1
|
Kojima K, Sakurai K, Ando T, Sakai Y, Ochiai M, Kato T, Ito H. Immunohistochemical investigation of the transcription factor PROX1 emphasizing on neuroendocrine neoplasms. Med Mol Morphol 2025:10.1007/s00795-025-00437-z. [PMID: 40234276 DOI: 10.1007/s00795-025-00437-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
Prospero homeobox protein 1 (PROX1) is aberrantly expressed in tumors, including neuroendocrine neoplasms (NENs); however, the detailed expression pattern remains elusive. This study aimed to immunohistochemically assess PROX1 expression. Immunohistochemistry (IHC) for PROX1 was performed on tissue microarrays of normal tissues (n = 107), NENs (n = 152) (small cell lung carcinoma [SCLC], lung carcinoid [LC], gastroenteropancreatic-NEN [GEP-NEN], esophageal neuroendocrine carcinoma [ENEC], medullary thyroid carcinoma [MTC], neuroblastoma [NB], and pheochromocytoma [PHEO]), and non-NENs (n = 469). In normal tissues, PROX1 was expressed in lymphatic endothelial cells and a subset of epithelial cells in the gastrointestinal tract and the distal convoluted tubules. In NENs, the positive expression was observed in the nucleus of tumor cells in 19/26 SCLC (73.1%), 13/16 LC (81.3%), 10/15 GEP-NEN (66.7%), 2/2 ENEC (100%), 17/43 MTC (39.5%), 1/25 NB (4.0%), and 0/25 PHEO (0%). Although PROX1 was negative in many non-NENs, our analysis revealed high expression in certain cases with medulloblastoma and one case with juvenile granulosa cell tumor. PROX1 was expressed in specific cases with epithelial NENs and some cases with non-NENs. Analysis of PROX1 should provide insights into the molecular characteristics of distinct tumors.
Collapse
Affiliation(s)
- Kanata Kojima
- Department of Clinical Laboratory, Fujita Health University Hospital, Toyoake, Aichi, 470-1192, Japan
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Kouhei Sakurai
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| | - Tatsuya Ando
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yasuhiro Sakai
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Mako Ochiai
- Department of Clinical Laboratory, Fujita Health University Hospital, Toyoake, Aichi, 470-1192, Japan
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Taku Kato
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Hiroyasu Ito
- Department of Clinical Laboratory, Fujita Health University Hospital, Toyoake, Aichi, 470-1192, Japan
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| |
Collapse
|
2
|
Sakurai K, Ando T, Sakai Y, Mori Y, Nakamura S, Kato T, Ito H. PROX1 is a regulator of neuroendocrine-related gene expression in lung carcinoid. Hum Cell 2024; 37:1559-1566. [PMID: 39066858 DOI: 10.1007/s13577-024-01109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024]
Abstract
Lung neuroendocrine neoplasms (NENs) are a diverse group of tumors characterized by neuroendocrine (NE) differentiation. Among lung NENs, lung carcinoid (LC) is a rare tumor with unique characteristics. Recent research has highlighted the importance of transcription factors (TFs) in establishing gene expression programs in lung NENs such as small cell lung carcinoma. However, the TFs that control the gene expression of LC are largely unknown. In this study, we report the expression and potential function of a TF called Prospero homeobox protein1 (PROX1) in LC. Publicly available transcriptome data suggested that PROX1 was highly expressed in LC tissues, which was confirmed by immunohistochemical analysis on a tissue microarray. Knockdown of PROX1 did not impact the cellular viability of an LC-derived cell line, NCI-H727. Meanwhile, transcriptome analysis revealed that PROX1 knockdown altered the expression of genes involved in NE differentiation. ASCL1, CHGA, CALCA, and LINC00261 were suggested as downstream genes of PROX1. These findings indicate that PROX1 may play an important role in the NE identity of LC by regulating the expression of key target genes.
Collapse
Affiliation(s)
- Kouhei Sakurai
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| | - Tatsuya Ando
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yasuhiro Sakai
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Yuichiro Mori
- School of Medicine, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Satoru Nakamura
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
- Central Research Laboratory, Nitto Fuji Flour Milling Co., Ltd., Tokyo, 143-0001, Japan
| | - Taku Kato
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Hiroyasu Ito
- Department of Joint Research Laboratory of Clinical Medicine, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| |
Collapse
|
3
|
Baur K, Carrillo-García C, Şan Ş, von Hahn M, Strelau J, Hölzl-Wenig G, Mandl C, Ciccolini F. Growth/differentiation factor 15 controls ependymal and stem cell number in the V-SVZ. Stem Cell Reports 2024; 19:351-365. [PMID: 38366596 PMCID: PMC10937156 DOI: 10.1016/j.stemcr.2024.01.008] [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: 07/28/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 02/18/2024] Open
Abstract
The expression of growth/differentiation factor (GDF) 15 increases in the ganglionic eminence (GE) late in neural development, especially in neural stem cells (NSCs). However, GDF15 function in this region remains unknown. We report that GDF15 receptor is expressed apically in the GE and that GDF15 ablation promotes proliferation and cell division in the embryonic GE and in the adult ventricular-subventricular zone (V-SVZ). This causes a transient generation of additional neuronal progenitors, compensated by cell death, and a lasting increase in the number of ependymal cells and apical NSCs. Finally, both GDF15 receptor and the epidermal growth factor receptor (EGFR) were expressed in progenitors and mutation of GDF15 affected EGFR signaling. However, only exposure to exogenous GDF15, but not to EGF, normalized proliferation and the number of apical progenitors. Thus, GDF15 regulates proliferation of apical progenitors in the GE, thereby affecting the number of ependymal cells and NSCs.
Collapse
Affiliation(s)
- Katja Baur
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Carmen Carrillo-García
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Şeydanur Şan
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; Sorbonne University, 21 Rue de l'École de Médecine, 75006 Paris, France
| | - Manja von Hahn
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Jens Strelau
- University of Heidelberg, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Gabriele Hölzl-Wenig
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Claudia Mandl
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Francesca Ciccolini
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany.
| |
Collapse
|
4
|
del Águila Á, Adam M, Ullom K, Shaw N, Qin S, Ehrman J, Nardini D, Salomone J, Gebelein B, Lu QR, Potter SS, Waclaw R, Campbell K, Nakafuku M. Olig2 defines a subset of neural stem cells that produce specific olfactory bulb interneuron subtypes in the subventricular zone of adult mice. Development 2022; 149:274286. [PMID: 35132995 PMCID: PMC8959153 DOI: 10.1242/dev.200028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
Distinct neural stem cells (NSCs) reside in different regions of the subventricular zone (SVZ) and generate multiple olfactory bulb (OB) interneuron subtypes in the adult brain. However, the molecular mechanisms underlying such NSC heterogeneity remain largely unknown. Here, we show that the basic helix-loop-helix transcription factor Olig2 defines a subset of NSCs in the early postnatal and adult SVZ. Olig2-expressing NSCs exist broadly but are most enriched in the ventral SVZ along the dorsoventral axis complementary to dorsally enriched Gsx2-expressing NSCs. Comparisons of Olig2-expressing NSCs from early embryonic to adult stages using single cell transcriptomics reveal stepwise developmental changes in their cell cycle and metabolic properties. Genetic studies further show that cross-repression contributes to the mutually exclusive expression of Olig2 and Gsx2 in NSCs/progenitors during embryogenesis, but that their expression is regulated independently from each other in adult NSCs. Finally, lineage-tracing and conditional inactivation studies demonstrate that Olig2 plays an important role in the specification of OB interneuron subtypes. Altogether, our study demonstrates that Olig2 defines a unique subset of adult NSCs enriched in the ventral aspect of the adult SVZ.
Collapse
Affiliation(s)
- Ángela del Águila
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Kristy Ullom
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Nicholas Shaw
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Medical Science, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA
| | - Shenyue Qin
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Jacqueline Ehrman
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Diana Nardini
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Joseph Salomone
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Brian Gebelein
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Q. Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA
| | - Steven S. Potter
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA
| | - Ronald Waclaw
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA
| | - Kenneth Campbell
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA,Division of Neurosurgery, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Masato Nakafuku
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA,Department of Pediatrics, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA,Department of Neurosurgery, University of Cincinnati College of Medicine, 3125 Eden Avenue, Cincinnati, OH 45267-0521, USA,Author for correspondence ()
| |
Collapse
|
5
|
Abstract
Adipose tissue, once thought to be an inert receptacle for energy storage, is now recognized as a complex tissue with multiple resident cell populations that actively collaborate in response to diverse local and systemic metabolic, thermal, and inflammatory signals. A key participant in adipose tissue homeostasis that has only recently captured broad scientific attention is the lymphatic vasculature. The lymphatic system's role in lipid trafficking and mediating inflammation makes it a natural partner in regulating adipose tissue, and evidence supporting a bidirectional relationship between lymphatics and adipose tissue has accumulated in recent years. Obesity is now understood to impair lymphatic function, whereas altered lymphatic function results in aberrant adipose tissue deposition, though the molecular mechanisms governing these phenomena have yet to be fully elucidated. We will review our current understanding of the relationship between adipose tissue and the lymphatic system here, focusing on known mechanisms of lymphatic-adipose crosstalk.
Collapse
Affiliation(s)
- Gregory P Westcott
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Joslin Diabetes Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
- Broad Institute, Cambridge, MA 02142, USA
- Correspondence: Evan D. Rosen, MD, PhD, Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
6
|
A suite of new Dre recombinase drivers markedly expands the ability to perform intersectional genetic targeting. Cell Stem Cell 2021; 28:1160-1176.e7. [PMID: 33567267 DOI: 10.1016/j.stem.2021.01.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/06/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
The use of the dual recombinase-mediated intersectional genetic approach involving Cre-loxP and Dre-rox has significantly enhanced the precision of in vivo lineage tracing, as well as gene manipulation. However, this approach is limited by the small number of Dre recombinase driver constructs available. Here, we developed more than 70 new intersectional drivers to better target diverse cell lineages. To highlight their applicability, we used these new tools to study the in vivo adipogenic fate of perivascular progenitors, which revealed that PDGFRa+ but not PDGFRa-PDGFRb+ perivascular cells are the endogenous progenitors of adult adipocytes. In addition to lineage tracing, we used members of this new suite of drivers to more specifically knock out genes in complex tissues, such as white adipocytes and lymphatic vessels, that heretofore cannot be selectively targeted by conventional Cre drivers alone. In summary, these new transgenic tools expand the intersectional genetic approach while enhancing its precision.
Collapse
|
7
|
Rudzińska M, Grzanka M, Stachurska A, Mikula M, Paczkowska K, Stępień T, Paziewska A, Ostrowski J, Czarnocka B. Molecular Signature of Prospero Homeobox 1 (PROX1) in Follicular Thyroid Carcinoma Cells. Int J Mol Sci 2019; 20:ijms20092212. [PMID: 31060342 PMCID: PMC6539481 DOI: 10.3390/ijms20092212] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/24/2019] [Accepted: 04/28/2019] [Indexed: 01/15/2023] Open
Abstract
The prospero homeobox 1 (PROX1) transcription factor is a product of one of the lymphangiogenesis master genes. It has also been suggested to play a role in carcinogenesis, although its precise role in tumour development and metastasis remains unclear. The aim of this study was to gain more knowledge on the PROX1 function in thyroid tumorigenesis. Follicular thyroid cancer-derived cells—CGTH-W-1—were transfected with PROX1-siRNA (small interfering RNA) and their proliferation, cell cycle, apoptosis and motility were then analysed. The transcriptional signature of PROX1 depletion was determined using RNA-Sequencing (RNA-Seq) and the expression of relevant genes was further validated using reverse transcriptase quantitative PCR (RT-qPCR), Western blot and immunocytochemistry. PROX1 depletion resulted in a decreased cell motility, with both migratory and invasive potential being significantly reduced. The cell morphology was also affected, while the other studied cancer-related cell characteristics were not significantly altered. RNA-seq analysis revealed significant changes in the expression of transcripts encoding genes involved in both motility and cytoskeleton organization. Our transcriptional analysis of PROX1-depleted follicular thyroid carcinoma cells followed by functional and phenotypical analyses provide, for the first time, evidence that PROX1 plays an important role in the metastasis of thyroid cancer cells by regulating genes involved in focal adhesion and cytoskeleton organization in tumour cells.
