1
|
Halmi CA, Leonard CE, McIntosh AT, Taneyhill LA. N-cadherin facilitates trigeminal sensory neuron outgrowth and target tissue innervation. Development 2025; 152:dev204369. [PMID: 40260574 PMCID: PMC12070061 DOI: 10.1242/dev.204369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/01/2025] [Indexed: 04/23/2025]
Abstract
The trigeminal ganglion emerges from the condensation of two distinct precursor cell populations, cranial placodes and neural crest. While its dual cellular origin is well understood, the molecules underlying its formation remain relatively obscure. Trigeminal ganglion assembly is mediated, in part, by neural cadherin (N-cadherin), which is initially expressed by placodal neurons and is required for their proper coalescence with neural crest cells. Axon outgrowth first occurs from placodal neurons, but as gangliogenesis proceeds, neural crest cells also differentiate into N-cadherin-expressing neurons, and both extend axons toward targets. However, the role of N-cadherin in axon outgrowth and target innervation has not been explored. Our data show that N-cadherin knockdown in chick trigeminal placode cells decreases trigeminal ganglion size, nerve growth and target innervation in vivo, and reduces neurite complexity of neural crest-derived neurons in vitro. Furthermore, blocking N-cadherin-mediated adhesion prevents axon extension in most placodal neurons in vitro. Collectively, these findings reveal cell- and non-cell autonomous functions for N-cadherin, highlighting its crucial role in mediating reciprocal interactions between neural crest- and placode-derived neurons throughout trigeminal ganglion development.
Collapse
Affiliation(s)
- Caroline A. Halmi
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Carrie E. Leonard
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Alec T. McIntosh
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Lisa A. Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
2
|
Leonard CE, McIntosh A, Sanyal J, Taneyhill LA. The transcriptional landscape of the developing chick trigeminal ganglion. Dev Biol 2025; 520:108-116. [PMID: 39719193 PMCID: PMC11863308 DOI: 10.1016/j.ydbio.2024.12.013] [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: 07/20/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
The trigeminal ganglion is a critical structure in the peripheral nervous system, responsible for transmitting sensations of touch, pain, and temperature from craniofacial regions to the brain. Trigeminal ganglion development depends upon intrinsic cellular programming as well as extrinsic signals exchanged by diverse cell populations. With its complex anatomy and dual cellular origin from cranial placodes and neural crest cells, the trigeminal ganglion offers a rich context for examining diverse biological processes, including cell migration, fate determination, adhesion, and axon guidance. Avian models have, so far, enabled key insights into craniofacial and peripheral nervous system development. Yet the molecular mechanisms driving trigeminal ganglion formation and subsequent nerve growth remain elusive. In this study, we performed RNA-sequencing at multiple stages of chick trigeminal ganglion development and generated a novel transcriptomic dataset that has been curated to illustrate temporally dynamic gene expression patterns. This publicly available resource identifies major pathways involved in trigeminal gangliogenesis, particularly with respect to the condensation and maturation of placode-derived neurons, thus inviting new lines of research into the essential processes governing trigeminal ganglion development.
Collapse
Affiliation(s)
- Carrie E Leonard
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Alec McIntosh
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20007, USA
| | - Johena Sanyal
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
3
|
Halmi CA, Leonard CE, McIntosh AT, Taneyhill LA. N-cadherin facilitates trigeminal sensory neuron outgrowth and target tissue innervation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.20.594965. [PMID: 38826314 PMCID: PMC11142107 DOI: 10.1101/2024.05.20.594965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The trigeminal ganglion emerges from the condensation of two distinct precursor cell populations, cranial placodes and neural crest. While its dual cellular origin is well understood, the molecules underlying its formation remain relatively obscure. Trigeminal ganglion assembly is mediated, in part, by neural cadherin (N-cadherin), which is initially expressed by placodal neurons and required for their proper coalescence with neural crest cells. Axon outgrowth first occurs from placodal neurons, but as gangliogenesis proceeds, neural crest cells also differentiate into N-cadherin-expressing neurons, and both extend axons toward targets. However, the role of N-cadherin in axon outgrowth and target innervation has not been explored. Our data show that N-cadherin knockdown in chick trigeminal placode cells decreases trigeminal ganglion size, nerve growth, and target innervation in vivo , and reduces neurite complexity of neural crest-derived neurons in vitro. Furthermore, blocking N-cadherin-mediated adhesion prevents axon extension in most placodal neurons in vitro . Collectively, these findings reveal cell- and non-cell autonomous functions for N-cadherin, highlighting its critical role in mediating reciprocal interactions between neural crest- and placode-derived neurons throughout trigeminal ganglion development.
Collapse
|
4
|
Cui HS, Zheng YX, Cho YS, Ro YM, Jeon K, Joo SY, Seo CH. Slit1 Promotes Hypertrophic Scar Formation Through the TGF-β Signaling Pathway. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2051. [PMID: 39768930 PMCID: PMC11678377 DOI: 10.3390/medicina60122051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
Background and objectives: Slit1 is a secreted protein that is closely related to cell movement and adhesion. Few studies related to fibrosis exist, and the preponderance of current research is confined to the proliferation and differentiation of neural systems. Hypertrophic scars (HTSs) are delineated by an overproduction of the extracellular matrix (ECM) by activated fibroblasts, leading to anomalous fibrosis, which is a severe sequela of burns. However, the functionality of Slit1 in HTS formation remains unknown. We aimed to investigate whether Slit1 regulates fibroblasts through a fibrosis-related mechanism derived from post-burn HTS tissues and normal patient tissues. Methods: Human normal fibroblasts (HNFs) and hypertrophic scar fibroblasts (HTSFs) were extracted from normal skin and post-burn HTS tissues, with settings grouped according to the patient of origin. Cell proliferation was evaluated using a CellTiter-Glo Luminescent Cell Viability Assay Kit. Cell migration experiments were carried out using a μ-Dish insert system. Protein and mRNA expression levels were quantified by Western blot and quantitative real-time polymerase chain reaction. Results: We found increased expressions of Slit1 in HTS tissues and HTSFs compared to normal tissues and HNFs. The treatment of human recombinant Slit1 protein (rSlit1) within HNFs promoted cell proliferation and differentiation, leading to an upregulation in ECM components such as α-SMA, type I and III collagen, and fibronectin. The treatment of rSlit1 in HNFs facilitated cell migration, concurrent with enhanced levels of N-cadherin and vimentin, and a diminished expression of E-cadherin. Treatment with rSlit1 resulted in the phosphorylation of SMAD pathway proteins, including SMAD2, SMAD3, and SMAD1/5/8, and non-SMAD pathway proteins, including TAK1, JNK1, ERK1/2, and p38, in HNFs. Conclusions: Exogenous Slit1 potentiates the epithelial-mesenchymal transition and upregulates SMAD and non-SMAD signaling pathways in HNFs, leading to the development of HTS, suggesting that Slit1 is a promising new target for the treatment of post-burn HTS.
Collapse
Affiliation(s)
- Hui Song Cui
- Burn Institute, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, 94-200 Yeongdeungpo-Dong, Yeongdeungpo-Ku, Seoul 07247, Republic of Korea; (H.S.C.); (Y.X.Z.); (Y.M.R.)
| | - Ya Xin Zheng
- Burn Institute, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, 94-200 Yeongdeungpo-Dong, Yeongdeungpo-Ku, Seoul 07247, Republic of Korea; (H.S.C.); (Y.X.Z.); (Y.M.R.)
| | - Yoon Soo Cho
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, 94-200 Yeongdeungpo-Dong, Yeongdeungpo-Ku, Seoul 07247, Republic of Korea;
| | - Yu Mi Ro
- Burn Institute, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, 94-200 Yeongdeungpo-Dong, Yeongdeungpo-Ku, Seoul 07247, Republic of Korea; (H.S.C.); (Y.X.Z.); (Y.M.R.)
| | - Kibum Jeon
- Department of Laboratory Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, 94-200 Yeongdeungpo-Dong, Yeongdeungpo-Ku, Seoul 07247, Republic of Korea;
| | - So Young Joo
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, 94-200 Yeongdeungpo-Dong, Yeongdeungpo-Ku, Seoul 07247, Republic of Korea;
| | - Cheong Hoon Seo
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, 94-200 Yeongdeungpo-Dong, Yeongdeungpo-Ku, Seoul 07247, Republic of Korea;
| |
Collapse
|
5
|
Leonard CE, McIntosh A, Sanyal J, Taneyhill LA. The transcriptional landscape of the developing chick trigeminal ganglion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.20.604400. [PMID: 39211243 PMCID: PMC11361123 DOI: 10.1101/2024.07.20.604400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The trigeminal ganglion is a critical structure in the peripheral nervous system, responsible for transmitting sensations of touch, pain, and temperature from craniofacial regions to the brain. Trigeminal ganglion development depends upon intrinsic cellular programming as well as extrinsic signals exchanged by diverse cell populations. With its complex anatomy and dual cellular origin from cranial placodes and neural crest cells, the trigeminal ganglion offers a rich context for examining diverse biological processes, including cell migration, fate determination, adhesion, and axon guidance. Avian models have, so far, enabled key insights into craniofacial and peripheral nervous system development. Yet, the molecular mechanisms driving trigeminal ganglion formation and subsequent nerve growth remain elusive. In this study, we performed RNA-sequencing at multiple stages of chick trigeminal ganglion development and generated a novel transcriptomic dataset that has been curated to illustrate temporally dynamic gene expression patterns. This publicly available resource identifies major pathways involved in trigeminal gangliogenesis, particularly with respect to the condensation and maturation of placode-derived neurons, thus inviting new lines of research into the essential processes governing trigeminal ganglion development.
Collapse
|
6
|
Urrutia HA, Stundl J, Bronner ME. Tlx3 mediates neuronal differentiation and proper condensation of the developing trigeminal ganglion. Dev Biol 2024; 515:79-91. [PMID: 39019425 PMCID: PMC11317220 DOI: 10.1016/j.ydbio.2024.07.005] [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: 02/08/2024] [Revised: 06/20/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
The trigeminal ganglion, the largest of the vertebrate cranial ganglia, is comprised of sensory neurons that relay sensations of pain, touch, and temperature to the brain. These neurons are derived from two embryonic cell types, the neural crest and ectodermal placodes, whose interactions are critical for proper ganglion formation. While the T-cell leukemia homeobox 3 (Tlx3) gene is known to be expressed in placodally-derived sensory neurons and necessary for their differentiation, little was known about Tlx3 expression and/or function in the neural crest-derived component of the developing trigeminal ganglion. By combining lineage labeling with in situ hybridization in the chick embryo, we show that neural crest-derived cells that contribute to the cranial trigeminal ganglion express Tlx3 at a time point that coincides with the onset of ganglion condensation. Importantly, loss of Tlx3 function in vivo diminishes the overall size and abundance of neurons within the trigeminal ganglion. Conversely, ectopic expression of Tlx3 in migrating cranial neural crest results in their premature neuronal differentiation. Taken together, our results demonstrate a critical role for Tlx3 in neural crest-derived cells during chick trigeminal gangliogenesis.
