1
|
Fadl S, Saleh AMM, Abou-Elmagd A, Abdel-Maksoud FM. Prehatching development of the adrenal gland in Japanese quail (Coturnix japonica): Histological, immunohistochemical, and electron microscopic studies. Microsc Res Tech 2024; 87:727-739. [PMID: 37990954 DOI: 10.1002/jemt.24462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/09/2023] [Accepted: 11/11/2023] [Indexed: 11/23/2023]
Abstract
The adrenal glands play a key role in maintaining the physiological balance of birds and helping them to survive environmental changes. The objective of the present work was to give a detailed investigation of the histological, ultrastructural, and immunohistochemical findings of the adrenal gland in Japanese quail during the prehatching phase. The current study was performed on 45 healthy Japanese quail embryos at different prehatching periods. Our results showed the primordium of the quail's adrenocortical tissue appeared at 3 days of incubation as a thickening of the splanchnic mesoderm. The prospective chromaffin cells appeared at 5 days as clusters of cells migrated from the neural crest cells along the dorsal aorta toward the interrenal tissue. TH immunoreactivity was observed in the neural crest cells during their migration toward the adrenal primordium. Furthermore, these TH immunopositive cells were intermingled with the developing interrenal cell cords that developed from the coelomic epithelium. NSE immunostaining was detected within the cytoplasm of interrenal cells, chromaffin cells, and ganglion cells. Sox10 is expressed in chromaffin and ganglion cells with different staining intensities. On the 13th day of prehatching, both interrenal and chromaffin cells were β-catenin immunonegative, but on the 17th day, both cells were immunopositively. Our findings show that during prenatal life, the adrenal gland undergoes significant morphological changes. Together, the present data suggest that studying the prenatal development of the adrenal gland in birds is important for advancing our understanding of this critical organ and its functions. RESEARCH HIGHLIGHTS: The present study aimed to give a detailed study of the histological, ultrastructural, and immunohistochemical investigations of the adrenal gland in Japanese quail during the prehatching period. The interrenal primordium was observed on the third embryonic day, on the fifth ED the primordium of the chromaffin tissue appeared as row of migrating neural crest cell. At the ultrastructural level, the interrenal cells take steroid-secreting cells characters, they have varying amounts of lipid droplets and abundant mitochondria at 15th ED contained moderate number of lysosomes and mitochondria.
Collapse
Affiliation(s)
- Saher Fadl
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Abdelmohaimen M M Saleh
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Ahmed Abou-Elmagd
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Fatma M Abdel-Maksoud
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
2
|
Soukup J, Manethova M, Stejskal V, Novakova M, Duskova J, Hornychova H, Hacova M, Staniczkova-Zambo I, Zelinka T, Kosak M, Cesak T, Netuka D, Ryska A, Gabalec F. Hand2 Immunohistochemistry in the Diagnosis of Paragangliomas and Other Neuroendocrine Neoplasms. Endocr Pathol 2024; 35:14-24. [PMID: 38416360 DOI: 10.1007/s12022-024-09803-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/08/2024] [Indexed: 02/29/2024]
Abstract
Hand2 is a core transcription factor responsible for chromaffin cell differentiation. However, its potential utility in surgical pathology has not been studied. Thus, we aimed to investigate its expression in paragangliomas, other neuroendocrine neoplasms (NENs), and additional non-neuroendocrine tumors. We calibrated Hand2 immunohistochemistry on adrenal medulla cells and analyzed H-scores in 46 paragangliomas (PGs), 9 metastatic PGs, 21 cauda equina neuroendocrine tumors (CENETs), 48 neuroendocrine carcinomas (NECs), 8 olfactory neuroblastomas (ONBs), 110 well-differentiated NETs (WDNETs), 10 adrenal cortical carcinomas, 29 adrenal cortical adenomas, 8 melanomas, 41 different carcinomas, and 10 gastrointestinal stromal tumors (GISTs). Both tissue microarrays (TMAs) and whole sections (WSs) were studied. In 171 NENs, previously published data on Phox2B and GATA3 were correlated with Hand2. Hand2 was positive in 98.1% (54/55) PGs, but only rarely in WDNETs (9.6%, 10/104), CENETs (9.5%, 2/21), NECs (4.2%, 2/48), or ONBs (12.5%, 1/8). Any Hand2 positivity was 98.1% sensitive and 91.7% specific for the diagnosis of PG. The Hand2 H-score was significantly higher in primary PGs compared to Hand2-positive WDNETs (median 166.3 vs. 7.5; p < 0.0001). Metastatic PGs were positive in 88.9% (8/9). No Hand2 positivity was observed in any adrenal cortical neoplasm or other non-neuroendocrine tumors, with exception of 8/10 GISTs. Parasympathetic PGs showed a higher Hand2 H-score compared to sympathetic PGs (median H-scores 280 vs. 104, p < 0.0001). Hand2 positivity in NENs serves as a reliable marker of primary and metastatic PG, since other NENs only rarely exhibit limited Hand2 positivity.
Collapse
Affiliation(s)
- Jiri Soukup
- Department of Pathology, Military University Hospital Prague, U Vojenske Nemocnice 1200, Praha 6, 169 02, Prague, Czech Republic.
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic.