Collapse
Affiliation(s)
- Magdalena Rudzińska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland.
| | - Małgorzata Grzanka
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland.
| | - Anna Stachurska
- Department of Immunohematology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland.
| | - Michał Mikula
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, 02-781 Warsaw, Poland.
| | - Katarzyna Paczkowska
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, 02-781 Warsaw, Poland.
| | - Tomasz Stępień
- Clinic of Endocrinological and General Surgery, Medical University of Lodz, 93-513 Lodz, Poland.
| | - Agnieszka Paziewska
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland.
| | - Jerzy Ostrowski
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, 02-781 Warsaw, Poland.
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland.
| | - Barbara Czarnocka
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland.
| |
Collapse
|
8
|
Dong ZY, Pei Z, Wang YL, Li Z, Khan A, Meng XT. Ascl1 Regulates Electric Field-Induced Neuronal Differentiation Through PI3K/Akt Pathway. Neuroscience 2019; 404:141-152. [PMID: 30771509 DOI: 10.1016/j.neuroscience.2019.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 12/14/2022]
Abstract
Directing differentiation of neural stem/progenitor cells (NSCs/NPCs) to produce functional neurons is one of the greatest challenges in regenerative medicine. Our previous paper has confirmed that electrical stimulation has a high efficiency of triggering neuronal differentiation by using isolated filum terminale (FT)-derived NPCs. To further clarify the intrinsic molecular mechanisms, protein-protein interaction (PPI) network analysis was applied to pinpoints novel hubs in electric field (EF)-induced neuronal differentiation. In this study, siRNA transfection of Achaete-scute homolog 1 (Ascl1) in NPCs or NPCs was followed by direct current stimulation at 150 mV/mm. Neuronal differentiation rate and protein expression level were analyzed after 7 or 14 days of electrical stimulation. The data showed that the expression level of Ascl1 was enhanced by electrical stimulation and positively correlated to EF strength. Moreover, we identified that the expression of Ascl1 positively regulated neuronal differentiation of NPCs and can be up-regulated by EF-stimulation through the activation of phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway. Therefore, this study provides new insights into the role of Ascl1 and its relevant PI3K/Akt pathway in regulating of EF-induced neuronal differentiation and pointed out that continuous expression of Ascl1 in NPCs is required for EF-induced neuronal differentiation.
Collapse
Affiliation(s)
- Zhi-Yong Dong
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Zhe Pei
- Department of Neuroscience and Pediatric, GSRB1 Duke University, Durham 27710, USA
| | - Yan-Ling Wang
- Laboratory Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Zhe Li
- Laboratory Teaching Center of Basic Medicine, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Amber Khan
- The Graduate Center and CUNY School of Medicine, CUNY, 85 St Nicholas Terrace, New York, NY 10027, USA.
| | - Xiao-Ting Meng
- Department of Histology & Embryology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
9
|
Arimura N, Dewa KI, Okada M, Yanagawa Y, Taya SI, Hoshino M. Comprehensive and cell-type-based characterization of the dorsal midbrain during development. Genes Cells 2018; 24:41-59. [PMID: 30422377 DOI: 10.1111/gtc.12656] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/15/2018] [Accepted: 10/24/2018] [Indexed: 01/09/2023]
Abstract
The layer structure has been intensively characterized in the developing neocortex and cerebellum based on the various molecular markers. However, as to the developing dorsal midbrain, comprehensive analyses have not been intensely carried out, and thus, the name as well as the definition of each layer is not commonly shared. Here, we redefined the three layers, such as the ventricular zone, intermediate zone and marginal zone, based on various markers for proliferation and differentiation in embryonic dorsal midbrain. Biphasic Ki67 expression defines the classical VZ, in which there is clear separation of the mitotic and interphase zones. Next, we mapped the distribution of immature neurons to the defined layers, based on markers for glutamatergic and GABAergic lineage. Interestingly, Tbr2 and Neurog2 were expressed in the postmitotic neurons. We also report that active (phosphorylated) JNK is a useful marker to demarcate layers during the embryonic stage. Finally, we validated the final arrival layers of the migratory glutamatergic and GABAergic neurons. These results form a foundation for analyses of brain development, especially in the proliferation and migration of excitatory and inhibitory neurons in the dorsal midbrain.
Collapse
Affiliation(s)
- Nariko Arimura
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Ken-Ichi Dewa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Pharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Mako Okada
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Pharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Shin-Ichiro Taya
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| |
Collapse
|
10
|
McClay DR, Miranda E, Feinberg SL. Neurogenesis in the sea urchin embryo is initiated uniquely in three domains. Development 2018; 145:dev167742. [PMID: 30413529 PMCID: PMC6240313 DOI: 10.1242/dev.167742] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/25/2018] [Indexed: 12/23/2022]
Abstract
Many marine larvae begin feeding within a day of fertilization, thus requiring rapid development of a nervous system to coordinate feeding activities. Here, we examine the patterning and specification of early neurogenesis in sea urchin embryos. Lineage analysis indicates that neurons arise locally in three regions of the embryo. Perturbation analyses showed that when patterning is disrupted, neurogenesis in the three regions is differentially affected, indicating distinct patterning requirements for each neural domain. Six transcription factors that function during proneural specification were identified and studied in detail. Perturbations of these proneural transcription factors showed that specification occurs differently in each neural domain prior to the Delta-Notch restriction signal. Though gene regulatory network state changes beyond the proneural restriction are largely unresolved, the data here show that the three neural regions already differ from each other significantly early in specification. Future studies that define the larval nervous system in the sea urchin must therefore separately characterize the three populations of neurons that enable the larva to feed, to navigate, and to move food particles through the gut.
Collapse
Affiliation(s)
- David R McClay
- Department of Biology, 124 Science Drive, Duke University, Durham, NC 27708, USA
| | - Esther Miranda
- Department of Biology, 124 Science Drive, Duke University, Durham, NC 27708, USA
| | - Stacy L Feinberg
- Department of Biology, 124 Science Drive, Duke University, Durham, NC 27708, USA
| |
Collapse
|
11
|
Shen W, Li Y, Cao L, Cai X, Ge Y, Zhu W. Decreased Expression of Prox1 Is Associated With Postoperative Recurrence in Crohn's Disease. J Crohns Colitis 2018; 12:1210-1218. [PMID: 29947772 DOI: 10.1093/ecco-jcc/jjy091] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Prox1 is a transcription factor necessary for lymphangiogenesis and lymphatic function. The aim of the study was to assess the correlation between the expression of Prox1 and postoperative recurrence in Crohn's disease [CD]. METHODS Forty CD patients who underwent ileo-colonic resection were included. Expression levels of Prox1 and D2-40 were detected using immunohistochemistry. Expression levels of Prox1, VEGFR3, and VEGFC protein were also detected in fresh CD specimens using western blotting and quantitative polymerase chain reaction [Q-PCR]. Endoscopic recurrence was used as the endpoint. Patients comprised two groups: endoscopic recurrence [Group R+] and no endoscopic recurrence [Group R-]. RESULTS Prox1 protein expression was significantly higher in CD than in normal tissues [p <0.05], as detected using both immunohistochemistry and western blotting. Analysis of inter-relationships revealed significant correlation between Prox1 expression and lymphatic vessel density [p <0.001, r = 0.823]. There was also significant correlation between Prox1 expression and the visceral fat area [VFA] [p = 0.002, r = -0.469]. The Group R- patients had significantly higher Prox1 expression than the Group R+ patients [21.08 ± 1.61 versus 15.64 ± 1.17, p = 0.011]. Also, the lymphatic vessel density value was lower in Group R+ than in Group R- patients [6.02 ± 0.39 versus 8.13 ± 0.59, p = 0.004]. Moreover, there was a significant difference in the VFA between Group R- and Group R+ patients [64.43 ± 7.76 versus 90.44 ± 6.11, p = 0.016]. In addition to Prox1, VEGFC/VEGFR3 was found to increase, which was further confirmed using Q-PCR. CONCLUSIONS Prox1 expression could be useful as a protective factor against recurrence in CD patients. The therapeutic role of Prox1 may lead to improved treatments.
Collapse
Affiliation(s)
- Weisong Shen
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yi Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lei Cao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xingchen Cai
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuanyuan Ge
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Weiming Zhu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
12
|
Kubová H, Folbergrová J, Rejchrtová J, Tsenov G, Pařízková M, Burchfiel J, Mikulecká A, Mareš P. The Free Radical Scavenger N-Tert-Butyl-α-Phenylnitrone (PBN) Administered to Immature Rats During Status Epilepticus Alters Neurogenesis and Has Variable Effects, Both Beneficial and Detrimental, on Long-Term Outcomes. Front Cell Neurosci 2018; 12:266. [PMID: 30210297 PMCID: PMC6121067 DOI: 10.3389/fncel.2018.00266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022] Open
Abstract
Status epilepticus (SE), especially in immature animals, is known to produce recurrent spontaneous seizures and behavioral comorbidities later in life. The cause of these adverse long-term outcomes is unknown, but it has been hypothesized that free radicals produced by SE may play a role. We tested this hypothesis by treating immature (P25) rats with the free radical scavenger N-tert-butyl-α-phenylnitrone (PBN) at the time of lithium chloride (LiCl)/pilocarpine (PILO)-induced SE. Later, long-term outcomes were assessed. Cognitive impairment (spatial memory) was tested in the Morris water maze (MWM). Emotional disturbances were assessed by the capture test (aggressiveness) and elevated plus maze's (EPM) test (anxiety). Next, the presence and severity of spontaneous seizures were assessed by continuous video/EEG monitoring for 5 days. Finally, immunochemistry, stereology and morphology were used to assess the effects of PBN on hippocampal neuropathology and neurogenesis. PBN treatment modified the long-term effects of SE in varying ways, some beneficial and some detrimental. Beneficially, PBN protected against severe anatomical damage in the hippocampus and associated spatial memory impairment. Detrimentally, PBN treated animals had more severe seizures later in life. PBN also made animals more aggressive and more anxious. Correlating with these detrimental long-term outcomes, PBN significantly modified post-natal neurogenesis. Treated animals had significantly increased numbers of mature granule cells (GCs) ectopically located in the dentate hilus (DH). These results raise the possibility that abnormal neurogenesis may significantly contribute to the development of post-SE epilepsy and behavioral comorbidities.
