1
|
Tagami A, Ikeda Y, Ishizuka K, Maekawa M. Conditional disruption of Nr5a1 directed by Sox9-Cre impairs adrenal development. Sci Rep 2024; 14:12297. [PMID: 38811798 PMCID: PMC11137078 DOI: 10.1038/s41598-024-63264-9] [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: 04/01/2024] [Accepted: 05/27/2024] [Indexed: 05/31/2024] Open
Abstract
The current study aimed to investigate the effect of Sox9-Cre-directed Nr5a1-conditional knockout (Sox9-Cre;Nr5a1flox/flox) on adrenal development. We showed that SOX9 is expressed by adrenocortical cells at E10.5-E11.5 but is extinguished no later than E12.5. The number of adrenocortical cells significantly reduced in Sox9-Cre;Nr5a1flox/flox mice while the number of cleaved caspase 3-positive cells increased compared to that in the controls at E11.5-E12.5, when the adrenal primordium (AP) is about to expand. This indicated that fetal adrenocortical cells are lost via apoptosis due to Nr5a1 ablation by E12.5. Both medulla formation and encapsulation were perturbed, accompanied by a smaller AP size, in Sox9-Cre;Nr5a1flox/flox mice during embryonic development. Adult Sox9-Cre;Nr5a1flox/flox adrenals were hypoplastic and exhibited irregular organization of the medulla with aberrant sex differentiation in the X zone. Additionally, there were histologically eosin-negative vacuolated cells, which were negative for both the X-zone marker 20αHSD and the steroidogenesis marker 3βHSD at the innermost cortex of Sox9-Cre;Nr5a1flox/flox adrenals. Although Nr5a1+/- adrenals were hypoplastic, a small number of chromaffin cells were properly located in the center, having normal sex differences in the X-zone. The results collectively provided in-vivo evidence that Nr5a1 plays a critical role in AP expansion and subsequent adrenal development.
Collapse
Affiliation(s)
- Ayako Tagami
- Department of Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi, 464-8650, Japan
| | - Yayoi Ikeda
- Department of Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi, 464-8650, Japan.
| | - Kyoko Ishizuka
- Department of Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi, 464-8650, Japan
| | - Mamiko Maekawa
- Department of Anatomy, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, Aichi, 464-8650, Japan
| |
Collapse
|
2
|
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
|
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
|
Abdel-Maksoud FM, Fadl S, Abou-Elmagd A, Saleh AMM. Post-hatching developmental changes in the adrenal gland of the Japanese quail (Coturnix coturnix japonica): Histological, immunohistochemical, and electron microscopic studies. Microsc Res Tech 2023; 86:1461-1474. [PMID: 37204121 DOI: 10.1002/jemt.24348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/21/2023] [Accepted: 05/06/2023] [Indexed: 05/20/2023]
Abstract
The adrenal glands are paired abdominal endocrine organs vital to the bird's health. The present research aimed to provide a comprehensive examination of the histological, ultrastructural, and immunohistochemical investigations of the adrenal gland in Japanese quail during the post hatching period. The current study was performed on 21 healthy Japanese quail chicks at different post hatching periods. Our results showed the adrenal gland is surrounded by a connective tissue capsule, which consists of dense collagen fibers containing large blood vessels, chromaffin cells, autonomic ganglia, fibroblasts, and migrating Schwann cells. The zonation of the adrenal gland is composed of a subcapsular layer, a peripheral zone, and a central zone, which gets more pronounced with age. At the ultrastructural level, the interrenal cells take the steroid-secreting cells characters; they have varying amounts of lipid droplets and abundant mitochondria. Adrenal medullary chromaffin cells showed positive immunoreactivity to the NSE. With increasing age, the chromaffin tissue's Sox10 immunoreactivity increased. β-catenin is expressed within the plasmalemma and the cytoplasm of the interrenal and chromaffin cells and its reactivity increased with age, especially in the chromaffin cells. Our results indicate the adrenal gland undergoes significant morphological changes during the postnatal life. Overall, the postnatal period is an important time for the development and maturation of the adrenal glands.
Collapse
Affiliation(s)
- Fatma M Abdel-Maksoud
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Saher Fadl
- 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
| | - Abdelmohaimen M M Saleh
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
5
|
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
|
6
|
Iwahashi N, Umakoshi H, Seki T, Gomez-Sanchez CE, Mukai K, Suematsu M, Umezawa Y, Oya M, Kosaka T, Seki M, Suzuki Y, Horiuchi Y, Ogawa Y, Nishimoto K. Characterization of Aldosterone-producing Cell Cluster (APCC) at Single-cell Resolution. J Clin Endocrinol Metab 2022; 107:2439-2448. [PMID: 35796577 PMCID: PMC9387688 DOI: 10.1210/clinem/dgac394] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT The adrenal cortex consists of zona glomerulosa (ZG), fasciculata (ZF), and reticularis. Aldosterone-producing cell clusters (APCCs) that strongly express aldosterone synthase (CYP11B2) are frequently found in adult adrenals and harbor somatic mutations that are also detected in aldosterone-producing adenomas (APAs). Primary aldosteronism is mainly caused by APAs or idiopathic hyperaldosteronism (IHA). We presume that APCCs are causing IHA and are precursors of APAs. However, the gene expression characteristics and especially the development of APCCs are not well understood. OBJECTIVE This study aimed to analyze the transcriptome of APCCs at single-cell resolution and infer the developmental trajectory. METHODS Single-cell RNA sequencing (scRNA-seq) of 2 adult adrenals was performed. RESULTS Immunohistochemical analyses confirmed the 2 adrenals had APCCs. scRNA-seq data of 2928 adrenal cells were obtained and 1765 adrenocortical cells were identified based on unsupervised clustering and the marker gene expression. The adrenocortical cells were divided into 6 clusters, of which 3 clusters (923 cells) were composed of APCC/ZG cells. By further subclustering, the APCC/ZG cells were divided into 3 clusters (clusters C1, C2, and C3), we finally identified APCC cluster (C3) and ZG cluster (C1). Cluster C2 seemed to be ZG-to-ZF transitional cells. RNA velocity analysis inferred the developmental direction from cluster ZG-cluster-C1 to APCC-cluster-C3. The scRNA-seq additionally revealed that many CYP11B2-positive cells were positive for CYP11B1 and/or CYP17A1, which were essential for cortisol but not for aldosterone production. CONCLUSIONS Our results revealed the gene expression characteristics of APCC at single-cell resolution and show that some ZG cells remodel to APCC.
