1
|
Sarnella A, Ferrara Y, Terlizzi C, Albanese S, Monti S, Licenziato L, Mancini M. The Chicken Embryo: An Old but Promising Model for In Vivo Preclinical Research. Biomedicines 2024; 12:2835. [PMID: 39767740 PMCID: PMC11673736 DOI: 10.3390/biomedicines12122835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The chicken embryo has emerged as a valuable model for preclinical studies due to its unique combination of accessibility, affordability, and relevance to human biology. Its rapid development, external growth environment, and clear structural visibility offer distinct advantages over traditional mammalian models. These features facilitate the study of real-time biological processes, including tissue development, tumor growth, angiogenesis, and drug delivery, using various imaging modalities, such as optical imaging, magnetic resonance imaging, positron emission tomography, computed tomography, and ultrasound. The chicken embryo model also minimizes ethical concerns compared to mammalian models, as it allows for early-stage research without the complexity of a fully developed animal. Moreover, its ability to integrate human tumor cells into xenograft models provides a reliable platform for cancer research, enabling high-throughput screening of therapeutic interventions and tracking molecular dynamics in vivo. Advances in molecular imaging techniques further enhance the resolution and depth of data obtained from these studies, offering insights into cellular and molecular mechanisms underlying disease. Given its versatility, cost-effectiveness, and translational potential, the chicken embryo represents a promising tool for advancing preclinical research, particularly in drug development, cancer biology, and regenerative medicine.
Collapse
Affiliation(s)
| | | | - Cristina Terlizzi
- Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy; (A.S.); (Y.F.); (S.A.); (S.M.); (L.L.); (M.M.)
| | | | | | | | | |
Collapse
|
2
|
Bertrand JU, Petit V, Aktary Z, de la Grange P, Elkoshi N, Sohier P, Delmas V, Levy C, Larue L. Loss of Dicer in Newborn Melanocytes Leads to Premature Hair Graying and Changes in Integrin Expression. J Invest Dermatol 2024; 144:601-611. [PMID: 37739336 DOI: 10.1016/j.jid.2023.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/24/2023]
Abstract
Premature hair graying occurs owing to the depletion of melanocyte stem cells in the hair follicle, which can be accelerated by stress caused by genetic or environmental factors. However, the connection between stress and melanocyte stem cell loss is not fully understood. MicroRNAs are molecules that control gene expression by regulating mRNA stability and translation and are produced by the enzyme Dicer, which is repressed under stress. In this study, using 2 mouse genetic models and human and mouse cell lines, we found that the inactivation of Dicer in melanocytes leads to misplacement of these cells within the hair follicle, resulting in a lack of melanin transfer to keratinocytes in the growing hair and the exhaustion of the melanocyte stem cell pool. We also show that miR-92b, which regulates ItgaV mRNA and protein levels, plays a role in altering melanocyte migration. Overall, our findings suggest that the Dicer-miR92b-ItgaV pathway serves as a major signaling pathway linking stress to premature hair greying.
Collapse
Affiliation(s)
- Juliette U Bertrand
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR 3347, Univ Paris-Sud, Univ Paris-Saclay, Orsay, France
| | - Valérie Petit
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR 3347, Univ Paris-Sud, Univ Paris-Saclay, Orsay, France
| | - Zackie Aktary
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR 3347, Univ Paris-Sud, Univ Paris-Saclay, Orsay, France
| | | | - Nadav Elkoshi
- Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Pierre Sohier
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR 3347, Univ Paris-Sud, Univ Paris-Saclay, Orsay, France
| | - Véronique Delmas
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR 3347, Univ Paris-Sud, Univ Paris-Saclay, Orsay, France
| | - Carmit Levy
- Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Lionel Larue
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Orsay, France; Centre National de la Recherche Scientifique (CNRS) UMR 3347, Univ Paris-Sud, Univ Paris-Saclay, Orsay, France.
