1
|
Yabut OR, Arela J, Gomez HG, Castillo JG, Ngo T, Pleasure SJ. Aberrant FGF signaling promotes granule neuron precursor expansion in SHH subgroup infantile medulloblastoma. eLife 2025; 13:RP100767. [PMID: 39835775 PMCID: PMC11750132 DOI: 10.7554/elife.100767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
Mutations in Sonic Hedgehog (SHH) signaling pathway genes, for example, Suppressor of Fused (SUFU), drive granule neuron precursors (GNP) to form medulloblastomas (MBSHH). However, how different molecular lesions in the Shh pathway drive transformation is frequently unclear, and SUFU mutations in the cerebellum seem distinct. In this study, we show that fibroblast growth factor 5 (FGF5) signaling is integral for many infantile MBSHH cases and that FGF5 expression is uniquely upregulated in infantile MBSHH tumors. Similarly, mice lacking SUFU (Sufu-cKO) ectopically express Fgf5 specifically along the secondary fissure where GNPs harbor preneoplastic lesions and show that FGFR signaling is also ectopically activated in this region. Treatment with an FGFR antagonist rescues the severe GNP hyperplasia and restores cerebellar architecture. Thus, direct inhibition of FGF signaling may be a promising and novel therapeutic candidate for infantile MBSHH.
Collapse
Affiliation(s)
- Odessa R Yabut
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Jessica Arela
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Hector G Gomez
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Jesse Garcia Castillo
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Thomas Ngo
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| | - Samuel J Pleasure
- Department of Neurology, Weill Institute for Neuroscience, University of California San FranciscoSan FranciscoUnited States
| |
Collapse
|
2
|
Rigueur D. A primer for Fibroblast Growth Factor 16 (FGF16). Differentiation 2024; 140:100817. [PMID: 39632143 DOI: 10.1016/j.diff.2024.100817] [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: 06/03/2024] [Revised: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024]
Abstract
During the discovery of the Fibroblast Growth Factor superfamily, scientists were determined to uncover all the genes that encoded FGF proteins. In 1998, FGF16 was discovered with classical cloning techniques in human and rat heart samples. FGF16 loss- and gain-of-function experiments in several organisms demonstrated a conserved function in vertebrates, and as a component of the FGF9 subfamily of ligands (FGF-E/-9/-20), is functionally conserved and sufficient to rescue loss-of-function phenotypes in invertebrates, like C. elegans. FGF16 has a broad expression pattern, predominantly expressed in brown adipose tissue, heart, with low but detectable levels in the brain, olfactory bulb, inner ear, muscle, thymus, pancreas, spleen, stomach, small intestine, and gonads (testis and ovary). FGF16 is also expressed moderately in the late developing limb bud. Despite its expression levels, this ligand plays notable roles in autopod metacarpal development; loss of one allele causes congenital metacarpal 4-5 fusion and hand deformities in humans. The broad expression pattern of FGF16 in several tissues underscores its multifaceted roles in stem cell maintenance, proliferation, cell fate specification, and metabolism.
Collapse
Affiliation(s)
- Diana Rigueur
- University of California, Los Angeles, Department of Molecular, Cell and Developmental Biology, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Anerillas C, Perramon-Güell A, Altés G, Cuesta S, Vaquero M, Olomí A, Rodríguez-Barrueco R, Llobet-Navàs D, Egea J, Dolcet X, Yeramian A, Encinas M. Sprouty1 is a broad mediator of cellular senescence. Cell Death Dis 2024; 15:296. [PMID: 38670941 PMCID: PMC11053034 DOI: 10.1038/s41419-024-06689-4] [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: 10/10/2023] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024]
Abstract
Genes of the Sprouty family (Spry1-4) restrain signaling by certain receptor tyrosine kinases. Consequently, these genes participate in several developmental processes and function as tumor suppressors in adult life. Despite these important roles, the biology of this family of genes still remains obscure. Here we show that Sprouty proteins are general mediators of cellular senescence. Induction of cellular senescence by several triggers in vitro correlates with upregulation of Sprouty protein levels. More importantly, overexpression of Sprouty genes is sufficient to cause premature cellular senescence, via a conserved N-terminal tyrosine (Tyrosine 53 of Sprouty1). Accordingly, fibroblasts from knockin animals lacking that tyrosine escape replicative senescence. In vivo, heterozygous knockin mice display delayed induction of cellular senescence during cutaneous wound healing and upon chemotherapy-induced cellular senescence. Unlike other functions of this family of genes, induction of cellular senescence appears to be independent of activation of the ERK1/2 pathway. Instead, we show that Sprouty proteins induce cellular senescence upstream of the p38 pathway in these in vitro and in vivo paradigms.
Collapse
Affiliation(s)
- Carlos Anerillas
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain.
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA.
- Homeostasis de tejidos y órganos program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Autónoma de Madrid, Madrid, Spain.
| | - Aida Perramon-Güell
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain
| | - Gisela Altés
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain
| | - Sara Cuesta
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain
- Fundación de Investigación Biomédica de Cádiz, Hospital Universitario Puerta del Mar, Novena Planta, Investigación, Av Ana de Viya, 21, Cádiz, Spain
| | - Marta Vaquero
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain
- Hospital Universitari Arnau de Vilanova, Rovira Roure, 80, Lleida, Spain
| | - Anna Olomí
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain
| | - Ruth Rodríguez-Barrueco
- Laboratory of Precision Medicine, Oncobell Program. Bellvitge Biomedical Research Institute (IDIBELL), Gran via De l'Hospitalet, Barcelona, Spain
| | - David Llobet-Navàs
- Laboratory of Precision Medicine, Oncobell Program. Bellvitge Biomedical Research Institute (IDIBELL), Gran via De l'Hospitalet, Barcelona, Spain
| | - Joaquim Egea
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain
| | - Xavi Dolcet
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain
| | - Andrée Yeramian
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain
| | - Mario Encinas
- Developmental and Oncogenic Signaling Group, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, Lleida, Spain.
| |
Collapse
|
4
|
Altés G, Vaquero M, Cuesta S, Anerillas C, Macià A, Espinet C, Ribera J, Bellusci S, Klein OD, Yeramian A, Dolcet X, Egea J, Encinas M. A dominant negative mutation uncovers cooperative control of caudal Wolffian duct development by Sprouty genes. Cell Mol Life Sci 2022; 79:514. [PMID: 36098804 PMCID: PMC9470706 DOI: 10.1007/s00018-022-04546-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 12/01/2022]
Abstract
The Wolffian ducts (WD) are paired epithelial tubules central to the development of the mammalian genitourinary tract. Outgrowths from the WD known as the ureteric buds (UB) generate the collecting ducts of the kidney. Later during development, the caudal portion of the WD will form the vas deferens, epididymis and seminal vesicle in males, and will degenerate in females. While the genetic pathways controlling the development of the UB are firmly established, less is known about those governing development of WD portions caudal to the UB. Sprouty proteins are inhibitors of receptor tyrosine kinase (RTK) signaling in vivo. We have recently shown that homozygous mutation of a conserved tyrosine (Tyr53) of Spry1 results in UB defects indistinguishable from that of Spry1 null mice. Here, we show that heterozygosity for the Spry1 Y53A allele causes caudal WD developmental defects consisting of ectopically branched seminal vesicles in males and persistent WD in females, without affecting kidney development. Detailed analysis reveals that this phenotype also occurs in Spry1+/– mice but with a much lower penetrance, indicating that removal of tyrosine 53 generates a dominant negative mutation in vivo. Supporting this notion, concomitant deletion of one allele of Spry1 and Spry2 also recapitulates the genital phenotype of Spry1Y53A/+ mice with high penetrance. Mechanistically, we show that unlike the effects of Spry1 in kidney development, these caudal WD defects are independent of Ret signaling, but can be completely rescued by lowering the genetic dosage of Fgf10. In conclusion, mutation of tyrosine 53 of Spry1 generates a dominant negative allele that uncovers fine-tuning of caudal WD development by Sprouty genes.
Collapse
Affiliation(s)
- Gisela Altés
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Marta Vaquero
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Sara Cuesta
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain.,Fundación de Investigación Biomédica de Cádiz, Hospital Universitario Puerta del Mar, Novena Planta, Investigación, Av Ana de Viya, 21, 11009, Cádiz, Spain
| | - Carlos Anerillas
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Anna Macià
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Carme Espinet
- Department of Basic Medical Sciences, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Joan Ribera
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | | | - Ophir D Klein
- Department of Orofacial Sciences, University of California, San Francisco, USA.,Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, USA
| | - Andree Yeramian
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain
| | - Xavi Dolcet
- Department of Basic Medical Sciences, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Joaquim Egea
- Department of Basic Medical Sciences, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Rovira Roure, 80, 25198, Lleida, Spain
| | - Mario Encinas
- Department of Experimental Medicine, Universitat de Lleida/Institut de Recerca Biomèdica de Lleida, Edifici Biomedicina I, Lab 2.8, Rovira Roure, 80, 25198, Lleida, Spain.
| |
Collapse
|
5
|
Kobayashi Y, Nouet J, Baljinnyam E, Siddiqui Z, Fine DH, Fraidenraich D, Kumar VA, Shimizu E. iPSC-derived cranial neural crest-like cells can replicate dental pulp tissue with the aid of angiogenic hydrogel. Bioact Mater 2022; 14:290-301. [PMID: 35310357 PMCID: PMC8897656 DOI: 10.1016/j.bioactmat.2021.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 12/18/2022] Open
Abstract
The dental pulp has irreplaceable roles in maintaining healthy teeth and its regeneration is a primary aim of regenerative endodontics. This study aimed to replicate the characteristics of dental pulp tissue by using cranial neural crest (CNC)-like cells (CNCLCs); these cells were generated by modifying several steps of a previously established method for deriving NC-like cells from induced pluripotent stem cells (iPSCs). CNC is the anterior region of the neural crest in vertebrate embryos, which contains the primordium of dental pulp cells or odontoblasts. The produced CNCLCs showed approximately 2.5–12,000-fold upregulations of major CNC marker genes. Furthermore, the CNCLCs exhibited remarkable odontoblastic differentiation ability, especially when treated with a combination of the fibroblast growth factors (FGFs) FGF4 and FGF9. The FGFs induced odontoblast marker genes by 1.7–5.0-fold, as compared to bone morphogenetic protein 4 (BMP4) treatment. In a mouse subcutaneous implant model, the CNCLCs briefly fated with FGF4 + FGF9 replicated dental pulp tissue characteristics, such as harboring odontoblast-like cells, a dentin-like layer, and vast neovascularization, induced by the angiogenic self-assembling peptide hydrogel (SAPH), SLan. SLan acts as a versatile biocompatible scaffold in the canal space. This study demonstrated a successful collaboration between regenerative medicine and SAPH technology. Cranial neural crest like cells (CNCLCs) were generated by simplifying a previously established method for deriving neural crest-like cells from iPSCs. The produced CNCLCs showed approximately ∼12,000-fold upregulations of major CNC marker genes. The combination of fibroblast growth factors, FGF4 and FGF9, induced the CNCLCs toward odontoblastic differentiation more effectively than BMP4. In a mice subcutaneous implant model, the CNCLCs replicated the characteristics of dental pulp harboring vast neovascularization with the aid of the angiogenic hydrogel, SLan.
Collapse
|
6
|
Hausott B, Glueckert R, Schrott-Fischer A, Klimaschewski L. Signal Transduction Regulators in Axonal Regeneration. Cells 2022; 11:cells11091537. [PMID: 35563843 PMCID: PMC9104247 DOI: 10.3390/cells11091537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
Intracellular signal transduction in response to growth factor receptor activation is a fundamental process during the regeneration of the nervous system. In this context, intracellular inhibitors of neuronal growth factor signaling have become of great interest in the recent years. Among them are the prominent signal transduction regulators Sprouty (SPRY) and phosphatase and tensin homolog deleted on chromosome 10 (PTEN), which interfere with major signaling pathways such as extracellular signal-regulated kinase (ERK) or phosphoinositide 3-kinase (PI3K)/Akt in neurons and glial cells. Furthermore, SPRY and PTEN are themselves tightly regulated by ubiquitin ligases such as c-casitas b-lineage lymphoma (c-CBL) or neural precursor cell expressed developmentally down-regulated protein 4 (NEDD4) and by different microRNAs (miRs) including miR-21 and miR-222. SPRY, PTEN and their intracellular regulators play an important role in the developing and the lesioned adult central and peripheral nervous system. This review will focus on the effects of SPRY and PTEN as well as their regulators in various experimental models of axonal regeneration in vitro and in vivo. Targeting these signal transduction regulators in the nervous system holds great promise for the treatment of neurological injuries in the future.
