1
|
Van Haver S, Fan Y, Bekaert SL, Everaert C, Van Loocke W, Zanzani V, Deschildre J, Maestre IF, Amaro A, Vermeirssen V, De Preter K, Zhou T, Kentsis A, Studer L, Speleman F, Roberts SS. Human iPSC modeling recapitulates in vivo sympathoadrenal development and reveals an aberrant developmental subpopulation in familial neuroblastoma. iScience 2024; 27:108096. [PMID: 38222111 PMCID: PMC10784699 DOI: 10.1016/j.isci.2023.108096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/12/2023] [Accepted: 09/26/2023] [Indexed: 01/16/2024] Open
Abstract
Studies defining normal and disrupted human neural crest cell development have been challenging given its early timing and intricacy of development. Consequently, insight into the early disruptive events causing a neural crest related disease such as pediatric cancer neuroblastoma is limited. To overcome this problem, we developed an in vitro differentiation model to recapitulate the normal in vivo developmental process of the sympathoadrenal lineage which gives rise to neuroblastoma. We used human in vitro pluripotent stem cells and single-cell RNA sequencing to recapitulate the molecular events during sympathoadrenal development. We provide a detailed map of dynamically regulated transcriptomes during sympathoblast formation and illustrate the power of this model to study early events of the development of human neuroblastoma, identifying a distinct subpopulation of cell marked by SOX2 expression in developing sympathoblast obtained from patient derived iPSC cells harboring a germline activating mutation in the anaplastic lymphoma kinase (ALK) gene.
Collapse
Affiliation(s)
- Stéphane Van Haver
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Yujie Fan
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Developmental Biology Program, MSKCC, New York, NY 10065, USA
- Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10065, USA
| | - Sarah-Lee Bekaert
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Celine Everaert
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Wouter Van Loocke
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Vittorio Zanzani
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Joke Deschildre
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Inés Fernandez Maestre
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adrianna Amaro
- Department of Pediatrics, MSKCC, New York, NY 10065, USA
| | - Vanessa Vermeirssen
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Katleen De Preter
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | - Ting Zhou
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Alex Kentsis
- Department of Pediatrics, MSKCC, New York, NY 10065, USA
- Molecular Pharmacology Program, MSKCC, New York, NY, USA
- Tow Center for Developmental Oncology, MSKCC, New York, NY 10065, USA
- Departments of Pediatrics, Pharmacology and Physiology & Biophysics, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA
- Developmental Biology Program, MSKCC, New York, NY 10065, USA
| | - Frank Speleman
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium
| | | |
Collapse
|
2
|
Brandon AA, Almeida D, Powder KE. Neural crest cells as a source of microevolutionary variation. Semin Cell Dev Biol 2023; 145:42-51. [PMID: 35718684 PMCID: PMC10482117 DOI: 10.1016/j.semcdb.2022.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 05/03/2022] [Accepted: 06/03/2022] [Indexed: 11/28/2022]
Abstract
Vertebrates have some of the most complex and diverse features in animals, from varied craniofacial morphologies to colorful pigmentation patterns and elaborate social behaviors. All of these traits have their developmental origins in a multipotent embryonic lineage of neural crest cells. This "fourth germ layer" is a vertebrate innovation and the source of a wide range of adult cell types. While others have discussed the role of neural crest cells in human disease and animal domestication, less is known about their role in contributing to adaptive changes in wild populations. Here, we review how variation in the development of neural crest cells and their derivatives generates considerable phenotypic diversity in nature. We focus on the broad span of traits under natural and sexual selection whose variation may originate in the neural crest, with emphasis on behavioral factors such as intraspecies communication that are often overlooked. In all, we encourage the integration of evolutionary ecology with developmental biology and molecular genetics to gain a more complete understanding of the role of this single cell type in trait covariation, evolutionary trajectories, and vertebrate diversity.
Collapse
Affiliation(s)
- A Allyson Brandon
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | - Daniela Almeida
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | - Kara E Powder
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA.
| |
Collapse
|
3
|
Bauer MB, Currie KPM. Serotonin and the serotonin transporter in the adrenal gland. VITAMINS AND HORMONES 2023; 124:39-78. [PMID: 38408804 PMCID: PMC11217909 DOI: 10.1016/bs.vh.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
The adrenal glands are key components of the mammalian endocrine system, helping maintain physiological homeostasis and the coordinated response to stress. Each adrenal gland has two morphologically and functionally distinct regions, the outer cortex and inner medulla. The cortex is organized into three concentric zones which secrete steroid hormones, including aldosterone and cortisol. Neural crest-derived chromaffin cells in the medulla are innervated by preganglionic sympathetic neurons and secrete catecholamines (epinephrine, norepinephrine) and neuropeptides into the bloodstream, thereby functioning as the neuroendocrine arm of the sympathetic nervous system. In this article we review serotonin (5-HT) and the serotonin transporter (SERT; SLC6A4) in the adrenal gland. In the adrenal cortex, 5-HT, primarily sourced from resident mast cells, acts as a paracrine signal to stimulate aldosterone and cortisol secretion through 5-HT4/5-HT7 receptors. Medullary chromaffin cells contain a small amount of 5-HT due to SERT-mediated uptake and express 5-HT1A receptors which inhibit secretion. The atypical mechanism of the 5-HT1A receptors and interaction with SERT fine tune this autocrine pathway to control stress-evoked catecholamine secretion. Receptor-independent signaling by SERT/intracellular 5-HT modulates the amount and kinetics of transmitter release from single vesicle fusion events. SERT might also influence stress-evoked upregulation of tyrosine hydroxylase transcription. Transient signaling via 5-HT3 receptors during embryonic development can limit the number of chromaffin cells found in the mature adrenal gland. Together, this emerging evidence suggests that the adrenal medulla is a peripheral hub for serotonergic control of the sympathoadrenal stress response.
Collapse
Affiliation(s)
- Mary Beth Bauer
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, South Broadway, Camden, NJ, United States
| | - Kevin P M Currie
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, South Broadway, Camden, NJ, United States.
| |
Collapse
|
4
|
Kameneva P, Melnikova VI, Kastriti ME, Kurtova A, Kryukov E, Murtazina A, Faure L, Poverennaya I, Artemov AV, Kalinina TS, Kudryashov NV, Bader M, Skoda J, Chlapek P, Curylova L, Sourada L, Neradil J, Tesarova M, Pasqualetti M, Gaspar P, Yakushov VD, Sheftel BI, Zikmund T, Kaiser J, Fried K, Alenina N, Voronezhskaya EE, Adameyko I. Serotonin limits generation of chromaffin cells during adrenal organ development. Nat Commun 2022; 13:2901. [PMID: 35614045 PMCID: PMC9133002 DOI: 10.1038/s41467-022-30438-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 04/23/2022] [Indexed: 11/12/2022] Open
Abstract
Adrenal glands are the major organs releasing catecholamines and regulating our stress response. The mechanisms balancing generation of adrenergic chromaffin cells and protecting against neuroblastoma tumors are still enigmatic. Here we revealed that serotonin (5HT) controls the numbers of chromaffin cells by acting upon their immediate progenitor "bridge" cells via 5-hydroxytryptamine receptor 3A (HTR3A), and the aggressive HTR3Ahigh human neuroblastoma cell lines reduce proliferation in response to HTR3A-specific agonists. In embryos (in vivo), the physiological increase of 5HT caused a prolongation of the cell cycle in "bridge" progenitors leading to a smaller chromaffin population and changing the balance of hormones and behavioral patterns in adulthood. These behavioral effects and smaller adrenals were mirrored in the progeny of pregnant female mice subjected to experimental stress, suggesting a maternal-fetal link that controls developmental adaptations. Finally, these results corresponded to a size-distribution of adrenals found in wild rodents with different coping strategies.
Collapse
Affiliation(s)
- Polina Kameneva
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Victoria I Melnikova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Maria Eleni Kastriti
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Anastasia Kurtova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Emil Kryukov
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Aliia Murtazina
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Irina Poverennaya
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Artem V Artemov
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- National Medical Research Center for Endocrinology, Moscow, Russia
| | - Tatiana S Kalinina
- Federal state budgetary institution "Research Zakusov Institute of Pharmacology" (FSBI "Zakusov Institute of Pharmacology"), Russian Academy of Sciences, Moscow, Russia
| | - Nikita V Kudryashov
- Federal state budgetary institution "Research Zakusov Institute of Pharmacology" (FSBI "Zakusov Institute of Pharmacology"), Russian Academy of Sciences, Moscow, Russia
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), 13125, Berlin-Buch, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
- Institute for Biology, University of Lübeck, 23562, Lübeck, Germany
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Petr Chlapek
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Lucie Curylova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Lukas Sourada
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Jakub Neradil
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Massimo Pasqualetti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
| | | | - Vasily D Yakushov
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Boris I Sheftel
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Natalia Alenina
- Max-Delbrück Center for Molecular Medicine (MDC), 13125, Berlin-Buch, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Elena E Voronezhskaya
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria.
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
5
|
Yaglova NV, Obernikhin SS, Tsomartova DA, Yaglov VV, Nazimova SV, Tsomartova ES, Timokhina EP, Chereshneva EV, Ivanova MY, Lomanovskaya TA. Impact of Prenatal and Postnatal Exposure to Endocrine Disrupter DDT on Adrenal Medulla Function. Int J Mol Sci 2022; 23:ijms23094912. [PMID: 35563302 PMCID: PMC9101091 DOI: 10.3390/ijms23094912] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 12/04/2022] Open
Abstract
Epinephrine is the most abundant catecholamine hormone, produced by the nervous system and adrenal glands. Endocrine disruption of epinephrine synthesis, secretion and signaling is less studied than steroid and thyroid hormones. Dichlorodiphenyltrichloroethane (DDT) is recognized as one of the most prominent environmental contaminants with a long half-life. It is a potent endocrine disrupter affecting sex steroid, mineralocorticoid, glucocorticoid and thyroid hormone production. Exposure to low doses of DDT is universal and begins in utero. Therefore, we studied adrenal medulla growth and function in male Wistar rats exposed to low doses of DDT during prenatal and postnatal development until puberty and adulthood, as well as rats exposed to DDT since the first day of postnatal development. All the exposed rats demonstrated lowered epinephrine blood levels, gradually reducing with age. DDT was found to inhibit the synthesis of tyrosine hydroxylase and affect the mitochondrial apparatus of epinephrine-producing cells during puberty and even after maturation. Low-dose exposure to DDT from birth resulted in more pronounced changes in adrenomedullary cells and a more profound decrease (up to 50%) in epinephrine secretion in adult rats. Prenatal onset of exposure demonstrated a mild effect on epinephrine-producing function (30% reduction), but was associated with lower rate of adrenal medulla growth during maturation and 25% smaller adrenal medullar size in adult rats. All subjects exposed to low doses of DDT failed to develop adaptive changes and restore proper epinephrine production. These results indicate a dysmorphogenetic effect of prenatal exposure and disruption of secretory function of adrenal chromaffin cells by postnatal exposure to DDT.
Collapse
Affiliation(s)
- Nataliya V. Yaglova
- Laboratory of Endocrine System Development, Research Institute of Human Morphology, FSBSI, Petrovsky National Research Centre of Surgery, 119991 Moscow, Russia; (S.S.O.); (D.A.T.); (V.V.Y.); (S.V.N.); (E.S.T.); (E.P.T.)