- Department of Pathology, Charles University, First Faculty of Medicine and General University Hospital in Prague, Studnickova, 2039, 128 00, Nové Mesto, Prague, Czech Republic.
| | - Monika Manethova
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - Vaclav Stejskal
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - Marie Novakova
- Department of Pathology, Military University Hospital Prague, U Vojenske Nemocnice 1200, Praha 6, 169 02, Prague, Czech Republic
| | - Jaroslava Duskova
- Department of Pathology, Charles University, First Faculty of Medicine and General University Hospital in Prague, Studnickova, 2039, 128 00, Nové Mesto, Prague, Czech Republic
| | - Helena Hornychova
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - Maria Hacova
- Department of Pathology, The Regional Hospital Pardubice, Pardubice, Czech Republic
| | - Iva Staniczkova-Zambo
- 1st Department of Pathology, St. Anne's University Hospital and Faculty of Medicine , Masaryk University, Pekarská 664/53, 602 00, Brno-stred, Brno, Czech Republic
| | - Tomas Zelinka
- Centre for Hypertension, 3rd Department of Medicine, Charles University, First Faculty of Medicine and General University Hospital in Prague, Prague, Czech Republic
| | - Mikulas Kosak
- Department of Internal Medicine, First Faculty of Medicine, Charles University and Military University Hospital, Prague, Czech Republic
| | - Tomas Cesak
- Department of Neurosurgery, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - David Netuka
- Department of Neurosurgery and Neurooncology, 1st Medical Faculty, Charles University, Military University Hospital Prague, U Vojenske Nemocnice 1200, Praha 6, 169 02, Prague, Czech Republic
| | - Ales Ryska
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| | - Filip Gabalec
- 4th Department of Internal Medicine, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Králové, Sokolska 582, 500 05, Hradec Kralove, Czech Republic
| |
Collapse
|
3
|
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
|
4
|
Capaldo A. The Adrenal Gland of Squamata (Reptilia): A Comparative Overview. Animals (Basel) 2023; 13:2686. [PMID: 37684950 PMCID: PMC10486442 DOI: 10.3390/ani13172686] [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: 07/27/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
The adrenal gland is a complex endocrine organ composed of two components: a steroidogenic tissue, which produces steroid hormones, and a chromaffin tissue, which mainly produces norepinephrine and epinephrine. Through evolution, their relationships with each other changed. They begin as isolated chromaffin and steroidogenic cell aggregates, typical of fish, and end with the advanced compact gland, typical of mammals, which consists of an external steroidogenic cortical zone and an internal chromaffin medullary zone. The adrenal gland of reptiles is unique because, with few exceptions, it is near the gonads and genital ducts, and the chromaffin and steroidogenic tissues are closely associated. However, the degree of mixing is variable. For example, in Squamata, the mixing degree of chromaffin and steroidogenic tissues, their reciprocal position in the gland, and the relative quantities of norepinephrine and epinephrine secreted by the chromaffin cells are extremely variable. This variability could be related to the phylogenetic history of the species. After a brief discussion of the adrenal gland and its main functions in vertebrates, this overview will examine the general characteristics of the adrenal gland of squamates, the differences in morphology of the gland, and the possible relationships with the phylogeny of the different species.
Collapse
Affiliation(s)
- Anna Capaldo
- Department of Biology, University of Naples Federico II, Via Cinthia, Edificio 7, 80126 Naples, Italy
| |
Collapse
|
5
|
Phox2B is a sensitive and reliable marker of paraganglioma-Phox2B immunohistochemistry in diagnosis of neuroendocrine neoplasms. Virchows Arch 2023; 482:679-686. [PMID: 36656393 DOI: 10.1007/s00428-023-03490-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/14/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023]
Abstract
Phox2B is a transcription factor responsible for chromaffin cell phenotype. Although it is used routinely for diagnosis of neuroblastoma, previous reports concerning its utility in the diagnosis of neuroendocrine neoplasms have been conflicting. We assessed Phox2b immunoreactivity in different neuroendocrine neoplasms. Tissue microarrays or whole sections of 36 paragangliomas (PGs), 91 well-differentiated neuroendocrine tumours of different organs (WDNETs), 31 neuroendocrine carcinomas (NECs), and 6 olfactory neuroblastomas (ONBs) were stained with Phox2B antibody (EP312) and GATA3. The percentage of positive cells and intensity was analysed using H-score. Phox2B immunoreactivity was seen in 97.2% (35/36) PGs, 11% (10/91) WDNETs, 9.7% (3/31) NECs, and 16.7% (1/6) ONBs. PGs were significantly more often positive (p < 0.001, χ2) than other neuroendocrine tumours, showing highest H-score (mean 144.9, SD ± 75.1) and percentage of positive cells (median 81.3%, IQR 62.5-92.5%). Compared to Phox2B-positive WDNETs, PGs showed significantly higher H-score (median 145 vs 7.5, p < 0.001) and percentage of positive cells (median 82.5% vs 4.5%, p < 0.001). Phox2B positivity was 97.2% sensitive and 89% specific for the diagnosis of PG. GATA3 was 100% sensitive and 88% specific for the diagnosis of PG. When combined, any Phox2B/GATA3 coexpression was 97.1% sensitive and 99.1% specific for the diagnosis of paraganglioma. Widespread Phox2B immunoreactivity is a highly characteristic feature of PGs and it can be used as an additional marker in differential diagnosis of neuroendocrine tumours.