Collapse
Affiliation(s)
| | | | - Tsugio Seki
- Department of Medical Education, School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA
| | - Celso E Gomez-Sanchez
- Department of Pharmacology and Toxicology and Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Kuniaki Mukai
- Medical Education Center, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yuta Umezawa
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of MedicineTokyo 160-8582, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of MedicineTokyo 160-8582, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-0882, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-0882, Japan
| | - Yutaka Horiuchi
- Department of Microbiology, Faculty of Medicine, Saitama Medical University, Saitama 350-0495, Japan
| | - Yoshihiro Ogawa
- Correspondence: Yoshihiro Ogawa, MD, PhD, Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Koshiro Nishimoto
- Correspondence: Koshiro Nishimoto, MD, PhD, Department of Uro-Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama 350-1298, Japan.
| |
Collapse
|
7
|
Moon S, Zhao YT. Convergent biological pathways underlying the Kallmann syndrome-linked genes Hs6st1 and Fgfr1. Hum Mol Genet 2022; 31:4207-4216. [PMID: 35899427 PMCID: PMC9759331 DOI: 10.1093/hmg/ddac172] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/05/2022] [Accepted: 07/24/2022] [Indexed: 01/21/2023] Open
Abstract
Kallmann syndrome (KS) is a congenital disorder characterized by idiopathic hypogonadotropic hypogonadism and olfactory dysfunction. KS is linked to variants in >34 genes, which are scattered across the human genome and show disparate biological functions. Although the genetic basis of KS is well studied, the mechanisms by which disruptions of these diverse genes cause the same outcome of KS are not fully understood. Here we show that disruptions of KS-linked genes affect the same biological processes, indicating convergent molecular mechanisms underlying KS. We carried out machine learning-based predictions and found that KS-linked mutations in heparan sulfate 6-O-sulfotransferase 1 (HS6ST1) are likely loss-of-function mutations. We next disrupted Hs6st1 and another KS-linked gene, fibroblast growth factor receptor 1 (Fgfr1), in mouse neuronal cells and measured transcriptome changes using RNA sequencing. We found that disruptions of Hs6st1 and Fgfr1 altered genes in the same biological processes, including the upregulation of genes in extracellular pathways and the downregulation of genes in chromatin pathways. Moreover, we performed genomics and bioinformatics analyses and found that Hs6st1 and Fgfr1 regulate gene transcription likely via the transcription factor Sox9/Sox10 and the chromatin regulator Chd7, which are also associated with KS. Together, our results demonstrate how different KS-linked genes work coordinately in a convergent signaling pathway to regulate the same biological processes, thus providing new insights into KS.
Collapse
Affiliation(s)
- Sohyun Moon
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, USA
| | - Ying-Tao Zhao
- To whom correspondence should be addressed: Tel: 516-686-3764; Fax: 516-686-3832;
| |
Collapse
|
8
|
Deal KK, Rosebrock JC, Eeds AM, DeKeyser JML, Musser MA, Ireland SJ, May-Zhang AA, Buehler DP, Southard-Smith EM. Sox10-cre BAC transgenes reveal temporal restriction of mesenchymal cranial neural crest and identify glandular Sox10 expression. Dev Biol 2020; 471:119-137. [PMID: 33316258 DOI: 10.1016/j.ydbio.2020.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/29/2022]
Abstract
Diversity of neural crest derivatives has been studied with a variety of approaches during embryonic development. In mammals Cre-LoxP lineage tracing is a robust means to fate map neural crest relying on cre driven from regulatory elements of early neural crest genes. Sox10 is an essential transcription factor for normal neural crest development. A variety of efforts have been made to label neural crest derivatives using partial Sox10 regulatory elements to drive cre expression. To date published Sox10-cre lines have focused primarily on lineage tracing in specific tissues or during early fetal development. We describe two new Sox10-cre BAC transgenes, constitutive (cre) and inducible (cre/ERT2), that contain the complete repertoire of Sox10 regulatory elements. We present a thorough expression profile of each, identifying a few novel sites of Sox10 expression not captured by other neural crest cre drivers. Comparative mapping of expression patterns between the Sox10-cre and Sox10-cre/ERT2 transgenes identified a narrow temporal window in which Sox10 expression is present in mesenchymal derivatives prior to becoming restricted to neural elements during embryogenesis. In more caudal structures, such as the intestine and lower urinary tract, our Sox10-cre BAC transgene appears to be more efficient in labeling neural crest-derived cell types than Wnt1-cre. The analysis reveals consistent expression of Sox10 in non-neural crest derived glandular epithelium, including salivary, mammary, and urethral glands of adult mice. These Sox10-cre and Sox10-cre/ERT2 transgenic lines are verified tools that will enable refined temporal and cell-type specific lineage analysis of neural crest derivatives as well as glandular tissues that rely on Sox10 for proper development and function.
Collapse
Affiliation(s)
- Karen K Deal
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jennifer C Rosebrock
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Angela M Eeds
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jean-Marc L DeKeyser
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Present address: Northwestern University, Dept. of Pharmacology, USA
| | - Melissa A Musser
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Present address: Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Sara J Ireland
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Aaron A May-Zhang
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Dennis P Buehler
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - E Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
9
|
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: 22] [Impact Index Per Article: 4.4] [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
|
10
|
The transcription factor Sox10 is an essential determinant of branching morphogenesis and involution in the mouse mammary gland. Sci Rep 2020; 10:17807. [PMID: 33082503 PMCID: PMC7575560 DOI: 10.1038/s41598-020-74664-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022] Open
Abstract
The high mobility group-domain containing transcription factor Sox10 is an essential regulator of developmental processes and homeostasis in the neural crest, several neural crest-derived lineages and myelinating glia. Recent studies have also implicated Sox10 as an important factor in mammary stem and precursor cells. Here we employ a series of mouse mutants with constitutive and conditional Sox10 deficiencies to show that Sox10 has multiple functions in the developing mammary gland. While there is no indication for a requirement of Sox10 in the specification of the mammary placode or descending mammary bud, it is essential for both the prenatal hormone-independent as well as the pubertal hormone-dependent branching of the mammary epithelium and for proper alveologenesis during pregnancy. It furthermore acts in a dosage-dependent manner. Sox10 also plays a role during the involution process at the end of the lactation period. Whereas its effect on epithelial branching and alveologenesis are likely causally related to its function in mammary stem and precursor cells, this is not the case for its function during involution where Sox10 seems to work at least in part through regulation of the miR-424(322)/503 cluster.
Collapse
|
11
|
Yang CL, Serra-Roma A, Gualandi M, Bodmer N, Niggli F, Schulte JH, Bode PK, Shakhova O. Lineage-restricted sympathoadrenal progenitors confer neuroblastoma origin and its tumorigenicity. Oncotarget 2020; 11:2357-2371. [PMID: 32595833 PMCID: PMC7299536 DOI: 10.18632/oncotarget.27636] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/20/2020] [Indexed: 01/18/2023] Open
Abstract
Neuroblastoma (NB) is the most common cancer in infants and it accounts for six percent of all pediatric malignancies. There are several hypotheses proposed on the origins of NB. While there is little genetic evidence to support this, the prevailing model is that NB originates from neural crest stem cells (NCSCs). Utilizing in vivo mouse models, we demonstrate that targeting MYCN oncogene to NCSCs causes perinatal lethality. During sympathoadrenal (SA) lineage development, SOX transcriptional factors drive the transition from NCSCs to lineage-specific progenitors, characterized by the sequential activation of Sox9/Sox10/Sox4/Sox11 genes. We find the NCSCs factor SOX10 is not expressed in neuroblasts, but rather restricted to the Schwannian stroma and is associated with a good prognosis. On the other hand, SOX9 expression in NB cells was associated with several key biological processes including migration, invasion and differentiation. Moreover, manipulating SOX9 gene predominantly affects lineage-restricted SA progenitors. Our findings highlight a unique molecular SOX signature associated with NB that is highly reminiscent of SA progenitor transcriptional program during embryonic development, providing novel insights into NB pathobiology. In summary, we provide multiple lines of evidence suggesting that multipotent NCSCs do not contribute to NB initiation and maintenance.
