1
|
Schiavinato A, Marcous F, Zuk AV, Keene DR, Tufa SF, Mosquera LM, Zigrino P, Mauch C, Eckes B, Francois K, De Backer J, Hunzelmann N, Moinzadeh P, Krieg T, Callewaert B, Sengle G. New insights into the structural role of EMILINs within the human skin microenvironment. Sci Rep 2024; 14:30345. [PMID: 39639116 PMCID: PMC11621341 DOI: 10.1038/s41598-024-81509-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Supramolecular extracellular matrix (ECM) networks play an essential role in skin architecture and function. Elastin microfibril interface-located proteins (EMILINs) comprise a family of three extracellular glycoproteins that serve as essential structural components of the elastin/fibrillin microfibril network, and exert crucial functions in cellular signaling. Little is known about the structural nature of EMILIN networks in skin. We therefore investigated the spatiotemporal localization of EMILIN-1, -2, -3 in human skin induced by aging, UV-exposure, fibrosis, and connective tissue disorder. Confocal immunofluorescence and immunogold electron microscopy analysis identified all EMILINs as components of elastic fibers and elastin-free oxytalan fibers inserted into the basement membrane (BM). Further, our ultrastructural analysis demonstrates cellular contacts of dermally localized EMILIN-1 positive fibers across the BM with the surface of basal keratinocytes. Analysis of skin biopsies and fibroblast cultures from fibrillin-1 deficient Marfan patients revealed that EMILINs require intact fibrillin-1 as deposition scaffold. In patients with scleroderma and the bleomycin-induced murine fibrosis model EMILIN-2 was upregulated. EMILIN-3 localizes to the tips of candelabra-like oxytalan fibers, and to specialized BMs engulfing hair follicles and sebaceous glands. Our data identify EMILINs as important markers to monitor rearrangements of the dermal ECM architecture induced by aging and pathological conditions.
Collapse
Affiliation(s)
- Alvise Schiavinato
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 52, 50931, Cologne, Germany
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Fady Marcous
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 52, 50931, Cologne, Germany
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Alexandra V Zuk
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Douglas R Keene
- Micro-Imaging Center, Shriners Children's, Portland, OR, 97239, USA
| | - Sara F Tufa
- Micro-Imaging Center, Shriners Children's, Portland, OR, 97239, USA
| | - Laura M Mosquera
- Center for Medical Genetics, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Pediatrics, Division of Pediatric Cardiology, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, 9000, Ghent, Belgium
| | - Paola Zigrino
- Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Cornelia Mauch
- Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Beate Eckes
- Translational Matrix Biology, Faculty of Medicine, University Hospital Cologne, 50931, Cologne, Germany
| | - Katrien Francois
- Department of Cardiovascular Surgery, Ghent University Hospital, 9000, Ghent, Belgium
| | - Julie De Backer
- Center for Medical Genetics, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Cardiology, Ghent University Hospital, 9000, Ghent, Belgium
| | - Nicolas Hunzelmann
- Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Pia Moinzadeh
- Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Thomas Krieg
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
- Translational Matrix Biology, Faculty of Medicine, University Hospital Cologne, 50931, Cologne, Germany
- Cologne Excellence Cluster On Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000, Ghent, Belgium
| | - Gerhard Sengle
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 52, 50931, Cologne, Germany.
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.
- Cologne Center for Musculoskeletal Biomechanics (CCMB), 50931, Cologne, Germany.
| |
Collapse
|
2
|
Truong BT, Shull LC, Zepeda BJ, Lencer E, Artinger KB. Human split hand/foot variants are not as functional as wildtype human PRDM1 in the rescue of craniofacial defects. Birth Defects Res 2024; 116:e2327. [PMID: 38456586 PMCID: PMC10949536 DOI: 10.1002/bdr2.2327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 01/24/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Split hand/foot malformation (SHFM) is a congenital limb disorder presenting with limb anomalies, such as missing, hypoplastic, or fused digits, and often craniofacial defects, including a cleft lip/palate, microdontia, micrognathia, or maxillary hypoplasia. We previously identified three novel variants in the transcription factor, PRDM1, that are associated with SHFM phenotypes. One individual also presented with a high arch palate. Studies in vertebrates indicate that PRDM1 is important for development of the skull; however, prior to our study, human variants in PRDM1 had not been associated with craniofacial anomalies. METHODS Using transient mRNA overexpression assays in prdm1a-/- mutant zebrafish, we tested whether the PRDM1 SHFM variants were functional and could lead to a rescue of the craniofacial defects observed in prdm1a-/- mutants. We also mined previously published CUT&RUN and RNA-seq datasets that sorted EGFP-positive cells from a Tg(Mmu:Prx1-EGFP) transgenic line that labels the pectoral fin, pharyngeal arches, and dorsal part of the head to examine Prdm1a binding and the effect of Prdm1a loss on craniofacial genes. RESULTS The prdm1a-/- mutants exhibit craniofacial defects including a hypoplastic neurocranium, a loss of posterior ceratobranchial arches, a shorter palatoquadrate, and an inverted ceratohyal. Injection of wildtype (WT) hPRDM1 in prdm1a-/- mutants partially rescues the palatoquadrate phenotype. However, injection of each of the three SHFM variants fails to rescue this skeletal defect. Loss of prdm1a leads to a decreased expression of important craniofacial genes by RNA-seq, including emilin3a, confirmed by hybridization chain reaction expression. Other genes including dlx5a/dlx6a, hand2, sox9b, col2a1a, and hoxb genes are also reduced. Validation by real-time quantitative PCR in the anterior half of zebrafish embryos failed to confirm the expression changes suggesting that the differences are enriched in prx1 expressing cells. CONCLUSION These data suggest that the three SHFM variants are likely not functional and may be associated with the craniofacial defects observed in the humans. Finally, they demonstrate how Prdm1a can directly bind and regulate genes involved in craniofacial development.
Collapse
Affiliation(s)
- Brittany T Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Craniofacial Development, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lomeli C Shull
- Department of Craniofacial Development, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
| | - Bryan J Zepeda
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, USA
| | - Ezra Lencer
- Biology Department, Lafayette College, Easton, Pennsylvania, USA
| | - Kristin B Artinger
- Department of Craniofacial Development, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, Minnesota, USA
| |
Collapse
|
3
|
Williams RJ, Laagland LT, Bach FC, Ward L, Chan W, Tam V, Medzikovic A, Basatvat S, Paillat L, Vedrenne N, Snuggs JW, Poramba-Liyanage DW, Hoyland JA, Chan D, Camus A, Richardson SM, Tryfonidou MA, Le Maitre CL. Recommendations for intervertebral disc notochordal cell investigation: From isolation to characterization. JOR Spine 2023; 6:e1272. [PMID: 37780826 PMCID: PMC10540834 DOI: 10.1002/jsp2.1272] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 10/03/2023] Open
Abstract
Background Lineage-tracing experiments have established that the central region of the mature intervertebral disc, the nucleus pulposus (NP), develops from the embryonic structure called "the notochord". However, changes in the cells derived from the notochord which form the NP (i.e., notochordal cells [NCs]), in terms of their phenotype and functional identity from early developmental stages to skeletal maturation are less understood. These key issues require further investigation to better comprehend the role of NCs in homeostasis and degeneration as well as their potential for regeneration. Progress in utilizing NCs is currently hampered due to poor consistency and lack of consensus methodology for in vitro NC extraction, manipulation, and characterization. Methods Here, an international group has come together to provide key recommendations and methodologies for NC isolation within key species, numeration, in vitro manipulation and culture, and characterization. Results Recommeded protocols are provided for isolation and culture of NCs. Experimental testing provided recommended methodology for numeration of NCs. The issues of cryopreservation are demonstrated, and a pannel of immunohistochemical markers are provided to inform NC characterization. Conclusions Together we hope this article provides a road map for in vitro studies of NCs to support advances in research into NC physiology and their potential in regenerative therapies.
