786.mcr-10-0334] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A critical problem in the treatment of malignant gliomas is the extensive infiltration of individual tumor cells into adjacent brain tissues. This invasive phenotype severely limits all current therapies, and to date, no treatment is available to control the spread of this disease. Members of the tumor necrosis factor (TNF) ligand superfamily and their cognate receptors regulate various cellular responses including proliferation, migration, differentiation, and apoptosis. Specifically, the TNFRSF19/TROY gene encodes a type I cell surface receptor that is expressed on migrating or proliferating progenitor cells of the hippocampus, thalamus, and cerebral cortex. Here, we show that levels of TROY mRNA expression directly correlate with increasing glial tumor grade. Among malignant gliomas, TROY expression correlates inversely with overall patient survival. In addition, we show that TROY overexpression in glioma cells activates Rac1 signaling in a Pyk2-dependent manner to drive glioma cell invasion and migration. Pyk2 coimmunoprecipitates with the TROY receptor, and depletion of Pyk2 expression by short hairpin RNA interference oligonucleotides inhibits TROY-induced Rac1 activation and subsequent cellular migration. These findings position aberrant expression and/or signaling by TROY as a contributor, and possibly as a driver, of the malignant dispersion of glioma cells.
Collapse
Affiliation(s)
- Vincent M Paulino
- The Translational Genomics Research Institute, 445 North Fifth Street, Suite 400, Phoenix, AZ 85004, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Age-dependent maintenance of motor control and corticostriatal innervation by death receptor 3. J Neurosci 2010; 30:3782-92. [PMID: 20220013 DOI: 10.1523/jneurosci.1928-09.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Death receptor 3 is a proinflammatory member of the immunomodulatory tumor necrosis factor receptor superfamily, which has been implicated in several inflammatory diseases such as arthritis and inflammatory bowel disease. Intriguingly however, constitutive DR3 expression has been detected in the brains of mice, rats, and humans, although its neurological function remains unknown. By mapping the normal brain expression pattern of DR3, we found that DR3 is expressed specifically by cells of the neuron lineage in a developmentally regulated and region-specific pattern. Behavioral studies on DR3-deficient (DR3(ko)) mice showed that constitutive neuronal DR3 expression was required for stable motor control function in the aging adult. DR3(ko) mice progressively developed behavioral defects characterized by altered gait, dyskinesia, and hyperactivity, which were associated with elevated dopamine and lower serotonin levels in the striatum. Importantly, retrograde tracing showed that absence of DR3 expression led to the loss of corticostriatal innervation without significant neuronal loss in aged DR3(ko) mice. These studies indicate that DR3 plays a key nonredundant role in the retention of normal motor control function during aging in mice and implicate DR3 in progressive neurological disease.
Collapse
|
37
|
Qiu W, Hu Y, Andersen TE, Jafari A, Li N, Chen W, Kassem M. Tumor necrosis factor receptor superfamily member 19 (TNFRSF19) regulates differentiation fate of human mesenchymal (stromal) stem cells through canonical Wnt signaling and C/EBP. J Biol Chem 2010; 285:14438-49. [PMID: 20223822 DOI: 10.1074/jbc.m109.052001] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mechanisms controlling human multipotent mesenchymal (stromal) stem cell (hMSC) differentiation into osteoblasts or adipocytes are poorly understood. We have previously demonstrated that Wnt signaling in hMSC enhanced osteoblast differentiation and inhibited adipogenesis by comparing two hMSC cell lines overexpressing mutated forms of the Wnt co-receptor LRP5: T253I (hMSC-LRP5(T253)) and T244M (hMSC-LRP5(T244)) conducting high and low level of Wnt signaling, respectively. To explore the underlying molecular mechanisms, we compared gene expression profiles of hMSC-LRP5(T253) and hMSC-LRP5(T244) treated with Wnt3a using whole genome expression microarrays and found that TNFRSF19 is differentially up-regulated between the two cells lines. Bioinformatic analysis and dual luciferase assay of its promoter revealed that TNFRSF19 transcript 2 (TNFRSF19.2) is a target of canonical Wnt signaling. Knocking down TNFRSF19 in hMSC-LRP5(T253) cells decreased Wnt3a-induced osteoblast differentiation marker alkaline phosphate activity and its overexpression in hMSC-LRP5(T244) cells increased alkaline phosphate activity. In addition, TNFRSF19 was negatively regulated by adipogenic transcription factor CCAAT/enhancer-binding proteins (C/EBP). Knocking down TNFRSF19 in hMSC-LRP5(T253) cells or its overexpression in hMSC-LRP5(T244) cells significantly increased or decreased adipogenesis, respectively. In conclusion, we revealed a novel function of TNFRSF19 as a factor mediating differentiation signals that determine the hMSC differentiating fate into osteoblasts or adipocytes.