- Correspondence: ; Tel.: +7-499-120-04-79
| | - Sergey S. Obernikhin
- Laboratory of Endocrine System Development, Research Institute of Human Morphology, FSBSI, Petrovsky National Research Centre of Surgery, 119991 Moscow, Russia; (S.S.O.); (D.A.T.); (V.V.Y.); (S.V.N.); (E.S.T.); (E.P.T.)
| | - Dibakhan A. Tsomartova
- Laboratory of Endocrine System Development, Research Institute of Human Morphology, FSBSI, Petrovsky National Research Centre of Surgery, 119991 Moscow, Russia; (S.S.O.); (D.A.T.); (V.V.Y.); (S.V.N.); (E.S.T.); (E.P.T.)
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Valentin V. Yaglov
- Laboratory of Endocrine System Development, Research Institute of Human Morphology, FSBSI, Petrovsky National Research Centre of Surgery, 119991 Moscow, Russia; (S.S.O.); (D.A.T.); (V.V.Y.); (S.V.N.); (E.S.T.); (E.P.T.)
| | - Svetlana V. Nazimova
- Laboratory of Endocrine System Development, Research Institute of Human Morphology, FSBSI, Petrovsky National Research Centre of Surgery, 119991 Moscow, Russia; (S.S.O.); (D.A.T.); (V.V.Y.); (S.V.N.); (E.S.T.); (E.P.T.)
| | - Elina S. Tsomartova
- Laboratory of Endocrine System Development, Research Institute of Human Morphology, FSBSI, Petrovsky National Research Centre of Surgery, 119991 Moscow, Russia; (S.S.O.); (D.A.T.); (V.V.Y.); (S.V.N.); (E.S.T.); (E.P.T.)
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Ekaterina P. Timokhina
- Laboratory of Endocrine System Development, Research Institute of Human Morphology, FSBSI, Petrovsky National Research Centre of Surgery, 119991 Moscow, Russia; (S.S.O.); (D.A.T.); (V.V.Y.); (S.V.N.); (E.S.T.); (E.P.T.)
| | - Elizaveta V. Chereshneva
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Marina Y. Ivanova
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| | - Tatiana A. Lomanovskaya
- Department of Histology, Cytology, and Embryology, Federal State Funded Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia; (E.V.C.); (M.Y.I.); (T.A.L.)
| |
Collapse
|
6
|
Guérineau NC, Campos P, Le Tissier PR, Hodson DJ, Mollard P. Cell Networks in Endocrine/Neuroendocrine Gland Function. Compr Physiol 2022; 12:3371-3415. [PMID: 35578964 DOI: 10.1002/cphy.c210031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Reproduction, growth, stress, and metabolism are determined by endocrine/neuroendocrine systems that regulate circulating hormone concentrations. All these systems generate rhythms and changes in hormone pulsatility observed in a variety of pathophysiological states. Thus, the output of endocrine/neuroendocrine systems must be regulated within a narrow window of effective hormone concentrations but must also maintain a capacity for plasticity to respond to changing physiological demands. Remarkably most endocrinologists still have a "textbook" view of endocrine gland organization which has emanated from 20th century histological studies on thin 2D tissue sections. However, 21st -century technological advances, including in-depth 3D imaging of specific cell types have vastly changed our knowledge. We now know that various levels of multicellular organization can be found across different glands, that organizational motifs can vary between species and can be modified to enhance or decrease hormonal release. This article focuses on how the organization of cells regulates hormone output using three endocrine/neuroendocrine glands that present different levels of organization and complexity: the adrenal medulla, with a single neuroendocrine cell type; the anterior pituitary, with multiple intermingled cell types; and the pancreas with multiple intermingled cell types organized into distinct functional units. We give an overview of recent methodologies that allow the study of the different components within endocrine systems, particularly their temporal and spatial relationships. We believe the emerging findings about network organization, and its impact on hormone secretion, are crucial to understanding how homeostatic regulation of endocrine axes is carried out within endocrine organs themselves. © 2022 American Physiological Society. Compr Physiol 12:3371-3415, 2022.
Collapse
Affiliation(s)
| | - Pauline Campos
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Paul R Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.,COMPARE University of Birmingham and University of Nottingham Midlands, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Patrice Mollard
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
7
|
Wang Y, Guo B, Guo Y, Qi N, Lv Y, Ye Y, Huang Y, Long X, Chen H, Su C, Zhang L, Zhang Q, Li M, Liao J, Yan Y, Mao X, Zeng Y, Jiang J, Chen Z, Guo Y, Gao S, Cheng J, Jiang Y, Mo Z. A spatiotemporal steroidogenic regulatory network in human fetal adrenal glands and gonads. Front Endocrinol (Lausanne) 2022; 13:1036517. [PMID: 36465633 PMCID: PMC9713933 DOI: 10.3389/fendo.2022.1036517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022] Open
Abstract
Human fetal adrenal glands produce substantial amounts of dehydroepiandrosterone (DHEA), which is one of the most important precursors of sex hormones. However, the underlying biological mechanism remains largely unknown. Herein, we sequenced human fetal adrenal glands and gonads from 7 to 14 gestational weeks (GW) via 10× Genomics single-cell transcriptome techniques, reconstructed their location information by spatial transcriptomics. Relative to gonads, adrenal glands begin to synthesize steroids early. The coordination among steroidogenic cells and multiple non-steroidogenic cells promotes adrenal cortex construction and steroid synthesis. Notably, during the window of sexual differentiation (8-12 GW), key enzyme gene expression shifts to accelerate DHEA synthesis in males and cortisol synthesis in females. Our research highlights the robustness of the action of fetal adrenal glands on gonads to modify the process of sexual differentiation.
Collapse
Affiliation(s)
- Yifu Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Bingqian Guo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yajie Guo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Nana Qi
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yufang Lv
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yu Ye
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Emergency, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yan Huang
- Department of Obstetrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xinyang Long
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- School of Public Health of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
| | - Hongfei Chen
- Department of Obstetrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Cheng Su
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Liying Zhang
- Department of Gynecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qingyun Zhang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Minxi Li
- Department of Gynecology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Liao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yunkun Yan
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xingning Mao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yanyu Zeng
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Jinghang Jiang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhongyuan Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
| | - Yi Guo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shuai Gao
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiwen Cheng
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yonghua Jiang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-Association of Southeast Asian Nations (ASEAN) Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, Nanning, Guangxi, China
- Department of Obstetrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, China
- *Correspondence: Zengnan Mo, ; Yonghua Jiang,
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, Guangxi, China
- Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- *Correspondence: Zengnan Mo, ; Yonghua Jiang,
| |
Collapse
|
8
|
Chromosome Imbalances in Neuroblastoma-Recent Molecular Insight into Chromosome 1p-deletion, 2p-gain, and 11q-deletion Identifies New Friends and Foes for the Future. Cancers (Basel) 2021; 13:cancers13235897. [PMID: 34885007 PMCID: PMC8657310 DOI: 10.3390/cancers13235897] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Neuroblastoma is a pediatric cancer that arises in the sympathetic nervous system. High-risk neuroblastoma is clinically challenging and identification of novel therapies, particularly those that offer a reduction in morbidity for these patients, is a high priority. Combining genetic analyses with investigation of molecular mechanisms, while considering recent advances in our understanding of key developmental events, provides avenues for future treatment. Here we review and highlight several recently published articles that address novel molecular mechanisms arising from chromosome 1p, 2p, and 11q aberrations, which likely contribute to high-risk neuroblastoma, and discusses their potential impact on treatment options. Abstract Neuroblastoma is the most common extracranial solid pediatric tumor, with around 15% childhood cancer-related mortality. High-risk neuroblastomas exhibit a range of genetic, morphological, and clinical heterogeneities, which add complexity to diagnosis and treatment with existing modalities. Identification of novel therapies is a high priority in high-risk neuroblastoma, and the combination of genetic analysis with increased mechanistic understanding—including identification of key signaling and developmental events—provides optimism for the future. This focused review highlights several recent findings concerning chromosomes 1p, 2p, and 11q, which link genetic aberrations with aberrant molecular signaling output. These novel molecular insights contribute important knowledge towards more effective treatment strategies for neuroblastoma.
Collapse
|
9
|
Solovieva T, Bronner M. Reprint of: Schwann cell precursors: Where they come from and where they go. Cells Dev 2021; 168:203729. [PMID: 34456178 DOI: 10.1016/j.cdev.2021.203729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 10/20/2022]
Abstract
Schwann cell precursors (SCPs) are a transient population in the embryo, closely associated with nerves along which they migrate into the periphery of the body. Long considered to be progenitors that only form Schwann cells-the myelinating cells of nerves, current evidence suggests that SCPs have much broader developmental potential. Indeed, different cell marking techniques employed over the past 20 years have identified multiple novel SCP derivatives throughout the body. It is now clear that SCPs represent a multipotent progenitor population, which also display a level of plasticity in response to injury. Moreover, they originate from multiple origins in the embryo and may reflect several distinct subpopulations in terms of molecular identity and fate. Here we review SCP origins, derivatives and plasticity in development, growth and repair.
Collapse
Affiliation(s)
- Tatiana Solovieva
- Division of Biology and Biological Engineering, California Institute of Technology, United States of America.
| | - Marianne Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, United States of America
| |
Collapse
|
10
|
Kamenev D, Sunadome K, Shirokov M, Chagin AS, Singh A, Irion U, Adameyko I, Fried K, Dyachuk V. Schwann cell precursors generate sympathoadrenal system during zebrafish development. J Neurosci Res 2021; 99:2540-2557. [PMID: 34184294 DOI: 10.1002/jnr.24909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 11/07/2022]
Abstract
The autonomic portion of the peripheral nervous system orchestrates tissue homeostasis through direct innervation of internal organs, and via release of adrenalin and noradrenalin into the blood flow. The developmental mechanisms behind the formation of autonomic neurons and chromaffin cells are not fully understood. Using genetic tracing, we discovered that a significant proportion of sympathetic neurons in zebrafish originates from Schwann cell precursors (SCPs) during a defined period of embryonic development. Moreover, SCPs give rise to the main portion of the chromaffin cells, as well as to a significant proportion of enteric and other autonomic neurons associated with internal organs. The conversion of SCPs into neuronal and chromaffin cells is ErbB receptor dependent, as the pharmacological inhibition of the ErbB pathway effectively perturbed this transition. Finally, using genetic ablations, we revealed that SCPs producing neurons and chromaffin cells migrate along spinal motor axons to reach appropriate target locations. This study reveals the evolutionary conservation of SCP-to-neuron and SCP-to-chromaffin cell transitions over significant growth periods in fish and highlights relevant cellular-genetic mechanisms. Based on this, we anticipate that multipotent SCPs might be present in postnatal vertebrate tissues, retaining the capacity to regenerate autonomic neurons and chromaffin cells.