Collapse
Affiliation(s)
- Barbara Hausott
- Institute of Neuroanatomy, Medical University Innsbruck, 6020 Innsbruck, Austria;
- Correspondence:
| | - Rudolf Glueckert
- Department of Otorhinolaryngology, Medical University Innsbruck, 6020 Innsbruck, Austria; (R.G.); (A.S.-F.)
| | - Anneliese Schrott-Fischer
- Department of Otorhinolaryngology, Medical University Innsbruck, 6020 Innsbruck, Austria; (R.G.); (A.S.-F.)
| | - Lars Klimaschewski
- Institute of Neuroanatomy, Medical University Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
7
|
Sugahara F, Pascual-Anaya J, Kuraku S, Kuratani S, Murakami Y. Genetic Mechanism for the Cyclostome Cerebellar Neurons Reveals Early Evolution of the Vertebrate Cerebellum. Front Cell Dev Biol 2021; 9:700860. [PMID: 34485287 PMCID: PMC8416312 DOI: 10.3389/fcell.2021.700860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/13/2021] [Indexed: 11/13/2022] Open
Abstract
The vertebrate cerebellum arises at the dorsal part of rhombomere 1, induced by signals from the isthmic organizer. Two major cerebellar neuronal subtypes, granule cells (excitatory) and Purkinje cells (inhibitory), are generated from the anterior rhombic lip and the ventricular zone, respectively. This regionalization and the way it develops are shared in all extant jawed vertebrates (gnathostomes). However, very little is known about early evolution of the cerebellum. The lamprey, an extant jawless vertebrate lineage or cyclostome, possesses an undifferentiated, plate-like cerebellum, whereas the hagfish, another cyclostome lineage, is thought to lack a cerebellum proper. In this study, we found that hagfish Atoh1 and Wnt1 genes are co-expressed in the rhombic lip, and Ptf1a is expressed ventrally to them, confirming the existence of r1's rhombic lip and the ventricular zone in cyclostomes. In later stages, lamprey Atoh1 is downregulated in the posterior r1, in which the NeuroD increases, similar to the differentiation process of cerebellar granule cells in gnathostomes. Also, a continuous Atoh1-positive domain in the rostral r1 is reminiscent of the primordium of valvula cerebelli of ray-finned fishes. Lastly, we detected a GAD-positive domain adjacent to the Ptf1a-positive ventricular zone in lampreys, suggesting that the Ptf1a-positive cells differentiate into some GABAergic inhibitory neurons such as Purkinje and other inhibitory neurons like in gnathostomes. Altogether, we conclude that the ancestral genetic programs for the formation of a distinct cerebellum were established in the last common ancestor of vertebrates.
Collapse
Affiliation(s)
- Fumiaki Sugahara
- Division of Biology, Hyogo College of Medicine, Nishinomiya, Japan.,Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), Kobe, Japan
| | - Juan Pascual-Anaya
- Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), Kobe, Japan.,Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain.,Andalusian Centre for Nanomedicine and Biotechnology (BIONAND), Málaga, Spain
| | - Shigehiro Kuraku
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan.,Molecular Life History Laboratory, Department of Genomics and Evolutionary Biology, National Institute of Genetics, Mishima, Japan
| | - Shigeru Kuratani
- Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), Kobe, Japan.,Laboratory for Evolutionary Morphology, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| | - Yasunori Murakami
- Graduate School of Science and Engineering, Ehime University, Matsuyama, Japan
| |
Collapse
|
8
|
Tian L, Xiao H, Li M, Wu X, Xie Y, Zhou J, Zhang X, Wang B. A novel Sprouty4-ERK1/2-Wnt/β-catenin regulatory loop in marrow stromal progenitor cells controls osteogenic and adipogenic differentiation. Metabolism 2020; 105:154189. [PMID: 32105664 DOI: 10.1016/j.metabol.2020.154189] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/16/2020] [Accepted: 02/24/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Sprouty (SPRY) proteins play critical roles in controlling cell proliferation, differentiation, and survival by inhibiting receptor tyrosine kinase (RTK)-mediated extracellular signal-regulated kinase (ERK) signaling. Recent studies have demonstrated that SPRY4 negatively regulates angiogenesis and tumor growth. However, whether SPRY4 regulates osteogenic and/or adipogenic differentiation of mesenchymal stem cells remains to be explored. RESULTS In this study, we investigated the expression pattern of Spry4 and found that its expression was regulated during the differentiation of mouse marrow stromal progenitor cells and increased in the metaphysis of ovariectomized mice. In vitro loss-of-function and gain-of-function studies demonstrated that SPRY4 inhibited osteogenic differentiation and stimulated adipogenic differentiation of progenitor cells. In vivo experiments showed that silencing of Spry4 in the marrow of C57BL/6 mice blocked fat accumulation and promoted osteoblast differentiation in ovariectomized mice. Mechanistic investigations revealed the inhibitory effect of SPRY4 on canonical wingless-type MMTV integration site (Wnt) signaling and ERK pathway. ERK1/2 was shown to interact with low-density lipoprotein receptor-related protein 6 (LRP6) and activate the canonical Wnt signaling pathway. Inactivation of Wnt signaling attenuated the inhibition of adipogenic differentiation and stimulation of osteogenic differentiation by Spry4 small interfering RNA (siRNA). Finally, promoter study revealed that β-catenin transcriptionally inhibited the expression of Spry4. CONCLUSIONS Our study for the first time suggests that a novel SPRY4-ERK1/2-Wnt/β-catenin regulatory loop exists in marrow stromal progenitor cells and plays a key role in cell fate determination. It also highlights the potential of SPRY4 as a novel therapeutic target for the treatment of metabolic bone disorders such as osteoporosis.
Collapse
Affiliation(s)
- Lijie Tian
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Hongyan Xiao
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Mengyue Li
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xiaowen Wu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Yan Xie
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Jie Zhou
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xin Zhang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Baoli Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
9
|
Sarić N, Selby M, Ramaswamy V, Kool M, Stockinger B, Hogstrand C, Williamson D, Marino S, Taylor MD, Clifford SC, Basson MA. The AHR pathway represses TGFβ-SMAD3 signalling and has a potent tumour suppressive role in SHH medulloblastoma. Sci Rep 2020; 10:148. [PMID: 31924815 PMCID: PMC6954114 DOI: 10.1038/s41598-019-56876-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 12/02/2019] [Indexed: 12/22/2022] Open
Abstract
Sonic Hedgehog (SHH) medulloblastomas are brain tumours that arise in the posterior fossa. Cancer-propagating cells (CPCs) provide a reservoir of cells capable of tumour regeneration and relapse post-treatment. Understanding and targeting the mechanisms by which CPCs are maintained and expanded in SHH medulloblastoma could present novel therapeutic opportunities. We identified the aryl hydrocarbon receptor (AHR) pathway as a potent tumour suppressor in a SHH medulloblastoma mouse model. Ahr-deficient tumours and CPCs grown in vitro, showed elevated activation of the TGFβ mediator, SMAD3. Pharmacological inhibition of the TGFβ/SMAD3 signalling axis was sufficient to inhibit the proliferation and promote the differentiation of Ahr-deficient CPCs. Human SHH medulloblastomas with high expression of the AHR repressor (AHRR) exhibited a significantly worse prognosis compared to AHRRlow tumours in two independent patient cohorts. Together, these findings suggest that reduced AHR pathway activity promotes SHH medulloblastoma progression, consistent with a tumour suppressive role for AHR. We propose that TGFβ/SMAD3 inhibition may represent an actionable therapeutic approach for a subset of aggressive SHH medulloblastomas characterised by reduced AHR pathway activity.
Collapse
Affiliation(s)
- Nemanja Sarić
- Centre for Craniofacial and Regenerative Biology, King's College London, Floor 27, Guy's Hospital Tower Wing, London, SE1 9RT, UK
| | - Matthew Selby
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| | - Vijay Ramaswamy
- Divisions of Hematology/Oncology and Neurosurgery, The Hospital for Sick Children, Toronto, ON, Canada
- Departments of Medical Biophysics and Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Marcel Kool
- Hopp Children's Cancer Center (KiTZ), Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), and German Cancer Consortium (DKTK), Heidelberg, Germany
| | | | - Christer Hogstrand
- Diabetes & Nutritional Sciences Division, King's College London, 3.85 Franklin-Wilkins Building, London, SE1 9NH, UK
| | - Daniel Williamson
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| | - Silvia Marino
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Michael D Taylor
- Divisions of Hematology/Oncology and Neurosurgery, The Hospital for Sick Children, Toronto, ON, Canada
- Departments of Medical Biophysics and Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Steven C Clifford
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| | - M Albert Basson
- Centre for Craniofacial and Regenerative Biology, King's College London, Floor 27, Guy's Hospital Tower Wing, London, SE1 9RT, UK.
- MRC Centre for Neurodevelopmental Disorders, King's College London, 4th floor, New Hunt's House, London, SE1 1UL, UK.
| |
Collapse
|
10
|
Sprouty3 and Sprouty4, Two Members of a Family Known to Inhibit FGF-Mediated Signaling, Exert Opposing Roles on Proliferation and Migration of Glioblastoma-Derived Cells. Cells 2019; 8:cells8080808. [PMID: 31374860 PMCID: PMC6721513 DOI: 10.3390/cells8080808] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/26/2022] Open
Abstract
Dysregulation of receptor tyrosine kinase-induced pathways is a critical step driving the oncogenic potential of brain cancer. In this study, we investigated the role of two members of the Sprouty (Spry) family in brain cancer-derived cell lines. Using immunoblot analyses we found essential differences in the pattern of endogenous Spry3 and Spry4 expression. While Spry4 expression was mitogen-dependent and repressed in a number of cells from higher malignant brain cancers, Spry3 levels neither fluctuated in response to serum withdrawal nor were repressed in glioblastoma (GBM)-derived cell lines. In accordance to the well-known inhibitory role of Spry proteins in fibroblast growth factor (FGF)-mediated signaling, both Spry proteins were able to interfere with FGF-induced activation of the MAPK pathway although to a different extent. In response to serum solely, Spry4 exerts its role as a negative regulator of MAPK activation. Ectopic expression of Spry4 inhibited proliferation and migration of GBM-originated cells, positioning it as a tumor suppressor in brain cancer. In contrast, elevated Spry3 levels accelerated both proliferation and migration of these cell lines, while repression of Spry3 levels using shRNA caused a significant diminished growth and migration velocity rate of a GBM-derived cell line. This argues for a tumor-promoting function of Spry3 in GBMs. Based on these data we conclude that Spry3 and Spry4 fulfill different if not opposing roles within the cancerogenesis of brain malignancies.
Collapse
|
11
|
Ning Z, Williams JM, Kumari R, Baranov PV, Moore T. Opposite Expression Patterns of Spry3 and p75NTR in Cerebellar Vermis Suggest a Male-Specific Mechanism of Autism Pathogenesis. Front Psychiatry 2019; 10:416. [PMID: 31275178 PMCID: PMC6591651 DOI: 10.3389/fpsyt.2019.00416] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/24/2019] [Indexed: 12/22/2022] Open
Abstract
Autism is a genetically complex neurobehavioral disorder with a population prevalence of more than 1%. Cerebellar abnormalities, including Purkinje cell deficits in the vermis, are consistently reported, and rodent models of cerebellar dysfunction exhibit features analogous to human autism. We previously analyzed the regulation and expression of the pseudoautosomal region 2 gene SPRY3, which is adjacent to X chromosome-linked TMLHE, a known autism susceptibility gene. SPRY3 is a regulator of branching morphogenesis and is strongly expressed in Purkinje cells. We previously showed that mouse Spry3 is not expressed in cerebellar vermis lobules VI-VII and X, regions which exhibit significant Purkinje cell loss or abnormalities in autism. However, these lobules have relatively high expression of p75NTR, which encodes a neurotrophin receptor implicated in autism. We propose a mechanism whereby inappropriate SPRY3 expression in these lobules could interact with TrkB and p75NTR signaling pathways resulting in Purkinje cell pathology. We report preliminary characterization of X and Y chromosome-linked regulatory sequences upstream of SPRY3, which are polymorphic in the general population. We suggest that an OREG-annotated region on chromosome Yq12 ∼60 kb from SPRY3 acts as a silencer of Y-linked SPRY3 expression. Deletion of a β-satellite repeat, or alterations in chromatin structure in this region due to trans-acting factors, could affect the proposed silencing function, leading to reactivation and inappropriate expression of Y-linked SPRY3. This proposed male-specific mechanism could contribute to the male bias in autism prevalence.