Collapse
Affiliation(s)
- Weimin Qiu
- Laboratory for Molecular Endocrinology (KMEB), Department of Endocrinology and Metabolism, University Hospital of Odense, J. B. Winsløws Vej 25, 1, DK-5000 Odense C, Denmark
| | | | | | | | | | | | | |
Collapse
|
38
|
Priolo M. Ectodermal dysplasias: an overview and update of clinical and molecular-functional mechanisms. Am J Med Genet A 2010; 149A:2003-13. [PMID: 19504607 DOI: 10.1002/ajmg.a.32804] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The ectodermal dysplasias (EDs) are a large and complex group of disorders. In various combinations, they all share anomalies in hair, teeth, nails, and sweat gland function. The anomalies affecting the epidermis and epidermal appendages are extremely variable. Many are associated with malformations in other organs and systems. Clinical overlap is present among EDs. Few causative genes have been identified, to date. Most of the EDs present multisystem involvement with abnormal development of structures also derived from mesoderm. In the last few years, it has become evident that gene expression in the EDs is not limited to the ectoderm and that there is a concomitant effect on developing mesenchymal structures, with modification or abolition of ectodermal-mesenchymal signaling. It is possible to approach this group of diseases basing on functional and molecular findings and to begin to explain the complex clinical consequences of mutations affecting specific developmental pathways. We have reviewed the molecular basis of ectodermal dysplasias applying this new clinical-functional classification. For each subset of the identified ED, we will now describe the genes and related proteins involved in terms of: (1) structure of the genes and their role in differentiation of the epidermis and the ectodermal derivatives; (2) genotype-phenotype correlation.
Collapse
Affiliation(s)
- Manuela Priolo
- Operative Unit of Medical Genetics Bianchi-Melacrino-Morelli Hospital, Reggio Calabria, Italy.
| |
Collapse
|
39
|
Cao Z, Gao Y, Deng K, Williams G, Doherty P, Walsh FS. Receptors for myelin inhibitors: Structures and therapeutic opportunities. Mol Cell Neurosci 2009; 43:1-14. [PMID: 19619659 DOI: 10.1016/j.mcn.2009.07.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 05/28/2009] [Accepted: 07/10/2009] [Indexed: 11/19/2022] Open
Abstract
Many studies have indicated that the inability of adult mammalian central nervous system (CNS) to regenerate after injury is partly due to the existence of growth-inhibitory molecules associated with CNS myelin. Studies over the years have led to the identification of multiple myelin-associated inhibitors, among which Nogo, myelin-associated glycoprotein (MAG) and oligodendrocyte-myelin glycoprotein (Omgp) represent potentially major contributors to CNS axon regeneration failure. Here we review in vitro and in vivo investigations into these inhibitory ligands and their functional mechanisms, focusing particularly on the neuronal receptors that mediate the inhibitory signals from these myelin molecules. A better understanding of the receptors for myelin-associated inhibitors could provide opportunities to decipher the mechanism of restriction in CNS regeneration, and lead to the development of potential therapeutic targets in neurodegenerative diseases and neurological injury. We will discuss the structures of the receptors and therapeutic opportunities that might arise based on this information.
Collapse
Affiliation(s)
- Zixuan Cao
- Neuroscience Discovery, Wyeth Research, Princeton, NJ 08543, USA
| | | | | | | | | | | |
Collapse
|
40
|
Pantalacci S, Prochazka J, Martin A, Rothova M, Lambert A, Bernard L, Charles C, Viriot L, Peterkova R, Laudet V. Patterning of palatal rugae through sequential addition reveals an anterior/posterior boundary in palatal development. BMC DEVELOPMENTAL BIOLOGY 2008; 8:116. [PMID: 19087265 PMCID: PMC2637861 DOI: 10.1186/1471-213x-8-116] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Accepted: 12/16/2008] [Indexed: 01/12/2023]
Abstract
Background The development of the secondary palate has been a main topic in craniofacial research, as its failure results in cleft palate, one of the most common birth defects in human. Nevertheless, palatal rugae (or rugae palatinae), which are transversal ridges developing on the secondary palate, received little attention. However, rugae could be useful as landmarks to monitor anterior/posterior (A/P) palatal growth, and they provide a simple model of mesenchymal-epithelial structures arranged in a serial pattern. Results We first determined in which order the nine mouse rugae appear during development. Our results revealed a reiterative process, which is coupled with A/P growth of palatal shelves, and by which rugae 3 to 7b are sequentially interposed, in the increasing distance between the second most anterior ruga, ruga 2, and the two most posterior rugae, rugae 8 and 9. We characterized the steps of ruga interposition in detail, showing that a new ruga forms from an active zone of high proliferation rate, next to the last formed ruga. Then, by analyzing the polymorphism of wild type and EdaTa mutant mice, we suggest that activation-inhibition mechanisms may be involved in positioning new rugae, like for other skin appendages. Finally, we show that the ruga in front of which new rugae form, i.e. ruga 8 in mouse, coincides with an A/P gene expression boundary in the palatal shelves (Shox2/Meox2-Tbx22). This coincidence is significant, since we also found it in hamster, despite differences in the adult ruga pattern of these two species. Conclusion We showed that palatal rugae are sequentially added to the growing palate, in an interposition process that appears to be dependent on activation-inhibition mechanisms and reveals a new developmental boundary in the growing palate. Further studies on rugae may help to shed light on both the development and evolution of structures arranged in regular patterns. Moreover, rugae will undoubtedly be powerful tools to further study the anteroposterior regionalization of the growing palate.