| |
Collapse
|
3
|
Tang H, Zhang Y, Yang L, Hong C, Chen K, Li Y, Wu H. Serotonin/5-HT7 receptor provides an adaptive signal to enhance pigmentation response to environmental stressors through cAMP-PKA-MAPK, Rab27a/RhoA, and PI3K/AKT signaling pathways. FASEB J 2023; 37:e22893. [PMID: 36961387 PMCID: PMC11977531 DOI: 10.1096/fj.202201352rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/05/2023] [Accepted: 03/13/2023] [Indexed: 03/25/2023]
Abstract
Serotonin (5-HT), a neurotransmitter, is essential for normal and pathological pigmentation processing, and its receptors may be therapeutical targets. The effect and behavior of the 5-HT7 receptor (5-HT7R) in melanogenesis in high vertebrates remain unknown. Herein, we examine the role and molecular mechanism of 5-HT7R in the pigmentation of human skin cells, human tissue, mice, and zebrafish models. Firstly, 5-HT7R protein expression decreased significantly in stress-induced depigmentation skin and vitiligo epidermis. Stressed mice received transdermal serotonin 5-HT7R selective agonists (LP-12, 0.01%) for 12 or 60 days. Mice might recover from persistent stress-induced depigmentation. The downregulation of tyrosinase (Tyr), microphthalmia-associated transcription factor (Mitf) expression, and 5-HT7R was consistently restored in stressed skin. High-throughput RNA sequencing showed that structural organization (dendrite growth and migration) and associated pathways were activated in the dorsal skin of LP-12-treated animals. 5-HT7R selective agonist, LP-12, had been demonstrated to enhance melanin production, dendrite growth, and chemotactic motility in B16F10 cells, normal human melanocytes (NHMCs), and zebrafish. Mechanistically, the melanogenic, dendritic, and migratory functions of 5-HT7R were dependent on the downstream signaling of cAMP-PKA-ERK1/2, JNK MAPK, RhoA/Rab27a, and PI3K/AKT pathway activation. Importantly, pharmacological inhibition and genetic siRNA of 5-HT7R by antagonist SB269970 partially/completely abolished these functional properties and the related activated pathways in both NHMCs and B16F10 cells. Consistently, htr7a/7b genetic knockdown in zebrafish could blockade melanogenic effects and abrogate 5-HT-induced melanin accumulation. Collectively, we have first identified that 5-HT7R regulates melanogenesis, which may be a targeted therapy for pigmentation disorders, especially those worsened by stress.
Collapse
Affiliation(s)
- Hui‐hao Tang
- Department of TCM Chemistry, School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yi‐fan Zhang
- Department of TCM Chemistry, School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Li‐li Yang
- Department of DermatologyShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chen Hong
- Department of TCM Chemistry, School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Kai‐xian Chen
- Department of TCM Chemistry, School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Yi‐ming Li
- Department of TCM Chemistry, School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Hua‐li Wu
- Department of TCM Chemistry, School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
4
|
Yahya I, Brand-Saberi B, Morosan-Puopolo G. Chicken embryo as a model in second heart field development. Heliyon 2023; 9:e14230. [PMID: 36923876 PMCID: PMC10009738 DOI: 10.1016/j.heliyon.2023.e14230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/30/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Previously, a single source of progenitor cells was thought to be responsible for the formation of the cardiac muscle. However, the second heart field has recently been identified as an additional source of myocardial progenitor cells. The chicken embryo, which develops in the egg, outside the mother can easily be manipulated in vivo and in vitro. Hence, it was an excellent model for establishing the concept of the second heart field. Here, our review will focus on the chicken model, specifically its role in understanding the second heart field. In addition to discussing historical aspects, we provide an overview of recent findings that have helped to define the chicken second heart field progenitor cells. A better understanding of the second heart field development will provide important insights into the congenital malformations affecting cardiac muscle formation and function.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801, Bochum, Germany
- Department of Anatomy, Faculty of Veterinary Medicine, University of Khartoum, Khartoum, 11115, Sudan
- Corresponding author. Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801 Bochum, Germany.
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801, Bochum, Germany
| | | |
Collapse
|
5
|
Haage A, Wagner K, Deng W, Venkatesh B, Mitchell C, Goodwin K, Bogutz A, Lefebvre L, Van Raamsdonk CD, Tanentzapf G. Precise coordination of cell-ECM adhesion is essential for efficient melanoblast migration during development. Development 2020; 147:dev.184234. [PMID: 32580934 DOI: 10.1242/dev.184234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 06/08/2020] [Indexed: 01/07/2023]
Abstract
Melanoblasts disperse throughout the skin and populate hair follicles through long-range cell migration. During migration, cells undergo cycles of coordinated attachment and detachment from the extracellular matrix (ECM). Embryonic migration processes that require cell-ECM attachment are dependent on the integrin family of adhesion receptors. Precise regulation of integrin-mediated adhesion is important for many developmental migration events. However, the mechanisms that regulate integrin-mediated adhesion in vivo in melanoblasts are not well understood. Here, we show that autoinhibitory regulation of the integrin-associated adapter protein talin coordinates cell-ECM adhesion during melanoblast migration in vivo Specifically, an autoinhibition-defective talin mutant strengthens and stabilizes integrin-based adhesions in melanocytes, which impinges on their ability to migrate. Mice with defective talin autoinhibition exhibit delays in melanoblast migration and pigmentation defects. Our results show that coordinated integrin-mediated cell-ECM attachment is essential for melanoblast migration and that talin autoinhibition is an important mechanism for fine-tuning cell-ECM adhesion during cell migration in development.