Collapse
Affiliation(s)
- Chia-Lung Yang
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - André Serra-Roma
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - Marco Gualandi
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - Nicole Bodmer
- Department of Oncology, Children Hospital of Zürich, Zürich, Switzerland
| | - Felix Niggli
- Department of Oncology, Children Hospital of Zürich, Zürich, Switzerland
| | | | - Peter Karl Bode
- Department of Surgical Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Olga Shakhova
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
12
|
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: 225] [Impact Index Per Article: 32.1] [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
|
13
|
Qiu W, Guo F, Glass K, Yuan GC, Quackenbush J, Zhou X, Tantisira KG. Differential connectivity of gene regulatory networks distinguishes corticosteroid response in asthma. J Allergy Clin Immunol 2017; 141:1250-1258. [PMID: 28736268 DOI: 10.1016/j.jaci.2017.05.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/02/2017] [Accepted: 05/03/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Variations in drug response between individuals have prevented us from achieving high drug efficacy in treating many complex diseases, including asthma. Genetics plays an important role in accounting for such interindividual variations in drug response. However, systematic approaches for addressing how genetic factors and their regulators determine variations in drug response in asthma treatment are lacking. OBJECTIVE We sought to identify key transcriptional regulators of corticosteroid response in asthma using a novel systems biology approach. METHODS We used Passing Attributes between Networks for Data Assimilations (PANDA) to construct the gene regulatory networks associated with good responders and poor responders to inhaled corticosteroids based on a subset of 145 white children with asthma who participated in the Childhood Asthma Management Cohort. PANDA uses gene expression profiles and published relationships among genes, transcription factors (TFs), and proteins to construct the directed networks of TFs and genes. We assessed the differential connectivity between the gene regulatory network of good responders versus that of poor responders. RESULTS When compared with poor responders, the network of good responders has differential connectivity and distinct ontologies (eg, proapoptosis enriched in network of good responders and antiapoptosis enriched in network of poor responders). Many of the key hubs identified in conjunction with clinical response are also cellular response hubs. Functional validation demonstrated abrogation of differences in corticosteroid-treated cell viability following siRNA knockdown of 2 TFs and differential downstream expression between good responders and poor responders. CONCLUSIONS We have identified and validated multiple TFs influencing asthma treatment response. Our results show that differential connectivity analysis can provide new insights into the heterogeneity of drug treatment effects.
Collapse
Affiliation(s)
- Weiliang Qiu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Feng Guo
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Guo Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - John Quackenbush
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Mass; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Kelan G Tantisira
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass.
| |
Collapse
|
14
|
Stolt CC, Wegner M. Schwann cells and their transcriptional network: Evolution of key regulators of peripheral myelination. Brain Res 2015; 1641:101-110. [PMID: 26423937 DOI: 10.1016/j.brainres.2015.09.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 09/18/2015] [Accepted: 09/20/2015] [Indexed: 11/29/2022]
Abstract
As derivatives of the neural crest, Schwann cells represent a vertebrate invention. Their development and differentiation is under control of a newly constructed, vertebrate-specific regulatory network that contains Sox10, Oct6 and Krox20 as cornerstones and central regulators of peripheral myelination. In this review, we discuss the function and relationship of these transcription factors among each other and in the context of their regulatory network, and present ideas of how neofunctionalization may have helped to recruit them to their novel task in Schwann cells. This article is part of a Special Issue entitled SI: Myelin Evolution.
Collapse
Affiliation(s)
- C Claus Stolt
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| |
Collapse
|
15
|
Vukicevic V, Rubin de Celis MF, Pellegata NS, Bornstein SR, Androutsellis-Theotokis A, Ehrhart-Bornstein M. Adrenomedullary progenitor cells: Isolation and characterization of a multi-potent progenitor cell population. Mol Cell Endocrinol 2015; 408:178-84. [PMID: 25575455 DOI: 10.1016/j.mce.2014.12.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/25/2014] [Accepted: 12/27/2014] [Indexed: 12/19/2022]
Abstract
The adrenal is a highly plastic organ with the ability to adjust to physiological needs by adapting hormone production but also by generating and regenerating both adrenocortical and adrenomedullary tissue. It is now apparent that many adult tissues maintain stem and progenitor cells that contribute to their maintenance and adaptation. Research from the last years has proven the existence of stem and progenitor cells also in the adult adrenal medulla throughout life. These cells maintain some neural crest properties and have the potential to differentiate to the endocrine and neural lineages. In this article, we discuss the evidence for the existence of adrenomedullary multi potent progenitor cells, their isolation and characterization, their differentiation potential as well as their clinical potential in transplantation therapies but also in pathophysiology.
Collapse
Affiliation(s)
- Vladimir Vukicevic
- Division of Molecular Endocrinology, Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden, 01307 Dresden, Germany
| | - Maria Fernandez Rubin de Celis
- Division of Molecular Endocrinology, Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden, 01307 Dresden, Germany
| | - Natalia S Pellegata
- Institute of Pathology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Stefan R Bornstein
- Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden, 01307 Dresden, Germany; Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Andreas Androutsellis-Theotokis
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany; Division of Stem Cell Biology, Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden, 01307 Dresden, Germany
| | - Monika Ehrhart-Bornstein
- Division of Molecular Endocrinology, Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden, 01307 Dresden, Germany; Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany.
| |
Collapse
|
16
|
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
|
17
|
Rubin de Celis MF, Garcia-Martin R, Wittig D, Valencia GD, Enikolopov G, Funk RH, Chavakis T, Bornstein SR, Androutsellis-Theotokis A, Ehrhart-Bornstein M. Multipotent Glia-Like Stem Cells Mediate Stress Adaptation. Stem Cells 2015; 33:2037-51. [DOI: 10.1002/stem.2002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 01/30/2015] [Accepted: 02/06/2015] [Indexed: 01/10/2023]
Affiliation(s)
- Maria F. Rubin de Celis
- Division of Molecular Endocrinology; Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden; Dresden Germany
| | - Ruben Garcia-Martin
- Department of Clinical Pathobiochemistry and Institute of Clinical Chemistry and Laboratory Medicine; Carl Gustav Carus University Clinic, Technische Universität Dresden; Dresden Germany
| | - Dierk Wittig
- Institute of Anatomy; Technische Universität Dresden; Dresden Germany
| | - Gabriela D. Valencia
- Division of Molecular Endocrinology; Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden; Dresden Germany
| | | | - Richard H. Funk
- Institute of Anatomy; Technische Universität Dresden; Dresden Germany
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry and Institute of Clinical Chemistry and Laboratory Medicine; Carl Gustav Carus University Clinic, Technische Universität Dresden; Dresden Germany
- Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden; Dresden Germany
- Center for Regenerative Therapies Dresden, Technische Universität Dresden; Dresden Germany
| | - Stefan R. Bornstein
- Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden; Dresden Germany
- Center for Regenerative Therapies Dresden, Technische Universität Dresden; Dresden Germany
| | - Andreas Androutsellis-Theotokis
- Division of Stem Cell Biology; Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden; Dresden Germany
| | - Monika Ehrhart-Bornstein
- Division of Molecular Endocrinology; Medical Clinic III, Carl Gustav Carus University Clinic, Technische Universität Dresden; Dresden Germany
- Center for Regenerative Therapies Dresden, Technische Universität Dresden; Dresden Germany
| |
Collapse
|
18
|
Shakhova O, Cheng P, Mishra PJ, Zingg D, Schaefer SM, Debbache J, Häusel J, Matter C, Guo T, Davis S, Meltzer P, Mihic-Probst D, Moch H, Wegner M, Merlino G, Levesque MP, Dummer R, Santoro R, Cinelli P, Sommer L. Antagonistic cross-regulation between Sox9 and Sox10 controls an anti-tumorigenic program in melanoma. PLoS Genet 2015; 11:e1004877. [PMID: 25629959 PMCID: PMC4309598 DOI: 10.1371/journal.pgen.1004877] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/04/2014] [Indexed: 12/20/2022] Open
Abstract
Melanoma is the most fatal skin cancer, but the etiology of this devastating disease is still poorly understood. Recently, the transcription factor Sox10 has been shown to promote both melanoma initiation and progression. Reducing SOX10 expression levels in human melanoma cells and in a genetic melanoma mouse model, efficiently abolishes tumorigenesis by inducing cell cycle exit and apoptosis. Here, we show that this anti-tumorigenic effect functionally involves SOX9, a factor related to SOX10 and upregulated in melanoma cells upon loss of SOX10. Unlike SOX10, SOX9 is not required for normal melanocyte stem cell function, the formation of hyperplastic lesions, and melanoma initiation. To the contrary, SOX9 overexpression results in cell cycle arrest, apoptosis, and a gene expression profile shared by melanoma cells with reduced SOX10 expression. Moreover, SOX9 binds to the SOX10 promoter and induces downregulation of SOX10 expression, revealing a feedback loop reinforcing the SOX10 low/SOX9 high ant,m/ii-tumorigenic program. Finally, SOX9 is required in vitro and in vivo for the anti-tumorigenic effect achieved by reducing SOX10 expression. Thus, SOX10 and SOX9 are functionally antagonistic regulators of melanoma development. For the development of future cancer therapies it is imperative to understand the molecular processes underlying tumor initiation and expansion. Many key factors involved in these processes have been identified based on cell culture and transplantation experiments, but their relevance for tumor formation and disease progression in the living organism is often unclear. Therefore, genetically modified mice spontaneously developing tumors present indispensable models for cancer research. Here, we address this issue by studying the formation of melanoma, the most fatal skin tumor in industrialized countries. To this end, we use a transgenic mouse model to elucidate cellular and molecular mechanisms regulating congenital nevus and melanoma initiation. We show that a transcription factor called SOX10 promotes melanoma formation by repressing an anti-tumorigenic program involving the activity of a related factor, SOX9. When SOX10 is inactivated, SOX9 becomes upregulated and induces cell cycle arrest and death in melanoma cells. Furthermore, upon experimental elevation of SOX9 levels, SOX10 activity is suppressed, revealing an antagonistic relationship between SOX9 and SOX10 in melanoma initiation. Knowledge of how an anti-tumorigenic program can be stimulated by modulating the activities of these key factors might help to design novel therapeutic strategies.