Collapse
Affiliation(s)
- Rebecca J Williams
- Department of Oncology and Metabolism Medical School, The University of Sheffield Sheffield UK
- Biomolecular Sciences Research Centre Sheffield Hallam University Sheffield UK
| | - Lisanne T Laagland
- Department of Clinical Sciences Faculty of Veterinary Medicine, Utrecht University Utrecht The Netherlands
| | - Frances C Bach
- Department of Clinical Sciences Faculty of Veterinary Medicine, Utrecht University Utrecht The Netherlands
| | - Lizzy Ward
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health The University of Manchester Manchester UK
| | - Wilson Chan
- School of Biomedical Sciences The University of Hong Kong Pokfulam Hong Kong China
| | - Vivian Tam
- School of Biomedical Sciences The University of Hong Kong Pokfulam Hong Kong China
| | - Adel Medzikovic
- Department of Clinical Sciences Faculty of Veterinary Medicine, Utrecht University Utrecht The Netherlands
| | - Shaghayegh Basatvat
- Department of Oncology and Metabolism Medical School, The University of Sheffield Sheffield UK
- Biomolecular Sciences Research Centre Sheffield Hallam University Sheffield UK
| | - Lily Paillat
- Regenerative Medicine and Skeleton, RMeS Nantes Université, Oniris, CHU Nantes, INSERM, UMR 1229 Nantes France
| | - Nicolas Vedrenne
- Regenerative Medicine and Skeleton, RMeS Nantes Université, Oniris, CHU Nantes, INSERM, UMR 1229 Nantes France
| | - Joseph W Snuggs
- Department of Oncology and Metabolism Medical School, The University of Sheffield Sheffield UK
- Biomolecular Sciences Research Centre Sheffield Hallam University Sheffield UK
| | - Deepani W Poramba-Liyanage
- Department of Clinical Sciences Faculty of Veterinary Medicine, Utrecht University Utrecht The Netherlands
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health The University of Manchester Manchester UK
- NIHR Manchester Biomedical Research Centre Central Manchester Foundation Trust, Manchester Academic Health Science Centre Manchester UK
| | - Danny Chan
- School of Biomedical Sciences The University of Hong Kong Pokfulam Hong Kong China
| | - Anne Camus
- Regenerative Medicine and Skeleton, RMeS Nantes Université, Oniris, CHU Nantes, INSERM, UMR 1229 Nantes France
| | - Stephen M Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health The University of Manchester Manchester UK
| | - Marianna A Tryfonidou
- Department of Clinical Sciences Faculty of Veterinary Medicine, Utrecht University Utrecht The Netherlands
| | - Christine L Le Maitre
- Department of Oncology and Metabolism Medical School, The University of Sheffield Sheffield UK
- Biomolecular Sciences Research Centre Sheffield Hallam University Sheffield UK
| |
Collapse
|
4
|
Lin YC, Sahoo BK, Gau SS, Yang RB. The biology of SCUBE. J Biomed Sci 2023; 30:33. [PMID: 37237303 PMCID: PMC10214685 DOI: 10.1186/s12929-023-00925-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
The SCUBE [Signal peptide-Complement C1r/C1s, Uegf, Bmp1 (CUB)-Epithelial growth factor domain-containing protein] family consists of three proteins in vertebrates, SCUBE1, 2 and 3, which are highly conserved in zebrafish, mice and humans. Each SCUBE gene encodes a polypeptide of approximately 1000 amino acids that is organized into five modular domains: (1) an N-terminal signal peptide sequence, (2) nine tandem epidermal growth factor (EGF)-like repeats, (3) a large spacer region, (4) three cysteine-rich (CR) motifs, and (5) a CUB domain at the C-terminus. Murine Scube genes are expressed individually or in combination during the development of various tissues, including those in the central nervous system and the axial skeleton. The cDNAs of human SCUBE orthologs were originally cloned from vascular endothelial cells, but SCUBE expression has also been found in platelets, mammary ductal epithelium and osteoblasts. Both soluble and membrane-associated SCUBEs have been shown to play important roles in physiology and pathology. For instance, upregulation of SCUBEs has been reported in acute myeloid leukemia, breast cancer and lung cancer. In addition, soluble SCUBE1 is released from activated platelets and can be used as a clinical biomarker for acute coronary syndrome and ischemic stroke. Soluble SCUBE2 enhances distal signaling by facilitating the secretion of dual-lipidated hedgehog from nearby ligand-producing cells in a paracrine manner. Interestingly, the spacer regions and CR motifs can increase or enable SCUBE binding to cell surfaces via electrostatic or glycan-lectin interactions. As such, membrane-associated SCUBEs can function as coreceptors that enhance the signaling activity of various serine/threonine kinase or tyrosine kinase receptors. For example, membrane-associated SCUBE3 functions as a coreceptor that promotes signaling in bone morphogenesis. In humans, SCUBE3 mutations are linked to abnormalities in growth and differentiation of both bones and teeth. In addition to studies on human SCUBE function, experimental results from genetically modified mouse models have yielded important insights in the field of systems biology. In this review, we highlight novel molecular discoveries and critical directions for future research on SCUBE proteins in the context of cancer, skeletal disease and cardiovascular disease.
Collapse
Affiliation(s)
- Yuh-Charn Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Binay K Sahoo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shiang-Shin Gau
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan.
- Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
5
|
Tesoriero C, Greco F, Cannone E, Ghirotto F, Facchinello N, Schiavone M, Vettori A. Modeling Human Muscular Dystrophies in Zebrafish: Mutant Lines, Transgenic Fluorescent Biosensors, and Phenotyping Assays. Int J Mol Sci 2023; 24:8314. [PMID: 37176020 PMCID: PMC10179009 DOI: 10.3390/ijms24098314] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of myopathies characterized by progressive muscle weakness leading to death from heart or respiratory failure. MDs are caused by mutations in genes involved in both the development and organization of muscle fibers. Several animal models harboring mutations in MD-associated genes have been developed so far. Together with rodents, the zebrafish is one of the most popular animal models used to reproduce MDs because of the high level of sequence homology with the human genome and its genetic manipulability. This review describes the most important zebrafish mutant models of MD and the most advanced tools used to generate and characterize all these valuable transgenic lines. Zebrafish models of MDs have been generated by introducing mutations to muscle-specific genes with different genetic techniques, such as (i) N-ethyl-N-nitrosourea (ENU) treatment, (ii) the injection of specific morpholino, (iii) tol2-based transgenesis, (iv) TALEN, (v) and CRISPR/Cas9 technology. All these models are extensively used either to study muscle development and function or understand the pathogenetic mechanisms of MDs. Several tools have also been developed to characterize these zebrafish models by checking (i) motor behavior, (ii) muscle fiber structure, (iii) oxidative stress, and (iv) mitochondrial function and dynamics. Further, living biosensor models, based on the expression of fluorescent reporter proteins under the control of muscle-specific promoters or responsive elements, have been revealed to be powerful tools to follow molecular dynamics at the level of a single muscle fiber. Thus, zebrafish models of MDs can also be a powerful tool to search for new drugs or gene therapies able to block or slow down disease progression.
Collapse
Affiliation(s)
- Chiara Tesoriero
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| | - Francesca Greco
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| | - Elena Cannone
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Francesco Ghirotto
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| | - Nicola Facchinello
- Neuroscience Institute, Italian National Research Council (CNR), 35131 Padua, Italy
| | - Marco Schiavone
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Andrea Vettori
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (C.T.); (F.G.); (F.G.); (A.V.)
| |
Collapse
|
6
|
Deng P, Huang J, Zhang Q, Li Y, Li J. The role of EMILIN-1 in the osteo/odontogenic differentiation of dental pulp stem cells. BMC Oral Health 2023; 23:203. [PMID: 37024847 PMCID: PMC10077624 DOI: 10.1186/s12903-023-02905-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Human dental pulp stem cells (hDPSCs) may be the best choice for self-repair and regeneration of teeth and maxillofacial bone tissue due to their homogeneous tissue origin, high proliferation and differentiation rates, and no obvious ethical restrictions. Recently, several studies have shown that extracellular matrix (ECM) proteins can effectively regulate the proliferation and differentiation fate of mesenchymal stem cells (MSCs). However, the role of elastin microfibril interface-located protein-1 (EMILIN-1), a new ECM glycoprotein, in osteo/odontogenic differentiation of hDPSCs has not been reported. The aim of this study was to explore the effect of EMILIN-1 during osteo/odontogenic differentiation of hDPSCs. METHODS hDPSCs were cultured in osteo/odontogenic induction medium. qPCR and Western blot analysis were performed to detect osteo/odonto-specific genes/proteins expression as well as the expression of EMILIN-1. After knockdown of Emilin-1 in hDPSCs with small interfering RNA and exogenous addition of recombinant human EMILIN-1 protein (rhEMILIN-1), Cell Counting Kit-8 assay, alkaline phosphatase staining, alizarin red S staining, qPCR and Western blot were performed to examine the effect of EMILIN-1 on proliferation and osteo/odontogenic differentiation of hDPSCs. RESULTS During the osteo/odontogenic induction of hDPSCs, the expression of osteo/odonto-specific genes/proteins increased, as did EMILIN-1 protein levels. More notably, knockdown of Emilin-1 decreased hDPSCs proliferation and osteo/odontogenic differentiation, whereas exogenous addition of rhEMILIN-1 increased them. CONCLUSIONS These findings suggested that EMILIN-1 is essential for the osteo/odontogenic differentiation of hDPSCs, which may provide new insights for teeth and bone tissue regeneration.
Collapse
Affiliation(s)
- Pingmeng Deng
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, People's Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Jing Huang
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, People's Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Qixuan Zhang
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, People's Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Yuejia Li
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, People's Republic of China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, People's Republic of China.
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, People's Republic of China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China.
| |
Collapse
|
7
|
Truong BT, Shull LC, Lencer E, Bend EG, Field M, Blue EE, Bamshad MJ, Skinner C, Everman D, Schwartz CE, Flanagan-Steet H, Artinger KB. PRDM1 DNA-binding zinc finger domain is required for normal limb development and is disrupted in split hand/foot malformation. Dis Model Mech 2023; 16:dmm049977. [PMID: 37083955 PMCID: PMC10151829 DOI: 10.1242/dmm.049977] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/09/2023] [Indexed: 04/22/2023] Open
Abstract
Split hand/foot malformation (SHFM) is a rare limb abnormality with clefting of the fingers and/or toes. For many individuals, the genetic etiology is unknown. Through whole-exome and targeted sequencing, we detected three novel variants in a gene encoding a transcription factor, PRDM1, that arose de novo in families with SHFM or segregated with the phenotype. PRDM1 is required for limb development; however, its role is not well understood and it is unclear how the PRDM1 variants affect protein function. Using transient and stable overexpression rescue experiments in zebrafish, we show that the variants disrupt the proline/serine-rich and DNA-binding zinc finger domains, resulting in a dominant-negative effect. Through gene expression assays, RNA sequencing, and CUT&RUN in isolated pectoral fin cells, we demonstrate that Prdm1a directly binds to and regulates genes required for fin induction, outgrowth and anterior/posterior patterning, such as fgfr1a, dlx5a, dlx6a and smo. Taken together, these results improve our understanding of the role of PRDM1 in the limb gene regulatory network and identified novel PRDM1 variants that link to SHFM in humans.