Collapse
Affiliation(s)
- Amanda Haage
- Department of Biomedical Sciences, University of North Dakota, 1301 N Columbia Rd, Grand Forks, ND 58202, ND, USA
| | - Kelsey Wagner
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| | - Wenjun Deng
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| | - Bhavya Venkatesh
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| | - Caitlin Mitchell
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| | - Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
| | - Aaron Bogutz
- Department of Medical Genetics, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Louis Lefebvre
- Department of Medical Genetics, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Catherine D Van Raamsdonk
- Department of Medical Genetics, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, 2350 Health Sciences Mall, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Vandamme N, Berx G. From neural crest cells to melanocytes: cellular plasticity during development and beyond. Cell Mol Life Sci 2019; 76:1919-1934. [PMID: 30830237 PMCID: PMC11105195 DOI: 10.1007/s00018-019-03049-w] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/25/2019] [Accepted: 02/18/2019] [Indexed: 01/07/2023]
Abstract
Here, we review melanocyte development and how the embryonic melanoblast, although specified to become a melanocyte, is prone to cellular plasticity and is not fully committed to the melanocyte lineage. Even fully differentiated and pigment-producing melanocytes do not always have a stable phenotype. The gradual lineage restriction of neural crest cells toward the melanocyte lineage is determined by both cell-intrinsic and extracellular signals in which differentiation and pathfinding ability reciprocally influence each other. These signals are leveraged by subtle differences in timing and axial positioning. The most extensively studied migration route is the dorsolateral path between the dermomyotome and the prospective epidermis, restricted to melanoblasts. In addition, the embryonic origin of the skin dermis through which neural crest derivatives migrate may also affect the segregation between melanogenic and neurogenic cells in embryos. It is widely accepted that, irrespective of the model organism studied, the immediate precursor of both melanoblast and neurogenic populations is a glial-melanogenic bipotent progenitor. Upon exposure to different conditions, melanoblasts may differentiate into other neural crest-derived lineages such as neuronal cells and vice versa. Key factors that regulate melanoblast migration and patterning will regulate melanocyte homeostasis during different stages of hair cycling in postnatal hair follicles.
Collapse
Affiliation(s)
- Niels Vandamme
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- DAMBI, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Geert Berx
- Molecular and Cellular Oncology Laboratory, Department of Biomedical Molecular Biology, Ghent University, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
7
|
Bessonnard S, Vandormael-Pournin S, Coqueran S, Cohen-Tannoudji M, Artus J. PDGF Signaling in Primitive Endoderm Cell Survival Is Mediated by PI3K-mTOR Through p53-Independent Mechanism. Stem Cells 2019; 37:888-898. [PMID: 30913328 DOI: 10.1002/stem.3008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/18/2019] [Accepted: 03/12/2019] [Indexed: 12/22/2022]
Abstract
Receptor tyrosine kinase signaling pathways are key regulators for the formation of the primitive endoderm (PrE) and the epiblast (Epi) from the inner cell mass (ICM) of the mouse preimplantation embryo. Among them, FGF signaling is critical for PrE cell specification, whereas PDGF signaling is critical for the survival of committed PrE cells. Here, we investigated possible functional redundancies among FGF, PDGF, and KIT signaling and showed that only PDGF signaling is involved in PrE cell survival. In addition, we analyzed the effectors downstream of PDGFRα. Our results suggest that the role of PDGF signaling in PrE cell survival is mediated through PI3K-mTOR and independently from p53. Lastly, we uncovered a role for PI3K-mTOR signaling in the survival of Epi cells. Taken together, we propose that survival of ICM cell lineages relies on the regulation of PI3K-mTOR signaling through the regulation of multiple signaling pathways. Stem Cells 2019;37:888-898.
Collapse
Affiliation(s)
- Sylvain Bessonnard
- Early Mammalian Development and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Paris, France
| | | | - Sabrina Coqueran
- Early Mammalian Development and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Paris, France
| | - Michel Cohen-Tannoudji
- Early Mammalian Development and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Paris, France
| | - Jérôme Artus
- Early Mammalian Development and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Paris, France
| |
Collapse
|
8
|
Zhou L, Cai M, Ren Y, Wu H, Liu M, Chen H, Shang J. The different roles of 5-HT1A/2A receptors in fluoxetine ameliorated pigmentation of C57BL/6 mouse skin in response to stress. J Dermatol Sci 2018; 92:222-229. [PMID: 30527375 DOI: 10.1016/j.jdermsci.2018.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 10/14/2018] [Accepted: 10/15/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND 5-HT1A receptor was participated in fluoxetine induced melanogenesis in melanocytes and in normal C57BL/6 mice, but we know little about whether other 5-HT receptors are involved in regulation of fluoxetine promotes pigmentation. OBJECTIVE To investigate the role of 5-HT receptors in regulation of fluoxetine ameliorates chronic unpredictable mild stress (CUMS) and chronic restraint stress (CRS) induce hypopigmentation in C57BL/6 mice. METHODS CUMS and CRS were used to induce depigmentation in mice and evaluate the effect of fluoxetine. Western blot, immunohistochemistry and Q-PCR assay were used to determine the levels of protein and mRNA. Masson Fontana staining was used for melanin staining and FITC-Phalloidin staining was used to detect the expression of F-actin. Zebrafish and B16F10 cells were used for the mechanism research. RESULTS Fluoxetine (2.6 mg/kg, ig) ameliorated hypopigmentation induced by CUMS and CRS in mice, significantly increased the mRNA and protein levels of 5-HT1 A and 5-HT2 A receptors in mice and B16F10 cells. The effect of fluoxetine on melanogenesis in B16F10 cells and zebrafish were inhibited by WAY100635 (a selective 5-HT1 A receptor antagonist) and ketanserin (a 5-HT2 A receptor antagonist), respectively. Activation of p38 MAPK signaling pathways was contributed to fluoxetine induced melanogenesis and inhibited by WAY100635, but not ketanserin. However, ketanserin selectively weakened the action of fluoxetine promoted migration and up-regulated Rab27a protein expression in B16F10 cells. CONCLUSIONS 5-HT1 A and 2 A receptors contribute to melanogenesis and migration property of fluoxetine. The newly revealed mechanism indicates that fluoxetine and its analogues may be a potential drug for treatment of depigmentation disorders.
