87 results on '"Heather C. Etchevers"'
Search Results
2. Sustained experimental activation of FGF8/ERK in the developing chicken spinal cord models early events in ERK-mediated tumorigenesis
- Author
-
Axelle Wilmerding, Lauranne Bouteille, Nathalie Caruso, Ghislain Bidaut, Heather C. Etchevers, Yacine Graba, and Marie-Claire Delfini
- Subjects
MAPK/ERK pathway ,MEK1 (MAP2K1) ,Developing spinal cord ,Neural crest cells ,Chicken embryo ,Cancer model ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The MAPK/ERK pathway regulates a variety of physiological cellular functions, including cell proliferation and survival. It is abnormally activated in many types of human cancers in response to driver mutations in regulators of this pathway that trigger tumor initiation. The early steps of oncogenic progression downstream of ERK overactivation are poorly understood due to a lack of appropriate models. We show here that ERK1/2 overactivation in the trunk neural tube of the chicken embryo through expression of a constitutively active form of the upstream kinase MEK1 (MEK1ca), rapidly provokes a profound change in the transcriptional signature of developing spinal cord cells. These changes are concordant with a previously established role of the tyrosine kinase receptor ligand FGF8 acting via the ERK1/2 effectors to maintain an undifferentiated state. Furthermore, we show that MEK1ca-transfected spinal cord cells lose neuronal identity, retain caudal markers, and ectopically express potential effector oncogenes, such as AQP1. MEK1ca expression in the developing spinal cord from the chicken embryo is thus a tractable in vivo model to identify the mechanisms fostering neoplasia and malignancy in ERK-induced tumorigenesis of neural origins.
- Published
- 2022
- Full Text
- View/download PDF
3. Multiple congenital malformations arise from somatic mosaicism for constitutively active Pik3ca signaling
- Author
-
Elise Marechal, Anne Poliard, Kilian Henry, Mathias Moreno, Mathilde Legrix, Nicolas Macagno, Grégoire Mondielli, Teddy Fauquier, Anne Barlier, and Heather C. Etchevers
- Subjects
neural crest ,embryo ,PI3K ,cancer ,birth defect ,cleft palate ,Biology (General) ,QH301-705.5 - Abstract
Recurrent missense mutations of the PIK3CA oncogene are among the most frequent drivers of human cancers. These often lead to constitutive activation of its product p110α, a phosphatidylinositol 3-kinase (PI3K) catalytic subunit. In addition to causing a broad range of cancers, the H1047R mutation is also found in affected tissues of a distinct set of congenital tumors and malformations. Collectively termed PIK3CA-related disorders (PRDs), these lead to overgrowth of brain, adipose, connective and musculoskeletal tissues and/or blood and lymphatic vessel components. Vascular malformations are frequently observed in PRD, due to cell-autonomous activation of PI3K signaling within endothelial cells. These, like most muscle, connective tissue and bone, are derived from the embryonic mesoderm. However, important organ systems affected in PRDs are neuroectodermal derivatives. To further examine their development, we drove the most common post-zygotic activating mutation of Pik3ca in neural crest and related embryonic lineages. Outcomes included macrocephaly, cleft secondary palate and more subtle skull anomalies. Surprisingly, Pik3ca-mutant subpopulations of neural crest origin were also associated with widespread cephalic vascular anomalies. Mesectodermal neural crest is a major source of non-endothelial connective tissue in the head, but not the body. To examine the response of vascular connective tissues of the body to constitutive Pik3ca activity during development, we expressed the mutation by way of an Egr2 (Krox20) Cre driver. Lineage tracing led us to observe new lineages that had normally once expressed Krox20 and that may be co-opted in pathogenesis, including vascular pericytes and perimysial fibroblasts. Finally, Schwann cell precursors having transcribed either Krox20 or Sox10 and induced to express constitutively active PI3K were associated with vascular and other tumors. These murine phenotypes may aid discovery of new candidate human PRDs affecting craniofacial and vascular smooth muscle development as well as the reciprocal paracrine signaling mechanisms leading to tissue overgrowth.
- Published
- 2022
- Full Text
- View/download PDF
4. Cutaneous Melanomas Arising during Childhood: An Overview of the Main Entities
- Author
-
Arnaud de la Fouchardière, Felix Boivin, Heather C. Etchevers, and Nicolas Macagno
- Subjects
congenital nevus ,melanoma ,childhood ,skin ,oncogenetics ,SSM ,Dermatology ,RL1-803 - Abstract
Cutaneous melanomas are exceptional in children and represent a variety of clinical situations, each with a different prognosis. In congenital nevi, the risk of transformation is correlated with the size of the nevus. The most frequent type is lateral transformation, extremely rare before puberty, reminiscent of a superficial spreading melanoma (SSM) ex-nevus. Deep nodular transformation is much rarer, can occur before puberty, and must be distinguished from benign proliferative nodules. Superficial spreading melanoma can also arise within small nevi, which were not visible at birth, usually after puberty, and can reveal a cancer predisposition syndrome (CDKN2A or CDK4 germline mutations). Prognosis is correlated with classical histoprognostic features (mainly Breslow thickness). Spitz tumors are frequent in adolescents and encompass benign (Spitz nevus), intermediate (atypical Spitz tumor), and malignant forms (malignant Spitz tumor). The whole spectrum is characterized by specific morphology with spindled and epithelioid cells, genetic features, and an overall favorable outcome even if a regional lymph node is involved. Nevoid melanomas are rare and difficult to diagnose clinically and histologically. They can arise in late adolescence. Their prognosis is currently not very well ascertained. A small group of melanomas remains unclassified after histological and molecular assessment.
- Published
- 2021
- Full Text
- View/download PDF
5. Outflow Tract Formation—Embryonic Origins of Conotruncal Congenital Heart Disease
- Author
-
Sonia Stefanovic, Heather C. Etchevers, and Stéphane Zaffran
- Subjects
congenital heart defects ,cardiac progenitor cells ,outflow tract ,second heart field ,neural crest ,endocardium ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Anomalies in the cardiac outflow tract (OFT) are among the most frequent congenital heart defects (CHDs). During embryogenesis, the cardiac OFT is a dynamic structure at the arterial pole of the heart. Heart tube elongation occurs by addition of cells from pharyngeal, splanchnic mesoderm to both ends. These progenitor cells, termed the second heart field (SHF), were first identified twenty years ago as essential to the growth of the forming heart tube and major contributors to the OFT. Perturbation of SHF development results in common forms of CHDs, including anomalies of the great arteries. OFT development also depends on paracrine interactions between multiple cell types, including myocardial, endocardial and neural crest lineages. In this publication, dedicated to Professor Andriana Gittenberger-De Groot and her contributions to the field of cardiac development and CHDs, we review some of her pioneering studies of OFT development with particular interest in the diverse origins of the many cell types that contribute to the OFT. We also discuss the clinical implications of selected key findings for our understanding of the etiology of CHDs and particularly OFT malformations.
- Published
- 2021
- Full Text
- View/download PDF
6. Supplementary Materials and Methods from Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma
- Author
-
Francis Mussai, Carmela De Santo, Michelle Haber, Susan A. Burchill, Ronny Schmidt, Louis Chesler, David S. Ziegler, Jayne Murray, Heather C. Etchevers, Murray D. Norris, Paul N. Cheng, Carmel M. McConville, Ashley Vardon, Samantha Brownhill, Sarah Booth, Georgina L. Eden, Orli Yogev, Fenna De Bie, Sharon A. Egan, Andrea M. Berry, Luciana Gneo, Laura D. Gamble, and Livingstone Fultang
- Abstract
Supplementary Materials and Methods CLEAN version
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figure 3 from Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma
- Author
-
Francis Mussai, Carmela De Santo, Michelle Haber, Susan A. Burchill, Ronny Schmidt, Louis Chesler, David S. Ziegler, Jayne Murray, Heather C. Etchevers, Murray D. Norris, Paul N. Cheng, Carmel M. McConville, Ashley Vardon, Samantha Brownhill, Sarah Booth, Georgina L. Eden, Orli Yogev, Fenna De Bie, Sharon A. Egan, Andrea M. Berry, Luciana Gneo, Laura D. Gamble, and Livingstone Fultang
- Abstract
Supplementary Figure 3 Neuroblastoma conditioning upregulates IL-1ï�¢ï€ and TNF-ï�¡ï€ expression in macrophages
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure Legends and Table from Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma
- Author
-
Francis Mussai, Carmela De Santo, Michelle Haber, Susan A. Burchill, Ronny Schmidt, Louis Chesler, David S. Ziegler, Jayne Murray, Heather C. Etchevers, Murray D. Norris, Paul N. Cheng, Carmel M. McConville, Ashley Vardon, Samantha Brownhill, Sarah Booth, Georgina L. Eden, Orli Yogev, Fenna De Bie, Sharon A. Egan, Andrea M. Berry, Luciana Gneo, Laura D. Gamble, and Livingstone Fultang
- Abstract
Supplementary Figure Legends and Tables
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 4 from Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma
- Author
-
Francis Mussai, Carmela De Santo, Michelle Haber, Susan A. Burchill, Ronny Schmidt, Louis Chesler, David S. Ziegler, Jayne Murray, Heather C. Etchevers, Murray D. Norris, Paul N. Cheng, Carmel M. McConville, Ashley Vardon, Samantha Brownhill, Sarah Booth, Georgina L. Eden, Orli Yogev, Fenna De Bie, Sharon A. Egan, Andrea M. Berry, Luciana Gneo, Laura D. Gamble, and Livingstone Fultang
- Abstract
Supplementary Figure 4 Macrophage infiltration and downstream effects on neuroblastoma tumours
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma
- Author
-
Francis Mussai, Carmela De Santo, Michelle Haber, Susan A. Burchill, Ronny Schmidt, Louis Chesler, David S. Ziegler, Jayne Murray, Heather C. Etchevers, Murray D. Norris, Paul N. Cheng, Carmel M. McConville, Ashley Vardon, Samantha Brownhill, Sarah Booth, Georgina L. Eden, Orli Yogev, Fenna De Bie, Sharon A. Egan, Andrea M. Berry, Luciana Gneo, Laura D. Gamble, and Livingstone Fultang
- Abstract
Neuroblastoma is the most common childhood solid tumor, yet the prognosis for high-risk disease remains poor. We demonstrate here that arginase 2 (ARG2) drives neuroblastoma cell proliferation via regulation of arginine metabolism. Targeting arginine metabolism, either by blocking cationic amino acid transporter 1 (CAT-1)–dependent arginine uptake in vitro or therapeutic depletion of arginine by pegylated recombinant arginase BCT-100, significantly delayed tumor development and prolonged murine survival. Tumor cells polarized infiltrating monocytes to an M1-macrophage phenotype, which released IL1β and TNFα in a RAC-alpha serine/threonine-protein kinase (AKT)–dependent manner. IL1β and TNFα established a feedback loop to upregulate ARG2 expression via p38 and extracellular regulated kinases 1/2 (ERK1/2) signaling in neuroblastoma and neural crest–derived cells. Proteomic analysis revealed that enrichment of IL1β and TNFα in stage IV human tumor microenvironments was associated with a worse prognosis. These data thus describe an immune-metabolic regulatory loop between tumor cells and infiltrating myeloid cells regulating ARG2, which can be clinically exploited.Significance:These findings illustrate that cross-talk between myeloid cells and tumor cells creates a metabolic regulatory loop that promotes neuroblastoma progression.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 2 from Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma
- Author
-
Francis Mussai, Carmela De Santo, Michelle Haber, Susan A. Burchill, Ronny Schmidt, Louis Chesler, David S. Ziegler, Jayne Murray, Heather C. Etchevers, Murray D. Norris, Paul N. Cheng, Carmel M. McConville, Ashley Vardon, Samantha Brownhill, Sarah Booth, Georgina L. Eden, Orli Yogev, Fenna De Bie, Sharon A. Egan, Andrea M. Berry, Luciana Gneo, Laura D. Gamble, and Livingstone Fultang
- Abstract
Supplementary Figure 2 Cytokine profile of tumour induced macrophages or granulocytes
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 1 from Macrophage-Derived IL1β and TNFα Regulate Arginine Metabolism in Neuroblastoma
- Author
-
Francis Mussai, Carmela De Santo, Michelle Haber, Susan A. Burchill, Ronny Schmidt, Louis Chesler, David S. Ziegler, Jayne Murray, Heather C. Etchevers, Murray D. Norris, Paul N. Cheng, Carmel M. McConville, Ashley Vardon, Samantha Brownhill, Sarah Booth, Georgina L. Eden, Orli Yogev, Fenna De Bie, Sharon A. Egan, Andrea M. Berry, Luciana Gneo, Laura D. Gamble, and Livingstone Fultang
- Abstract
Supplementary Figure 1: M1-Macrophage infiltrate neuroblastoma
- Published
- 2023
- Full Text
- View/download PDF
13. Domains and outcomes of the core outcome set of congenital melanocytic naevi for clinical practice and research (the OCOMEN project)
- Author
-
Ph.I. Spuls, A C Fledderus, A. Wolkerstorfer, Heather C. Etchevers, M S van Kessel, C. M. A. M. van der Horst, W Oei, Suzanne G.M.A. Pasmans, Dermatology, Plastic, Reconstructive and Hand Surgery, AMS - Musculoskeletal Health, AMS - Rehabilitation & Development, APH - Methodology, APH - Quality of Care, ACS - Diabetes & metabolism, ACS - Atherosclerosis & ischemic syndromes, University of Amsterdam [Amsterdam] (UvA), Erasmus University Medical Center [Rotterdam] (Erasmus MC), Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Naevus International Patient Representative Working Group Leader
- Subjects
medicine.medical_specialty ,Melanocytic naevi ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Nevus, Pigmented ,Consensus ,Skin Neoplasms ,Delphi Technique ,MEDLINE ,Dermatology ,Computer-assisted web interviewing ,Outcome (game theory) ,Clinical Practice ,Core (game theory) ,Treatment Outcome ,Research Design ,Family medicine ,Outcome Assessment, Health Care ,medicine ,Humans ,Set (psychology) ,Psychology ,Inclusion (education) ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
International audience; Background: Congenital melanocytic naevi (CMN) can have a great impact on patients' lives owing to perceived stigmatization, and the risk of melanoma development and neurological complications. Development of a core outcome set (COS) for care and research in CMN will allow standard reporting of outcomes. This will enable comparison of outcomes, allowing professionals to offer advice about the best management options. In previous research, stakeholders (patients, parents and professionals) reached consensus on the core domains of the COS. To select the appropriate measurement instruments, the domains should be specified by outcomes.Objectives: To reach consensus on the specific core outcomes describing the core domains pertaining to clinical care and research in CMN.Methods: A list of provisional outcomes (obtained earlier) was critically reviewed by the Outcomes for COngenital MElanocytic Naevi (OCOMEN) research team and by relevant stakeholders through an online questionnaire, to refine this list and provide clear definitions for every outcome. When needed, discussion with individual participants was undertaken over the telephone or by email. During an online consensus meeting, stakeholders discussed the inclusion of potential outcomes. After the meeting, participants voted in two rounds for the inclusion of outcomes.Results: Forty-four stakeholders from 19 countries participated. Nine core outcomes were included in the COS relative to clinical care and 10 core outcomes for research.Conclusions: These core outcomes will enable standard reporting in future care and research of CMN. This study facilitates the next step of COS development: selecting the appropriate measurement instruments for every outcome.