Collapse
Affiliation(s)
- Hana Kubová
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jaroslava Folbergrová
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jana Rejchrtová
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Grygoriy Tsenov
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martina Pařízková
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - James Burchfiel
- Strong Epilepsy Center, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Anna Mikulecká
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Pavel Mareš
- Department of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
13
|
Goudarzi KM, Espinoza JA, Guo M, Bartek J, Nistér M, Lindström MS, Hägerstrand D. Reduced Expression of PROX1 Transitions Glioblastoma Cells into a Mesenchymal Gene Expression Subtype. Cancer Res 2018; 78:5901-5916. [PMID: 30135192 DOI: 10.1158/0008-5472.can-18-0320] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/01/2018] [Accepted: 08/16/2018] [Indexed: 11/16/2022]
Abstract
The homeodomain transcription factor PROX1 has been linked to several cancer types, including gliomas, but its functions remain to be further elucidated. Here we describe a functional role and the prognostic value of PROX1 in glioblastoma. Low expression of PROX1 correlated with poor overall survival and the mesenchymal glioblastoma subtype signature. The latter finding was recapitulated in vitro, where suppression or overexpression of PROX1 in glioma cell cultures transitioned cells to a mesenchymal or to a nonmesenchymal glioblastoma gene expression signature, respectively. PROX1 modulation affected proliferation rates that coincided with changes in protein levels of CCNA1 and CCNE1 as well as the cyclin inhibitors CDKN1A, CDKN1B, and CDKN1C. Overexpression of SOX2 increased PROX1 expression, but treatment with a CDK2 inhibitor subsequently decreased PROX1 expression, which was paralleled by decreased SOX2 levels. The THRAP3 protein was a novel binding partner for PROX1, and suppression of THRAP3 increased both transcript and protein levels of PROX1. Together, these findings highlight the prognostic value of PROX1 and its role as a regulator of glioblastoma gene expression subtypes, intratumoral heterogeneity, proliferation, and cell-cycle control.Significance: These findings demonstrate the role and prognostic value of PROX1 in glioblastomas; low PROX1 levels correlate with a mesenchymal gene expression subtype and shorter survival in glioblastoma tumors. Cancer Res; 78(20); 5901-16. ©2018 AACR.
Collapse
Affiliation(s)
- Kaveh M Goudarzi
- SciLifeLab, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jaime A Espinoza
- SciLifeLab, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Min Guo
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital at Solna, Stockholm, Sweden
| | - Jiri Bartek
- SciLifeLab, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- The Danish Cancer Society Research Centre, Copenhagen, Denmark
| | - Monica Nistér
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital at Solna, Stockholm, Sweden
| | - Mikael S Lindström
- SciLifeLab, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Hägerstrand
- Cancer Center Karolinska, Department of Oncology-Pathology, Karolinska Institutet and Karolinska University Hospital at Solna, Stockholm, Sweden.
| |
Collapse
|
14
|
Shaikh MN, Tejedor FJ. Mnb/Dyrk1A orchestrates a transcriptional network at the transition from self-renewing neurogenic progenitors to postmitotic neuronal precursors. J Neurogenet 2018; 32:37-50. [PMID: 29495936 DOI: 10.1080/01677063.2018.1438427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The Down syndrome and microcephaly related gene Mnb/Dyrk1A encodes an evolutionary conserved protein kinase subfamily that plays important roles in neurodevelopment. minibrain (mnb) mutants of Drosophila melanogaster (Dm) exhibit reduced adult brains due to neuronal deficits generated during larval development. These deficits are the consequence of the apoptotic cell death of numerous neuronal precursors that fail to properly exit the cell cycle and differentiate. We have recently found that in both the Dm larval brain and the embryonic vertebrate central nervous system (CNS), a transient expression of Mnb/Dyrk1A promotes the cell cycle exit of newborn neuronal precursors by upregulating the expression of the cyclin-dependent kinase inhibitor p27kip1 (called Dacapo in Dm). In the larval brain, Mnb performs this action by regulating the expression of three transcription factors, Asense (Ase), Deadpan (Dpn) and Prospero (Pros), which are key regulators of the self-renewal, proliferation, and terminal differentiation of neural progenitor cells. We have here studied in detail the cellular/temporal expression pattern of Ase, Dpn, Pros and Mnb, and have analyzed possible regulatory effects among them at the transitions from neurogenic progenitors to postmitotic neuronal precursors in the Dm larval brain. The emerging picture of this analysis reveals an intricate regulatory network in which Mnb appears to play a pivotal role helping to delineate the dynamics of the expression patterns of Ase, Dpn and Pros, as well as their specific functions in the aforementioned transitions. Our results also show that Ase, Dpn and Pros perform several cross-regulatory actions and contribute to shape the precise cellular/temporal expression pattern of Mnb. We propose that Mnb/Dyrk1A plays a central role in CNS neurogenesis by integrating molecular mechanisms that regulate progenitor self-renewal, cell cycle progression and neuronal differentiation.
Collapse
Affiliation(s)
- Mirja N Shaikh
- a Instituto de Neurociencias , CSIC and Universidad Miguel Hernandez , Alicante , Spain
| | - Francisco J Tejedor
- a Instituto de Neurociencias , CSIC and Universidad Miguel Hernandez , Alicante , Spain
| |
Collapse
|
15
|
Yang Y, Shen W, Ni Y, Su Y, Yang Z, Zhao C. Impaired Interneuron Development after Foxg1 Disruption. Cereb Cortex 2018; 27:793-808. [PMID: 26620267 DOI: 10.1093/cercor/bhv297] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Interneurons play pivotal roles in the modulation of cortical function; however, the mechanisms that control interneuron development remain unclear. This study aimed to explore a new role for Foxg1 in interneuron development. By crossing Foxg1fl/fl mice with a Dlx5/6-Cre line, we determined that conditional disruption of Foxg1 in the subpallium results in defects in interneuron development. In developing interneurons, the expression levels of several receptors, including roundabout-1, Eph receptor A4, and C-X-C motif receptor 4/7, were strongly downregulated, which led to migration defects after Foxg1 ablation. The transcription factors Dlx1/2 and Mash1, which have been reported to be involved in interneuron development, were significantly upregulated at the mRNA levels. Foxg1 mutant cells developed shorter neurites and fewer branches and displayed severe migration defects in vitro. Notably, Prox1, which is a transcription factor that functions as a key regulator in the development of excitatory neurons, was also dramatically upregulated at both the mRNA and protein levels, suggesting that Prox1 is also important for interneuron development. Our work demonstrates that Foxg1 may act as a critical upstream regulator of Dlx1/2, Mash1, and Prox1 to control interneuron development. These findings will further our understanding of the molecular mechanisms of interneuron development.
Collapse
Affiliation(s)
- Ying Yang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wei Shen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yang Ni
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yan Su
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhengang Yang
- Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Anatomy and Neuroscience, School of Medicine, Southeast University, Nanjing 210009, China.,Center of Depression, Beijing Institute for Brain Disorders, Beijing 100069, China
| |
Collapse
|
16
|
Sur A, Magie CR, Seaver EC, Meyer NP. Spatiotemporal regulation of nervous system development in the annelid Capitella teleta. EvoDevo 2017; 8:13. [PMID: 28775832 PMCID: PMC5539756 DOI: 10.1186/s13227-017-0076-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/20/2017] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND How nervous systems evolved remains an unresolved question. Previous studies in vertebrates and arthropods revealed that homologous genes regulate important neurogenic processes such as cell proliferation and differentiation. However, the mechanisms through which such homologs regulate neurogenesis across different bilaterian clades are variable, making inferences about nervous system evolution difficult. A better understanding of neurogenesis in the third major bilaterian clade, Spiralia, would greatly contribute to our ability to deduce the ancestral mechanism of neurogenesis. RESULTS Using whole-mount in situ hybridization, we examined spatiotemporal gene expression for homologs of soxB, musashi, prospero, achaete-scute, neurogenin, and neuroD in embryos and larvae of the spiralian annelid Capitella teleta, which has a central nervous system (CNS) comprising a brain and ventral nerve cord. For all homologs examined, we found expression in the neuroectoderm and/or CNS during neurogenesis. Furthermore, the onset of expression and localization within the developing neural tissue for each of these genes indicates putative roles in separate phases of neurogenesis, e.g., in neural precursor cells (NPCs) versus in cells that have exited the cell cycle. Ct-soxB1, Ct-soxB, and Ct-ngn are the earliest genes expressed in surface cells in the anterior and ventral neuroectoderm, while Ct-ash1 expression initiates slightly later in surface neuroectoderm. Ct-pros is expressed in single cells in neural and non-neural ectoderm, while Ct-msi and Ct-neuroD are localized to differentiating neural cells in the brain and ventral nerve cord. CONCLUSIONS These results suggest that the genes investigated in this article are involved in a neurogenic gene regulatory network in C. teleta. We propose that Ct-SoxB1, Ct-SoxB, and Ct-Ngn are involved in maintaining NPCs in a proliferative state. Ct-Pros may function in division of NPCs, Ct-Ash1 may promote cell cycle exit and ingression of NPC daughter cells, and Ct-NeuroD and Ct-Msi may control neuronal differentiation. Our results support the idea of a common genetic toolkit driving neural development whose molecular architecture has been rearranged within and across clades during evolution. Future functional studies should help elucidate the role of these homologs during C. teleta neurogenesis and identify which aspects of bilaterian neurogenesis may have been ancestral or were derived within Spiralia.
Collapse
Affiliation(s)
- Abhinav Sur
- Biology Department, Clark University, 950 Main St., Worcester, MA 01610-1400 USA
| | - Craig R. Magie
- Department of Biological Sciences, Quinnipiac University, 275 Mount Carmel Ave., Hamden, CT 06518-1905 USA
| | - Elaine C. Seaver
- Whitney Laboratory for Marine Bioscience, 9505 Ocean Shore Blvd., St. Augustine, FL 32080-8610 USA
| | - Néva P. Meyer
- Biology Department, Clark University, 950 Main St., Worcester, MA 01610-1400 USA
| |
Collapse
|
17
|
Hayashi T, Uchida K, Mine Y, Yamada M, Kawase T. Feasibility of Using Early Mesencephalic Neural Plate for Intracerebral Grafting. Cell Transplant 2017. [DOI: 10.3727/000000002783985639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The purpose of this study was to elucidate the biological significance and the possibility of intracerebral grafting of neuroepithelial stem cells derived from the mesencephalic neural plate. Immunohistological studies of embryonic day 10.5 (E10.5) Wister rats revealed strong nestin expression in the mesencephalic part of the neural plate. Mesencephalic neural plates removed from E10.5 rats were processed to either tissue or cell dissociation culture. They were cultured in vitro under various conditions and were analyzed 7 days after the primary culture. When they were cultured as a tissue, cell proliferation and differentiation into neurons extending long neurites were obvious in a serum-free medium, in a medium containing 3% serum, and in a medium containing 20 ng/ml epidermal growth factor. On the other hand, in a medium containing 10 ng/ml basic fibroblast growth factor (bFGF), both vigorous cell proliferation and sphere formation were recognized. Furthermore, marked neurite growth was rarely seen in this culture. When they were plated in a dissociation culture, cell proliferation and neurosphere generation were also recognized only in a medium containing bFGF, depending on the initial cell concentration. The spheres, generated 7 days after the primary cell culture, were positively stained by nestin. These data suggested that bFGF was able to amplify the stem cell population present in the mesencephalic neural plate derived from early embryos. This might make it possible to obtain a large number of stem cells as donor material for neural transplantation on demand.
Collapse
Affiliation(s)
- Takuro Hayashi
- Department of Neurosurgery, School of Medicine, Keio University, Shinanomachi 35, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Koichi Uchida
- Department of Neurosurgery, School of Medicine, Keio University, Shinanomachi 35, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yutaka Mine
- Department of Neurosurgery, School of Medicine, Keio University, Shinanomachi 35, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Motoyuki Yamada
- Department of Neurosurgery, School of Medicine, Keio University, Shinanomachi 35, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takeshi Kawase
- Department of Neurosurgery, School of Medicine, Keio University, Shinanomachi 35, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
18
|
Nuclear receptor NR5A2 controls neural stem cell fate decisions during development. Nat Commun 2016; 7:12230. [PMID: 27447294 PMCID: PMC4961839 DOI: 10.1038/ncomms12230] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 06/14/2016] [Indexed: 02/08/2023] Open
Abstract
The enormous complexity of mammalian central nervous system (CNS) is generated by highly synchronized actions of diverse factors and signalling molecules in neural stem/progenitor cells (NSCs). However, the molecular mechanisms that integrate extrinsic and intrinsic signals to control proliferation versus differentiation decisions of NSCs are not well-understood. Here we identify nuclear receptor NR5A2 as a central node in these regulatory networks and key player in neural development. Overexpression and loss-of-function experiments in primary NSCs and mouse embryos suggest that NR5A2 synchronizes cell-cycle exit with induction of neurogenesis and inhibition of astrogliogenesis by direct regulatory effects on Ink4/Arf locus, Prox1, a downstream target of proneural genes, as well as Notch1 and JAK/STAT signalling pathways. Upstream of NR5a2, proneural genes, as well as Notch1 and JAK/STAT pathways control NR5a2 endogenous expression. Collectively, these observations render NR5A2 a critical regulator of neural development and target gene for NSC-based treatments of CNS-related diseases. The molecular signals regulating the decision of neural stem cells (NSC) to proliferate versus differentiate are unclear. Here, the authors identify the nuclear receptor NR5A2 as coordinating cell-cycle exit with differentiation of NSCs via direct actions on Ink4, Prox1, Notch1 and JAK/STAT.