Collapse
Affiliation(s)
- Liangliang Zhou
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Minxuan Cai
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yingying Ren
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Huali Wu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Meng Liu
- Cancer Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, PR China
| | - Haijuan Chen
- Qinghai Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, University of the Chinese Academy of Sciences, Xining 810008, PR China
| | - Jing Shang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
9
|
Fane ME, Chhabra Y, Smith AG, Sturm RA. BRN2, a POUerful driver of melanoma phenotype switching and metastasis. Pigment Cell Melanoma Res 2018; 32:9-24. [PMID: 29781575 DOI: 10.1111/pcmr.12710] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/18/2018] [Accepted: 04/25/2018] [Indexed: 12/30/2022]
Abstract
The POU domain family of transcription factors play a central role in embryogenesis and are highly expressed in neural crest cells and the developing brain. BRN2 is a class III POU domain protein that is a key mediator of neuroendocrine and melanocytic development and differentiation. While BRN2 is a central regulator in numerous developmental programs, it has also emerged as a major player in the biology of tumourigenesis. In melanoma, BRN2 has been implicated as one of the master regulators of the acquisition of invasive behaviour within the phenotype switching model of progression. As a mediator of melanoma cell phenotype switching, it coordinates the transition to a dedifferentiated, slow cycling and highly motile cell type. Its inverse expression relationship with MITF is believed to mediate tumour progression and metastasis within this model. Recent evidence has now outlined a potential epigenetic switching mechanism in melanoma cells driven by BRN2 expression that induces melanoma cell invasion. We summarize the role of BRN2 in tumour cell dissemination and metastasis in melanoma, while also examining it as a potential metastatic regulator in other tumour models.
Collapse
Affiliation(s)
- Mitchell E Fane
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia.,Dermatology Research Centre, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Yash Chhabra
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia.,Dermatology Research Centre, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Aaron G Smith
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Richard A Sturm
- Dermatology Research Centre, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
10
|
van Geel N, Bosma S, Boone B, Speeckaert R. Classification of segmental vitiligo on the trunk. Br J Dermatol 2014; 170:322-7. [PMID: 24117138 DOI: 10.1111/bjd.12652] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2013] [Indexed: 01/04/2023]
Abstract
BACKGROUND Segmental vitiligo is characterized by a unilateral and localized distribution. A classification for segmental vitiligo on the face has been proposed, while this is still unavailable for lesions on the trunk. OBJECTIVES To design a classification for segmental vitiligo lesions on the trunk by evaluating their distribution pattern. METHODS In this retrospective observational study, the distribution pattern of 106 segmental vitiligo lesions on the trunk was analysed and classified into recurring subtypes. RESULTS Segmental vitiligo was more frequently observed on the ventral side (85·8%) than the lateral side (52·8%) or the back (36·8%) of the trunk. Based on recurring similarities in distribution patterns, lesions were categorized into six distinct subtypes: types 1, 2 and 3 involved the upper part of the trunk, type 4 and 5 the middle part and type 6 the lower part of the trunk. In total, 67·9% of all segmental vitiligo lesions fitted into these subtypes. The most frequent type of lesion was subtype 3 (22·6%), which showed a characteristic V-shaped pattern on the upper trunk, followed by subtype 5 (17·9%), with a band-like pattern on the lateral side, and subtype 6 (13·2%), with a rectangular depigmentation on the lower abdomen. CONCLUSIONS Based on our observations we suggest a new classification into six subtypes for segmental vitiligo lesions on the trunk, allowing categorization of the majority of segmental vitiligo patterns on the trunk. This may have important prognostic and diagnostic significance in early developing lesions.