Collapse
Affiliation(s)
- Dmitrii Kamenev
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kazunori Sunadome
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Maxim Shirokov
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia
| | - Andrey S Chagin
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Ajeet Singh
- Max-Planck-Institut für Entwicklungsbiologie, Tübingen, Germany
| | - Uwe Irion
- Max-Planck-Institut für Entwicklungsbiologie, Tübingen, Germany
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Vyacheslav Dyachuk
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia
| |
Collapse
|
11
|
Siaw JT, Javanmardi N, Van den Eynden J, Lind DE, Fransson S, Martinez-Monleon A, Djos A, Sjöberg RM, Östensson M, Carén H, Trøen G, Beiske K, Berbegall AP, Noguera R, Lai WY, Kogner P, Palmer RH, Hallberg B, Martinsson T. 11q Deletion or ALK Activity Curbs DLG2 Expression to Maintain an Undifferentiated State in Neuroblastoma. Cell Rep 2021; 32:108171. [PMID: 32966799 DOI: 10.1016/j.celrep.2020.108171] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/09/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
High-risk neuroblastomas typically display an undifferentiated or poorly differentiated morphology. It is therefore vital to understand molecular mechanisms that block the differentiation process. We identify an important role for oncogenic ALK-ERK1/2-SP1 signaling in the maintenance of undifferentiated neural crest-derived progenitors through the repression of DLG2, a candidate tumor suppressor gene in neuroblastoma. DLG2 is expressed in the murine "bridge signature" that represents the transcriptional transition state when neural crest cells or Schwann cell precursors differentiate to chromaffin cells of the adrenal gland. We show that the restoration of DLG2 expression spontaneously drives neuroblastoma cell differentiation, highlighting the importance of DLG2 in this process. These findings are supported by genetic analyses of high-risk 11q deletion neuroblastomas, which identified genetic lesions in the DLG2 gene. Our data also suggest that further exploration of other bridge genes may help elucidate the mechanisms underlying the differentiation of NC-derived progenitors and their contribution to neuroblastomas.
Collapse
Affiliation(s)
- Joachim Tetteh Siaw
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Niloufar Javanmardi
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530 Gothenburg, Sweden
| | - Jimmy Van den Eynden
- Department of Human Structure and Repair, Anatomy and Embryology Unit, Ghent University, 9000 Ghent, Belgium
| | - Dan Emil Lind
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Susanne Fransson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530 Gothenburg, Sweden
| | - Angela Martinez-Monleon
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530 Gothenburg, Sweden
| | - Anna Djos
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530 Gothenburg, Sweden
| | - Rose-Marie Sjöberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530 Gothenburg, Sweden
| | - Malin Östensson
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Helena Carén
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Gunhild Trøen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Klaus Beiske
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Ana P Berbegall
- Department of Pathology, Medical School, University of Valencia/INCLIVA, Valencia/CIBER of Cancer, Madrid, Spain
| | - Rosa Noguera
- Department of Pathology, Medical School, University of Valencia/INCLIVA, Valencia/CIBER of Cancer, Madrid, Spain
| | - Wei-Yun Lai
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ruth H Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden.
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden.
| | - Tommy Martinsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530 Gothenburg, Sweden.
| |
Collapse
|
12
|
Solovieva T, Bronner M. Schwann cell precursors: Where they come from and where they go. Cells Dev 2021; 166:203686. [PMID: 33994354 DOI: 10.1016/j.cdev.2021.203686] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 11/30/2022]
Abstract
Schwann cell precursors (SCPs) are a transient population in the embryo, closely associated with nerves along which they migrate into the periphery of the body. Long considered to be progenitors that only form Schwann cells-the myelinating cells of nerves, current evidence suggests that SCPs have much broader developmental potential. Indeed, different cell marking techniques employed over the past 20 years have identified multiple novel SCP derivatives throughout the body. It is now clear that SCPs represent a multipotent progenitor population, which also display a level of plasticity in response to injury. Moreover, they originate from multiple origins in the embryo and may reflect several distinct subpopulations in terms of molecular identity and fate. Here we review SCP origins, derivatives and plasticity in development, growth and repair.
Collapse
Affiliation(s)
- Tatiana Solovieva
- Division of Biology and Biological Engineering, California Institute of Technology, United States of America.
| | - Marianne Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, United States of America
| |
Collapse
|
13
|
Kameneva P, Artemov AV, Kastriti ME, Faure L, Olsen TK, Otte J, Erickson A, Semsch B, Andersson ER, Ratz M, Frisén J, Tischler AS, de Krijger RR, Bouderlique T, Akkuratova N, Vorontsova M, Gusev O, Fried K, Sundström E, Mei S, Kogner P, Baryawno N, Kharchenko PV, Adameyko I. Single-cell transcriptomics of human embryos identifies multiple sympathoblast lineages with potential implications for neuroblastoma origin. Nat Genet 2021; 53:694-706. [PMID: 33833454 PMCID: PMC7610777 DOI: 10.1038/s41588-021-00818-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 02/16/2021] [Indexed: 02/01/2023]
Abstract
Characterization of the progression of cellular states during human embryogenesis can provide insights into the origin of pediatric diseases. We examined the transcriptional states of neural crest- and mesoderm-derived lineages differentiating into adrenal glands, kidneys, endothelium and hematopoietic tissue between post-conception weeks 6 and 14 of human development. Our results reveal transitions connecting the intermediate mesoderm and progenitors of organ primordia, the hematopoietic system and endothelial subtypes. Unexpectedly, by using a combination of single-cell transcriptomics and lineage tracing, we found that intra-adrenal sympathoblasts at that stage are directly derived from nerve-associated Schwann cell precursors, similarly to local chromaffin cells, whereas the majority of extra-adrenal sympathoblasts arise from the migratory neural crest. In humans, this process persists during several weeks of development within the large intra-adrenal ganglia-like structures, which may also serve as reservoirs of originating cells in neuroblastoma.
Collapse
Affiliation(s)
- Polina Kameneva
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
| | - Artem V Artemov
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Maria Eleni Kastriti
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Molecular Neurosciences, Medical University of Vienna, Vienna, Austria
| | - Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Thale K Olsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Jörg Otte
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Alek Erickson
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
| | - Bettina Semsch
- Department of Comparative Medicine, Karolinska Institutet, Solna, Sweden
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Michael Ratz
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institutet, Solna, Sweden
| | - Arthur S Tischler
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, MA, USA
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology CS, Utrecht, the Netherlands
- Deptartment of Pathology, University Medical Center Utrecht CX, Utrecht, the Netherlands
| | - Thibault Bouderlique
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Natalia Akkuratova
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
- Institute of Translational Biomedicine, St. Petersburg University, St. Petersburg, Russia
| | - Maria Vorontsova
- Endocrinology Research Centre, Moscow, Russian Federation
- Moscow Institute of Physics and Technology, Dolgoprudniy, Russian Federation
- Institute for Regenerative Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Oleg Gusev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
- RIKEN Innovation Center, RIKEN, Yokohama, Japan
- Center for Life Science Technologies, RIKEN, Yokohama, Japan
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Shenglin Mei
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Peter V Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden.
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
14
|
Siaw JT, Gabre JL, Uçkun E, Vigny M, Zhang W, Van den Eynden J, Hallberg B, Palmer RH, Guan J. Loss of RET Promotes Mesenchymal Identity in Neuroblastoma Cells. Cancers (Basel) 2021; 13:cancers13081909. [PMID: 33921066 PMCID: PMC8071449 DOI: 10.3390/cancers13081909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/21/2021] [Accepted: 04/12/2021] [Indexed: 12/18/2022] Open
Abstract
Aberrant activation of anaplastic lymphoma kinase (ALK) drives neuroblastoma (NB). Previous work identified the RET receptor tyrosine kinase (RTK) as a downstream target of ALK activity in NB models. We show here that ALK activation in response to ALKAL2 ligand results in the rapid phosphorylation of RET in NB cells, providing additional insight into the contribution of RET to the ALK-driven gene signature in NB. To further address the role of RET in NB, RET knockout (KO) SK-N-AS cells were generated by CRISPR/Cas9 genome engineering. Gene expression analysis of RET KO NB cells identified a reprogramming of NB cells to a mesenchymal (MES) phenotype that was characterized by increased migration and upregulation of the AXL and MNNG HOS transforming gene (MET) RTKs, as well as integrins and extracellular matrix components. Strikingly, the upregulation of AXL in the absence of RET reflects the development timeline observed in the neural crest as progenitor cells undergo differentiation during embryonic development. Together, these findings suggest that a MES phenotype is promoted in mesenchymal NB cells in the absence of RET, reflective of a less differentiated developmental status.
Collapse
Affiliation(s)
- Joachim T. Siaw
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden; (J.T.S.); (J.L.G.); (E.U.); (B.H.); (R.H.P.)
| | - Jonatan L. Gabre
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden; (J.T.S.); (J.L.G.); (E.U.); (B.H.); (R.H.P.)
- Anatomy and Embryology Unit, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium;
| | - Ezgi Uçkun
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden; (J.T.S.); (J.L.G.); (E.U.); (B.H.); (R.H.P.)
| | - Marc Vigny
- Université Pierre et Marie Curie, UPMC, INSERM UMRS-839, 75005 Paris, France;
| | - Wancun Zhang
- Department of Pediatric Oncology Surgery, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China;
| | - Jimmy Van den Eynden
- Anatomy and Embryology Unit, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium;
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden; (J.T.S.); (J.L.G.); (E.U.); (B.H.); (R.H.P.)
| | - Ruth H. Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden; (J.T.S.); (J.L.G.); (E.U.); (B.H.); (R.H.P.)
| | - Jikui Guan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden; (J.T.S.); (J.L.G.); (E.U.); (B.H.); (R.H.P.)
- Department of Pediatric Oncology Surgery, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China;
- Correspondence:
| |
Collapse
|
15
|
Hanemaaijer ES, Margaritis T, Sanders K, Bos FL, Candelli T, Al-Saati H, van Noesel MM, Meyer-Wentrup FAG, van de Wetering M, Holstege FCP, Clevers H. Single-cell atlas of developing murine adrenal gland reveals relation of Schwann cell precursor signature to neuroblastoma phenotype. Proc Natl Acad Sci U S A 2021; 118:e2022350118. [PMID: 33500353 PMCID: PMC7865168 DOI: 10.1073/pnas.2022350118] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor and accounts for ∼10% of pediatric cancer-related deaths. The exact cell of origin has yet to be elucidated, but it is generally accepted that neuroblastoma derives from the neural crest and should thus be considered an embryonal malignancy. About 50% of primary neuroblastoma tumors arise in the adrenal gland. Here, we present an atlas of the developing mouse adrenal gland at a single-cell level. Five main cell cluster groups (medulla, cortex, endothelial, stroma, and immune) make up the mouse adrenal gland during fetal development. The medulla group, which is of neural crest origin, is further divided into seven clusters. Of interest is the Schwann cell precursor ("SCP") and the "neuroblast" cluster, a highly cycling cluster that shares markers with sympathoblasts. The signature of the medullary SCP cluster differentiates neuroblastoma patients based on disease phenotype: The SCP signature score anticorrelates with ALK and MYCN expression, two indicators of poor prognosis. Furthermore, a high SCP signature score is associated with better overall survival rates. This study provides an insight into the developing adrenal gland and introduces the SCP gene signature as being of interest for further research in understanding neuroblastoma phenotype.
Collapse
Affiliation(s)
- Evelyn S Hanemaaijer
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Thanasis Margaritis
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Karin Sanders
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - Frank L Bos
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Tito Candelli
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Hanin Al-Saati
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | | | - Marc van de Wetering
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Frank C P Holstege
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Hans Clevers
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
16
|
Chiba T, Takaguri A, Maeda T. Norepinephrine transporter expressed on mammary epithelial cells incorporates norepinephrine in milk into the cells. Biochem Biophys Res Commun 2021; 545:1-7. [PMID: 33529804 DOI: 10.1016/j.bbrc.2021.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/07/2021] [Indexed: 11/16/2022]
Abstract
Mammary epithelial cells synthesize and secrete norepinephrine (NE) into breast milk to regulate β-casein expression through the adrenergic β2 receptor. We investigated the expression, localization, and roles of NE transporter (NET) in the mammary epithelium during lactation. mRNA and protein levels of NET were determined in primary normal human mammary epithelial cells (pHMECs) and non-malignant human mammary epithelial MCF-12A cells. In nursing CD1 mice, NET localized to the apical membranes of the mammary epithelium. The intracellular NE content of pHMECs incubated with NE increased. Although the β-casein concentration in milk was slightly higher at day 10 than at day 2 of lactation, the NE concentration and lactation-related proteins were only slightly changed on days 2-10. Restraint stress increased the NE concentration in milk from nursing mice and NET protein levels were significantly higher than in non-stressed nursing mice. NET is expressed on the apical membrane of mammary epithelial cells and incorporates NE in milk into cells, potentially regulating the NE concentration in milk.