Collapse
Affiliation(s)
| | | | | | | | - Tom Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| |
Collapse
|
12
|
Bashford AL, Subramanian V. Mice with a conditional deletion of Talpid3 (KIAA0586) - a model for Joubert syndrome. J Pathol 2019; 248:396-408. [PMID: 30924151 PMCID: PMC6767539 DOI: 10.1002/path.5271] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 03/07/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
Joubert syndrome (JS) is a ciliopathy associated with mutations in numerous genes encoding cilia components. TALPID3 encoded by KIAA0856 in man (2700049A03Rik in mouse) is a centrosomal protein essential for the assembly of primary cilia. Mutations in KIAA0856 have been recently identified in JS patients. Herein, we describe a novel mouse JS model with a conditional deletion of the conserved exons 11–12 of Talpid3 in the central nervous system which recapitulates the complete cerebellar phenotype seen in JS. Talpid3 mutant mice exhibit key hallmarks of JS including progressive ataxia, severely hypoplastic cerebellar hemispheres and vermis, together with abnormal decussation of the superior cerebellar peduncles. The Purkinje cell layer is disorganised with abnormal dendritic arborisation. The external granule layer (EGL) is thinner, lacks primary cilia, and has a reduced level of proliferation. Furthermore, we describe novel cellular defects including ectopic clusters of mature granule neurons, and abnormal parallel fibre‐derived synapses and disorientation of cells in the EGL. The defective glial scaffold results in abnormal granule cell migration which manifests as ectopic clusters of granule neurons. In addition, we show a reduction in Wnt7a expression suggesting that defects may arise not only from deficiencies in the Hedgehog (Hh) pathway but also due to the additional roles of Talpid3. The Talpid3 conditional knockout mouse is a novel JS model which fully recapitulates the JS cerebellar phenotype. These findings reveal a role for Talpid3 in granule precursor cell migration in the cerebellum (either direct or indirect) which together with defective Hh signalling underlies the JS phenotype. Our findings also illustrate the utility of creating conditional mouse models to assist in unravelling the molecular and cellular mechanisms underlying JS. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Andrew L Bashford
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| | | |
Collapse
|
13
|
Miterko LN, White JJ, Lin T, Brown AM, O'Donovan KJ, Sillitoe RV. Persistent motor dysfunction despite homeostatic rescue of cerebellar morphogenesis in the Car8 waddles mutant mouse. Neural Dev 2019; 14:6. [PMID: 30867000 PMCID: PMC6417138 DOI: 10.1186/s13064-019-0130-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
Background Purkinje cells play a central role in establishing the cerebellar circuit. Accordingly, disrupting Purkinje cell development impairs cerebellar morphogenesis and motor function. In the Car8wdl mouse model of hereditary ataxia, severe motor deficits arise despite the cerebellum overcoming initial defects in size and morphology. Methods To resolve how this compensation occurs, we asked how the loss of carbonic anhydrase 8 (CAR8), a regulator of IP3R1 Ca2+ signaling in Purkinje cells, alters cerebellar development in Car8wdl mice. Using a combination of histological, physiological, and behavioral analyses, we determined the extent to which the loss of CAR8 affects cerebellar anatomy, neuronal firing, and motor coordination during development. Results Our results reveal that granule cell proliferation is reduced in early postnatal mutants, although by the third postnatal week there is enhanced and prolonged proliferation, plus an upregulation of Sox2 expression in the inner EGL. Modified circuit patterning of Purkinje cells and Bergmann glia accompany these granule cell adjustments. We also find that although anatomy eventually normalizes, the abnormal activity of neurons and muscles persists. Conclusions Our data show that losing CAR8 only transiently restricts cerebellar growth, but permanently damages its function. These data support two current hypotheses about cerebellar development and disease: (1) Sox2 expression may be upregulated at sites of injury and contribute to the rescue of cerebellar structure and (2) transient delays to developmental processes may precede permanent motor dysfunction. Furthermore, we characterize waddles mutant mouse morphology and behavior during development and propose a Sox2-positive, cell-mediated role for rescue in a mouse model of human motor diseases. Electronic supplementary material The online version of this article (10.1186/s13064-019-0130-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lauren N Miterko
- Department of Pathology and Immunology, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA
| | - Joshua J White
- Department of Pathology and Immunology, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA.,Department of Neuroscience, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA
| | - Tao Lin
- Department of Pathology and Immunology, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA
| | - Amanda M Brown
- Department of Pathology and Immunology, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA.,Department of Neuroscience, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA.,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA
| | - Kevin J O'Donovan
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York, 10996, USA.,Burke Neurological Institute, Weill Cornell Medicine, White Plains, 10605, USA
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA. .,Department of Neuroscience, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA. .,Program in Developmental Biology, Baylor College of Medicine, Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA. .,Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX, 77030, USA.
| |
Collapse
|
14
|
|
15
|
Kurtzeborn K, Cebrian C, Kuure S. Regulation of Renal Differentiation by Trophic Factors. Front Physiol 2018; 9:1588. [PMID: 30483151 PMCID: PMC6240607 DOI: 10.3389/fphys.2018.01588] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
Classically, trophic factors are considered as proteins which support neurons in their growth, survival, and differentiation. However, most neurotrophic factors also have important functions outside of the nervous system. Especially essential renal growth and differentiation regulators are glial cell line-derived neurotrophic factor (GDNF), bone morphogenetic proteins (BMPs), and fibroblast growth factors (FGFs). Here we discuss how trophic factor-induced signaling contributes to the control of ureteric bud (UB) branching morphogenesis and to maintenance and differentiation of nephrogenic mesenchyme in embryonic kidney. The review includes recent advances in trophic factor functions during the guidance of branching morphogenesis and self-renewal versus differentiation decisions, both of which dictate the control of kidney size and nephron number. Creative utilization of current information may help better recapitulate renal differentiation in vitro, but it is obvious that significantly more basic knowledge is needed for development of regeneration-based renal therapies.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
| | - Cristina Cebrian
- Developmental Biology Division, Cincinnati Children’s Hospital, Cincinnati, OH, United States
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
- GM-Unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
16
|
Hausott B, Klimaschewski L. Sprouty2-a Novel Therapeutic Target in the Nervous System? Mol Neurobiol 2018; 56:3897-3903. [PMID: 30225774 PMCID: PMC6505497 DOI: 10.1007/s12035-018-1338-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/29/2018] [Indexed: 02/08/2023]
Abstract
Clinical trials applying growth factors to alleviate symptoms of patients with neurological disorders have largely been unsuccessful in the past. As an alternative approach, growth factor receptors or components of their signal transduction machinery may be targeted directly. In recent years, the search for intracellular signaling integrator downstream of receptor tyrosine kinases provided valuable novel substrates. Among them are the Sprouty proteins which mainly act as inhibitors of growth factor-dependent neuronal and glial signaling pathways. In this review, we summarize the role of Sprouties in the lesioned central and peripheral nervous system with particular reference to Sprouty2 that is upregulated in various experimental models of neuronal degeneration and regeneration. Increased synthesis under pathological conditions makes Sprouty2 an attractive pharmacological target to enhance intracellular signaling activities, notably the ERK pathway, in affected neurons or activated astrocytes. Interestingly, high Sprouty2 levels are also found in malignant glioma cells. We recently demonstrated that abrogating Sprouty2 function strongly inhibits intracranial tumor growth and leads to significantly prolonged survival of glioblastoma bearing mice by induction of ERK-dependent DNA replication stress. On the contrary, knockdown of Sprouty proteins increases proliferation of activated astrocytes and, consequently, reduces secondary brain damage in neuronal lesion models such as kainic acid-induced epilepsy or endothelin-induced ischemia. Furthermore, downregulation of Sprouty2 improves nerve regeneration in the lesioned peripheral nervous system. Taken together, targeting Sprouties as intracellular inhibitors of the ERK pathway holds great promise for the treatment of various neurological disorders including gliomas. Since the protein lacks enzymatic activities, it will be difficult to develop chemical compounds capable to directly and specifically modulate Sprouty functions. However, interfering with Sprouty expression by gene therapy or siRNA treatment provides a realistic approach to evaluate the therapeutic potential of indirectly stimulating ERK activities in neurological disease.
Collapse
Affiliation(s)
- Barbara Hausott
- Department of Anatomy, Histology and Embyrology, Division of Neuroanatomy, Medical University Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria
| | - Lars Klimaschewski
- Department of Anatomy, Histology and Embyrology, Division of Neuroanatomy, Medical University Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria. .,Division for Neuroanatomy, Medical University of Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria.
| |
Collapse
|
17
|
Secretome Screening Reveals Fibroblast Growth Factors as Novel Inhibitors of Viral Replication. J Virol 2018; 92:JVI.00260-18. [PMID: 29899088 DOI: 10.1128/jvi.00260-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/05/2018] [Indexed: 12/17/2022] Open
Abstract
Cellular antiviral programs can efficiently inhibit viral infection. These programs are often initiated through signaling cascades induced by secreted proteins, such as type I interferons, interleukin-6 (IL-6), or tumor necrosis factor alpha (TNF-α). In the present study, we generated an arrayed library of 756 human secreted proteins to perform a secretome screen focused on the discovery of novel modulators of viral entry and/or replication. The individual secreted proteins were tested for the capacity to inhibit infection by two replication-competent recombinant vesicular stomatitis viruses (VSVs) with distinct glycoproteins utilizing different entry pathways. Fibroblast growth factor 16 (FGF16) was identified and confirmed as the most prominent novel inhibitor of both VSVs and therefore of viral replication, not entry. Importantly, an antiviral interferon signature was completely absent in FGF16-treated cells. Nevertheless, the antiviral effect of FGF16 is broad, as it was evident on multiple cell types and also on infection by coxsackievirus. In addition, other members of the FGF family also inhibited viral infection. Thus, our unbiased secretome screen revealed a novel protein family capable of inducing a cellular antiviral state. This previously unappreciated role of the FGF family may have implications for the development of new antivirals and the efficacy of oncolytic virus therapy.IMPORTANCE Viruses infect human cells in order to replicate, while human cells aim to resist infection. Several cellular antiviral programs have therefore evolved to resist infection. Knowledge of these programs is essential for the design of antiviral therapeutics in the future. The induction of antiviral programs is often initiated by secreted proteins, such as interferons. We hypothesized that other secreted proteins may also promote resistance to viral infection. Thus, we tested 756 human secreted proteins for the capacity to inhibit two pseudotypes of vesicular stomatitis virus (VSV). In this secretome screen on viral infection, we identified fibroblast growth factor 16 (FGF16) as a novel antiviral against multiple VSV pseudotypes as well as coxsackievirus. Subsequent testing of other FGF family members revealed that FGF signaling generally inhibits viral infection. This finding may lead to the development of new antivirals and may also be applicable for enhancing oncolytic virus therapy.