Collapse
Affiliation(s)
- N van Geel
- Department of Dermatology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | | | | | | |
Collapse
|
11
|
Goldstein RS. Transplantation of mammalian embryonic stem cells and their derivatives to avian embryos. Stem Cell Rev Rep 2010; 6:473-83. [PMID: 20533000 DOI: 10.1007/s12015-010-9161-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Xenografting of normal and transformed mammalian tissues and cells to chick embryos has been performed for almost 100 years. Embryonic stem cells, derived more than 25 years ago from murine, and more than 10 years ago from human blastocysts, have transformed many fields of biological research. There is a growing body of studies combining these two widely-used experimental systems. This review surveys those reports in which murine or human embryonic stem cells, or differentiated derivatives of these pluripotent stem cells, were transplanted to embryonated chick eggs. Many of these studies have utilized the unique characteristics of both experimental models to obtain answers to developmental questions that are difficult or impossible to approach with xenografting to adult rodents or tissue culture-only techniques.
Collapse
Affiliation(s)
- Ronald S Goldstein
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel.
| |
Collapse
|
12
|
Abstract
Animal models play a crucial role in fundamental and medical research. Progress in the fields of drug discovery, regenerative medicine and cancer research among others are heavily dependent on in vivo models to validate in vitro observations, and develop new therapeutic approaches. However, conventional rodent and large animal experiments face ethical, practical and technical issues that limit their usage. The chick embryo represents an accessible and economical in vivo model, which has long been used in developmental biology, gene expression analysis and loss/gain of function experiments. It is also an established model for tissue/cell transplantation, and because of its lack of immune system in early development, the chick embryo is increasingly recognised as a model of choice for mammalian biology with new applications for stem cell and cancer research. Here, we review novel applications of the chick embryo model, and discuss future developments of this in vivo model for biomedical research.
Collapse
Affiliation(s)
- Hassan Rashidi
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), School of Clinical Sciences, The University of Nottingham, Nottingham, UK
| | | |
Collapse
|
13
|
Harris ML, Hall R, Erickson CA. Directing pathfinding along the dorsolateral path - the role of EDNRB2 and EphB2 in overcoming inhibition. Development 2008; 135:4113-22. [PMID: 19004859 DOI: 10.1242/dev.023119] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neural crest cells that become pigment cells migrate along a dorsolateral route between the ectoderm and the somite, whereas most other neural crest cells are inhibited from entering this space. This pathway choice has been attributed to unique, cell-autonomous migratory properties acquired by neural crest cells when they become specified as melanoblasts. By shRNA knockdown and overexpression experiments, we investigated the roles of three transmembrane receptors in regulating dorsolateral pathfinding in the chick trunk. We show that Endothelin receptor B2 (EDNRB2) and EphB2 are both determinants in this process, and that, unlike in other species, c-KIT is not. We demonstrate that the overexpression of EDNRB2 can maintain normal dorsolateral migration of melanoblasts in the absence of EphB2, and vice versa, suggesting that changes in receptor expression levels regulate the invasion of this pathway. Furthermore, by heterotopic grafting, we show that neural crest cell populations that do not rely on the activation of these receptors can migrate dorsolaterally only if this path is free of inhibitory molecules. We conclude that the requirement for EDNRB2 and EphB2 expression by melanoblasts is to support their migration by helping them to overcome repulsive or non-permissive cues in the dorsolateral environment.
Collapse
Affiliation(s)
- Melissa L Harris
- University of California, Davis, Department of Molecular and Cellular Biology, One Shields Avenue, Davis, CA 95616, USA
| | | | | |
Collapse
|
14
|
Silver DL, Hou L, Pavan WJ. The genetic regulation of pigment cell development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 589:155-69. [PMID: 17076280 DOI: 10.1007/978-0-387-46954-6_9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pigment cells in developing vertebrates are derived from a transient and pluripotent population of cells called neural crest. The neural crest delaminates from the developing neural tube and overlying ectoderm early in development. The pigment cells are the only derivative to migrate along the dorso-lateral pathway. As they migrate, the precursor pigment cell population differentiates and expands through proliferation and pro-survival processes, ultimately contributing to the coloration of organisms. The types of pigment cells that develop, timing of these processes, and final destination can vary between organisms. Studies from mice, chick, Xenopus, zebrafish, and medaka have led to the identification of many genes that regulate pigment cell development. These include several classes of proteins: transcription factors, transmembrane receptors, and extracellular ligands. This chapter discusses an overview of pigment cell development and the genes that regulate this important process.