Collapse
|
6
|
Parasiliti-Caprino M, Lopez C, Bollati M, Bioletto F, Sola C, Di Carlo MC, Ponzetto F, Gesmundo I, Settanni F, Ghigo E, Mengozzi G, Maccario M, Giordano R. A retrospective study on the association between urine metanephrines and cardiometabolic risk in patients with nonfunctioning adrenal incidentaloma. Sci Rep 2022; 12:14913. [PMID: 36050396 PMCID: PMC9436965 DOI: 10.1038/s41598-022-19321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/26/2022] [Indexed: 11/09/2022] Open
Abstract
Several studies argued that cardiovascular evaluation of patients with nonfunctioning adrenal incidentaloma is of particular importance. Therefore, we aimed to evaluate the possibility of stratifying the cardiometabolic risk using metanephrine levels in this setting of patients. A retrospective cross-sectional study was designed, collecting data of metanephrine values in 828 patients with nonfunctioning adrenal incidentaloma, referred to our Division within the University of Turin between 2007 and 2021. The univariate analysis showed associations between urine metanephrines and cardiometabolic variables/parameters, particularly considering the noradrenaline metabolite. At the univariate regression, normetanephrine was associated with metabolic syndrome (OR = 1.13, p = 0.002), hypertensive cardiomyopathy (OR = 1.09, p = 0.026), microalbuminuria (OR = 1.14, p = 0.024), and eGFR < 60 mL/min/1.73 m2 (OR = 1.11, p = 0.013), while metanephrine was associated with microalbuminuria (OR = 1.50, p = 0.008). At multivariate regression, considering all major cardiovascular risk factors as possible confounders, normetanephrine retained a significant association with metabolic syndrome (OR = 1.10, p = 0.037). Moreover, metanephrine retained a significant association with the presence of microalbuminuria (OR = 1.66, p = 0.003). The present study showed a further role for metanephrines in the cardiovascular risk stratification of patients with nonfunctioning adrenal incidentaloma. Individuals with high levels of these indirect markers of sympathetic activity should be carefully monitored and may benefit from an aggressive treatment to reduce their additional cardiometabolic burden.
Collapse
Affiliation(s)
- Mirko Parasiliti-Caprino
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy.
| | - Chiara Lopez
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Martina Bollati
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Fabio Bioletto
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Chiara Sola
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Maria Chiara Di Carlo
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Federico Ponzetto
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Iacopo Gesmundo
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Fabio Settanni
- Clinical Biochemistry Laboratory, City of Health and Science University Hospital, Turin, Italy
| | - Ezio Ghigo
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Giulio Mengozzi
- Clinical Biochemistry Laboratory, City of Health and Science University Hospital, Turin, Italy
| | - Mauro Maccario
- Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, City of Health and Science University Hospital, University of Turin, Corso Dogliotti 14, 10126, Turin, Italy
| | - Roberta Giordano
- Department of Biological and Clinical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
7
|
Bechmann N, Berger I, Bornstein SR, Steenblock C. Adrenal medulla development and medullary-cortical interactions. Mol Cell Endocrinol 2021; 528:111258. [PMID: 33798635 DOI: 10.1016/j.mce.2021.111258] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/12/2021] [Accepted: 03/22/2021] [Indexed: 01/10/2023]
Abstract
The mammalian adrenal gland is composed of two distinct tissue types in a bidirectional connection, the catecholamine-producing medulla derived from the neural crest and the mesoderm-derived cortex producing steroids. The medulla mainly consists of chromaffin cells derived from multipotent nerve-associated descendants of Schwann cell precursors. Already during adrenal organogenesis, close interactions between cortex and medulla are necessary for proper differentiation and morphogenesis of the gland. Moreover, communication between the cortex and the medulla ensures a regular function of the adult adrenal. In tumor development, interfaces between the two parts are also common. Here, we summarize the development of the mammalian adrenal medulla and the current understanding of the cortical-medullary interactions under development and in health and disease.
Collapse
Affiliation(s)
- Nicole Bechmann
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Ilona Berger
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Diabetes and Nutritional Sciences Division, King's College London, London, UK
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
8
|
Jansky S, Sharma AK, Körber V, Quintero A, Toprak UH, Wecht EM, Gartlgruber M, Greco A, Chomsky E, Grünewald TGP, Henrich KO, Tanay A, Herrmann C, Höfer T, Westermann F. Single-cell transcriptomic analyses provide insights into the developmental origins of neuroblastoma. Nat Genet 2021; 53:683-693. [PMID: 33767450 DOI: 10.1038/s41588-021-00806-1] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 01/29/2021] [Indexed: 01/31/2023]
Abstract
Neuroblastoma is a pediatric tumor of the developing sympathetic nervous system. However, the cellular origin of neuroblastoma has yet to be defined. Here we studied the single-cell transcriptomes of neuroblastomas and normal human developing adrenal glands at various stages of embryonic and fetal development. We defined normal differentiation trajectories from Schwann cell precursors over intermediate states to neuroblasts or chromaffin cells and showed that neuroblastomas transcriptionally resemble normal fetal adrenal neuroblasts. Importantly, neuroblastomas with varying clinical phenotypes matched different temporal states along normal neuroblast differentiation trajectories, with the degree of differentiation corresponding to clinical prognosis. Our work highlights the roles of oncogenic MYCN and loss of TFAP2B in blocking differentiation and may provide the basis for designing therapeutic interventions to overcome differentiation blocks.