Collapse
Affiliation(s)
- Hugo A Urrutia
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Jan Stundl
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
7
|
Hines MA, Taneyhill LA. Elp1 function in placode-derived neurons is critical for proper trigeminal ganglion development. Dev Dyn 2024:10.1002/dvdy.749. [PMID: 39381860 PMCID: PMC11978919 DOI: 10.1002/dvdy.749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/06/2024] [Accepted: 09/14/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND The trigeminal nerve is the largest cranial nerve and functions in somatosensation. Cell bodies of this nerve are positioned in the trigeminal ganglion, which arises from the coalescence of neural crest and placode cells. While this dual cellular origin has been known for decades, the molecular mechanisms controlling trigeminal ganglion development remain obscure. We performed RNA sequencing on the forming chick trigeminal ganglion and identified Elongator acetyltransferase complex subunit 1 (Elp1) for further study. Mutations in ELP1 cause familial dysautonomia (FD), a fatal disorder characterized by the presence of smaller trigeminal nerves and sensory deficits. While Elp1 has established roles in neurogenesis, its function in placode cells during trigeminal gangliogenesis has not been investigated. RESULTS To this end, we used morpholinos to deplete Elp1 from chick trigeminal placode cells. Elp1 knockdown decreased trigeminal ganglion size and led to aberrant innervation of the eye by placode-derived neurons. Trigeminal nerve branches also appeared to exhibit reduced axon outgrowth to target tissues. CONCLUSIONS These findings reveal a new role for Elp1 in placode-derived neurons during chick trigeminal ganglion development. These results have potential high significance to provide new insights into trigeminal ganglion development and the etiology of FD.
Collapse
Affiliation(s)
- Margaret A. Hines
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742 USA
| | - Lisa A. Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742 USA
| |
Collapse
|
8
|
Hines MA, Taneyhill LA. Elp1 function in placode-derived neurons is critical for proper trigeminal ganglion development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603323. [PMID: 39071383 PMCID: PMC11275904 DOI: 10.1101/2024.07.12.603323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background The trigeminal nerve is the largest cranial nerve and functions in somatosensation. Cell bodies of this nerve are positioned in the trigeminal ganglion, which arises from the coalescence of neural crest and placode cells. While this dual cellular origin has been known for decades, the molecular mechanisms controlling trigeminal ganglion development remain obscure. We performed RNAsequencing on the forming chick trigeminal ganglion and identified Elongator acetyltransferase complex subunit 1 ( Elp1 ) for further study. Mutations in ELP1 cause familial dysautonomia (FD), a fatal disorder characterized by the presence of smaller trigeminal nerves and sensory deficits. While Elp1 has established roles in neurogenesis, its functions in placode cells during trigeminal gangliogenesis have not been investigated. Results To this end, we used morpholinos to deplete Elp1 from chick trigeminal placode cells. Elp1 knockdown decreased trigeminal ganglion size and led to aberrant innervation of the eye by placode-derived neurons. Trigeminal nerve branches exhibited fewer axons, and abnormal interactions between placode-derived neurons and neural crest cells were observed. Conclusions These findings reveal a new role for Elp1 in chick placode-derived neurons during trigeminal ganglion development. These results have potential high significance to provide new insights into trigeminal ganglion development and the etiology of FD. Bullet points Elp1 is expressed in undifferentiated neural crest cells and placode-derived neurons contributing to the trigeminal ganglion.Elp1 knockdown in trigeminal placode cells reduces trigeminal ganglion size.Elp1 depletion from trigeminal placode cells leads to aberrant target tissue innervation and disrupts proper neural crest-placodal neuron interactions in the trigeminal ganglion. Grant sponsor and number NIH R01DE024217 and NIH R03HD108480.
Collapse
|
9
|
Bernardi YE, Sanchez-Vasquez E, Márquez RB, Piacentino ML, Urrutia H, Rossi I, Alcântara Saraiva KL, Pereira-Neves A, Ramirez MI, Bronner ME, de Miguel N, Strobl-Mazzulla PH. miR-203 secreted in extracellular vesicles mediates the communication between neural crest and placode cells required for trigeminal ganglia formation. PLoS Biol 2024; 22:e3002074. [PMID: 39038054 PMCID: PMC11293684 DOI: 10.1371/journal.pbio.3002074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 08/01/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
While interactions between neural crest and placode cells are critical for the proper formation of the trigeminal ganglion, the mechanisms underlying this process remain largely uncharacterized. Here, by using chick embryos, we show that the microRNA (miR)-203, whose epigenetic repression is required for neural crest migration, is reactivated in coalescing and condensing trigeminal ganglion cells. Overexpression of miR-203 induces ectopic coalescence of neural crest cells and increases ganglion size. By employing cell-specific electroporations for either miR-203 sponging or genomic editing using CRISPR/Cas9, we elucidated that neural crest cells serve as the source, while placode cells serve as the site of action for miR-203 in trigeminal ganglion condensation. Demonstrating intercellular communication, overexpression of miR-203 in the neural crest in vitro or in vivo represses an miR-responsive sensor in placode cells. Moreover, neural crest-secreted extracellular vesicles (EVs), visualized using pHluorin-CD63 vector, become incorporated into the cytoplasm of placode cells. Finally, RT-PCR analysis shows that small EVs isolated from condensing trigeminal ganglia are selectively loaded with miR-203. Together, our findings reveal a critical role in vivo for neural crest-placode communication mediated by sEVs and their selective microRNA cargo for proper trigeminal ganglion formation.
Collapse
Affiliation(s)
- Yanel E. Bernardi
- Laboratory of Developmental Biology, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM, Chascomús, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Argentina
| | - Estefania Sanchez-Vasquez
- Laboratory of Developmental Biology, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM, Chascomús, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Argentina
| | - Rocío Belén Márquez
- Laboratory of Developmental Biology, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM, Chascomús, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Argentina
| | - Michael L. Piacentino
- Division of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - Hugo Urrutia
- Division of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - Izadora Rossi
- Laboratorio de biologia molecular e sistematica de tripanossomatideos, Instituto Carlos Chagas, Fiocruz Parana, Curitiba, Brazil
| | | | | | - Marcel I. Ramirez
- Laboratorio de biologia molecular e sistematica de tripanossomatideos, Instituto Carlos Chagas, Fiocruz Parana, Curitiba, Brazil
| | - Marianne E. Bronner
- Division of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - Natalia de Miguel
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Argentina
- Laboratorio de Parásitos Anaerobios, Instituto Tecnológico Chascomús (INTECH), CONICET-UNSAM, Chascomús, Argentina
| | - Pablo H. Strobl-Mazzulla
- Laboratory of Developmental Biology, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM, Chascomús, Argentina
- Escuela de Bio y Nanotecnologías (UNSAM), Chascomús, Argentina
| |
Collapse
|
10
|
Edens BM, Bronner ME. Making developmental sense of the senses, their origin and function. Curr Top Dev Biol 2024; 159:132-167. [PMID: 38729675 DOI: 10.1016/bs.ctdb.2024.01.015] [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] [Indexed: 05/12/2024]
Abstract
The primary senses-touch, taste, sight, smell, and hearing-connect animals with their environments and with one another. Aside from the eyes, the primary sense organs of vertebrates and the peripheral sensory pathways that relay their inputs arise from two transient stem cell populations: the neural crest and the cranial placodes. In this chapter we consider the senses from historical and cultural perspectives, and discuss the senses as biological faculties. We begin with the embryonic origin of the neural crest and cranial placodes from within the neural plate border of the ectodermal germ layer. Then, we describe the major chemical (i.e. olfactory and gustatory) and mechanical (i.e. vestibulo-auditory and somatosensory) senses, with an emphasis on the developmental interactions between neural crest and cranial placodes that shape their structures and functions.
Collapse
Affiliation(s)
- Brittany M Edens
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States.
| |
Collapse
|
11
|
Inoue K, Bostan H, Browne MR, Bevis OF, Bortner CD, Moore SA, Stence AA, Martin NP, Chen SH, Burkholder AB, Li JL, Shaw ND. DUX4 double whammy: The transcription factor that causes a rare muscular dystrophy also kills the precursors of the human nose. SCIENCE ADVANCES 2023; 9:eabq7744. [PMID: 36800423 PMCID: PMC9937577 DOI: 10.1126/sciadv.abq7744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 01/12/2023] [Indexed: 05/19/2023]
Abstract
SMCHD1 mutations cause congenital arhinia (absent nose) and a muscular dystrophy called FSHD2. In FSHD2, loss of SMCHD1 repressive activity causes expression of double homeobox 4 (DUX4) in muscle tissue, where it is toxic. Studies of arhinia patients suggest a primary defect in nasal placode cells (human nose progenitors). Here, we show that upon SMCHD1 ablation, DUX4 becomes derepressed in H9 human embryonic stem cells (hESCs) as they differentiate toward a placode cell fate, triggering cell death. Arhinia and FSHD2 patient-derived induced pluripotent stem cells (iPSCs) express DUX4 when converted to placode cells and demonstrate variable degrees of cell death, suggesting an environmental disease modifier. HSV-1 may be one such modifier as herpesvirus infection amplifies DUX4 expression in SMCHD1 KO hESC and patient iPSC. These studies suggest that arhinia, like FSHD2, is due to compromised SMCHD1 repressive activity in a cell-specific context and provide evidence for an environmental modifier.
Collapse
Affiliation(s)
- Kaoru Inoue
- Pediatric Neuroendocrinology Group, Clinical Research Branch, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC, USA
| | - Hamed Bostan
- Integrative Bioinformatics, NIEHS, Research Triangle Park, NC, USA
| | - MaKenna R. Browne
- Pediatric Neuroendocrinology Group, Clinical Research Branch, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC, USA
| | - Owen F. Bevis
- Pediatric Neuroendocrinology Group, Clinical Research Branch, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC, USA
| | - Carl D. Bortner
- Signal Transduction Laboratory, NIEHS, Research Triangle Park, NC, USA
| | - Steven A. Moore
- Department of Pathology, University of Iowa Carver College of Medicine and Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Iowa City, IA, USA
| | - Aaron A. Stence
- University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | | | - Shih-Heng Chen
- Viral Vector Core, NIEHS, Research Triangle Park, NC, USA
| | | | - Jian-Liang Li
- Integrative Bioinformatics, NIEHS, Research Triangle Park, NC, USA
| | - Natalie D. Shaw
- Pediatric Neuroendocrinology Group, Clinical Research Branch, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC, USA
| |
Collapse
|
12
|
Halmi CA, Wu CY, Taneyhill LA. Neural crest cell-placodal neuron interactions are mediated by Cadherin-7 and N-cadherin during early chick trigeminal ganglion assembly. F1000Res 2022; 11:741. [PMID: 36128560 PMCID: PMC9475207 DOI: 10.12688/f1000research.122686.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 02/12/2024] Open
Abstract
Background: Arising at distinct positions in the head, the cranial ganglia are crucial for integrating various sensory inputs. The largest of these ganglia is the trigeminal ganglion, which relays pain, touch and temperature information through its three primary nerve branches to the central nervous system. The trigeminal ganglion and its nerves are composed of derivatives of two critical embryonic cell types, neural crest cells and placode cells, that migrate from different anatomical locations, coalesce together, and differentiate to form trigeminal sensory neurons and supporting glia. While the dual cellular origin of the trigeminal ganglion has been known for over 60 years, molecules expressed by neural crest cells and placode cells that regulate initial ganglion assembly remain obscure. Prior studies revealed the importance of cell surface cadherin proteins during early trigeminal gangliogenesis, with Cadherin-7 and neural cadherin (N-cadherin) expressed in neural crest cells and placode cells, respectively. Although cadherins typically interact in a homophilic ( i.e., like) fashion, the presence of different cadherins on these intermingling cell populations raises the question as to whether heterophilic cadherin interactions may also be occurring during initial trigeminal ganglion formation, which was the aim of this study. Methods: To assess potential interactions between Cadherin-7 and N-cadherin, we used biochemistry and innovative imaging assays conducted in vitro and in vivo, including in the forming chick trigeminal ganglion. Results: Our data revealed a physical interaction between Cadherin-7 and N-cadherin. Conclusions: These studies identify a new molecular basis by which neural crest cells and placode cells can aggregate in vivo to build the trigeminal ganglion during embryogenesis.