Collapse
Affiliation(s)
- Debra L Silver
- Genetic Diseases Branch, NHGRI, NIH, Room 4A51, Bldg. 49, 49 Convent Drive, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
15
|
Pla P, Alberti C, Solov'eva O, Pasdar M, Kunisada T, Larue L. Ednrb2 orients cell migration towards the dorsolateral neural crest pathway and promotes melanocyte differentiation. ACTA ACUST UNITED AC 2005; 18:181-7. [PMID: 15892714 DOI: 10.1111/j.1600-0749.2005.00230.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Endothelin receptors B (Ednrb) are involved in the development of the enteric and melanocytic lineages, which originate from neural crest cells (NCCs). In mice, trunk NCCs and their derivatives express only one Ednrb. In quail, trunk NCCs express two Ednrb: Ednrb and Ednrb2. Quail Ednrb is expressed in NCCs migrating along the ventral pathway, which gives rise to the peripheral nervous system, including enteric ganglia. Ednrb2 is upregulated in NCCs before these cells enter the dorsolateral pathway. The NCCs migrating along the dorsolateral pathway are melanocyte precursors. We analyzed the in vitro differentiation and in ovo migration of mouse embryonic stem (ES) cells expressing and not expressing Ednrb2. We generated a series of transfected ES cell lines expressing Ednrb2. This receptor, like Ednrb, oriented genuine ES cells towards melanocyte lineage differentiation in vitro. The in ovo migration of Ednrb2-expressing ES cells was massively oriented towards the dorsolateral pathway, unlike that of WT or Ednrb-expressing ES cells. Thus, Ednrb2 is involved in melanoblast differentiation and migration.
Collapse
Affiliation(s)
- Patrick Pla
- Developmental Genetics of Melanocytes, UMR 146 CNRS-Institut Curie, Orsay, France
| | | | | | | | | | | |
Collapse
|
16
|
Slominski A, Tobin DJ, Shibahara S, Wortsman J. Melanin pigmentation in mammalian skin and its hormonal regulation. Physiol Rev 2004; 84:1155-228. [PMID: 15383650 DOI: 10.1152/physrev.00044.2003] [Citation(s) in RCA: 1414] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cutaneous melanin pigment plays a critical role in camouflage, mimicry, social communication, and protection against harmful effects of solar radiation. Melanogenesis is under complex regulatory control by multiple agents interacting via pathways activated by receptor-dependent and -independent mechanisms, in hormonal, auto-, para-, or intracrine fashion. Because of the multidirectional nature and heterogeneous character of the melanogenesis modifying agents, its controlling factors are not organized into simple linear sequences, but they interphase instead in a multidimensional network, with extensive functional overlapping with connections arranged both in series and in parallel. The most important positive regulator of melanogenesis is the MC1 receptor with its ligands melanocortins and ACTH, whereas among the negative regulators agouti protein stands out, determining intensity of melanogenesis and also the type of melanin synthesized. Within the context of the skin as a stress organ, melanogenic activity serves as a unique molecular sensor and transducer of noxious signals and as regulator of local homeostasis. In keeping with these multiple roles, melanogenesis is controlled by a highly structured system, active since early embryogenesis and capable of superselective functional regulation that may reach down to the cellular level represented by single melanocytes. Indeed, the significance of melanogenesis extends beyond the mere assignment of a color trait.
Collapse
Affiliation(s)
- Andrzej Slominski
- Dept. of Pathology, Suite 599, University of Tennessee Health Science Center, 930 Madison Avenue, Memphis, TN 38163, USA.
| | | | | | | |
Collapse
|
17
|
Franceschini I, Vitry S, Padilla F, Casanova P, Tham TN, Fukuda M, Rougon G, Durbec P, Dubois-Dalcq M. Migrating and myelinating potential of neural precursors engineered to overexpress PSA-NCAM. Mol Cell Neurosci 2004; 27:151-62. [PMID: 15485771 DOI: 10.1016/j.mcn.2004.05.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Revised: 05/19/2004] [Accepted: 05/25/2004] [Indexed: 11/17/2022] Open
Abstract
Polysialic acid (PSA) on NCAM is an important modulator of cell-cell interactions during development and regeneration. Here we investigated whether PSA overexpression influences neural cell migration and myelination. We stably expressed a GFP-tagged polysialytransferase, PSTGFP, in mouse neurospheres and induced prolonged PSA synthesis. Using a chick xenograft assay for migration, we show that PSA can instruct precursor migration along the ventral pathway. PSA persistence did not change neural precursor multipotentiality in vitro but induced a delay in oligodendrocyte differentiation. PSTGFP+ precursors showed widespread engraftment in shiverer brain, closely similar to that observed with control precursors expressing a fluorescent protein. Initially, myelination by oligodendrocytes was delayed but, eventually, down-regulation of PSTGFP occurred, allowing myelination to proceed. Thus down-regulation of polysialyltransferases takes place even in cells where its RNA is under the control of a heterologous promoter and engineering PSA overexpression in neural precursors does not cause irreversible unphysiological effects.