Collapse
Affiliation(s)
- Selina Jansky
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Ashwini Kumar Sharma
- Health Data Science Unit, Medical Faculty University Heidelberg and BioQuant, Heidelberg, Germany
| | - Verena Körber
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrés Quintero
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Health Data Science Unit, Medical Faculty University Heidelberg and BioQuant, Heidelberg, Germany
| | - Umut H Toprak
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elisa M Wecht
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Moritz Gartlgruber
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alessandro Greco
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.,Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elad Chomsky
- Department of Computer Science and Applied Mathematics and Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Thomas G P Grünewald
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kai-Oliver Henrich
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Amos Tanay
- Department of Computer Science and Applied Mathematics and Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Carl Herrmann
- Health Data Science Unit, Medical Faculty University Heidelberg and BioQuant, Heidelberg, Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Westermann
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany. .,Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
9
|
Godefroy D, Boukhzar L, Dubessy C, Montero-Hadjadje M, Yon L, Eiden LE, Anouar Y. Three-dimensional mapping of tyrosine hydroxylase in the transparent brain and adrenal of prenatal and pre-weaning mice: Comprehensive methodological flowchart and quantitative aspects of 3D mapping. J Neurosci Methods 2020; 335:108596. [DOI: 10.1016/j.jneumeth.2020.108596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 10/25/2022]
|
10
|
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
|
11
|
Abstract
The mitochondrial enzyme succinate dehydrogenase (SDH) acts as a tumor suppressor. Biallelic inactivation of one of the genes encoding for SDH subunits (collectively named SDHx) leads to complete loss of the protein function and the development of diverse group of tumors. Pheochromocytomas-paragangliomas are the prime example of hereditary tumors caused by SDH deficiency. In this review, we discuss the roles of imaging examinations, and illustrate new insights into genotype-imaging phenotype relationships.
Collapse
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France
| | - Henri Timmers
- Department of Internal Medicine, Division of Endocrinology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
12
|
Mohlin S, Kunttas E, Persson CU, Abdel-Haq R, Castillo A, Murko C, Bronner ME, Kerosuo L. Maintaining multipotent trunk neural crest stem cells as self-renewing crestospheres. Dev Biol 2019; 447:137-146. [PMID: 30664880 DOI: 10.1016/j.ydbio.2019.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 01/15/2023]
Abstract
Neural crest cells have broad migratory and differentiative ability that differs according to their axial level of origin. However, their transient nature has limited understanding of their stem cell and self-renewal properties. While an in vitro culture method has made it possible to maintain cranial neural crest cells as self-renewing multipotent crestospheres (Kerosuo et al., 2015), these same conditions failed to preserve trunk neural crest in a stem-like state. Here we optimize culture conditions for maintenance of avian trunk crestospheres, comprised of both neural crest stem and progenitor cells. Our trunk-derived crestospheres are multipotent and display self-renewal capacity over several weeks. Trunk crestospheres display elevated expression of neural crest cell markers as compared to those characteristic of ventrolateral neural tube or mesodermal fates. Moreover, trunk crestospheres express increased levels of trunk neural crest-enriched markers as compared to cranial crestospheres. Finally, we use lentiviral transduction as a tool to manipulate gene expression in trunk crestospheres. Taken together, this method enables long-term in vitro maintenance and manipulation of multipotent trunk neural crest cells in a premigratory stem or early progenitor state. Trunk crestospheres are a valuable resource for probing mechanisms underlying neural crest stemness and lineage decisions as well as accompanying diseases.
Collapse
Affiliation(s)
- Sofie Mohlin
- Department of Pediatrics, Clinical Sciences, Lund University, Lund, Sweden.
| | - Ezgi Kunttas
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Camilla U Persson
- Translational Cancer Research, Lund University Cancer Center at Medicon Village, Lund University, Lund, Sweden
| | - Reem Abdel-Haq
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Aldo Castillo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Christina Murko
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Laura Kerosuo
- Neural Crest Development and Disease Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA; Department of Biochemistry and Developmental Biology, Medicum, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
13
|
Oakley RH, Ramamoorthy S, Foley JF, Busada JT, Lu NZ, Cidlowski JA. Glucocorticoid receptor isoform-specific regulation of development, circadian rhythm, and inflammation in mice. FASEB J 2018; 32:5258-5271. [PMID: 29672221 DOI: 10.1096/fj.201701153r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Glucocorticoids are primary stress hormones, and their synthetic derivatives are widely used clinically. The therapeutic efficacy of these steroids is limited by side effects and glucocorticoid resistance. Multiple glucocorticoid receptor (GR) isoforms are produced from a single gene by alternative translation initiation; however, the role individual isoforms play in tissue-specific responses to glucocorticoids is unknown. We have generated knockin mice that exclusively express the most active receptor isoform, GR-C3. GR-C3 knockin mice die at birth due to respiratory distress. Microarray analysis of fibroblasts from wild-type and GR-C3 mice indicated that most genes regulated by GR-C3 were unique to this isoform. Antenatal glucocorticoid administration rescued GR-C3 knockin mice from neonatal death. Dual-energy X-ray absorptiometry revealed no major alterations in body composition for rescued knockin mice. Rescued female, but not male, GR-C3 mice exhibited increased wheel running activity in the light portion of the day. LPS administration induced premature mortality in rescued GR-C3 knockin mice, and gene expression studies revealed a deficiency in the ability of GR-C3 to repress a large cohort of immune and inflammatory response genes. These findings demonstrate that specific GR translational isoforms can influence development, circadian rhythm, and inflammation through the regulation of distinct gene networks.-Oakley, R. H., Ramamoorthy, S., Foley, J. F., Busada, J. T., Lu, N. Z., Cidlowski, J. A. Glucocorticoid receptor isoform-specific regulation of development, circadian rhythm, and inflammation in mice.