Collapse
Affiliation(s)
- Brittany T. Truong
- Human Medical Genetics & Genomics Graduate Program, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lomeli C. Shull
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ezra Lencer
- Biology Department, Lafayette College, Easton, PA 18042, USA
| | - Eric G. Bend
- Greenwood Genetics Center, Greenwood, SC 29646, USA
| | - Michael Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW 2298, AUS
| | - Elizabeth E. Blue
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
| | - Michael J. Bamshad
- Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | - Kristin B. Artinger
- Department of Craniofacial Biology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
8
|
Risato G, Celeghin R, Brañas Casas R, Dinarello A, Zuppardo A, Vettori A, Pilichou K, Thiene G, Basso C, Argenton F, Visentin S, Cosmi E, Tiso N, Beffagna G. Hyperactivation of Wnt/β-catenin and Jak/Stat3 pathways in human and zebrafish foetal growth restriction models: Implications for pharmacological rescue. Front Cell Dev Biol 2022; 10:943127. [PMID: 36051436 PMCID: PMC9424487 DOI: 10.3389/fcell.2022.943127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Foetal Growth Restriction (FGR), previously known as Intrauterine Growth Restriction (IUGR), is an obstetrical condition due to placental insufficiency, affecting yearly about 30 million newborns worldwide. In this work, we aimed to identify and pharmacologically target signalling pathways specifically involved in the FGR condition, focusing on FGR-related cardiovascular phenotypes. The transcriptional profile of human umbilical cords from FGR and control cases was compared with the response to hypoxia of zebrafish (Danio rerio) transgenic lines reporting in vivo the activity of twelve signalling pathways involved in embryonic development. Wnt/β-catenin and Jak/Stat3 were found as key pathways significantly dysregulated in both human and zebrafish samples. This information was used in a chemical-genetic analysis to test drugs targeting Wnt/β-catenin and Jak/Stat3 pathways to rescue a set of FGR phenotypes, including growth restriction and cardiovascular modifications. Treatments with the Wnt/β-catenin agonist SB216763 successfully rescued body dimensions, cardiac shape, and vessel organization in zebrafish FGR models. Our data support the Wnt/β-catenin pathway as a key FGR marker and a promising target for pharmacological intervention in the FGR condition.
Collapse
Affiliation(s)
- Giovanni Risato
- Department of Biology, University of Padova, Padova, Italy
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Rudy Celeghin
- Department of Biology, University of Padova, Padova, Italy
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | | | | | | | - Andrea Vettori
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | | | - Silvia Visentin
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Erich Cosmi
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova, Italy
| | - Giorgia Beffagna
- Department of Biology, University of Padova, Padova, Italy
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
9
|
Serifi I, Besta S, Karetsou Z, Giardoglou P, Beis D, Niewiadomski P, Papamarcaki T. Targeting of SET/I2PP2A oncoprotein inhibits Gli1 transcription revealing a new modulator of Hedgehog signaling. Sci Rep 2021; 11:13940. [PMID: 34230583 PMCID: PMC8260731 DOI: 10.1038/s41598-021-93440-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 06/18/2021] [Indexed: 01/17/2023] Open
Abstract
The Hedgehog (Hh)/Gli signaling pathway controls cell proliferation and differentiation, is critical for the development of nearly every tissue and organ in vertebrates and is also involved in tumorigenesis. In this study, we characterize the oncoprotein SET/I2PP2A as a novel regulator of Hh signaling. Our previous work has shown that the zebrafish homologs of SET are expressed during early development and localized in the ciliated organs. In the present work, we show that CRISPR/Cas9-mediated knockdown of setb gene in zebrafish embryos resulted in cyclopia, a characteristic patterning defect previously reported in Hh mutants. Consistent with these findings, targeting setb gene using CRISPR/Cas9 or a setb morpholino, reduced Gli1-dependent mCherry expression in the Hedgehog reporter zebrafish line Tg(12xGliBS:mCherry-NLS). Likewise, SET loss of function by means of pharmacological inhibition and gene knockdown prevented the increase of Gli1 expression in mammalian cells in vitro. Conversely, overexpression of SET resulted in an increase of the expression of a Gli-dependent luciferase reporter, an effect likely attributable to the relief of the Sufu-mediated inhibition of Gli1. Collectively, our data support the involvement of SET in Gli1-mediated transcription and suggest the oncoprotein SET/I2PP2A as a new modulator of Hedgehog signaling.
Collapse
Affiliation(s)
- Iliana Serifi
- Laboratory of Biological Chemistry, Medical Department, School of Health Sciences, University of Ioannina, 451 10, Ioannina, Greece.,Department of Biomedical Research, Foundation for Research and Technology-Hellas, Institute of Molecular Biology and Biotechnology, 451 10, Ioannina, Greece
| | - Simoni Besta
- Laboratory of Biological Chemistry, Medical Department, School of Health Sciences, University of Ioannina, 451 10, Ioannina, Greece
| | - Zoe Karetsou
- Laboratory of Biological Chemistry, Medical Department, School of Health Sciences, University of Ioannina, 451 10, Ioannina, Greece
| | - Panagiota Giardoglou
- Developmental Biology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 115 27, Athens, Greece
| | - Dimitris Beis
- Developmental Biology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 115 27, Athens, Greece
| | | | - Thomais Papamarcaki
- Laboratory of Biological Chemistry, Medical Department, School of Health Sciences, University of Ioannina, 451 10, Ioannina, Greece. .,Department of Biomedical Research, Foundation for Research and Technology-Hellas, Institute of Molecular Biology and Biotechnology, 451 10, Ioannina, Greece.
| |
Collapse
|
10
|
Itoh K, Ossipova O, Sokol SY. Pinhead antagonizes Admp to promote notochord formation. iScience 2021; 24:102520. [PMID: 34142034 PMCID: PMC8188501 DOI: 10.1016/j.isci.2021.102520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/18/2021] [Accepted: 05/05/2021] [Indexed: 12/05/2022] Open
Abstract
Dorsoventral patterning of a vertebrate embryo critically depends on the activity of Smad1 that mediates signaling by BMP proteins, anti-dorsalizing morphogenetic protein (Admp), and their antagonists. Pinhead (Pnhd), a cystine-knot-containing secreted protein, is expressed in the ventrolateral mesoderm during Xenopus gastrulation; however, its molecular targets and signaling mechanisms have not been fully elucidated. Our mass spectrometry-based screen of the gastrula secretome identified Admp as Pnhd-associated protein. We show that Pnhd binds Admp and inhibits its ventralizing activity by reducing Smad1 phosphorylation and its transcriptional targets. Importantly, Pnhd depletion further increased phospho-Smad1 levels in the presence of Admp. Furthermore, Pnhd synergized with Chordin and a truncated BMP4 receptor in the induction of notochord markers in ectoderm cells, and Pnhd-depleted embryos displayed notochord defects. Our findings suggest that Pnhd binds and inactivates Admp to promote notochord development. We propose that the interaction between Admp and Pnhd refines Smad1 activity gradients during vertebrate gastrulation.
Collapse
Affiliation(s)
- Keiji Itoh
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Olga Ossipova
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
11
|
Wang Z, Mizoguchi T, Kuribara T, Nakajima M, Iwata M, Sakamoto Y, Nakamura H, Murayama T, Nemoto T, Itoh M. Py 3-FITC: a new fluorescent probe for live cell imaging of collagen-rich tissues and ionocytes. Open Biol 2021; 11:200241. [PMID: 33561382 PMCID: PMC8061698 DOI: 10.1098/rsob.200241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 01/04/2021] [Indexed: 11/12/2022] Open
Abstract
Polypyrrole-based polyamides are used as sequence-specific DNA probes. However, their cellular uptake and distribution are affected by several factors and have not been extensively studied in vivo. Here, we generated a series of fluorescence-conjugated polypyrrole compounds and examined their cellular distribution using live zebrafish and cultured human cells. Among the evaluated compounds, Py3-FITC was able to visualize collagen-rich tissues, such as the jaw cartilage, opercle and bulbus arteriosus, in early-stage living zebrafish embryos. Then, we stained cultured human cells with Py3-FITC and found that the staining became more intense as the amount of collagen was increased. In addition, Py3-FITC-stained HR cells, which represent a type of ionocyte on the body surface of living zebrafish embryos. Py3-FITC has low toxicity, and collagen-rich tissues and ionocytes can be visualized when soaked in Py3-FITC solution. Therefore, Py3-FITC may be a useful live imaging tool for detecting changes in collagen-rich tissue and ionocytes, including their mammalian analogues, during both normal development and disease progression.