Collapse
Affiliation(s)
- Caroline A. Halmi
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
| | - Chyong-Yi Wu
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
- U.S. Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Lisa A. Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
| |
Collapse
|
13
|
Halmi CA, Wu CY, Taneyhill LA. Neural crest cell-placodal neuron interactions are mediated by Cadherin-7 and N-cadherin during early chick trigeminal ganglion assembly. F1000Res 2022. [PMID: 36128560 DOI: 10.13016/llyh-dppy] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Arising at distinct positions in the head, the cranial ganglia are crucial for integrating various sensory inputs. The largest of these ganglia is the trigeminal ganglion, which relays pain, touch and temperature information through its three primary nerve branches to the central nervous system. The trigeminal ganglion and its nerves are composed of derivatives of two critical embryonic cell types, neural crest cells and placode cells, that migrate from different anatomical locations, coalesce together, and differentiate to form trigeminal sensory neurons and supporting glia. While the dual cellular origin of the trigeminal ganglion has been known for over 60 years, molecules expressed by neural crest cells and placode cells that regulate initial ganglion assembly remain obscure. Prior studies revealed the importance of cell surface cadherin proteins during early trigeminal gangliogenesis, with Cadherin-7 and neural cadherin (N-cadherin) expressed in neural crest cells and placode cells, respectively. Although cadherins typically interact in a homophilic ( i.e., like) fashion, the presence of different cadherins on these intermingling cell populations raises the question as to whether heterophilic cadherin interactions may also be occurring during initial trigeminal ganglion formation, which was the aim of this study. Methods: To assess potential interactions between Cadherin-7 and N-cadherin, we used biochemistry and innovative imaging assays conducted in vitro and in vivo, including in the forming chick trigeminal ganglion. Results: Our data revealed a physical interaction between Cadherin-7 and N-cadherin. Conclusions: These studies identify a new molecular basis by which neural crest cells and placode cells can aggregate in vivo to build the trigeminal ganglion during embryogenesis.
Collapse
Affiliation(s)
- Caroline A Halmi
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
| | - Chyong-Yi Wu
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA.,U.S. Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
| |
Collapse
|
14
|
Halmi CA, Wu CY, Taneyhill LA. Neural crest cell-placodal neuron interactions are mediated by Cadherin-7 and N-cadherin during early chick trigeminal ganglion assembly. F1000Res 2022; 11:741. [PMID: 36128560 PMCID: PMC9475207 DOI: 10.12688/f1000research.122686.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Arising at distinct positions in the head, the cranial ganglia are crucial for integrating various sensory inputs. The largest of these ganglia is the trigeminal ganglion, which relays pain, touch and temperature information through its three primary nerve branches to the central nervous system. The trigeminal ganglion and its nerves are composed of derivatives of two critical embryonic cell types, neural crest cells and placode cells, that migrate from different anatomical locations, coalesce together, and differentiate to form trigeminal sensory neurons and supporting glia. While the dual cellular origin of the trigeminal ganglion has been known for over 60 years, molecules expressed by neural crest cells and placode cells that regulate initial ganglion assembly remain obscure. Prior studies revealed the importance of cell surface cadherin proteins during early trigeminal gangliogenesis, with Cadherin-7 and neural cadherin (N-cadherin) expressed in neural crest cells and placode cells, respectively. Although cadherins typically interact in a homophilic ( i.e., like) fashion, the presence of different cadherins expressed in neural crest cells and placode cells raises the question as to whether heterophilic cadherin interactions may also be occurring. Given this, the aim of the study was to understand whether Cadherin-7 and N-cadherin were interacting during initial trigeminal ganglion formation. Methods: To assess potential interactions between Cadherin-7 and N-cadherin, we used biochemistry and innovative imaging assays conducted in vitro and in vivo, including in the forming chick trigeminal ganglion. Results: Our data revealed a physical interaction between Cadherin-7 and N-cadherin. Conclusions: These studies identify a new molecular basis by which neural crest cells and placode cells can aggregate in vivo to build the trigeminal ganglion during embryogenesis.
Collapse
Affiliation(s)
- Caroline A. Halmi
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
| | - Chyong-Yi Wu
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
- U.S. Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Lisa A. Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
| |
Collapse
|
15
|
Friedmacher F, Rolle U, Puri P. Genetically Modified Mouse Models of Congenital Diaphragmatic Hernia: Opportunities and Limitations for Studying Altered Lung Development. Front Pediatr 2022; 10:867307. [PMID: 35633948 PMCID: PMC9136148 DOI: 10.3389/fped.2022.867307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/18/2022] [Indexed: 11/21/2022] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a relatively common and life-threatening birth defect, characterized by an abnormal opening in the primordial diaphragm that interferes with normal lung development. As a result, CDH is accompanied by immature and hypoplastic lungs, being the leading cause of morbidity and mortality in patients with this condition. In recent decades, various animal models have contributed novel insights into the pathogenic mechanisms underlying CDH and associated pulmonary hypoplasia. In particular, the generation of genetically modified mouse models, which show both diaphragm and lung abnormalities, has resulted in the discovery of multiple genes and signaling pathways involved in the pathogenesis of CDH. This article aims to offer an up-to-date overview on CDH-implicated transcription factors, molecules regulating cell migration and signal transduction as well as components contributing to the formation of extracellular matrix, whilst also discussing the significance of these genetic models for studying altered lung development with regard to the human situation.
Collapse
Affiliation(s)
- Florian Friedmacher
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Udo Rolle
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Prem Puri
- Beacon Hospital, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Karpinski BA, Maynard TM, Bryan CA, Yitsege G, Horvath A, Lee NH, Moody SA, LaMantia AS. Selective disruption of trigeminal sensory neurogenesis and differentiation in a mouse model of 22q11.2 deletion syndrome. Dis Model Mech 2022; 15:dmm047357. [PMID: 33722956 PMCID: PMC8126478 DOI: 10.1242/dmm.047357] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
22q11.2 Deletion Syndrome (22q11DS) is a neurodevelopmental disorder associated with cranial nerve anomalies and disordered oropharyngeal function, including pediatric dysphagia. Using the LgDel 22q11DS mouse model, we investigated whether sensory neuron differentiation in the trigeminal ganglion (CNgV), which is essential for normal orofacial function, is disrupted. We did not detect changes in cranial placode cell translocation or neural crest migration at early stages of LgDel CNgV development. However, as the ganglion coalesces, proportions of placode-derived LgDel CNgV cells increase relative to neural crest cells. In addition, local aggregation of placode-derived cells increases and aggregation of neural crest-derived cells decreases in LgDel CNgV. This change in cell-cell relationships was accompanied by altered proliferation of placode-derived cells at embryonic day (E)9.5, and premature neurogenesis from neural crest-derived precursors, reflected by an increased frequency of asymmetric neurogenic divisions for neural crest-derived precursors by E10.5. These early differences in LgDel CNgV genesis prefigure changes in sensory neuron differentiation and gene expression by postnatal day 8, when early signs of cranial nerve dysfunction associated with pediatric dysphagia are observed in LgDel mice. Apparently, 22q11 deletion destabilizes CNgV sensory neuron genesis and differentiation by increasing variability in cell-cell interaction, proliferation and sensory neuron differentiation. This early developmental divergence and its consequences may contribute to oropharyngeal dysfunction, including suckling, feeding and swallowing disruptions at birth, and additional orofacial sensory/motor deficits throughout life.
Collapse
Affiliation(s)
- Beverly A. Karpinski
- Department of Anatomy and Cell Biology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Thomas M. Maynard
- The Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA
| | - Corey A. Bryan
- Department of Anatomy and Cell Biology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Gelila Yitsege
- Department of Anatomy and Cell Biology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Anelia Horvath
- Department of Pharmacology and Physiology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Norman H. Lee
- Department of Pharmacology and Physiology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Sally A. Moody
- Department of Anatomy and Cell Biology, The George Washington School of Medicine and Health Sciences, Washington DC, 20037, USA
| | - Anthony-Samuel LaMantia
- The Fralin Biomedical Research Institute, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060, USA
| |
Collapse
|
17
|
Leonard CE, Quiros J, Lefcort F, Taneyhill LA. Loss of Elp1 disrupts trigeminal ganglion neurodevelopment in a model of familial dysautonomia. eLife 2022; 11:71455. [PMID: 35713404 PMCID: PMC9273214 DOI: 10.7554/elife.71455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 06/17/2022] [Indexed: 01/28/2023] Open
Abstract
Familial dysautonomia (FD) is a sensory and autonomic neuropathy caused by mutations in elongator complex protein 1 (ELP1). FD patients have small trigeminal nerves and impaired facial pain and temperature perception. These signals are relayed by nociceptive neurons in the trigeminal ganglion, a structure that is composed of both neural crest- and placode-derived cells. Mice lacking Elp1 in neural crest derivatives ('Elp1 CKO') are born with small trigeminal ganglia, suggesting Elp1 is important for trigeminal ganglion development, yet the function of Elp1 in this context is unknown. We demonstrate that Elp1, expressed in both neural crest- and placode-derived neurons, is not required for initial trigeminal ganglion formation. However, Elp1 CKO trigeminal neurons exhibit abnormal axon outgrowth and deficient target innervation. Developing nociceptors expressing the receptor TrkA undergo early apoptosis in Elp1 CKO, while TrkB- and TrkC-expressing neurons are spared, indicating Elp1 supports the target innervation and survival of trigeminal nociceptors. Furthermore, we demonstrate that specific TrkA deficits in the Elp1 CKO trigeminal ganglion reflect the neural crest lineage of most TrkA neurons versus the placodal lineage of most TrkB and TrkC neurons. Altogether, these findings explain defects in cranial gangliogenesis that may lead to loss of facial pain and temperature sensation in FD.