Collapse
Affiliation(s)
- Robert H Oakley
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Sivapriya Ramamoorthy
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Julie F Foley
- Cellular and Molecular Pathology Branch, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Jonathan T Busada
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Nick Z Lu
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - John A Cidlowski
- Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| |
Collapse
|
14
|
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
|
15
|
Zhu S, Thomas Look A. Neuroblastoma and Its Zebrafish Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:451-78. [PMID: 27165366 DOI: 10.1007/978-3-319-30654-4_20] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Neuroblastoma, an important developmental tumor arising in the peripheral sympathetic nervous system (PSNS), accounts for approximately 10 % of all cancer-related deaths in children. Recent genomic analyses have identified a spectrum of genetic alterations in this tumor. Amplification of the MYCN oncogene is found in 20 % of cases and is often accompanied by mutational activation of the ALK (anaplastic lymphoma kinase) gene, suggesting their cooperation in tumor initiation and spread. Understanding how complex genetic changes function together in oncogenesis has been a continuing and daunting task in cancer research. This challenge was addressed in neuroblastoma by generating a transgenic zebrafish model that overexpresses human MYCN and activated ALK in the PSNS, leading to tumors that closely resemble human neuroblastoma and new opportunities to probe the mechanisms that underlie the pathogenesis of this tumor. For example, coexpression of activated ALK with MYCN in this model triples the penetrance of neuroblastoma and markedly accelerates tumor onset, demonstrating the interaction of these modified genes in tumor development. Further, MYCN overexpression induces adrenal sympathetic neuroblast hyperplasia, blocks chromaffin cell differentiation, and ultimately triggers a developmentally-timed apoptotic response in the hyperplastic sympathoadrenal cells. In the context of MYCN overexpression, activated ALK provides prosurvival signals that block this apoptotic response, allowing continued expansion and oncogenic transformation of hyperplastic neuroblasts, thus promoting progression to neuroblastoma. This application of the zebrafish model illustrates its value in rational assessment of the multigenic changes that define neuroblastoma pathogenesis and points the way to future studies to identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Cancer Center and Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55902, USA.
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Santana MM, Rosmaninho-Salgado J, Cortez V, Pereira FC, Kaster MP, Aveleira CA, Ferreira M, Álvaro AR, Cavadas C. Impaired adrenal medullary function in a mouse model of depression induced by unpredictable chronic stress. Eur Neuropsychopharmacol 2015; 25:1753-66. [PMID: 26187454 DOI: 10.1016/j.euroneuro.2015.06.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/17/2015] [Accepted: 06/22/2015] [Indexed: 12/16/2022]
Abstract
Stress has been considered determinant in the etiology of depression. The adrenal medulla plays a key role in response to stress by releasing catecholamines, which are important to maintain homeostasis. We aimed to study the adrenal medulla in a mouse model of depression induced by 21 days of unpredictable chronic stress (UCS). We observed that UCS induced a differential and time-dependent change in adrenal medulla. After 7 days of UCS, mice did not show depressive-like behavior, but the adrenal medullae show increased protein and/or mRNA levels of catecholamine biosynthetic enzymes (TH, DβH and PNMT), Neuropeptide Y, the SNARE protein SNAP-25, the catecholamine transporter VMAT2 and the chromaffin progenitor cell markers, Mash1 and Phox2b. Moreover, 7 days of UCS induced a decrease in the chromaffin progenitor cell markers, Sox9 and Notch1. This suggests an increased capacity of chromaffin cells to synthesize, store and release catecholamines. In agreement, after 7 days, UCS mice had higher NE and EP levels in adrenal medulla. Opposite, when mice were submitted to 21 days of UCS, and showed a depressive like behavior, adrenal medullae had lower protein and/or mRNA levels of catecholamine biosynthetic enzymes (TH, DβH, PNMT), catecholamine transporters (NET, VMAT1), SNARE proteins (synthaxin1A, SNAP25, VAMP2), catecholamine content (EP, NE), and lower EP serum levels, indicating a reduction in catecholamine synthesis, re-uptake, storage and release. In conclusion, this study suggests that mice exposed to UCS for a period of 21 days develop a depressive-like behavior accompanied by an impairment of adrenal medullary function.
Collapse
Affiliation(s)
- Magda M Santana
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | | | - Vera Cortez
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Frederico C Pereira
- IBILI - Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal
| | - Manuella P Kaster
- Department of Biochemistry, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Célia A Aveleira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Marisa Ferreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ana Rita Álvaro
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
17
|
Coste C, Neirinckx V, Gothot A, Wislet S, Rogister B. Are neural crest stem cells the missing link between hematopoietic and neurogenic niches? Front Cell Neurosci 2015; 9:218. [PMID: 26136659 PMCID: PMC4469833 DOI: 10.3389/fncel.2015.00218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/22/2015] [Indexed: 12/24/2022] Open
Abstract
Hematopoietic niches are defined as cellular and molecular microenvironments that regulate hematopoietic stem cell (HSC) function together with stem cell autonomous mechanisms. Many different cell types have been characterized as contributors to the formation of HSC niches, such as osteoblasts, endothelial cells, Schwann cells, and mesenchymal progenitors. These mesenchymal progenitors have themselves been classified as CXC chemokine ligand (CXCL) 12-abundant reticular (CAR) cells, stem cell factor expressing cells, or nestin-positive mesenchymal stem cells (MSCs), which have been recently identified as neural crest-derived cells (NCSCs). Together, these cells are spatially associated with HSCs and believed to provide appropriate microenvironments for HSC self-renewal, differentiation, mobilization and hibernation both by cell-cell contact and soluble factors. Interestingly, it appears that regulatory pathways governing the hematopoietic niche homeostasis are operating in the neurogenic niche as well. Therefore, this review paper aims to compare both the regulation of hematopoietic and neurogenic niches, in order to highlight the role of NCSCs and nervous system components in the development and the regulation of the hematopoietic system.