Collapse
Affiliation(s)
- Takeshi Chiba
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 15-4-1 Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, 006-8585, Japan; Creation Research Institute of Life Science in KITA-no-DAICHI, Hokkaido University of Science, 15-4-1 Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, 006-8585, Japan.
| | - Akira Takaguri
- Creation Research Institute of Life Science in KITA-no-DAICHI, Hokkaido University of Science, 15-4-1 Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, 006-8585, Japan; Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 15-4-1 Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, 006-8585, Japan
| | - Tomoji Maeda
- Department of Clinical Pharmacology and Pharmaceutics, Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama, 362-0862, Japan
| |
Collapse
|
17
|
Kastriti ME, Kameneva P, Adameyko I. Stem cells, evolutionary aspects and pathology of the adrenal medulla: A new developmental paradigm. Mol Cell Endocrinol 2020; 518:110998. [PMID: 32818585 DOI: 10.1016/j.mce.2020.110998] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/20/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
The mammalian adrenal gland is composed of two main components; the catecholaminergic neural crest-derived medulla, found in the center of the gland, and the mesoderm-derived cortex producing steroidogenic hormones. The medulla is composed of neuroendocrine chromaffin cells with oxygen-sensing properties and is dependent on tissue interactions with the overlying cortex, both during development and in adulthood. Other relevant organs include the Zuckerkandl organ containing extra-adrenal chromaffin cells, and carotid oxygen-sensing bodies containing glomus cells. Chromaffin and glomus cells reveal a number of important similarities and are derived from the multipotent nerve-associated descendants of the neural crest, or Schwann cell precursors. Abnormalities in complex developmental processes during differentiation of nerve-associated and other progenitors into chromaffin and oxygen-sensing populations may result in different subtypes of paraganglioma, neuroblastoma and pheochromocytoma. Here, we summarize recent findings explaining the development of chromaffin and oxygen-sensing cells, as well as the potential mechanisms driving neuroendocrine tumor initiation.
Collapse
Affiliation(s)
- Maria Eleni Kastriti
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden; Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Polina Kameneva
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden; National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden; Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria; Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
18
|
Rocha M, Beiriger A, Kushkowski EE, Miyashita T, Singh N, Venkataraman V, Prince VE. From head to tail: regionalization of the neural crest. Development 2020; 147:dev193888. [PMID: 33106325 PMCID: PMC7648597 DOI: 10.1242/dev.193888] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neural crest is regionalized along the anteroposterior axis, as demonstrated by foundational lineage-tracing experiments that showed the restricted developmental potential of neural crest cells originating in the head. Here, we explore how recent studies of experimental embryology, genetic circuits and stem cell differentiation have shaped our understanding of the mechanisms that establish axial-specific populations of neural crest cells. Additionally, we evaluate how comparative, anatomical and genomic approaches have informed our current understanding of the evolution of the neural crest and its contribution to the vertebrate body.
Collapse
Affiliation(s)
- Manuel Rocha
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Anastasia Beiriger
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| | - Elaine E Kushkowski
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Tetsuto Miyashita
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
- Canadian Museum of Nature, Ottawa, ON K1P 6P4, Canada
| | - Noor Singh
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| | - Vishruth Venkataraman
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| | - Victoria E Prince
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL 60637, USA
- Department of Organismal Biology and Anatomy, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
19
|
Abstract
Investigations of the cellular and molecular mechanisms that mediate the development of the autonomic nervous system have identified critical genes and signaling pathways that, when disrupted, cause disorders of the autonomic nervous system. This review summarizes our current understanding of how the autonomic nervous system emerges from the organized spatial and temporal patterning of precursor cell migration, proliferation, communication, and differentiation, and discusses potential clinical implications for developmental disorders of the autonomic nervous system, including familial dysautonomia, Hirschsprung disease, Rett syndrome, and congenital central hypoventilation syndrome.
Collapse
Affiliation(s)
- Frances Lefcort
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, Montana
| |
Collapse
|
20
|
Murtazina AR, Nikishina YO, Ugrumov MV. The Role of the Brain in the Regulation of Peripheral Organs-Noradrenaline Sources in Neonatal Rats: Noradrenaline Synthesis Enzyme Activity. DOKL BIOCHEM BIOPHYS 2020; 493:201-204. [PMID: 32894465 DOI: 10.1134/s1607672920040109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 11/22/2022]
Abstract
This work is aimed at studying the mechanisms of reciprocal humoral regulation of noradrenaline-producing organs in rats in the perinatal period of development. The activity of noradrenaline synthesis enzymes tyrosine hydroxylase and dopamine-beta-hydroxylase was measured in the brain and adrenal glands 48 and 72 h after the injection of immunotoxin (anti-dopamine-beta-hydroxylase-saporin) into the rat brain ventricles. It was shown that, 48 h after the immunotoxin injection into the brain, the activity of tyrosine hydroxylase in the brain decreased; however, 72 h after the injection it reached the control levels. This fact indicates that noradrenaline synthesis in the survived neurons increases. In the adrenal glands, 72 h after the immunotoxin injection into the brain, the activity of dopamine-beta-hydroxylase increased. This points to a compensatory increase in the rate of noradrenaline synthesis in the adrenal glands when the synthesis of noradrenaline in the brain is inhibited.
Collapse
Affiliation(s)
- A R Murtazina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Yu O Nikishina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.
| | - M V Ugrumov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
21
|
Mariniello K, Ruiz-Babot G, McGaugh EC, Nicholson JG, Gualtieri A, Gaston-Massuet C, Nostro MC, Guasti L. Stem Cells, Self-Renewal, and Lineage Commitment in the Endocrine System. Front Endocrinol (Lausanne) 2019; 10:772. [PMID: 31781041 PMCID: PMC6856655 DOI: 10.3389/fendo.2019.00772] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
The endocrine system coordinates a wide array of body functions mainly through secretion of hormones and their actions on target tissues. Over the last decades, a collective effort between developmental biologists, geneticists, and stem cell biologists has generated a wealth of knowledge related to the contribution of stem/progenitor cells to both organogenesis and self-renewal of endocrine organs. This review provides an up-to-date and comprehensive overview of the role of tissue stem cells in the development and self-renewal of endocrine organs. Pathways governing crucial steps in both development and stemness maintenance, and that are known to be frequently altered in a wide array of endocrine disorders, including cancer, are also described. Crucially, this plethora of information is being channeled into the development of potential new cell-based treatment modalities for endocrine-related illnesses, some of which have made it through clinical trials.
Collapse
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Emily C. McGaugh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - James G. Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Angelica Gualtieri
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
22
|
Zakrevska MV, Tybinka AM. Peculiarities of microstructure of the suprarenal glands of rabbits with different types of autonomic tone. REGULATORY MECHANISMS IN BIOSYSTEMS 2019. [DOI: 10.15421/021962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The article investigates the structure of the suprarenal (adrenal) glands of male rabbits (Oryctolagus cuniculus), in which, on the basis of electrocardiographic and variational-pulsometric studies, different types of autonomic tone were observed. This allowed the animals to be divided into three groups: 1) sympathicotonic rabbits; 2) normotonic rabbits; 3) parasympathicotonic rabbits. The animals of the first two groups were characterized by almost the same body weight, while weight of the rabbits of the third group was slightly higher. After euthanasia, the suprarenal glands were extracted for histological and histochemical analyses. Morphometric study of histopreparations revealed that in the normotonic rabbits the thickness of the zona glomerulosa and zona fasciculata of the suprarenal glands were of average sizes, and the area of the medulla was the smallest. The parasympathicotonic rabbits had the thickest zona glomerulosa and greatest area of the medulla, but the thinnest zona fasciculata. The sympathicotonic rabbits were observed to have the greatest thickness of the zona fasciculata of the suprarenal glands, the area of the medulla was of average values, and the thickness of the zona glumerulosa was of minimum value. The type of autonomic tone also manifests in the saturation of each of the zones with cells. The normotonic rabbits were observed to have the highest number of cells per area of 1,000 µm² in the zona fasciculata and the medulla, sympathicotonic rabbits – in the zona glomerulosa and zona reticularis, and in parasympathicotonic rabbits this parameter had average or lowest values in all the zones. The sizes of cells and their structural parts were characterized on the basis of nuclear-cytoplasmic ratio. In the zona fasciculata and medulla this parameter was highest among parasympathicotonic rabbits, and lowest in sympathicotonic rabbits. In the zona glomerulosa, almost equal values were observed in the normotonic and parasympathicotonic rabbits, while being reliably lower in sympathicotonic rabbits. By the value of nuclear-cytoplasmic ratio in the zona reticularis, the normotonic rabbits dominated, followed by the sympathicotonic animals, and the parasympathicotonic rabbits had the lowest parameters.
Collapse
|
23
|
Wang H, Krishnan C, Charville GW. INSM1 Expression in Peripheral Neuroblastic Tumors and Other Embryonal Neoplasms. Pediatr Dev Pathol 2019; 22:440-448. [PMID: 30975032 DOI: 10.1177/1093526619843725] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Insulinoma-associated protein 1 (INSM1) is a transcription factor that functions in neuroepithelial tissue development and shows expression in neuroendocrine neoplasms. Given the role of INSM1 in controlling differentiation of the sympatho-adrenal lineage, we hypothesized that INSM1 expression would define a subset of neuroblastic tumors. This study aimed to characterize the immunohistochemical profile of INSM1 in a cohort of peripheral neuroblastic tumors and compare INSM1 expression in these tumors to that seen in other embryonal neoplasms, using both tissue microarrays and whole-slide histologic sections. INSM1 showed nuclear expression in 39/50 (78%) peripheral neuroblastic tumors, including 27/32 (84%) neuroblastomas, 9/9 (100%) ganglioneuroblastomas, and 3/9 (33%) ganglioneuromas. Altogether, 70% of peripheral neuroblastic tumors showed anti-INSM1 immunoreactivity in more than 20% of tumor nuclei. Although no non-neuroblastic tumors in this study exhibited INSM1 expression in more than 20% of nuclei, focal or patchy staining was identified in 7/14 (50%) rhabdomyosarcomas, 7/22 (32%) nephroblastomas, and 4/20 (20%) Ewing sarcomas. The absence of INSM1 expression in peripheral neuroblastic tumors was restricted to undifferentiated and poorly differentiated neuroblastomas, as well as mature ganglioneuromas, mimicking the transient INSM1 expression seen in sympatho-adrenal differentiation during normal development. No significant association between MYCN amplification status and INSM1 expression was observed. We found that all 3 INSM1-negative neuroblastoma patients with available follow-up were alive at a median of 15 years, in comparison to 9 of 13 INSM1-positive neuroblastoma patients living at a median of 5 years. Additional studies are needed to determine whether INSM1 expression is indicative of a clinically significant differentiation state in neuroblastoma.