- Published
- 2021
- Full Text
- View/download PDF
14. Development of an international core domain set for medium, large and giant congenital melanocytic naevi as a first step towards a core outcome set for clinical practice and research
- Author
-
M S van Kessel, Sven Krengel, C. M. A. M. van der Horst, Heather C. Etchevers, W Oei, Albert Wolkerstorfer, A C Fledderus, Ph.I. Spuls, Suzanne G.M.A. Pasmans, I.J. Korfage, C.A.M. Eggen, Jan Kottner, Dermatology, APH - Methodology, APH - Quality of Care, Plastic, Reconstructive and Hand Surgery, ACS - Atherosclerosis & ischemic syndromes, AMS - Musculoskeletal Health, AMS - Rehabilitation & Development, Amsterdam Movement Sciences, ACS - Diabetes & metabolism, Erasmus University Medical Center [Rotterdam] (Erasmus MC), Charité - UniversitätsMedizin = Charité - University Hospital [Berlin], Naevus International Patient Representative Working Group Leader, Dermatological group practice, Lübeck, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Association du Naevus Géant Congénital, Naevus 2000 France-Europe and the Asociación Española de Nevus Gigante Congénito, Gall, Valérie, and Public Health
- Subjects
medicine.medical_specialty ,Consensus ,Delphi Technique ,Best practice ,MEDLINE ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Dermatology ,Outcome (game theory) ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Quality of life (healthcare) ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,melanoma ,Humans ,Medicine ,Patient Reported Outcome Measures ,Set (psychology) ,Nevus, Pigmented ,congenital nevi ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,business.industry ,outcome domains ,Core outcome set ,Focus group ,3. Good health ,Clinical trial ,Treatment Outcome ,clinical research ,Research Design ,Family medicine ,Quality of Life ,business ,Psychosocial ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Background: Medium, large and giant congenital melanocytic naevi (CMN) can impose a psychosocial burden on patients and families, and are associated with increased risk of developing melanoma or neurological symptoms. Lack of consensus on what outcomes to measure makes it difficult to advise patients and families about treatment and to set up best practice for CMN. Objectives: Fostering consensus among patient representatives and professionals, we aim to develop a core outcome set, defined as the minimum set of outcomes to measure and report in care and all clinical trials of a specific health condition. We focused on the ‘what to measure’ aspect, the so-called core domain set (CDS), following the COMET and CS-COUSIN guidelines. Methods: We conducted a systematic review to identify outcomes reported in the literature. Focus groups with patient representatives identified patient-reported outcomes. All these outcomes were classified into domains. Through e-Delphi surveys, 144 stakeholders from 27 countries iteratively rated the importance of domains and outcomes. An online consensus meeting attended by seven patient representatives and seven professionals finalized the CDS. Results: We reached consensus on six domains, four of which were applied to both care and research: ‘quality of life’, ‘neoplasms’, ‘nervous system’ and ‘anatomy of skin’. ‘Adverse events’ was specific to care and ‘pathology’ to research. Conclusions: We have developed a CDS for medium-to-giant CMN. Its application in reporting care and research of CMN will facilitate treatment comparisons. The next step will be to reach consensus on the specific outcomes for each of the domains and what instruments should be used to measure these domains and outcomes.
- Published
- 2021
- Full Text
- View/download PDF
15. Somatotroph Tumors and the Epigenetic Status of the GNAS Locus
- Author
-
Catherine Roche, Guillaume Assié, Justine Galluso, Grégory Mougel, Henry Dufour, Pauline Romanet, Thomas Cuny, Mirella Hage, Dominique Figarella-Branger, Daniel De Murat, Heather C. Etchevers, Marily Theodoropoulou, Peter Kamenicky, Anne Barlier, Thomas Graillon, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Institut National de la Santé et de la Recherche Médicale (INSERM)-Aix Marseille Université (AMU), Institut Marseille Maladies Rares (MarMaRa), Aix Marseille Université (AMU), Laboratoire de Biochimie et de Biologie Moléculaire [Hôpital de la Conception - APHM], Hôpital de la Conception [CHU - APHM] (LA CONCEPTION), Université Paris-Saclay, Physiologie et physiopathologie endocriniennes, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay, AP-HP Hôpital Bicêtre (Le Kremlin-Bicêtre), Ludwig Maximilian University [Munich] (LMU), Département de Neurochirurgie [CHU Timone], Hôpital de la Timone [CHU - APHM] (TIMONE), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Paris (UP), Institut de neurophysiopathologie (INP), Aix Marseille Université (AMU)-Centre National de la Recherche Scientifique (CNRS), Service d'endocrinologie, diabète, maladies métaboliques [Hôpital de la Conception - APHM], All phases of this study were supported by grants from the Institut National de lutte contrele Cancer (INCa), and the French Ministry of Health. The project resulting in this publication receivedfunding from the Groupement de Recherche CREST-NET (GDR2031) and Excellence Initiative of AixMarseille University—A*Midex—a French 'Investissement d’Avenir'—Institute MarMaRa AMX-19-IET- 007., Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Physiologie et physiopathologie endocriniennes (PHYSENDO), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), and Gall, Valérie
- Subjects
0301 basic medicine ,[SDV]Life Sciences [q-bio] ,gsp oncogene ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,medicine.disease_cause ,pituitary ,0302 clinical medicine ,Somatostatin receptor 2 ,Biology (General) ,Imprinting (psychology) ,Spectroscopy ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,General Medicine ,Methylation ,Computer Science Applications ,[SDV] Life Sciences [q-bio] ,Chemistry ,PitNET ,030220 oncology & carcinogenesis ,imprinting ,hormones, hormone substitutes, and hormone antagonists ,epigenetic ,musculoskeletal diseases ,endocrine system ,QH301-705.5 ,Biology ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,GNAS ,stomatognathic system ,relaxation ,[SDV.BBM] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,medicine ,GNAS complex locus ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Epigenetics ,Physical and Theoretical Chemistry ,Allele ,QD1-999 ,Molecular Biology ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,somatotroph ,Oncogene ,Organic Chemistry ,tumorigenesis ,030104 developmental biology ,nervous system ,Cancer research ,biology.protein ,Carcinogenesis ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Background: Forty percent of somatotroph tumors harbor recurrent activating GNAS mutations, historically called the gsp oncogene. In gsp-negative somatotroph tumors, GNAS expression itself is highly variable, those with GNAS overexpression most resemble phenotypically those carrying the gsp oncogene. GNAS is monoallelically expressed in the normal pituitary due to methylation-based imprinting. We hypothesize that changes in GNAS imprinting of gsp-negative tumors affect GNAS expression levels and tumorigenesis. Methods: We characterized the GNAS locus in two independent somatotroph tumor cohorts: one of 23 tumors previously published (PMID: 31883967) and classified by pan-genomic analysis, and a second with 82 tumors. Results: Multi-omics analysis of the first cohort identified a significant difference between gsp-negative and gsp-positive tumors in the methylation index at the known differentially methylated region (DMR) of the GNAS A/B transcript promoter, which was confirmed in the larger series of 82 tumors. GNAS allelic expression was analyzed using a polymorphic Fok1 cleavage site in 32 heterozygous gsp-negative tumors. GNAS expression was significantly reduced in the 14 tumors with relaxed GNAS imprinting and biallelic expression, compared to 18 tumors with monoallelic expression. Tumors with relaxed GNAS imprinting showed significantly lower SSTR2 and AIP expression levels. Conclusion: Altered A/B DMR methylation was found exclusively in gsp-negative somatotroph tumors. 43% of gsp-negative tumors showed GNAS imprinting relaxation, which correlated with lower GNAS, SSTR2 and AIP expression, indicating lower sensitivity to somatostatin analogues and potentially aggressive behavior.
- Published
- 2021
- Full Text
- View/download PDF
16. Somatotroph Tumors and the Epigenetic Status of the
- Author
-
Pauline, Romanet, Justine, Galluso, Peter, Kamenicky, Mirella, Hage, Marily, Theodoropoulou, Catherine, Roche, Thomas, Graillon, Heather C, Etchevers, Daniel, De Murat, Grégory, Mougel, Dominique, Figarella-Branger, Henry, Dufour, Thomas, Cuny, Guillaume, Assié, and Anne, Barlier
- Subjects
musculoskeletal diseases ,Adult ,Male ,gsp oncogene ,pituitary ,Article ,Epigenesis, Genetic ,Genomic Imprinting ,Young Adult ,GNAS ,stomatognathic system ,relaxation ,Chromogranins ,GTP-Binding Protein alpha Subunits, Gs ,Humans ,Pituitary Neoplasms ,Alleles ,Aged ,Aged, 80 and over ,somatotroph ,DNA Methylation ,Middle Aged ,Somatotrophs ,Gene Expression Regulation, Neoplastic ,tumorigenesis ,PitNET ,Mutation ,Female ,imprinting ,epigenetic - Abstract
Forty percent of somatotroph tumors harbor recurrent activating GNAS mutations, historically called the gsp oncogene. In gsp-negative somatotroph tumors, GNAS expression itself is highly variable; those with GNAS overexpression most resemble phenotypically those carrying the gsp oncogene. GNAS is monoallelically expressed in the normal pituitary due to methylation-based imprinting. We hypothesize that changes in GNAS imprinting of gsp-negative tumors affect GNAS expression levels and tumorigenesis. We characterized the GNAS locus in two independent somatotroph tumor cohorts: one of 23 tumors previously published (PMID: 31883967) and classified by pan-genomic analysis, and a second with 82 tumors. Multi-omics analysis of the first cohort identified a significant difference between gsp-negative and gsp-positive tumors in the methylation index at the known differentially methylated region (DMR) of the GNAS A/B transcript promoter, which was confirmed in the larger series of 82 tumors. GNAS allelic expression was analyzed using a polymorphic Fok1 cleavage site in 32 heterozygous gsp-negative tumors. GNAS expression was significantly reduced in the 14 tumors with relaxed GNAS imprinting and biallelic expression, compared to 18 tumors with monoallelic expression. Tumors with relaxed GNAS imprinting showed significantly lower SSTR2 and AIP expression levels. Altered A/B DMR methylation was found exclusively in gsp-negative somatotroph tumors. 43% of gsp-negative tumors showed GNAS imprinting relaxation, which correlated with lower GNAS, SSTR2 and AIP expression, indicating lower sensitivity to somatostatin analogues and potentially aggressive behavior.
- Published
- 2021
17. Pericyte Ontogeny: The Use of Chimeras to Track a Cell Lineage of Diverse Germ Line Origins
- Author
-
Heather C, Etchevers
- Subjects
Germ Cells ,Biological Ontologies ,Chimera ,Stem Cells ,Transplantation, Heterologous ,Animals ,Humans ,Cell Differentiation ,Cell Lineage ,Chick Embryo ,Endothelium, Vascular ,Pericytes - Abstract
The goal of lineage tracing is to understand body formation over time by discovering which cells are the progeny of a specific, identified, ancestral progenitor. Subsidiary questions include unequivocal identification of what they have become, how many descendants develop, whether they live or die, and where they are located in the tissue or body at the end of the window examined. A classical approach in experimental embryology, lineage tracing continues to be used in developmental biology and stem cell and cancer research, wherever cellular potential and behavior need to be studied in multiple dimensions, of which one is time. Each technical approach has its advantages and drawbacks. This chapter, with some previously unpublished data, will concentrate nonexclusively on the use of interspecies chimeras to explore the origins of perivascular (or mural) cells, of which those adjacent to the vascular endothelium are termed pericytes for this purpose. These studies laid the groundwork for our understanding that pericytes derive from progenitor mesenchymal pools of multiple origins in the vertebrate embryo, some of which persist into adulthood. The results obtained through xenografting, like in the methodology described here, complement those obtained through genetic lineage-tracing techniques within a given species.
- Published
- 2021
18. Pericyte Ontogeny: The Use of Chimeras to Track a Cell Lineage of Diverse Germ Line Origins
- Author
-
Heather C. Etchevers
- Subjects
0301 basic medicine ,Neural crest ,Biology ,Germline ,03 medical and health sciences ,Chimera (genetics) ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Evolutionary biology ,Fate mapping ,medicine ,Pericyte ,Stem cell ,Developmental biology ,030217 neurology & neurosurgery ,Progenitor - Abstract
The goal of lineage tracing is to understand body formation over time by discovering which cells are the progeny of a specific, identified, ancestral progenitor. Subsidiary questions include unequivocal identification of what they have become, how many descendants develop, whether they live or die, and where they are located in the tissue or body at the end of the window examined. A classical approach in experimental embryology, lineage tracing continues to be used in developmental biology, stem cell and cancer research, wherever cellular potential and behavior need to be studied in multiple dimensions, of which one is time. Each technical approach has its advantages and drawbacks. This brief review, with some previously unpublished data, will concentrate non-exclusively on the use of interspecies chimeras to explore the origins of perivascular (or mural) cells, of which those adjacent to the vascular endothelium are termed pericytes for this purpose. These studies laid the groundwork for our understanding that pericytes derive from progenitor mesenchymal pools of multiple origins in the vertebrate embryo, some of which persist into adulthood. The results obtained through xenografting complement and abut those obtained through genetic lineage tracing techniques within a given species.
- Published
- 2021
- Full Text
- View/download PDF
19. A subpopulation of smooth muscle cells, derived from melanocyte-competent precursors, prevents patent ductus arteriosus.