Collapse
Affiliation(s)
- Cécile Coste
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Virginie Neirinckx
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - André Gothot
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Cardiovascular Sciences, University of Liège Liège, Belgium ; Hematology Department, University Hospital Liège, Belgium
| | - Sabine Wislet
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Bernard Rogister
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium ; Groupe Interdisciplinaire de Génoprotéomique Appliquée-Development, Stem Cells and Regenerative Medicine, University of Liège Liège, Belgium ; Neurology Department, University Hospital Liège, Belgium
| |
Collapse
|
18
|
Chan WH, Gonsalvez DG, Young HM, Southard-Smith EM, Cane KN, Anderson CR. Differences in CART expression and cell cycle behavior discriminate sympathetic neuroblast from chromaffin cell lineages in mouse sympathoadrenal cells. Dev Neurobiol 2015; 76:137-49. [PMID: 25989220 DOI: 10.1002/dneu.22304] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 04/22/2015] [Accepted: 05/13/2015] [Indexed: 12/31/2022]
Abstract
Adrenal medullary chromaffin cells and peripheral sympathetic neurons originate from a common sympathoadrenal (SA) progenitor cell. The timing and phenotypic changes that mark this lineage diversification are not fully understood. The present study investigated the expression patterns of phenotypic markers, and cell cycle dynamics, in the adrenal medulla and the neighboring suprarenal ganglion of embryonic mice. The noradrenergic marker, tyrosine hydroxylase (TH), was detected in both presumptive adrenal medulla and sympathetic ganglion cells, but with significantly stronger immunostaining in the former. There was intense cocaine and amphetamine-regulated transcript (CART) peptide immunostaining in most neuroblasts, whereas very few adrenal chromaffin cells showed detectable CART immunostaining. This phenotypic segregation appeared as early as E12.5, before anatomical segregation of the two cell types. Cell cycle dynamics were also examined. Initially, 88% of Sox10 positive (+) neural crest progenitors were proliferating at E10.5. Many SA progenitor cells withdrew from the cell cycle at E11.5 as they started to express TH. Whereas 70% of neuroblasts (TH+/CART+ cells) were back in the cell cycle at E12.5, only around 20% of chromaffin (CART negative) cells were in the cell cycle at E12.5 and subsequent days. Thus, chromaffin cell and neuroblast lineages showed differences in proliferative behavior from their earliest appearance. We conclude that the intensity of TH immunostaining and the expression of CART permit early discrimination of chromaffin cells and sympathetic neuroblasts, and that developing chromaffin cells exhibit significantly lower proliferative activity relative to sympathetic neuroblasts.
Collapse
Affiliation(s)
- Wing Hei Chan
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| | - David G Gonsalvez
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| | - Heather M Young
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| | - E Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, 529 Light Hall, 2215 Garland Avenue, Nashville, Tennessee
| | - Kylie N Cane
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| | - Colin R Anderson
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
19
|
Lumb R, Schwarz Q. Sympathoadrenal neural crest cells: the known, unknown and forgotten? Dev Growth Differ 2015; 57:146-57. [PMID: 25581786 DOI: 10.1111/dgd.12189] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 10/30/2014] [Accepted: 11/02/2014] [Indexed: 12/30/2022]
Abstract
Neural crest cells (NCCs) are highly migratory progenitor cells that give rise to a vast array of differentiated cell types. One of their key derivatives is the autonomic nervous system (ANS) that is comprised in part from chromaffin cells of the adrenal medulla and organ of Zuckerkandl, the sympathetic chain and additional prevertebral ganglia such as the celiac ganglia, suprarenal ganglia and mesenteric ganglia. In this review we discuss recent advances toward our understanding of how the NCC precursors of the ANS migrate to their target regions, how they are instructed to differentiate into the correct cell types, and the morphogenetic signals controlling their development. Many of these processes remain enigmatic to developmental biologists worldwide. Taking advantage of lineage tracing mouse models one of our own aims is to address the morphogenetic events underpinning the formation of the ANS and to identify the molecular mechanisms that help to segregate a mixed population of NCCs into pathways specific for the sympathetic ganglia, sensory ganglia or adrenal medulla.
Collapse
Affiliation(s)
- Rachael Lumb
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, 5000, Australia; Medical School, University of Adelaide, Adelaide, South Australia, 5000, Australia
| | | |
Collapse
|
20
|
Lenders JWM, Eisenhofer G. Pathophysiology and diagnosis of disorders of the adrenal medulla: focus on pheochromocytoma. Compr Physiol 2014; 4:691-713. [PMID: 24715564 DOI: 10.1002/cphy.c130034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The principal function of the adrenal medulla is the production and secretion of catecholamines. During stressful challenging conditions, catecholamines exert a pivotal homeostatic role. Although the main adrenomedullary catecholamine, epinephrine, has a wide array of adrenoreceptor-mediated effects, its absence does not cause life-threatening problems. In contrast, excess production of catecholamines due to an adrenomedullary tumor, specifically pheochromocytoma, results in significant morbidity and mortality. Despite being rare, pheochromocytoma has a notoriously bad reputation because of its potential devastating effects if undetected and untreated. The paroxysmal signs and symptoms and the risks of missing or delaying the diagnosis are well known for most physicians. Nevertheless, even today the diagnosis is still overlooked in a considerable number of patients. Prevention and complete cure are however possible by early diagnosis and appropriate treatment but these patients remain a challenge for physicians. Yet, biochemical proof of presence or absence of catecholamine excess has become more easy and straightforward due to developments in assay methodology. This also applies to radiological and functional imaging techniques for locating the tumor. The importance of genetic testing for underlying germline mutations in susceptibility genes for patients and relatives is increasingly recognized. Yet, the effectiveness of genetic testing, in terms of costs and benefits to health, has not been definitively established. Further improvement in knowledge of genotype-phenotype relationships in pheochromocytoma will open new avenues to a more rationalized and personalized diagnostic approach of affected patients.