Collapse
Affiliation(s)
- Zhaotong Wang
- Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | | | | | - Masaya Nakajima
- Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | - Mayuu Iwata
- Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | - Yuka Sakamoto
- Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | | | | | - Tetsuhiro Nemoto
- Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | - Motoyuki Itoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| |
Collapse
|
12
|
Abstract
The vertebrate body plan is characterized by the presence of a segmented spine along its main axis. Here, we examine the current understanding of how the axial tissues that are formed during embryonic development give rise to the adult spine and summarize recent advances in the field, largely focused on recent studies in zebrafish, with comparisons to amniotes where appropriate. We discuss recent work illuminating the genetics and biological mechanisms mediating extension and straightening of the body axis during development, and highlight open questions. We specifically focus on the processes of notochord development and cerebrospinal fluid physiology, and how defects in those processes may lead to scoliosis.
Collapse
Affiliation(s)
- Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC, 27710, USA
| | - Ryan S Gray
- Department of Nutritional Sciences, University of Texas at Austin, Dell Pediatrics Research Institute, Austin, TX, 78723, USA
| |
Collapse
|
13
|
Adamo CS, Zuk AV, Sengle G. The fibrillin microfibril/elastic fibre network: A critical extracellular supramolecular scaffold to balance skin homoeostasis. Exp Dermatol 2020; 30:25-37. [PMID: 32920888 DOI: 10.1111/exd.14191] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 01/08/2023]
Abstract
Supramolecular networks composed of fibrillins (fibrillin-1 and fibrillin-2) and associated ligands form intricate cellular microenvironments which balance skin homoeostasis and direct remodelling. Fibrillins assemble into microfibrils which are not only indispensable for conferring elasticity to the skin, but also control the bioavailability of growth factors targeted to the extracellular matrix architecture. Fibrillin microfibrils (FMF) represent the core scaffolds for elastic fibre formation, and they also decorate the surface of elastic fibres and form independent networks. In normal dermis, elastic fibres are suspended in a three-dimensional basket-like lattice of FMF intersecting basement membranes at the dermal-epidermal junction and thus conferring pliability to the skin. The importance of FMF for skin homoeostasis is illustrated by the clinical features caused by mutations in the human fibrillin genes (FBN1, FBN2), summarized as "fibrillinopathies." In skin, fibrillin mutations result in phenotypes ranging from thick, stiff and fibrotic skin to thin, lax and hyperextensible skin. The most plausible explanation for this spectrum of phenotypic outcomes is that FMF regulate growth factor signalling essential for proper growth and homoeostasis of the skin. Here, we will give an overview about the current understanding of the underlying pathomechanisms leading to fibrillin-dependent fibrosis as well as forms of cutis laxa caused by mutational inactivation of FMF-associated ligands.
Collapse
Affiliation(s)
- Christin S Adamo
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alexandra V Zuk
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), Cologne, Germany
| |
Collapse
|
14
|
Autophagic flux inhibition enhances cytotoxicity of the receptor tyrosine kinase inhibitor ponatinib. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:195. [PMID: 32962733 PMCID: PMC7507635 DOI: 10.1186/s13046-020-01692-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022]
Abstract
Background Despite reported advances, acquired resistance to tyrosine kinase inhibitors still represents a serious problem in successful cancer treatment. Among this class of drugs, ponatinib (PON) has been shown to have notable long-term efficacy, although its cytotoxicity might be hampered by autophagy. In this study, we examined the likelihood of PON resistance evolution in neuroblastoma and assessed the extent to which autophagy might provide survival advantages to tumor cells. Methods The effects of PON in inducing autophagy were determined both in vitro, using SK-N-BE(2), SH-SY5Y, and IMR-32 human neuroblastoma cell lines, and in vivo, using zebrafish and mouse models. Single and combined treatments with chloroquine (CQ)—a blocking agent of lysosomal metabolism and autophagic flux—and PON were conducted, and the effects on cell viability were determined using metabolic and immunohistochemical assays. The activation of the autophagic flux was analyzed through immunoblot and protein arrays, immunofluorescence, and transmission electron microscopy. Combination therapy with PON and CQ was tested in a clinically relevant neuroblastoma mouse model. Results Our results confirm that, in neuroblastoma cells and wild-type zebrafish embryos, PON induces the accumulation of autophagy vesicles—a sign of autophagy activation. Inhibition of autophagic flux by CQ restores the cytotoxic potential of PON, thus attributing to autophagy a cytoprotective nature. In mice, the use of CQ as adjuvant therapy significantly improves the anti-tumor effects obtained by PON, leading to ulterior reduction of tumor masses. Conclusions Together, these findings support the importance of autophagy monitoring in the treatment protocols that foresee PON administration, as this may predict drug resistance acquisition. The findings also establish the potential for combined use of CQ and PON, paving the way for their consideration in upcoming treatment protocols against neuroblastoma.
Collapse
|
15
|
EMILIN proteins are novel extracellular constituents of the dentin-pulp complex. Sci Rep 2020; 10:15320. [PMID: 32948785 PMCID: PMC7501263 DOI: 10.1038/s41598-020-72123-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Odontoblasts and pulp stroma cells are embedded within supramolecular networks of extracellular matrix (ECM). Fibrillin microfibrils and associated proteins are crucial constituents of these networks, serving as contextual scaffolds to regulate tissue development and homeostasis by providing both structural and mechanical properties and sequestering growth factors of the TGF-β superfamily. EMILIN-1, -2, and -3 are microfibril-associated glycoproteins known to modulate cell behaviour, growth factor activity, and ECM assembly. So far their expression in the various cells of the dentin-pulp complex during development, in the adult stage, and during inflammation has not been investigated. Confocal immunofluorescence microscopy and western blot analysis of developing and adult mouse molars and incisors revealed an abundant presence of EMILINs in the entire dental papilla, at early developmental stages. Later in development the signal intensity for EMILIN-3 decreases, while EMILIN-1 and -2 staining appears to increase in the pre-dentin and in the ECM surrounding odontoblasts. Our data also demonstrate new specific interactions of EMILINs with fibulins in the dentin enamel junction. Interestingly, in dentin caries lesions the signal for EMILIN-3 was significantly increased in inflamed odontoblasts. Overall our findings point for the first time to a role of EMILINs in dentinogenesis, pulp biology, and inflammation.
Collapse
|
16
|
Dstyk mutation leads to congenital scoliosis-like vertebral malformations in zebrafish via dysregulated mTORC1/TFEB pathway. Nat Commun 2020; 11:479. [PMID: 31980602 PMCID: PMC6981171 DOI: 10.1038/s41467-019-14169-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Congenital scoliosis (CS) is a complex genetic disorder characterized by vertebral malformations. The precise etiology of CS is not fully defined. Here, we identify that mutation in dual serine/threonine and tyrosine protein kinase (dstyk) lead to CS-like vertebral malformations in zebrafish. We demonstrate that the scoliosis in dstyk mutants is related to the wavy and malformed notochord sheath formation and abnormal axial skeleton segmentation due to dysregulated biogenesis of notochord vacuoles and notochord function. Further studies show that DSTYK is located in late endosomal/lysosomal compartments and is involved in the lysosome biogenesis in mammalian cells. Dstyk knockdown inhibits notochord vacuole and lysosome biogenesis through mTORC1-dependent repression of TFEB nuclear translocation. Inhibition of mTORC1 activity can rescue the defect in notochord vacuole biogenesis and scoliosis in dstyk mutants. Together, our findings reveal a key role of DSTYK in notochord vacuole biogenesis, notochord morphogenesis and spine development through mTORC1/TFEB pathway. Congenital scoliosis is a complex genetic disorder characterized by vertebral malformation. Here, the authors demonstrate that loss of dstyk leads to scoliosis in zebrafish due to dysregulated biogenesis of notochord vacuoles and that DSTYK is required for lysosome biogenesis through mTORC1 regulation.
Collapse
|
17
|
Tonelli F, Bek JW, Besio R, De Clercq A, Leoni L, Salmon P, Coucke PJ, Willaert A, Forlino A. Zebrafish: A Resourceful Vertebrate Model to Investigate Skeletal Disorders. Front Endocrinol (Lausanne) 2020; 11:489. [PMID: 32849280 PMCID: PMC7416647 DOI: 10.3389/fendo.2020.00489] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
Animal models are essential tools for addressing fundamental scientific questions about skeletal diseases and for the development of new therapeutic approaches. Traditionally, mice have been the most common model organism in biomedical research, but their use is hampered by several limitations including complex generation, demanding investigation of early developmental stages, regulatory restrictions on breeding, and high maintenance cost. The zebrafish has been used as an efficient alternative vertebrate model for the study of human skeletal diseases, thanks to its easy genetic manipulation, high fecundity, external fertilization, transparency of rapidly developing embryos, and low maintenance cost. Furthermore, zebrafish share similar skeletal cells and ossification types with mammals. In the last decades, the use of both forward and new reverse genetics techniques has resulted in the generation of many mutant lines carrying skeletal phenotypes associated with human diseases. In addition, transgenic lines expressing fluorescent proteins under bone cell- or pathway- specific promoters enable in vivo imaging of differentiation and signaling at the cellular level. Despite the small size of the zebrafish, many traditional techniques for skeletal phenotyping, such as x-ray and microCT imaging and histological approaches, can be applied using the appropriate equipment and custom protocols. The ability of adult zebrafish to remodel skeletal tissues can be exploited as a unique tool to investigate bone formation and repair. Finally, the permeability of embryos to chemicals dissolved in water, together with the availability of large numbers of small-sized animals makes zebrafish a perfect model for high-throughput bone anabolic drug screening. This review aims to discuss the techniques that make zebrafish a powerful model to investigate the molecular and physiological basis of skeletal disorders.