Collapse
Affiliation(s)
- Olga Shakhova
- Cell and Developmental Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Phil Cheng
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Pravin J. Mishra
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Daniel Zingg
- Cell and Developmental Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Simon M. Schaefer
- Cell and Developmental Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Julien Debbache
- Cell and Developmental Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Jessica Häusel
- Cell and Developmental Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Claudia Matter
- Department of Oncology, University Hospital Zurich, Schlieren, Switzerland
| | - Theresa Guo
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Sean Davis
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Paul Meltzer
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Daniela Mihic-Probst
- Department of Pathology, Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology, Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Wegner
- Institute of Biochemistry, Emil Fischer Center, FAU University of Erlangen-Nuernberg, Erlangen, Germany
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | | | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Raffaella Santoro
- Institute of Veterinary Biochemistry and Molecular Biology, University of Zurich, Zurich, Switzerland
| | - Paolo Cinelli
- Division of Trauma Surgery, Center for Clinical Research, University Hospital Zurich, Zurich, Switzerland
| | - Lukas Sommer
- Cell and Developmental Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
- * E-mail:
| |
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
|
Can the ‘neuron theory’ be complemented by a universal mechanism for generic neuronal differentiation. Cell Tissue Res 2014; 359:343-84. [DOI: 10.1007/s00441-014-2049-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/23/2014] [Indexed: 12/19/2022]
|
21
|
Segregation of neuronal and neuroendocrine differentiation in the sympathoadrenal lineage. Cell Tissue Res 2014; 359:333-41. [PMID: 25038743 DOI: 10.1007/s00441-014-1947-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 06/06/2014] [Indexed: 10/25/2022]
Abstract
Neuronal and neuroendocrine cells possess the capacity for Ca(2+)-regulated discharge of messenger molecules, which they release into synapses or the blood stream, respectively. The neural-crest-derived sympathoadrenal lineage gives rise to the sympathetic neurons of the autonomic nervous system and the neuroendocrine chromaffin cells of the adrenal medulla. These cells provide an excellent model system for studying common and distinct developmental mechanisms underlying the acquisition of neuroendocrine and neuronal properties. As catecholaminergic cells, they possess common markers related to noradrenaline synthesis, storage and release, but they also display diverging gene expression patterns and are morphologically and functionally different. The precise mechanisms that underlie the diversification of sympathoadrenal cells into neurons and neuroendocrine cells are not fully understood. However, in the past we could show that the establishment of a chromaffin phenotype does not depend on signals from the adrenal cortex and that chromaffin cells and sympathetic neurons apparently differ from the onset of their catecholaminergic differentiation. Nevertheless, the cues that specifically induce neuroendocrine features remain elusive. The early development of the progenitors of chromaffin cells and sympathetic neurons depends on a common set of transcription factors with overlapping but distinct influences on their development. In addition to the well-defined role of transcription factors as developmental regulators, our understanding of post-transcriptional gene regulation by microRNAs has substantially increased within the last few decades. This review highlights the major similarities and differences between chromaffin cells and sympathetic neurons, summarizes our current knowledge of the roles of selected transcription factors, microRNAs and environmental signals for the neuroendocrine differentiation of sympathoadrenal cells, and draws comparisons with the development of other endocrine and neuronal cells.
Collapse
|
22
|
Weider M, Reiprich S, Wegner M. Sox appeal - Sox10 attracts epigenetic and transcriptional regulators in myelinating glia. Biol Chem 2014; 394:1583-93. [PMID: 23729567 DOI: 10.1515/hsz-2013-0146] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 05/15/2013] [Indexed: 01/09/2023]
Abstract
Sox10 belongs to the Sox family of high-mobility group-box transcription factors. It fulfils widespread and essential functions in myelinating glia at multiple stages of development such as glial specification, survival and terminal differentiation. To a large extent, these diverse activities can be attributed to its capacity to interact with different transcription factors in distinct regulatory networks. Beyond transcription factors, an increasing number of interaction partners are emerging with alternative impact on gene expression. These include components of the mediator complex, the Brahma-associated factor complex and histone deacetylases. Here, we discuss interactions with functional relevance in myelinating glia and link Sox10 function in these cells not only to gene transcription, but also to epigenetics and chromatin remodeling.
Collapse
|
23
|
Targeted deletion of Sox10 by Wnt1-cre defects neuronal migration and projection in the mouse inner ear. PLoS One 2014; 9:e94580. [PMID: 24718611 PMCID: PMC3981815 DOI: 10.1371/journal.pone.0094580] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/18/2014] [Indexed: 12/31/2022] Open
Abstract
Sensory nerves of the brainstem are mostly composed of placode-derived neurons, neural crest-derived neurons and neural crest-derived Schwann cells. This mixed origin of cells has made it difficult to dissect interdependence for fiber guidance. Inner ear-derived neurons are known to connect to the brain after delayed loss of Schwann cells in ErbB2 mutants. However, the ErbB2 mutant related alterations in the ear and the brain compound interpretation of the data. We present here a new model to evaluate exclusively the effect of Schwann cell loss on inner ear innervation. Conditional deletion of the neural crest specific transcription factor, Sox10, using the rhombic lip/neural crest specific Wnt1-cre driver spares Sox10 expression in the ear. We confirm that neural crest-derived cells provide a stop signal for migrating spiral ganglion neurons. In the absence of Schwann cells, spiral ganglion neurons migrate into the center of the cochlea and even out of the ear toward the brain. Spiral ganglion neuron afferent processes reach the organ of Corti, but many afferent fibers bypass the organ of Corti to enter the lateral wall of the cochlea. In contrast to this peripheral disorganization, the central projection to cochlear nuclei is normal. Compared to ErbB2 mutants, conditional Sox10 mutants have limited cell death in spiral ganglion neurons, indicating that the absence of Schwann cells alone contributes little to the embryonic survival of neurons. These data suggest that neural crest-derived cells are dispensable for all central and some peripheral targeting of inner ear neurons. However, Schwann cells provide a stop signal for migratory spiral ganglion neurons and facilitate proper targeting of the organ of Corti by spiral ganglion afferents.