Collapse
Affiliation(s)
- Carrie E Leonard
- Department of Avian and Animal Sciences, University of Maryland, College ParkCollege ParkUnited States
| | - Jolie Quiros
- Department of Avian and Animal Sciences, University of Maryland, College ParkCollege ParkUnited States
| | - Frances Lefcort
- Department of Microbiology and Cell Biology, Montana State UniversityBozemanUnited States
| | - Lisa A Taneyhill
- Department of Avian and Animal Sciences, University of Maryland, College ParkCollege ParkUnited States
| |
Collapse
|
18
|
Slit2 is necessary for optic axon organization in the zebrafish ventral midline. Cells Dev 2021; 166:203677. [PMID: 33994352 DOI: 10.1016/j.cdev.2021.203677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Slit-Robo signaling has been implicated in regulating several steps of retinal ganglion cell axon guidance, with a central role assigned to Slit2. We report here the phenotypical characterization of a CRISPR-Cas9-generated zebrafish null mutant for this gene, along with a detailed analysis of its expression pattern by WM-FISH. All evident defects in the optic axons in slit2-/- mutants were detected outside the retina, coincident with the major sites of expression at the ventral forebrain, around the developing optic nerve and anterior to the optic chiasm/proximal tract. Anterograde axon tracing experiments in zygotic and maternal-zygotic mutants, as well as morphants, showed the occurrence of axon sorting defects, which appeared mild at the optic nerve level, but more severe in the optic chiasm and the proximal tract. A remarkable sorting defect was the usual splitting of one of the optic nerves in two branches that surrounded the contralateral nerve at the chiasm. Although all axons eventually crossed the midline, the retinotopic order appeared lost at the proximal optic tract, to eventually correct distally. Time-lapse analysis demonstrated the sporadic occurrence of axon misrouting at the chiasm level, which could be responsible for the sorting errors. Our results support previous evidence of a channeling role for Slit molecules in retinal ganglion cell axons at the optic nerve, in addition to a function in the segregation of axons coming from each nerve and from different retinal regions at the medio-ventral area of the forebrain.
Collapse
|
19
|
Nakamura H, Doi T, Puri P, Friedmacher F. Transgenic animal models of congenital diaphragmatic hernia: a comprehensive overview of candidate genes and signaling pathways. Pediatr Surg Int 2020; 36:991-997. [PMID: 32591848 PMCID: PMC7385019 DOI: 10.1007/s00383-020-04705-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2020] [Indexed: 01/10/2023]
Abstract
Congenital diaphragmatic hernia (CDH) is a relatively common and life-threatening birth defect, characterized by incomplete formation of the diaphragm. Because CDH herniation occurs at the same time as preacinar airway branching, normal lung development becomes severely disrupted, resulting almost invariably in pulmonary hypoplasia. Despite various research efforts over the past decades, the pathogenesis of CDH and associated lung hypoplasia remains poorly understood. With the advent of molecular techniques, transgenic animal models of CDH have generated a large number of candidate genes, thus providing a novel basis for future research and treatment. This review article offers a comprehensive overview of genes and signaling pathways implicated in CDH etiology, whilst also discussing strengths and limitations of transgenic animal models in relation to the human condition.
Collapse
Affiliation(s)
- Hiroki Nakamura
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland ,Department of Pediatric Surgery, Kansai Medical University, Osaka, Japan
| | - Takashi Doi
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland ,Department of Pediatric Surgery, Kansai Medical University, Osaka, Japan
| | - Prem Puri
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland ,Beacon Hospital, University College Dublin, Dublin, Ireland
| | - Florian Friedmacher
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland ,Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
20
|
Trans-Axonal Signaling in Neural Circuit Wiring. Int J Mol Sci 2020; 21:ijms21145170. [PMID: 32708320 PMCID: PMC7404203 DOI: 10.3390/ijms21145170] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/24/2022] Open
Abstract
The development of neural circuits is a complex process that relies on the proper navigation of axons through their environment to their appropriate targets. While axon–environment and axon–target interactions have long been known as essential for circuit formation, communication between axons themselves has only more recently emerged as another crucial mechanism. Trans-axonal signaling governs many axonal behaviors, including fasciculation for proper guidance to targets, defasciculation for pathfinding at important choice points, repulsion along and within tracts for pre-target sorting and target selection, repulsion at the target for precise synaptic connectivity, and potentially selective degeneration for circuit refinement. This review outlines the recent advances in identifying the molecular mechanisms of trans-axonal signaling and discusses the role of axon–axon interactions during the different steps of neural circuit formation.
Collapse
|
21
|
Yuan T, York JR, McCauley DW. Neural crest and placode roles in formation and patterning of cranial sensory ganglia in lamprey. Genesis 2020; 58:e23356. [PMID: 32049434 DOI: 10.1002/dvg.23356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 11/07/2022]
Abstract
Vertebrates possess paired cranial sensory ganglia derived from two embryonic cell populations, neural crest and placodes. Cranial sensory ganglia arose prior to the divergence of jawed and jawless vertebrates, but the developmental mechanisms that facilitated their evolution are unknown. Using gene expression and cell lineage tracing experiments in embryos of the sea lamprey, Petromyzon marinus, we find that in the cranial ganglia we targeted, development consists of placode-derived neuron clusters in the core of ganglia, with neural crest cells mostly surrounding these neuronal clusters. To dissect functional roles of neural crest and placode cell associations in these developing cranial ganglia, we used CRISPR/Cas9 gene editing experiments to target genes critical for the development of each population. Genetic ablation of SoxE2 and FoxD-A in neural crest cells resulted in differentiated cranial sensory neurons with abnormal morphologies, whereas deletion of DlxB in cranial placodes resulted in near-total loss of cranial sensory neurons. Taken together, our cell-lineage, gene expression, and gene editing results suggest that cranial neural crest cells may not be required for cranial ganglia specification but are essential for shaping the morphology of these sensory structures. We propose that the association of neural crest and placodes in the head of early vertebrates was a key step in the organization of neurons and glia into paired sensory ganglia.
Collapse
Affiliation(s)
- Tian Yuan
- Department of Biology, University of Oklahoma, Norman, Oklahoma
| | - Joshua R York
- Department of Biology, University of Oklahoma, Norman, Oklahoma
| | | |
Collapse
|
22
|
Kosti A, Du L, Shivram H, Qiao M, Burns S, Garcia JG, Pertsemlidis A, Iyer VR, Kokovay E, Penalva LOF. ELF4 Is a Target of miR-124 and Promotes Neuroblastoma Proliferation and Undifferentiated State. Mol Cancer Res 2020; 18:68-78. [PMID: 31624087 PMCID: PMC6942226 DOI: 10.1158/1541-7786.mcr-19-0187] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/06/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022]
Abstract
13-Cis-retinoic acid (RA) is typically used in postremission maintenance therapy in patients with neuroblastoma. However, side effects and recurrence are often observed. We investigated the use of miRNAs as a strategy to replace RA as promoters of differentiation. miR-124 was identified as the top candidate in a functional screen. Genomic target analysis indicated that repression of a network of transcription factors (TF) could be mediating most of miR-124's effect in driving differentiation. To advance miR-124 mimic use in therapy and better define its mechanism of action, a high-throughput siRNA morphologic screen focusing on its TF targets was conducted and ELF4 was identified as a leading candidate for miR-124 repression. By altering its expression levels, we showed that ELF4 maintains neuroblastoma in an undifferentiated state and promotes proliferation. Moreover, ELF4 transgenic expression was able to counteract the neurogenic effect of miR-124 in neuroblastoma cells. With RNA sequencing, we established the main role of ELF4 to be regulation of cell-cycle progression, specifically through the DREAM complex. Interestingly, several cell-cycle genes activated by ELF4 are repressed by miR-124, suggesting that they might form a TF-miRNA regulatory loop. Finally, we showed that high ELF4 expression is often observed in neuroblastomas and is associated with poor survival. IMPLICATIONS: miR-124 induces neuroblastoma differentiation partially through the downregulation of TF ELF4, which drives neuroblastoma proliferation and its undifferentiated phenotype.
Collapse
Affiliation(s)
- Adam Kosti
- Department of Cell Systems and Anatomy, UT Health Science Center at San Antonio, San Antonio, Texas
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas
| | - Liqin Du
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas
| | - Haridha Shivram
- Department of Molecular Biosciences and Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, Texas
| | - Mei Qiao
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas
| | - Suzanne Burns
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas
| | - Juan Gabriel Garcia
- Department of Cell Systems and Anatomy, UT Health Science Center at San Antonio, San Antonio, Texas
| | - Alexander Pertsemlidis
- Department of Cell Systems and Anatomy, UT Health Science Center at San Antonio, San Antonio, Texas
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas
- Department of Pediatrics, UT Health Science Center at San Antonio, San Antonio, Texas
| | - Vishwanath R Iyer
- Department of Molecular Biosciences and Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, Texas
| | - Erzsebet Kokovay
- Department of Cell Systems and Anatomy, UT Health Science Center at San Antonio, San Antonio, Texas
| | - Luiz O F Penalva
- Department of Cell Systems and Anatomy, UT Health Science Center at San Antonio, San Antonio, Texas.
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
23
|
Kim M, Lee CH, Barnum SJ, Watson RC, Li J, Mastick GS. Slit/Robo signals prevent spinal motor neuron emigration by organizing the spinal cord basement membrane. Dev Biol 2019; 455:449-457. [PMID: 31356769 PMCID: PMC6842423 DOI: 10.1016/j.ydbio.2019.07.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 01/30/2023]
Abstract
The developing spinal cord builds a boundary between the CNS and the periphery, in the form of a basement membrane. The spinal cord basement membrane is a barrier that retains CNS neuron cell bodies, while being selectively permeable to specific axon types. Spinal motor neuron cell bodies are located in the ventral neural tube next to the floor plate and project their axons out through the basement membrane to peripheral targets. However, little is known about how spinal motor neuron cell bodies are retained inside the ventral neural tube, while their axons can exit. In previous work, we found that disruption of Slit/Robo signals caused motor neuron emigration outside the spinal cord. In the current study, we investigate how Slit/Robo signals are necessary to keep spinal motor neurons within the neural tube. Our findings show that when Slit/Robo signals were removed from motor neurons, they migrated outside the spinal cord. Furthermore, this emigration was associated with abnormal basement membrane protein expression in the ventral spinal cord. Using Robo2 and Slit2 conditional mutants, we found that motor neuron-derived Slit/Robo signals were required to set up a normal basement membrane in the spinal cord. Together, our results suggest that motor neurons produce Slit signals that are required for the basement membrane assembly to retain motor neuron cell bodies within the spinal cord.