Collapse
|
18
|
Morgani SM, Saiz N, Garg V, Raina D, Simon CS, Kang M, Arias AM, Nichols J, Schröter C, Hadjantonakis AK. A Sprouty4 reporter to monitor FGF/ERK signaling activity in ESCs and mice. Dev Biol 2018; 441:104-126. [PMID: 29964027 DOI: 10.1016/j.ydbio.2018.06.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 06/25/2018] [Accepted: 06/25/2018] [Indexed: 12/31/2022]
Abstract
The FGF/ERK signaling pathway is highly conserved throughout evolution and plays fundamental roles during embryonic development and in adult organisms. While a plethora of expression data exists for ligands, receptors and pathway regulators, we know little about the spatial organization or dynamics of signaling in individual cells within populations. To this end we developed a transcriptional readout of FGF/ERK activity by targeting a histone H2B-linked Venus fluorophore to the endogenous locus of Spry4, an early pathway target, and generated Spry4H2B-Venus embryonic stem cells (ESCs) and a derivative mouse line. The Spry4H2B-Venus reporter was heterogeneously expressed within ESC cultures and responded to FGF/ERK signaling manipulation. In vivo, the Spry4H2B-Venus reporter recapitulated the expression pattern of Spry4 and localized to sites of known FGF/ERK activity including the inner cell mass of the pre-implantation embryo and the limb buds, somites and isthmus of the post-implantation embryo. Additionally, we observed highly localized reporter expression within adult organs. Genetic and chemical disruption of FGF/ERK signaling, in vivo in pre- and post-implantation embryos, abrogated Venus expression establishing the reporter as an accurate signaling readout. This tool will provide new insights into the dynamics of the FGF/ERK signaling pathway during mammalian development.
Collapse
Affiliation(s)
- Sophie M Morgani
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Nestor Saiz
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vidur Garg
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Dhruv Raina
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Claire S Simon
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Minjung Kang
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | | | - Jennifer Nichols
- Wellcome Trust-Medical Research Council Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Christian Schröter
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA.
| |
Collapse
|
19
|
Schilling K. Moving into shape: cell migration during the development and histogenesis of the cerebellum. Histochem Cell Biol 2018; 150:13-36. [DOI: 10.1007/s00418-018-1677-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2018] [Indexed: 12/31/2022]
|
20
|
Company V, Moreno-Bravo JA, Perez-Balaguer A, Puelles E. The Amniote Oculomotor Complex. Anat Rec (Hoboken) 2018; 302:446-451. [DOI: 10.1002/ar.23827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Verónica Company
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC; Sant Joan d'Alacant, Alicante 03550 Spain
| | - Juan Antonio Moreno-Bravo
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC; Sant Joan d'Alacant, Alicante 03550 Spain
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Institut de la Vision; 17 Rue Moreau, Paris 75012 France
| | - Ariadna Perez-Balaguer
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC; Sant Joan d'Alacant, Alicante 03550 Spain
| | - Eduardo Puelles
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC; Sant Joan d'Alacant, Alicante 03550 Spain
| |
Collapse
|
21
|
Donovan APA, Yu T, Ellegood J, Riegman KLH, de Geus C, van Ravenswaaij-Arts C, Fernandes C, Lerch JP, Basson MA. Cerebellar Vermis and Midbrain Hypoplasia Upon Conditional Deletion of Chd7 from the Embryonic Mid-Hindbrain Region. Front Neuroanat 2017; 11:86. [PMID: 29046629 PMCID: PMC5632662 DOI: 10.3389/fnana.2017.00086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/14/2017] [Indexed: 01/24/2023] Open
Abstract
Reduced fibroblast growth factor (FGF) signaling from the mid-hindbrain or isthmus organizer (IsO) during early embryonic development results in hypoplasia of the midbrain and cerebellar vermis. We previously reported evidence for reduced Fgf8 expression and FGF signaling in the mid-hindbrain region of embryos heterozygous for Chd7, the gene mutated in CHARGE (Coloboma, Heart defects, choanal Atresia, Retarded growth and development, Genitourinary anomalies and Ear defects) syndrome. However, Chd7+/- animals only exhibit mild cerebellar vermis anomalies. As homozygous deletion of Chd7 is embryonic lethal, we conditionally deleted Chd7 from the early embryonic mid-hindbrain region to identify the function of CHD7 in mid-hindbrain development. Using a combination of high resolution structural MRI and histology, we report striking midbrain and cerebellar vermis hypoplasia in the homozygous conditional mutants. We show that cerebellar vermis hypoplasia is associated with reduced embryonic Fgf8 expression and an expanded roof plate in rhombomere 1 (r1). These findings identify an essential role for Chd7 in regulating mid-hindbrain development via Fgf8.
Collapse
Affiliation(s)
- Alex P A Donovan
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Tian Yu
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Jacob Ellegood
- Department of Medical Biophysics, University of Toronto, Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada
| | - Kimberley L H Riegman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Christa de Geus
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Conny van Ravenswaaij-Arts
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Cathy Fernandes
- MRC Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Jason P Lerch
- Department of Medical Biophysics, University of Toronto, Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - M Albert Basson
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| |
Collapse
|
22
|
Scheuer T, Sharkovska Y, Tarabykin V, Marggraf K, Brockmöller V, Bührer C, Endesfelder S, Schmitz T. Neonatal Hyperoxia Perturbs Neuronal Development in the Cerebellum. Mol Neurobiol 2017; 55:3901-3915. [PMID: 28547531 DOI: 10.1007/s12035-017-0612-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/11/2017] [Indexed: 12/22/2022]
Abstract
Impaired postnatal brain development of preterm infants often results in neurological deficits. Besides pathologies of the forebrain, maldeveolopment of the cerebellum is increasingly recognized to contribute to psychomotor impairments of many former preterm infants. However, causes are poorly defined. We used a hyperoxia model to define neonatal damage in cerebellar granule cell precursors (GCPs) and in Purkinje cells (PCs) known to be essential for interaction with GCPs during development. We exposed newborn rats to 24 h 80% O2 from age P6 to P7 to identify postnatal and long-term damage in cerebellar GCPs at age P7 after hyperoxia and also after recovery in room air thereafter until P11 and P30. We determined proliferation and apoptosis of GCPs and immature neurons by immunohistochemistry, quantified neuronal damage by qPCR and Western blots for neuronal markers, and measured dendrite outgrowth of PCs by CALB1 immunostainings and by Sholl analysis of Golgi stainings. After hyperoxia, proliferation of PAX6+ GCPs was decreased at P7, while DCX + CASP3+ cells were increased at P11. Neuronal markers Pax6, Tbr2, and Prox1 were downregulated at P11 and P30. Neuronal damage was confirmed by reduced NeuN protein expression at P30. Sonic hedgehog (SHH) was significantly decreased at P7 and P11 after hyperoxia and coincided with lower CyclinD2 and Hes1 expression at P7. The granule cell injury was accompanied by hampered PC maturation with delayed dendrite formation and impaired branching. Neonatal injury induced by hyperoxia inhibits PC functioning and impairs granule cell development. As a conclusion, maldevelopment of the cerebellar neurons found in preterm infants could be caused by postnatal oxygen toxicity.
Collapse
Affiliation(s)
- Till Scheuer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany. .,Institute of Bioanalytics, Technische Universität Berlin, 13355, Berlin, Germany. .,Klinik für Neonatologie, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Yuliya Sharkovska
- Department for Neonatology, Charité University Medical Center, Berlin, Germany.,Institute for Cell and Neurobiology, Center for Anatomy, Charité University Medical Center, Berlin, Germany
| | - Victor Tarabykin
- Institute for Cell and Neurobiology, Center for Anatomy, Charité University Medical Center, Berlin, Germany
| | - Katharina Marggraf
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Vivien Brockmöller
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Christoph Bührer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | | | - Thomas Schmitz
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| |
Collapse
|
23
|
Zahir FR, Mwenifumbo JC, Chun HJE, Lim EL, Van Karnebeek CDM, Couse M, Mungall KL, Lee L, Makela N, Armstrong L, Boerkoel CF, Langlois SL, McGillivray BM, Jones SJM, Friedman JM, Marra MA. Comprehensive whole genome sequence analyses yields novel genetic and structural insights for Intellectual Disability. BMC Genomics 2017; 18:403. [PMID: 28539120 PMCID: PMC5442678 DOI: 10.1186/s12864-017-3671-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/29/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Intellectual Disability (ID) is among the most common global disorders, yet etiology is unknown in ~30% of patients despite clinical assessment. Whole genome sequencing (WGS) is able to interrogate the entire genome, providing potential to diagnose idiopathic patients. METHODS We conducted WGS on eight children with idiopathic ID and brain structural defects, and their normal parents; carrying out an extensive data analyses, using standard and discovery approaches. RESULTS We verified de novo pathogenic single nucleotide variants (SNV) in ARID1B c.1595delG and PHF6 c.820C > T, potentially causative de novo two base indels in SQSTM1 c.115_116delinsTA and UPF1 c.1576_1577delinsA, and de novo SNVs in CACNB3 c.1289G > A, and SPRY4 c.508 T > A, of uncertain significance. We report results from a large secondary control study of 2081 exomes probing the pathogenicity of the above genes. We analyzed structural variation by four different algorithms including de novo genome assembly. We confirmed a likely contributory 165 kb de novo heterozygous 1q43 microdeletion missed by clinical microarray. The de novo assembly resulted in unmasking hidden genome instability that was missed by standard re-alignment based algorithms. We also interrogated regulatory sequence variation for known and hypothesized ID genes and present useful strategies for WGS data analyses for non-coding variation. CONCLUSION This study provides an extensive analysis of WGS in the context of ID, providing genetic and structural insights into ID and yielding diagnoses.
Collapse
Affiliation(s)
- Farah R Zahir
- Canada's Michael Smith Genome Sciences Center, Vancouver, BC, V5Z 4S6, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada. .,Qatar Biomedical Research Institute, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar.
| | - Jill C Mwenifumbo
- Canada's Michael Smith Genome Sciences Center, Vancouver, BC, V5Z 4S6, Canada
| | - Hye-Jung E Chun
- Canada's Michael Smith Genome Sciences Center, Vancouver, BC, V5Z 4S6, Canada
| | - Emilia L Lim
- Canada's Michael Smith Genome Sciences Center, Vancouver, BC, V5Z 4S6, Canada
| | - Clara D M Van Karnebeek
- Department of Pediatrics, Centre for Molecular Medicine & Therapeutics Child & Family Research Institute, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Madeline Couse
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Center, Vancouver, BC, V5Z 4S6, Canada
| | - Leora Lee
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Nancy Makela
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Linlea Armstrong
- Provincial Medical Genetics Programme, Children's & Women's Health Centre of British Columbia, Vancouver, BC, V6H 3N1, Canada
| | - Cornelius F Boerkoel
- Provincial Medical Genetics Programme, Children's & Women's Health Centre of British Columbia, Vancouver, BC, V6H 3N1, Canada
| | - Sylvie L Langlois
- Provincial Medical Genetics Programme, Children's & Women's Health Centre of British Columbia, Vancouver, BC, V6H 3N1, Canada
| | - Barbara M McGillivray
- Provincial Medical Genetics Programme, Children's & Women's Health Centre of British Columbia, Vancouver, BC, V6H 3N1, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Center, Vancouver, BC, V5Z 4S6, Canada
| | - Jan M Friedman
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Center, Vancouver, BC, V5Z 4S6, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
24
|
Hibi M, Matsuda K, Takeuchi M, Shimizu T, Murakami Y. Evolutionary mechanisms that generate morphology and neural-circuit diversity of the cerebellum. Dev Growth Differ 2017; 59:228-243. [DOI: 10.1111/dgd.12349] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 01/10/2023]
Affiliation(s)
- Masahiko Hibi
- Bioscience and Biotechnology Center; Nagoya University; Nagoya 464-8601 Japan
- Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Koji Matsuda
- Bioscience and Biotechnology Center; Nagoya University; Nagoya 464-8601 Japan
- Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Miki Takeuchi
- Bioscience and Biotechnology Center; Nagoya University; Nagoya 464-8601 Japan
| | - Takashi Shimizu
- Bioscience and Biotechnology Center; Nagoya University; Nagoya 464-8601 Japan
- Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Yasunori Murakami
- Graduate School of Science and Engineering; Ehime University; Matsuyama 790-8577 Japan
| |
Collapse
|
25
|
Whittaker DE, Riegman KL, Kasah S, Mohan C, Yu T, Sala BP, Hebaishi H, Caruso A, Marques AC, Michetti C, Smachetti MES, Shah A, Sabbioni M, Kulhanci O, Tee WW, Reinberg D, Scattoni ML, Volk H, McGonnell I, Wardle FC, Fernandes C, Basson MA. The chromatin remodeling factor CHD7 controls cerebellar development by regulating reelin expression. J Clin Invest 2017; 127:874-887. [PMID: 28165338 PMCID: PMC5330721 DOI: 10.1172/jci83408] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
The mechanisms underlying the neurodevelopmental deficits associated with CHARGE syndrome, which include cerebellar hypoplasia, developmental delay, coordination problems, and autistic features, have not been identified. CHARGE syndrome has been associated with mutations in the gene encoding the ATP-dependent chromatin remodeler CHD7. CHD7 is expressed in neural stem and progenitor cells, but its role in neurogenesis during brain development remains unknown. Here we have shown that deletion of Chd7 from cerebellar granule cell progenitors (GCps) results in reduced GCp proliferation, cerebellar hypoplasia, developmental delay, and motor deficits in mice. Genome-wide expression profiling revealed downregulated expression of the gene encoding the glycoprotein reelin (Reln) in Chd7-deficient GCps. Recessive RELN mutations have been associated with severe cerebellar hypoplasia in humans. We found molecular and genetic evidence that reductions in Reln expression contribute to GCp proliferative defects and cerebellar hypoplasia in GCp-specific Chd7 mouse mutants. Finally, we showed that CHD7 is necessary for maintaining an open, accessible chromatin state at the Reln locus. Taken together, this study shows that Reln gene expression is regulated by chromatin remodeling, identifies CHD7 as a previously unrecognized upstream regulator of Reln, and provides direct in vivo evidence that a mammalian CHD protein can control brain development by modulating chromatin accessibility in neuronal progenitors.