- Author
-
Ichiro Yajima, Sophie Colombo, Isabel Puig, Delphine Champeval, Mayuko Kumasaka, Elodie Belloir, Jacky Bonaventure, Manuel Mark, Hiroaki Yamamoto, Mark M Taketo, Philippe Choquet, Heather C Etchevers, Friedrich Beermann, Véronique Delmas, Laurent Monassier, and Lionel Larue
- Subjects
Medicine ,Science - Abstract
BACKGROUND: Patent ductus arteriosus is a life-threatening condition frequent in premature newborns but also present in some term infants. Current mouse models of this malformation generally lead to perinatal death, not reproducing the full phenotypic spectrum in humans, in whom genetic inheritance appears complex. The ductus arteriosus (DA), a temporary fetal vessel that bypasses the lungs by shunting the aortic arch to the pulmonary artery, is constituted by smooth muscle cells of distinct origins (SMC1 and SMC2) and many fewer melanocytes. To understand novel mechanisms preventing DA closure at birth, we evaluated the importance of cell fate specification in SMC that form the DA during embryonic development. Upon specific Tyr::Cre-driven activation of Wnt/β-catenin signaling at the time of cell fate specification, melanocytes replaced the SMC2 population of the DA, suggesting that SMC2 and melanocytes have a common precursor. The number of SMC1 in the DA remained similar to that in controls, but insufficient to allow full DA closure at birth. Thus, there was no cellular compensation by SMC1 for the loss of SMC2. Mice in which only melanocytes were genetically ablated after specification from their potential common precursor with SMC2, demonstrated that differentiated melanocytes themselves do not affect DA closure. Loss of the SMC2 population, independent of the presence of melanocytes, is therefore a cause of patent ductus arteriosus and premature death in the first months of life. Our results indicate that patent ductus arteriosus can result from the insufficient differentiation, proliferation, or contractility of a specific smooth muscle subpopulation that shares a common neural crest precursor with cardiovascular melanocytes.
- Published
- 2013
- Full Text
- View/download PDF
20. Human Developmental Cell Atlas: milestones achieved and the roadmap ahead
- Author
-
Mats Nilsson, Kathy K. Niakan, Anna Hupalowska, Kerstin B. Meyer, Dana Pe'er, Jennifer Rood, Andrew J. Copp, Bruce J. Aronow, Pablo G. Camara, Sten Linnarsson, Paolo Giacobini, Emily Kirby, Rahul Satija, Berthold Göttgens, Heather C. Etchevers, Bayanne Olabi, Joakim Lundeberg, Orit Rozenblatt-Rosen, Roser Vento-Tormo, Aviv Regev, John C. Marioni, Arnold R. Kriegstein, Gary D. Bader, Deanne Taylor, Simone Webb, Barbara Treutlein, J. Gray Camp, Alain Chédotal, Muzlifah Haniffa, Guoji Guo, Kylie R. James, and Sarah A. Teichmann
- Subjects
Engineering ,business.industry ,Atlas (topology) ,Developmental cell ,book.journal ,business ,book ,Neuroscience - Abstract
The Human Developmental Cell Atlas (HDCA), as part of the Human Cell Atlas, aims to generate a comprehensive reference map of cells during development. This detailed study of development will be critical for understanding normal organogenesis, the impact of mutations, environmental factors and infectious agents on congenital and childhood disorders, and the molecular cellular basis of ageing, cancer and regenerative medicine. In this perspective, we outline the challenges of mapping and modelling human development using state of the art technologies to create a reference atlas across gestation for scientific and clinical benefit. We discuss the potential value of HDCA to enhance human pluripotent stem cell-derived organoid model systems and, in turn, the use of organoids and animal models to inform HDCA. Finally, we provide a roadmap towards a complete atlas of human development.
- Published
- 2020
- Full Text
- View/download PDF
21. Cutaneous Melanocytic Tumors With Concomitant NRASQ61R and IDH1R132C Mutations: A Report of 6 Cases
- Author
-
Nicolas Macagno, Franck Tirode, Arnaud de la Fouchardière, Sandrine Sierra-Fortuny, Béatrice Vergier, Heather C. Etchevers, Véronique Haddad, and Daniel Pissaloux
- Subjects
0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Adult ,Male ,Pathology ,medicine.medical_specialty ,IDH1 ,Skin Neoplasms ,Adolescent ,Biology ,Pathology and Forensic Medicine ,Malignant transformation ,GTP Phosphohydrolases ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Dermis ,medicine ,Nevus ,Humans ,Benign melanocytic nevus ,Aged ,Aged, 80 and over ,Nevus, Pigmented ,Membrane Proteins ,Middle Aged ,medicine.disease ,Isocitrate Dehydrogenase ,Staining ,030104 developmental biology ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Melanocytes ,Surgery ,Female ,Anatomy ,Melanocytoma - Abstract
We report a series of 6 melanocytic proliferations harboring both NRAS and IDH1 hotspot mutations. Clinically, there was no specific sex-ratio, ages ranged from 18 to 85 years, and the trunk and limbs were the most affected localizations. In half of the cases, progressive modification of a pre-existing nevus was reported. Morphologically, all tumors were predominantly based in the dermis and the most striking pathologic finding was the presence of a background architecture of congenital-type nevi with a superimposed biphasic pattern formed by dendritic pigmented melanocytes surrounding areas of nevoid melanocytes. This finding was further underscored by HMB45 staining, which was positive in the dendritic cells and negative in the nevoid melanocytes. Four cases displayed increased cellularity and 1 case showed increased dermal mitotic activity. DNA and RNA sequencing revealed NRAS and IDH1 comutations in all 6 cases, with homogenous expression data according to unsupervised clustering analysis. Array-comparative genomic hybridization revealed no copy number alteration for the 2 most cellular and mitogenic cases. All were surgically excised, available follow-up for 2 patients showed no relapse nor metastases. We hypothesize that the IDH1 mutation is a secondary event in a pre-existing NRAS-mutated nevus and could be in part responsible for the emergence of a pigmented dendritic dermal component. So far, such comutations have been reported in one benign melanocytic nevus and several melanomas. This combination could represent a new subgroup of intermediate prognosis (melanocytoma) with a distinctive morphology. Further acquisition of genomic anomalies could progressively lead to malignant transformation.
- Published
- 2020
22. Melanocortin-1 receptor (MC1R) genotypes do not correlate with size in two cohorts of medium-to-giant congenital melanocytic nevi
- Author
-
Isabelle James, Gemma Tell-Marti, Cristina Carrera, Heather C. Etchevers, Alicia Barreiro, Josep Malvehy, Vanessa Martins da Silva, Neus Calbet-Llopart, Mirella Pascini-Garrigos, Susana Puig, Guillaume Captier, Miriam Potrony, Joan Anton Puig-Butille, and Nathalie Degardin
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Heterozygote ,Skin Neoplasms ,Adolescent ,Genotype ,Population ,Dermatology ,Biology ,Compound heterozygosity ,Genetic polymorphisms ,General Biochemistry, Genetics and Molecular Biology ,Germline ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Internal medicine ,Epidemiology ,medicine ,Genetic predisposition ,Humans ,Genetic Predisposition to Disease ,Risk factor ,education ,education.field_of_study ,Nevus, Pigmented ,Polimorfisme genètic ,Homozygote ,Phenotype ,Fenotip ,Skin diseases ,030104 developmental biology ,Malalties de la pell ,Spain ,030220 oncology & carcinogenesis ,Case-Control Studies ,Cohort ,Female ,Receptor, Melanocortin, Type 1 ,Melanocortin 1 receptor - Abstract
Congenital melanocytic nevi (CMN) are cutaneous malformations whose prevalence is inversely correlated with projected adult size. CMN are caused by somatic mutations, but epidemiological studies suggest that germline genetic factors may influence CMN development. In CMN patients from the U.K., genetic variants in the MC1R gene, such as p.V92M and loss-of-function variants, have been previously associated with larger CMN. We analyzed the association of MC1R variants with CMN characteristics in 113 medium-to-giant CMN patients from Spain and from a distinct cohort of 53 patients from France, Norway, Canada and the U.S. These cohorts were similar at the clinical and phenotypical level, except for the number of nevi per patient. We found that the p.V92M or loss-of-function MC1R variants either alone or in combination did not correlate with CMN size, in contrast to the U.K. CMN patients. An additional case-control analysis with 259 unaffected Spanish individuals, showed a higher frequency of MC1R compound heterozygous or homozygous variant genotypes in Spanish CMN patients compared to the control population (15.9% vs. 9.3%; P=0.075). Altogether, this study suggests that MC1R variants are not associated with CMN size in these non-U.K. cohorts. Additional studies are required to define the potential role of MC1R as a risk factor in CMN development.SIGNIFICANCECongenital melanocytic nevi (CMN) are common pigmented lesions that originate during prenatal life, without clear evidence of a genetic predisposition. To date, limited data exist regarding the role of the MC1R gene, a key regulator of human pigmentation, in the development of the class of rarer CMN that are greater than 10 cm diameter at projected adult size and associated with increased morbidity or mortality risks. This study provides data from a large set of such CMN patients to support the hypothesis that MC1R could be involved in the development of these types of lesions, but at the same time discounting its influence on the size of CMN across distinct populations. Improving our understanding of genetic susceptibility to rare types of CMN is necessary to determine whether routine germline genotyping is relevant in clinical practice.
- Published
- 2020
23. Epigenetic deregulation of GATA3 in neuroblastoma is associated with increased GATA3 protein expression and with poor outcomes
- Author
-
Jessica Charlet, Keith W. Brown, Karim Malik, Bader Almutairi, Heather C. Etchevers, Anthony R. Dallosso, Marianna Szemes, King Saud University [Riyadh] (KSU), Bristol Genetics Laboratory, Southmead Hospital, North Bristol NHS Trust, Bayer Corporation, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Institut National de la Santé et de la Recherche Médicale (INSERM)-Aix Marseille Université (AMU), and CLIC Sargent UK, the John James Bristol Foundation and the University of Bristol Cancer Research Fund,INSERM
- Subjects
0301 basic medicine ,[SDV]Life Sciences [q-bio] ,Cellular differentiation ,lcsh:Medicine ,GATA3 Transcription Factor ,Biology ,Article ,Epigenesis, Genetic ,Paediatric cancer ,Neuroblastoma ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Transcriptional regulation ,medicine ,cancer ,Humans ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Epigenetics ,lcsh:Science ,neoplasms ,Cancer ,Regulation of gene expression ,Multidisciplinary ,lcsh:R ,GATA3 ,DNA, Neoplasm ,Methylation ,DNA Methylation ,Prognosis ,medicine.disease ,Neoplasm Proteins ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,paediatric cancer ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,lcsh:Q ,[SDV.MHEP.DERM]Life Sciences [q-bio]/Human health and pathology/Dermatology - Abstract
International audience; To discover epigenetic changes that may underly neuroblastoma pathogenesis, we identified differentially methylated genes in neuroblastoma cells compared to neural crest cells, the presumptive precursors cells for neuroblastoma, by using genome-wide DNA methylation analysis. We previously described genes that were hypermethylated in neuroblastoma; in this paper we report on 67 hypomethylated genes, which were filtered to select genes that showed transcriptional over-expression and an association with poor prognosis in neuroblastoma, highlighting GATA3 for detailed studies. Specific methylation assays confirmed the hypomethylation of GATA3 in neuroblastoma, which correlated with high expression at both the RNA and protein level. Demethylation with azacytidine in cultured sympathetic ganglia cells led to increased GATA3 expression, suggesting a mechanistic link between GATA3 expression and DNA methylation. Neuroblastomas that had completely absent GATA3 methylation and/or very high levels of protein expression, were associated with poor prognosis. Knock-down of GATA3 in neuroblastoma cells lines inhibited cell proliferation and increased apoptosis but had no effect on cellular differentiation. These results identify GATA3 as an epigenetically regulated component of the neuroblastoma transcriptional control network, that is essential for neuroblastoma proliferation. This suggests that the GATA3 transcriptional network is a promising target for novel neuroblastoma therapies. Neuroblastoma (NB) is one of the commonest extra-cranial solid malignancies of childhood, which arises as a result of disordered development of the sympathetic nervous system from neural crest cells 1,2. Neuroblastoma is clinically heterogeneous, with younger patients (18months) mostly have disseminated tumours at diagnosis and poor outcomes 3. The clinical heterogeneity of neuroblastoma is reflected in its molecular pathogenesis, where no single pathway has been identified as being critical for tumour development. Oncogene activations were the first genetic alterations identified in neuroblastoma; initially MYCN amplification was identified in high-risk tumours 4 and later ALK mutations were discovered in inherited neuroblastoma and some sporadic high-risk tumours 5,6. Mutations in tumour suppressor genes such as PHOX2B 7 and NF1 8 have also been reported. Recent genome-wide analyses have identified genomic mutations and other alterations in chromatin remodelling genes such as ATRX, ARID1A and ARID1B, in components of the RAC-RHO pathway 9-11 and in TERT 12,13 , with relapsed tumours demonstrating an increased mutation rate 14,15. Like most childhood cancers, neuroblastomas contain fewer mutations than adult cancers 16,17 , with some tumours apparently containing no detectable driver mutations 9-11. In low-risk neuroblastomas, copy-number changes may drive tumorigenesis 18 , but the lack of driver mutations in many cases, emphasises the need to consider other mechanisms of pathogenesis, such as epigenetic alterations 19 .
- Published
- 2019
- Full Text
- View/download PDF
24. A severe clinical phenotype of Noonan syndrome with neonatal hypertrophic cardiomyopathy in the second case worldwide with RAF1 S259Y neomutation
- Author
-
Sonia Blibech, Lilia Kraoua, Sonia Abdelhak, Laurent Argiro, R. M’rad, Heather C. Etchevers, Hager Jaouadi, Amel Ben Chehida, Nicolas Lévy, Rym Benkhalifa, Neji Tebib, Nadia Kasdallah, Valérie Delague, Stéphane Zaffran, Laboratoire de Génomique Biomédicale et Oncogénétique - Biomedical Genomics and Oncogenetics Laboratory (LR11IPT05), Université de Tunis El Manar (UTM)-Institut Pasteur de Tunis, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP), Hôpital La Rabta [Tunis], Université de Tunis El Manar (UTM), Hôpital Charles Nicolle [Tunis], Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital militaire de Tunis, Military Hospital of Tunis, Laboratoire des Venins et Toxines, Institut Pasteur de Tunis, Institut Pasteur de Tunis, The project leading to this publication has received funding from the Excellence Initiative of Aix-Marseille University – A*MIDEX, a French ‘Investissements d'Avenir’ programme (RARE-MED project)., The authors would like to thank the family for their collaboration. This work was supported by the Tunisian Ministry of Public Health, the Ministry of Higher Education and Scientific Research (LR16IPT05)., and ANR-11-IDEX-0001,Amidex,INITIATIVE D'EXCELLENCE AIX MARSEILLE UNIVERSITE(2011)
- Subjects
Pathology ,RAF1 mutation ,030204 cardiovascular system & hematology ,medicine.disease_cause ,whole exome sequencing ,Exon ,0302 clinical medicine ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Noonan syndrome ,Exome sequencing ,2. Zero hunger ,0303 health sciences ,Mutation ,Hypertrophic cardiomyopathy ,General Medicine ,3. Good health ,Phenotype ,Failure to thrive ,RAS/MAPK pathway ,Female ,medicine.symptom ,Research Paper ,medicine.medical_specialty ,Tunisia ,macromolecular substances ,Protein Serine-Threonine Kinases ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Pectus excavatum ,Proto-Oncogene Proteins ,Genetics ,medicine ,Humans ,cardiovascular diseases ,[SDV.MHEP.OS]Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,030304 developmental biology ,[SDV.MHEP.PED]Life Sciences [q-bio]/Human health and pathology/Pediatrics ,Genetic heterogeneity ,business.industry ,Infant ,Cardiomyopathy, Hypertrophic ,hypertrophic cardiomyopathy ,medicine.disease ,Proto-Oncogene Proteins c-raf ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,business ,[SDV.MHEP.DERM]Life Sciences [q-bio]/Human health and pathology/Dermatology - Abstract
Noonan syndrome and related disorders are a group of clinically and genetically heterogeneous conditions caused by mutations in genes of the RAS/MAPK pathway. Noonan syndrome causes multiple congenital anomalies, which are frequently accompanied by hypertrophic cardiomyopathy (HCM). We report here a Tunisian patient with a severe phenotype of Noonan syndrome including neonatal HCM, facial dysmorphism, severe failure to thrive, cutaneous abnormalities, pectus excavatum and severe stunted growth, who died in her eighth month of life. Using whole exome sequencing, we identified a de novo mutation in exon 7 of the RAF1 gene: c.776C > A (p.Ser259Tyr). This mutation affects a highly conserved serine residue, a main mediator of Raf-1 inhibition via phosphorylation. To our knowledge the c.776C > A mutation has been previously reported in only one case with prenatally diagnosed Noonan syndrome. Our study further supports the striking correlation of RAF1 mutations with HCM and highlights the clinical severity of Noonan syndrome associated with a RAF1 p.Ser259Tyr mutation.