Collapse
Affiliation(s)
- Francesca Tonelli
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Jan Willem Bek
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Roberta Besio
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Adelbert De Clercq
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Laura Leoni
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Paul J. Coucke
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Andy Willaert
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Antonella Forlino
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Antonella Forlino
| |
Collapse
|
18
|
Corallo D, Donadon M, Pantile M, Sidarovich V, Cocchi S, Ori M, De Sarlo M, Candiani S, Frasson C, Distel M, Quattrone A, Zanon C, Basso G, Tonini GP, Aveic S. LIN28B increases neural crest cell migration and leads to transformation of trunk sympathoadrenal precursors. Cell Death Differ 2019; 27:1225-1242. [PMID: 31601998 DOI: 10.1038/s41418-019-0425-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 09/04/2019] [Accepted: 09/12/2019] [Indexed: 01/25/2023] Open
Abstract
The RNA-binding protein LIN28B regulates developmental timing and determines stem cell identity by suppressing the let-7 family of microRNAs. Postembryonic reactivation of LIN28B impairs cell commitment to differentiation, prompting their transformation. In this study, we assessed the extent to which ectopic lin28b expression modulates the physiological behavior of neural crest cells (NCC) and governs their transformation in the trunk region of developing embryos. We provide evidence that the overexpression of lin28b inhibits sympathoadrenal cell differentiation and accelerates NCC migration in two vertebrate models, Xenopus leavis and Danio rerio. Our results highlight the relevance of ITGA5 and ITGA6 in the LIN28B-dependent regulation of the invasive motility of tumor cells. The results also establish that LIN28B overexpression supports neuroblastoma onset and the metastatic potential of malignant cells through let-7a-dependent and let-7a-independent mechanisms.
Collapse
Affiliation(s)
- Diana Corallo
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.
| | - Michael Donadon
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Marcella Pantile
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Viktoryia Sidarovich
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Simona Cocchi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Michela Ori
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Miriam De Sarlo
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Simona Candiani
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Genova, Italy
| | - Chiara Frasson
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Martin Distel
- Innovative Cancer Models, Children's Cancer Research Institute (CCRI), Wien, Austria
| | - Alessandro Quattrone
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Carlo Zanon
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Giuseppe Basso
- Department of Women and Child Health, Haematology-Oncology Clinic, University of Padua, Padova, Italy
| | - Gian Paolo Tonini
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Sanja Aveic
- Neuroblastoma Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy. .,Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
19
|
Wei L, Al Oustah A, Blader P, Roussigné M. Notch signaling restricts FGF pathway activation in parapineal cells to promote their collective migration. eLife 2019; 8:46275. [PMID: 31498774 PMCID: PMC6733574 DOI: 10.7554/elife.46275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 07/18/2019] [Indexed: 12/16/2022] Open
Abstract
Coordinated migration of cell collectives is important during embryonic development and relies on cells integrating multiple mechanical and chemical cues. Recently, we described that focal activation of the FGF pathway promotes the migration of the parapineal in the zebrafish epithalamus. How FGF activity is restricted to leading cells in this system is, however, unclear. Here, we address the role of Notch signaling in modulating FGF activity within the parapineal. While Notch loss-of-function results in an increased number of parapineal cells activating the FGF pathway, global activation of Notch signaling decreases it; both contexts result in defects in parapineal migration and specification. Decreasing or increasing FGF signaling in a Notch loss-of-function context respectively rescues or aggravates parapineal migration defects without affecting parapineal cells specification. We propose that Notch signaling controls the migration of the parapineal through its capacity to restrict FGF pathway activation to a few leading cells.
Collapse
Affiliation(s)
- Lu Wei
- Centre de Biologie Intégrative (CBI), Centre de Biologie du Développement (CBD), Université de Toulouse, CNRS (UMR 5547), Toulouse, France
| | - Amir Al Oustah
- Centre de Biologie Intégrative (CBI), Centre de Biologie du Développement (CBD), Université de Toulouse, CNRS (UMR 5547), Toulouse, France
| | - Patrick Blader
- Centre de Biologie Intégrative (CBI), Centre de Biologie du Développement (CBD), Université de Toulouse, CNRS (UMR 5547), Toulouse, France
| | - Myriam Roussigné
- Centre de Biologie Intégrative (CBI), Centre de Biologie du Développement (CBD), Université de Toulouse, CNRS (UMR 5547), Toulouse, France
| |
Collapse
|
20
|
Giuliodori A, Beffagna G, Marchetto G, Fornetto C, Vanzi F, Toppo S, Facchinello N, Santimaria M, Vettori A, Rizzo S, Della Barbera M, Pilichou K, Argenton F, Thiene G, Tiso N, Basso C. Loss of cardiac Wnt/β-catenin signalling in desmoplakin-deficient AC8 zebrafish models is rescuable by genetic and pharmacological intervention. Cardiovasc Res 2019. [PMID: 29522173 DOI: 10.1093/cvr/cvy057] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aims Arrhythmogenic cardiomyopathy (AC) is an inherited heart disease characterized by life-threatening ventricular arrhythmias and fibro-fatty replacement of the myocardium. More than 60% of AC patients show pathogenic mutations in genes encoding for desmosomal proteins. By focusing our attention on the AC8 form, linked to the junctional protein desmoplakin (DSP), we present here a zebrafish model of DSP deficiency, exploited to identify early changes of cell signalling in the cardiac region. Methods and results To obtain an embryonic model of Dsp deficiency, we first confirmed the orthologous correspondence of zebrafish Dsp genes (dspa and dspb) to the human DSP counterpart. Then, we verified their cardiac expression, at embryonic and adult stages, and subsequently we targeted them by antisense morpholino strategy, confirming specific and disruptive effects on desmosomes, like those identified in AC patients. Finally, we exploited our Dsp-deficient models for an in vivo cell signalling screen, using pathway-specific reporter transgenes. Out of nine considered, three pathways (Wnt/β-catenin, TGFβ/Smad3, and Hippo/YAP-TAZ) were significantly altered, with Wnt as the most dramatically affected. Interestingly, under persistent Dsp deficiency, Wnt signalling is rescuable both by a genetic and a pharmacological approach. Conclusion Our data point to Wnt/β-catenin as the final common pathway underlying different desmosomal AC forms and support the zebrafish as a suitable model for detecting early signalling pathways involved in the pathogenesis of DSP-associated diseases, possibly responsive to pharmacological or genetic rescue.
Collapse
Affiliation(s)
- Alice Giuliodori
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Giorgia Beffagna
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Giulia Marchetto
- European Laboratory for Non-Linear Spectroscopy, via N. Carrara, 1, Sesto Fiorentino (FI) 50019, Italy
| | - Chiara Fornetto
- European Laboratory for Non-Linear Spectroscopy, via N. Carrara, 1, Sesto Fiorentino (FI) 50019, Italy
| | - Francesco Vanzi
- European Laboratory for Non-Linear Spectroscopy, via N. Carrara, 1, Sesto Fiorentino (FI) 50019, Italy.,Department of Biology, University of Florence, via Madonna del Piano, 6, Sesto Fiorentino (FI) 50019, Italy
| | - Stefano Toppo
- Department of Molecular Medicine University of Padova, viale G. Colombo, 3, Padova 35131, Italy; and
| | - Nicola Facchinello
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Mattia Santimaria
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Andrea Vettori
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Stefania Rizzo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Mila Della Barbera
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Kalliopi Pilichou
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Francesco Argenton
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Gaetano Thiene
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, via U. Bassi, 58/B, Padova 35131, Italy
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, via A. Gabelli, 61, Padova 35121, Italy
| |
Collapse
|
21
|
Norman J, Sorrell EL, Hu Y, Siripurapu V, Garcia J, Bagwell J, Charbonneau P, Lubkin SR, Bagnat M. Tissue self-organization underlies morphogenesis of the notochord. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2017.0320. [PMID: 30249771 DOI: 10.1098/rstb.2017.0320] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
The notochord is a conserved axial structure that in vertebrates serves as a hydrostatic scaffold for embryonic axis elongation and, later on, for proper spine assembly. It consists of a core of large fluid-filled vacuolated cells surrounded by an epithelial sheath that is encased in extracellular matrix. During morphogenesis, the vacuolated cells inflate their vacuole and arrange in a stereotypical staircase pattern. We investigated the origin of this pattern and found that it can be achieved purely by simple physical principles. We are able to model the arrangement of vacuolated cells within the zebrafish notochord using a physical model composed of silicone tubes and water-absorbing polymer beads. The biological structure and the physical model can be accurately described by the theory developed for the packing of spheres and foams in cylinders. Our experiments with physical models and numerical simulations generated several predictions on key features of notochord organization that we documented and tested experimentally in zebrafish. Altogether, our data reveal that the organization of the vertebrate notochord is governed by the density of the osmotically swelling vacuolated cells and the aspect ratio of the notochord rod. We therefore conclude that self-organization underlies morphogenesis of the vertebrate notochord.This article is part of the Theo Murphy meeting issue on 'Mechanics of development'.