Collapse
Affiliation(s)
- Sophie Pantalacci
- Molecular Zoology, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, Lyon, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mégarbané H, Cluzeau C, Bodemer C, Fraïtag S, Chababi-Atallah M, Mégarbané A, Smahi A. Unusual presentation of a severe autosomal recessive anhydrotic ectodermal dysplasia with a novel mutation in the EDAR gene. Am J Med Genet A 2008; 146A:2657-62. [PMID: 18816645 DOI: 10.1002/ajmg.a.32509] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We report on an 18-year-old woman, born to first-cousin parents, presenting with a severe form of anhydrotic ectodermal dysplasia (EDA/HED). She had sparse hair, absent limb hair, absent sweating, episodes of hyperpyrexia, important hypodontia, and hyperconvex nails. She also showed unusual clinical manifestations such as an absence of breasts, a rudimentary extranumerary areola and nipple on the left side, and marked palmo-plantar hyperkeratosis. Light microscopy of skin biopsies showed orthokeratotic hyperkeratosis and absence of sweat glands. A novel homozygous mutation (IVS9 + 1G > A) in the EDAR gene was identified. This mutation results in a total absence of EDAR transcripts and consequently of the EDAR protein, which likely results in abolition of all ectodysplasin-mediated NF-kappaB signaling. This is the first complete loss-of-function mutation in the EDAR gene reported to date, which may explain the unusual presentation of HED in this patient, enlarging the clinical spectrum linked to the dysfunction of the ectodysplasin mediated NF-kappaB signaling.
Collapse
Affiliation(s)
- Hala Mégarbané
- Unité de Génétique Médicale, Faculté de Médecine, Université Saint-Joseph, Beirut, Lebanon
| | | | | | | | | | | | | |
Collapse
|
42
|
Harris MP, Rohner N, Schwarz H, Perathoner S, Konstantinidis P, Nüsslein-Volhard C. Zebrafish eda and edar mutants reveal conserved and ancestral roles of ectodysplasin signaling in vertebrates. PLoS Genet 2008; 4:e1000206. [PMID: 18833299 PMCID: PMC2542418 DOI: 10.1371/journal.pgen.1000206] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2007] [Accepted: 08/22/2008] [Indexed: 12/28/2022] Open
Abstract
The genetic basis of the development and variation of adult form of vertebrates is not well understood. To address this problem, we performed a mutant screen to identify genes essential for the formation of adult skeletal structures of the zebrafish. Here, we describe the phenotypic and molecular characterization of a set of mutants showing loss of adult structures of the dermal skeleton, such as the rays of the fins and the scales, as well as the pharyngeal teeth. The mutations represent adult-viable, loss of function alleles in the ectodysplasin (eda) and ectodysplasin receptor (edar) genes. These genes are frequently mutated in the human hereditary disease hypohidrotic ectodermal dysplasia (HED; OMIM 224900, 305100) that affects the development of integumentary appendages such as hair and teeth. We find mutations in zebrafish edar that affect similar residues as mutated in human cases of HED and show similar phenotypic consequences. eda and edar are not required for early zebrafish development, but are rather specific for the development of adult skeletal and dental structures. We find that the defects of the fins and scales are due to the role of Eda signaling in organizing epidermal cells into discrete signaling centers of the scale epidermal placode and fin fold. Our genetic analysis demonstrates dose-sensitive and organ-specific response to alteration in levels of Eda signaling. In addition, we show substantial buffering of the effect of loss of edar function in different genetic backgrounds, suggesting canalization of this developmental system. We uncover a previously unknown role of Eda signaling in teleosts and show conservation of the developmental mechanisms involved in the formation and variation of both integumentary appendages and limbs. Lastly, our findings point to the utility of adult genetic screens in the zebrafish in identifying essential developmental processes involved in human disease and in morphological evolution.