Collapse
Affiliation(s)
- Isabelle Franceschini
- Unité de Neurovirologie et Régénération du Système Nerveux, Institut Pasteur, 75724 Paris cedex 15, France
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Pla P, Solov'eva O, Moore R, Alberti C, Kunisada T, Larue L. Dct::lacZ ES Cells: A Novel Cellular Model to Study Melanocyte Determination and Differentiation. ACTA ACUST UNITED AC 2004; 17:142-9. [PMID: 15016303 DOI: 10.1046/j.1600-0749.2003.00121.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Embryonic stem (ES) cells differentiate into various cell lineages in vitro. A procedure was previously designed to promote the differentiation of ES cells towards the melanocyte lineage and to obtain large and reproducible amounts of melanocytes. To elucidate the main events that lead to the development of melanocytes in vitro, we used transgenic Dct::lacZ mouse blastocysts to establish ES cell lines expressing the lacZ reporter gene under the control of the Dct promoter. Dct, a melanoblast marker, is expressed just after melanoblast determination in vivo. We evaluated the importance of recruitment, proliferation and differentiation during melanocyte ontogeny after the in vitro differentiation of Dct::lacZ ES cells into melanocytes. We showed that bFGF and cholera toxin induce precocious melanoblast determination, associated with early melanocyte differentiation. Edn3 induced melanoblast proliferation and long-term melanoblast recruitment, but not precocious determination. The lack of basic Fibroblast Growth Factor (bFGF) and cholera toxin can be partially compensated by Edn3. Thus, Dct::lacZ ES cells can be used as a model to study determination, proliferation and differentiation in the melanocyte lineage in vitro.
Collapse
Affiliation(s)
- Patrick Pla
- Developmental Genetics of Melanocytes, UMR 146 CNRS-Institut Curie, Bat. 110, 91405, Orsay Cedex, France
| | | | | | | | | | | |
Collapse
|
19
|
Moore R, Larue L. Cell surface molecules and truncal neural crest ontogeny: A perspective. ACTA ACUST UNITED AC 2004; 72:140-50. [PMID: 15269888 DOI: 10.1002/bdrc.20014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The neural crest cell is synonymous with vertebrates and can be viewed as a transitory, mobile vector that conveys neuroepithelial stem cells to a diverse number of remote locations in the embryo. Neural crest cells have been studied intensively over the past 30 years, and it is increasingly apparent that their fate is, at least in part, directed extrinsically by the environment to which they are exposed in vivo. The interface between the cell surface and the opposing environment is clearly an important compartment for the correct deployment of the neural crest. Here, we review some of the molecules present in this location and how they influence the fate of the neural crest and generate disease.
Collapse
Affiliation(s)
- Robert Moore
- Human Genetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA.
| | | |
Collapse
|
20
|
Goldstein RS, Drukker M, Reubinoff BE, Benvenisty N. Integration and differentiation of human embryonic stem cells transplanted to the chick embryo. Dev Dyn 2002; 225:80-6. [PMID: 12203723 DOI: 10.1002/dvdy.10108] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Human embryonic stem (ES) cells are pluripotent cells that can differentiate into a large array of cell types and, thus, hold promise for advancing our understanding of human embryology and for contributing to transplantation medicine. In this study, differentiation of human ES cells was examined in vivo by in ovo transplantation to organogenesis-stage embryos. Colonies of human ES cells were grafted into or in place of epithelial-stage somites of chick embryos of 1.5 to 2 days of development. The grafted human ES cells survived in the chick host and were identified by vital staining with carboxyfluorescein diacetate or use of a green fluorescent protein-expressing cells. Histologic analysis showed that human ES cells are easily distinguished from host cells by their larger, more intensely staining nuclei. Some grafted cells differentiated en masse into epithelia, whereas others migrated and mingled with host tissues, including the dorsal root ganglion. Colonies grafted directly adjacent to the host neural tube produced primarily structures with the morphology and molecular characteristics of neural rosettes. These structures contain differentiated neurons as shown by beta-3-tubulin and neurofilament expression in axons and cell bodies. Axons derived from the grafted cells penetrate the host nervous system, and host axons enter the structures derived from the graft. Our results show that human ES cells transplanted in ovo survive, divide, differentiate, and integrate with host tissues and that the host embryonic environment may modulate their differentiation. The chick embryo, therefore, may serve as an accessible and unique experimental system for the study of in vivo development of human ES cells.
Collapse
Affiliation(s)
- Ronald S Goldstein
- Gonda Research Center, Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel.