Collapse
Affiliation(s)
- James Norman
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Emma L Sorrell
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.,Department of Mathematics, North Carolina State University, Raleigh, NC 27695-8205, USA
| | - Yi Hu
- Department of Chemistry, Duke University, Durham, NC 27710, USA
| | - Vaishnavi Siripurapu
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.,North Carolina School of Science and Mathematics, Durham, NC 27705, USA
| | - Jamie Garcia
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Jennifer Bagwell
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | | | - Sharon R Lubkin
- Department of Mathematics, North Carolina State University, Raleigh, NC 27695-8205, USA
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
22
|
Chen JH, Kuo KT, Bamodu OA, Lin YC, Yang RB, Yeh CT, Chao TY. Upregulated SCUBE2 expression in breast cancer stem cells enhances triple negative breast cancer aggression through modulation of notch signaling and epithelial-to-mesenchymal transition. Exp Cell Res 2018; 370:444-453. [PMID: 29981340 DOI: 10.1016/j.yexcr.2018.07.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/01/2018] [Accepted: 07/03/2018] [Indexed: 01/16/2023]
Abstract
BACKGROUND Metastatic and/or recurrent breast carcinomas are leading causes of cancer-related death worldwide. Breast cancer stem cells (BCSCs) have been implicated in cancer metastases and progression, thus, the need for the discovery and development of effective BCSCs-specific therapies against metastatic and triple negative breast cancer (TNBC). The expression of SCUBE2, originally identified in vascular endothelia, then in several non-endothelial cell types, is downregulated in invasive breast carcinomas. However, the role of SCUBE2 in BCSCs remains unknown. This present study investigated the probable involvements of SCUBE2 in BCSCs and TNBC metastasis. METHODS The mRNA expression of SCUBE2, stemness and EMT markers in MDA-MB-231 and Hs578T tumorspheres or adherent cells were evaluated by qRT-PCR and microarray analyses. Using gene overexpression, in vitro migration and invasion assays, as well as in vivo bioluminescence imaging, we evaluated the role of SCUBE2 in MDA-MB-231 or Hs578T BCSCs. Western blot and cytotoxicity assays helped identify and validate SCUBE2 molecular target(s) and inhibitor(s). RESULTS Concurrently increased SCUBE2 expression and cell motility were observed in TNBC tumorspheres compared to the parental adherent cells. SCUBE2 overexpression augmented BCSCs motility in vitro, and enhanced TNBC metastasis in vivo. While SCUBE2 overexpression activated Notch signaling its downregulation suppressed Notch signal effectors NICD, Jagged 1, HEY1, and HES1. CONCLUSIONS We demonstrate that SCUBE2 expression is upregulated in BCSCs, promote EMT and enhance TNBC metastasis by activating Notch signaling. This reveals a potential druggable molecular target and an effective therapeutic strategy against metastatic and aggressive TNBC.
Collapse
Affiliation(s)
- Jia-Hong Chen
- Division of Medical Oncology and Hematology, Tri-Service General Hospital, National Defense Medical Centre, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Kuang-Tai Kuo
- Division of Thoracic Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Division of Thoracic Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Oluwaseun Adebayo Bamodu
- Department of Hematology and Oncology, Cancer Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Medical Research and Education, Taipei Medical University, Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yuh-Charn Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ruey-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Chi-Tai Yeh
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan; Department of Hematology and Oncology, Cancer Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Medical Research and Education, Taipei Medical University, Shuang Ho Hospital, New Taipei City, Taiwan.
| | - Tsu-Yi Chao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan; Department of Hematology and Oncology, Cancer Center, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan; Department of Medical Research and Education, Taipei Medical University, Shuang Ho Hospital, New Taipei City, Taiwan.
| |
Collapse
|
23
|
De Clercq A, Perrott MR, Davie PS, Preece MA, Huysseune A, Witten PE. The external phenotype-skeleton link in post-hatch farmed Chinook salmon (Oncorhynchus tshawytscha). JOURNAL OF FISH DISEASES 2018; 41:511-527. [PMID: 29159824 DOI: 10.1111/jfd.12753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/28/2017] [Accepted: 10/02/2017] [Indexed: 06/07/2023]
Abstract
Skeletal deformities in farmed fish are a recurrent problem. External malformations are easily recognized, but there is little information on how external malformations relate to malformations of the axial skeleton: the external phenotype-skeleton link. Here, this link is studied in post-hatch to first-feed life stages of Chinook salmon (Oncorhynchus tshawytscha) raised at 4, 8 and 12°C. Specimens were whole-mount-stained for cartilage and bone, and analysed by histology. In all temperature groups, externally normal specimens can have internal malformations, predominantly fused vertebral centra. Conversely, externally malformed fish usually display internal malformations. Externally curled animals typically have malformed haemal and neural arches. External malformations affecting a single region (tail malformation and bent neck) relate to malformed notochords and early fusion of fused vertebral centra. The frequencies of internal malformations in both externally normal and malformed specimens show a U-shaped response, with lowest frequency in 8°C specimens. The fused vertebral centra that occur in externally normal specimens represent a malformation that can be contained and could be carried through into harvest size animals. This study highlights the relationship between external phenotype and axial skeleton and may help to set the framework for the early identification of skeletal malformations on fish farms.
Collapse
Affiliation(s)
- A De Clercq
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North, New Zealand
- Evolutionary Developmental Biology, Ghent University, Ghent, Belgium
| | - M R Perrott
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North, New Zealand
| | - P S Davie
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North, New Zealand
| | - M A Preece
- New Zealand King Salmon, Nelson, New Zealand
| | - A Huysseune
- Evolutionary Developmental Biology, Ghent University, Ghent, Belgium
| | - P E Witten
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North, New Zealand
- Evolutionary Developmental Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
24
|
Trapani V, Bonaldo P, Corallo D. Role of the ECM in notochord formation, function and disease. J Cell Sci 2017; 130:3203-3211. [PMID: 28883093 DOI: 10.1242/jcs.175950] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The notochord is a midline structure common to all chordate animals; it provides mechanical and signaling cues for the developing embryo. In vertebrates, the notochord plays key functions during embryogenesis, being a source of developmental signals that pattern the surrounding tissues. It is composed of a core of vacuolated cells surrounded by an epithelial-like sheath of cells that secrete a thick peri-notochordal basement membrane made of different extracellular matrix (ECM) proteins. The correct deposition and organization of the ECM is essential for proper notochord morphogenesis and function. Work carried out in the past two decades has allowed researchers to dissect the contribution of different ECM components to this embryonic tissue. Here, we will provide an overview of these genetic and mechanistic studies. In particular, we highlight the specific functions of distinct matrix molecules in regulating notochord development and notochord-derived signals. Moreover, we also discuss the involvement of ECM synthesis and its remodeling in the pathogenesis of chordoma, a malignant bone cancer that originates from remnants of notochord remaining after embryogenesis.
Collapse
Affiliation(s)
- Valeria Trapani
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy .,CRIBI Biotechnology Center, University of Padova, Padova, 35131, Italy
| | - Diana Corallo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy .,Pediatric Research Institute, Città della Speranza, 35127 Padova, Italy
| |
Collapse
|
25
|
Wang F, Gao ZX, Cai F, Sinkemani A, Xie ZY, Shi R, Wei JN, Wu XT. Formation, function, and exhaustion of notochordal cytoplasmic vacuoles within intervertebral disc: current understanding and speculation. Oncotarget 2017; 8:57800-57812. [PMID: 28915712 PMCID: PMC5593684 DOI: 10.18632/oncotarget.18101] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 05/01/2017] [Indexed: 01/08/2023] Open
Abstract
Notochord nucleus pulposus cells are characteristic of containing abundant and giant cytoplasmic vacuoles. This review explores the embryonic formation, biological function, and postnatal exhaustion of notochord vacuoles, aiming to characterize the signal network transforming the vacuolated nucleus pulposus cells into the vacuole-less chondrocytic cells. Embryonically, the cytoplasmic vacuoles within vertebrate notochord originate from an evolutionarily conserved vacuolation process during neurulation, which may continue to provide mechanical and signal support in constructing a mammalian intervertebral disc. For full vacuolation, a vacuolating specification from dorsal organizer cells, synchronized convergent extension, well-structured notochord sheath, and sufficient post-Golgi trafficking in notochord cells are required. Postnatally, age-related and species-specific exhaustion of vacuolated nucleus pulposus cells could be potentiated by Fas- and Fas ligand-induced apoptosis, intolerance to mechanical stress and nutrient deficiency, vacuole-mediated proliferation check, and gradual de-vacuolation within the avascular and compression-loaded intervertebral disc. These results suggest that the notochord vacuoles are active and versatile organelles for both embryonic notochord and postnatal nucleus pulposus, and may provide novel information on intervertebral disc degeneration to guide cell-based regeneration.