Collapse
Affiliation(s)
- Hannah Wang
- Department of Pathology, Stanford University School of Medicine, Stanford, California.,Stanford Hospital and Clinics, Stanford, California
| | - Chandra Krishnan
- Department of Pathology, Dell Children's Medical Center, Austin, Texas
| | - Gregory W Charville
- Department of Pathology, Stanford University School of Medicine, Stanford, California.,Stanford Hospital and Clinics, Stanford, California
| |
Collapse
|
24
|
Murtazina AR, Nikishina YO, Bondarenko NS, Dil'mukhametova LK, Sapronova AY, Ugrumov MV. Developing brain as a source of circulating norepinephrine in rats during the critical period of morphogenesis. Brain Struct Funct 2019; 224:3059-3073. [PMID: 31493024 DOI: 10.1007/s00429-019-01950-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/28/2019] [Indexed: 11/29/2022]
Abstract
The development of individual organs and the whole organism is under the control by morphogenetic factors over the critical period of morphogenesis. This study was aimed to test our hypothesis that the developing brain operates as an endocrine organ during morphogenesis, in rats during the perinatal period (Ugrumov in Neuro Chem 35:837-850, 2010). Norepinephrine, which is a morphogenetic factor, was used as a marker of the endocrine activity of the developing brain, although it is also secreted by peripheral organs. In this study, it was first shown that the concentration of norepinephrine in the peripheral blood of neonatal rats is sufficient to ensure the morphogenetic effect on the peripheral organs and the brain itself. Using pharmacological suppression of norepinephrine production in the brain, but not in peripheral organs, it was shown that norepinephrine is delivered from the brain to the general circulation in neonatal rats, that is, during morphogenesis. In fact, even partial suppression of norepinephrine production in the brain of neonatal rats led to a significant decrease of norepinephrine concentration in plasma, suggesting that at this time the brain is an important source of circulating norepinephrine. Conversely, the suppression of the production of norepinephrine in the brain of prepubertal rats did not cause a change in its concentration in plasma, showing no secretion of brain-derived norepinephrine to the bloodstream after morphogenesis. The above data support our hypothesis that morphogenetic factors, including norepinephrine, are delivered from the developing brain to the bloodstream, which occurs only during the critical period of morphogenesis.
Collapse
Affiliation(s)
- Aliia R Murtazina
- Laboratory of Neural and Neuroendocrine Regulations, Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov str., 119334, Moscow, Russia
| | - Yulia O Nikishina
- Laboratory of Neural and Neuroendocrine Regulations, Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov str., 119334, Moscow, Russia
| | - Nadezhda S Bondarenko
- Laboratory of Neural and Neuroendocrine Regulations, Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov str., 119334, Moscow, Russia
| | - Liliya K Dil'mukhametova
- Laboratory of Neural and Neuroendocrine Regulations, Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov str., 119334, Moscow, Russia
| | - Anna Ya Sapronova
- Laboratory of Neural and Neuroendocrine Regulations, Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov str., 119334, Moscow, Russia
| | - Michael V Ugrumov
- Laboratory of Neural and Neuroendocrine Regulations, Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov str., 119334, Moscow, Russia.
| |
Collapse
|
25
|
Chiba T, Maeda T, Fujita Y, Takeda R, Kikuchi A, Kudo K. Stress-Induced Suppression of Milk Protein Is Involved in a Noradrenergic Mechanism in the Mammary Gland. Endocrinology 2019; 160:2074-2084. [PMID: 31150047 DOI: 10.1210/en.2019-00300] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/23/2019] [Indexed: 01/19/2023]
Abstract
Stress decreases milk components such as milk protein and milk yield. The objective of this study was to investigate whether noradrenaline (NA) in milk constituted a factor associated with stress-induced changes in milk proteins such as β-casein. Breast milk obtained from eight healthy, nursing women contained NA at concentrations ranging from 12.7 to 115.5 nM. The expression of tyrosine hydroxylase (TH), a rate-limiting enzyme of NA synthesis, was observed in primary normal human mammary epithelial cells (HMECs), and in MCF-12A and MCF-10A cell lines. The mean NA concentration in culture medium used by MCF-12A transfected with TH small interfering RNA (siRNA) was significantly lower than that of cells transfected with control siRNA. NA concentration in milk in restraint-stressed nursing mice was significantly higher than that in nonstressed nursing mice, owing to elevated TH expression in the mammary epithelium. The mean β-casein concentration in milk in restraint-stressed mice was significantly lower than that in nonstressed mice. NA treatment resulted in a concentration-dependent decrease in β-casein expression in HMECs. β2 adrenergic receptor (ADRB2) expression was observed in HMECs, MCF-12A, and MCF-10A, and immunohistochemical analysis of ADRB2 using mammary epithelium sections obtained from mice at day 10 of lactation showed that ADRB2 was expressed at the apical membrane of mammary epithelium. Treatment with salbutamol, an ADRB2 stimulant, decreased β-casein expression in a concentration-dependent manner in MCF-12A. Our results showed that endogenous NA derived from mammary epithelial cells likely comprises one of the factors involved in stress-induced changes in milk proteins such as β-casein.
Collapse
Affiliation(s)
- Takeshi Chiba
- Department of Clinical Pharmacy, Division of Clinical Pharmaceutics and Pharmacy Practice, School of Pharmacy, Iwate Medical University, Iwate, Japan
| | - Tomoji Maeda
- Department of Pharmacology, Nihon Pharmaceutical University, Komuro, Ina-machi, Kitaadachi-gun, Saitama, Japan
| | - Yu Fujita
- Department of Biological Pharmacy, Division of Neuroscience, School of Pharmacy, Iwate Medical University, Iwate, Japan
| | - Rika Takeda
- Department of Nursing, Iwate Medical University Hospital, Iwate, Japan
| | - Akihiko Kikuchi
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Kenzo Kudo
- Department of Clinical Pharmacy, Division of Clinical Pharmaceutics and Pharmacy Practice, School of Pharmacy, Iwate Medical University, Iwate, Japan
| |
Collapse
|
26
|
Murtazina AR, Nikishina YO, Dil'mukhametova LK, Sapronova AY, Ugrumov MV. The Role of the Brain in the Regulation of Peripheral Noradrenaline-producing Organs in Rats During Morphogenesis. DOKL BIOCHEM BIOPHYS 2019; 486:243-246. [PMID: 31367831 DOI: 10.1134/s1607672919030207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Indexed: 11/23/2022]
Abstract
This work represents one part of our research project, in which we attempted to prove that a humoral regulation between noradrenaline-producing organs exist in the perinatal period. In this study, we used a rat model that allowed blocking the synthesis of noradrenalin in the brain and evaluated gene expression and protein levels of noradrenaline key synthesis enzymes such as tyrosine hydroxylase (TH) and dopamine beta-hydroxylase (DBH) in peripheral noradrenaline-producing organs. As a result, we showed an increased gene expression of TH and DBH in adrenal glands. These data indicate that, if neonatal rat brain lacks the ability to produce noradrenaline, then the synthesis of noradrenaline in adrenal glands increased as a compensatory process, so that the concentration levels in blood are maintained at normal levels. This indicates that there is a humoral regulation between brain and adrenal glands, which is not fully understood yet.
Collapse
Affiliation(s)
- A R Murtazina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, 119334, Moscow, Russia
| | - Yu O Nikishina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, 119334, Moscow, Russia.
| | - L K Dil'mukhametova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, 119334, Moscow, Russia
| | - A Ya Sapronova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, 119334, Moscow, Russia
| | - M V Ugrumov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, 119334, Moscow, Russia
| |
Collapse
|
27
|
Koopman K, Gaal J, de Krijger RR. Pheochromocytomas and Paragangliomas: New Developments with Regard to Classification, Genetics, and Cell of Origin. Cancers (Basel) 2019; 11:cancers11081070. [PMID: 31362359 PMCID: PMC6721302 DOI: 10.3390/cancers11081070] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
Pheochromocytomas (PCC) and paragangliomas (PGL) are rare neuroendocrine tumors that arise in the adrenal medulla and in extra-adrenal locations, such as the head, neck, thorax, abdomen, and pelvis. Classification of these tumors into those with or without metastatic potential on the basis of gross or microscopic features is challenging. Recent insights and scoring systems have attempted to develop solutions for this, as described in the latest World Health Organization (WHO) edition on endocrine tumor pathology. PCC and PGL are amongst the tumors most frequently accompanied by germline mutations. More than 20 genes are responsible for a hereditary background in up to 40% of these tumors; somatic mutations in the same and several additional genes form the basis for another 30%. However, this does not allow for a complete understanding of the pathogenesis or targeted treatment of PCC and PGL, for which surgery is the primary treatment and for which metastasis is associated with poor outcome. This review describes recent insights into the cell of origin of these tumors, the latest developments with regard to the genetic background, and the current status of tumor classification including proposed scoring systems.
Collapse
Affiliation(s)
- Karen Koopman
- Martini Hospital, 9728 NT Groningen, The Netherlands
| | - Jose Gaal
- Department of Pathology, Isala Hospital, 8025AB Zwolle, The Netherlands
| | - Ronald R de Krijger
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands.
- Princess Maxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands.
| |
Collapse
|
28
|
Kastriti ME, Kameneva P, Kamenev D, Dyachuk V, Furlan A, Hampl M, Memic F, Marklund U, Lallemend F, Hadjab S, Calvo-Enrique L, Ernfors P, Fried K, Adameyko I. Schwann Cell Precursors Generate the Majority of Chromaffin Cells in Zuckerkandl Organ and Some Sympathetic Neurons in Paraganglia. Front Mol Neurosci 2019; 12:6. [PMID: 30740044 PMCID: PMC6355685 DOI: 10.3389/fnmol.2019.00006] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/09/2019] [Indexed: 11/13/2022] Open
Abstract
In humans, neurosecretory chromaffin cells control a number of important bodily functions, including those related to stress response. Chromaffin cells appear as a distinct cell type at the beginning of midgestation and are the main cellular source of adrenalin and noradrenalin released into the blood stream. In mammals, two different chromaffin organs emerge at a close distance to each other, the adrenal gland and Zuckerkandl organ (ZO). These two structures are found in close proximity to the kidneys and dorsal aorta, in a region where paraganglioma, pheochromocytoma and neuroblastoma originate in the majority of clinical cases. Recent studies showed that the chromaffin cells comprising the adrenal medulla are largely derived from nerve-associated multipotent Schwann cell precursors (SCPs) arriving at the adrenal anlage with the preganglionic nerve fibers, whereas the migratory neural crest cells provide only minor contribution. However, the embryonic origin of the ZO, which differs from the adrenal medulla in a number of aspects, has not been studied in detail. The ZO is composed of chromaffin cells in direct contact with the dorsal aorta and the intraperitoneal cavity and disappears through an autophagy-mediated mechanism after birth. In contrast, the adrenal medulla remains throughout the entire life and furthermore, is covered by the adrenal cortex. Using a combination of lineage tracing strategies with nerve- and cell type-specific ablations, we reveal that the ZO is largely SCP-derived and forms in synchrony with progressively increasing innervation. Moreover, the ZO develops hand-in-hand with the adjacent sympathetic ganglia that coalesce around the dorsal aorta. Finally, we were able to provide evidence for a SCP-contribution to a small but significant proportion of sympathetic neurons of the posterior paraganglia. Thus, this cellular source complements the neural crest, which acts as a main source of sympathetic neurons. Our discovery of a nerve-dependent origin of chromaffin cells and some sympathoblasts may help to understand the origin of pheochromocytoma, paraganglioma and neuroblastoma, all of which are currently thought to be derived from the neural crest or committed sympathoadrenal precursors.