| | | | | | | |
Collapse
|
21
|
Peters EMJ, Tobin DJ, Botchkareva N, Maurer M, Paus R. Migration of melanoblasts into the developing murine hair follicle is accompanied by transient c-Kit expression. J Histochem Cytochem 2002; 50:751-66. [PMID: 12019292 DOI: 10.1177/002215540205000602] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Disruption of the c-Kit/stem cell factor (SCF) signaling pathway interferes with the survival, migration, and differentiation of melanocytes during generation of the hair follicle pigmentary unit. We examined c-Kit, SCF, and S100 (a marker for precursor melanocytic cells) expression, as well as melanoblast/melanocyte ultrastructure, in perinatal C57BL/6 mouse skin. Before the onset of hair bulb melanogenesis (i.e., stages 0-4 of hair follicle morphogenesis), strong c-Kit immunoreactivity (IR) was seen in selected non-melanogenic cells in the developing hair placode and hair plug. Many of these cells were S100-IR and were ultrastructurally identified as melanoblasts with migratory appearance. During the subsequent stages (5 and 6), increasingly dendritic c-Kit-IR cells successively invaded the hair bulb, while S100-IR gradually disappeared from these cells. Towards the completion of hair follicle morphogenesis (stages 7 and 8), several distinct follicular melanocytic cell populations could be defined and consisted broadly of (a) undifferentiated, non-pigmented c-Kit-negative melanoblasts in the outer root sheath and bulge and (b) highly differentiated melanocytes adjacent to the hair follicle dermal papilla above Auber's line. Widespread epithelial SCF-IR was seen throughout hair follicle morphogenesis. These findings suggest that melanoblasts express c-Kit as a prerequisite for migration into the SCF-supplying hair follicle epithelium. In addition, differentiated c-Kit-IR melanocytes target the bulb, while non-c-Kit-IR melanoblasts invade the outer root sheath and bulge in fully developed hair follicles.
Collapse
Affiliation(s)
- Eva M J Peters
- Department of Dermatology, University Hospital Eppendorf, University of Hamburg, Germany
| | | | | | | | | |
Collapse
|
22
|
Mansouri A, Pla P, Larue L, Gruss P. Pax3acts cell autonomously in the neural tube and somites by controlling cell surface properties. Development 2001; 128:1995-2005. [PMID: 11493522 DOI: 10.1242/dev.128.11.1995] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Pax3 is a member of the paired-box-containing transcription factors. It is expressed in the developing somites, dorsal spinal cord, mesencephalon and neural crest derivatives. Several loss-of-function mutations are correlated with the Splotch phenotype in mice and Waardenburg syndrome in humans. Malformations include a lack of muscle in the limb, a failure of neural tube closure and dysgenesis of numerous neural crest derivatives. In this study we have used embryonic stem (ES) cells to generate a lacZ knock-in into the Pax3 locus. The Pax3 knock-in Splotch allele (Sp2G) was used to generate Pax3-deficient ES cells in order to investigate whether, in chimeric embryos, Pax3 is acting cell autonomously in the somites and the neural tube. We found that while Pax3 function is essential for the neuroepithelium and somites, a wild-type environment rescues mutant neural crest cells. In the two affected embryonic tissues, mutant and wild-type cells undergo segregation and do not intermingle.The contribution of mutant cells to the neural tube and the somites displayed temporal differences. All chimeric embryos showed a remarkable contribution of blue cells to the neural tube at all stages analyzed, indicating that the Pax3-deficient cells are not excluded from the neural epithelium while development proceeds. In contrast, this is not true for the paraxial mesoderm. The somite contribution of Pax3−/− ES cells becomes less frequent in older embryos as compared to controls with Pax3+/− ES cells. We propose that although Pax3 function is related to cell surface properties, its role may differ in various tissues. In fact, apoptosis was found in Pax3-deficient cells of the lateral dermomyotome but not in the neural tube.
Collapse
Affiliation(s)
- A Mansouri
- Max-Planck Institute for Biophysical Chemistry, Department of Molecular Cell Biology, Am Fassberg 11, D-37077 Göttingen, Germany.
| | | | | | | |
Collapse
|
23
|
Durbec P, Rougon G. Transplantation of mammalian olfactory progenitors into chick hosts reveals migration and differentiation potentials dependent on cell commitment. Mol Cell Neurosci 2001; 17:561-76. [PMID: 11273650 DOI: 10.1006/mcne.2000.0951] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In vertebrates, interneurons of the olfactory bulb are continuously generated postnatally and throughout life at the subventricular zone of the forebrain. From there, the neuronal progenitors migrate tangentially in a typical chain-like structure to the olfactory bulb in which they differentiate as interneurons. We have used a mouse/chick xenograft strategy to explore the migration and differentiation potential of the mouse olfactory progenitors in a heterochronic and heterotypic environment. We compared the migration of primary cells derived from the subventricular zone of adult or newborn lateral ventricule with the behavior of in vitro amplified cells derived from the same structures. We show that in the chick environment, olfactory bulb progenitors from newborn brain tissue perform chain migration along the neural crest cell routes, whereas grafted neurosphere-derived-cells migrate as isolated cells. These results, together with in vitro observations, allow us to propose that neuronal chain migration is a community effect independent of environmental cues but which is closely regulated by the differentiation program of the cells. We established that the progenitor cells performing chain migration are already committed, while neurosphere-derived-cells are able to integrate and differentiate as components of the peripheral nervous system.
Collapse
Affiliation(s)
- P Durbec
- Laboratoire de Génétique et Physiologie du Développement, IBDM, CNRS/INSERM/Université de la Méditérranée/AP de Marseille, Parc Scientifique de Luminy, Marseille Cedex 9, 13288, France
| | | |
Collapse
|