Collapse
|
24
|
Guasti L, Cavlan D, Cogger K, Banu Z, Shakur A, Latif S, King PJ. Dlk1 up-regulates Gli1 expression in male rat adrenal capsule cells through the activation of β1 integrin and ERK1/2. Endocrinology 2013; 154:4675-84. [PMID: 24064361 DOI: 10.1210/en.2013-1211] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development and maintenance of the zones of the adrenal cortex and their steroidal output are extremely important in the control of gluconeogenesis, the stress response, and blood volume. Sonic Hedgehog (Shh) is expressed in the adrenal cortex and signals to capsular cells, which can respond by migrating into the cortex and converting into a steroidogenic phenotype. Delta-like homologue 1 (Dlk1), a member of the Notch/Delta/Serrate family of epidermal growth factor-like repeat-containing proteins, has a well-established role in inhibiting adipocyte differentiation. We demonstrate that Shh and Dlk1 are coexpressed in the outer undifferentiated zone of the male rat adrenal and that Dlk1 signals to the adrenal capsule, activating glioma-associated oncogene homolog 1 transcription in a β1 integrin- and Erk1/2-dependent fashion. Moreover, Shh and Dlk1 expression inversely correlates with the size of the zona glomerulosa in rats after manipulation of the renin-angiotensin system, suggesting a role in the homeostatic maintenance of the gland.
Collapse
Affiliation(s)
- Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
25
|
Van Otterloo E, Cornell RA, Medeiros DM, Garnett AT. Gene regulatory evolution and the origin of macroevolutionary novelties: insights from the neural crest. Genesis 2013; 51:457-70. [PMID: 23712931 DOI: 10.1002/dvg.22403] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 05/10/2013] [Accepted: 05/14/2013] [Indexed: 11/07/2022]
Abstract
The appearance of novel anatomic structures during evolution is driven by changes to the networks of transcription factors, signaling pathways, and downstream effector genes controlling development. The nature of the changes to these developmental gene regulatory networks (GRNs) is poorly understood. A striking test case is the evolution of the GRN controlling development of the neural crest (NC). NC cells emerge from the neural plate border (NPB) and contribute to multiple adult structures. While all chordates have a NPB, only in vertebrates do NPB cells express all the genes constituting the neural crest GRN (NC-GRN). Interestingly, invertebrate chordates express orthologs of NC-GRN components in other tissues, revealing that during vertebrate evolution new regulatory connections emerged between transcription factors primitively expressed in the NPB and genes primitively expressed in other tissues. Such interactions could have evolved by two mechanisms. First, transcription factors primitively expressed in the NPB may have evolved new DNA and/or cofactor binding properties (protein neofunctionalization). Alternately, cis-regulatory elements driving NPB expression may have evolved near genes primitively expressed in other tissues (cis-regulatory neofunctionalization). Here we discuss how gene duplication can, in principle, promote either form of neofunctionalization. We review recent published examples of interspecies gene-swap, or regulatory-element-swap, experiments that test both models. Such experiments have yielded little evidence to support the importance of protein neofunctionalization in the emergence of the NC-GRN, but do support the importance of novel cis-regulatory elements in this process. The NC-GRN is an excellent model for the study of gene regulatory and macroevolutionary innovation.
Collapse
Affiliation(s)
- Eric Van Otterloo
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
| | | | | | | |
Collapse
|
26
|
Pingault V, Bodereau V, Baral V, Marcos S, Watanabe Y, Chaoui A, Fouveaut C, Leroy C, Vérier-Mine O, Francannet C, Dupin-Deguine D, Archambeaud F, Kurtz FJ, Young J, Bertherat J, Marlin S, Goossens M, Hardelin JP, Dodé C, Bondurand N. Loss-of-function mutations in SOX10 cause Kallmann syndrome with deafness. Am J Hum Genet 2013; 92:707-24. [PMID: 23643381 DOI: 10.1016/j.ajhg.2013.03.024] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 02/25/2013] [Accepted: 03/29/2013] [Indexed: 12/11/2022] Open
Abstract
Transcription factor SOX10 plays a role in the maintenance of progenitor cell multipotency, lineage specification, and cell differentiation and is a major actor in the development of the neural crest. It has been implicated in Waardenburg syndrome (WS), a rare disorder characterized by the association between pigmentation abnormalities and deafness, but SOX10 mutations cause a variable phenotype that spreads over the initial limits of the syndrome definition. On the basis of recent findings of olfactory-bulb agenesis in WS individuals, we suspected SOX10 was also involved in Kallmann syndrome (KS). KS is defined by the association between anosmia and hypogonadotropic hypogonadism due to incomplete migration of neuroendocrine gonadotropin-releasing hormone (GnRH) cells along the olfactory, vomeronasal, and terminal nerves. Mutations in any of the nine genes identified to date account for only 30% of the KS cases. KS can be either isolated or associated with a variety of other symptoms, including deafness. This study reports SOX10 loss-of-function mutations in approximately one-third of KS individuals with deafness, indicating a substantial involvement in this clinical condition. Study of SOX10-null mutant mice revealed a developmental role of SOX10 in a subpopulation of glial cells called olfactory ensheathing cells. These mice indeed showed an almost complete absence of these cells along the olfactory nerve pathway, as well as defasciculation and misrouting of the nerve fibers, impaired migration of GnRH cells, and disorganization of the olfactory nerve layer of the olfactory bulbs.
Collapse
Affiliation(s)
- Veronique Pingault
- Equipe 11, Institut National de la Santé et de la Recherche Médicale Unité 955, 94000 Créteil, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Xia X, Chen J, Zhang L, Du Q, Sun J, Chang Z. Molecular cloning and mRNA expression pattern of Sox10 in Paramisgurnus dabryanus. Mol Biol Rep 2012; 40:3123-34. [DOI: 10.1007/s11033-012-2386-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 12/17/2012] [Indexed: 02/06/2023]
|
28
|
Santana MM, Chung KF, Vukicevic V, Rosmaninho-Salgado J, Kanczkowski W, Cortez V, Hackmann K, Bastos CA, Mota A, Schrock E, Bornstein SR, Cavadas C, Ehrhart-Bornstein M. Isolation, characterization, and differentiation of progenitor cells from human adult adrenal medulla. Stem Cells Transl Med 2012. [PMID: 23197690 DOI: 10.5966/sctm.2012-0022] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chromaffin cells, sympathetic neurons of the dorsal ganglia, and the intermediate small intensely fluorescent cells derive from a common neural crest progenitor cell. Contrary to the closely related sympathetic nervous system, within the adult adrenal medulla a subpopulation of undifferentiated progenitor cells persists, and recently, we established a method to isolate and differentiate these progenitor cells from adult bovine adrenals. However, no studies have elucidated the existence of adrenal progenitor cells within the human adrenal medulla. Here we describe the isolation, characterization, and differentiation of chromaffin progenitor cells obtained from adult human adrenals. Human chromaffin progenitor cells were cultured in low-attachment conditions for 10-12 days as free-floating spheres in the presence of fibroblast growth factor-2 (FGF-2) and epidermal growth factor. These primary human chromosphere cultures were characterized by the expression of several progenitor markers, including nestin, CD133, Notch1, nerve growth factor receptor, Snai2, Sox9, Sox10, Phox2b, and Ascl1 on the molecular level and of Sox9 on the immunohistochemical level. In opposition, phenylethanolamine N-methyltransferase (PNMT), a marker for differentiated chromaffin cells, significantly decreased after 12 days in culture. Moreover, when plated on poly-l-lysine/laminin-coated slides in the presence of FGF-2, human chromaffin progenitor cells were able to differentiate into two distinct neuron-like cell types, tyrosine hydroxylase (TH)(+)/β-3-tubulin(+) cells and TH(-)/β-3-tubulin(+) cells, and into chromaffin cells (TH(+)/PNMT(+)). This study demonstrates the presence of progenitor cells in the human adrenal medulla and reveals their potential use in regenerative medicine, especially in the treatment of neuroendocrine and neurodegenerative diseases.