Collapse
|
19
|
Götz M, Nakafuku M, Petrik D. Neurogenesis in the Developing and Adult Brain-Similarities and Key Differences. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a018853. [PMID: 27235475 DOI: 10.1101/cshperspect.a018853] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Adult neurogenesis in the mammalian brain is often viewed as a continuation of neurogenesis at earlier, developmental stages. Here, we will critically review the extent to which this is the case highlighting similarities as well as key differences. Although many transcriptional regulators are shared in neurogenesis at embryonic and adult stages, recent findings on the molecular mechanisms by which these neuronal fate determinants control fate acquisition and maintenance have revealed profound differences between development and adulthood. Importantly, adult neurogenesis occurs in a gliogenic environment, hence requiring adult-specific additional and unique mechanisms of neuronal fate specification and maintenance. Thus, a better understanding of the molecular logic for continuous adult neurogenesis provides important clues to develop strategies to manipulate endogenous stem cells for the purpose of repair.
Collapse
Affiliation(s)
- Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Munich, Germany Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, 80336 Munich, Germany Synergy, Munich Cluster for Systems Neurology, 81377 Munich, Germany
| | - Masato Nakafuku
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45140 Departments of Pediatrics and Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - David Petrik
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Neuherberg, Munich, Germany Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, 80336 Munich, Germany
| |
Collapse
|
20
|
Ishii J, Yazawa T, Chiba T, Shishido-Hara Y, Arimasu Y, Sato H, Kamma H. PROX1 Promotes Secretory Granule Formation in Medullary Thyroid Cancer Cells. Endocrinology 2016; 157:1289-98. [PMID: 26760117 DOI: 10.1210/en.2015-1973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mechanisms of endocrine secretory granule (SG) formation in thyroid C cells and medullary thyroid cancer (MTC) cells have not been fully elucidated. Here we directly demonstrated that PROX1, a developmental homeobox gene, is transcriptionally involved in SG formation in MTC, which is derived from C cells. Analyses using gene expression databases on web sites revealed that, among thyroid cancer cells, MTC cells specifically and highly express PROX1 as well as several SG-forming molecule genes. Immunohistochemical analyses showed that in vivo MTC and C cells expressed PROX1, although follicular thyroid cancer and papillary thyroid cancer cells, normal follicular cells did not. Knockdown of PROX1 in an MTC cells reduced SGs detected by electron microscopy, and decreased expression of SG-related genes (chromogranin A, chromogranin B, secretogranin II, secretogranin III, synaptophysin, and carboxypeptidase E). Conversely, the introduction of a PROX1 transgene into a papillary thyroid cancer and anaplastic thyroid cancer cells induced the expression of SG-related genes. Reporter assays using the promoter sequence of chromogranin A showed that PROX1 activates the chromogranin A gene in addition to the known regulatory mechanisms, which are mediated via the cAMP response element binding protein and the repressor element 1-silencing transcription factor. Furthermore, chromatin immunoprecipitation-PCR assays demonstrated that PROX1 binds to the transcriptional regulatory element of the chromogranin A gene. In conclusion, PROX1 is an important regulator of endocrine SG formation in MTC cells.
Collapse
Affiliation(s)
- Jun Ishii
- Department of Pathology (J.I., T.C., Y.A., H.K.), Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan; Department of Diagnostic Pathology (T.Y.), Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; Department of Anatomic Pathology (Y.S.-H.), Tokyo Medical University, Shinjuku, Tokyo 101-0062, Japan; and Department of Anatomy (H.S.), St Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | - Takuya Yazawa
- Department of Pathology (J.I., T.C., Y.A., H.K.), Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan; Department of Diagnostic Pathology (T.Y.), Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; Department of Anatomic Pathology (Y.S.-H.), Tokyo Medical University, Shinjuku, Tokyo 101-0062, Japan; and Department of Anatomy (H.S.), St Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | - Tomohiro Chiba
- Department of Pathology (J.I., T.C., Y.A., H.K.), Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan; Department of Diagnostic Pathology (T.Y.), Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; Department of Anatomic Pathology (Y.S.-H.), Tokyo Medical University, Shinjuku, Tokyo 101-0062, Japan; and Department of Anatomy (H.S.), St Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | - Yukiko Shishido-Hara
- Department of Pathology (J.I., T.C., Y.A., H.K.), Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan; Department of Diagnostic Pathology (T.Y.), Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; Department of Anatomic Pathology (Y.S.-H.), Tokyo Medical University, Shinjuku, Tokyo 101-0062, Japan; and Department of Anatomy (H.S.), St Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | - Yuu Arimasu
- Department of Pathology (J.I., T.C., Y.A., H.K.), Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan; Department of Diagnostic Pathology (T.Y.), Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; Department of Anatomic Pathology (Y.S.-H.), Tokyo Medical University, Shinjuku, Tokyo 101-0062, Japan; and Department of Anatomy (H.S.), St Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | - Hanako Sato
- Department of Pathology (J.I., T.C., Y.A., H.K.), Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan; Department of Diagnostic Pathology (T.Y.), Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; Department of Anatomic Pathology (Y.S.-H.), Tokyo Medical University, Shinjuku, Tokyo 101-0062, Japan; and Department of Anatomy (H.S.), St Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | - Hiroshi Kamma
- Department of Pathology (J.I., T.C., Y.A., H.K.), Kyorin University School of Medicine, Mitaka, Tokyo 181-8611, Japan; Department of Diagnostic Pathology (T.Y.), Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; Department of Anatomic Pathology (Y.S.-H.), Tokyo Medical University, Shinjuku, Tokyo 101-0062, Japan; and Department of Anatomy (H.S.), St Marianna University School of Medicine, Kanagawa 216-8511, Japan
| |
Collapse
|
21
|
Shaikh MN, Gutierrez-Aviño F, Colonques J, Ceron J, Hämmerle B, Tejedor FJ. Minibrain drives the Dacapo dependent cell cycle exit of neurons in the Drosophila brain by promoting asense and prospero expression. Development 2016; 143:3195-205. [DOI: 10.1242/dev.134338] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 07/25/2016] [Indexed: 01/20/2023]
Abstract
A key issue in neurodevelopment is to understand how precursor cells decide to stop dividing and commence their terminal differentiation at the correct time and place. Here, we show that minibrain (mnb), the Drosophila ortholog of the Down syndrome candidate gene MNB/DYRK1A, is transiently expressed in newborn neuronal precursors known as ganglion cells (GCs). Mnb promotes the cell cycle exit of GCs through a dual mechanism that regulates the expression of the cyclin-dependent kinase inhibitor Dacapo, the homolog of vertebrate p27kip1. On the one hand, Mnb upregulates the expression of the proneural transcription factor (TF) Asense, which promotes Dacapo expression. On the other, Mnb induces the expression of Prospero, a homeodomain TF that in turn inhibits the expression of Deadpan, a pan-neural TF that represses dacapo. In addition to its effects on Asense and Prospero, Mnb also promotes the expression of the neuronal-specific RNA regulator Elav, strongly suggesting that Mnb facilitates neuronal differentiation. These actions of Mnb ensure the precise timing of neuronal birth, coupling the mechanisms that regulate neurogenesis, cell cycle control and terminal differentiation of neurons.
Collapse
Affiliation(s)
- Mirja N. Shaikh
- Instituto de Neurociencias, CSIC and Universidad Miguel Hernandez, Alicante, Spain
| | | | - Jordi Colonques
- Instituto de Neurociencias, CSIC and Universidad Miguel Hernandez, Alicante, Spain
| | - Julian Ceron
- Instituto de Neurociencias, CSIC and Universidad Miguel Hernandez, Alicante, Spain
| | - Barbara Hämmerle
- Instituto de Neurociencias, CSIC and Universidad Miguel Hernandez, Alicante, Spain
| | - Francisco J. Tejedor
- Instituto de Neurociencias, CSIC and Universidad Miguel Hernandez, Alicante, Spain
| |
Collapse
|
22
|
Holzmann J, Hennchen M, Rohrer H. Prox1 identifies proliferating neuroblasts and nascent neurons during neurogenesis in sympathetic ganglia. Dev Neurobiol 2015; 75:1352-67. [PMID: 25788138 DOI: 10.1002/dneu.22289] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/12/2015] [Indexed: 12/28/2022]
Abstract
Neurogenesis in embryonic sympathetic ganglia involves neuroblasts that resume proliferation following neuronal differentiation. As cell cycle exit is not associated with neuronal differentiation, the identity of proliferating neuroblasts is incompletely understood. Here, we use sympathetic ganglia of chick embryos to define the timing of neurogenesis and neuroblast identity focusing on the expression and function of the transcription factor Prox1. We show that a large fraction of neuroblasts has initially withdrawn from the cell cycle at embryonic day 3 (E3), which is reflected by a high proportion of p27(+)/Islet1(+) neuroblasts (63%) and low numbers of EdU(+)/Islet1(+) cells (12%). The proportion of proliferating Islet1(+) neuroblasts, identified by EdU pulse labeling and by the absence of the postmitotic marker p27 increases to reach maximal levels at E5, when virtually all neuroblasts are in the cell cycle (95%). Subsequently, the proportion of EdU-labeled and p27(-) neuroblasts is reduced to reach low levels at E11. Interestingly, the expression of the transcription factor Prox1 is restricted to the neuronal lineage, that is, Sox10(+)/Phox2b(+) neuron progenitors, proliferating p27(-)/Islet1(+) neuroblasts and nascent neurons but is rapidly lost in postmitotic neurons. In vitro and in vivo knockdown and overexpression experiments demonstrate effects of Prox1 in the support of neuroblast proliferation and survival. Taken together, these results define the neurogenesis period in the chick paravertebral sympathetic ganglia including an initial cell cycle withdrawal and identify Prox1 as a marker and regulator of proliferating sympathetic neuroblasts.