Collapse
Affiliation(s)
- Jacques W M Lenders
- Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|
21
|
Abstract
Recent genomic and biological studies of neuroblastoma have shed light on the dramatic heterogeneity in the clinical behaviour of this disease, which spans from spontaneous regression or differentiation in some patients, to relentless disease progression in others, despite intensive multimodality therapy. This evidence also suggests several possible mechanisms to explain the phenomena of spontaneous regression in neuroblastomas, including neurotrophin deprivation, humoral or cellular immunity, loss of telomerase activity and alterations in epigenetic regulation. A better understanding of the mechanisms of spontaneous regression might help to identify optimal therapeutic approaches for patients with these tumours. Currently, the most druggable mechanism is the delayed activation of developmentally programmed cell death regulated by the tropomyosin receptor kinase A pathway. Indeed, targeted therapy aimed at inhibiting neurotrophin receptors might be used in lieu of conventional chemotherapy or radiation in infants with biologically favourable tumours that require treatment. Alternative approaches consist of breaking immune tolerance to tumour antigens or activating neurotrophin receptor pathways to induce neuronal differentiation. These approaches are likely to be most effective against biologically favourable tumours, but they might also provide insights into treatment of biologically unfavourable tumours. We describe the different mechanisms of spontaneous neuroblastoma regression and the consequent therapeutic approaches.
Collapse
Affiliation(s)
- Garrett M Brodeur
- Division of Oncology, The Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, Philadelphia, PA 19104-4302, USA
| | - Rochelle Bagatell
- Division of Oncology, The Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, Philadelphia, PA 19104-4302, USA
| |
Collapse
|
22
|
Taïeb D, Kaliski A, Boedeker CC, Martucci V, Fojo T, Adler JR, Pacak K. Current approaches and recent developments in the management of head and neck paragangliomas. Endocr Rev 2014; 35:795-819. [PMID: 25033281 PMCID: PMC4167435 DOI: 10.1210/er.2014-1026] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Head and neck paragangliomas (HNPGLs) are rare neuroendocrine tumors belonging to the family of pheochromocytoma/paraganglioma neoplasms. Despite advances in understanding the pathogenesis of these tumors, the growth potential and clinical outcome of individual cases remains largely unpredictable. Over several decades, surgical resection has long been the treatment of choice for HNPGLs. However, increasing experience in various forms of radiosurgery has been reported to result in curative-like outcomes, even for tumors localized in the most inaccessible anatomical areas. The emergence of such new therapies challenges the traditional paradigm for the management of HNPGLs. This review will assist and guide physicians who encounter patients with such tumors, either from a diagnostic or therapeutic standpoint. This review will also particularly emphasize current and emerging knowledge in genetics, imaging, and therapeutic options as well as the health-related quality of life for patients with HNPGLs.
Collapse
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine (D.T.), La Timone University Hospital, CERIMED, Aix-Marseille Univ, F-13385 Marseille, France; Department of Radiation Oncology (A.K.), Besançon University Hospital, F-25030 Besançon, France; Department of Otorhinolaryngology/Head and Neck Surgery (C.C.B.), HELIOS Hanseklinikum Stralsund, D-18435 Stralsund, Germany; Department of Otorhinolaryngology/Head and Neck Surgery (C.C.B.), University Hospital, Freiburg, Germany; Program in Reproductive and Adult Endocrinology (V.M., K.P.), Eunice Kennedy Shriver National Institute of Child Health and Human Development and Medical Oncology Branch (T.F.), National Institutes of Health, Bethesda, Maryland 20892; Department of Neurosurgery (J.R.A.), Stanford Hospital and Clinics, Stanford University, Stanford, California 94305
| | | | | | | | | | | | | |
Collapse
|
23
|
Masjkur J, Levenfus I, Lange S, Arps-Forker C, Poser S, Qin N, Vukicevic V, Chavakis T, Eisenhofer G, Bornstein SR, Ehrhart-Bornstein M, Androutsellis-Theotokis A. A defined, controlled culture system for primary bovine chromaffin progenitors reveals novel biomarkers and modulators. Stem Cells Transl Med 2014; 3:801-8. [PMID: 24855275 DOI: 10.5966/sctm.2013-0211] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
We present a method to efficiently culture primary chromaffin progenitors from the adult bovine adrenal medulla in a defined, serum-free monolayer system. Tissue is dissociated and plated for expansion under support by the mitogen basic fibroblast growth factor (bFGF). The cultures, although not homogenous, contain a subpopulation of cells expressing the neural stem cell marker Hes3 that also propagate. In addition, Hes3 is also expressed in the adult adrenal medulla from where the tissue is taken. Differentiation is induced by bFGF withdrawal and switching to Neurobasal medium containing B27. Following differentiation, Hes3 expression is lost, and cells acquire morphologies and biomarker expression patterns of chromaffin cells and dopaminergic neurons. We tested the effect of different treatments that we previously showed regulate Hes3 expression and cell number in cultures of fetal and adult rodent neural stem cells. Treatment of the cultures with a combination of Delta4, Angiopoietin2, and a Janus kinase inhibitor increases cell number during the expansion phase without significantly affecting catecholamine content levels. Treatment with cholera toxin does not significantly affect cell number but reduces the ratio of epinephrine to norepinephrine content and increases the dopamine content relative to total catecholamines. These data suggest that this defined culture system can be used for target identification in drug discovery programs and that the transcription factor Hes3 may serve as a new biomarker of putative adrenomedullary chromaffin progenitor cells.