Collapse
Affiliation(s)
- Matthew P Harris
- Max Planck Institute for Developmental Biology, Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|
43
|
Pispa J, Pummila M, Barker PA, Thesleff I, Mikkola ML. Edar and Troy signalling pathways act redundantly to regulate initiation of hair follicle development. Hum Mol Genet 2008; 17:3380-91. [DOI: 10.1093/hmg/ddn232] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
44
|
Abstract
Binding of myelin inhibitors to the NgR1/p75/LINGO-1 signaling complex activates RhoA to mediate the inhibition of axonal outgrowth. The nerve growth factor receptor p75, a TNF family receptor, is absent or poorly expressed in certain types of neurons that respond to myelin inhibitors, thereby prompting speculation that other TNF family receptors are involved in the NgR1 complex. Troy/Taj is an orphan TNF family receptor that is broadly expressed in postnatal and adult neurons. Troy binds to NgR1 and can functionally replace p75 in the p75/NgR1/LINGO-1 complex to activate RhoA and block neurite outgrowth in the presence of myelin inhibitors. Neurons from Troy-deficient mice are more resistant to the suppressive action of the myelin inhibitors. The discovery of TROY function in axon growth is an important step for understanding the complex regulation of axonal regeneration by diverse members of the TNF receptor family.
Collapse
Affiliation(s)
- Sha Mi
- Biogen Idec Inc., 14 Cambridge Center, Cambridge, MA 02142, United States.
| |
Collapse
|
45
|
|
46
|
Fliniaux I, Mikkola ML, Lefebvre S, Thesleff I. Identification of dkk4 as a target of Eda-A1/Edar pathway reveals an unexpected role of ectodysplasin as inhibitor of Wnt signalling in ectodermal placodes. Dev Biol 2008; 320:60-71. [PMID: 18508042 DOI: 10.1016/j.ydbio.2008.04.023] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 03/26/2008] [Accepted: 04/14/2008] [Indexed: 12/23/2022]
Abstract
The development of epithelial appendages, including hairs, glands and teeth starts from ectodermal placodes, and is regulated by interplay of stimulatory and inhibitory signals. Ectodysplasin-A1 (Eda-A1) and Wnts are high in hierarchy of placode activators. To identify direct targets of ectodysplasin pathway, we performed microarray profiling of genes differentially regulated by short exposure to recombinant Eda-A1 in embryonic eda(-/-) skin explants. Surprisingly, there were only two genes with obvious involvement in Wnt pathway: dkk4 (most highly induced gene in the screen), and lrp4. Both genes colocalized with Eda-A1 receptor Edar in placodes of ectodermal organs. They were upregulated upon Edar activation while several other Wnt associated genes previously suggested as Edar targets were unaffected. However, low dkk4 and lrp4 expression was retained in the absence of NF-kappaB signalling in eda(-/-) hair placodes. We provide evidence that this expression was dependent on Wnt activity present prior to Eda-A1/Edar signalling. Dkk4 was recently suggested as a key Wnt antagonist regulating lateral inhibition essential for correct patterning of hair follicles. Several pieces of evidence suggest Lrp4 as a Wnt inhibitor, as well. The finding that Eda-A1 induces placode inhibitors was unexpected, and underlines the importance of delicate fine-tuning of signalling during placode formation.
Collapse
Affiliation(s)
- Ingrid Fliniaux
- Institute of Biotechnology, Developmental Biology Program, University of Helsinki, 00014, Helsinki, Finland
| | | | | | | |
Collapse
|
47
|
Pantalacci S, Chaumot A, Benoît G, Sadier A, Delsuc F, Douzery EJP, Laudet V. Conserved features and evolutionary shifts of the EDA signaling pathway involved in vertebrate skin appendage development. Mol Biol Evol 2008; 25:912-28. [PMID: 18304980 DOI: 10.1093/molbev/msn038] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
It is widely accepted that evolutionary changes in conserved developmental signaling pathways play an important role in morphological evolution. However, few in silico studies were interested in tracking such changes in a signaling pathway. The Ectodysplasin (EDA) pathway provides an opportunity to fill this gap because it is involved in vertebrate skin appendage development such as scales, teeth, hair, and feathers that take an obvious part in the adaptation of species to their environment. We benefited from the large amount of genomic data now available to explore the evolution of the upstream genes of the EDA pathway. In mammals, these genes are eda (encoding 2 ligands, EDA-A1 and EDA-A2), edar (EDA-A1 receptor), edaradd (EDA receptor [EDAR] adapter), xedar (EDA-A2 receptor), and troy (a XEDAR-related receptor). We show that the evolution of EDA pathway genes combines both strongly conserved features and evolutionary shifts. These shifts are found at different signaling levels (from the ligand to intracellular signaling) and at different taxonomic levels (class, suborder, and genera). Although conserved features likely participate to the similarities found in the early development of vertebrate skin appendages, these shifts might account for innovations and specializations. Moreover, our study demonstrates that we can now benefit from the large number of sequenced vertebrate genomes to explore the evolution of specific signaling pathways and thereby to open new perspectives for developmental biology and evolutionary developmental biology.