Collapse
Affiliation(s)
- Julia Holzmann
- Max-Planck-Institute for Brain Research; Research Group Developmental Neurobiology, Max-von-Laue-Str. 4, 60438 Frankfurt/Main, Germany
| | - Melanie Hennchen
- Max-Planck-Institute for Brain Research; Research Group Developmental Neurobiology, Max-von-Laue-Str. 4, 60438 Frankfurt/Main, Germany
| | - Hermann Rohrer
- Max-Planck-Institute for Brain Research; Research Group Developmental Neurobiology, Max-von-Laue-Str. 4, 60438 Frankfurt/Main, Germany.,Institute of Clinical Neuroanatomy, Goethe-University Frankfurt, Theodor-Stern-Kai 7, Frankfurt/Main, Germany
| |
Collapse
|
23
|
Niu W, Zang T, Smith DK, Vue TY, Zou Y, Bachoo R, Johnson JE, Zhang CL. SOX2 reprograms resident astrocytes into neural progenitors in the adult brain. Stem Cell Reports 2015; 4:780-94. [PMID: 25921813 PMCID: PMC4437485 DOI: 10.1016/j.stemcr.2015.03.006] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/24/2015] [Accepted: 03/26/2015] [Indexed: 01/14/2023] Open
Abstract
Glial cells can be in vivo reprogrammed into functional neurons in the adult CNS; however, the process by which this reprogramming occurs is unclear. Here, we show that a distinct cellular sequence is involved in SOX2-driven in situ conversion of adult astrocytes to neurons. This includes ASCL1+ neural progenitors and DCX+ adult neuroblasts (iANBs) as intermediates. Importantly, ASCL1 is required, but not sufficient, for the robust generation of iANBs in the adult striatum. These progenitor-derived iANBs predominantly give rise to calretinin+ interneurons when supplied with neurotrophic factors or the small-molecule valproic acid. Patch-clamp recordings from the induced neurons reveal subtype heterogeneity, though all are functionally mature, fire repetitive action potentials, and receive synaptic inputs. Together, these results show that SOX2-mediated in vivo reprogramming of astrocytes to neurons passes through proliferative intermediate progenitors, which may be exploited for regenerative medicine. SOX2 induces ASCL1-positive neural progenitors in the adult mouse brain Ascl1 in resident astrocytes is required for SOX2-mediated in vivo reprogramming Induced ASCL1-positive neural progenitors generate mature calretinin neurons
Collapse
Affiliation(s)
- Wenze Niu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Tong Zang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Derek K Smith
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Tou Yia Vue
- Department of Neuroscience, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Yuhua Zou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Robert Bachoo
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jane E Johnson
- Department of Neuroscience, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
24
|
Stergiopoulos A, Elkouris M, Politis PK. Prospero-related homeobox 1 (Prox1) at the crossroads of diverse pathways during adult neural fate specification. Front Cell Neurosci 2015; 8:454. [PMID: 25674048 PMCID: PMC4306308 DOI: 10.3389/fncel.2014.00454] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/16/2014] [Indexed: 12/11/2022] Open
Abstract
Over the last decades, adult neurogenesis in the central nervous system (CNS) has emerged as a fundamental process underlying physiology and disease. Recent evidence indicates that the homeobox transcription factor Prox1 is a critical intrinsic regulator of neurogenesis in the embryonic CNS and adult dentate gyrus (DG) of the hippocampus, acting in multiple ways and instructed by extrinsic cues and intrinsic factors. In the embryonic CNS, Prox1 is mechanistically involved in the regulation of proliferation vs. differentiation decisions of neural stem cells (NSCs), promoting cell cycle exit and neuronal differentiation, while inhibiting astrogliogenesis. During the complex differentiation events in adult hippocampal neurogenesis, Prox1 is required for maintenance of intermediate progenitors (IPs), differentiation and maturation of glutamatergic interneurons, as well as specification of DG cell identity over CA3 pyramidal fate. The mechanism by which Prox1 exerts multiple functions involves distinct signaling pathways currently not fully highlighted. In this mini-review, we thoroughly discuss the Prox1-dependent phenotypes and molecular pathways in adult neurogenesis in relation to different upstream signaling cues and cell fate determinants. In addition, we discuss the possibility that Prox1 may act as a cross-talk point between diverse signaling cascades to achieve specific outcomes during adult neurogenesis.
Collapse
Affiliation(s)
- Athanasios Stergiopoulos
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece
| | - Maximilianos Elkouris
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens Athens, Greece
| |
Collapse
|
25
|
Vue TY, Kim EJ, Parras CM, Guillemot F, Johnson JE. Ascl1 controls the number and distribution of astrocytes and oligodendrocytes in the gray matter and white matter of the spinal cord. Development 2014; 141:3721-31. [PMID: 25249462 DOI: 10.1242/dev.105270] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Glia constitute the majority of cells in the mammalian central nervous system and are crucial for neurological function. However, there is an incomplete understanding of the molecular control of glial cell development. We find that the transcription factor Ascl1 (Mash1), which is best known for its role in neurogenesis, also functions in both astrocyte and oligodendrocyte lineages arising in the mouse spinal cord at late embryonic stages. Clonal fate mapping in vivo reveals heterogeneity in Ascl1-expressing glial progenitors and shows that Ascl1 defines cells that are restricted to either gray matter (GM) or white matter (WM) as astrocytes or oligodendrocytes. Conditional deletion of Ascl1 post-neurogenesis shows that Ascl1 is required during oligodendrogenesis for generating the correct numbers of WM but not GM oligodendrocyte precursor cells, whereas during astrocytogenesis Ascl1 functions in balancing the number of dorsal GM protoplasmic astrocytes with dorsal WM fibrous astrocytes. Thus, in addition to its function in neurogenesis, Ascl1 marks glial progenitors and controls the number and distribution of astrocytes and oligodendrocytes in the GM and WM of the spinal cord.
Collapse
Affiliation(s)
- Tou Yia Vue
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Euiseok J Kim
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carlos M Parras
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, London NW7 1AA, UK
| | - Francois Guillemot
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, London NW7 1AA, UK
| | - Jane E Johnson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
26
|
Kasim M, Benko E, Winkelmann A, Mrowka R, Staudacher JJ, Persson PB, Scholz H, Meier JC, Fähling M. Shutdown of achaete-scute homolog-1 expression by heterogeneous nuclear ribonucleoprotein (hnRNP)-A2/B1 in hypoxia. J Biol Chem 2014; 289:26973-26988. [PMID: 25124043 DOI: 10.1074/jbc.m114.579391] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The basic helix-loop-helix transcription factor hASH1, encoded by the ASCL1 gene, plays an important role in neurogenesis and tumor development. Recent findings indicate that local oxygen tension is a critical determinant for the progression of neuroblastomas. Here we investigated the molecular mechanisms underlying the oxygen-dependent expression of hASH1 in neuroblastoma cells. Exposure of human neuroblastoma-derived Kelly cells to 1% O2 significantly decreased ASCL1 mRNA and hASH1 protein levels. Using reporter gene assays, we show that the response of hASH1 to hypoxia is mediated mainly by post-transcriptional inhibition via the ASCL1 mRNA 5'- and 3'-UTRs, whereas additional inhibition of the ASCL1 promoter was observed under prolonged hypoxia. By RNA pulldown experiments followed by MALDI/TOF-MS analysis, we identified heterogeneous nuclear ribonucleoprotein (hnRNP)-A2/B1 and hnRNP-R as interactors binding directly to the ASCL1 mRNA 5'- and 3'-UTRs and influencing its expression. We further demonstrate that hnRNP-A2/B1 is a key positive regulator of ASCL1, findings that were also confirmed by analysis of a large compilation of gene expression data. Our data suggest that a prominent down-regulation of hnRNP-A2/B1 during hypoxia is associated with the post-transcriptional suppression of hASH1 synthesis. This novel post-transcriptional mechanism for regulating hASH1 levels will have important implications in neural cell fate development and disease.
Collapse
Affiliation(s)
- Mumtaz Kasim
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, D-10117 Berlin
| | - Edgar Benko
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, D-10117 Berlin
| | - Aline Winkelmann
- RNA Editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, D-13125 Berlin, and
| | - Ralf Mrowka
- Klinik für Innere Medizin III, AG Experimentelle Nephrologie, Universitätsklinikum Jena, D-07743 Jena, Germany
| | - Jonas J Staudacher
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, D-10117 Berlin
| | - Pontus B Persson
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, D-10117 Berlin
| | - Holger Scholz
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, D-10117 Berlin
| | - Jochen C Meier
- RNA Editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, D-13125 Berlin, and
| | - Michael Fähling
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, D-10117 Berlin,.
| |
Collapse
|
27
|
Abstract
Granule neurons in the hippocampal dentate gyrus (DG) receive their primary inputs from the cortex and are known to be continuously generated throughout adult life. Ongoing integration of newborn neurons into the existing hippocampal neural circuitry provides enhanced neuroplasticity, which plays a crucial role in learning and memory; deficits in this process have been associated with cognitive decline under neuropathological conditions. In this Primer, we summarize the developmental principles that regulate the process of DG neurogenesis and discuss recent advances in harnessing these developmental cues to generate DG granule neurons from human pluripotent stem cells.
Collapse
Affiliation(s)
- Diana X Yu
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Maria C Marchetto
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Fred H Gage
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
28
|
Yasugi T, Fischer A, Jiang Y, Reichert H, Knoblich JA. A regulatory transcriptional loop controls proliferation and differentiation in Drosophila neural stem cells. PLoS One 2014; 9:e97034. [PMID: 24804774 PMCID: PMC4013126 DOI: 10.1371/journal.pone.0097034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/14/2014] [Indexed: 12/04/2022] Open
Abstract
Neurogenesis is initiated by a set of basic Helix-Loop-Helix (bHLH) transcription factors that specify neural progenitors and allow them to generate neurons in multiple rounds of asymmetric cell division. The Drosophila Daughterless (Da) protein and its mammalian counterparts (E12/E47) act as heterodimerization factors for proneural genes and are therefore critically required for neurogenesis. Here, we demonstrate that Da can also be an inhibitor of the neural progenitor fate whose absence leads to stem cell overproliferation and tumor formation. We explain this paradox by demonstrating that Da induces the differentiation factor Prospero (Pros) whose asymmetric segregation is essential for differentiation in one of the two daughter cells. Da co-operates with the bHLH transcription factor Asense, whereas the other proneural genes are dispensible. After mitosis, Pros terminates Asense expression in one of the two daughter cells. In da mutants, pros is not expressed, leading to the formation of lethal transplantable brain tumors. Our results define a transcriptional feedback loop that regulates the balance between self-renewal and differentiation in Drosophila optic lobe neuroblasts. They indicate that initiation of a neural differentiation program in stem cells is essential to prevent tumorigenesis.
Collapse
Affiliation(s)
- Tetsuo Yasugi
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Anja Fischer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
| | - Yanrui Jiang
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Juergen A. Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
- * E-mail:
| |
Collapse
|
29
|
Kameda Y. Signaling molecules and transcription factors involved in the development of the sympathetic nervous system, with special emphasis on the superior cervical ganglion. Cell Tissue Res 2014; 357:527-48. [PMID: 24770894 DOI: 10.1007/s00441-014-1847-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 02/12/2014] [Indexed: 12/16/2022]
Abstract
The cells that constitute the sympathetic nervous system originate from the neural crest. This review addresses the current understanding of sympathetic ganglion development viewed from molecular and morphological perspectives. Development of the sympathetic nervous system is categorized into three main steps, as follows: (1) differentiation and migration of cells in the neural crest lineage for formation of the primary sympathetic chain, (2) differentiation of sympathetic progenitors, and (3) growth and survival of sympathetic ganglia. The signaling molecules and transcription factors involved in each of these developmental stages are elaborated mostly on the basis of the results of targeted mutation of respective genes. Analyses in mutant mice revealed differences between the superior cervical ganglion (SCG) and the other posterior sympathetic ganglia. This review provides a summary of the similarities and differences in the development of the SCG and other posterior sympathetic ganglia. Relevant to the development of sympathetic ganglia is the demonstration that neuroendocrine cells, such as adrenal chromaffin cells and carotid body glomus cells, share a common origin with the sympathetic ganglia. Neural crest cells at the trunk level give rise to common sympathoadrenal progenitors of sympathetic neurons and chromaffin cells, while progenitors segregated from the SCG give rise to glomus cells. After separation from the sympathetic primordium, the progenitors of both chromaffin cells and glomus cells colonize the anlage of the adrenal gland and carotid body, respectively. This review highlights the biological properties of chromaffin cells and glomus cells, because, although both cell types are derivatives of sympathetic primordium, they are distinct in many respects.