Collapse
Affiliation(s)
- Minkyung Kim
- Department of Biology, University of Nevada, Reno, NV, 89557, USA.
| | - Clare H Lee
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Sarah J Barnum
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Roland Cj Watson
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Jennifer Li
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Grant S Mastick
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| |
Collapse
|
24
|
Wu CY, Taneyhill LA. Cadherin-7 mediates proper neural crest cell-placodal neuron interactions during trigeminal ganglion assembly. Genesis 2018; 57:e23264. [PMID: 30461190 DOI: 10.1002/dvg.23264] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/18/2018] [Accepted: 11/18/2018] [Indexed: 12/28/2022]
Abstract
The cranial trigeminal ganglia play a vital role in the peripheral nervous system through their relay of sensory information from the vertebrate head to the brain. These ganglia are generated from the intermixing and coalescence of two distinct cell populations: cranial neural crest cells and placodal neurons. Trigeminal ganglion assembly requires the formation of cadherin-based adherens junctions within the neural crest cell and placodal neuron populations; however, the molecular composition of these adherens junctions is still unknown. Herein, we aimed to define the spatio-temporal expression pattern and function of Cadherin-7 during early chick trigeminal ganglion formation. Our data reveal that Cadherin-7 is expressed exclusively in migratory cranial neural crest cells and is absent from trigeminal neurons. Using molecular perturbation experiments, we demonstrate that modulation of Cadherin-7 in neural crest cells influences trigeminal ganglion assembly, including the organization of neural crest cells and placodal neurons within the ganglionic anlage. Moreover, alterations in Cadherin-7 levels lead to changes in the morphology of trigeminal neurons. Taken together, these findings provide additional insight into the role of cadherin-based adhesion in trigeminal ganglion formation, and, more broadly, the molecular mechanisms that orchestrate the cellular interactions essential for cranial gangliogenesis.
Collapse
Affiliation(s)
- Chyong-Yi Wu
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland
| |
Collapse
|
25
|
Rogers CD. Data on the effects of N-cadherin perturbation on the expression of type II cadherin proteins and major signaling pathways. Data Brief 2018; 20:419-425. [PMID: 30175208 PMCID: PMC6116335 DOI: 10.1016/j.dib.2018.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/01/2018] [Accepted: 08/09/2018] [Indexed: 11/28/2022] Open
Abstract
This article contains data related to the research article entitled, "A catenin-dependent balance between N-cadherin and E-cadherin controls neuroectodermal cell fate choices" (Rogers et. al., 2018) [1]. The data presented here include (1) proximity ligation assays using antibodies that recognize type I cadherins (N-cadherin and E-cadherin) co-incubated with antibodies against type II cadherins (Cadherin-6B and Cadherin-11) to test heterotypic interactions in vivo; (2) expression of Cadherin-6B and Cadherin-7 after electroporation with full length N-cadherin and N-cadherin translation-blocking morpholino; and (3) expression of WNT, Notch and TGF-β signaling reporters and effectors after loss of N-cadherin protein in chicken embryos.
Collapse
|
26
|
Rogers CD, Sorrells LK, Bronner ME. A catenin-dependent balance between N-cadherin and E-cadherin controls neuroectodermal cell fate choices. Mech Dev 2018; 152:44-56. [PMID: 30009960 PMCID: PMC6112866 DOI: 10.1016/j.mod.2018.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/16/2018] [Accepted: 07/02/2018] [Indexed: 01/19/2023]
Abstract
Characterizing endogenous protein expression, interaction and function, this study identifies in vivo interactions and competitive balance between N-cadherin and E-cadherin in developing avian (Gallus gallus) neural and neural crest cells. Numerous cadherin proteins, including neural cadherin (Ncad) and epithelial cadherin (Ecad), are expressed in the developing neural plate as well as in neural crest cells as they delaminate from the newly closed neural tube. To clarify independent or coordinate function during development, we examined their expression in the cranial region. The results revealed surprising overlap and distinct localization of Ecad and Ncad in the neural tube. Using a proximity ligation assay and co-immunoprecipitation, we found that Ncad and Ecad formed heterotypic complexes in the developing neural tube, and that modulation of Ncad levels led to reciprocal gain or reduction of Ecad protein, which then alters ectodermal cell fate. Here, we demonstrate that the balance of Ecad and Ncad is dependent upon the availability of β-catenin proteins, and that alteration of either classical cadherin modifies the proportions of the neural crest and neuroectodermal cells that are specified.
Collapse
Affiliation(s)
- Crystal D Rogers
- Department of Biology, California State University, Northridge, Northridge, CA 91330, United States of America.
| | - Lisa K Sorrells
- Department of Biology, California State University, Northridge, Northridge, CA 91330, United States of America.
| | - Marianne E Bronner
- Division of Biology and Biological Engineering 139-74, California Institute of Technology, Pasadena, CA 91125, United States of America.
| |
Collapse
|
27
|
Martinez D, Zuhdi N, Reyes M, Ortega B, Giovannone D, Lee VM, de Bellard ME. Screen for Slit/Robo signaling in trunk neural cells reveals new players. Gene Expr Patterns 2018; 28:22-33. [PMID: 29427758 PMCID: PMC5980643 DOI: 10.1016/j.gep.2018.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/19/2018] [Accepted: 01/20/2018] [Indexed: 01/15/2023]
Abstract
Slits ligands and their Robo receptors are involved in quite disparate cell signaling pathways that include axon guidance, cell proliferation, cell motility and angiogenesis. Neural crest cells emerge by delamination from neural cells in the dorsal neural tube, and give rise to various components of the peripheral nervous system in vertebrates. It is well established that these cells change from a non-migratory to a highly migratory state allowing them to reach distant regions before they differentiate. However, but the mechanism controlling this delamination and subsequent migration are still not fully understood. The repulsive Slit ligand family members, have been classified also as true tumor suppressor molecules. The present study explored in further detail what possible Slit/Robo signals are at play in the trunk neural cells and neural crest cells by carrying out a microarray after Slit2 gain of function in trunk neural tubes. We found that in addition to molecules known to be downstream of Slit/Robo signaling, there were a large set of molecules known to be important in maintaining cells in non-motile, epithelia phenotype. Furthermore, we found new molecules previously not associated with Slit/Robo signaling: cell proliferation markers, Ankyrins and RAB intracellular transporters. Our findings suggest that neural crest cells use and array of different Slit/Robo pathways during their transformation from non-motile to highly motile cells.
Collapse
Affiliation(s)
- Darwin Martinez
- California State University Northridge, Biology Dept., MC 8303, 18111 Nordhoff Street, Northridge, CA, 91330, United States
| | - Nora Zuhdi
- California State University Northridge, Biology Dept., MC 8303, 18111 Nordhoff Street, Northridge, CA, 91330, United States
| | - Michelle Reyes
- California State University Northridge, Biology Dept., MC 8303, 18111 Nordhoff Street, Northridge, CA, 91330, United States
| | - Blanca Ortega
- California State University Northridge, Biology Dept., MC 8303, 18111 Nordhoff Street, Northridge, CA, 91330, United States
| | - Dion Giovannone
- California State University Northridge, Biology Dept., MC 8303, 18111 Nordhoff Street, Northridge, CA, 91330, United States
| | - Vivian M Lee
- California State University Northridge, Biology Dept., MC 8303, 18111 Nordhoff Street, Northridge, CA, 91330, United States
| | - Maria Elena de Bellard
- California State University Northridge, Biology Dept., MC 8303, 18111 Nordhoff Street, Northridge, CA, 91330, United States.
| |
Collapse
|
28
|
Li Y, Zhang XT, Wang XY, Wang G, Chuai M, Münsterberg A, Yang X. Robo signaling regulates the production of cranial neural crest cells. Exp Cell Res 2017; 361:73-84. [PMID: 28987541 DOI: 10.1016/j.yexcr.2017.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/08/2017] [Accepted: 10/03/2017] [Indexed: 11/27/2022]
Abstract
Slit/Robo signaling plays an important role in the guidance of developing neurons in developing embryos. However, it remains obscure whether and how Slit/Robo signaling is involved in the production of cranial neural crest cells. In this study, we examined Robo1 deficient mice to reveal developmental defects of mouse cranial frontal and parietal bones, which are derivatives of cranial neural crest cells. Therefore, we determined the production of HNK1+ cranial neural crest cells in early chick embryo development after knock-down (KD) of Robo1 expression. Detection of markers for pre-migratory and migratory neural crest cells, PAX7 and AP-2α, showed that production of both was affected by Robo1 KD. In addition, we found that the transcription factor slug is responsible for the aberrant delamination/EMT of cranial neural crest cells induced by Robo1 KD, which also led to elevated expression of E- and N-Cadherin. N-Cadherin expression was enhanced when blocking FGF signaling with dominant-negative FGFR1 in half of the neural tube. Taken together, we show that Slit/Robo signaling influences the delamination/EMT of cranial neural crest cells, which is required for cranial bone development.
Collapse
Affiliation(s)
- Yan Li
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China; The key Laboratory of Assisted Circulation, Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiao-Tan Zhang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Xiao-Yu Wang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Guang Wang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Manli Chuai
- Division of Cell and Developmental Biology, University of Dundee, Dundee DD1 5EH, UK
| | - Andrea Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Xuesong Yang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
29
|
Extrinsic mechanical forces mediate retrograde axon extension in a developing neuronal circuit. Nat Commun 2017; 8:282. [PMID: 28819208 PMCID: PMC5561127 DOI: 10.1038/s41467-017-00283-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 06/19/2017] [Indexed: 12/26/2022] Open
Abstract
To form functional neural circuits, neurons migrate to their final destination and extend axons towards their targets. Whether and how these two processes are coordinated in vivo remains elusive. We use the zebrafish olfactory placode as a system to address the underlying mechanisms. Quantitative live imaging uncovers a choreography of directed cell movements that shapes the placode neuronal cluster: convergence of cells towards the centre of the placodal domain and lateral cell movements away from the brain. Axon formation is concomitant with lateral movements and occurs through an unexpected, retrograde mode of extension, where cell bodies move away from axon tips attached to the brain surface. Convergence movements are active, whereas cell body lateral displacements are of mainly passive nature, likely triggered by compression forces from converging neighbouring cells. These findings unravel a previously unknown mechanism of neuronal circuit formation, whereby extrinsic mechanical forces drive the retrograde extension of axons.How neuronal migration and axon growth coordinate during development is only partially understood. Here the authors use quantitative imaging to characterise the morphogenesis of the zebrafish olfactory placode and report an unexpected phenomenon, whereby axons extend through the passive movement of neuron cell bodies away from tethered axon tips.
Collapse
|
30
|
Friedl P, Mayor R. Tuning Collective Cell Migration by Cell-Cell Junction Regulation. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a029199. [PMID: 28096261 DOI: 10.1101/cshperspect.a029199] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Collective cell migration critically depends on cell-cell interactions coupled to a dynamic actin cytoskeleton. Important cell-cell adhesion receptor systems implicated in controlling collective movements include cadherins, immunoglobulin superfamily members (L1CAM, NCAM, ALCAM), Ephrin/Eph receptors, Slit/Robo, connexins and integrins, and an adaptive array of intracellular adapter and signaling proteins. Depending on molecular composition and signaling context, cell-cell junctions adapt their shape and stability, and this gradual junction plasticity enables different types of collective cell movements such as epithelial sheet and cluster migration, branching morphogenesis and sprouting, collective network migration, as well as coordinated individual-cell migration and streaming. Thereby, plasticity of cell-cell junction composition and turnover defines the type of collective movements in epithelial, mesenchymal, neuronal, and immune cells, and defines migration coordination, anchorage, and cell dissociation. We here review cell-cell adhesion systems and their functions in different types of collective cell migration as key regulators of collective plasticity.