Collapse
Affiliation(s)
- Maria Eleni Kastriti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Polina Kameneva
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia
| | - Dmitry Kamenev
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia.,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Viacheslav Dyachuk
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia.,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Alessandro Furlan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| | - Marek Hampl
- Institute of Animal Physiology and Genetics, CAS, Brno, Czechia.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Fatima Memic
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Marklund
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Laura Calvo-Enrique
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Patrik Ernfors
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Kaj Fried
- National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
29
|
Catalano-Iniesta L, Iglesias-Osma MC, Sánchez-Robledo V, Carretero-Hernández M, Blanco EJ, Carretero J, García-Barrado MJ. Variations in adrenal gland medulla and dopamine effects induced by the lack of Irs2. J Physiol Biochem 2018; 74:667-677. [PMID: 30367392 DOI: 10.1007/s13105-018-0655-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 10/18/2018] [Indexed: 01/26/2023]
Abstract
The adrenomedullary chromaffin cells' hormonal pathway has been related to the pathophysiology of diabetes mellitus. In mice, the deletion of insulin receptor substrate type 2 (Irs2) causes peripheral insulin resistance and reduction in β-cell mass, leading to overt diabetes, with gender differences on adrenergic signaling. To further unravel the relevance of Irs2 on glycemic control, we analyzed in adult Irs2 deficient (Irs2-/-) mice, of both sexes but still normoglycemic, dopamine effects on insulin secretion and glycerol release, as well as their adrenal medulla by an immunohistochemical and morphologic approach. In isolated islets, 10 μM dopamine significantly inhibited insulin release in wild-type (WT) and female Irs2-/- mice; however, male Irs2-/- islets were insensitive to that catecholamine. Similarly, on isolated adipocytes, gender differences were observed between WT and Irs2-/- mice in basal and evoked glycerol release with crescent concentrations of dopamine. By immunohistochemistry, reactivity to tyrosine hydroxylase (TH) in female mice was significantly higher in the adrenal medulla of Irs2-/- compared to WT; although no differences for TH-immunopositivity were observed between the male groups of mice. However, compared to their corresponding WT animals, adrenomedullary chromaffin cells of Irs2-/- mice showed a significant decrease in the cellular and nuclear areas, and even in their percentage of apoptosis. Therefore, our observations suggest that, together with gender differences on dopamine responses in Irs2-/- mice, disturbances in adrenomedullary chromaffin cells could be related to deficiency of Irs2. Accordingly, Irs2 could be necessary for adequate glucose homeostasis and maintenance of the population of the adrenomedullary chromaffin cells.
Collapse
Affiliation(s)
- Leonardo Catalano-Iniesta
- Department of Physiology and Pharmacology, INCyL and IBSAL, Faculty of Medicine, University of Salamanca, Avda. Alfonso X el Sabio, s/n, E-37007, Salamanca, Spain.,Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain.,Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - María Carmen Iglesias-Osma
- Department of Physiology and Pharmacology, INCyL and IBSAL, Faculty of Medicine, University of Salamanca, Avda. Alfonso X el Sabio, s/n, E-37007, Salamanca, Spain.,Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain.,Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Virginia Sánchez-Robledo
- Department of Physiology and Pharmacology, INCyL and IBSAL, Faculty of Medicine, University of Salamanca, Avda. Alfonso X el Sabio, s/n, E-37007, Salamanca, Spain.,Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain.,Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Marta Carretero-Hernández
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain.,Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain.,Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Enrique J Blanco
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain.,Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain.,Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - José Carretero
- Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain.,Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain.,Department of Human Anatomy and Histology, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - María José García-Barrado
- Department of Physiology and Pharmacology, INCyL and IBSAL, Faculty of Medicine, University of Salamanca, Avda. Alfonso X el Sabio, s/n, E-37007, Salamanca, Spain. .,Laboratory of Neuroendocrinology, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain. .,Laboratory of Neuroendocrinology and Obesity, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain.
| |
Collapse
|
30
|
Hockman D, Adameyko I, Kaucka M, Barraud P, Otani T, Hunt A, Hartwig AC, Sock E, Waithe D, Franck MCM, Ernfors P, Ehinger S, Howard MJ, Brown N, Reese J, Baker CVH. Striking parallels between carotid body glomus cell and adrenal chromaffin cell development. Dev Biol 2018; 444 Suppl 1:S308-S324. [PMID: 29807017 PMCID: PMC6453021 DOI: 10.1016/j.ydbio.2018.05.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/20/2018] [Accepted: 05/20/2018] [Indexed: 12/31/2022]
Abstract
Carotid body glomus cells mediate essential reflex responses to arterial blood hypoxia. They are dopaminergic and secrete growth factors that support dopaminergic neurons, making the carotid body a potential source of patient-specific cells for Parkinson's disease therapy. Like adrenal chromaffin cells, which are also hypoxia-sensitive, glomus cells are neural crest-derived and require the transcription factors Ascl1 and Phox2b; otherwise, their development is little understood at the molecular level. Here, analysis in chicken and mouse reveals further striking molecular parallels, though also some differences, between glomus and adrenal chromaffin cell development. Moreover, histology has long suggested that glomus cell precursors are ‘émigrés’ from neighbouring ganglia/nerves, while multipotent nerve-associated glial cells are now known to make a significant contribution to the adrenal chromaffin cell population in the mouse. We present conditional genetic lineage-tracing data from mice supporting the hypothesis that progenitors expressing the glial marker proteolipid protein 1, presumably located in adjacent ganglia/nerves, also contribute to glomus cells. Finally, we resolve a paradox for the ‘émigré’ hypothesis in the chicken - where the nearest ganglion to the carotid body is the nodose, in which the satellite glia are neural crest-derived, but the neurons are almost entirely placode-derived - by fate-mapping putative nodose neuronal 'émigrés' to the neural crest. Glomus cell precursors express the neuron-specific marker Elavl3/4 (HuC/D). Developing glomus cells express multiple ‘sympathoadrenal' genes. Glomus cell development requires Hand2 and Sox4/11, but not Ret or Tfap2b. Multipotent progenitors with a glial phenotype contribute to glomus cells. Fate-mapping resolves a paradox for the ganglionic 'émigré' hypothesis in birds.
Collapse
Affiliation(s)
- Dorit Hockman
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, United Kingdom; Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, United Kingdom; Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institute, S-171 77 Stockholm, Sweden; Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria
| | - Marketa Kaucka
- Department of Physiology and Pharmacology, Karolinska Institute, S-171 77 Stockholm, Sweden
| | - Perrine Barraud
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, United Kingdom
| | - Tomoki Otani
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, United Kingdom
| | - Adam Hunt
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, United Kingdom
| | - Anna C Hartwig
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen, Germany
| | - Dominic Waithe
- Wolfson Imaging Centre, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, United Kingdom
| | - Marina C M Franck
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, S-171 77 Stockholm, Sweden
| | - Patrik Ernfors
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, S-171 77 Stockholm, Sweden
| | - Sean Ehinger
- Department of Neurosciences and Program in Neurosciences and Neurodegenerative Diseases, University of Toledo Health Sciences Campus, Toledo, OH 43614, USA
| | - Marthe J Howard
- Department of Neurosciences and Program in Neurosciences and Neurodegenerative Diseases, University of Toledo Health Sciences Campus, Toledo, OH 43614, USA
| | - Naoko Brown
- Depts. of Pediatrics, Cell and Developmental Biology, Vanderbilt University Medical Center, 2215 B Garland Avenue, Nashville, TN 37232, USA
| | - Jeffrey Reese
- Depts. of Pediatrics, Cell and Developmental Biology, Vanderbilt University Medical Center, 2215 B Garland Avenue, Nashville, TN 37232, USA
| | - Clare V H Baker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, United Kingdom.
| |
Collapse
|
31
|
Furlan A, Adameyko I. Schwann cell precursor: a neural crest cell in disguise? Dev Biol 2018; 444 Suppl 1:S25-S35. [PMID: 29454705 DOI: 10.1016/j.ydbio.2018.02.008] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 01/19/2023]
Abstract
Schwann cell precursors (SCPs) are multipotent embryonic progenitors covering all developing peripheral nerves. These nerves grow and navigate with unprecedented precision, delivering SCP progenitors to almost all locations in the embryonic body. Within specific developing tissues, SCPs detach from nerves and generate neuroendocrine cells, autonomic neurons, mature Schwann cells, melanocytes and other cell types. These properties of SCPs evoke resemblances between them and their parental population, namely, neural crest cells. Neural crest cells are incredibly multipotent migratory cells that revolutionized the course of evolution in the lineage of early chordate animals. Given this similarity and recent data, it is possible to hypothesize that proto-neural crest cells are similar to SCPs spreading along the nerves. Here, we review the multipotency of SCPs, the signals that govern them, their potential therapeutic value, SCP's embryonic origin and their evolutionary connections. We dedicate this article to the memory of Wilhelm His, the father of the microtome and "Zwischenstrang", currently known as the neural crest.
Collapse
Affiliation(s)
- Alessandro Furlan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724 USA
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria.
| |
Collapse
|
32
|
Fishbein L, Wilkerson MD. Chromaffin cell biology: inferences from The Cancer Genome Atlas. Cell Tissue Res 2018; 372:339-346. [PMID: 29450724 DOI: 10.1007/s00441-018-2795-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 01/16/2018] [Indexed: 12/28/2022]
Abstract
Pheochromocytomas and paragangliomas (PCC/PGLs) are rare neuroendocrine tumors that are unusually diverse in metabolic profiles, in classes of molecular alterations and across a large number of altered genes. The Cancer Genome Atlas (TCGA) comprehensively profiled the molecular landscape of PCC/PGLs and identified novel genomic alterations and a new molecular classification of PCC/PGLs. In this review, we discuss the significant clinico-molecular findings of this integrated profiling study. We then review the molecular data of the TCGA cohort centering around known markers of sympathoadrenal cell lineage to better understand chromaffin cell biology. This analysis adds a new layer, that of chromaffin cell type, onto the published molecular classifications and in doing so provides inferences about underlying chromaffin cell biology and diversity.
Collapse
Affiliation(s)
- Lauren Fishbein
- Division of Endocrinology, Metabolism and Diabetes, Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, 12801 E. 17th Ave, MS 8106, Aurora, CO, 80045, USA
| | - Matthew D Wilkerson
- The American Genome Center, Collaborative Health Initiative Research Program, Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
33
|
Furlan A, Dyachuk V, Kastriti ME, Calvo-Enrique L, Abdo H, Hadjab S, Chontorotzea T, Akkuratova N, Usoskin D, Kamenev D, Petersen J, Sunadome K, Memic F, Marklund U, Fried K, Topilko P, Lallemend F, Kharchenko PV, Ernfors P, Adameyko I. Multipotent peripheral glial cells generate neuroendocrine cells of the adrenal medulla. Science 2018; 357:357/6346/eaal3753. [PMID: 28684471 DOI: 10.1126/science.aal3753] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 06/05/2017] [Indexed: 12/23/2022]
Abstract
Adrenaline is a fundamental circulating hormone for bodily responses to internal and external stressors. Chromaffin cells of the adrenal medulla (AM) represent the main neuroendocrine adrenergic component and are believed to differentiate from neural crest cells. We demonstrate that large numbers of chromaffin cells arise from peripheral glial stem cells, termed Schwann cell precursors (SCPs). SCPs migrate along the visceral motor nerve to the vicinity of the forming adrenal gland, where they detach from the nerve and form postsynaptic neuroendocrine chromaffin cells. An intricate molecular logic drives two sequential phases of gene expression, one unique for a distinct transient cellular state and another for cell type specification. Subsequently, these programs down-regulate SCP-gene and up-regulate chromaffin cell-gene networks. The AM forms through limited cell expansion and requires the recruitment of numerous SCPs. Thus, peripheral nerves serve as a stem cell niche for neuroendocrine system development.