- Published
- 2019
- Full Text
- View/download PDF
25. Macrophage-Derived IL1 beta and TNF alpha Regulate Arginine Metabolism in Neuroblastoma
- Author
-
Murray D. Norris, Ashley Vardon, Andrea M. Berry, Francis Mussai, Sarah Booth, Jayne Murray, Louis Chesler, Samantha C. Brownhill, Susan A. Burchill, Laura D. Gamble, Luciana Gneo, Ronny Schmidt, Orli Yogev, P. N.M. Cheng, Michelle Haber, Sharon A. Egan, David S. Ziegler, Carmela De Santo, Livingstone Fultang, Fenna De Bie, Carmel McConville, Georgina L. Eden, Heather C. Etchevers, Division of Stem Cell Biology and Developmental Genetics, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), and Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Cancer Research ,Arginine ,MAP Kinase Signaling System ,Interleukin-1beta ,Mice, Transgenic ,Sarcoma, Ewing ,Article ,03 medical and health sciences ,Mice ,Neuroblastoma ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Myeloid Cells ,ARG2 ,Protein kinase B ,Tumor microenvironment ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Arginase ,Chemistry ,Tumor Necrosis Factor-alpha ,Macrophages ,medicine.disease ,3. Good health ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Tumor necrosis factor alpha - Abstract
Neuroblastoma is the most common childhood solid tumor, yet the prognosis for high-risk disease remains poor. We demonstrate here that arginase 2 (ARG2) drives neuroblastoma cell proliferation via regulation of arginine metabolism. Targeting arginine metabolism, either by blocking cationic amino acid transporter 1 (CAT-1)–dependent arginine uptake in vitro or therapeutic depletion of arginine by pegylated recombinant arginase BCT-100, significantly delayed tumor development and prolonged murine survival. Tumor cells polarized infiltrating monocytes to an M1-macrophage phenotype, which released IL1β and TNFα in a RAC-alpha serine/threonine-protein kinase (AKT)–dependent manner. IL1β and TNFα established a feedback loop to upregulate ARG2 expression via p38 and extracellular regulated kinases 1/2 (ERK1/2) signaling in neuroblastoma and neural crest–derived cells. Proteomic analysis revealed that enrichment of IL1β and TNFα in stage IV human tumor microenvironments was associated with a worse prognosis. These data thus describe an immune-metabolic regulatory loop between tumor cells and infiltrating myeloid cells regulating ARG2, which can be clinically exploited. Significance: These findings illustrate that cross-talk between myeloid cells and tumor cells creates a metabolic regulatory loop that promotes neuroblastoma progression.
- Published
- 2019
- Full Text
- View/download PDF
26. Outflow Tract Formation—Embryonic Origins of Conotruncal Congenital Heart Disease
- Author
-
Stéphane Zaffran, Heather C. Etchevers, Sonia Stefanovic, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), ANR-09-JCJC-0079,CROCRODIEL,Croissance Cristalline d'Oxydes Dielectriques(2009), ANR-18-CE13-0011,HEARTBOX,HEARTBOX(2018), and Gall, Valérie
- Subjects
0301 basic medicine ,lcsh:Diseases of the circulatory (Cardiovascular) system ,medicine.medical_specialty ,Cell type ,Heart disease ,[SDV]Life Sciences [q-bio] ,cardiac progenitor cells ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,Review ,03 medical and health sciences ,0302 clinical medicine ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Internal medicine ,medicine ,Pharmacology (medical) ,outflow tract ,General Pharmacology, Toxicology and Pharmaceutics ,Progenitor cell ,Endocardium ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,business.industry ,Lateral plate mesoderm ,Embryogenesis ,Neural crest ,second heart field ,medicine.disease ,congenital heart defects ,[SDV.MHEP.CSC] Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,[SDV] Life Sciences [q-bio] ,030104 developmental biology ,lcsh:RC666-701 ,Great arteries ,endocardium ,Cardiology ,business ,valve ,neural crest ,030217 neurology & neurosurgery ,cushion - Abstract
International audience; Anomalies in the cardiac outflow tract (OFT) are among the most frequent congenital heart defects (CHDs). During embryogenesis, the cardiac OFT is a dynamic structure at the arterial pole of the heart. Heart tube elongation occurs by addition of cells from pharyngeal, splanchnic mesoderm to both ends. These progenitor cells, termed the second heart field (SHF), were first identified twenty years ago as essential to the growth of the forming heart tube and major contributors to the OFT. Perturbation of SHF development results in common forms of CHDs, including anomalies of the great arteries. OFT development also depends on paracrine interactions between multiple cell types, including myocardial, endocardial and neural crest lineages. In this publication, dedicated to Professor Andriana Gittenberger-De Groot and her contributions to the field of cardiac development and CHDs, we review some of her pioneering studies of OFT development with particular interest in the diverse origins of the many cell types that contribute to the OFT. We also discuss the clinical implications of selected key findings for our understanding of the etiology of CHDs and particularly OFT malformations.
- Published
- 2021
- Full Text
- View/download PDF
27. The hedgehog pathway and ocular developmental anomalies
- Author
-
Heather C. Etchevers, Florencia Cavodeassi, Sophie Creuzet, St George's University of London, Institut des Neurosciences Paris-Saclay (NeuroPSI), Université Paris-Sud - Paris 11 (UP11)-Centre National de la Recherche Scientifique (CNRS), Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Institut National de la Santé et de la Recherche Médicale (INSERM)-Aix Marseille Université (AMU), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Etchevers, Heather
- Subjects
genetic structures ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Context (language use) ,Review ,Eye ,Hedgehog pathway ,03 medical and health sciences ,Neural crest ,Human genetics ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Genetics ,Animals ,Humans ,Hedgehog Proteins ,Sonic hedgehog ,[SDV.MHEP.OS]Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,Hedgehog ,Genetics (clinical) ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,biology ,Eye development ,030305 genetics & heredity ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,[SDV.BDD.EO] Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,Gene Expression Regulation, Developmental ,Hedgehog signaling pathway ,eye diseases ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,[SDV.MHEP.OS] Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,embryonic structures ,biology.protein ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,sense organs ,Signal transduction ,Neuroscience ,Signal Transduction - Abstract
International audience; Mutations in effectors of the Hedgehog signaling pathway are responsible for a wide variety of ocular developmental anomalies (ODA). These range from massive malformations of the brain and ocular primordia, not always compatible with postnatal life, to subtle but damaging functional effects on specific eye components. This review will concentrate on the effects and effectors of the major vertebrate Hedgehog ligand for eye and brain formation, Sonic hedgehog (SHH), in tissues that constitute the eye directly and also in those tissues that exert indirect influence on eye formation. After a brief overview of human eye development, the many roles of the SHH signaling pathway during both early and later morphogenetic processes in the brain and then eye and periocular 30 primordia will be evoked. Some of the unique molecular biology of this pathway in vertebrates, particularly ciliary signal transduction, will also be broached within this developmental cellular context.
- Published
- 2018
- Full Text
- View/download PDF
28. Widespread dynamic and pleiotropic expression of the melanocortin-1-receptor (MC1R) system is conserved across chick, mouse and human embryonic development
- Author
-
Chloe Santos, Nita Solanky, Dianne Gerrelli, Anna C. Thomas, Magalie E. Carey, Pauline Heux, Heather C. Etchevers, Veronica A. Kinsler, Wisenave Arulvasan, Genetics and Genomic Medicine, University College of London [London] (UCL), Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), UCL Institute of Child Health, and Great Ormond Street Hospital for Children [London] (GOSH)
- Subjects
0301 basic medicine ,Embryology ,medicine.medical_specialty ,Health, Toxicology and Mutagenesis ,Pomc ,Embryonic Development ,Locus (genetics) ,In situ hybridization ,Chick Embryo ,Biology ,Toxicology ,Avian Proteins ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Internal medicine ,medicine ,Genetics ,Human embryogenesis ,Animals ,Humans ,Prenatal ,Receptor ,Gene ,Nevus ,Research Articles ,030304 developmental biology ,Skin ,0303 health sciences ,Gene Expression Regulation, Developmental ,Brain ,Heart ,medicine.disease ,Embryo, Mammalian ,Phenotype ,Oculocutaneous albinism ,Hormone ,030104 developmental biology ,Endocrinology ,Liver ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Pediatrics, Perinatology and Child Health ,Melanocortin ,Receptor, Melanocortin, Type 1 ,030217 neurology & neurosurgery ,Developmental Biology ,Melanocortin 1 receptor ,Research Article - Abstract
BackgroundMC1R, a G-protein coupled receptor with high affinity for alpha-melanocyte stimulating hormone (αMSH), modulates pigment production in melanocytes from many species and is associated with human melanoma risk.MC1Rmutations affecting human skin and hair color also have pleiotropic effects on the immune response and analgesia. Variants affecting human pigmentationin uteroalter the congenital phenotype of both oculocutaneous albinism and congenital melanocytic naevi, and have a possible effect on birthweight.Methods and ResultsByin situhybridization, RT-PCR and immunohistochemistry, we show thatMC1Ris widely expressed during human, chick and mouse embryonic and fetal stages in many somatic tissues, particularly in the musculoskeletal and nervous systems, and conserved across evolution in these three amniotes. Its dynamic pattern differs from that ofTUBB3, a gene overlapping the same locus in humans and encoding class III β-tubulin. The αMSH peptide and the transcript for its precursor, pro-opiomelanocortin (POMC), are similarly present in numerous extra-cutaneous tissues.MC1Rgenotyping of variants p.(V60M) and p.(R151C) was undertaken for 867 healthy children from the Avon Longitudinal Study of Parent and Children (ALSPAC) cohort, and birthweight modelled using multiple logistic regression analysis. A significant positive association initially found between R151C and birth weight, independent of known birth weight modifiers, was not reproduced when combined with data from an independent genome-wide association study of 6,459 additional members of the same cohort.ConclusionsThese data clearly show a new and hitherto unsuspected role for MC1R in non-cutaneous solid tissues before birth.
- Published
- 2018
- Full Text
- View/download PDF
29. Giant congenital melanocytic nevus with vascular malformation and epidermal cysts associated with a somatic activating mutation in BRAF
- Author
-
Frédéric Fina, Helmuth Vorbringer, Christian Rose, Pauline Heux, Benjamin Schwarz, Nicolas Macagno, Birgit Kahle, Stéphane Zaffran, Sven Krengel, Irina Berger, Heather C. Etchevers, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Catholique de Lille - Faculté de Médecine, Maïeutique, Sciences de la santé (FMMS), Institut Catholique de Lille (ICL), Université catholique de Lille (UCL)-Université catholique de Lille (UCL), Universitätsklinikum Schleswig-Holstein, Service d'Anatomo-Cyto-Pathologie et de NeuroPathologie [Hôpital de la Timone - APHM] (ACPNP), Aix Marseille Université (AMU)- Hôpital de la Timone [CHU - APHM] (TIMONE), Centre de Recherches en Oncologie biologique et Oncopharmacologie (CRO2), Aix Marseille Université (AMU)- Hôpital de la Timone [CHU - APHM] (TIMONE)-Institut National de la Santé et de la Recherche Médicale (INSERM), This study was supported by grants from Nevus Outreach, Inc., Asociación Española de afectados por Nevus Gigante Congénito, Association du Naevus Géant Congénital, Naevus 2000 France-Europe, Caring Matters Now, the RE(ACT) Community and aid-in-kind from the BeHeard Rare Disease Science Challenge., Université catholique de Lille - Faculté de médecine et de maïeutique (UCL FMM), Université catholique de Lille (UCL), and Etchevers, Heather
- Subjects
Male ,Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Neuroblastoma RAS viral oncogene homolog ,Pathology ,medicine.medical_specialty ,melanocyte ,Vascular Malformations ,[SDV.MHEP.PHY] Life Sciences [q-bio]/Human health and pathology/Tissues and Organs [q-bio.TO] ,Epidermal Cyst ,Dermatology ,[SDV.GEN.GH] Life Sciences [q-bio]/Genetics/Human genetics ,Biology ,General Biochemistry, Genetics and Molecular Biology ,BRAF ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Congenital melanocytic nevus ,medicine ,[SDV.MHEP.PHY]Life Sciences [q-bio]/Human health and pathology/Tissues and Organs [q-bio.TO] ,Humans ,Nevus ,HRAS ,skin and connective tissue diseases ,neoplasms ,Aged ,Nevus, Pigmented ,venous ,GNA11 ,integumentary system ,Vascular malformation ,congenital ,[SDV.MHEP.DERM] Life Sciences [q-bio]/Human health and pathology/Dermatology ,medicine.disease ,malformation ,030104 developmental biology ,Oncology ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Mutation ,Venous malformation ,GNAQ ,[SDV.MHEP.DERM]Life Sciences [q-bio]/Human health and pathology/Dermatology ,nevus - Abstract
International audience; Giant congenital melanocytic nevi may be symptomatically isolated, or syndromic. Associations with capillary malformations are exceptional, and development of epidermal cysts has not been described. A 71-year old patient with a giant congenital melanocytic nevus of the lower back, buttocks and thighs was asymptomatic except for unexpected hemorrhage during partial surgical excision years before. Blunt trauma at age 64 initiated recurrent, severe pain under the nevus; multiple large epidermal cysts developed within it. Imaging and biopsy showed a large, non-pulsatile venous malformation intermingled with the deep nevus. A low-abundance, heterozygous BRAF c.1799T>A (p.V600E) mutation was present in both the gluteal and occipital “satellite” nevi; additional mutations in NRAS, GNAQ, GNA11, HRAS and PIK3CA were undetectable. This is the first demonstration of an identical BRAF mutation in multiple congenital nevi from the same individual, confirming genetic heterogeneity in giant nevi. This exceptional case indicates that constitutive activation of BRAF can be an underlying cause of unusual associations of giant nevi with vascular malformations, and that the latter may be included among the somatic RASopathies.