Collapse
Affiliation(s)
- Peter Friedl
- Department of Cell Biology, Radboud University Medical Centre, Nijmegen 6525GA, The Netherlands.,David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030.,Cancer Genomics Center, 3584 CG Utrecht, The Netherlands
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
31
|
Wu MF, Liao CY, Wang LY, Chang JT. The role of Slit-Robo signaling in the regulation of tissue barriers. Tissue Barriers 2017; 5:e1331155. [PMID: 28598714 PMCID: PMC5501134 DOI: 10.1080/21688370.2017.1331155] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 01/12/2023] Open
Abstract
The role of Slit/Robo signaling has extended from initial axon repulsion in the developing nervous system to organ morphogenesis, cancer development and angiogenesis. Slit/Robo signaling regulates similar pathways within these processes. Slit/Robo ensures the homeostasis of the dynamic interaction between cell-cell and cell-matrix interactions. The dysregulation of Slit/Robo signaling damages the tissue barrier, resulting in developmental abnormalities or disease. Here, we summarize how Slit/Robo controls kidney morphogenesis and describe the dual roles of Slit/Robo signaling in the regulation of tumorigenesis and angiogenesis.
Collapse
Affiliation(s)
- Ming-Fang Wu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.
- Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C.
| | - Chen-Yi Liao
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.
| | - Ling-Yi Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.
| | - Jinghua Tsai Chang
- Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan, R.O.C.
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, R.O.C.
| |
Collapse
|
32
|
Bjorke B, Shoja-Taheri F, Kim M, Robinson GE, Fontelonga T, Kim KT, Song MR, Mastick GS. Contralateral migration of oculomotor neurons is regulated by Slit/Robo signaling. Neural Dev 2016; 11:18. [PMID: 27770832 PMCID: PMC5075191 DOI: 10.1186/s13064-016-0073-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 10/11/2016] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Oculomotor neurons develop initially like typical motor neurons, projecting axons out of the ventral midbrain to their ipsilateral targets, the extraocular muscles. However, in all vertebrates, after the oculomotor nerve (nIII) has reached the extraocular muscle primordia, the cell bodies that innervate the superior rectus migrate to join the contralateral nucleus. This motor neuron migration represents a unique strategy to form a contralateral motor projection. Whether migration is guided by diffusible cues remains unknown. METHODS We examined the role of Slit chemorepellent signals in contralateral oculomotor migration by analyzing mutant mouse embryos. RESULTS We found that the ventral midbrain expresses high levels of both Slit1 and 2, and that oculomotor neurons express the repellent Slit receptors Robo1 and Robo2. Therefore, Slit signals are in a position to influence the migration of oculomotor neurons. In Slit 1/2 or Robo1/2 double mutant embryos, motor neuron cell bodies migrated into the ventral midbrain on E10.5, three days prior to normal migration. These early migrating neurons had leading projections into and across the floor plate. In contrast to the double mutants, embryos which were mutant for single Slit or Robo genes did not have premature migration or outgrowth on E10.5, demonstrating a cooperative requirement of Slit1 and 2, as well as Robo1 and 2. To test how Slit/Robo midline repulsion is modulated, we found that the normal migration did not require the receptors Robo3 and CXCR4, or the chemoattractant, Netrin 1. The signal to initiate contralateral migration is likely autonomous to the midbrain because oculomotor neurons migrate in embryos that lack either nerve outgrowth or extraocular muscles, or in cultured midbrains that lacked peripheral tissue. CONCLUSION Overall, our results demonstrate that a migratory subset of motor neurons respond to floor plate-derived Slit repulsion to properly control the timing of contralateral migration.
Collapse
Affiliation(s)
- Brielle Bjorke
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | | | - Minkyung Kim
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - G Eric Robinson
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | | | - Kyung-Tai Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | - Mi-Ryoung Song
- School of Life Sciences, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, 500-712, Republic of Korea
| | - Grant S Mastick
- Department of Biology, University of Nevada, Reno, NV, 89557, USA.
| |
Collapse
|
33
|
Katow H, Katow T, Yoshida H, Kiyomoto M, Uemura I. Immunohistochemical and ultrastructural properties of the larval ciliary band-associated strand in the sea urchin Hemicentrotus pulcherrimus. Front Zool 2016; 13:27. [PMID: 27313654 PMCID: PMC4910247 DOI: 10.1186/s12983-016-0159-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/02/2016] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The swimming activity of sea urchin larvae is dependent on the ciliary band (CB) on the larval surface and is regulated by several neurotransmitters, including serotonin (5HT), dopamine, and γ-aminobutyric acid (GABA). However, the CB signal transmission mechanism remains unknown. The present study investigated the structural relationship between the CB and external signal receptors by immunohistochemical and transmission electron microscopic analyses of sea urchin, Hemicentrotus pulcherrimus, larvae. RESULTS Glutamate decarboxylase (GAD; GABA synthetase) was detected in a strand of multiple cells along the circumoral CB in 6-arm plutei. The GAD-expressing strand was closely associated with the CB on the oral ectoderm side. The ciliary band-associated strand (CBAS) also expressed the 5HT receptor (5HThpr) and encephalopsin (ECPN) throughout the cytoplasm and comprised 1- to 2-μm diameter axon-like long stretched regions and sporadic 6- to 7-μm diameter bulbous nucleated regions (perikarya) that protruded into the oral ectoderm side. Besides the laterally polarized morphology of the CBAS cells, Epith-2, which is the epithelial lateral cell surface-specific protein of the sea urchin embryo and larva, was expressed exclusively by perikarya but not by the axon-like regions. The CBAS exposed its narrow apical surface on the larval epithelium between the CB and squamous cells and formed adherens junctions (AJs) on the apical side between them. Despite the presence of the CBAS axon-like regions, tubulins, such as α-, β-, and acetylated α-tubulins, were not detected. However, the neuroendocrine cell marker protein synaptophysin was detected in the axon-like regions and in bouton-like protrusions that contained numerous small ultrastructural vesicles. CONCLUSIONS The unique morphology of the CBAS in the sea urchin larva epithelium had not been reported. The CBAS expresses a remarkable number of receptors to environmental stimuli and proteins that are probably involved in signal transmission to the CB. The properties of the CBAS explain previous reports that larval swimming is triggered by environmental stimuli and suggest crosstalk among receptors and potential plural sensory functions of the CBAS.
Collapse
Affiliation(s)
- Hideki Katow
- />Research Center for Marine Biology, Tohoku University, Asamushi, Aomori, Aomori 039-3501 Japan
- />Center of Research Instruments, Institute of Development, Aging and Cancer, Tohoku University, Sendai, 980-8575 Japan
| | - Tomoko Katow
- />Research Center for Marine Biology, Tohoku University, Asamushi, Aomori, Aomori 039-3501 Japan
| | - Hiromi Yoshida
- />Center of Research Instruments, Institute of Development, Aging and Cancer, Tohoku University, Sendai, 980-8575 Japan
| | - Masato Kiyomoto
- />Center of Research Instruments, Institute of Development, Aging and Cancer, Tohoku University, Sendai, 980-8575 Japan
- />Marine and Coastal Research Center, Ochanomizu University, Tateyama, Chiba 294-0301 Japan
| | - Isao Uemura
- />Division of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo 192-0397 Japan
| |
Collapse
|
34
|
Patthey C, Clifford H, Haerty W, Ponting CP, Shimeld SM, Begbie J. Identification of molecular signatures specific for distinct cranial sensory ganglia in the developing chick. Neural Dev 2016; 11:3. [PMID: 26819088 PMCID: PMC4730756 DOI: 10.1186/s13064-016-0057-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/08/2016] [Indexed: 11/22/2022] Open
Abstract
Background The cranial sensory ganglia represent populations of neurons with distinct functions, or sensory modalities. The production of individual ganglia from distinct neurogenic placodes with different developmental pathways provides a powerful model to investigate the acquisition of specific sensory modalities. To date there is a limited range of gene markers available to examine the molecular pathways underlying this process. Results Transcriptional profiles were generated for populations of differentiated neurons purified from distinct cranial sensory ganglia using microdissection in embryonic chicken followed by FAC-sorting and RNAseq. Whole transcriptome analysis confirmed the division into somato- versus viscerosensory neurons, with additional evidence for subdivision of the somatic class into general and special somatosensory neurons. Cross-comparison of distinct ganglia transcriptomes identified a total of 134 markers, 113 of which are novel, which can be used to distinguish trigeminal, vestibulo-acoustic and epibranchial neuronal populations. In situ hybridisation analysis provided validation for 20/26 tested markers, and showed related expression in the target region of the hindbrain in many cases. Conclusions One hundred thirty-four high-confidence markers have been identified for placode-derived cranial sensory ganglia which can now be used to address the acquisition of specific cranial sensory modalities. Electronic supplementary material The online version of this article (doi:10.1186/s13064-016-0057-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cedric Patthey
- Department of Zoology, University of Oxford, Oxford, UK. .,Umeå Center for Molecular Medicine, Umeå University, Umeå, Sweden.
| | - Harry Clifford
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK. .,MRC Functional Genomics, University of Oxford, Oxford, UK.
| | - Wilfried Haerty
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK. .,MRC Functional Genomics, University of Oxford, Oxford, UK.
| | - Chris P Ponting
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK. .,MRC Functional Genomics, University of Oxford, Oxford, UK.
| | | | - Jo Begbie
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
35
|
The Order and Place of Neuronal Differentiation Establish the Topography of Sensory Projections and the Entry Points within the Hindbrain. J Neurosci 2015; 35:7475-86. [PMID: 25972174 DOI: 10.1523/jneurosci.3743-14.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Establishing topographical maps of the external world is an important but still poorly understood feature of the vertebrate sensory system. To study the selective innervation of hindbrain regions by sensory afferents in the zebrafish embryo, we mapped the fine-grained topographical representation of sensory projections at the central level by specific photoconversion of sensory neurons. Sensory ganglia located anteriorly project more medially than do ganglia located posteriorly, and this relates to the order of sensory ganglion differentiation. By single-plane illumination microscopy (SPIM) in vivo imaging, we show that (1) the sequence of arrival of cranial ganglion inputs predicts the topography of central projections, and (2) delaminated neuroblasts differentiate in close contact with the neural tube, and they never loose contact with the neural ectoderm. Afferent entrance points are established by plasma membrane interactions between primary differentiated peripheral sensory neurons and neural tube border cells with the cooperation of neural crest cells. These first contacts remain during ensuing morphological growth to establish pioneer axons. Neural crest cells and repulsive slit1/robo2 signals then guide axons from later-differentiating neurons toward the neural tube. Thus, this study proposes a new model by which the topographical representation of cranial sensory ganglia is established by entrance order, with the entry points determined by cell contact between the sensory ganglion cell bodies and the hindbrain.