Collapse
Affiliation(s)
- Sophie Pantalacci
- Molecular Zoology Team, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Institut Fédératif Biosciences 128 Gerland Lyon Sud, CNRS, INRA, Université Claude Bernard Lyon 1, 69364 Lyon cedex 07, France
| | | | | | | | | | | | | |
Collapse
|
48
|
Charles C, Pantalacci S, Peterkova R, Peterka M, Laudet V, Viriot L. Disruption of the palatal rugae pattern in Tabby (eda) mutant mice. Eur J Oral Sci 2007; 115:441-8. [DOI: 10.1111/j.1600-0722.2007.00482.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
49
|
Zhang YD, Chen Z, Song YQ, Liu C, Chen YP. Making a tooth: growth factors, transcription factors, and stem cells. Cell Res 2007; 15:301-16. [PMID: 15916718 DOI: 10.1038/sj.cr.7290299] [Citation(s) in RCA: 216] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Mammalian tooth development is largely dependent on sequential and reciprocal epithelial-mesenchymal interactions. These processes involve a series of inductive and permissive interactions that result in the determination, differentiation, and organization of odontogenic tissues. Multiple signaling molecules, including BMPs, FGFs, Shh, and Wnt proteins, have been implicated in mediating these tissue interactions. Transcription factors participate in epithelial-mesenchymal interactions via linking the signaling loops between tissue layers by responding to inductive signals and regulating the expression of other signaling molecules. Adult stem cells are highly plastic and multipotent. These cells including dental pulp stem cells and bone marrow stromal cells could be reprogrammed into odontogenic fate and participated in tooth formation. Recent progress in the studies of molecular basis of tooth development, adult stem cell biology, and regeneration will provide fundamental knowledge for the realization of human tooth regeneration in the near future.
Collapse
|
50
|
Knecht AK, Hosemann KE, Kingsley DM. Constraints on utilization of the EDA-signaling pathway in threespine stickleback evolution. Evol Dev 2007; 9:141-54. [PMID: 17371397 DOI: 10.1111/j.1525-142x.2007.00145.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Many traits evolve in parallel in widely separated populations. The evolutionary radiation of threespine sticklebacks provides a powerful model for testing the molecular basis of parallel evolution in vertebrates. Although marine sticklebacks are completely covered with bony armor plates, most freshwater populations have dramatic reductions in plates. Recent genetic studies have shown that major changes in armor patterning are likely due to regulatory alterations in the gene encoding the secreted signaling molecule ectodysplasin (EDA). In mammals, mutations in many different components of the EDA-signaling pathway produce similar changes in hair, teeth, sweat glands, and dermal bones. To test whether other genes in the EDA pathway also control natural variation in armor plates, we identified and mapped stickleback EDA Receptor (EDAR), the EDAR-Associated Death Domain adaptor, Tumor Necrosis Factor Receptor (TNFR) SuperFamily member 19, its adaptor TNFR-Associated Factor 6, and the downstream regulator nuclear factor kappa B Essential Modulator (NEMO). In contrast to the diversity of genes underlying ectodermal dysplasia disease phenotypes in humans, none of these EDA pathway components map to chromosomes previously shown to modify armor plates in natural populations, though EDAR showed a small but significant effect on plate number. We further investigated whether these genes exhibit differences in copy number, target size, or genomic organization that might make them less suitable targets for evolutionary change. In comparison with EDA, all these genes have smaller surrounding noncoding (putative regulatory) regions, with fewer evolutionarily conserved regions. We suggest that the presence of highly modular cis-acting control sequences may be a key factor influencing the likelihood that particular genes will serve as the basis of major phenotypic changes in nature.
Collapse
Affiliation(s)
- Anne K Knecht
- Department of Developmental Biology and, Stanford University School of Medicine, Stanford, CA 94305-5329, USA
| | | | | |
Collapse
|