Collapse
Affiliation(s)
- Danielle E. Whittaker
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
- Department of Comparative Biomedical Sciences, Royal Veterinary College, and
| | - Kimberley L.H. Riegman
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Sahrunizam Kasah
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Conor Mohan
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Tian Yu
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Blanca Pijuan Sala
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Husam Hebaishi
- King’s College London, Randall Division, New Hunt’s House, London, United Kingdom
| | - Angela Caruso
- Neurotoxicology and Neuroendocrinology Section, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, and
- School of Behavioural Neuroscience, Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Ana Claudia Marques
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Caterina Michetti
- Neurotoxicology and Neuroendocrinology Section, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, and
- Department of Physiology and Pharmacology “V. Erspamer,” Sapienza University of Rome, Rome, Italy
| | | | - Apar Shah
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
| | - Mara Sabbioni
- Neurotoxicology and Neuroendocrinology Section, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, and
| | - Omer Kulhanci
- MRC Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Wee-Wei Tee
- Howard Hughes Medical Institute, Department of Molecular Pharmacology and Biochemistry, New York University School of Medicine, New York, New York, USA
| | - Danny Reinberg
- Howard Hughes Medical Institute, Department of Molecular Pharmacology and Biochemistry, New York University School of Medicine, New York, New York, USA
| | - Maria Luisa Scattoni
- Neurotoxicology and Neuroendocrinology Section, Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, and
| | - Holger Volk
- Department of Comparative Biomedical Sciences, Royal Veterinary College, and
| | - Imelda McGonnell
- Department of Comparative Biomedical Sciences, Royal Veterinary College, and
| | - Fiona C. Wardle
- King’s College London, Randall Division, New Hunt’s House, London, United Kingdom
| | - Cathy Fernandes
- MRC Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- King’s College London, MRC Centre for Neurodevelopmental Disorders, New Hunt’s House, London, United Kingdom
| | - M. Albert Basson
- King’s College London, Department of Craniofacial Development and Stem Cell Biology, Guy’s Hospital Tower Wing
- King’s College London, MRC Centre for Neurodevelopmental Disorders, New Hunt’s House, London, United Kingdom
| |
Collapse
|
26
|
Mitra I, Lavillaureix A, Yeh E, Traglia M, Tsang K, Bearden CE, Rauen KA, Weiss LA. Reverse Pathway Genetic Approach Identifies Epistasis in Autism Spectrum Disorders. PLoS Genet 2017; 13:e1006516. [PMID: 28076348 PMCID: PMC5226683 DOI: 10.1371/journal.pgen.1006516] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/01/2016] [Indexed: 02/08/2023] Open
Abstract
Although gene-gene interaction, or epistasis, plays a large role in complex traits in model organisms, genome-wide by genome-wide searches for two-way interaction have limited power in human studies. We thus used knowledge of a biological pathway in order to identify a contribution of epistasis to autism spectrum disorders (ASDs) in humans, a reverse-pathway genetic approach. Based on previous observation of increased ASD symptoms in Mendelian disorders of the Ras/MAPK pathway (RASopathies), we showed that common SNPs in RASopathy genes show enrichment for association signal in GWAS (P = 0.02). We then screened genome-wide for interactors with RASopathy gene SNPs and showed strong enrichment in ASD-affected individuals (P < 2.2 x 10-16), with a number of pairwise interactions meeting genome-wide criteria for significance. Finally, we utilized quantitative measures of ASD symptoms in RASopathy-affected individuals to perform modifier mapping via GWAS. One top region overlapped between these independent approaches, and we showed dysregulation of a gene in this region, GPR141, in a RASopathy neural cell line. We thus used orthogonal approaches to provide strong evidence for a contribution of epistasis to ASDs, confirm a role for the Ras/MAPK pathway in idiopathic ASDs, and to identify a convergent candidate gene that may interact with the Ras/MAPK pathway.
Collapse
Affiliation(s)
- Ileena Mitra
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Alinoë Lavillaureix
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Université Paris Descartes, Sorbonne Paris Cité, Faculty of Medicine, Paris, France
| | - Erika Yeh
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Michela Traglia
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Kathryn Tsang
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Psychology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Katherine A. Rauen
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Department of Pediatrics, School of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Lauren A. Weiss
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
27
|
Abstract
Autism Spectrum Disorders (ASDs) are a heterogeneous group of neurodevelopmental disorders that are diagnosed solely on the basis of behaviour. A large body of work has reported neuroanatomical differences between individuals with ASD and neurotypical controls. Despite the huge clinical and genetic heterogeneity that typifies autism, some of these anatomical features appear to be either present in most cases or so dramatically altered in some that their presence is now reasonably well replicated in a number of studies. One such finding is the tendency towards overgrowth of the frontal cortex during the early postnatal period. Although these reports have been focused primarily on the presumed pathological anatomy, they are providing us with important insights into normal brain anatomy and are stimulating new ideas and hypotheses about the normal trajectory of brain development and the function of specific anatomical brain structures. The use of model systems that include genetic model organisms such as the mouse and, more recently, human induced pluripotent stem cell-derived brain organoids to model normal and pathological human cortical development, is proving particularly informative. Here we review some of the neuroanatomical alterations reported in autism, with a particular focus on well-validated findings and recent advances in the field, and ask what these observations can tell us about normal and abnormal brain development.
Collapse
Affiliation(s)
- Alex P. A. Donovan
- Department of Craniofacial Development and Stem Cell Biology, and MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
| | - M. Albert Basson
- Department of Craniofacial Development and Stem Cell Biology, and MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
| |
Collapse
|
28
|
Ning Z, McLellan AS, Ball M, Wynne F, O'Neill C, Mills W, Quinn JP, Kleinjan DA, Anney RJ, Carmody RJ, O'Keeffe G, Moore T. Regulation of SPRY3 by X chromosome and PAR2-linked promoters in an autism susceptibility region. Hum Mol Genet 2015; 24:5126-41. [PMID: 26089202 DOI: 10.1093/hmg/ddv231] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/15/2015] [Indexed: 11/13/2022] Open
Abstract
Sprouty proteins are regulators of cell growth and branching morphogenesis. Unlike mouse Spry3, which is X-linked, human SPRY3 maps to the pseudoautosomal region 2; however, the human Y-linked allele is not expressed due to epigenetic silencing by an unknown mechanism. SPRY3 maps adjacent to X-linked Trimethyllysine hydroxylase epsilon (TMLHE), recently identified as an autism susceptibility gene. We report that Spry3 is highly expressed in central and peripheral nervous system ganglion cells in mouse and human, including cerebellar Purkinje cells and retinal ganglion cells. Transient over-expression or knockdown of Spry3 in cultured mouse superior cervical ganglion cells inhibits and promotes, respectively, neurite growth and branching. A 0.7 kb gene fragment spanning the human SPRY3 transcriptional start site recapitulates the endogenous Spry3-expression pattern in LacZ reporter mice. In the human and mouse the SPRY3 promoter contains an AG-rich repeat and we found co-expression, and promoter binding and/or regulation of SPRY3 expression by transcription factors MAZ, EGR1, ZNF263 and PAX6. We identified eight alleles of the human SPRY3 promoter repeat in Caucasians, and similar allele frequencies in autism families. We characterized multiple SPRY3 transcripts originating at two CpG islands in the X-linked F8A3-TMLHE region, suggesting X chromosome regulation of SPRY3. These findings provide an explanation for differential regulation of X and Y-linked SPRY3 alleles. In addition, the presence of a SPRY3 transcript exon in a previously described X chromosome deletion associated with autism, and the cerebellar interlobular variation in Spry3 expression coincident with the reported pattern of Purkinje cell loss in autism, suggest SPRY3 as a candidate susceptibility locus for autism.
Collapse
Affiliation(s)
- Zhenfei Ning
- School of Biochemistry and Cell Biology, University College Cork, Western Gateway Building, Western Road, Cork, Ireland
| | - Andrew S McLellan
- School of Biochemistry and Cell Biology, University College Cork, Western Gateway Building, Western Road, Cork, Ireland
| | - Melanie Ball
- School of Biochemistry and Cell Biology, University College Cork, Western Gateway Building, Western Road, Cork, Ireland
| | - Freda Wynne
- School of Biochemistry and Cell Biology, University College Cork, Western Gateway Building, Western Road, Cork, Ireland
| | - Cora O'Neill
- School of Biochemistry and Cell Biology, University College Cork, Western Gateway Building, Western Road, Cork, Ireland
| | - Walter Mills
- School of Biochemistry and Cell Biology, University College Cork, Western Gateway Building, Western Road, Cork, Ireland
| | - John P Quinn
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, UK
| | - Dirk A Kleinjan
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Richard J Anney
- Department of Psychiatry, Neuropsychiatric Genetics Research Group, Institute of Molecular Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Ruaidhre J Carmody
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Sir Graeme Davies Building, Glasgow G12 8TA, UK and
| | - Gerard O'Keeffe
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, College Road, Cork, Ireland
| | - Tom Moore
- School of Biochemistry and Cell Biology, University College Cork, Western Gateway Building, Western Road, Cork, Ireland,
| |
Collapse
|
29
|
Knosp WM, Knox SM, Lombaert IMA, Haddox CL, Patel VN, Hoffman MP. Submandibular parasympathetic gangliogenesis requires sprouty-dependent Wnt signals from epithelial progenitors. Dev Cell 2015; 32:667-77. [PMID: 25805134 DOI: 10.1016/j.devcel.2015.01.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/21/2014] [Accepted: 01/21/2015] [Indexed: 10/23/2022]
Abstract
Parasympathetic innervation is critical for submandibular gland (SMG) development and regeneration. Parasympathetic ganglia (PSG) are derived from Schwann cell precursors that migrate along nerves, differentiate into neurons, and coalesce within their target tissue to form ganglia. However, signals that initiate gangliogenesis after the precursors differentiate into neurons are unknown. We found that deleting negative regulators of FGF signaling, Sprouty1 and Sprouty2 (Spry1/2DKO), resulted in a striking loss of gangliogenesis, innervation, and keratin 5-positive (K5+) epithelial progenitors in the SMG. Here we identify Wnts produced by K5+ progenitors in the SMG as key mediators of gangliogenesis. Wnt signaling increases survival and proliferation of PSG neurons, and inhibiting Wnt signaling disrupts gangliogenesis and organ innervation. Activating Wnt signaling and reducing FGF gene dosage rescues gangliogenesis and innervation in both the Spry1/2DKO SMG and pancreas. Thus, K5+ progenitors produce Wnt signals to establish the PSG-epithelial communication required for organ innervation and progenitor cell maintenance.