Collapse
|
29
|
Abstract
Schwann cells are the main glial cell type in the PNS. They develop along nerves during embryogenesis and rely on the HMG domain containing Sox10 transcription factor for specification, lineage progression, and terminal differentiation. Sox10 deletion in immature Schwann cells caused peripheral nerve defects in mice that were not restricted to this glial cell type, although expression in the nerve and gene loss were. Formation of the perineurium as the protecting sheath was, for instance, heavily compromised. This resembled the defect observed after loss of Desert hedgehog (Dhh) in mice. Here we show that Sox10 activates Dhh expression in Schwann cells via an enhancer that is located in intron 1 of the Dhh gene. Sox10 binds this enhancer in monomeric form via several sites. Mutation of these sites abolishes both Schwann-cell-specific activity and Sox10 responsiveness in vitro and in transgenic mouse embryos. This argues that Sox10 activates Dhh expression by direct binding to the enhancer and by increasing Dhh levels promotes formation of the perineurial sheath. This represents the first mechanism for a non-cell-autonomous function of Sox10 during peripheral nerve development.
Collapse
|
30
|
Kerosuo L, Bronner-Fraser M. What is bad in cancer is good in the embryo: importance of EMT in neural crest development. Semin Cell Dev Biol 2012; 23:320-32. [PMID: 22430756 PMCID: PMC3345076 DOI: 10.1016/j.semcdb.2012.03.010] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 02/14/2012] [Accepted: 03/01/2012] [Indexed: 11/18/2022]
Abstract
Although the epithelial to mesenchymal transition (EMT) is famous for its role in cancer metastasis, it also is a normal developmental event in which epithelial cells are converted into migratory mesenchymal cells. A prime example of EMT during development occurs when neural crest (NC) cells emigrate from the neural tube thus providing an excellent model to study the principles of EMT in a nonmalignant environment. NC cells start life as neuroepithelial cells intermixed with precursors of the central nervous system. After EMT, they delaminate and begin migrating, often to distant sites in the embryo. While proliferating and maintaining multipotency and cell survival the transitioning neural crest cells lose apicobasal polarity and the basement membrane is broken down. This review discusses how these events are coordinated and regulated, by series of events involving signaling factors, gene regulatory interactions, as well as epigenetic and post-transcriptional modifications. Even though the series of events involved in NC EMT are well known, the sequence in which these steps take place remains a subject of debate, raising the intriguing possibility that, rather than being a single event, neural crest EMT may involve multiple parallel mechanisms.
Collapse
Affiliation(s)
- Laura Kerosuo
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, United States
| | | |
Collapse
|
31
|
Vukicevic V, Schmid J, Hermann A, Lange S, Qin N, Gebauer L, Chunk KF, Ravens U, Eisenhofer G, Storch A, Ader M, Bornstein SR, Ehrhart-Bornstein M. Differentiation of chromaffin progenitor cells to dopaminergic neurons. Cell Transplant 2012; 21:2471-86. [PMID: 22507143 DOI: 10.3727/096368912x638874] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The differentiation of dopamine-producing neurons from chromaffin progenitors might represent a new valuable source for replacement therapies in Parkinson's disease. However, characterization of their differentiation potential is an important prerequisite for efficient engraftment. Based on our previous studies on isolation and characterization of chromaffin progenitors from adult adrenals, this study investigates their potential to produce dopaminergic neurons and means to enhance their dopaminergic differentiation. Chromaffin progenitors grown in sphere culture showed an increased expression of nestin and Mash1, indicating an increase of the progenitor subset. Proneurogenic culture conditions induced the differentiation into neurons positive for neural markers β-III-tubulin, MAP2, and TH accompanied by a decrease of Mash1 and nestin. Furthermore, Notch2 expression decreased concomitantly with a downregulation of downstream effectors Hes1 and Hes5 responsible for self-renewal and proliferation maintenance of progenitor cells. Chromaffin progenitor-derived neurons secreted dopamine upon stimulation by potassium. Strikingly, treatment of differentiating cells with retinoic and ascorbic acid resulted in a twofold increase of dopamine secretion while norepinephrine and epinephrine were decreased. Initiation of dopamine synthesis and neural maturation is controlled by Pitx3 and Nurr1. Both Pitx3 and Nurr1 were identified in differentiating chromaffin progenitors. Along with the gained dopaminergic function, electrophysiology revealed features of mature neurons, such as sodium channels and the capability to fire multiple action potentials. In summary, this study elucidates the capacity of chromaffin progenitor cells to generate functional dopaminergic neurons, indicating their potential use in cell replacement therapies.
Collapse
Affiliation(s)
- Vladimir Vukicevic
- Molecular Endocrinology, Medical Clinic III, University Clinic Dresden, Dresden University of Technology, Fetscherstrasse 74, Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Modulation of Dopaminergic Neuronal Differentiation from Sympathoadrenal Progenitors. J Mol Neurosci 2012; 48:420-6. [DOI: 10.1007/s12031-012-9746-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 03/05/2012] [Indexed: 12/25/2022]
|
33
|
Simon C, Lickert H, Götz M, Dimou L. Sox10-iCreERT2 : a mouse line to inducibly trace the neural crest and oligodendrocyte lineage. Genesis 2012; 50:506-15. [PMID: 22173870 DOI: 10.1002/dvg.22003] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 12/02/2011] [Accepted: 12/12/2011] [Indexed: 11/11/2022]
Abstract
SOX10 is a well-conserved and widely expressed transcription factor involved in the regulation of embryonic development and in the determination of cell fate. As it is expressed in neural crest cells, their derivatives and the oligodendrocyte lineage, mutations of the protein contribute to a variety of diseases like neurocristopathies, peripheral demyelinating neuropathies, and melanoma. Here, we report the generation of an inducible Sox10-iCreER(T2) BAC transgenic mouse line that labels, depending on the timepoint of induction, distinct derivatives of the otic placode and the neural crest as well as cells of the oligodendrocyte lineage. Surprisingly, we could show a neural crest origin of pericytes in the brain. Besides its use for fate-mapping, the Sox10-iCreER(T2) mouse line is a powerful tool to conditionally inactivate genes in the neural crest cells, their progeny and/or the oligodendrocyte lineage in a time-dependent fashion to gain further insights into their function and contribution to diseases.