Collapse
Affiliation(s)
- Yoko Kameda
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan,
| |
Collapse
|
30
|
Markitantova YV, Zinovieva RD. Intracellular localization of transcription factor PROX1 in the human retina in ontogeny. BIOL BULL+ 2014. [DOI: 10.1134/s106235901402006x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
31
|
Hirayama T, Asano Y, Iida H, Watanabe T, Nakamura T, Goitsuka R. Meis1 is required for the maintenance of postnatal thymic epithelial cells. PLoS One 2014; 9:e89885. [PMID: 24594519 PMCID: PMC3942356 DOI: 10.1371/journal.pone.0089885] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 01/24/2014] [Indexed: 12/19/2022] Open
Abstract
Most epithelial tissues retain stem/progenitor cells to maintain homeostasis of the adult tissues; however, the existence of a thymic epithelial cell (TEC) progenitor capable of maintaining homeostasis of the postnatal thymus remains unclear. Here, we show that a cell population expressing high levels of Meis1, a homeodomain transcription factor, is enriched in TECs with an immature cellular phenotype. These TECs selectively express genes involved in embryonic thymic organogenesis and epithelial stem cell maintenance, and also have the potential to proliferate and differentiate into mature TEC populations. Furthermore, postnatal inactivation of Meis1 in TECs caused disorganization of the thymic architecture, which ultimately leads to premature disappearance of the thymus. There was an age-associated reduction in the proportion of the TEC population expressing high levels of Meis1, which may also be related to thymic involution. These findings indicate that Meis1 is potentially involved in the maintenance of postnatal TECs with progenitor activity that is required for homeostasis of the postnatal thymus.
Collapse
Affiliation(s)
- Takehiro Hirayama
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yusuke Asano
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Hajime Iida
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Takeshi Watanabe
- Center for Innovation in Immunoregulative Technology and Therapeutics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takuro Nakamura
- Department of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryo Goitsuka
- Division of Development and Aging, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
- * E-mail:
| |
Collapse
|
32
|
Yu DX, Di Giorgio FP, Yao J, Marchetto MC, Brennand K, Wright R, Mei A, McHenry L, Lisuk D, Grasmick JM, Silberman P, Silberman G, Jappelli R, Gage FH. Modeling hippocampal neurogenesis using human pluripotent stem cells. Stem Cell Reports 2014; 2:295-310. [PMID: 24672753 PMCID: PMC3964286 DOI: 10.1016/j.stemcr.2014.01.009] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 01/17/2014] [Accepted: 01/20/2014] [Indexed: 02/07/2023] Open
Abstract
The availability of human pluripotent stem cells (hPSCs) offers the opportunity to generate lineage-specific cells to investigate mechanisms of human diseases specific to brain regions. Here, we report a differentiation paradigm for hPSCs that enriches for hippocampal dentate gyrus (DG) granule neurons. This differentiation paradigm recapitulates the expression patterns of key developmental genes during hippocampal neurogenesis, exhibits characteristics of neuronal network maturation, and produces PROX1+ neurons that functionally integrate into the DG. Because hippocampal neurogenesis has been implicated in schizophrenia (SCZD), we applied our protocol to SCZD patient-derived human induced pluripotent stem cells (hiPSCs). We found deficits in the generation of DG granule neurons from SCZD hiPSC-derived hippocampal NPCs with lowered levels of NEUROD1, PROX1, and TBR1, reduced neuronal activity, and reduced levels of spontaneous neurotransmitter release. Our approach offers important insights into the neurodevelopmental aspects of SCZD and may be a promising tool for drug screening and personalized medicine. Hippocampal neurogenesis is modeled using human pluripotent stem cells Differentiated DG neurons are detected using lentiviral PROX1-GFP reporter construct Differentiated granule neurons functionally integrate into the dentate gyrus in vivo SCZD hiPSC-derived hippocampal NPCs present deficits in hippocampal neurogenesis
Collapse
Affiliation(s)
- Diana Xuan Yu
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Francesco Paolo Di Giorgio
- Neuroscience Discovery, Novartis Pharma AG, Novartis Institute for Biomedical Research, Postfach, Basel CH-4002, Switzerland
| | - Jun Yao
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Maria Carolina Marchetto
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Kristen Brennand
- Department of Neuroscience/Psychiatry, Mount Sinai School of Medicine, 1425 Madison Ave, New York, NY 10059, USA
| | - Rebecca Wright
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Arianna Mei
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Lauren McHenry
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - David Lisuk
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jaeson Michael Grasmick
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Pedro Silberman
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Giovanna Silberman
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Roberto Jappelli
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Fred H Gage
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
33
|
Cunningham D, Casey ES. Spatiotemporal development of the embryonic nervous system of Saccoglossus kowalevskii. Dev Biol 2013; 386:252-63. [PMID: 24333176 DOI: 10.1016/j.ydbio.2013.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 11/21/2013] [Accepted: 12/03/2013] [Indexed: 11/16/2022]
Abstract
Defining the organization and temporal onset of key steps in neurogenesis in invertebrate deuterostomes is critical to understand the evolution of the bilaterian and deuterostome nervous systems. Although recent studies have revealed the organization of the nervous system in adult hemichordates, little attention has been paid to neurogenesis during embryonic development in this third major phylum of deuterostomes. We examine the early events of neural development in the enteropneust hemichordate Saccoglossus kowalevskii by analyzing the expression of 11 orthologs of key genes associated with neurogenesis in an expansive range of bilaterians. Using in situ hybridization (ISH) and RT-PCR, we follow the course of neural development to track the transition of the early embryonic diffuse nervous system to the more regionalized midline nervous system of the adult. We show that in Saccoglossus, neural progenitor markers are expressed maternally and broadly encircle the developing embryo. An increase in their expression and the onset of pan neural markers, indicate that neural specification occurs in late blastulae - early gastrulae. By mid-gastrulation, punctate expression of markers of differentiating neurons encircling the embryo indicate the presence of immature neurons, and at the end of gastrulation when the embryo begins to elongate, markers of mature neurons are expressed. At this stage, expression of a subset of neuronal markers is concentrated along the trunk ventral and dorsal midlines. These data indicate that the diffuse embryonic nervous system of Saccoglossus is transient and quickly reorganizes before hatching to resemble the adult regionalized, centralized nervous system. This regionalization occurs at a much earlier developmental stage than anticipated indicating that centralization is not linked in S. kowalevskii to a lifestyle change of a swimming larva metamorphosing to a crawling worm-like adult.
Collapse
Affiliation(s)
- Doreen Cunningham
- Department of Biology, Georgetown University, Washington DC 20057, United States.
| | - Elena Silva Casey
- Department of Biology, Georgetown University, Washington DC 20057, United States
| |
Collapse
|
34
|
Chaitanya GV, Omura S, Sato F, Martinez NE, Minagar A, Ramanathan M, Guttman BW, Zivadinov R, Tsunoda I, Alexander JS. Inflammation induces neuro-lymphatic protein expression in multiple sclerosis brain neurovasculature. J Neuroinflammation 2013; 10:125. [PMID: 24124909 PMCID: PMC3854084 DOI: 10.1186/1742-2094-10-125] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/24/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is associated with ectopic lymphoid follicle formation. Podoplanin+ (lymphatic marker) T helper17 (Th17) cells and B cell aggregates have been implicated in the formation of tertiary lymphoid organs (TLOs) in MS and experimental autoimmune encephalitis (EAE). Since podoplanin expressed by Th17 cells in MS brains is also expressed by lymphatic endothelium, we investigated whether the pathophysiology of MS involves inductions of lymphatic proteins in the inflamed neurovasculature. METHODS We assessed the protein levels of lymphatic vessel endothelial hyaluronan receptor and podoplanin, which are specific to the lymphatic system and prospero-homeobox protein-1, angiopoietin-2, vascular endothelial growth factor-D, vascular endothelial growth factor receptor-3, which are expressed by both lymphatic endothelium and neurons. Levels of these proteins were measured in postmortem brains and sera from MS patients, in the myelin proteolipid protein (PLP)-induced EAE and Theiler's murine encephalomyelitis virus (TMEV) induced demyelinating disease (TMEV-IDD) mouse models and in cell culture models of inflamed neurovasculature. RESULTS AND CONCLUSIONS Intense staining for LYVE-1 was found in neurons of a subset of MS patients using immunohistochemical approaches. The lymphatic protein, podoplanin, was highly expressed in perivascular inflammatory lesions indicating signaling cross-talks between inflamed brain vasculature and lymphatic proteins in MS. The profiles of these proteins in MS patient sera discriminated between relapsing remitting MS from secondary progressive MS and normal patients. The in vivo findings were confirmed in the in vitro cell culture models of neuroinflammation.
Collapse
Affiliation(s)
- Ganta Vijay Chaitanya
- Department of Molecular & Cellular Physiology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Seiichi Omura
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Fumitaka Sato
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Nicholas E Martinez
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Alireza Minagar
- Department of Neurology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Murali Ramanathan
- Department of Neurology, State University of New York, Buffalo, NY, USA
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY, USA
| | | | - Robert Zivadinov
- Department of Neurology, State University of New York, Buffalo, NY, USA
- Buffalo Neuroimaging Analysis Center, State University of New York, Buffalo, NY, USA
| | - Ikuo Tsunoda
- Department of Microbiology and Immunology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| | - Jonathan S Alexander
- Department of Molecular & Cellular Physiology, School of Medicine, Louisiana State University Health-Shreveport, 1501 Kings Highway, Shreveport, LA, 71130, USA
| |
Collapse
|
35
|
PROX1: a lineage tracer for cortical interneurons originating in the lateral/caudal ganglionic eminence and preoptic area. PLoS One 2013; 8:e77339. [PMID: 24155945 PMCID: PMC3796451 DOI: 10.1371/journal.pone.0077339] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 08/31/2013] [Indexed: 11/19/2022] Open
Abstract
The homeobox-encoding gene Prox1 and its Drosophila homologue prospero are key regulators of cell fate-specification. In the developing rodent cortex a sparse population of cells thought to correspond to late-generated cortical pyramidal neuron precursors expresses PROX1. Using a series of transgenic mice that mark cell lineages in the subcortical telencephalon and, more specifically, different populations of cortical interneurons, we demonstrate that neurons expressing PROX1 do not represent pyramidal neurons or their precursors but are instead subsets of cortical interneurons. These correspond to interneurons originating in the lateral/caudal ganglionic eminence (LGE/CGE) and a small number of preoptic area (POA)-derived neurons. Expression within the cortex can be detected from late embryonic stages onwards when cortical interneurons are still migrating. There is persistent expression in postmitotic cells in the mature brain mainly in the outer cortical layers. PROX1(+ve) interneurons express neurochemical markers such as calretinin, neuropeptide Y, reelin and vasoactive intestinal peptide, all of which are enriched in LGE/CGE- and some POA-derived cells. Unlike in the cortex, in the striatum PROX1 marks nearly all interneurons regardless of their origin. Weak expression of PROX1 can also be detected in oligodendrocyte lineage cells throughout the forebrain. Our data show that PROX1 can be used as a genetic lineage tracer of nearly all LGE/CGE- and subsets POA-derived cortical interneurons at all developmental and postnatal stages in vivo.
Collapse
|
36
|
Elsir T, Smits A, Lindström MS, Nistér M. Transcription factor PROX1: its role in development and cancer. Cancer Metastasis Rev 2013; 31:793-805. [PMID: 22733308 DOI: 10.1007/s10555-012-9390-8] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The homeobox gene PROX1 is critical for organ development during embryogenesis. The Drosophila homologue, known as prospero has been shown to act as a tumor suppressor by controlling asymmetric cell division of neuroblasts. Likewise, alterations in PROX1 expression and function are associated with a number of human cancers including hematological malignancies, carcinomas of the pancreas, liver and the biliary system, sporadic breast cancer, Kaposiform hemangioendothelioma, colon cancer, and brain tumors. PROX1 is involved in cancer development and progression and has been ascribed both tumor suppressive and oncogenic properties in a variety of different cancer types. However, the exact mechanisms through which PROX1 regulates proliferation, migration, and invasion of cancer cells are by large unknown. This review provides an update on the role of PROX1 in organ development and on its emerging functions in cancer, with special emphasis on the central nervous system and glial brain tumors.