Collapse
Affiliation(s)
- Wendy M Knosp
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Sarah M Knox
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA; Department of Cell and Tissue Biology, UCSF, San Francisco, CA 94143, USA
| | | | - Candace L Haddox
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Vaishali N Patel
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, NIDCR, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2015; 4:215-66. [PMID: 25772309 PMCID: PMC4393358 DOI: 10.1002/wdev.176] [Citation(s) in RCA: 1457] [Impact Index Per Article: 145.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/23/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
The signaling component of the mammalian Fibroblast Growth Factor (FGF) family is comprised of eighteen secreted proteins that interact with four signaling tyrosine kinase FGF receptors (FGFRs). Interaction of FGF ligands with their signaling receptors is regulated by protein or proteoglycan cofactors and by extracellular binding proteins. Activated FGFRs phosphorylate specific tyrosine residues that mediate interaction with cytosolic adaptor proteins and the RAS-MAPK, PI3K-AKT, PLCγ, and STAT intracellular signaling pathways. Four structurally related intracellular non-signaling FGFs interact with and regulate the family of voltage gated sodium channels. Members of the FGF family function in the earliest stages of embryonic development and during organogenesis to maintain progenitor cells and mediate their growth, differentiation, survival, and patterning. FGFs also have roles in adult tissues where they mediate metabolic functions, tissue repair, and regeneration, often by reactivating developmental signaling pathways. Consistent with the presence of FGFs in almost all tissues and organs, aberrant activity of the pathway is associated with developmental defects that disrupt organogenesis, impair the response to injury, and result in metabolic disorders, and cancer. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of MedicineSt. Louis, MO, USA
- *
Correspondence to:
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto UniversitySakyo, Kyoto, Japan
| |
Collapse
|
31
|
Butts T, Green MJ, Wingate RJT. Development of the cerebellum: simple steps to make a 'little brain'. Development 2014; 141:4031-41. [PMID: 25336734 DOI: 10.1242/dev.106559] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cerebellum is a pre-eminent model for the study of neurogenesis and circuit assembly. Increasing interest in the cerebellum as a participant in higher cognitive processes and as a locus for a range of disorders and diseases make this simple yet elusive structure an important model in a number of fields. In recent years, our understanding of some of the more familiar aspects of cerebellar growth, such as its territorial allocation and the origin of its various cell types, has undergone major recalibration. Furthermore, owing to its stereotyped circuitry across a range of species, insights from a variety of species have contributed to an increasingly rich picture of how this system develops. Here, we review these recent advances and explore three distinct aspects of cerebellar development - allocation of the cerebellar anlage, the significance of transit amplification and the generation of neuronal diversity - each defined by distinct regulatory mechanisms and each with special significance for health and disease.
Collapse
Affiliation(s)
- Thomas Butts
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, UK
| | - Mary J Green
- National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Richard J T Wingate
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK
| |
Collapse
|
32
|
Kim E, Wang Y, Kim SJ, Bornhorst M, Jecrois ES, Anthony TE, Wang C, Li YE, Guan JL, Murphy GG, Zhu Y. Transient inhibition of the ERK pathway prevents cerebellar developmental defects and improves long-term motor functions in murine models of neurofibromatosis type 1. eLife 2014; 3. [PMID: 25535838 PMCID: PMC4297949 DOI: 10.7554/elife.05151] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/22/2014] [Indexed: 12/14/2022] Open
Abstract
Individuals with neurofibromatosis type 1 (NF1) frequently exhibit cognitive and motor impairments and characteristics of autism. The cerebellum plays a critical role in motor control, cognition, and social interaction, suggesting that cerebellar defects likely contribute to NF1-associated neurodevelopmental disorders. Here we show that Nf1 inactivation during early, but not late stages of cerebellar development, disrupts neuronal lamination, which is partially caused by overproduction of glia and subsequent disruption of the Bergmann glia (BG) scaffold. Specific Nf1 inactivation in glutamatergic neuronal precursors causes premature differentiation of granule cell (GC) precursors and ectopic production of unipolar brush cells (UBCs), indirectly disrupting neuronal migration. Transient MEK inhibition during a neonatal window prevents cerebellar developmental defects and improves long-term motor performance of Nf1-deficient mice. This study reveals essential roles of Nf1 in GC/UBC migration by generating correct numbers of glia and controlling GC/UBC fate-specification/differentiation, identifying a therapeutic prevention strategy for multiple NF1-associcated developmental abnormalities. DOI:http://dx.doi.org/10.7554/eLife.05151.001 Neurofibromatosis type 1 is a condition characterized by the growth of tumors along the nerves of the body. It is caused by mutations in a gene called NF1, which codes for a protein that normally works to inhibit the activity of another protein called Ras. In healthy cells, Ras is needed to stimulate the cells to grow and divide. However, if the Ras protein is not turned off at the right time or if it is activated at the wrong time, it can force cells to keep growing and dividing; this leads to the growth of tumors. Along with being prone to developing cancer, individuals with neurofibromatosis type 1 also develop a range of neurodevelopmental disorders that alter their learning, motor skills and social interactions. Some also exhibit behaviors that are associated with autism. This led Kim, Wang et al. to investigate whether a region of the brain—called the cerebellum—that has recently been associated with autism is also affected in a mouse model of neurofibromatosis type 1. The cerebellum is best known for its role in coordinating movement, although it also has functions in cognition, behavior and other processes. Ras is involved in the development of the cerebellum; and so Kim, Wang et al. asked whether the loss of the Nf1 gene from cells in the mouse cerebellum might cause the neurodevelopmental defects associated with neurofibromatosis type 1. Loss of Nf1 during early (but not in late) development of the cerebellum disrupted the normal organization of the nerve cells (or neurons) into specific cell layers. These defects were caused, in part, by the over-growth of a type of supporting cell—called glia cells—at a specific developmental stage—that would normally form a scaffold to help neurons migrate to their correct position. Nf1 also controls the generation of the correct types of neurons in the right time and at right location during the early development of the cerebellum. Next, Kim, Wang et al. treated newborn mice with a compound that inhibits Ras signaling via their mother's milk for 3 weeks. In mice with an inactive Nf1 gene, the treatment helped to prevent some defects in the cerebellum and the mice had improved motor coordination several months later. Whether this could form the basis of a preventative treatment for neurodevelopmental disorders associated with neurofibromatosis type 1 in humans remains a question for future work. DOI:http://dx.doi.org/10.7554/eLife.05151.002
Collapse
Affiliation(s)
- Edward Kim
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| | - Yuan Wang
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| | - Sun-Jung Kim
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| | - Miriam Bornhorst
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| | - Emmanuelle S Jecrois
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| | - Todd E Anthony
- Laboratory of Molecular Biology, Rockefeller University, New York, United States
| | - Chenran Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, United States
| | - Yi E Li
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| | - Jun-Lin Guan
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| | - Geoffrey G Murphy
- Molecular and Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, United States
| | - Yuan Zhu
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|
33
|
Donnison M, Broadhurst R, Pfeffer PL. Elf5 and Ets2 maintain the mouse extraembryonic ectoderm in a dosage dependent synergistic manner. Dev Biol 2014; 397:77-88. [PMID: 25446535 DOI: 10.1016/j.ydbio.2014.10.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/08/2014] [Accepted: 10/14/2014] [Indexed: 01/14/2023]
Abstract
The ETS superfamily transcription factors Elf5 and Ets2 have both been implicated in the maintenance of the extraembryonic ectoderm (ExE) of the mouse embryo. While homozygous mutants of either gene result in various degrees of ExE tissue loss, heterozygotes are without phenotype. We show here that compound heterozygous mutants exhibit a phenotype intermediate to that of the more severe Elf5-/- and the milder Ets2-/- mutants. Functional redundancy is shown via commonalities in expression patterns, in target gene expression, and by partial rescue of Elf5-/- mutants through overexpressing Ets2 in an Elf5-like fashion. A model is presented suggesting the functional division of the ExE region into a proximal and distal domain based on gene expression patterns and the proximal to distal increasing sensitivity to threshold levels of combined Elf5 and Ets2 activity.
Collapse
Affiliation(s)
- Martyn Donnison
- AgResearch Ruakura, 10 Bisley Road, Hamilton 3214, New Zealand
| | - Ric Broadhurst
- AgResearch Ruakura, 10 Bisley Road, Hamilton 3214, New Zealand
| | - Peter L Pfeffer
- AgResearch Ruakura, 10 Bisley Road, Hamilton 3214, New Zealand; School of Biological Sciences, Victoria University of Wellington, PO Box 600, Wellington 6140, New Zealand.
| |
Collapse
|
34
|
Gazzellone MJ, Zhou X, Lionel AC, Uddin M, Thiruvahindrapuram B, Liang S, Sun C, Wang J, Zou M, Tammimies K, Walker S, Selvanayagam T, Wei J, Wang Z, Wu L, Scherer SW. Copy number variation in Han Chinese individuals with autism spectrum disorder. J Neurodev Disord 2014; 6:34. [PMID: 25170348 PMCID: PMC4147384 DOI: 10.1186/1866-1955-6-34] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/13/2014] [Indexed: 12/11/2022] Open
Abstract
Background Autism spectrum disorders (ASDs) are a group of neurodevelopmental conditions with a demonstrated genetic etiology. Rare (<1% frequency) copy number variations (CNVs) account for a proportion of the genetic events involved, but the contribution of these events in non-European ASD populations has not been well studied. Here, we report on rare CNVs detected in a cohort of individuals with ASD of Han Chinese background. Methods DNA samples were obtained from 104 ASD probands and their parents who were recruited from Harbin, China. Samples were genotyped on the Affymetrix CytoScan HD platform. Rare CNVs were identified by comparing data with 873 technology-matched controls from Ontario and 1,235 additional population controls of Han Chinese ethnicity. Results Of the probands, 8.6% had at least 1 de novo CNV (overlapping the GIGYF2, SPRY1, 16p13.3, 16p11.2, 17p13.3-17p13.2, DMD, and NAP1L6 genes/loci). Rare inherited CNVs affected other plausible neurodevelopmental candidate genes including GRID2, LINGO2, and SLC39A12. A 24-kb duplication was also identified at YWHAE, a gene previously implicated in ASD and other developmental disorders. This duplication is observed at a similar frequency in cases and in population controls and is likely a benign Asian-specific copy number polymorphism. Conclusions Our findings help define genomic features relevant to ASD in the Han Chinese and emphasize the importance of using ancestry-matched controls in medical genetic interpretations.
Collapse
Affiliation(s)
- Matthew J Gazzellone
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto Ontario M5S 1A1, Canada
| | - Xue Zhou
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada.,Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin, Heilongjiang 150086, People's Republic of China.,Heilongjiang Provincial Centre for Disease Control and Prevention, Harbin, Heilongjiang 150030, People's Republic of China
| | - Anath C Lionel
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto Ontario M5S 1A1, Canada
| | - Mohammed Uddin
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada
| | - Bhooma Thiruvahindrapuram
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada
| | - Shuang Liang
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin, Heilongjiang 150086, People's Republic of China
| | - Caihong Sun
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin, Heilongjiang 150086, People's Republic of China
| | - Jia Wang
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin, Heilongjiang 150086, People's Republic of China
| | - Mingyang Zou
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin, Heilongjiang 150086, People's Republic of China
| | - Kristiina Tammimies
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada.,Center of Neurodevelopmental Disorders, Department of Women's and Children's Health, Karolinska Institutet, Stockholm 113 30, Sweden
| | - Susan Walker
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada
| | - Thanuja Selvanayagam
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada
| | - John Wei
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada
| | - Zhuozhi Wang
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada
| | - Lijie Wu
- Department of Children's and Adolescent Health, Public Health College of Harbin Medical University, Harbin, Heilongjiang 150086, People's Republic of China
| | - Stephen W Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Peter Gilgan Centre for Research and Learning, Room 139800, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto Ontario M5S 1A1, Canada
| |
Collapse
|
35
|
Marvaldi L, Thongrong S, Kozłowska A, Irschick R, Pritz CO, Bäumer B, Ronchi G, Geuna S, Hausott B, Klimaschewski L. Enhanced axon outgrowth and improved long-distance axon regeneration in sprouty2 deficient mice. Dev Neurobiol 2014; 75:217-31. [PMID: 25104556 DOI: 10.1002/dneu.22224] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 07/16/2014] [Accepted: 08/06/2014] [Indexed: 12/31/2022]
Abstract
Sprouty (Spry) proteins are negative feedback inhibitors of receptor tyrosine kinase signaling. Downregulation of Spry2 has been demonstrated to promote elongative axon growth of cultured peripheral and central neurons. Here, we analyzed Spry2 global knockout mice with respect to axon outgrowth in vitro and peripheral axon regeneration in vivo. Neurons dissociated from adult Spry2 deficient sensory ganglia revealed stronger extracellular signal-regulated kinase activation and enhanced axon outgrowth. Prominent axon elongation was observed in heterozygous Spry2(+/-) neuron cultures, whereas homozygous Spry2(-/-) neurons predominantly exhibited a branching phenotype. Following sciatic nerve crush, Spry2(+/-) mice recovered faster in motor but not sensory testing paradigms (Spry2(-/-) mice did not tolerate anesthesia required for nerve surgery). We attribute the improvement in the rotarod test to higher numbers of myelinated fibers in the regenerating sciatic nerve, higher densities of motor endplates in hind limb muscles and increased levels of GAP-43 mRNA, a downstream target of extracellular regulated kinase signaling. Conversely, homozygous Spry2(-/-) mice revealed enhanced mechanosensory function (von Frey's test) that was accompanied by an increased innervation of the epidermis, elevated numbers of nonmyelinated axons and more IB4-positive neurons in dorsal root ganglia. The present results corroborate the functional significance of receptor tyrosine kinase signaling inhibitors for axon outgrowth during development and nerve regeneration and propose Spry2 as a novel potential target for pharmacological inhibition to accelerate long-distance axon regeneration in injured peripheral nerves.