Collapse
Affiliation(s)
- Jimmy Masjkur
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ian Levenfus
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Sven Lange
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Carina Arps-Forker
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Steve Poser
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Nan Qin
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Vladimir Vukicevic
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Graeme Eisenhofer
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Monika Ehrhart-Bornstein
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Andreas Androutsellis-Theotokis
- Department of Internal Medicine III and Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav Carus, University of Dresden, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
24
|
Stubbusch J, Narasimhan P, Huber K, Unsicker K, Rohrer H, Ernsberger U. Synaptic protein and pan-neuronal gene expression and their regulation by Dicer-dependent mechanisms differ between neurons and neuroendocrine cells. Neural Dev 2013; 8:16. [PMID: 23961995 PMCID: PMC3766641 DOI: 10.1186/1749-8104-8-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/19/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Neurons in sympathetic ganglia and neuroendocrine cells in the adrenal medulla share not only their embryonic origin from sympathoadrenal precursors in the neural crest but also a range of functional features. These include the capacity for noradrenaline biosynthesis, vesicular storage and regulated release. Yet the regulation of neuronal properties in early neuroendocrine differentiation is a matter of debate and the developmental expression of the vesicle fusion machinery, which includes components found in both neurons and neuroendocrine cells, is not resolved. RESULTS Analysis of synaptic protein and pan-neuronal marker mRNA expression during mouse development uncovers profound differences between sympathetic neurons and adrenal chromaffin cells, which result in qualitatively similar but quantitatively divergent transcript profiles. In sympathetic neurons embryonic upregulation of synaptic protein mRNA follows early and persistent induction of pan-neuronal marker transcripts. In adrenal chromaffin cells pan-neuronal marker expression occurs only transiently and synaptic protein messages remain at distinctly low levels throughout embryogenesis. Embryonic induction of synaptotagmin I (Syt1) in sympathetic ganglia and postnatal upregulation of synaptotagmin VII (Syt7) in adrenal medulla results in a cell type-specific difference in isoform prevalence. Dicer 1 inactivation in catecholaminergic cells reduces high neuronal synaptic protein mRNA levels but not their neuroendocrine low level expression. Pan-neuronal marker mRNAs are induced in chromaffin cells to yield a more neuron-like transcript pattern, while ultrastructure is not altered. CONCLUSIONS Our study demonstrates that remarkably different gene regulatory programs govern the expression of synaptic proteins in the neuronal and neuroendocrine branch of the sympathoadrenal system. They result in overlapping but quantitatively divergent transcript profiles. Dicer 1-dependent regulation is required to establish high neuronal mRNA levels for synaptic proteins and to maintain repression of neurofilament messages in neuroendocrine cells.
Collapse
Affiliation(s)
- Jutta Stubbusch
- Max Planck Institute for Brain Research, Deutschordenstrasse 46 D-60528, Frankfurt, Germany.
| | | | | | | | | | | |
Collapse
|
25
|
Shtukmaster S, Schier MC, Huber K, Krispin S, Kalcheim C, Unsicker K. Sympathetic neurons and chromaffin cells share a common progenitor in the neural crest in vivo. Neural Dev 2013; 8:12. [PMID: 23777568 PMCID: PMC3693940 DOI: 10.1186/1749-8104-8-12] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 05/17/2013] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The neural crest (NC) is a transient embryonic structure unique to vertebrates, which generates peripheral sensory and autonomic neurons, glia, neuroendocrine chromaffin and thyroid C-cells, melanocytes, and mesenchymal derivatives such as parts of the skull, heart, and meninges. The sympathoadrenal (SA) cell lineage is one major sub-lineage of the NC that gives rise to sympathetic neurons, chromaffin cells, and the intermediate small intensely fluorescent (SIF) cells. A key question is when during NC ontogeny do multipotent progenitors segregate into the different NC-derived lineages. Recent evidence suggested that sympathetic, sensory, and melanocyte progenitors delaminate from the thoracic neural tube (NT) in successive, largely non-overlapping waves and that at least certain NC progenitors are already fate-restricted within the NT. Whether sympathetic neurons and chromaffin cells, suggested by cell culture studies to share a common progenitor, are also fate segregated in ovo prior to emigration, is not known. RESULTS We have conducted single cell electroporations of a GFP-encoding plasmid into the dorsal midline of E2 chick NTs at the adrenomedullary level of the NC. Analysis of their derivatives, performed at E6, revealed that in most cases, labelled progeny was detected in both sympathetic ganglia and adrenal glands, where cells co-expressed characteristic marker combinations. CONCLUSIONS Our results show that sympathetic neurons and adrenal chromaffin cells share a common progenitor in the NT. Together with previous findings we suggest that phenotypic diversification of these sublineages is likely to occur after delamination from the NT and prior to target encounter.
Collapse
Affiliation(s)
- Stella Shtukmaster
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology University of Freiburg, Albertstr, 17, Freiburg D-79104, Germany
| | | | | | | | | | | |
Collapse
|