Collapse
Affiliation(s)
- Tamador Elsir
- Department of Oncology-Pathology, Karolinska Institutet, CCK R8:05, Karolinska University Hospital, 17176 Stockholm, Sweden.
| | | | | | | |
Collapse
|
37
|
Takakuwa O, Maeno K, Kunii E, Ozasa H, Hijikata H, Uemura T, Kasai D, Ohkubo H, Miyazaki M, Oguri T, Niimi A. Involvement of intermediate filament nestin in cell growth of small-cell lung cancer. Lung Cancer 2013; 81:174-9. [PMID: 23706418 DOI: 10.1016/j.lungcan.2013.04.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 04/04/2013] [Accepted: 04/27/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND Nestin is a class VI intermediate filament protein expressed in stem/progenitor cells during the development of the central nervous system. Nestin is detected in various types of tumors and is involved in malignant processes. This study investigated the expression and function of nestin in small-cell lung cancer (SCLC). METHODS Expression of nestin and achaete-scute homolog 1 (ASH1) was studied in 21 lung cancer cell lines. To assess the function of nestin, a short hairpin RNA (shRNA) targeting nestin was transfected into two SCLC cell lines (DMS53 and SBC3), and cloned cells that showed apparent down-regulation of nestin were obtained. Nestin expression was also studied immunohistochemically in surgically resected SCLC primary tumors and metastatic SCLC tumors obtained from autopsy cases. RESULT Nestin was expressed in nine of 10 SCLC cell lines. The nestin expression level was significantly higher in SCLC cell lines than in NSCLC cell lines (P < 0.01). There was a statistically significant positive correlation between the expression levels of nestin and ASH1 in SCLC cell lines. Nestin knock-down cells created by transfection with shRNA exhibited decreased invasion and cell proliferation capabilities. Furthermore, nestin was detected in SCLC tumor cells and tumor vessels in all clinical tumor specimens. CONCLUSION Nestin is expressed in SCLC in association with neuroendocrine features and participates in malignant phenotypes, including cell growth. Therefore, nestin may be a novel therapeutic target for SCLC.
Collapse
Affiliation(s)
- Osamu Takakuwa
- Department of Medical Oncology and Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sousa-Nunes R, Somers WG. Mechanisms of asymmetric progenitor divisions in the Drosophila central nervous system. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:79-102. [PMID: 23696353 DOI: 10.1007/978-94-007-6621-1_6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Drosophila central nervous system develops from polarised asymmetric divisions of precursor cells, called neuroblasts. Decades of research on neuroblasts have resulted in a substantial understanding of the factors and molecular events responsible for fate decisions of neuroblasts and their progeny. Furthermore, the cell-cycle dependent mechanisms responsible for asymmetric cortical protein localisation, resulting in the unequal partitioning between daughters, are beginning to be exposed. Disruption to the appropriate partitioning of proteins between neuroblasts and differentiation-committed daughters can lead to supernumerary neuroblast-like cells and the formation of tumours. Many of the factors responsible for regulating asymmetric division of Drosophila neuroblasts are evolutionarily conserved and, in many cases, have been shown to play a functionally conserved role in mammalian neurogenesis. Recent genome-wide studies coupled with advancements in live-imaging technologies have opened further avenues of research into neuroblast biology. We review our current understanding of the molecular mechanisms regulating neuroblast divisions, a powerful system to model mammalian neurogenesis and tumourigenesis.
Collapse
Affiliation(s)
- Rita Sousa-Nunes
- MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, London, SE1 1UL, UK.
| | | |
Collapse
|
39
|
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
|
40
|
Ungerer P, Eriksson BJ, Stollewerk A. Unravelling the evolution of neural stem cells in arthropods: notch signalling in neural stem cell development in the crustacean Daphnia magna. Dev Biol 2012; 371:302-11. [PMID: 22964415 DOI: 10.1016/j.ydbio.2012.08.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 08/14/2012] [Accepted: 08/23/2012] [Indexed: 01/13/2023]
Abstract
The genetic regulatory networks controlling major developmental processes seem to be conserved in bilaterians regardless of an independent or a common origin of the structures. This has been explained by the employment of a genetic toolkit that was repeatedly used during bilaterian evolution to build the various forms and body plans. However, it is not clear how genetic networks were incorporated into the formation of novel structures and how homologous genes can regulate the disparate morphological processes. Here we address this question by analysing the role of Notch signalling, which is part of the bilaterian toolkit, in neural stem cell evolution in arthropods. Within arthropods neural stem cells have evolved in the last common ancestor of insects and crustaceans (Tetraconata). We analyse here for the first time the role of Notch signalling in a crustacean, the branchiopod Daphnia magna, and show that it is required in neural stem cells for regulating the time of neural precursor production and for binary cell fate decisions in the ventral neuroectoderm. The function of Notch signalling has diverged in the ventral neuroectoderm of insects and crustaceans accompanied by changes in the morphogenetic processes. In the crustacean, Notch controlled mechanisms of neuroblast regulation have evolved that are surprisingly similar to vertebrates and thus present a remarkable case of parallel evolution. These new data on a representative of crustaceans complete the arthropod data set on Notch signalling in the nervous system and allow for reconstructing how the Notch signalling pathway has been co-opted from pre-existing structures to the development of the evolving neural stem cells in the Tetraconata ancestor.
Collapse
Affiliation(s)
- Petra Ungerer
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, UK.
| | | | | |
Collapse
|
41
|
Kang SS, Keasey MP, Arnold SA, Reid R, Geralds J, Hagg T. Endogenous CNTF mediates stroke-induced adult CNS neurogenesis in mice. Neurobiol Dis 2012; 49:68-78. [PMID: 22960105 DOI: 10.1016/j.nbd.2012.08.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/16/2012] [Accepted: 08/22/2012] [Indexed: 11/24/2022] Open
Abstract
Focal brain ischemia in adult rats rapidly and robustly induces neurogenesis in the subventricular zone (SVZ) but there are few and inconsistent reports in mice, presenting a hurdle to genetically investigate the endogenous neurogenic regulators such as ciliary neurotrophic factor (CNTF). Here, we first provide a platform for further studies by showing that middle cerebral artery occlusion in adult male C57BL/6 mice robustly enhances neurogenesis in the SVZ only under very specific conditions, i.e., 14days after a 30min occlusion. CNTF expression paralleled changes in the number of proliferated, BrdU-positive, SVZ cells. Stroke-induced proliferation was absent in CNTF-/- mice, suggesting that it is mediated by CNTF. MCAO-increased CNTF appears to act on C cell proliferation and by inducing FGF2 expression but not via EGF expression or Notch1 signaling of neural stem cells in the SVZ. CNTF is unique, as expression of other gp130 ligands, IL-6 and LIF, did not predict SVZ proliferation or showed no or only small compensatory increases in CNTF-/- mice. Expression of tumor necrosis factor-α, which can inhibit neurogenesis, and the presence of leukocytes in the SVZ were inversely correlated with neurogenesis, but pro-inflammatory cytokines did not affect CNTF expression in cultured astrocytes. These results suggest that slowly up-regulated CNTF in the SVZ mediates stroke-induced neurogenesis and is counteracted by inflammation. Further pharmacological stimulation of endogenous CNTF might be a good therapeutic strategy for cell replacement after stroke as CNTF regulates normal patterns of neurogenesis and is expressed almost exclusively in the nervous system.
Collapse
Affiliation(s)
- Seong Su Kang
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA
| | - Matthew P Keasey
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA
| | - Sheila A Arnold
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA; Department of Pharmacology and Toxicology, University of Louisville, KY 40292, USA
| | - Rollie Reid
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA
| | - Justin Geralds
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA
| | - Theo Hagg
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY 40292, USA; Department of Neurological Surgery, University of Louisville, KY 40292, USA; Department of Pharmacology and Toxicology, University of Louisville, KY 40292, USA.
| |
Collapse
|
42
|
Ramani P, Norton A, Somerville MS, May MT. PROX1 lymphatic density correlates with adverse clinicopathological factors, lymph node metastases and survival in neuroblastomas. J Neurooncol 2012; 108:375-83. [DOI: 10.1007/s11060-012-0838-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Accepted: 02/20/2012] [Indexed: 12/25/2022]
|
43
|
Mooney R, Haeger S, Lawal R, Mason M, Shrestha N, Laperle A, Bjugstad K, Mahoney M. Control of neural cell composition in poly(ethylene glycol) hydrogel culture with soluble factors. Tissue Eng Part A 2011; 17:2805-15. [PMID: 21823990 DOI: 10.1089/ten.tea.2010.0654] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Poly(ethylene glycol) (PEG) hydrogels are being developed as cell delivery vehicles that have great potential to improve neuronal replacement therapies. Current research priorities include (1) characterizing neural cell growth within PEG hydrogels relative to standard culture systems and (2) generating neuronal-enriched populations within the PEG hydrogel environment. This study compares the percentage of neural precursor cells (NPCs), neurons, and glia present when dissociated neural cells are seeded within PEG hydrogels relative to standard monolayer culture. Results demonstrate that PEG hydrogels enriched the initial cell population for NPCs, which subsequently gave rise to neurons, then to glia. Relative to monolayer culture, PEG hydrogels maintained an increased percentage of NPCs and a decreased percentage of glia. This neurogenic advantage of PEG hydrogels is accentuated in the presence of basic fibroblast growth factor and epidermal growth factor, which more potently increase NPC and neuronal expression markers when applied to cells cultured within PEG hydrogels. Finally, this work demonstrates that glial differentiation can be selectively eliminated upon supplementation with a γ-secretase inhibitor. Together, this study furthers our understanding of how the PEG hydrogel environment influences neural cell composition and also describes select soluble factors that are useful in generating neuronal-enriched populations within the PEG hydrogel environment.
Collapse
Affiliation(s)
- Rachael Mooney
- Department of Biochemistry, University of Colorado, Boulder, Colorado 80309, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
A transient expression of Prospero promotes cell cycle exit of Drosophila postembryonic neurons through the regulation of Dacapo. PLoS One 2011; 6:e19342. [PMID: 21552484 PMCID: PMC3084296 DOI: 10.1371/journal.pone.0019342] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 03/28/2011] [Indexed: 11/29/2022] Open
Abstract
Cell proliferation, specification and terminal differentiation must be precisely coordinated during brain development to ensure the correct production of different neuronal populations. Most Drosophila neuroblasts (NBs) divide asymmetrically to generate a new NB and an intermediate progenitor called ganglion mother cell (GMC) which divides only once to generate two postmitotic cells called ganglion cells (GCs) that subsequently differentiate into neurons. During the asymmetric division of NBs, the homeodomain transcription factor PROSPERO is segregated into the GMC where it plays a key role as cell fate determinant. Previous work on embryonic neurogenesis has shown that PROSPERO is not expressed in postmitotic neuronal progeny. Thus, PROSPERO is thought to function in the GMC by repressing genes required for cell-cycle progression and activating genes involved in terminal differentiation. Here we focus on postembryonic neurogenesis and show that the expression of PROSPERO is transiently upregulated in the newly born neuronal progeny generated by most of the larval NBs of the OL and CB. Moreover, we provide evidence that this expression of PROSPERO in GCs inhibits their cell cycle progression by activating the expression of the cyclin-dependent kinase inhibitor (CKI) DACAPO. These findings imply that PROSPERO, in addition to its known role as cell fate determinant in GMCs, provides a transient signal to ensure a precise timing for cell cycle exit of prospective neurons, and hence may link the mechanisms that regulate neurogenesis and those that control cell cycle progression in postembryonic brain development.