Collapse
Affiliation(s)
- Letizia Marvaldi
- Division of Neuroanatomy, Department of Anatomy and Histology, Medical University Innsbruck, 6020, Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Meier F, Giesert F, Delic S, Faus-Kessler T, Matheus F, Simeone A, Hölter SM, Kühn R, Weisenhorn DMV, Wurst W, Prakash N. FGF/FGFR2 signaling regulates the generation and correct positioning of Bergmann glia cells in the developing mouse cerebellum. PLoS One 2014; 9:e101124. [PMID: 24983448 PMCID: PMC4077754 DOI: 10.1371/journal.pone.0101124] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 06/03/2014] [Indexed: 12/02/2022] Open
Abstract
The normal cellular organization and layering of the vertebrate cerebellum is established during embryonic and early postnatal development by the interplay of a complex array of genetic and signaling pathways. Disruption of these processes and of the proper layering of the cerebellum usually leads to ataxic behaviors. Here, we analyzed the relative contribution of Fibroblast growth factor receptor 2 (FGFR2)-mediated signaling to cerebellar development in conditional Fgfr2 single mutant mice. We show that during embryonic mouse development, Fgfr2 expression is higher in the anterior cerebellar primordium and excluded from the proliferative ventricular neuroepithelium. Consistent with this finding, conditional Fgfr2 single mutant mice display the most prominent defects in the anterior lobules of the adult cerebellum. In this context, FGFR2-mediated signaling is required for the proper generation of Bergmann glia cells and the correct positioning of these cells within the Purkinje cell layer, and for cell survival in the developing cerebellar primordium. Using cerebellar microexplant cultures treated with an FGFR agonist (FGF9) or antagonist (SU5402), we also show that FGF9/FGFR-mediated signaling inhibits the outward migration of radial glia and Bergmann glia precursors and cells, and might thus act as a positioning cue for these cells. Altogether, our findings reveal the specific functions of the FGFR2-mediated signaling pathway in the generation and positioning of Bergmann glia cells during cerebellar development in the mouse.
Collapse
Affiliation(s)
- Florian Meier
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Florian Giesert
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Sabit Delic
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Theresa Faus-Kessler
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Friederike Matheus
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Antonio Simeone
- Centre of Genetics Engineering (CEINGE) Biotecnologie Avanzate, European School of Molecular Medicine and Institute of Genetics and Biophysics “A. Buzzati-Traverso”, Naples, Italy
| | - Sabine M. Hölter
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Ralf Kühn
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik c/o Helmholtz Zentrum München, Neuherberg, Germany
| | - Daniela M. Vogt. Weisenhorn
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik c/o Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Standort München, München, Germany
- Max-Planck Institute of Psychiatry, München, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik c/o Helmholtz Zentrum München, Neuherberg, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Standort München, München, Germany
- Max-Planck Institute of Psychiatry, München, Germany
- Munich Cluster for Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, München, Germany
- * E-mail: (WW) (WW); (NP) (NP)
| | - Nilima Prakash
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik c/o Helmholtz Zentrum München, Neuherberg, Germany
- * E-mail: (WW) (WW); (NP) (NP)
| |
Collapse
|
37
|
Saritas-Yildirim B, Silva EM. The role of targeted protein degradation in early neural development. Genesis 2014; 52:287-99. [PMID: 24623518 DOI: 10.1002/dvg.22771] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 11/08/2022]
Abstract
As neural stem cells differentiate into neurons during neurogenesis, the proteome of the cells is restructured by de novo expression and selective removal of regulatory proteins. The control of neurogenesis at the level of gene regulation is well documented and the regulation of protein abundance through protein degradation via the Ubiquitin/26S proteasome pathway is a rapidly developing field. This review describes our current understanding of the role of the proteasome pathway in neurogenesis. Collectively, the studies show that targeted protein degradation is an important regulatory mechanism in the generation of new neurons.
Collapse
|
38
|
Yu T, Meiners LC, Danielsen K, Wong MT, Bowler T, Reinberg D, Scambler PJ, van Ravenswaaij-Arts CM, Basson MA. Deregulated FGF and homeotic gene expression underlies cerebellar vermis hypoplasia in CHARGE syndrome. eLife 2013; 2:e01305. [PMID: 24368733 PMCID: PMC3870572 DOI: 10.7554/elife.01305] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mutations in CHD7 are the major cause of CHARGE syndrome, an autosomal dominant disorder with an estimated prevalence of 1/15,000. We have little understanding of the disruptions in the developmental programme that underpin brain defects associated with this syndrome. Using mouse models, we show that Chd7 haploinsufficiency results in reduced Fgf8 expression in the isthmus organiser (IsO), an embryonic signalling centre that directs early cerebellar development. Consistent with this observation, Chd7 and Fgf8 loss-of-function alleles interact during cerebellar development. CHD7 associates with Otx2 and Gbx2 regulatory elements and altered expression of these homeobox genes implicates CHD7 in the maintenance of cerebellar identity during embryogenesis. Finally, we report cerebellar vermis hypoplasia in 35% of CHARGE syndrome patients with a proven CHD7 mutation. These observations provide key insights into the molecular aetiology of cerebellar defects in CHARGE syndrome and link reduced FGF signalling to cerebellar vermis hypoplasia in a human syndrome. DOI:http://dx.doi.org/10.7554/eLife.01305.001 CHARGE syndrome is a rare genetic condition that causes various developmental abnormalities, including heart defects, deafness and neurological defects. In most cases, it is caused by mutations in a human gene called CHD7. CHD7 is known to control the expression of other genes during embryonic development, but the molecular mechanisms by which mutations in CHD7 lead to the neural defects found in CHARGE syndrome are unclear. During embryonic development, the neural tube—the precursor to the nervous system—is divided into segments, which give rise to different neural structures. The r1 segment, for example, forms the cerebellum, and the secretion of a protein called FGF8 (short for fibroblast growth factor 8) by a nearby structure called the isthmus organiser has an important role in this process. Since a reduction in FGF8 causes defects similar to those found in CHARGE syndrome, Yu et al. decided to investigate if the FGF signalling pathway was involved in this syndrome. Mice should have two working copies of the Chd7 gene, and mice that lack one of these suffer from symptoms similar to those of humans with CHARGE syndrome. Yu et al. examined the embryos of these mice and found that the isthmus organiser produced less FGF8. Embryos with no working copies of the gene completely lost the r1 segment. The loss of this segment appeared to be caused by changes in the expression of homeobox genes (the genes that determine the identity of brain segments). Embryos that did not have any working copies of the Chd7 gene died early in development, which made further studies impossible. However, embryos that had one working copy of the Chd7 gene survived, and Yu et al. took advantage of this to study the effects of reduced FGF8 expression on these mice. These experiments showed that mice with just one working copy of the Fgf8 gene and one working copy of the Chd7 gene had a small cerebellar vermis. This part of the cerebellum is known to be very sensitive to changes in FGF8 signalling. Yu et al. then used an MRI scanner to look at the cerebellar vermis in patients with CHARGE syndrome, and found that more than half of the patients had abnormal cerebella. In addition to confirming that studies on mouse embryos can provide insights into human disease, the work of Yu et al. add defects in the cerebellar vermis to the list of developmental abnormalities associated with CHARGE syndrome. The next step will be to test if any mutations in the human FGF8 gene can contribute to cerebellar defects in CHARGE syndrome, and to investigate if any other developmental defects in CHARGE syndrome are associated with abnormal FGF8 levels. DOI:http://dx.doi.org/10.7554/eLife.01305.002
Collapse
Affiliation(s)
- Tian Yu
- Department of Craniofacial Development and Stem Cell Biology, King's College London, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dyer C, Blanc E, Hanisch A, Roehl H, Otto GW, Yu T, Basson MA, Knight R. A bi-modal function of Wnt signalling directs an FGF activity gradient to spatially regulate neuronal differentiation in the midbrain. Development 2013; 141:63-72. [PMID: 24284206 DOI: 10.1242/dev.099507] [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] [Indexed: 12/28/2022]
Abstract
FGFs and Wnts are important morphogens during midbrain development, but their importance and potential interactions during neurogenesis are poorly understood. We have employed a combination of genetic and pharmacological manipulations in zebrafish to show that during neurogenesis FGF activity occurs as a gradient along the anterior-posterior axis of the dorsal midbrain and directs spatially dynamic expression of the Hairy gene her5. As FGF activity diminishes during development, Her5 is lost and differentiation of neuronal progenitors occurs in an anterior-posterior manner. We generated mathematical models to explain how Wnt and FGFs direct the spatial differentiation of neurons in the midbrain through Wnt regulation of FGF signalling. These models suggested that a negative-feedback loop controlled by Wnt is crucial for regulating FGF activity. We tested Sprouty genes as mediators of this regulatory loop using conditional mouse knockouts and pharmacological manipulations in zebrafish. These reveal that Sprouty genes direct the positioning of early midbrain neurons and are Wnt responsive in the midbrain. We propose a model in which Wnt regulates FGF activity at the isthmus by driving both FGF and Sprouty gene expression. This controls a dynamic, posteriorly retracting expression of her5 that directs neuronal differentiation in a precise spatiotemporal manner in the midbrain.
Collapse
Affiliation(s)
- Carlene Dyer
- Craniofacial Development and Stem Cell Biology, King's College London, Guy's Hospital, London SE1 9RT, UK
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Basson MA, Wingate RJ. Congenital hypoplasia of the cerebellum: developmental causes and behavioral consequences. Front Neuroanat 2013; 7:29. [PMID: 24027500 PMCID: PMC3759752 DOI: 10.3389/fnana.2013.00029] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/09/2013] [Indexed: 12/14/2022] Open
Abstract
Over the last 60 years, the spotlight of research has periodically returned to the cerebellum as new techniques and insights have emerged. Because of its simple homogeneous structure, limited diversity of cell types and characteristic behavioral pathologies, the cerebellum is a natural home for studies of cell specification, patterning, and neuronal migration. However, recent evidence has extended the traditional range of perceived cerebellar function to include modulation of cognitive processes and implicated cerebellar hypoplasia and Purkinje neuron hypo-cellularity with autistic spectrum disorder. In the light of this emerging frontier, we review the key stages and genetic mechanisms behind cerebellum development. In particular, we discuss the role of the midbrain hindbrain isthmic organizer in the development of the cerebellar vermis and the specification and differentiation of Purkinje cells and granule neurons. These developmental processes are then considered in relation to recent insights into selected human developmental cerebellar defects: Joubert syndrome, Dandy–Walker malformation, and pontocerebellar hypoplasia. Finally, we review current research that opens up the possibility of using the mouse as a genetic model to study the role of the cerebellum in cognitive function.