Collapse
Affiliation(s)
- Alessandro Furlan
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Vyacheslav Dyachuk
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden.,National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia.,Department of Nanophotonics and Metamaterials, ITMO University, St. Petersburg 197101, Russia
| | - Maria Eleni Kastriti
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Laura Calvo-Enrique
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Hind Abdo
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Tatiana Chontorotzea
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Natalia Akkuratova
- Skolkovo Institute of Science and Technology, Moscow 143005, Russia.,Institute of Translational Biomedicine, Saint Petersburg State University, St. Petersburg 199034, Russia
| | - Dmitry Usoskin
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Dmitry Kamenev
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Julian Petersen
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden.,Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria
| | - Kazunori Sunadome
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Fatima Memic
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ulrika Marklund
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Piotr Topilko
- Institut de Biologie de l'Ecole Normale Supérieure, Ecole Normale Supérieure, INSERM U1024, CNRS UMR 8197, 46 Rue d'Ulm, 75005 Paris, France
| | - Francois Lallemend
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Peter V Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Patrik Ernfors
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden.
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden. .,Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria
| |
Collapse
|
34
|
Abu-Bonsrah KD, Zhang D, Bjorksten AR, Dottori M, Newgreen DF. Generation of Adrenal Chromaffin-like Cells from Human Pluripotent Stem Cells. Stem Cell Reports 2018; 10:134-150. [PMID: 29233551 PMCID: PMC5768882 DOI: 10.1016/j.stemcr.2017.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 11/29/2022] Open
Abstract
Adrenomedullary chromaffin cells are catecholamine (CA)-producing cells originating from trunk neural crest (NC) via sympathoadrenal progenitors (SAPs). We generated NC and SAPs from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) in vitro via BMP2/FGF2 exposure, ascertained by qPCR and immunoexpression of SOX10, ASCL1, TFAP2α, and PHOX2B, and by fluorescence-activated cell sorting selection for p75NTR and GD2, and confirmed their trunk-like HOX gene expression. We showed that continuing BMP4 and curtailing FGF2 in vitro, augmented with corticosteroid mimetic, induced these cells to upregulate the chromaffin cell-specific marker PNMT and other CA synthesis and storage markers, and we demonstrated noradrenaline and adrenaline by Faglu and high-performance liquid chromatography. We showed these human cells' SAP-like property of migration and differentiation into cells expressing chromaffin cell markers by implanting them into avian embryos in vivo and in chorio-allantoic membrane grafts. These cells have the potential for investigating differentiation of human chromaffin cells and for modeling diseases involving this cell type.
Collapse
Affiliation(s)
- Kwaku Dad Abu-Bonsrah
- The Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052 VIC, Australia; Centre for Neural Engineering, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Dongcheng Zhang
- The Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052 VIC, Australia
| | - Andrew R Bjorksten
- Department of Anaesthesia and Pain Management, The Royal Melbourne Hospital Grattan Street, Parkville, 3052 VIC, Australia
| | - Mirella Dottori
- Centre for Neural Engineering, University of Melbourne, Parkville, 3010 VIC, Australia; Department of Anatomy and Neurosciences, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Donald F Newgreen
- The Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052 VIC, Australia.
| |
Collapse
|
35
|
Murtazina AR, Nikishina YO, Bondarenko NS, Sapronova AY, Volina EV, Ugryumov MV. Gene expression and the contents of noradrenaline synthesis enzymes in the rat brain during the critical period of morphogenesis. NEUROCHEM J+ 2017. [DOI: 10.1134/s1819712417030072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Hadoux J, Desterke C, Féraud O, Guibert M, De Rose RF, Opolon P, Divers D, Gobbo E, Griscelli F, Schlumberger M, Bennaceur-Griscelli A, Turhan AG. Transcriptional landscape of a RET C634Y-mutated iPSC and its CRISPR-corrected isogenic control reveals the putative role of EGR1 transcriptional program in the development of multiple endocrine neoplasia type 2A-associated cancers. Stem Cell Res 2017; 26:8-16. [PMID: 29197744 DOI: 10.1016/j.scr.2017.11.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/14/2017] [Accepted: 11/20/2017] [Indexed: 12/15/2022] Open
Abstract
MEN2A is a hereditary cancer-predisposing syndrome that affects patients with germline RET mutations. The effects of this oncogenic tyrosine kinase in the context of primitive stem cells are not known. In order to study these events, we generated a MEN2A induced Pluripotent Stem Cell (iPSC) line from a patient with RET mutation and an isogenic counterpart by CRISPR-Cas9 correction of the mutation. Whole exome sequencing of iPSC before and after CRISPR-Cas9 genome edition revealed no major exonic off target effect of the CRISPR correction. However, an integrative differential gene expression analysis of iPSC with oncogenic RETC634Y and its gene-corrected iPSC with RETY634C as well as RETwt iPSCs revealed activation of the Early Growth Response 1 (EGR1) transcriptional program in RET-mutated iPSC, a pathway shown to be involved in RET-induced oncogenesis. These data constitute the first proof of concept of the feasibility of the use of an iPSC and its genome-corrected counterpart to unravel the molecular mechanisms underlying the development of the hereditary MEN2A cancer predisposing syndrome.
Collapse
Affiliation(s)
- Julien Hadoux
- Inserm UMRS 935, Université Paris Sud, Villejuif, France; Gustave Roussy, Department of Nuclear medicine and Endocrine Oncology, 94800 Villejuif, France
| | | | - Olivier Féraud
- Inserm UMRS 935, Université Paris Sud, Villejuif, France; ESTeam Paris Sud, Infrastructure INGESTEM, Villejuif, France
| | - Mathieu Guibert
- Inserm UMRS 935, Université Paris Sud, Villejuif, France; ESTeam Paris Sud, Infrastructure INGESTEM, Villejuif, France
| | - Roberta Francesca De Rose
- Inserm UMRS 935, Université Paris Sud, Villejuif, France; Department of Health Sciences, University of Catanzaro, Catanzaro, Italy
| | - Paule Opolon
- Gustave Roussy, Laboratoire de Pathologie Expérimentale, F-94800 Villejuif, France
| | - Dominique Divers
- Inserm UMRS 935, Université Paris Sud, Villejuif, France; ESTeam Paris Sud, Infrastructure INGESTEM, Villejuif, France
| | - Emilie Gobbo
- ESTeam Paris Sud, Infrastructure INGESTEM, Villejuif, France
| | - Frank Griscelli
- Inserm UMRS 935, Université Paris Sud, Villejuif, France; ESTeam Paris Sud, Infrastructure INGESTEM, Villejuif, France; Paris Descartes University, & Gustave Roussy, Villejuif, France
| | - Martin Schlumberger
- Gustave Roussy, Laboratoire de Pathologie Expérimentale, F-94800 Villejuif, France
| | - Annelise Bennaceur-Griscelli
- Inserm UMRS 935, Université Paris Sud, Villejuif, France; ESTeam Paris Sud, Infrastructure INGESTEM, Villejuif, France; APHP, Division of Hematology-of Paris Sud University Hospitals, University Paris Sud, Le Kremlin Bicêtre, France
| | - Ali G Turhan
- Inserm UMRS 935, Université Paris Sud, Villejuif, France; ESTeam Paris Sud, Infrastructure INGESTEM, Villejuif, France; APHP, Division of Hematology-of Paris Sud University Hospitals, University Paris Sud, Le Kremlin Bicêtre, France.
| |
Collapse
|
37
|
Chan WH, Anderson CR, Gonsalvez DG. From proliferation to target innervation: signaling molecules that direct sympathetic nervous system development. Cell Tissue Res 2017; 372:171-193. [PMID: 28971249 DOI: 10.1007/s00441-017-2693-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023]
Abstract
The sympathetic division of the autonomic nervous system includes a variety of cells including neurons, endocrine cells and glial cells. A recent study (Furlan et al. 2017) has revised thinking about the developmental origin of these cells. It now appears that sympathetic neurons and chromaffin cells of the adrenal medulla do not have an immediate common ancestor in the form a "sympathoadrenal cell", as has been long believed. Instead, chromaffin cells arise from Schwann cell precursors. This review integrates the new findings with the expanding body of knowledge on the signalling pathways and transcription factors that regulate the origin of cells of the sympathetic division of the autonomic nervous system.
Collapse
Affiliation(s)
- W H Chan
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia
| | - C R Anderson
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia
| | - David G Gonsalvez
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia.
| |
Collapse
|
38
|
Murtazina AR, Dilmukhametova LK, Nikishina YO, Sapronova AY, Volina EV, Ugrumov MV. Changes in the secretory activity of organs producing noradrenaline upon inhibition of its synthesis in neonatal rat brain. Russ J Dev Biol 2017. [DOI: 10.1134/s1062360417050058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
39
|
Gómez-Paz A, Drucker-Colín R, Milán-Aldaco D, Palomero-Rivero M, Ambriz-Tututi M. Intrastriatal Chromospheres' Transplant Reduces Nociception in Hemiparkinsonian Rats. Neuroscience 2017; 387:123-134. [PMID: 28890053 DOI: 10.1016/j.neuroscience.2017.08.052] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 08/20/2017] [Accepted: 08/29/2017] [Indexed: 10/18/2022]
Abstract
The present study evaluates the possible antinociceptive effect of chromosphere transplants in rats injected with 6-hydroxydopamine (6-OHDA), a model of Parkinson's disease. Male adult Wistar rats received 40μg/0.5μl of 6-OHDA or 0.5μl of vehicle into the left substantia nigra (SNc). Rats were evaluated for mechanical allodynia, cold allodynia, thermal hyperalgesia and formalin. Rats with altered nociceptive threshold were transplanted with chromospheres. After transplant, rats were evaluated every week. Our results confirm that 6-OHDA injection into rat's SNc reduces mechanical, thermal, and chemical thresholds. Interestingly, chromospheres' transplant reverted 6-OHDA-induced allodynia and hyperalgesia. The antinociceptive effect induced by chromospheres was dopamine D2- and opioid-receptor dependent since sulpiride or naltrexone reverted its effect.