Collapse
|
45
|
Kaltezioti V, Kouroupi G, Oikonomaki M, Mantouvalou E, Stergiopoulos A, Charonis A, Rohrer H, Matsas R, Politis PK. Prox1 regulates the notch1-mediated inhibition of neurogenesis. PLoS Biol 2010; 8:e1000565. [PMID: 21203589 PMCID: PMC3006385 DOI: 10.1371/journal.pbio.1000565] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 11/03/2010] [Indexed: 11/18/2022] Open
Abstract
During development of the spinal cord, Prox1 controls the balance between proliferation and differentiation of neural progenitor cells via suppression of Notch1 gene expression. Activation of Notch1 signaling in neural progenitor cells (NPCs) induces self-renewal and inhibits neurogenesis. Upon neuronal differentiation, NPCs overcome this inhibition, express proneural genes to induce Notch ligands, and activate Notch1 in neighboring NPCs. The molecular mechanism that coordinates Notch1 inactivation with initiation of neurogenesis remains elusive. Here, we provide evidence that Prox1, a transcription repressor and downstream target of proneural genes, counteracts Notch1 signaling via direct suppression of Notch1 gene expression. By expression studies in the developing spinal cord of chick and mouse embryo, we showed that Prox1 is limited to neuronal precursors residing between the Notch1+ NPCs and post-mitotic neurons. Physiological levels of Prox1 in this tissue are sufficient to allow binding at Notch1 promoter and they are critical for proper Notch1 transcriptional regulation in vivo. Gain-of-function studies in the chick neural tube and mouse NPCs suggest that Prox1-mediated suppression of Notch1 relieves its inhibition on neurogenesis and allows NPCs to exit the cell cycle and differentiate. Moreover, loss-of-function in the chick neural tube shows that Prox1 is necessary for suppression of Notch1 outside the ventricular zone, inhibition of active Notch signaling, down-regulation of NPC markers, and completion of neuronal differentiation program. Together these data suggest that Prox1 inhibits Notch1 gene expression to control the balance between NPC self-renewal and neuronal differentiation. Early during development, neural progenitor cells (NPCs) can either proliferate or differentiate into neurons. Thus, generation of the correct number of neurons is governed by a tightly regulated balance between proliferation and differentiation, and disruption of this balance can result in severe developmental deficits, malformations, or cancers. Notch1 is a member of the Notch family of receptors, which make up a highly conserved cell signaling system. Notch1 signaling has been shown to inhibit NPC differentiation and to promote self-renewal, thereby allowing NPCs to divide and progressively generate the enormous number of neurons present in the central nervous system. The molecular mechanism by which NPCs overcome Notch1-mediated inhibition in order to differentiate into neurons, however, is not completely understood. In this study, we show that Prox1, a homeobox transcriptional repressor, plays a fundamental role in the switch to differentiation by suppressing the expression of Notch1 receptor, thereby preventing newly produced neuronal precursors from receiving inhibitory signals from Notch ligands present in neighboring cells. This transcriptional repression may regulate cell cycle exit and differentiation of NPCs as they migrate towards different regions and adopt their final cell fates. We suggest that Prox1 may exert its known influence on embryonic development, organ morphogenesis, and cancer through its ability to counteract Notch1 signaling.
Collapse
Affiliation(s)
- Valeria Kaltezioti
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Georgia Kouroupi
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Maria Oikonomaki
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Evangelia Mantouvalou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Athanasios Stergiopoulos
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Aristidis Charonis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Hermann Rohrer
- Department of Neurochemistry, Max-Planck Institute for Brain Research, Frankfurt/Main, Germany
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Panagiotis K. Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- * E-mail:
| |
Collapse
|
46
|
Bayraktar OA, Boone JQ, Drummond ML, Doe CQ. Drosophila type II neuroblast lineages keep Prospero levels low to generate large clones that contribute to the adult brain central complex. Neural Dev 2010; 5:26. [PMID: 20920301 PMCID: PMC2958855 DOI: 10.1186/1749-8104-5-26] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 10/01/2010] [Indexed: 01/01/2023] Open
Abstract
Tissue homeostasis depends on the ability of stem cells to properly regulate self-renewal versus differentiation. Drosophila neural stem cells (neuroblasts) are a model system to study self-renewal and differentiation. Recent work has identified two types of larval neuroblasts that have different self-renewal/differentiation properties. Type I neuroblasts bud off a series of small basal daughter cells (ganglion mother cells) that each generate two neurons. Type II neuroblasts bud off small basal daughter cells called intermediate progenitors (INPs), with each INP generating 6 to 12 neurons. Type I neuroblasts and INPs have nuclear Asense and cytoplasmic Prospero, whereas type II neuroblasts lack both these transcription factors. Here we test whether Prospero distinguishes type I/II neuroblast identity or proliferation profile, using several newly characterized Gal4 lines. We misexpress prospero using the 19H09-Gal4 line (expressed in type II neuroblasts but no adjacent type I neuroblasts) or 9D11-Gal4 line (expressed in INPs but not type II neuroblasts). We find that differential prospero expression does not distinguish type I and type II neuroblast identities, but Prospero regulates proliferation in both type I and type II neuroblast lineages. In addition, we use 9D11 lineage tracing to show that type II lineages generate both small-field and large-field neurons within the adult central complex, a brain region required for locomotion, flight, and visual pattern memory.
Collapse
Affiliation(s)
- Omer Ali Bayraktar
- Howard Hughes Medical Institute, University of Oregon, Eugene, 97403, USA
| | | | | | | |
Collapse
|
47
|
Elmi M, Matsumoto Y, Zeng ZJ, Lakshminarasimhan P, Yang W, Uemura A, Nishikawa SI, Moshiri A, Tajima N, Agren H, Funa K. TLX activates MASH1 for induction of neuronal lineage commitment of adult hippocampal neuroprogenitors. Mol Cell Neurosci 2010; 45:121-31. [PMID: 20599619 DOI: 10.1016/j.mcn.2010.06.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Revised: 06/01/2010] [Accepted: 06/09/2010] [Indexed: 02/06/2023] Open
Abstract
The orphan nuclear receptor TLX has been proposed to act as a repressor of cell cycle inhibitors to maintain the neural stem cells in an undifferentiated state, and prevents commitment into astrocyte lineages. However, little is known about the mechanism of TLX in neuronal lineage commitment and differentiation. A majority of adult rat hippocampus-derived progenitors (AHPs) cultured in the presence of FGF express a high level of TLX and a fraction of these cells also express the proneural gene MASH1. Upon FGF withdrawal, TLX rapidly decreased, while MASH1 was intensely expressed within 1h, decreasing gradually to disappear at 24h. Adenoviral transduction of TLX in AHP cells in the absence of FGF transiently increased cell proliferation, however, later resulted in neuronal differentiation by inducing MASH1, Neurogenin1, DCX, and MAP2ab. Furthermore, TLX directly targets and activates the MASH1 promoter through interaction with Sp1, recruiting co-activators whereas dismissing the co-repressor HDAC4. Conversely, silencing of TLX in AHPs decreased beta-III tubulin and DCX expression and promoted glial differentiation. Our results thus suggest that TLX not only acts as a repressor of cell cycle and glial differentiation but also activates neuronal lineage commitment in AHPs.
Collapse
Affiliation(s)
- Muna Elmi
- Institute of Biomedicine, Department of Medical Chemistry and Cell Biology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hanai S, Saito T, Nakagawa E, Arai A, Otsuki T, Sasaki M, Goto YI, Itoh M. Abnormal maturation of non-dysmorphic neurons in focal cortical dysplasia: Immunohistochemical considerations. Seizure 2010; 19:274-9. [DOI: 10.1016/j.seizure.2010.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 03/18/2010] [Accepted: 04/01/2010] [Indexed: 10/19/2022] Open
|
49
|
Osório J, Mueller T, Rétaux S, Vernier P, Wullimann MF. Phylotypic expression of the bHLH genes Neurogenin2, Neurod, and Mash1 in the mouse embryonic forebrain. J Comp Neurol 2010; 518:851-71. [PMID: 20058311 DOI: 10.1002/cne.22247] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In the anamniote model animals, zebrafish and Xenopus laevis, highly comparable early forebrain expression patterns of proneural basic helix-loop-helix (bHLH) genes relevant for neurogenesis (atonal homologs, i.e., neurogenins/NeuroD and achaete-scute homologs, i.e., Ascl/ash) were previously revealed during a particular period of development (zebrafish: 3 days; frog: stage 48). Neurogenins/NeuroD on the one hand and Ascl1/ash1 on the other hand exhibit essentially mutually exclusive spatial patterns, probably reflecting different positional information received within the neural tube, and appear to underlie glutamatergic versus GABAergic neuronal differentiation, respectively. Significant data suggest that similar complementary localizations of these proneural genes and corresponding differentiation pathways also exist in the mouse, the prominent mammalian model. The present article reports on detailed mouse brain bHLH gene expression patterns to fill existing gaps in the identification of expression domains, especially outside the telencephalon. Clearly, there are strong similarities in the complementarity of territories expressing Ascl1/Mash 1 versus neurogenins/NeuroD in the entire mouse forebrain, except for the pretectal alar plate and basal plate of prosomeres 1-3. The analysis substantiates localization of neurogenins/NeuroD in the pallium, eminentia thalami, and dorsal thalamus, and expression of Ascl1/Mash 1 in the striatal and septal subpallium, preoptic region, ventral thalamus, and hypothalamus, which is highly similar to the situation described in Xenopus and zebrafish. Thus, all three vertebrate model species display a "phylotypic stage or period" corresponding to a temporally and spatially defined control of neurogenesis during forebrain development, ultimately resulting in the differentiation of distinct populations of glutamatergic versus GABAergic neurons.
Collapse
Affiliation(s)
- Joana Osório
- CNRS, Institute of Neurobiology A. Fessard, Development, Evolution, Plasticity of the Nervous System, Research Unit 2197, 91198 Gif-sur-Yvette, France
| | | | | | | | | |
Collapse
|
50
|
Expression of PROX1 Is a common feature of high-grade malignant astrocytic gliomas. J Neuropathol Exp Neurol 2010; 69:129-38. [PMID: 20084020 DOI: 10.1097/nen.0b013e3181ca4767] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PROX1 is a prospero-related transcription factor that plays a critical role in the development of various organs including the mammalian lymphatic and central nervous systems; it controls cell proliferation and differentiation through different transcription pathwaysand has both oncogenic and tumor-suppressive functions. We investigated PROX1 expression patterns in 56 human astrocytic gliomas of different grades using immunohistochemistry. An average of 79% of cells in World Health Organization Grade IV (glioblastoma, n = 15) and 57% of cells in World Health Organization Grade III (anaplastic astrocytoma, n = 13) were strongly PROX1 positive; low-grade diffuse astrocytomas (Grade II, n = 13) had 21% of cells that were strongly positive; Grade I tumors (n = 15) had 1.5%; and non-neoplastic brain tissue (n = 15) had 3.7% of cells that were PROX1 positive. Double immunolabeling showed that PROX1+ cells in glioblastomas frequently coexpressed early neuronal proteins MAP2 and betaIII-tubulin but not the mature neuronal marker protein NeuN. Analyses of coexpression with proliferation markers suggest that PROX1+ cells have a marginally lower rate of proliferation than other tumor cells but are still mitotically active. We conclude that PROX1 may constitute a useful tool for the diagnosis and grading ofastrocytic gliomas and for distinguishing Grade III and Grade IV tumors from Grade I and Grade II tumors.
Collapse
|