- Published
- 2018
- Full Text
- View/download PDF
30. Ectopic expression of Hoxb1 induces cardiac and craniofacial malformations
- Author
-
Heather C. Etchevers, Sonia Stefanovic, Fabienne Lescroart, Gaëlle Odelin, Stéphane Zaffran, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Etchevers, Heather
- Subjects
0301 basic medicine ,maxillary aplasia ,PAX3 ,[SDV.BC.IC] Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Mouse models ,Ectopic Gene Expression ,Craniofacial Abnormalities ,Mice ,Hox genes ,0302 clinical medicine ,Endocrinology ,exencephaly ,Cell Movement ,Cell Behavior (q-bio.CB) ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Hox gene ,cleft palate ,0303 health sciences ,education.field_of_study ,Genes, Homeobox ,Neural crest ,[SDV.BDD.EO] Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,Gene Expression Regulation, Developmental ,Heart ,3. Good health ,Cell biology ,[SDV.MHEP.CSC] Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,[SDV.MHEP.OS] Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,Neural Crest ,embryonic structures ,Neural crest stem cells ,valve ,Signal Transduction ,double outlet right ventricle ,Heart Defects, Congenital ,Agnathia ,animal structures ,Population ,Cre recombinase ,Mice, Transgenic ,[SDV.GEN.GA] Life Sciences [q-bio]/Genetics/Animal genetics ,Biology ,[SDV.GEN.GH] Life Sciences [q-bio]/Genetics/Human genetics ,mandibular agenesis ,03 medical and health sciences ,[SDV.MHEP.PED] Life Sciences [q-bio]/Human health and pathology/Pediatrics ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,Genetics ,Animals ,Cell Lineage ,[SDV.MHEP.OS]Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,education ,Transcription factor ,030304 developmental biology ,Homeodomain Proteins ,[SDV.MHEP.PED]Life Sciences [q-bio]/Human health and pathology/Pediatrics ,Cell Biology ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,030104 developmental biology ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,FOS: Biological sciences ,Quantitative Biology - Cell Behavior ,Ectopic expression ,Head ,030217 neurology & neurosurgery - Abstract
Members of the large family of Hox transcription factors are encoded by genes whose tightly regulated expression in development and in space within different embryonic tissues confer positional identity from the neck to the tips of the limbs. Many structures of the face, head, and heart10 develop from cell populations expressing few or no Hox genes. Hoxb1 is the member of its chromosomal cluster expressed in the most rostral domain during vertebrate development, but never by the multipotent neural crest cell population anterior to the cerebellum. We have developed a novel floxed transgenic mouse line, CAG-Hoxb1,-EGFP (CAG-Hoxb1), which upon recombination by Cre recombinase conditionally induces robust Hoxb1 and eGFP overexpression. When induced within the neural crest lineage, pups die at birth. A variable phenotype develops from E11.5 on, associating frontonasal hypoplasia/aplasia, micrognathia/agnathia, major ocular and forebrain anomalies, and cardiovascular malformations. Neural crest derivatives in the body appear unaffected. Transcription of effectors of developmental signaling pathways (Bmp, Shh, Vegfa) and transcription factors (Pax3, Sox9) is altered in mutants. These outcomes emphasize that repression of Hoxb1, along with other paralog group 1 and 2 Hox genes, is strictly necessary in anterior cephalic NC for craniofacial, visual, auditory, and cardiovascular development., Comment: Genesis, Wiley-Blackwell, In press
- Published
- 2018
- Full Text
- View/download PDF
31. The diverse neural crest: from embryology to human pathology
- Author
-
Heather C. Etchevers, Nicole M. Le Douarin, Elisabeth Dupin, Marseille medical genetics - Centre de génétique médicale de Marseille (MMG), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de la Vision, Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
EDN ,Hirschsprung disease ,rhombomere ,PNS ,Chick Embryo ,0302 clinical medicine ,enteric nervous system ,Cell Movement ,Embryonic Structure ,Neoplasms ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,multiple endocrine neoplasia ,WS ,0303 health sciences ,education.field_of_study ,biology ,Vertebrate ,Neural crest ,Gene Expression Regulation, Developmental ,Cell Differentiation ,MEN ,Quail non Chick Peri-Nuclear ,Biological Evolution ,HSCR ,Embryology ,endothelin ,neural crest ,Schwann cell precursor ,dorsal root ganglion ,ENS ,Population ,Rhombomere ,embryo ,Coturnix ,Cell fate determination ,03 medical and health sciences ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,peripheral nervous system ,biology.animal ,Animals ,Humans ,Cell Lineage ,Genetic Predisposition to Disease ,[SDV.MHEP.OS]Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,education ,Molecular Biology ,Waardenburg syndrome ,030304 developmental biology ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,embryonic day ,[SDV.MHEP.PED]Life Sciences [q-bio]/Human health and pathology/Pediatrics ,QCPN ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,SCP ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,DRG ,NC ,Neuroscience ,Human Pathology ,030217 neurology & neurosurgery ,[SDV.MHEP.DERM]Life Sciences [q-bio]/Human health and pathology/Dermatology ,Developmental Biology - Abstract
We review here some of the historical highlights in exploratory studies of the vertebrate embryonic structure known as the neural crest. The study of the molecular properties of the cells that it produces, their migratory capacities and plasticity, and the still-growing list of tissues that depend on their presence for form and function, continue to enrich our understanding of congenital malformations, paediatric cancers and evolutionary biology. Developmental biology has been key to our understanding of the neural crest, starting with the early days of experimental embryology and through to today, when increasingly powerful technologies contribute to further insight into this fascinating vertebrate cell population.
- Published
- 2018
- Full Text
- View/download PDF
32. Cardiac outflow morphogenesis depends on effects of retinoic acid signaling on multiple cell lineages
- Author
-
Karen Niederreither, Emilie Faure, Nathalie Eudes, Nicolas Bertrand, Lucile Ryckebüsch, Nicolas El Robrini, Heather C. Etchevers, and Stéphane Zaffran
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Heart development ,Cardiac neural crest cells ,Mesenchyme ,Retinoic acid ,Morphogenesis ,Neural crest ,Biology ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Tretinoin ,Internal medicine ,embryonic structures ,cardiovascular system ,medicine ,Endocardium ,Developmental Biology ,medicine.drug - Abstract
Background: Retinoic acid (RA), the bioactive derivative of vitamin A, is essential for vertebrate heart development. Both excess and reduced RA signaling lead to cardiovascular malformations affecting the outflow tract (OFT). To address the cellular mechanisms underlying the effects of RA signaling during OFT morphogenesis, we used transient maternal RA supplementation to rescue the early lethality resulting from inactivation of the murine retinaldehyde dehydrogenase 2 (Raldh2) gene. Results: By embryonic day 13.5, all rescued Raldh2(-/-) hearts exhibit severe, reproducible OFT septation defects, although wild-type and Raldh2(+/-) littermates have normal hearts. Cardiac neural crest cells (cNCC) were present in OFT cushions of Raldh2(-/-) mutant embryos but ectopically located in the periphery of the endocardial cushions, rather than immediately underlying the endocardium. Excess mesenchyme was generated by Raldh2(-/-) mutant endocardium, which displaced cNCC derivatives from their subendocardial, medial position. Conclusions: RA signaling affects not only cNCC numbers but also their position relative to endocardial mesenchyme during the septation process. Our study shows that inappropriate coordination between the different cell types of the OFT perturbs its morphogenesis and leads to a severe congenital heart defect, persistent truncus arteriosus. Developmental Dynamics 245:388-401, 2016. (c) 2015 Wiley Periodicals, Inc.
- Published
- 2015
- Full Text
- View/download PDF
33. Pericyte ontogeny: the use of chimeras to track a cell lineage of diverse germ line origins
- Author
-
Heather C. Etchevers, Génétique Médicale et Génomique Fonctionnelle (GMGF), and Aix Marseille Université (AMU)-Assistance Publique - Hôpitaux de Marseille (APHM)- Hôpital de la Timone [CHU - APHM] (TIMONE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Germline ,03 medical and health sciences ,0302 clinical medicine ,Blood vessels ,[SDV.MHEP.CSC]Life Sciences [q-bio]/Human health and pathology/Cardiology and cardiovascular system ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,medicine ,030304 developmental biology ,Progenitor ,0303 health sciences ,[SDV.MHEP.PED]Life Sciences [q-bio]/Human health and pathology/Pediatrics ,Fate mapping ,Mesenchymal stem cell ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,medicine.anatomical_structure ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Evolutionary biology ,Embryology ,Immunology ,Identification (biology) ,Pericyte ,Stem cell ,Pericytes ,Developmental biology ,030217 neurology & neurosurgery - Abstract
The goal of lineage tracing is to understand body formation over time by discovering which cells are the progeny of a specific, identified, ancestral progenitor. Subsidiary questions include unequivocal identification of what they have become, how many descendants develop, whether they live or die, and where they are located in the tissue or body at the end of the window examined. A classical approach in experimental embryology, lineage tracing continues to be used in developmental biology, stem cell and cancer research, wherever cellular potential and behavior need to be studied in multiple dimensions, of which one is time. Each technical approach has its advantages and drawbacks. This chapter, with some previously unpublished data, will concentrate non-exclusively on the use of interspecies chimeras to explore the origins of perivascular (or mural) cells, of which those adjacent to the vascular endothelium are termed pericytes for this purpose. These studies laid the groundwork for our understanding that pericytes derive from progenitor mesenchymal pools of multiple origins in the vertebrate embryo, some of which persist into adulthood. The results obtained through xenografting, like in the methodology described here, complement those obtained through genetic lineage tracing techniques within a given species.
- Published
- 2017
34. Heterogeneity of neuroblastoma cell identity defined by transcriptional circuitries
- Author
-
Angel M. Carcaboso, Thomas G. P. Grunewald, Sandrine Grossetête-Lalami, Gaëlle Pierron, Alban Lermine, Cécile Pierre-Eugène, Bertrand Ducos, Valentina Boeva, Heather C. Etchevers, Virginie Raynal, Eve Lapouble, Estelle Daudigeos-Dubus, Caroline Louis-Brennetot, Birgit Geoerger, Thomas Deller, Isabelle Janoueix-Lerosey, Sophie Thomas, Valérie Combaret, Didier Surdez, Irina V. Medvedeva, Olivier Delattre, Agathe Peltier, Elise Diaz, Emmanuel Barillot, Hermann Rohrer, Simon Durand, Martin F. Orth, Gudrun Schleiermacher, Sylvain Baulande, Cancer et génome: Bioinformatique, biostatistiques et épidémiologie d'un système complexe, Institut Curie [Paris]-MINES ParisTech - École nationale supérieure des mines de Paris, Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Unité de génétique et biologie des cancers (U830), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5), Plateforme de sequencage ICGEX, Institut Curie [Paris], Génétique Médicale et Génomique Fonctionnelle (GMGF), Institut National de la Santé et de la Recherche Médicale (INSERM)-Aix Marseille Université (AMU)-Assistance Publique - Hôpitaux de Marseille (APHM)- Hôpital de la Timone [CHU - APHM] (TIMONE)-Centre National de la Recherche Scientifique (CNRS), Embryology and genetics of human malformation (Equipe Inserm U1163), Imagine - Institut des maladies génétiques (IMAGINE - U1163), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Vectorologie et thérapeutiques anti-cancéreuses [Villejuif] (UMR 8203), Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Centre National de la Recherche Scientifique (CNRS), Ludwig-Maximilians-Universität München (LMU), Institut de biologie de l'ENS Paris (IBENS), Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Département de Biologie - ENS Paris, École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Laboratoire de Physique Statistique de l'ENS (LPS), Université Paris Diderot - Paris 7 (UPD7)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Centre National de la Recherche Scientifique (CNRS)-Fédération de recherche du Département de physique de l'Ecole Normale Supérieure - ENS Paris (FRDPENS), Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL), Centre interdisciplinaire de recherche en biologie (CIRB), Labex MemoLife, Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Goethe-University Frankfurt am Main, Centre Léon Bérard [Lyon], Unité de Génétique Somatique, Goethe-Universität Frankfurt am Main, Mines Paris - PSL (École nationale supérieure des mines de Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Descartes - Paris 5 (UPD5)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Aix Marseille Université (AMU)-Assistance Publique - Hôpitaux de Marseille (APHM)- Hôpital de la Timone [CHU - APHM] (TIMONE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Département de Biologie - ENS Paris, École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Fédération de recherche du Département de physique de l'Ecole Normale Supérieure - ENS Paris (FRDPENS), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Université Paris sciences et lettres (PSL)-École normale supérieure - Paris (ENS-PSL), Centre National de la Recherche Scientifique (CNRS)-Institut Gustave Roussy (IGR)-Université Paris-Sud - Paris 11 (UP11), Institut de biologie de l'ENS Paris (UMR 8197/1024) (IBENS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Département de Biologie - ENS Paris, Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Gall, Valérie
- Subjects
0301 basic medicine ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Blotting, Western ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Mice, SCID ,Genetic Heterogeneity ,Neuroblastoma ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Mice, Inbred NOD ,Cell Line, Tumor ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Genetics ,medicine ,Animals ,Humans ,Cell Lineage ,Epigenetics ,Transcription factor ,neoplasms ,Homeodomain Proteins ,Mice, Knockout ,Regulation of gene expression ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,HEK 293 cells ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,GATA3 ,Neural crest ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,HEK293 Cells ,RNAi Therapeutics ,030104 developmental biology ,Doxycycline ,Immunology ,biology.protein ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,RNA Interference ,Single-Cell Analysis ,HAND2 ,Neuroscience ,Transcription Factors - Abstract
International audience; Neuroblastoma is a tumor of the peripheral sympathetic nervous system(1), derived from multipotent neural crest cells (NCCs). To define core regulatory circuitries (CRCs) controlling the gene expression program of neuroblastoma, we established and analyzed the neuroblastoma super-enhancer landscape. We discovered three types of identity in neuroblastoma cell lines: a sympathetic noradrenergic identity, defined by a CRC module including the PHOX2B, HAND2 and GATA3 transcription factors (TFs); an NCC-like identity, driven by a CRC module containing AP-1 TFs; and a mixed type, further deconvoluted at the single-cell level. Treatment of the mixed type with chemotherapeutic agents resulted in enrichment of NCC-like cells. The noradrenergic module was validated by ChIP-seq. Functional studies demonstrated dependency of neuroblastoma with noradrenergic identity on PHOX2B, evocative of lineage addiction. Most neuroblastoma primary tumors express TFs from the noradrenergic and NCC-like modules. Our data demonstrate a previously unknown aspect of tumor heterogeneity relevant for neuroblastoma treatment strategies.