Collapse
Affiliation(s)
- M Albert Basson
- Department of Craniofacial Development and Stem Cell Biology, King's College London London, UK ; Medical Research Council Centre for Developmental Neurobiology, King's College London London, UK
| | | |
Collapse
|
41
|
Annenkov A. Receptor tyrosine kinase (RTK) signalling in the control of neural stem and progenitor cell (NSPC) development. Mol Neurobiol 2013; 49:440-71. [PMID: 23982746 DOI: 10.1007/s12035-013-8532-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 08/09/2013] [Indexed: 01/04/2023]
Abstract
Important developmental responses are elicited in neural stem and progenitor cells (NSPC) by activation of the receptor tyrosine kinases (RTK), including the fibroblast growth factor receptors, epidermal growth factor receptor, platelet-derived growth factor receptors and insulin-like growth factor receptor (IGF1R). Signalling through these RTK is necessary and sufficient for driving a number of developmental processes in the central nervous system. Within each of the four RTK families discussed here, receptors are activated by sets of ligands that do not cross-activate receptors of the other three families, and therefore, their activation can be independently regulated by ligand availability. These RTK pathways converge on a conserved core of signalling molecules, but differences between the receptors in utilisation of signalling molecules and molecular adaptors for intracellular signal propagation become increasingly apparent. Intracellular inhibitors of RTK signalling are widely involved in the regulation of developmental signalling in NSPC and often determine developmental outcomes of RTK activation. In addition, cellular responses of NSPC to the activation of a given RTK may be significantly modulated by signal strength. Cellular propensity to respond also plays a role in developmental outcomes of RTK signalling. In combination, these mechanisms regulate the balance between NSPC maintenance and differentiation during development and in adulthood. Attribution of particular developmental responses of NSPC to specific pathways of RTK signalling becomes increasingly elucidated. Co-activation of several RTK in developing NSPC is common, and analysis of co-operation between their signalling pathways may advance knowledge of RTK role in NSPC development.
Collapse
Affiliation(s)
- Alexander Annenkov
- Bone and Joint Research Unit, William Harvey Research Institute, Bart's and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK,
| |
Collapse
|
42
|
Mor E, Kano SI, Colantuoni C, Sawa A, Navon R, Shomron N. MicroRNA-382 expression is elevated in the olfactory neuroepithelium of schizophrenia patients. Neurobiol Dis 2013; 55:1-10. [PMID: 23542694 DOI: 10.1016/j.nbd.2013.03.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 03/17/2013] [Accepted: 03/20/2013] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia is a common neuropsychiatric disorder that has a strong genetic component. MicroRNAs (miRNAs) have been implicated in neurodevelopmental and psychiatric disorders including schizophrenia, as indicated by their dysregulation in post-mortem brain tissues and in peripheral blood of schizophrenia patients. The olfactory epithelium (OE) is one of the few accessible neural tissues that contain neurons and their stem cells. Previous studies showed that OE-derived tissues and cells can be safely and easily collected from live human subjects and may provide a "window" into neuronal processes involved in disorders such as schizophrenia, while avoiding the limitations of using postmortem brain samples or non-neuronal tissues. In this study, we found that the brain-enriched miR-382 (miR-382-5p) expression was elevated in in vitro cultured olfactory cells, in a cohort of seven schizophrenia patients compared with seven non-schizophrenic controls. MiR-382 elevation was further confirmed in laser-capture microdissected OE neuronal tissue (LCM-OE), enriched for mature olfactory neurons, in a cohort of 18 schizophrenia patients and 18 non-schizophrenic controls. In sharp contrast, miR-382 expression could not be detected in lymphoblastoid cell lines generated from schizophrenic or non-schizophrenic individuals. We further found that miR-382 directly regulates the expression of two genes, FGFR1 and SPRY4, which are downregulated in both the cultured olfactory cells and LCM-OE derived from schizophrenia patients. These genes are involved in the fibroblast growth factor (FGF) signaling pathway, while impairment of this pathway may underlie abnormal brain development and function associated with schizophrenia. Our data suggest that miR-382 elevation detected in patients' OE-derived samples might serve to strengthen current biomarker studies in schizophrenia. This study also illustrates the potential utility of OE-derived tissues and cells as surrogate samples for the brain.
Collapse
Affiliation(s)
- Eyal Mor
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | | | | | | | | | | |
Collapse
|
43
|
Gardiner JR, Jackson AL, Gordon J, Lickert H, Manley NR, Basson MA. Localised inhibition of FGF signalling in the third pharyngeal pouch is required for normal thymus and parathyroid organogenesis. Development 2012; 139:3456-66. [PMID: 22912418 DOI: 10.1242/dev.079400] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The thymus and parathyroid glands are derived from the third pharyngeal pouch endoderm. The mechanisms that establish distinct molecular domains in the third pouch and control the subsequent separation of these organ primordia from the pharynx are poorly understood. Here, we report that mouse embryos that lack two FGF feedback antagonists, Spry1 and Spry2, display parathyroid and thymus hypoplasia and a failure of these organ primordia to completely separate from the pharynx. We show that FGF ligands and downstream reporter genes are expressed in highly regionalised patterns in the third pouch and that sprouty gene deletion results in upregulated FGF signalling throughout the pouch endoderm. As a consequence, the initiation of markers of parathyroid and thymus fate is altered. In addition, a normal apoptotic programme that is associated with the separation of the primordia from the pharynx is disrupted, resulting in the maintenance of a thymus-pharynx attachment and a subsequent inability of the thymus to migrate to its appropriate position above the heart. We demonstrate that the sprouty genes function in the pharyngeal endoderm itself to control these processes and that the defects in sprouty-deficient mutants are, at least in part, due to hyper-responsiveness to Fgf8. Finally, we provide evidence to suggest that parathyroid hypoplasia in these mutants is due to early gene expression defects in the third pouch, whereas thymus hypoplasia is caused by reduced proliferation of thymic epithelial cells in the thymus primordium.
Collapse
Affiliation(s)
- Jennifer R Gardiner
- Department of Craniofacial Development, King's College London, 27th floor, Guy's Tower, London SE1 9RT, UK
| | | | | | | | | | | |
Collapse
|
44
|
Activation of Wnt/β-catenin signalling affects differentiation of cells arising from the cerebellar ventricular zone. PLoS One 2012; 7:e42572. [PMID: 22880037 PMCID: PMC3411831 DOI: 10.1371/journal.pone.0042572] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 07/09/2012] [Indexed: 11/26/2022] Open
Abstract
Development of the cerebellum proceeds under the precise spatio-temporal control of several key developmental signalling pathways, including the Wnt/β-catenin pathway. We recently reported the activity of Wnt/β-catenin signalling in the perinatal cerebellar ventricular zone (VZ), a germinal centre in the developing cerebellum that gives rise to GABAergic and glial cells. In order to investigate the normal function of Wnt/β-catenin signalling in the VZ and the cell lineages it gives rise to, we used a combination of ex vivo cerebellar slice culture and in vivo genetic manipulation to dysregulate its activity during late embryonic development. Activation of the pathway at the cerebellar ventricular zone led to a reduction in the number of cells expressing the glial lineage markers Sox9 and GFAP and the interneuron marker Pax2, but had no consistent effect on either proliferation or apoptosis. Our findings suggest that activation of the Wnt/β-catenin pathway in the cerebellar ventricular zone causes a shift in the cell types produced, most likely due to disruption of normal differentiation. Thus, we propose that regulation of Wnt/β-catenin signalling levels are required for normal development of cells arising from the cerebellar ventricular zone during late embryogenesis.
Collapse
|
45
|
Shin EHH, Basson MA, Robinson ML, McAvoy JW, Lovicu FJ. Sprouty is a negative regulator of transforming growth factor β-induced epithelial-to-mesenchymal transition and cataract. Mol Med 2012; 18:861-73. [PMID: 22517312 DOI: 10.2119/molmed.2012.00111] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Accepted: 04/10/2012] [Indexed: 01/06/2023] Open
Abstract
Fibrosis affects an extensive range of organs and is increasingly acknowledged as a major component of many chronic disorders. It is now well accepted that the elevated expression of certain inflammatory cell-derived cytokines, especially transforming growth factor β (TGFβ), is involved in the epithelial-to-mesenchymal transition (EMT) leading to the pathogenesis of a diverse range of fibrotic diseases. In lens, aberrant TGFβ signaling has been shown to induce EMT leading to cataract formation. Sproutys (Sprys) are negative feedback regulators of receptor tyrosine kinase (RTK)-signaling pathways in many vertebrate systems, and in this study we showed that they are important in the murine lens for promoting the lens epithelial cell phenotype. Conditional deletion of Spry1 and Spry2 specifically from the lens leads to an aberrant increase in RTK-mediated extracellular signal-regulated kinase 1/2 phosphorylation and, surprisingly, elevated TGFβ-related signaling in lens epithelial cells, leading to an EMT and subsequent cataract formation. Conversely, increased Spry overexpression in lens cells can suppress not only TGFβ-induced signaling, but also the accompanying EMT and cataract formation. On the basis of these findings, we propose that a better understanding of the relationship between Spry and TGFβ signaling will not only elucidate the etiology of lens pathology, but will also lead to the development of treatments for other fibrotic-related diseases associated with TGFβ-induced EMT.
Collapse
Affiliation(s)
- Eun Hye H Shin
- Anatomy and Histology, Bosch Institute & Save Sight Institute, University of Sydney, Sydney, Australia
| | | | | | | | | |
Collapse
|
46
|
Crespo-Enriquez I, Partanen J, Martinez S, Echevarria D. Fgf8-related secondary organizers exert different polarizing planar instructions along the mouse anterior neural tube. PLoS One 2012; 7:e39977. [PMID: 22792203 PMCID: PMC3391221 DOI: 10.1371/journal.pone.0039977] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 05/30/2012] [Indexed: 12/11/2022] Open
Abstract
Early brain patterning depends on proper arrangement of positional information. This information is given by gradients of secreted signaling molecules (morphogens) detected by individual cells within the responding tissue, leading to specific fate decisions. Here we report that the morphogen FGF8 exerts initially a differential signal activity along the E9.5 mouse neural tube. We demonstrate that this polarizing activity codes by RAS-regulated ERK1/2 signaling and depends on the topographical location of the secondary organizers: the isthmic organizer (IsO) and the anterior neural ridge (anr) but not on zona limitans intrathalamica (zli). Our results suggest that Sprouty2, a negative modulator of RAS/ERK pathway, is important for regulating Fgf8 morphogenetic signal activity by controlling Fgf8-induced signaling pathways and positional information during early brain development.
Collapse
Affiliation(s)
- Ivan Crespo-Enriquez
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas–Universidad Miguel Hernandez, Alicante, Spain
| | - Juha Partanen
- Department of Biosciences, Division of Genetics, University of Helsinki, Helsinki, Finland
| | - Salvador Martinez
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas–Universidad Miguel Hernandez, Alicante, Spain
| | - Diego Echevarria
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas–Universidad Miguel Hernandez, Alicante, Spain
- * E-mail:
| |
Collapse
|
47
|
Basson MA. Signaling in cell differentiation and morphogenesis. Cold Spring Harb Perspect Biol 2012; 4:cshperspect.a008151. [PMID: 22570373 DOI: 10.1101/cshperspect.a008151] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
All the information to make a complete, fully functional living organism is encoded within the genome of the fertilized oocyte. How is this genetic code translated into the vast array of cellular behaviors that unfold during the course of embryonic development, as the zygote slowly morphs into a new organism? Studies over the last 30 years or so have shown that many of these cellular processes are driven by secreted or membrane-bound signaling molecules. Elucidating how the genetic code is translated into instructions or signals during embryogenesis, how signals are generated at the correct time and place and at the appropriate level, and finally, how these instructions are interpreted and put into action, are some of the central questions of developmental biology. Our understanding of the causes of congenital malformations and disease has improved substantially with the rapid advances in our knowledge of signaling pathways and their regulation during development. In this article, I review some of the signaling pathways that play essential roles during embryonic development. These examples show some of the mechanisms used by cells to receive and interpret developmental signals. I also discuss how signaling pathways downstream from these signals are regulated and how they induce specific cellular responses that ultimately affect cell fate and morphogenesis.
Collapse
Affiliation(s)
- M Albert Basson
- Department of Craniofacial Development, King's College London, United Kingdom.
| |
Collapse
|
48
|
White JJ, Sillitoe RV. Development of the cerebellum: from gene expression patterns to circuit maps. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:149-64. [DOI: 10.1002/wdev.65] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|