Collapse
|
36
|
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
|
37
|
Kim M, Fontelonga T, Roesener AP, Lee H, Gurung S, Mendonca PRF, Mastick GS. Motor neuron cell bodies are actively positioned by Slit/Robo repulsion and Netrin/DCC attraction. Dev Biol 2015; 399:68-79. [PMID: 25530182 PMCID: PMC4339514 DOI: 10.1016/j.ydbio.2014.12.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/22/2014] [Accepted: 12/10/2014] [Indexed: 11/29/2022]
Abstract
Motor neurons differentiate from a ventral column of progenitors and settle in static clusters, the motor nuclei, next to the floor plate. Within these cell clusters, motor neurons receive afferent input and project their axons out to muscle targets. The molecular mechanisms that position motor neurons in the neural tube remain poorly understood. The floor plate produces several types of guidance cues with well-known roles in attracting and repelling axons, including the Slit family of chemorepellents via their Robo receptors, and Netrin1 via its DCC attractive receptor. In the present study we found that Islet1(+) motor neuron cell bodies invaded the floor plate of Robo1/2 double mutant mouse embryos or Slit1/2/3 triple mutants. Misplaced neurons were born in their normal progenitor column, but then migrated tangentially into the ventral midline. Robo1 and 2 receptor expression in motor neurons was confirmed by reporter gene staining and anti-Robo antibody labeling. Mis-positioned motor neurons projected their axons longitudinally within the floor plate, and failed to reach their normal exit points. To test for potential counteracting ventral attractive signals, we examined Netrin-1 and DCC mutants, and found that motor neurons shifted dorsally in the hindbrain and spinal cord, suggesting that Netrin-1/DCC signaling normally attracts motor neurons closer to the floor plate. Our results show that motor neurons are actively migrating cells, and are normally trapped in a static position by Slit/Robo repulsion and Netrin-1/DCC attraction.
Collapse
Affiliation(s)
- Minkyung Kim
- Department of Biology, University of Nevada, Reno, NV, USA
| | | | | | - Haeram Lee
- Department of Biology, University of Nevada, Reno, NV, USA
| | - Suman Gurung
- Department of Biology, University of Nevada, Reno, NV, USA
| | | | | |
Collapse
|
38
|
Breau MA, Schneider-Maunoury S. Cranial placodes: models for exploring the multi-facets of cell adhesion in epithelial rearrangement, collective migration and neuronal movements. Dev Biol 2014; 401:25-36. [PMID: 25541234 DOI: 10.1016/j.ydbio.2014.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/08/2014] [Accepted: 12/09/2014] [Indexed: 01/16/2023]
Abstract
Key to morphogenesis is the orchestration of cell movements in the embryo, which requires fine-tuned adhesive interactions between cells and their close environment. The neural crest paradigm has provided important insights into how adhesion dynamics control epithelium-to-mesenchyme transition and mesenchymal cell migration. Much less is known about cranial placodes, patches of ectodermal cells that generate essential parts of vertebrate sensory organs and ganglia. In this review, we summarise the known functions of adhesion molecules in cranial placode morphogenesis, and discuss potential novel implications of adhesive interactions in this crucial developmental process. The great repertoire of placodal cell behaviours offers new avenues for exploring the multiple roles of adhesion complexes in epithelial remodelling, collective migration and neuronal movements.
Collapse
Affiliation(s)
- Marie Anne Breau
- Sorbonne Universités, UPMC Univ Paris 06, IBPS-UMR7622, F-75005 Paris, France; CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, F-75005 Paris, France; INSERM, U1156, F-75005 Paris, France.
| | - Sylvie Schneider-Maunoury
- Sorbonne Universités, UPMC Univ Paris 06, IBPS-UMR7622, F-75005 Paris, France; CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, F-75005 Paris, France; INSERM, U1156, F-75005 Paris, France
| |
Collapse
|
39
|
Migratory neural crest cell αN-catenin impacts chick trigeminal ganglia formation. Dev Biol 2014; 392:295-307. [PMID: 24882712 DOI: 10.1016/j.ydbio.2014.05.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 04/18/2014] [Accepted: 05/21/2014] [Indexed: 01/19/2023]
Abstract
Neural crest cells are an embryonic cell population that is crucial for proper vertebrate development. Initially localized to the dorsal neural folds, premigratory neural crest cells undergo an epithelial-to-mesenchymal transition (EMT) and migrate to their final destinations in the developing embryo. Together with epidermally-derived placode cells, neural crest cells then form the cranial sensory ganglia of the peripheral nervous system. Our prior work has shown that αN-catenin, the neural subtype of the adherens junction α-catenin protein, regulates cranial neural crest cell EMT by controlling premigratory neural crest cell cadherin levels. Although αN-catenin down-regulation is critical for initial neural crest cell EMT, a potential role for αN-catenin in later neural crest cell migration, and formation of the cranial ganglia, has not been examined. In this study, we show for the first time that migratory neural crest cells that will give rise to the cranial trigeminal ganglia express αN-catenin and Cadherin-7. αN-catenin loss- and gain-of-function experiments reveal effects on the migratory neural crest cell population that include subsequent defects in trigeminal ganglia assembly. Moreover, αN-catenin perturbation in neural crest cells impacts the placode cell contribution to the trigeminal ganglia and also changes neural crest cell Cadherin-7 levels and localization. Together, these results highlight a novel function for αN-catenin in migratory neural crest cells that form the trigeminal ganglia.
Collapse
|
40
|
Steventon B, Mayor R, Streit A. Neural crest and placode interaction during the development of the cranial sensory system. Dev Biol 2014; 389:28-38. [PMID: 24491819 PMCID: PMC4439187 DOI: 10.1016/j.ydbio.2014.01.021] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/19/2014] [Accepted: 01/25/2014] [Indexed: 01/13/2023]
Abstract
In the vertebrate head, the peripheral components of the sensory nervous system are derived from two embryonic cell populations, the neural crest and cranial sensory placodes. Both arise in close proximity to each other at the border of the neural plate: neural crest precursors abut the future central nervous system, while placodes originate in a common preplacodal region slightly more lateral. During head morphogenesis, complex events organise these precursors into functional sensory structures, raising the question of how their development is coordinated. Here we review the evidence that neural crest and placode cells remain in close proximity throughout their development and interact repeatedly in a reciprocal manner. We also review recent controversies about the relative contribution of the neural crest and placodes to the otic and olfactory systems. We propose that a sequence of mutual interactions between the neural crest and placodes drives the coordinated morphogenesis that generates functional sensory systems within the head.
Collapse
Affiliation(s)
- Ben Steventon
- Department of Developmental and Stem Cell Biology, Insitut Pasteur, France
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Andrea Streit
- Department of Craniofacial Development and Stem Cell Biology, King׳s College London, London, UK.
| |
Collapse
|
41
|
Fort P, Théveneau E. PleiotRHOpic: Rho pathways are essential for all stages of Neural Crest development. Small GTPases 2014; 5:e27975. [PMID: 24614304 DOI: 10.4161/sgtp.27975] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neural Crest (NC) cells are a multipotent migratory stem cell population unique to vertebrates, which contributes extensively to the formation of a wide array of neural and non-neural structures in the embryo. NC cells originate in the ectoderm at the border of the neural tube, undergo an epithelial-mesenchymal transition and acquire outstanding individual and collective migratory properties that allow them to disseminate and differentiate to different parts of the body. This exquisite capacity to switch from an epithelium to motile cells represents both a puzzling biological issue and an attractive model to address the basic mechanisms of cell migration and their alteration during cancer progression. Here we review how signaling pathways controlled by Rho GTPases, key players in cell adhesion, contraction, migration and polarity, contribute to the control the different phases of NC development.
Collapse
Affiliation(s)
- Philippe Fort
- CNRS; University Montpellier 2; CRBM-UMR5237; Montpellier, France
| | - Eric Théveneau
- CNRS; University Toulouse III; Centre de Biologie du Développement; UMR5547; Toulouse, France
| |
Collapse
|
42
|
Patthey C, Schlosser G, Shimeld SM. The evolutionary history of vertebrate cranial placodes--I: cell type evolution. Dev Biol 2014; 389:82-97. [PMID: 24495912 DOI: 10.1016/j.ydbio.2014.01.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/21/2014] [Accepted: 01/24/2014] [Indexed: 10/25/2022]
Abstract
Vertebrate cranial placodes are crucial contributors to the vertebrate cranial sensory apparatus. Their evolutionary origin has attracted much attention from evolutionary and developmental biologists, yielding speculation and hypotheses concerning their putative homologues in other lineages and the developmental and genetic innovations that might have underlain their origin and diversification. In this article we first briefly review our current understanding of placode development and the cell types and structures they form. We next summarise previous hypotheses of placode evolution, discussing their strengths and caveats, before considering the evolutionary history of the various cell types that develop from placodes. In an accompanying review, we also further consider the evolution of ectodermal patterning. Drawing on data from vertebrates, tunicates, amphioxus, other bilaterians and cnidarians, we build these strands into a scenario of placode evolutionary history and of the genes, cells and developmental processes that underlie placode evolution and development.
Collapse
Affiliation(s)
- Cedric Patthey
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK.
| | - Gerhard Schlosser
- Zoology, School of Natural Sciences & Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, University Road, Galway, Ireland
| | - Sebastian M Shimeld
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| |
Collapse
|
43
|
Abstract
Roundabout receptors (Robo) and their Slit ligands were discovered in the 1990s and found to be key players in axon guidance. Slit was initially described s an extracellular matrix protein that was expressed by midline glia in Drosophila. A few years later, it was shown that, in vertebrates and invertebrates, Slits acted as chemorepellents for axons crossing the midline. Robo proteins were originally discovered in Drosophila in a mutant screen for genes involved in the regulation of midline crossing. This ligand-receptor pair has since been implicated in a variety of other neuronal and non-neuronal processes ranging from cell migration to angiogenesis, tumourigenesis and even organogenesis of tissues such as kidneys, lungs and breasts.