- Published
- 2017
- Full Text
- View/download PDF
35. Genome-wide DNA methylation analysis identifies MEGF10 as a novel epigenetically repressed candidate tumor suppressor gene in neuroblastoma
- Author
-
Jessica, Charlet, Ayumi, Tomari, Anthony R, Dallosso, Marianna, Szemes, Martina, Kaselova, Thomas J, Curry, Bader, Almutairi, Heather C, Etchevers, Carmel, McConville, Karim T A, Malik, and Keith W, Brown
- Subjects
DNA methylation ,epigenetics ,Membrane Proteins ,Articles ,Article ,Epigenesis, Genetic ,Gene Expression Regulation, Neoplastic ,Histone Code ,neuroblastoma ,Cell Line, Tumor ,MEGF10 ,Humans ,Genes, Tumor Suppressor ,histone methylation ,Child - Abstract
Neuroblastoma is a childhood cancer in which many children still have poor outcomes, emphasising the need to better understand its pathogenesis. Despite recent genome‐wide mutation analyses, many primary neuroblastomas do not contain recognizable driver mutations, implicating alternate molecular pathologies such as epigenetic alterations. To discover genes that become epigenetically deregulated during neuroblastoma tumorigenesis, we took the novel approach of comparing neuroblastomas to neural crest precursor cells, using genome‐wide DNA methylation analysis. We identified 93 genes that were significantly differentially methylated of which 26 (28%) were hypermethylated and 67 (72%) were hypomethylated. Concentrating on hypermethylated genes to identify candidate tumor suppressor loci, we found the cell engulfment and adhesion factor gene MEGF10 to be epigenetically repressed by DNA hypermethylation or by H3K27/K9 methylation in neuroblastoma cell lines. MEGF10 showed significantly down‐regulated expression in neuroblastoma tumor samples; furthermore patients with the lowest‐expressing tumors had reduced relapse‐free survival. Our functional studies showed that knock‐down of MEGF10 expression in neuroblastoma cell lines promoted cell growth, consistent with MEGF10 acting as a clinically relevant, epigenetically deregulated neuroblastoma tumor suppressor gene. © 2016 The Authors. Molecular Carcinogenesis Published by Wiley Periodicals, Inc.
- Published
- 2016
36. Targeted resequencing identifies PTCH1 as a major contributor to ocular developmental anomalies and extends the SOX2 regulatory network
- Author
-
Nicolas Chassaing, Josseline Kaplan, Catherine Vincent-Delorme, Adrienne R. Niederriter, Sophie Lamarre, Stanislas Faguer, Nicholas Katsanis, Kelly L. McKnight, Erica E. Davis, Didier Lacombe, Annaïck Desmaison, Jean-Louis Dufier, Massimiliano Rossi, Christine Coubes, Laurent Pasquier, Heather C. Etchevers, Alexandre Causse, Hélène Dollfus, Patrick Calvas, Véronique David, Hôpital Purpan [Toulouse], CHU Toulouse [Toulouse], Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées, Unité différenciation épidermique et auto-immunité rhumatoïde (UDEAR), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Génétique et Développement de Rennes (IGDR), Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Centre National de la Recherche Scientifique (CNRS)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Service de biologie moléculaire, Hôpital Pontchaillou, Institut des Technologies Avancées en sciences du Vivant (ITAV), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique (CNRS), Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés (LISBP), Centre National de la Recherche Scientifique (CNRS)-Institut National des Sciences Appliquées - Toulouse (INSA Toulouse), Institut National des Sciences Appliquées (INSA)-Institut National des Sciences Appliquées (INSA)-Institut National de la Recherche Agronomique (INRA), Centre de Maladies Rares, Anomalies du Développement Nord de France-CH Arras - CHRU Lille, CHU Pontchaillou [Rennes], Service de Génétique Clinique, Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Service de génétique médicale, Université de Bordeaux (UB)-CHU Bordeaux [Bordeaux]-Groupe hospitalier Pellegrin, Centre de recherche en neurosciences de Lyon (CRNL), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet [Saint-Étienne] (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Service d'ophtalmologie [CHU Necker], CHU Necker - Enfants Malades [AP-HP]-Assistance publique - Hôpitaux de Paris (AP-HP) (APHP), Centre de Référence pour les Affections Rares en Génétique Ophtalmologique (CARGO) et Service de Génétique Médicale, Hôpitaux Universitaires de Strasbourg, Imagine - Institut des maladies génétiques (IMAGINE - U1163), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Descartes - Paris 5 (UPD5), Center for Human Disease Modeling, Duke University [Durham], Département de Néphrologie et Transplantation d'organes, Hôpital de Rangueil, CHU Toulouse [Toulouse]-CHU Toulouse [Toulouse], Centre de référence des maladies rénales rares, CHU Toulouse [Toulouse]-Hôpital de Rangueil, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), PHRC 09 109 01, Clinical Research Hospital Program from the French Ministry of Health, Retina France, Université de Toulouse (UT)-Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM), Duke University Medical Center, Université de Toulouse (UT), Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), Université de Rennes (UR)-Centre National de la Recherche Scientifique (CNRS)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Université de Toulouse (UT)-Université de Toulouse (UT)-Centre National de la Recherche Scientifique (CNRS), Institut National de la Recherche Agronomique (INRA)-Institut National des Sciences Appliquées - Toulouse (INSA Toulouse), Institut National des Sciences Appliquées (INSA)-Université de Toulouse (UT)-Institut National des Sciences Appliquées (INSA)-Université de Toulouse (UT)-Centre National de la Recherche Scientifique (CNRS), Hôpital Jeanne de Flandres, Université de Lille, Droit et Santé-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Centre de recherche en neurosciences de Lyon - Lyon Neuroscience Research Center (CRNL), Université de Lyon-Université de Lyon-Université Jean Monnet - Saint-Étienne (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Département de Néphrologie et Transplantation d'organes [CHU Toulouse], Pôle Urologie - Néphrologie - Dialyse - Transplantations - Brûlés - Chirurgie plastique - Explorations fonctionnelles et physiologiques [CHU Toulouse], Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse), Centre de Référence du sud-Ouest des maladies rénales rares [CHU Toulouse] (SODARE), Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique )-Centre National de la Recherche Scientifique (CNRS)-Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES), Centre National de la Recherche Scientifique (CNRS)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées, Institut National des Sciences Appliquées (INSA)-Institut National des Sciences Appliquées (INSA)-Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Toulouse III - Paul Sabatier (UT3), Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-CHU Necker - Enfants Malades [AP-HP], Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Universitaire de Toulouse, Institut de Génétique et Développement de Rennes ( IGDR ), Université de Rennes 1 ( UR1 ), Université de Rennes ( UNIV-RENNES ) -Université de Rennes ( UNIV-RENNES ) -Centre National de la Recherche Scientifique ( CNRS ) -Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Institut des Technologies Avancées en sciences du Vivant ( ITAV ), Université Toulouse III - Paul Sabatier ( UPS ), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre National de la Recherche Scientifique ( CNRS ), Laboratoire d'Ingénierie des Systèmes Biologiques et des Procédés ( LISBP ), Institut National de la Recherche Agronomique ( INRA ) -Institut National des Sciences Appliquées - Toulouse ( INSA Toulouse ), Institut National des Sciences Appliquées ( INSA ) -Institut National des Sciences Appliquées ( INSA ) -Centre National de la Recherche Scientifique ( CNRS ), Centre Hospitalier Régional Universitaire [Montpellier] ( CHRU Montpellier ), Université de Bordeaux ( UB ) -CHU Bordeaux [Bordeaux]-Groupe hospitalier Pellegrin, Inserm U1028, équipe TIGER, Centre de recherche en neurosciences de Lyon ( CRNL ), Université Claude Bernard Lyon 1 ( UCBL ), Université de Lyon-Université de Lyon-Université Jean Monnet [Saint-Étienne] ( UJM ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ) -Université Claude Bernard Lyon 1 ( UCBL ), Université de Lyon-Université de Lyon-Université Jean Monnet [Saint-Étienne] ( UJM ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ) -Service de Génétique, Hospices Civils de Lyon ( HCL ) -Hôpital Louis Pradel [CHU - HCL], Hospices Civils de Lyon ( HCL ) -Groupe Hospitalier Est-Hospices Civils de Lyon ( HCL ) -Hôpital Louis Pradel [CHU - HCL], Hospices Civils de Lyon ( HCL ) -Groupe Hospitalier Est, Assistance publique - Hôpitaux de Paris (AP-HP)-CHU Necker - Enfants Malades [AP-HP], Imagine - Institut des maladies génétiques ( IMAGINE - U1163 ), Centre National de la Recherche Scientifique ( CNRS ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Université Paris Descartes - Paris 5 ( UPD5 ), Duke university [Durham], Institut des Maladies Métaboliques et Cardiovasculaires ( I2MC ), Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Hôpital de Rangueil, and CHU Toulouse [Toulouse]-CHU Toulouse [Toulouse]-Université Toulouse III - Paul Sabatier ( UPS )
- Subjects
0301 basic medicine ,Patched ,Candidate gene ,Heterozygote ,endocrine system ,Biology ,Microphthalmia ,Frameshift mutation ,03 medical and health sciences ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Genetics ,medicine ,Missense mutation ,Animals ,Humans ,Gene Regulatory Networks ,Eye Abnormalities ,Sonic hedgehog ,[SDV.BDD]Life Sciences [q-bio]/Development Biology ,Genetics (clinical) ,Alleles ,Zebrafish ,Anophthalmia ,[ SDV ] Life Sciences [q-bio] ,SOXB1 Transcription Factors ,Research ,Sequence Analysis, DNA ,medicine.disease ,Patched-1 Receptor ,Disease Models, Animal ,030104 developmental biology ,Phenotype ,PTCH1 ,Gene Expression Regulation ,Genetic Loci ,Case-Control Studies ,Mutation ,embryonic structures ,biology.protein - Abstract
Ocular developmental anomalies (ODA) such as anophthalmia/microphthalmia (AM) or anterior segment dysgenesis (ASD) have an estimated combined prevalence of 3.7 in 10,000 births. Mutations in SOX2 are the most frequent contributors to severe ODA, yet account for a minority of the genetic drivers. To identify novel ODA loci, we conducted targeted high-throughput sequencing of 407 candidate genes in an initial cohort of 22 sporadic ODA patients. Patched 1 (PTCH1), an inhibitor of sonic hedgehog (SHH) signaling, harbored an enrichment of rare heterozygous variants in comparison to either controls, or to the other candidate genes (four missense and one frameshift); targeted resequencing of PTCH1 in a second cohort of 48 ODA patients identified two additional rare nonsynonymous changes. Using multiple transient models and a CRISPR/Cas9-generated mutant, we show physiologically relevant phenotypes altering SHH signaling and eye development upon abrogation of ptch1 in zebrafish for which in vivo complementation assays using these models showed that all six patient missense mutations affect SHH signaling. Finally, through transcriptomic and ChIP analyses, we show that SOX2 binds to an intronic domain of the PTCH1 locus to regulate PTCH1 expression, findings that were validated both in vitro and in vivo. Together, these results demonstrate that PTCH1 mutations contribute to as much as 10% of ODA, identify the SHH signaling pathway as a novel effector of SOX2 activity during human ocular development, and indicate that ODA is likely the result of overactive SHH signaling in humans harboring mutations in either PTCH1 or SOX2.
- Published
- 2016
- Full Text
- View/download PDF
37. OTX2mutations contribute to the otocephaly-dysgnathia complex
- Author
-
Patrick Calvas, Susanna Sorrentino, Ahmet Yesilyurt, Consolato Sergi, Dominique Carles, Hülya Kayserili, Ona Faye-Petersen, Dominique Martin-Coignard, Anthony J. Iacovelli, Adeline Vigouroux, Surasak Puvabanditsin, Nicolas Chassaing, Nicholas Katsanis, Philippe Loget, Ethylin Wang Jabs, William A. Paznekas, Férechté Encha-Razavi, Erica E. Davis, Simeon A. Boyadjiev, Catherine Mercer, Bryn D. Webb, Leopoldine Lequeux, Heather C. Etchevers, and Chih Ping Chen
- Subjects
Otocephaly ,Embryo, Nonmammalian ,Molecular Sequence Data ,Locus (genetics) ,Microphthalmia ,Jaw Abnormalities ,Holoprosencephaly ,Genetics ,medicine ,Animals ,Humans ,Zebrafish ,Genetics (clinical) ,Loss function ,Otx Transcription Factors ,Anophthalmia ,Base Sequence ,biology ,Genetic heterogeneity ,Sequence Analysis, DNA ,medicine.disease ,Dysgnathia ,biology.organism_classification ,Pedigree ,Disease Models, Animal ,Female - Abstract
Background Otocephaly or dysgnathia complex is characterised by mandibular hypoplasia/agenesis, ear anomalies, microstomia, and microglossia; the molecular basis of this developmental defect is largely unknown in humans. Methods and results This study reports a large family in which two cousins with micro/anophthalmia each gave birth to at least one child with otocephaly, suggesting a genetic relationship between anophthalmia and otocephaly. OTX2 , a known microphthalmia locus, was screened in this family and a frameshifting mutation was found. The study subsequently identified in one unrelated otocephalic patient a sporadic OTX2 mutation. Because OTX2 mutations may not be sufficient to cause otocephaly, the study assayed the potential of otx2 to modify craniofacial phenotypes in the context of known otocephaly gene suppression in vivo. It was found that otx2 can interact genetically with pgap1 , prrx1 , and msx1 to exacerbate mandibular and midline defects during zebrafish development. However, sequencing of these loci in the OTX2-positive families did not unearth likely pathogenic lesions, suggesting further genetic heterogeneity and complexity. Conclusion Identification of OTX2 involvement in otocephaly/dysgnathia in humans, even if loss of function mutations at this locus does not sufficiently explain the complex anatomical defects of these patients, suggests the requirement for a second genetic hit. Consistent with this notion, trans suppression of otx2 and other developmentally related genes recapitulate aspects of the otocephaly phenotype in zebrafish. This study highlights the combined utility of genetics and functional approaches to dissect both the regulatory pathways that govern craniofacial development and the genetics of this disease group.