Collapse
Affiliation(s)
- Feng Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Zeng-Xin Gao
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Feng Cai
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Soochow, Jiangsu Province, China
| | - Arjun Sinkemani
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Zhi-Yang Xie
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Rui Shi
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Ji-Nan Wei
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Xiao-Tao Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| |
Collapse
|
26
|
Schiavinato A, Keene DR, Imhof T, Doliana R, Sasaki T, Sengle G. Fibulin-4 deposition requires EMILIN-1 in the extracellular matrix of osteoblasts. Sci Rep 2017; 7:5526. [PMID: 28717224 PMCID: PMC5514116 DOI: 10.1038/s41598-017-05835-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 06/05/2017] [Indexed: 11/09/2022] Open
Abstract
Tissue microenvironments formed by extracellular matrix networks play an important role in regulating tissue structure and function. Extracellular microfibrillar networks composed of fibrillins and their associated ligands such as LTBPs, fibulins, and EMILINs are of particular interest in this regard since they provide a specialized cellular microenvironment guiding proper morphology and functional behavior of specialized cell types. To understand how cellular microenvironments composed of intricate microfibrillar networks influence cell fate decisions in a contextual manner, more information about the spatiotemporal localization, deposition, and function of their components is required. By employing confocal immunofluorescence and electron microscopy we investigated the localization and extracellular matrix deposition of EMILIN-1 and -2 in tissues of the skeletal system such as cartilage and bone as well as in in vitro cultures of osteoblasts. We found that upon RNAi mediated depletion of EMILIN-1 in primary calvarial osteoblasts and MC3T3-E1 cells only fibulin-4 matrix deposition was lost while other fibulin family members or LTBPs remained unaffected. Immunoprecipitation and ELISA-style binding assays confirmed a direct interaction between EMILIN-1 and fibulin-4. Our data suggest a new function for EMILIN-1 which implies the guidance of linear fibulin-4 matrix deposition and thereby fibulin-4 fiber formation.
Collapse
Affiliation(s)
- Alvise Schiavinato
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | | | - Thomas Imhof
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany.,Institute for Dental Research and Oral Musculoskeletal Biology, Medical Faculty, University of Cologne, 50931, Cologne, Germany
| | - Roberto Doliana
- Experimental Oncology 2, CRO, IRCCS, National Cancer Institute, Aviano, PN, Italy
| | - Takako Sasaki
- Department of Biochemistry II, Faculty of Medicine, Oita University, Oita, 879-5593, Japan
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany. .,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
27
|
Jacob AE, Amack JD, Turner CE. Paxillin genes and actomyosin contractility regulate myotome morphogenesis in zebrafish. Dev Biol 2017; 425:70-84. [PMID: 28315297 DOI: 10.1016/j.ydbio.2017.03.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/10/2017] [Accepted: 03/12/2017] [Indexed: 02/07/2023]
Abstract
Paxillin (Pxn) is a key adapter protein and signaling regulator at sites of cell-extracellular matrix (ECM) adhesion. Here, we investigated the role of Pxn during vertebrate development using the zebrafish embryo as a model system. We have characterized two Pxn genes, pxna and pxnb, in zebrafish that are maternally supplied and expressed in multiple tissues. Gene editing and antisense gene knockdown approaches were used to uncover Pxn functions during zebrafish development. While mutation of either pxna or pxnb alone did not cause gross embryonic phenotypes, double mutants lacking maternally supplied pxna or pxnb displayed defects in cardiovascular, axial, and skeletal muscle development. Transient knockdown of Pxn proteins resulted in similar defects. Irregular myotome shape and ECM composition were observed, suggesting an "inside-out" signaling role for Paxillin genes in the development of myotendinous junctions. Inhibiting non-muscle Myosin-II during somitogenesis altered the subcellular localization of Pxn protein and phenocopied pxn gene loss-of-function. This indicates that Paxillin genes are effectors of actomyosin contractility-driven morphogenesis of trunk musculature in zebrafish. Together, these results reveal new functions for Pxn during muscle development and provide novel genetic models to elucidate Pxn functions.
Collapse
Affiliation(s)
- Andrew E Jacob
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, United States
| | - Jeffrey D Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, United States.
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, United States.
| |
Collapse
|
28
|
Corallo D, Schiavinato A, Bizzotto D, Milanetto M, Guljelmovic M, Keene DR, Sengle G, Braghetta P, Bonaldo P. EMILIN3, an extracellular matrix molecule with restricted distribution in skin. Exp Dermatol 2017; 26:435-438. [DOI: 10.1111/exd.13254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Diana Corallo
- Department of Molecular Medicine; University of Padova; Padova Italy
| | | | - Dario Bizzotto
- Department of Molecular Medicine; University of Padova; Padova Italy
| | - Martina Milanetto
- Department of Molecular Medicine; University of Padova; Padova Italy
| | | | | | - Gerhard Sengle
- Center for Biochemistry; Medical Faculty, University of Cologne; Cologne Germany
- Center for Molecular Medicine Cologne (CMMC); University of Cologne; Cologne Germany
| | - Paola Braghetta
- Department of Molecular Medicine; University of Padova; Padova Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine; University of Padova; Padova Italy
- CRIBI Biotechnology Center; University of Padova; Padova Italy
| |
Collapse
|
29
|
Salva JE, Merrill AE. Signaling networks in joint development. Dev Dyn 2016; 246:262-274. [PMID: 27859991 DOI: 10.1002/dvdy.24472] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/09/2016] [Accepted: 11/14/2016] [Indexed: 12/21/2022] Open
Abstract
Here we review studies identifying regulatory networks responsible for synovial, cartilaginous, and fibrous joint development. Synovial joints, characterized by the fluid-filled synovial space between the bones, are found in high-mobility regions and are the most common type of joint. Cartilaginous joints such as the intervertebral disc unite adjacent bones through either a hyaline cartilage or a fibrocartilage intermediate. Fibrous joints, which include the cranial sutures, form a direct union between bones through fibrous connective tissue. We describe how the distinct morphologic and histogenic characteristics of these joint classes are established during embryonic development. Collectively, these studies reveal that despite the heterogeneity of joint strength and mobility, joint development throughout the skeleton utilizes common signaling networks via long-range morphogen gradients and direct cell-cell contact. This suggests that different joint types represent specialized variants of homologous developmental modules. Identifying the unifying aspects of the signaling networks between joint classes allows a more complete understanding of the signaling code for joint formation, which is critical to improving strategies for joint regeneration and repair. Developmental Dynamics 246:262-274, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joanna E Salva
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
30
|
Extracellular Matrix, a Hard Player in Angiogenesis. Int J Mol Sci 2016; 17:ijms17111822. [PMID: 27809279 PMCID: PMC5133823 DOI: 10.3390/ijms17111822] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/30/2016] [Accepted: 10/21/2016] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins, glycoproteins, proteoglycans, and polysaccharides. Through multiple interactions with each other and the cell surface receptors, not only the ECM determines the physical and mechanical properties of the tissues, but also profoundly influences cell behavior and many physiological and pathological processes. One of the functions that have been extensively explored is its impingement on angiogenesis. The strong impact of the ECM in this context is both direct and indirect by virtue of its ability to interact and/or store several growth factors and cytokines. The aim of this review is to provide some examples of the complex molecular mechanisms that are elicited by these molecules in promoting or weakening the angiogenic processes. The scenario is intricate, since matrix remodeling often generates fragments displaying opposite effects compared to those exerted by the whole molecules. Thus, the balance will tilt towards angiogenesis or angiostasis depending on the relative expression of pro- or anti-angiogenetic molecules/fragments composing the matrix of a given tissue. One of the vital aspects of this field of research is that, for its endogenous nature, the ECM can be viewed as a reservoir to draw from for the development of new more efficacious therapies to treat angiogenesis-dependent pathologies.
Collapse
|
31
|
Schiavinato A, Keene DR, Wohl AP, Corallo D, Colombatti A, Wagener R, Paulsson M, Bonaldo P, Sengle G. Targeting of EMILIN-1 and EMILIN-2 to Fibrillin Microfibrils Facilitates their Incorporation into the Extracellular Matrix. J Invest Dermatol 2016; 136:1150-1160. [PMID: 26945878 DOI: 10.1016/j.jid.2016.02.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/03/2016] [Accepted: 02/18/2016] [Indexed: 10/22/2022]
Abstract
Elastin microfibril interface-located proteins (EMILINs) 1 and 2 belong to a family of structurally related extracellular glycoproteins with unique functions in the extracellular space, such as modulation of pro-transforming growth factor-β processing, activation of the extrinsic apoptotic pathway, and regulation of Hedgehog and Wnt ligand bioavailability. However, little is known about how EMILINs may exert their extracellular functions. We therefore investigated the spatiotemporal localization and deposition of EMILIN-1 and -2 within the extracellular space. By using immunoelectron and immunofluorescence microscopy together with biochemical extraction, we showed that EMILIN-1 and -2 are targeted to fibrillin microfibrils in the skin. In addition, during skin wound healing and in vitro matrix fiber assembly by primary dermal fibroblasts, EMILIN-1 and -2 are deposited on and coregulated with fibrillin. Analysis of wounds and mouse embryonic fibroblast cultures showed that EMILIN-1 and -2 network formation also requires the presence of fibronectin. Disruption of microfibrils in fibrillin-1-deficient mice leads to fragmentation of the EMILIN-1 and -2 networks, suggesting an involvement of EMILINs in fibrillin-related skin disorders. The addition of EMILINs to the ligand repertoire of fibrillin strengthens the concept of fibrillin microfibrils as extracellular scaffolds integrating cellular force transmission and growth factor bioactivity.