Collapse
Affiliation(s)
- Christiane Simon
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University, Munich, Germany
| | | | | | | |
Collapse
|
34
|
Lakiza O, Miller S, Bunce A, Lee EMJ, McCauley DW. SoxE gene duplication and development of the lamprey branchial skeleton: Insights into development and evolution of the neural crest. Dev Biol 2011; 359:149-161. [PMID: 21889937 DOI: 10.1016/j.ydbio.2011.08.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 08/06/2011] [Accepted: 08/19/2011] [Indexed: 11/25/2022]
Abstract
SoxE genes are multifunctional transcriptional regulators that play key roles in specification and differentiation of neural crest. Three members (Sox8, Sox9, Sox10) are expressed in the neural crest and are thought to modulate the expression and activity of each other. In addition to regulating the expression of other early neural crest marker genes, SoxE genes are required for development of cartilage. Here we investigated the role of SoxE genes in development of the neural crest-derived branchial skeleton in the sea lamprey. Using a morpholino knockdown approach, we show that all three SoxE genes described in lamprey are required for branchial basket development. Our results suggest that SoxE1 and SoxE2 are required for specification of the chondrogenic neural crest. SoxE3 plays a morphogenetic role in patterning of the branchial basket and may be required for the development of mucocartilage, a tissue unique to larval lampreys. While the lamprey branchial basket develops primarily from an elastin-like major extracellular matrix protein that is specific to lampreys, fibrillar collagen is also expressed in developing branchial cartilage and may be regulated by the lamprey SoxE genes. Our data suggest that the regulation of Type II collagen by Sox9 might have been co-opted by the neural crest in development of the branchial skeleton following the divergence of agnathan and gnathostome vertebrates. Finally, our results also have implications for understanding the independent evolution of duplicated SoxE genes among agnathan and gnathostome vertebrates.
Collapse
Affiliation(s)
- Olga Lakiza
- Department of Zoology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, United States
| | - Sarah Miller
- Department of Zoology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, United States
| | - Ashley Bunce
- Department of Zoology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, United States
| | - Eric Myung-Jae Lee
- Department of Zoology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, United States
| | - David W McCauley
- Department of Zoology, University of Oklahoma, 730 Van Vleet Oval, Norman, OK 73019, United States.
| |
Collapse
|
35
|
Lee FY, Faivre EJ, Suzawa M, Lontok E, Ebert D, Cai F, Belsham DD, Ingraham HA. Eliminating SF-1 (NR5A1) sumoylation in vivo results in ectopic hedgehog signaling and disruption of endocrine development. Dev Cell 2011; 21:315-27. [PMID: 21820362 DOI: 10.1016/j.devcel.2011.06.028] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 03/23/2011] [Accepted: 06/24/2011] [Indexed: 11/16/2022]
Abstract
Sumoylation is generally considered a repressive mark for many transcription factors. However, the in vivo importance of sumoylation for any given substrate remains unclear and is questionable because the extent of sumoylation appears exceedingly low for most substrates. Here, we permanently eliminated SF-1/NR5A1 sumoylation in mice (Sf-1(K119R, K194R, or 2KR)) and found that Sf-1(2KR/2KR) mice failed to phenocopy a simple gain of SF-1 function or show elevated levels of well-established SF-1 target genes. Instead, mutant mice exhibited marked endocrine abnormalities and changes in cell fate that reflected an inappropriate activation of hedgehog signaling and other potential SUMO-sensitive targets. Furthermore, unsumoylatable SF-1 mutants activated Shh and exhibited preferential recruitment to Shh genomic elements in cells. We conclude that the sumoylation cycle greatly expands the functional capacity of transcription factors such as SF-1 and is leveraged during development to achieve cell-type-specific gene expression in multicellular organisms.
Collapse
Affiliation(s)
- Florence Y Lee
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Bremer M, Fröb F, Kichko T, Reeh P, Tamm ER, Suter U, Wegner M. Sox10 is required for Schwann-cell homeostasis and myelin maintenance in the adult peripheral nerve. Glia 2011; 59:1022-32. [PMID: 21491499 DOI: 10.1002/glia.21173] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 03/16/2011] [Indexed: 11/07/2022]
Abstract
The transcription factor Sox10 functions during multiple consecutive stages of Schwann-cell development in the peripheral nervous system (PNS). Although Sox10 continues to be expressed in mature Schwann cells of the adult peripheral nerve, it is currently unclear whether it is still functional. Here, we used a genetic strategy to selectively delete Sox10 in glia of adult mice in a tamoxifen-dependent manner. The tamoxifen-treated mice developed a severe peripheral neuropathy that was associated with dramatic alterations in peripheral nerve structure and function. Demyelination and axonal degeneration were as much evident as signs of neuroinflammation. Compound action potentials exhibited pathophysiological alterations. Sox10-deleted Schwann cells persisted in the peripheral nerve, but did not exhibit a mature, myelinating phenotype arguing that Sox10 is rather required for differentiation and maintenance of the differentiated state than for survival. Our report is the first evidence that Sox10 is still essentially required for Schwann-cell function in the adult PNS and establishes a useful model in which to study human peripheral neuropathies.
Collapse
Affiliation(s)
- Magdalena Bremer
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Ehrhart-Bornstein M, Vukicevic V, Chung KF, Ahmad M, Bornstein SR. Chromaffin progenitor cells from the adrenal medulla. Cell Mol Neurobiol 2010; 30:1417-23. [PMID: 21080061 PMCID: PMC11498770 DOI: 10.1007/s10571-010-9571-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 09/02/2010] [Indexed: 11/26/2022]
Abstract
Chromaffin cells of the adrenal medulla are neural crest-derived cells of the sympathoadrenal lineage. Different lines of evidence suggest the existence of a subpopulation of proliferation-competent progenitor cells even in the adult state. The identification of sympathoadrenal progenitors in the adrenal would greatly enhance the understanding of adrenal physiology and their potential role in adrenal pathogenesis. Isolation and differentiation of these progenitor cells in culture would provide a tool to understand their development in vitro. Furthermore, due to the close relation to sympathetic neurons, these cells might provide an expandable source of cells for cell therapy in the treatment of neurodegenerative diseases. We therefore aim to establish protocols for the efficient isolation, enrichment and differentiation of chromaffin progenitor cells to dopaminergic neurons in culture.
Collapse
|
38
|
Inoue S, Cho BH, Song CH, Fujimiya M, Murakami G, Matsubara A. Migration and distribution of neural crest-derived cells in the human adrenal cortex at 9-16 weeks of gestation: an immunohistochemical study. Okajimas Folia Anat Jpn 2010; 87:11-16. [PMID: 20715567 DOI: 10.2535/ofaj.87.11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Neural crest-derived cells are believed to migrate into the fetal adrenal cortex from the medially-located hilus. However, there appears to be a paucity of observations of the migration and distribution of medullary cells in humans. In sagittal as well as horizontal sections of human fetuses between 9 and 16 weeks of gestation, we identified chromaffin, ganglion and Schwann-like cells in the developing adrenal gland using immunohistochemistry. Cells showing tyrosine hydroxylase (TH) immunoreactivity (i.e., candidate ganglion cells) entered the fetal cortex mainly from the medial half of the adrenal, but the path of entry also included the ventral, dorsal and caudal aspects. These cells displayed linear arrangements, forming a connection between the peripheral and central areas of the gland. S100 protein-immunoreactive cells (i.e., Schwann-like cells) accompanied most (but not all) of the TH-positive cells. The distribution of chromogranin A-immunoreactive cells (i.e., chromaffin cells) was similar to and overlapped with that of TH-positive cells. Chromogranin A-positive cells were observed around the aorta as well as in the adrenal. The entry of neural crest-derived cells does not appear to be restricted to a hypothetical medial hilus, but occurs widely around the cortex, with or without the accompaniment of Schwann-like cells. These cells advance in lines through the fetal cortex in a cord-like arrangement without destruction of the cortical architecture. Some of the TH-positive cells very likely express chromogranin A before entry into the adrenal.