Collapse
|
44
|
Karpinski BA, Maynard TM, Fralish MS, Nuwayhid S, Zohn IE, Moody SA, LaMantia AS. Dysphagia and disrupted cranial nerve development in a mouse model of DiGeorge (22q11) deletion syndrome. Dis Model Mech 2013; 7:245-57. [PMID: 24357327 PMCID: PMC3917245 DOI: 10.1242/dmm.012484] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We assessed feeding-related developmental anomalies in the LgDel mouse model of chromosome 22q11 deletion syndrome (22q11DS), a common developmental disorder that frequently includes perinatal dysphagia – debilitating feeding, swallowing and nutrition difficulties from birth onward – within its phenotypic spectrum. LgDel pups gain significantly less weight during the first postnatal weeks, and have several signs of respiratory infections due to food aspiration. Most 22q11 genes are expressed in anlagen of craniofacial and brainstem regions critical for feeding and swallowing, and diminished expression in LgDel embryos apparently compromises development of these regions. Palate and jaw anomalies indicate divergent oro-facial morphogenesis. Altered expression and patterning of hindbrain transcriptional regulators, especially those related to retinoic acid (RA) signaling, prefigures these disruptions. Subsequently, gene expression, axon growth and sensory ganglion formation in the trigeminal (V), glossopharyngeal (IX) or vagus (X) cranial nerves (CNs) that innervate targets essential for feeding, swallowing and digestion are disrupted. Posterior CN IX and X ganglia anomalies primarily reflect diminished dosage of the 22q11DS candidate gene Tbx1. Genetic modification of RA signaling in LgDel embryos rescues the anterior CN V phenotype and returns expression levels or pattern of RA-sensitive genes to those in wild-type embryos. Thus, diminished 22q11 gene dosage, including but not limited to Tbx1, disrupts oro-facial and CN development by modifying RA-modulated anterior-posterior hindbrain differentiation. These disruptions likely contribute to dysphagia in infants and young children with 22q11DS.
Collapse
Affiliation(s)
- Beverly A Karpinski
- Department of Anatomy and Regenerative Biology, The George Washington University School of Medicine and Health Sciences, Washington DC 20037, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Rogers CD, Saxena A, Bronner ME. Sip1 mediates an E-cadherin-to-N-cadherin switch during cranial neural crest EMT. ACTA ACUST UNITED AC 2013; 203:835-47. [PMID: 24297751 PMCID: PMC3857483 DOI: 10.1083/jcb.201305050] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Sip1 promotes the mesenchymalization stage of the neural crest epithelial-to-mesenchymal transition by inducing a transcriptional switch in cells from expression of E-cadherin to N-cadherin. The neural crest, an embryonic stem cell population, initially resides within the dorsal neural tube but subsequently undergoes an epithelial-to-mesenchymal transition (EMT) to commence migration. Although neural crest and cancer EMTs are morphologically similar, little is known regarding conservation of their underlying molecular mechanisms. We report that Sip1, which is involved in cancer EMT, plays a critical role in promoting the neural crest cell transition to a mesenchymal state. Sip1 transcripts are expressed in premigratory/migrating crest cells. After Sip1 loss, the neural crest specifier gene FoxD3 was abnormally retained in the dorsal neuroepithelium, whereas Sox10, which is normally required for emigration, was diminished. Subsequently, clumps of adherent neural crest cells remained adjacent to the neural tube and aberrantly expressed E-cadherin while lacking N-cadherin. These findings demonstrate two distinct phases of neural crest EMT, detachment and mesenchymalization, with the latter involving a novel requirement for Sip1 in regulation of cadherin expression during completion of neural crest EMT.
Collapse
Affiliation(s)
- Crystal D Rogers
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125
| | | | | |
Collapse
|
46
|
Freter S, Fleenor SJ, Freter R, Liu KJ, Begbie J. Cranial neural crest cells form corridors prefiguring sensory neuroblast migration. Development 2013; 140:3595-600. [PMID: 23942515 PMCID: PMC3742142 DOI: 10.1242/dev.091033] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The majority of cranial sensory neurons originate in placodes in the surface ectoderm, migrating to form ganglia that connect to the central nervous system (CNS). Interactions between inward-migrating sensory neuroblasts and emigrant cranial neural crest cells (NCCs) play a role in coordinating this process, but how the relationship between these two cell populations is established is not clear. Here, we demonstrate that NCCs generate corridors delineating the path of migratory neuroblasts between the placode and CNS in both chick and mouse. In vitro analysis shows that NCCs are not essential for neuroblast migration, yet act as a superior substrate to mesoderm, suggesting provision of a corridor through a less-permissive mesodermal territory. Early organisation of NCC corridors occurs prior to sensory neurogenesis and can be recapitulated in vitro; however, NCC extension to the placode requires placodal neurogenesis, demonstrating reciprocal interactions. Together, our data indicate that NCC corridors impose physical organisation for precise ganglion formation and connection to the CNS, providing a local environment to enclose migrating neuroblasts and axonal processes as they migrate through a non-neural territory.
Collapse
Affiliation(s)
- Sabine Freter
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | | | | | | | | |
Collapse
|
47
|
Domyan ET, Branchfield K, Gibson DA, Naiche L, Lewandoski M, Tessier-Lavigne M, Ma L, Sun X. Roundabout receptors are critical for foregut separation from the body wall. Dev Cell 2013; 24:52-63. [PMID: 23328398 PMCID: PMC3551250 DOI: 10.1016/j.devcel.2012.11.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 10/27/2012] [Accepted: 11/27/2012] [Indexed: 01/08/2023]
Abstract
In mammals, precise placement of organs is essential for survival. We show here that inactivation of Roundabout (Robo) receptors 1 and 2 in mice leads to mispositioning of the stomach in the thoracic instead of the abdominal cavity, which likely contributes to poor lung inflation and lethality at birth, reminiscent of congenital diaphragmatic hernia (CDH) cases in humans. Unexpectedly, in Robo mutant mice, the primary defect preceding organ misplacement and diaphragm malformation is a delayed separation of foregut from the dorsal body wall. Foregut separation is a rarely considered morphogenetic event, and our data indicate that it occurs via repulsion of Robo-expressing foregut cells away from the Slit ligand source. In humans, genomic lesions containing Robo genes have been documented in CDH. Our findings suggest that separation of the foregut from the body wall is genetically controlled and that defects in this event may contribute to CDH.
Collapse
Affiliation(s)
- Eric Thomas Domyan
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kelsey Branchfield
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Daniel A. Gibson
- Department of Cell and Neurobiology, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - L.A. Naiche
- Cancer and Developmental Biology Lab, National Cancer Institute, Frederick, MD 21702, USA
| | - Mark Lewandoski
- Cancer and Developmental Biology Lab, National Cancer Institute, Frederick, MD 21702, USA
| | | | - Le Ma
- Department of Cell and Neurobiology, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033, USA
| | - Xin Sun
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
48
|
McKeown SJ, Wallace AS, Anderson RB. Expression and function of cell adhesion molecules during neural crest migration. Dev Biol 2012; 373:244-57. [PMID: 23123967 DOI: 10.1016/j.ydbio.2012.10.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/18/2012] [Accepted: 10/25/2012] [Indexed: 01/13/2023]
Abstract
Neural crest cells are highly migratory cells that give rise to many derivatives including peripheral ganglia, craniofacial structures and melanocytes. Neural crest cells migrate along defined pathways to their target sites, interacting with each other and their environment as they migrate. Cell adhesion molecules are critical during this process. In this review we discuss the expression and function of cell adhesion molecules during the process of neural crest migration, in particular cadherins, integrins, members of the immunoglobulin superfamily of cell adhesion molecules, and the proteolytic enzymes that cleave these cell adhesion molecules. The expression and function of these cell adhesion molecules and proteases are compared across neural crest emigrating from different axial levels, and across different species of vertebrates.
Collapse
Affiliation(s)
- Sonja J McKeown
- Department of Anatomy and Neuroscience, University of Melbourne, 3010 VIC, Australia.
| | | | | |
Collapse
|
49
|
Sakai N, Insolera R, Sillitoe RV, Shi SH, Kaprielian Z. Axon sorting within the spinal cord marginal zone via Robo-mediated inhibition of N-cadherin controls spinocerebellar tract formation. J Neurosci 2012; 32:15377-87. [PMID: 23115176 PMCID: PMC3511830 DOI: 10.1523/jneurosci.2225-12.2012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 08/23/2012] [Accepted: 09/03/2012] [Indexed: 11/21/2022] Open
Abstract
The axons of spinal projection neurons transmit sensory information to the brain by ascending within highly organized longitudinal tracts. However, the molecular mechanisms that control the sorting of these axons within the spinal cord and their directed growth to poorly defined targets are not understood. Here, we show that an interplay between Robo and the cell adhesion molecule, N-cadherin, sorts spinal commissural axons into appropriate longitudinal tracts within the spinal cord, and thereby facilitates their brain targeting. Specifically, we show that d1 and d2 spinal commissural axons join the lateral funiculus within the spinal cord and target the cerebellum in chick embryos, and that these axons contribute to the spinocerebellar projection in transgenic reporter mice. Disabling Robo signaling or overexpressing N-cadherin on these axons prevents the formation of the lateral funiculus and the spinocerebellar tract, and simultaneously perturbing Robo and N-cadherin function rescues both phenotypes in chick embryos. Consistent with these observations, disabling Robo function in conditional N-cadherin knock-out mice results in a wild-type-like lateral funiculus. Together, these findings suggest that spinal projection axons must be sorted into distinct longitudinal tracts within the spinal cord proper to project to their brain targets.
Collapse
Affiliation(s)
- Nozomi Sakai
- Dominick P. Purpura Department of Neuroscience and
| | - Ryan Insolera
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; and
- Neuroscience Graduate Program, Weill Cornell Medical College, New York, New York 10065
| | | | - Song-Hai Shi
- Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; and
- Neuroscience Graduate Program, Weill Cornell Medical College, New York, New York 10065
| | - Zaven Kaprielian
- Dominick P. Purpura Department of Neuroscience and
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, 10461
| |
Collapse
|
50
|
Giovannone D, Reyes M, Reyes R, Correa L, Martinez D, Ra H, Gomez G, Kaiser J, Ma L, Stein MP, de Bellard ME. Slits affect the timely migration of neural crest cells via Robo receptor. Dev Dyn 2012; 241:1274-88. [PMID: 22689303 PMCID: PMC3632352 DOI: 10.1002/dvdy.23817] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2012] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Neural crest cells emerge by delamination from the dorsal neural tube and give rise to various components of the peripheral nervous system in vertebrate embryos. These cells change from non-motile into highly motile cells migrating to distant areas before further differentiation. Mechanisms controlling delamination and subsequent migration of neural crest cells are not fully understood. Slit2, a chemorepellant for axonal guidance that repels and stimulates motility of trunk neural crest cells away from the gut has recently been suggested to be a tumor suppressor molecule. The goal of this study was to further investigate the role of Slit2 in trunk neural crest cell migration by constitutive expression in neural crest cells. RESULTS We found that Slit gain-of-function significantly impaired neural crest cell migration while Slit loss-of-function favored migration. In addition, we observed that the distribution of key cytoskeletal markers was disrupted in both gain and loss of function instances. CONCLUSIONS These findings suggest that Slit molecules might be involved in the processes that allow neural crest cells to begin migrating and transitioning to a mesenchymal type.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gustavo Gomez
- Division of Biology, 139-74. California Institute of Technology, Pasadena, CA 91125
| | | | - Le Ma
- Zilkha Neurogenetic Institute. KSOM of USC. 1501 San Pablo St. Los Angeles, CA 90033
| | | | | |
Collapse
|