Collapse
Affiliation(s)
- Alvise Schiavinato
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany; Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Alexander P Wohl
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Diana Corallo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Alfonso Colombatti
- Experimental Oncology 2, CRO, IRCCS, National Cancer Institute, Aviano, PN, Italy
| | - Raimund Wagener
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Mats Paulsson
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
32
|
Abstract
The intervertebral disc is a complex structure responsible for flexibility, multi-axial motion, and load transmission throughout the spine. Importantly, degeneration of the intervertebral disc is thought to be an initiating factor for back pain. Due to a lack of understanding of the pathways that govern disc degeneration, there are currently no disease-modifying treatments to delay or prevent degenerative disc disease. This review presents an overview of our current understanding of the developmental processes that regulate intervertebral disc formation, with particular emphasis on the role of the notochord and notochord-derived cells in disc homeostasis and how their loss can result in degeneration. We then describe the role of small animal models in understanding the development of the disc and their use to interrogate disc degeneration and associated pathologies. Finally, we highlight essential development pathways that are associated with disc degeneration and/or implicated in the reparative response of the tissue that might serve as targets for future therapeutic approaches.
Collapse
|
33
|
Corallo D, Trapani V, Bonaldo P. The notochord: structure and functions. Cell Mol Life Sci 2015; 72:2989-3008. [PMID: 25833128 PMCID: PMC11114051 DOI: 10.1007/s00018-015-1897-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/23/2015] [Accepted: 03/26/2015] [Indexed: 01/08/2023]
Abstract
The notochord is an embryonic midline structure common to all members of the phylum Chordata, providing both mechanical and signaling cues to the developing embryo. In vertebrates, the notochord arises from the dorsal organizer and it is critical for proper vertebrate development. This evolutionary conserved structure located at the developing midline defines the primitive axis of embryos and represents the structural element essential for locomotion. Besides its primary structural function, the notochord is also a source of developmental signals that patterns surrounding tissues. Among the signals secreted by the notochord, Hedgehog proteins play key roles during embryogenesis. The Hedgehog signaling pathway is a central regulator of embryonic development, controlling the patterning and proliferation of a wide variety of organs. In this review, we summarize the current knowledge on notochord structure and functions, with a particular emphasis on the key developmental events that take place in vertebrates. Moreover, we discuss some genetic studies highlighting the phenotypic consequences of impaired notochord development, which enabled to understand the molecular basis of different human congenital defects and diseases.
Collapse
Affiliation(s)
- Diana Corallo
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| | - Valeria Trapani
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| |
Collapse
|
34
|
Benato F, Colletti E, Skobo T, Moro E, Colombo L, Argenton F, Dalla Valle L. A living biosensor model to dynamically trace glucocorticoid transcriptional activity during development and adult life in zebrafish. Mol Cell Endocrinol 2014; 392:60-72. [PMID: 24796658 DOI: 10.1016/j.mce.2014.04.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 04/24/2014] [Accepted: 04/24/2014] [Indexed: 11/18/2022]
Abstract
Glucocorticoids (GCs) modulate many cellular processes through the binding of the glucocorticoid receptor (GR) to specific responsive elements located upstream of the transcription starting site or within an intron of GC target genes. Here we describe a transgenic fish line harboring a construct with nine GC-responsive elements (GREs) upstream of a reporter (EGFP) coding sequence. Transgenic fish exhibit strong fluorescence in many known GC-responsive organs. Moreover, its enhanced sensitivity allowed the discovery of novel GC-responsive tissue compartments, such as fin, eyes, and otic vesicles. Long-term persistence of transgene expression is seen during adult stages in several organs. Pharmacological and genetic analysis demonstrates that the transgenic line is highly responsive to drug administration and molecular manipulation. Moreover, reporter expression is sensitively and dynamically modulated by the photoperiod, thus proving that these fish are an in vivo valuable platform to explore GC responsiveness to both endogenous and exogenous stimuli.
Collapse
Affiliation(s)
- Francesca Benato
- Department of Biology, University of Padua, via U. Bassi 58/B, 35131 Padua, Italy
| | - Elisa Colletti
- Department of Biology, University of Padua, via U. Bassi 58/B, 35131 Padua, Italy
| | - Tatjana Skobo
- Department of Biology, University of Padua, via U. Bassi 58/B, 35131 Padua, Italy
| | - Enrico Moro
- Department of Molecular Medicine, University of Padua, via U. Bassi 58/B, 35131 Padua, Italy
| | - Lorenzo Colombo
- Department of Biology, University of Padua, via U. Bassi 58/B, 35131 Padua, Italy
| | - Francesco Argenton
- Department of Biology, University of Padua, via U. Bassi 58/B, 35131 Padua, Italy.
| | - Luisa Dalla Valle
- Department of Biology, University of Padua, via U. Bassi 58/B, 35131 Padua, Italy.
| |
Collapse
|
35
|
Schiavone M, Rampazzo E, Casari A, Battilana G, Persano L, Moro E, Liu S, Leach SD, Tiso N, Argenton F. Zebrafish reporter lines reveal in vivo signaling pathway activities involved in pancreatic cancer. Dis Model Mech 2014; 7:883-94. [PMID: 24878567 PMCID: PMC4073277 DOI: 10.1242/dmm.014969] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pancreatic adenocarcinoma, one of the worst malignancies of the exocrine pancreas, is a solid tumor with increasing incidence and mortality in industrialized countries. This condition is usually driven by oncogenic KRAS point mutations and evolves into a highly aggressive metastatic carcinoma due to secondary gene mutations and unbalanced expression of genes involved in the specific signaling pathways. To examine in vivo the effects of KRASG12D during pancreatic cancer progression and time correlation with cancer signaling pathway activities, we have generated a zebrafish model of pancreatic adenocarcinoma in which eGFP-KRASG12D expression was specifically driven to the pancreatic tissue by using the GAL4/UAS conditional expression system. Outcrossing the inducible oncogenic KRASG12D line with transgenic zebrafish reporters, harboring specific signaling responsive elements of transcriptional effectors, we were able to follow TGFβ, Notch, Bmp and Shh activities during tumor development. Zebrafish transgenic lines expressing eGFP-KRASG12D showed normal exocrine pancreas development until 3 weeks post fertilization (wpf). From 4 to 24 wpf we observed several degrees of acinar lesions, characterized by an increase in mesenchymal cells and mixed acinar/ductal features, followed by progressive bowel and liver infiltrations and, finally, highly aggressive carcinoma. Moreover, live imaging analysis of the exocrine pancreatic tissue revealed an increasing number of KRAS-positive cells and progressive activation of TGFβ and Notch pathways. Increase in TGFβ, following KRASG12D activation, was confirmed in a concomitant model of medulloblastoma (MDB). Notch and Shh signaling activities during tumor onset were different between MDB and pancreatic adenocarcinoma, indicating a tissue-specific regulation of cell signaling pathways. Moreover, our results show that a living model of pancreatic adenocarcinoma joined with cell signaling reporters is a suitable tool for describing in vivo the signaling cascades and molecular mechanisms involved in tumor development and a potential platform to screen for novel oncostatic drugs.
Collapse
Affiliation(s)
- Marco Schiavone
- Department of Biology, University of Padua, 35131 Padua, Italy
| | - Elena Rampazzo
- Department of Molecular Medicine, University of Padua, 35131 Padua, Italy
| | | | - Giusy Battilana
- Department of Molecular Medicine, University of Padua, 35131 Padua, Italy
| | - Luca Persano
- Department of Woman and Child Health, University of Padua, 35131 Padua, Italy
| | - Enrico Moro
- Department of Molecular Medicine, University of Padua, 35131 Padua, Italy
| | - Shu Liu
- Department of Surgery and The McKusick-Nathans Institute of Genetic Medicine Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Steve D Leach
- Department of Surgery and The McKusick-Nathans Institute of Genetic Medicine Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Natascia Tiso
- Department of Biology, University of Padua, 35131 Padua, Italy
| | | |
Collapse
|
36
|
Ferent J, Traiffort E. Hedgehog: Multiple Paths for Multiple Roles in Shaping the Brain and Spinal Cord. Neuroscientist 2014; 21:356-71. [PMID: 24743306 DOI: 10.1177/1073858414531457] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Since the discovery of the segment polarity gene Hedgehog in Drosophila three decades ago, our knowledge of Hedgehog signaling pathway has considerably improved and paved the way to a wide field of investigations in the developing and adult central nervous system. Its peculiar transduction mechanism together with its implication in tissue patterning, neural stem cell biology, and neural tissue homeostasis make Hedgehog pathway of interest in a high number of normal or pathological contexts. Consistent with its role during brain development, misregulation of Hedgehog signaling is associated with congenital diseases and tumorigenic processes while its recruitment in damaged neural tissue may be part of the repairing process. This review focuses on the most recent data regarding the Hedgehog pathway in the developing and adult central nervous system and also its relevance as a therapeutic target in brain and spinal cord diseases.
Collapse
Affiliation(s)
- Julien Ferent
- IRCM, Molecular Biology of Neural Development, Montreal, Quebec, Canada
| | - Elisabeth Traiffort
- INSERM-Université Paris Sud, Neuroprotection and Neuroregeneration: Small Neuroactive Molecules UMR 788, Le Kremlin-Bicêtre, France
| |
Collapse
|