- Published
- 2012
- Full Text
- View/download PDF
38. Primary culture of chick, mouse or human neural crest cells
- Author
-
Heather C. Etchevers
- Subjects
animal structures ,Cellular differentiation ,Population ,Cell Culture Techniques ,Chick Embryo ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Embryo Culture Techniques ,Mice ,Cell Movement ,medicine ,Animals ,Humans ,Cell Lineage ,education ,education.field_of_study ,Neural tube ,Neural crest ,Cell Differentiation ,Embryo ,Embryonic stem cell ,Cell biology ,medicine.anatomical_structure ,Neural Crest ,Cell culture ,Karyotyping ,embryonic structures ,Immunology ,Developmental biology - Abstract
A highly enriched population of neural crest cells (NCCs) from amniote embryos, such as from chicks, mice and humans, is desirable for experiments in fate determination. NCCs are also useful for testing the functional effects of molecular changes underlying numerous human diseases of neural crest derivatives and for investigating their potential for therapeutic compensation. This protocol details embryonic microdissection followed by neural tube explantation. Conditions favoring NCC expansion and the maintenance of their stem cell-like properties are described. Although neural crest-like cells can be derived from a number of sites in the mature organism, full potential is best ensured by their purification from their source tissue at the outset of migration. Going from embryo to established cell line takes 4 d; the first is the most labor-intensive day, but minimal intervention is required thereafter.
- Published
- 2011
- Full Text
- View/download PDF
39. Meeting report from the 2011 international expert meeting on large congenital melanocytic nevi and neurocutaneous melanocytosis, Tübingen
- Author
-
Mark Beckwith, Heather C. Etchevers, Sven Krengel, and Helmut Breuninger
- Subjects
medicine.medical_specialty ,Congenital diseases ,business.industry ,education ,Dermatology ,medicine.disease ,Patient advocacy ,humanities ,General Biochemistry, Genetics and Molecular Biology ,Oncology ,Family medicine ,Pediatric surgery ,Medicine ,Nevus ,Pediatric Neurology ,business ,Neurocutaneous melanocytosis - Abstract
An unconventional symposium on the subject of pathogenetic, clinical, and therapeutic aspects of large and giant congenital melanocytic nevi and neurocutaneous melanocytosis, was held at the University of Tubingen, Germany, on May 6-7, 2011. Exchanges were made between physicians from a wide range of clinical disciplines, including pathology, dermatology, plastic and pediatric surgery, neurosurgery, pediatric neurology and genetics; basic scientists in cell and developmental biology; psychologists; and an unprecedented gathering of international patient advocacy group representatives. This diversity of outlooks brought fresh perspectives to the discussions about current scientific and therapeutic advances in the field of these rare congenital diseases and their oncogenic associations. A roadmap for future actions sketched out promising therapeutic developments to follow and fostering of interdisciplinary collaboration among all the involved parties.
- Published
- 2011
- Full Text
- View/download PDF
40. Phenotypic spectrum ofSTRA6mutations: from Matthew-Wood syndrome to non-lethal anophthalmia
- Author
-
Nicolas Chassaing, Christelle Golzio, Heather C. Etchevers, Leopoldine Lequeux, Adeline Vigouroux, Sylvie Odent, Tania Attié-Bitach, Lucia Masini, Sylvie Manouvrier-Hanu, Patrick Calvas, Giovanna Maragliano, Francesca Piro, Francesco Danilo Tiziano, Jelena Martinovic-Bouriel, and Anne-Lise Delezoide
- Subjects
Genetics ,0303 health sciences ,Pediatrics ,medicine.medical_specialty ,Anophthalmia ,Biology ,medicine.disease ,Phenotype ,Microphthalmia ,3. Good health ,03 medical and health sciences ,Retinol binding protein ,Dysgenesis ,0302 clinical medicine ,medicine ,Diaphragmatic hernia ,Matthew Wood syndrome ,030217 neurology & neurosurgery ,Genetics (clinical) ,030304 developmental biology ,Tetralogy of Fallot - Abstract
Matthew-Wood, Spear, PDAC or MCOPS9 syndrome are alternative names used to refer to combinations of microphthalmia/anophthalmia, malformative cardiac defects, pulmonary dysgenesis, and diaphragmatic hernia. Recently, mutations in STRA6, encoding a membrane receptor for vitamin A-bearing plasma retinol binding protein, have been identified in such patients. We performed STRA6 molecular analysis in three fetuses and one child diagnosed with Matthew-Wood syndrome and in three siblings where two adult living brothers are affected with combinations of clinical anophthalmia, tetralogy of Fallot, and mental retardation. Among these patients, six novel mutations were identified, bringing the current total of known STRA6 mutations to seventeen. We extensively reviewed clinical data pertaining to all twenty-one reported patients with STRA6 mutations (the seven of this report and fourteen described elsewhere) and discuss additional features that may be part of the syndrome. The clinical spectrum associated with STRA6 deficiency is even more variable than initially described.
- Published
- 2009
- Full Text
- View/download PDF
41. Analysis of mouse models carrying the I26T and R160C substitutions in the transcriptional repressor HESX1 as models for septo-optic dysplasia and hypopituitarism
- Author
-
Juan Pedro Martinez-Barbera, Carles Gaston-Massuet, Massimo Signore, Sandra B.R. Castro, Heather C. Etchevers, Dianne Gerrelli, Ezat Sajedi, Mehul T. Dattani, Daniel Kelberman, and Cynthia L. Andoniadou
- Subjects
medicine.medical_specialty ,Mutant ,Neuroscience (miscellaneous) ,Medicine (miscellaneous) ,Repressor ,Locus (genetics) ,Hypopituitarism ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Prosencephalon ,Septo-Optic Dysplasia ,Immunology and Microbiology (miscellaneous) ,Internal medicine ,medicine ,Animals ,Body Patterning ,Homeodomain Proteins ,Optic nerve hypoplasia ,Septo-optic dysplasia ,medicine.disease ,Null allele ,Molecular biology ,Repressor Proteins ,Disease Models, Animal ,Endocrinology ,Mutation ,Forebrain ,Research Article - Abstract
SUMMARY A homozygous substitution of the highly conserved isoleucine at position 26 by threonine (I26T) in the transcriptional repressor HESX1 has been associated with anterior pituitary hypoplasia in a human patient, with no forebrain or eye defects. Two individuals carrying a homozygous substitution of the conserved arginine at position 160 by cysteine (R160C) manifest septo-optic dysplasia (SOD), a condition characterised by pituitary abnormalities associated with midline telencephalic structure defects and optic nerve hypoplasia. We have generated two knock-in mouse models containing either the I26T or R160C substitution in the genomic locus. Hesx1I26T/I26T embryos show pituitary defects comparable with Hesx1−/− mouse mutants, with frequent occurrence of ocular abnormalities, although the telencephalon develops normally. Hesx1R160C/R160C mutants display forebrain and pituitary defects that are identical to those observed in Hesx1−/− null mice. We also show that the expression pattern of HESX1 during early human development is very similar to that described in the mouse, suggesting that the function of HESX1 is conserved between the two species. Together, these results suggest that the I26T mutation yields a hypomorphic allele, whereas R160C produces a null allele and, consequently, a more severe phenotype in both mice and humans.
- Published
- 2008
- Full Text
- View/download PDF
42. Confirmation of RAX gene involvement in human anophthalmia
- Author
-
Heather C. Etchevers, François Malecaze, Nicolas Chassaing, Marlène Rio, Armelle Vigouroux, Patrick Calvas, Leopoldine Lequeux, and Matthias Titeux
- Subjects
endocrine system ,congenital, hereditary, and neonatal diseases and abnormalities ,Molecular Sequence Data ,Nonsense mutation ,Biology ,Compound heterozygosity ,Microphthalmia ,Article ,Cornea ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Anophthalmos ,Genetics ,medicine ,Humans ,Sclerocornea ,Eye Proteins ,Genetics (clinical) ,030304 developmental biology ,Homeodomain Proteins ,0303 health sciences ,Anophthalmia ,Sequence Analysis, DNA ,medicine.disease ,eye diseases ,3. Good health ,Child, Preschool ,Eye development ,Female ,Orbit ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Microphthalmia and anophthalmia are at the severe end of the spectrum of abnormalities in ocular development. Mutations in several genes have been involved in syndromic and non-syndromic anophthalmia. Previously, RAX recessive mutations were implicated in a single patient with right anophthalmia, left microphthalmia and sclerocornea. In this study, we report the findings of novel compound heterozygous RAX mutations in a child with bilateral anophthalmia. Both mutations are located in exon 3. c.664delT is a frameshifting deletion predicted to introduce a premature stop codon (p.Ser222ArgfsX62), and c.909C>G is a nonsense mutation with similar consequences (p.Tyr303X). This is the second report of a patient with anophthalmia caused by RAX mutations. These findings confirm that RAX plays a major role in the early stages of eye development and is involved in human anophthalmia.
- Published
- 2008
- Full Text
- View/download PDF
43. Human neural crest cells display molecular and phenotypic hallmarks of stem cells
- Author
-
Patrick Wincker, Pu-Ting Xu, Sophie Thomas, Michel Vekemans, Marie K Thomas, Arnold Munnich, Marcy C. Speer, Candice Babarit, Heather C. Etchevers, and Stanislas Lyonnet
- Subjects
Homeobox protein NANOG ,Cell type ,Mesenchyme ,Biology ,Cell Line ,SOXC Transcription Factors ,Transcriptome ,Mice ,SOX2 ,Genetics ,medicine ,Animals ,Cluster Analysis ,Humans ,Molecular Biology ,Embryonic Stem Cells ,Genetics (clinical) ,Gene Expression Profiling ,Gene Expression Regulation, Developmental ,Reproducibility of Results ,Neural crest ,Articles ,General Medicine ,Embryo, Mammalian ,Embryonic stem cell ,Cell biology ,DNA-Binding Proteins ,Embryo Research ,Phenotype ,medicine.anatomical_structure ,Neural Crest ,Connexin 43 ,Stem cell ,Biomarkers ,Transcription Factors - Abstract
The fields of both developmental and stem cell biology explore how functionally distinct cell types arise from a self-renewing founder population. Multipotent, proliferative human neural crest cells (hNCC) develop toward the end of the first month of pregnancy. It is assumed that most differentiate after migrating throughout the organism, although in animal models neural crest stem cells reportedly persist in postnatal tissues. Molecular pathways leading over time from an invasive mesenchyme to differentiated progeny such as the dorsal root ganglion, the maxillary bone or the adrenal medulla are altered in many congenital diseases. To identify additional components of such pathways, we derived and maintained self-renewing hNCC lines from pharyngulas. We show that, unlike their animal counterparts, hNCC are able to self-renew ex vivo under feeder-free conditions. While cross species comparisons showed extensive overlap between human, mouse and avian NCC transcriptomes, some molecular cascades are only active in the human cells, correlating with phenotypic differences. Furthermore, we found that the global hNCC molecular profile is highly similar to that of pluripotent embryonic stem cells when compared with other stem cell populations or hNCC derivatives. The pluripotency markers NANOG, POU5F1 and SOX2 are also expressed by hNCC, and a small subset of transcripts can unambiguously identify hNCC among other cell types. The hNCC molecular profile is thus both unique and globally characteristic of uncommitted stem cells.
- Published
- 2008
- Full Text
- View/download PDF
44. Bases génétiques et moléculaires des neurocristopathies
- Author
-
Jeanne Amiel, Heather C. Etchevers, and Stanislas Lyonnet
- Subjects
business.industry ,Pediatrics, Perinatology and Child Health ,Medicine ,business - Published
- 2007
- Full Text
- View/download PDF
45. Matthew-Wood Syndrome Is Caused by Truncating Mutations in the Retinol-Binding Protein Receptor Gene STRA6
- Author
-
Michel Vekemans, Christelle Golzio, Bettina Grattagliano-Bessières, Jelena Martinovic-Bouriel, Arnold Munnich, Sophie Delahaye, Tania Attié-Bitach, Stanislas Lyonnet, Heather C. Etchevers, Soumaya Mougou-Zrelli, Maryse Bonnière, Férechté Encha-Razavi, Sophie Thomas, IFR Necker-Enfants Malades (IRNEM), Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Génétique et épigénétique des maladies métaboliques, neurosensorielles et du développement (Inserm U781), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Necker - Enfants Malades [AP-HP], Institut de Puériculture de Paris (IPP), Assistance publique - Hôpitaux de Paris (AP-HP) (APHP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), IFR Necker-Enfants Malades ( IRNEM ), Assistance publique - Hôpitaux de Paris (AP-HP)-Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), Génétique et épigénétique des maladies métaboliques, neurosensorielles et du développement ( Inserm U781 ), Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ), Institut de Puériculture de Paris ( IPP ), and Assistance publique - Hôpitaux de Paris (AP-HP)
- Subjects
Lung Diseases ,MESH: Sequence Analysis, DNA ,Vitamin A transport ,MESH : Polymorphism, Genetic ,MESH : Receptors, Cell Surface ,MESH : Gene Deletion ,MESH: Genetic Markers ,MESH : Syndrome ,MESH : Frameshift Mutation ,MESH : Microphthalmos ,Consanguinity ,MESH: Lung Diseases ,Exon ,MESH : Exons ,0302 clinical medicine ,Microphthalmos ,MESH : Genetic Markers ,MESH: Syndrome ,Genetics(clinical) ,Frameshift Mutation ,Genetics (clinical) ,MESH: Receptors, Cell Surface ,0303 health sciences ,Fetal Growth Retardation ,MESH : Lung Diseases ,Homozygote ,MESH: Frameshift Mutation ,Exons ,Syndrome ,Pedigree ,MESH: Membrane Proteins ,MESH : Mutation ,MESH: Homozygote ,Genetic Markers ,MESH: Abnormalities, Multiple ,MESH: Mutation ,MESH: Pedigree ,MESH: Microphthalmos ,MESH: Fetal Growth Retardation ,Receptors, Cell Surface ,Biology ,Frameshift mutation ,03 medical and health sciences ,Pulmonary hypoplasia ,MESH : Homozygote ,Report ,MESH : Consanguinity ,MESH: Polymorphism, Genetic ,Genetics ,medicine ,Humans ,Abnormalities, Multiple ,MESH : Fetal Growth Retardation ,MESH : Haplotypes ,030304 developmental biology ,MESH: Consanguinity ,MESH : Mutagenesis, Insertional ,Polymorphism, Genetic ,MESH: Humans ,Anophthalmia ,MESH : Abnormalities, Multiple ,Genetic heterogeneity ,MESH : Humans ,Pulmonary Agenesis ,Membrane Proteins ,Sequence Analysis, DNA ,MESH: Haplotypes ,medicine.disease ,Molecular biology ,Mutagenesis, Insertional ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,Haplotypes ,MESH: Mutagenesis, Insertional ,MESH : Membrane Proteins ,MESH: Gene Deletion ,MESH : Pedigree ,[ SDV.BDD.EO ] Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,Mutation ,MESH: Exons ,Matthew Wood syndrome ,Gene Deletion ,030217 neurology & neurosurgery ,MESH : Sequence Analysis, DNA - Abstract
International audience; Retinoic acid (RA) is a potent teratogen in all vertebrates when tight homeostatic controls on its endogenous dose, location, or timing are perturbed during early embryogenesis. STRA6 encodes an integral cell-membrane protein that favors RA uptake from soluble retinol-binding protein; its transcription is directly regulated by RA levels. Molecular analysis of STRA6 was undertaken in two human fetuses from consanguineous families we previously described with Matthew-Wood syndrome in a context of severe microphthalmia, pulmonary agenesis, bilateral diaphragmatic eventration, duodenal stenosis, pancreatic malformations, and intrauterine growth retardation. The fetuses had either a homozygous insertion/deletion in exon 2 or a homozygous insertion in exon 7 predicting a premature stop codon in STRA6 transcripts. Five other fetuses presenting at least one of the two major signs of clinical anophthalmia or pulmonary hypoplasia with at least one of the two associated signs of diaphragmatic closure defect or cardiopathy had no STRA6 mutations. These findings suggest a molecular basis for the prenatal manifestations of Matthew-Wood syndrome and suggest that phenotypic overlap with other associations may be due to genetic heterogeneity of elements common to the RA- and fibroblast growth factor-signaling cascades.