Collapse
Affiliation(s)
- Alejandra Gómez-Paz
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico
| | - René Drucker-Colín
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico
| | - Diana Milán-Aldaco
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico
| | - Marcela Palomero-Rivero
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico
| | - Mónica Ambriz-Tututi
- Hospital General Ajusco Medio "Dra. Obdulia Rodriguez Rodriguez", Unidad de, Trastornos de Movimiento y Sueño, Mexico.
| |
Collapse
|
40
|
Bondarenko NS, Murtazina AR, Nikishina YO, Sapronova AY, Ugrumov MV. Molecular mechanisms of synthesis of noradrenaline as an inducer of development in the adrenal glands of rats in ontogenesis. DOKL BIOCHEM BIOPHYS 2017; 472:23-26. [DOI: 10.1134/s1607672917010070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Indexed: 11/23/2022]
|
41
|
Bondarenko NS, Dilmukhametova LK, Kurina AY, Murtazina AR, Sapronova AY, Sysoeva AP, Ugrumov MV. Plasticity of central and peripheral sources of noradrenaline in rats during ontogenesis. BIOCHEMISTRY (MOSCOW) 2017; 82:373-379. [DOI: 10.1134/s0006297917030166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
42
|
Steenblock C, Rubin de Celis MF, Androutsellis-Theotokis A, Sue M, Delgadillo Silva LF, Eisenhofer G, Andoniadou CL, Bornstein SR. Adrenal cortical and chromaffin stem cells: Is there a common progeny related to stress adaptation? Mol Cell Endocrinol 2017; 441:156-163. [PMID: 27637345 DOI: 10.1016/j.mce.2016.09.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
The adrenal gland is a highly plastic organ with the capacity to adapt the body homeostasis to different physiological needs. The existence of stem-like cells in the adrenal cortex has been revealed in many studies. Recently, we identified and characterized in mice a pool of glia-like multipotent Nestin-expressing progenitor cells, which contributes to the plasticity of the adrenal medulla. In addition, we found that these Nestin progenitors are actively involved in the stress response by giving rise to chromaffin cells. Interestingly, we also observed a Nestin-GFP-positive cell population located under the adrenal capsule and scattered through the cortex. In this article, we discuss the possibility of a common progenitor giving rise to subpopulations of cells both in the adrenal cortex and medulla, the isolation and characterization of this progenitor as well as its clinical potential in transplantation therapies and in pathophysiology.
Collapse
Affiliation(s)
- Charlotte Steenblock
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany.
| | | | - Andreas Androutsellis-Theotokis
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany; Stem Cells, Tissue Engineering and Modelling (STEM), Division of Cancer and Stem Cells, University of Nottingham, Nottingham, UK
| | - Mariko Sue
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | | | - Graeme Eisenhofer
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Cynthia L Andoniadou
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany; Department of Craniofacial Development and Stem Cell Biology, King's College London, London, UK
| | - Stefan R Bornstein
- Department of Internal Medicine III, Technische Universität Dresden, Dresden, Germany; Department of Endocrinology and Diabetes, King's College London, London, UK
| |
Collapse
|
43
|
Kurolap A, Armbruster A, Hershkovitz T, Hauf K, Mory A, Paperna T, Hannappel E, Tal G, Nijem Y, Sella E, Mahajnah M, Ilivitzki A, Hershkovitz D, Ekhilevitch N, Mandel H, Eulenburg V, Baris HN. Loss of Glycine Transporter 1 Causes a Subtype of Glycine Encephalopathy with Arthrogryposis and Mildly Elevated Cerebrospinal Fluid Glycine. Am J Hum Genet 2016; 99:1172-1180. [PMID: 27773429 DOI: 10.1016/j.ajhg.2016.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/07/2016] [Indexed: 01/02/2023] Open
Abstract
Glycine is a major neurotransmitter that activates inhibitory glycine receptors and is a co-agonist for excitatory glutamatergic N-methyl-D-aspartate (NMDA) receptors. Two transporters, GLYT1 and GLYT2, regulate extracellular glycine concentrations within the CNS. Dysregulation of the extracellular glycine has been associated with hyperekplexia and nonketotic hyperglycinemia. Here, we report four individuals from two families who presented at birth with facial dysmorphism, encephalopathy, arthrogryposis, hypotonia progressing to hypertonicity with startle-like clonus, and respiratory failure. Only one individual survived the respiratory failure and was weaned off ventilation but has significant global developmental delay. Mildly elevated cerebrospinal fluid (CSF) glycine and normal serum glycine were observed in two individuals. In both families, we identified truncating mutations in SLC6A9, encoding GLYT1. We demonstrate that pharmacologic or genetic abolishment of GlyT1 activity in mice leads to mildly elevated glycine in the CSF but not in blood. Additionally, previously reported slc6a9-null mice and zebrafish mutants also display phenotypes consistent with the affected individuals we examined. Our data suggest that truncating SLC6A9 mutations lead to a distinct human neurological syndrome hallmarked by mildly elevated CSF glycine and normal serum glycine.
Collapse
|
44
|
Nikishina YO, Murtazina AR, Sapronova AY, Melnikova VI, Bondarenko NS, Ugryumov MV. Reciprocal humoral regulation of endocrine noradrenaline sources in perinatal development of rats. Russ J Dev Biol 2016. [DOI: 10.1134/s1062360416050076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Intrastriatal Grafting of Chromospheres: Survival and Functional Effects in the 6-OHDA Rat Model of Parkinson's Disease. PLoS One 2016; 11:e0160854. [PMID: 27525967 PMCID: PMC4985142 DOI: 10.1371/journal.pone.0160854] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/26/2016] [Indexed: 11/19/2022] Open
Abstract
Cell replacement therapy in Parkinson’s disease (PD) aims at re-establishing dopamine neurotransmission in the striatum by grafting dopamine-releasing cells. Chromaffin cell (CC) grafts produce some transitory improvements of functional motor deficits in PD animal models, and have the advantage of allowing autologous transplantation. However, CC grafts have exhibited low survival, poor functional effects and dopamine release compared to other cell types. Recently, chromaffin progenitor-like cells were isolated from bovine and human adult adrenal medulla. Under low-attachment conditions, these cells aggregate and grow as spheres, named chromospheres. Here, we found that bovine-derived chromosphere-cell cultures exhibit a greater fraction of cells with a dopaminergic phenotype and higher dopamine release than CC. Chromospheres grafted in a rat model of PD survived in 57% of the total grafted animals. Behavioral tests showed that surviving chromosphere cells induce a reduction in motor alterations for at least 3 months after grafting. Finally, we found that compared with CC, chromosphere grafts survive more and produce more robust and consistent motor improvements. However, further experiments would be necessary to determine whether the functional benefits induced by chromosphere grafts can be improved, and also to elucidate the mechanisms underlying the functional effects of the grafts.
Collapse
|
46
|
Bondarenko NS, Murtazina AR, Dil'mukhametova LK, Ikonopistseva MA, Volina EV, Ugrumov MV. Secretory activity of the brain and peripheral organs: Spontaneous and stimulated release of noradrenaline in the ontogenesis of rats. DOKL BIOCHEM BIOPHYS 2016; 467:153-6. [PMID: 27193722 DOI: 10.1134/s1607672916020204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Indexed: 11/22/2022]
Abstract
Spontaneous and K(+)-stimulated release of noradrenaline from the hypothalamus, adrenal gland, and organ of Zuckerkandl under their flowing incubation was investigated in the perinatal period of ontogenesis of rats. The results suggest that, during the investigated period of ontogenesis, adrenal glands are the main source of noradrenaline in the blood, whereas the contributions of the organ of Zuckerkandl and the brain are not as significant and change during this period.
Collapse
Affiliation(s)
- N S Bondarenko
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia.
| | - A R Murtazina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia
| | - L K Dil'mukhametova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia
| | | | - E V Volina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia
| | - M V Ugrumov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia.,National Research University "Higher School of Economics,", Moscow, Russia
| |
Collapse
|
47
|
MiR-124 is differentially expressed in derivatives of the sympathoadrenal cell lineage and promotes neurite elongation in chromaffin cells. Cell Tissue Res 2016; 365:225-32. [DOI: 10.1007/s00441-016-2395-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/09/2016] [Indexed: 01/02/2023]
|
48
|
Murtazina AR, Nikishina YO, Bondarenko NS, Sapronova AJ, Ugrumov MV. Signal molecules during the organism development: Central and peripheral sources of noradrenaline in rat ontogenesis. DOKL BIOCHEM BIOPHYS 2016; 466:74-6. [PMID: 27025493 DOI: 10.1134/s160767291601018x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Indexed: 11/23/2022]
Abstract
Using the method of high performance liquid chromatography with electrochemical detection, the age dynamics of the content of noradrenaline (NA) in the brain, adrenal gland, and the organ of Zuckerkandl in prenatal (18th and 21st days of embryogenesis) and early postnatal (3, 7, 15, and 30th days) periods of development was studied. The potential contribution of these organs to the formation of physiologically active concentration of noradrenalin in the blood was also assessed. The results suggest that, during the development of the organism, the activity of the sources of noradrenaline in the general circulation changes, which gives a reason to assume the existence of humoral interaction between NA-producing organs in the perinatal period of ontogenesis.
Collapse
Affiliation(s)
- A R Murtazina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia.
| | - Y O Nikishina
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia
| | - N S Bondarenko
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia
| | - A Ja Sapronova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia
| | - M V Ugrumov
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, ul. Vavilova 26, Moscow, 119334, Russia
| |
Collapse
|
49
|
Morrison MA, Zimmerman MW, Look AT, Stewart RA. Studying the peripheral sympathetic nervous system and neuroblastoma in zebrafish. Methods Cell Biol 2016; 134:97-138. [PMID: 27312492 DOI: 10.1016/bs.mcb.2015.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The zebrafish serves as an excellent model to study vertebrate development and disease. Optically clear embryos, combined with tissue-specific fluorescent reporters, permit direct visualization and measurement of peripheral nervous system formation in real time. Additionally, the model is amenable to rapid cellular, molecular, and genetic approaches to determine how developmental mechanisms contribute to disease states, such as cancer. In this chapter, we describe the development of the peripheral sympathetic nervous system (PSNS) in general, and our current understanding of genetic pathways important in zebrafish PSNS development specifically. We also illustrate how zebrafish genetics is used to identify new mechanisms controlling PSNS development and methods for interrogating the potential role of PSNS developmental pathways in neuroblastoma pathogenesis in vivo using the zebrafish MYCN-driven neuroblastoma model.
Collapse
Affiliation(s)
- M A Morrison
- University of Utah, Salt Lake City, UT, United States
| | | | - A T Look
- Harvard Medical School, Boston, MA, United States
| | - R A Stewart
- University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
50
|
Generating trunk neural crest from human pluripotent stem cells. Sci Rep 2016; 6:19727. [PMID: 26812940 PMCID: PMC4728437 DOI: 10.1038/srep19727] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/17/2015] [Indexed: 12/17/2022] Open
Abstract
Neural crest cells (NCC) are stem cells that generate different lineages, including neuroendocrine, melanocytic, cartilage, and bone. The differentiation potential of NCC varies according to the level from which cells emerge along the neural tube. For example, only anterior “cranial” NCC form craniofacial bone, whereas solely posterior “trunk” NCC contribute to sympathoadrenal cells. Importantly, the isolation of human fetal NCC carries ethical and scientific challenges, as NCC induction typically occur before pregnancy is detectable. As a result, current knowledge of NCC biology derives primarily from non-human organisms. Important differences between human and non-human NCC, such as expression of HNK1 in human but not mouse NCC, suggest a need to study human NCC directly. Here, we demonstrate that current protocols to differentiate human pluripotent stem cells (PSC) to NCC are biased toward cranial NCC. Addition of retinoic acid drove trunk-related markers and HOX genes characteristic of a posterior identity. Subsequent treatment with bone morphogenetic proteins (BMPs) enhanced differentiation to sympathoadrenal cells. Our approach provides methodology for detailed studies of human NCC, and clarifies roles for retinoids and BMPs in the differentiation of human PSC to trunk NCC and to sympathoadrenal lineages.
Collapse
|