Collapse
Affiliation(s)
- Shogo Inoue
- Department of Urology, Graduate School of Biomedical Sciences, Hiroshima University, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Cossais F, Wahlbuhl M, Kriesch J, Wegner M. SOX10 structure-function analysis in the chicken neural tube reveals important insights into its role in human neurocristopathies. Hum Mol Genet 2010; 19:2409-20. [PMID: 20308050 DOI: 10.1093/hmg/ddq124] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The HMG-domain containing transcription factor Sox10 is essential for neural crest (NC) development and for oligodendrocyte differentiation. Heterozygous SOX10 mutations in humans lead to corresponding defects in several NC-derived lineages and to leukodystrophies. Disease phenotypes range from Waardenburg syndrome and Waardenburg-Hirschsprung disease to Peripheral demyelinating neuropathy, Central dysmyelination, Waardenburg syndrome and Hirschsprung disease (PCWH). The phenotypic variability can partly be explained by the action of modifier genes, but is also influenced by the mutation that leads to haploinsufficiency in some and to mutant SOX10 proteins with altered properties in other cases. Here, we used in ovo electroporation in the developing neural tube of chicken to determine which regions and properties of SOX10 are required for early NC development. We found a strict reliance on the DNA-binding activity and the presence of the C-terminal transactivation domain and a lesser influence of the dimerization function and a conserved domain in the center of the protein. Intriguingly, dominant-negative effects on early NC development were mostly observed for truncated SOX10 proteins whose production in patients is probably prevented by nonsense-mediated decay. In contrast, mutant SOX10 proteins that occur in patients were usually inactive. Any dominant negative activity which some of these mutants undoubtedly possess must, therefore, be restricted to single NC-derived cell lineages or oligodendrocytes at later times. This contributes to the phenotypic variability of human SOX10 mutations.
Collapse
Affiliation(s)
- François Cossais
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen, Erlangen, Germany
| | | | | | | |
Collapse
|
40
|
Potzner MR, Tsarovina K, Binder E, Penzo-Méndez A, Lefebvre V, Rohrer H, Wegner M, Sock E. Sequential requirement of Sox4 and Sox11 during development of the sympathetic nervous system. Development 2010; 137:775-84. [PMID: 20147379 DOI: 10.1242/dev.042101] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The highly related transcription factors Sox4 and Sox11 are expressed in the developing sympathetic nervous system. In the mouse, Sox11 appears first, whereas Sox4 is prevalent later. Using mouse mutagenesis and overexpression strategies in chicken, we studied the role of both SoxC proteins in this tissue. Neither Sox4 nor Sox11 predominantly functioned by promoting pan-neuronal or noradrenergic differentiation of sympathetic neurons as might have been expected from studies in neuronal precursors of the central nervous system. The transcriptional network that regulates the differentiation of sympathetic neurons remained intact and expression of noradrenergic markers showed only minor alterations. Instead, Sox11 was required in early sympathetic ganglia for proliferation of tyrosine hydroxylase-expressing cells, whereas Sox4 ensured the survival of these cells at later stages. In the absence of both Sox4 and Sox11, sympathetic ganglia remained hypoplastic throughout embryogenesis because of consecutive proliferation and survival defects. As a consequence, sympathetic ganglia were rudimentary in the adult and sympathetic innervation of target tissues was impaired leading to severe dysautonomia.
Collapse
Affiliation(s)
- Michaela R Potzner
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Cossais F, Sock E, Hornig J, Schreiner S, Kellerer S, Bösl MR, Russell S, Wegner M. Replacement of mouse Sox10 by the Drosophila ortholog Sox100B provides evidence for co-option of SoxE proteins into vertebrate-specific gene-regulatory networks through altered expression. Dev Biol 2010; 341:267-81. [PMID: 20144603 DOI: 10.1016/j.ydbio.2010.01.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 01/29/2010] [Accepted: 01/29/2010] [Indexed: 11/26/2022]
Abstract
Neural crest cells and oligodendrocytes as the myelinating glia of the central nervous system exist only in vertebrates. Their development is regulated by complex regulatory networks, of which the SoxE-type high-mobility-group domain transcription factors Sox8, Sox9 and Sox10 are essential components. Here we analyzed by in ovo electroporation in chicken and by gene replacement in the mouse whether the Drosophila ortholog Sox100B can functionally substitute for vertebrate SoxE proteins. Sox100B overexpression in the chicken neural tube led to the induction of neural crest cells as previously observed for vertebrate SoxE proteins. Furthermore, many aspects of neural crest and oligodendrocyte development were surprisingly normal in mice in which the Sox10 coding information was replaced by Sox100B arguing that Sox100B integrates well into the gene-regulatory networks that drive these processes. Our results therefore provide strong evidence for a model in which SoxE proteins were co-opted to these gene-regulatory networks mainly through the acquisition of novel expression patterns. However, later developmental defects in several neural crest derived lineages in mice homozygous for the Sox100B replacement allele indicate that some degree of functional specialization and adaptation of SoxE protein properties have taken place in addition to the co-option event.
Collapse
Affiliation(s)
- François Cossais
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen, Fahrstrasse 17, D-91054 Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
|
43
|
|
44
|
Giuliani L, Lenzini L, Antonello M, Aldighieri E, Belloni AS, Fassina A, Gomez-Sanchez C, Rossi GP. Expression and functional role of urotensin-II and its receptor in the adrenal cortex and medulla: novel insights for the pathophysiology of primary aldosteronism. J Clin Endocrinol Metab 2009; 94:684-90. [PMID: 19001524 DOI: 10.1210/jc.2008-1131] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT The involvement of urotensin II, a vasoactive peptide acting via the G protein-coupled urotensin II receptor, in arterial hypertension remains contentious. OBJECTIVE We investigated the expression of urotensin II and urotensin II receptor in adrenocortical and adrenomedullary tumors and the functional effects of urotensin II receptor activation. DESIGN The expression of urotensin II and urotensin II receptor was measured by real time RT-PCR in aldosterone-producing adenoma (n = 22) and pheochromocytoma (n = 10), using histologically normal adrenocortical (n = 6) and normal adrenomedullary (n = 5) tissue as control. Urotensin II peptide and urotensin II receptor protein were investigated with immunohistochemistry and immunoblotting. To identify urotensin II-related and urotensin II receptor-related pathways, a whole transcriptome analysis was used. The adrenocortical effects of urotensin II receptor activation were also assessed by urotensin II infusion with/without the urotensin II receptor antagonist palosuran in rats. RESULTS Urotensin II was more expressed in pheochromocytoma than in aldosterone-producing adenoma tissue; the opposite was seen for the urotensin II receptor expression. Urotensin II receptor activation in vivo in rats enhanced (by 182 +/- 9%; P < 0.007) the adrenocortical expression of immunoreactive aldosterone synthase. CONCLUSIONS Urotensin II is a putative mediator of the effects of the adrenal medulla and pheochromocytoma on the adrenocortical zona glomerulosa. This pathophysiological link might account for the reported causal relationship between pheochromocytoma and primary aldosteronism.
Collapse
Affiliation(s)
- Luisa Giuliani
- Department of Clinical and Experimental Medicine, Internal Medicine 4, School of Medicine, University of Padua, Padua, Italy
| | | | | | | | | | | | | | | |
Collapse
|