- Published
- 2007
- Full Text
- View/download PDF
46. Matthew-Wood syndrome: Report of two new cases supporting autosomal recessive inheritance and exclusion ofFGF10 andFGFR2
- Author
-
Catherine Fallet-Bianco, Michel Vekemans, Chantal Esculpavit, Jerôme Le Bidois, Véronique Mirlesse, Alexandra Benachi, Christelle Golzio, Nicole Morichon, Férechté Encha-Razavi, Bettina Grattagliano-Bessières, Heather C. Etchevers, Maryse Bonnière, Valérie Malan, Jelena Martinovic-Bouriel, Céline Bernabé-Dupont, Tania Attié-Bitach, and Marie-Cécile Aubry
- Subjects
Adult ,Male ,Chromosome Disorders ,Genes, Recessive ,Prenatal diagnosis ,Biology ,Microphthalmia ,Pulmonary hypoplasia ,Pregnancy ,Prenatal Diagnosis ,Genetics ,medicine ,Humans ,Microphthalmos ,Abnormalities, Multiple ,Receptor, Fibroblast Growth Factor, Type 2 ,Lung ,Genetics (clinical) ,Anophthalmia ,Pulmonary Agenesis ,Anophthalmos ,Syndrome ,medicine.disease ,Phenotype ,Hypoplasia ,Female ,Fibroblast Growth Factor 10 ,Matthew Wood syndrome - Abstract
We describe two fetal cases of microphthalmia/anophthalmia, pulmonary agenesis, and diaphragmatic defect. This rare association is known as Matthew-Wood syndrome (MWS; MIM 601186) or by the acronym “PMD” (Pulmonary agenesis, Microphthalmia, Diaphragmatic defect). Fewer than ten pre- and perinatal diagnoses of Matthew-Wood syndrome have been described to date. The cause is unknown, and the mode of transmission remains unclear. Most cases have been reported as isolated and sporadic, although recurrence among sibs has been observed once. Our two cases both occurred in consanguineous families, further supporting autosomal recessive transmission. In addition, in one family at least one of the elder sibs presented an evocatively similar phenotype. The spatiotemporal expression pattern of the FGF10 and FGFR2 genes in human embryos and the reported phenotypes of knockout mice for these genes spurred us to examine their coding sequences in our two cases of MWS. While in our patients, no causative sequence variations were identified in FGF10 or FGFR2, this cognate ligand-receptor pair and its downstream effectors remain functional candidates for MWS and similar associations of congenital ocular, diaphragmatic and pulmonary malformations. © 2007 Wiley-Liss, Inc.
- Published
- 2007
- Full Text
- View/download PDF
47. Cardiac outflow morphogenesis depends on effects of retinoic acid signaling on multiple cell lineages
- Author
-
Nicolas, El Robrini, Heather C, Etchevers, Lucile, Ryckebüsch, Emilie, Faure, Nathalie, Eudes, Karen, Niederreither, Stéphane, Zaffran, and Nicolas, Bertrand
- Subjects
Mice, Knockout ,Mice ,Organogenesis ,Animals ,Cell Lineage ,Heart ,Tretinoin ,Aldehyde Oxidoreductases ,Signal Transduction - Abstract
Retinoic acid (RA), the bioactive derivative of vitamin A, is essential for vertebrate heart development. Both excess and reduced RA signaling lead to cardiovascular malformations affecting the outflow tract (OFT). To address the cellular mechanisms underlying the effects of RA signaling during OFT morphogenesis, we used transient maternal RA supplementation to rescue the early lethality resulting from inactivation of the murine retinaldehyde dehydrogenase 2 (Raldh2) gene.By embryonic day 13.5, all rescued Raldh2(-/-) hearts exhibit severe, reproducible OFT septation defects, although wild-type and Raldh2(+/-) littermates have normal hearts. Cardiac neural crest cells (cNCC) were present in OFT cushions of Raldh2(-/-) mutant embryos but ectopically located in the periphery of the endocardial cushions, rather than immediately underlying the endocardium. Excess mesenchyme was generated by Raldh2(-/-) mutant endocardium, which displaced cNCC derivatives from their subendocardial, medial position.RA signaling affects not only cNCC numbers but also their position relative to endocardial mesenchyme during the septation process. Our study shows that inappropriate coordination between the different cell types of the OFT perturbs its morphogenesis and leads to a severe congenital heart defect, persistent truncus arteriosus.
- Published
- 2015
48. Phenotypic spectrum of CHARGE syndrome in fetuses with CHD7 truncating mutations correlates with expression during human development
- Author
-
N. Joye, Tania Attié-Bitach, Jelena Martinovic, Damien Sanlaville, Chantal Esculpavit, Catherine Ozilou, Marie-Cécile Aubry, Heather C. Etchevers, Arnold Munnich, Jeanne Amiel, Corinne Cruaud, Catherine Fredouille, Sophie Audollent, Michel Vekemans, Anna Pelet, Nicole Morichon-Delvallez, Yves Dumez, Stanislas Lyonnet, Mathieu Clément-Ziza, Anne-Lise Delezoide, Sophie Chemouny, Marie Gonzales, Férechté Encha-Razavi, Jean Weissenbach, and Géraldine Goudefroye
- Subjects
medicine.medical_specialty ,Pathology ,Heart malformation ,Embryonic Development ,Prenatal diagnosis ,Choanal atresia ,Biology ,CHARGE syndrome ,Pregnancy ,Prenatal Diagnosis ,Molecular genetics ,Genetics ,medicine ,Humans ,Abnormalities, Multiple ,Promoter Regions, Genetic ,In Situ Hybridization ,Genetics (clinical) ,DNA Primers ,Sequence Deletion ,Coloboma ,Base Sequence ,DNA Helicases ,Neural crest ,DNA ,Syndrome ,medicine.disease ,DNA-Binding Proteins ,Fetal Diseases ,Phenotype ,Agenesis ,Mutation ,Female ,Original Article - Abstract
Background: The acronym CHARGE refers to a non-random cluster of malformations including coloboma, heart malformation, choanal atresia, retardation of growth and/or development, genital anomalies, and ear anomalies. This set of multiple congenital anomalies is frequent, despite rare patients with normal intelligence, and prognosis remains poor. Recently, CHD7 gene mutations have been identified in CHARGE patients; however, the function of CHD7 during development remains unknown. Methods: We studied a series of 10 antenatal cases in whom the diagnosis of CHARGE syndrome was suspected, considering that a careful pathological description would shed light on the CHD7 function during development. CHD7 sequence analysis and in situ hybridisation were employed. Results: The diagnosis of CHARGE syndrome was confirmed in all 10 fetuses by the identification of a CHD7 heterozygous truncating mutation. Interestingly, arhinencephaly and semi-circular canal agenesis were two constant features which are not included in formal diagnostic criteria so far. In situ hybridisation analysis of the CHD7 gene during early human development emphasised the role of CHD7 in the development of the central nervous system, internal ear, and neural crest of pharyngeal arches, and more generally showed a good correlation between specific CHD7 expression pattern and the developmental anomalies observed in CHARGE syndrome. Conclusions: These results allowed us to further refine the phenotypic spectrum of developmental anomalies resulting from CHD7 dysfunction.
- Published
- 2005
- Full Text
- View/download PDF
49. Antenatal Presentation of Bardet-Biedl Syndrome May Mimic Meckel Syndrome
- Author
-
Géraldine Goudefroye, Marie-Odile Peter, Philippe Loget, Heather C. Etchevers, Dominique Gaillard, Houda Karmous-Benailly, Marie Gonzales, Madeleine Joubert, Ghislaine Plessis, Martine Le Merrer, Férechté Encha-Razavi, Marie-Claire Gubler, Joelle Augé, Chantal Esculpavit, Nora Brahimi, Eric Detrait, Catherine Ozilou, Yoann Sirot, Tania Attié-Bitach, B. Simon-Bouy, Michel Vekemans, Julia Tantau, Hélène Dollfus, Arnold Munnich, Sophie Audollent, Laure Clech, Jelena Martinovic, Corinne Jeanne-Pasquier, and Anne-Lise Delezoide
- Subjects
Male ,BBS2 ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Pathology ,BBS1 ,Heart malformation ,DNA Mutational Analysis ,BBS5 ,Kidney ,Diagnosis, Differential ,Fetus ,Bardet–Biedl syndrome ,Pregnancy ,Prenatal Diagnosis ,Internal medicine ,medicine ,Genetics ,Humans ,Abnormalities, Multiple ,Genetics(clinical) ,Meckel syndrome ,Bardet-Biedl Syndrome ,Genetics (clinical) ,Encephalocele ,Cystic kidney ,Base Sequence ,Polydactyly ,business.industry ,Infant, Newborn ,Articles ,Syndrome ,medicine.disease ,Pedigree ,Endocrinology ,Liver ,Female ,business - Abstract
Bardet-Biedl syndrome (BBS) is a multisystemic disorder characterized by postaxial polydactyly, progressive retinal dystrophy, obesity, hypogonadism, renal dysfunction, and learning difficulty. Other manifestations include diabetes mellitus, heart disease, hepatic fibrosis, and neurological features. The condition is genetically heterogeneous, and eight genes (BBS1-BBS8) have been identified to date. A mutation of the BBS1 gene on chromosome 11q13 is observed in 30%-40% of BBS cases. In addition, a complex triallelic inheritance has been established in this disorder--that is, in some families, three mutations at two BBS loci are necessary for the disease to be expressed. The clinical features of BBS that can be observed at birth are polydactyly, kidney anomaly, hepatic fibrosis, and genital and heart malformations. Interestingly, polydactyly, cystic kidneys, and liver anomalies (hepatic fibrosis with bile-duct proliferation) are also observed in Meckel syndrome, along with occipital encephalocele. Therefore, we decided to sequence the eight BBS genes in a series of 13 antenatal cases presenting with cystic kidneys and polydactyly and/or hepatic fibrosis but no encephalocele. These fetuses were mostly diagnosed as having Meckel or "Meckel-like" syndrome. In six cases, we identified a recessive mutation in a BBS gene (three in BBS2, two in BBS4, and one in BBS6). We found a heterozygous BBS6 mutation in three additional cases. No BBS1, BBS3, BBS5, BBS7, or BBS8 mutations were identified in our series. These results suggest that the antenatal presentation of BBS may mimic Meckel syndrome.
- Published
- 2005
- Full Text
- View/download PDF
50. PAX8,TITF1, andFOXE1Gene Expression Patterns during Human Development: New Insights into Human Thyroid Development and Thyroid Dysgenesis-Associated Malformations
- Author
-
Tania Attié-Bitach, Paul Czernichow, Sylvia Sura Trueba, Joelle Augé, Michel Vekemans, Jélééna Martinovic, Heather C. Etchevers, Michel Polak, and Geraldine Mattei
- Subjects
Thyroid Nuclear Factor 1 ,medicine.medical_specialty ,Pathology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Clinical Biochemistry ,Thyroid Gland ,Embryonic Development ,Gene Expression ,Biology ,Thyroglobulin ,Biochemistry ,Thyroid dysgenesis ,Fetal Development ,Mice ,PAX8 Transcription Factor ,Endocrinology ,Internal medicine ,medicine ,Animals ,Humans ,Paired Box Transcription Factors ,Biochemistry (medical) ,Thyroid ,Nuclear Proteins ,Forkhead Transcription Factors ,medicine.disease ,Immunohistochemistry ,Congenital hypothyroidism ,DNA-Binding Proteins ,Repressor Proteins ,medicine.anatomical_structure ,Ureteric bud ,Trans-Activators ,PAX8 ,Transcription Factors ,FOXE1 - Abstract
Thyroid dysgenesis (TD) is responsible for most cases of congenital hypothyroidism, a condition that affects about one in 4000 newborns. Mutations in PAX8, TITF1, or FOXE1 may account for congenital hypothyroidism in patients with either isolated TD or TD with associated malformations involving kidney, lung, forebrain, and palate. Pax8, titf1, and foxe1 are expressed in the mouse thyroid bud as soon as it differentiates on the pharyngeal floor. Because the spatio-temporal expression of these genes is unknown in humans, we decided to study them at different stages of human embryonic and fetal development. PAX8 and TITF1 were first expressed in the median thyroid primordium. Interestingly, PAX8 was also expressed in the thyroglossal duct and the ultimobranchial bodies. Human FOXE1 expression was detected later than in the mouse. PAX8 was also expressed in the developing central nervous system and kidney, including the ureteric bud and the main collecting ducts. TITF1 was expressed in the ventral forebrain and lung. FOXE1 expression was detected in the oropharyngeal epithelium and thymus. In conclusion, the expression patterns described here show some differences from those reported in the mouse. They explain the malformations associated with TD in patients carrying PAX8, TITF1, and FOXE1 gene mutations.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.