1
|
Jacinto JGP, Letko A, Häfliger IM, Drögemüller C, Agerholm JS. Congenital syndromic Chiari-like malformation (CSCM) in Holstein cattle: towards unravelling of possible genetic causes. Acta Vet Scand 2024; 66:29. [PMID: 38965607 PMCID: PMC11229497 DOI: 10.1186/s13028-024-00752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Chiari malformation type II (CMII) was originally reported in humans as a rare disorder characterized by the downward herniation of the hindbrain and towering cerebellum. The congenital brain malformation is usually accompanied by spina bifida, a congenital spinal anomaly resulting from incomplete closure of the dorsal aspect of the spinal neural tube, and occasionally by other lesions. A similar disorder has been reported in several animal species, including cattle, particularly as a congenital syndrome. A cause of congenital syndromic Chiari-like malformation (CSCM) in cattle has not been reported to date. We collected a series of 14 CSCM-affected Holstein calves (13 purebred, one Red Danish Dairy F1 cross) and performed whole-genome sequencing (WGS). WGS was performed on 33 cattle, including eight cases with parents (trio-based; group 1), three cases with one parent (group 2), and three single cases (solo-based; group 3). RESULTS Sequencing-based genome-wide association study of the 13 Holstein calves with CSCM and 166 controls revealed no significantly associated genome region. Assuming a single Holstein breed-specific recessive allele, no region of shared homozygosity was detected suggesting heterogeneity. Subsequent filtering for protein-changing variants that were only homozygous in the genomes of the individual cases allowed the identification of two missense variants affecting different genes, SHC4 in case 4 in group 1 and WDR45B in case 13 in group 3. Furthermore, these two variants were only observed in Holstein cattle when querying WGS data of > 5,100 animals. Alternatively, potential de novo mutational events were assessed in each case. Filtering for heterozygous private protein-changing variants identified one DYNC1H1 frameshift variant as a candidate causal dominant acting allele in case 12 in group 3. Finally, the presence of larger structural DNA variants and chromosomal abnormalities was investigated in all cases. Depth of coverage analysis revealed two different partial monosomies of chromosome 2 segments in cases 1 and 7 in group 1 and a trisomy of chromosome 12 in the WDR45B homozygous case 13 in group 3. CONCLUSIONS This study presents for the first time a detailed genomic evaluation of CSCM in Holstein cattle and suggests an unexpected genetic and allelic heterogeneity considering the mode of inheritance, as well as the type of variant. For the first time, we propose candidate causal variants that may explain bovine CSCM in a certain proportion of affected calves. We present cattle as a large animal model for human CMII and propose new genes and genomic variants as possible causes for related diseases in both animals and humans.
Collapse
Affiliation(s)
- Joana Goncalves Pontes Jacinto
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, Bern, 3012, Switzerland
- Clinic for Ruminants, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, Bern, 3012, Switzerland
| | - Anna Letko
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, Bern, 3012, Switzerland
| | - Irene Monika Häfliger
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, Bern, 3012, Switzerland
| | - Cord Drögemüller
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, Bern, 3012, Switzerland.
| | - Jørgen Steen Agerholm
- Department of Veterinary Clinical Sciences, University of Copenhagen, Højbakkegaard Allé 5A, Taastrup, 2630, Denmark
| |
Collapse
|
2
|
Kidner RQ, Goldstone EB, Rodefeld HJ, Brokaw LP, Gonzalez AM, Ros-Rocher N, Gerdt JP. Exogenous lipid vesicles induce endocytosis-mediated cellular aggregation in a close unicellular relative of animals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.593945. [PMID: 38798415 PMCID: PMC11118469 DOI: 10.1101/2024.05.14.593945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Capsaspora owczarzaki is a protozoan that may both reveal aspects of animal evolution and also curtail the spread of schistosomiasis, a neglected tropical disease. Capsaspora exhibits a chemically regulated aggregative behavior that resembles cellular aggregation in some animals. This behavior may have played a key role in the evolution of animal multicellularity. Additionally, this aggregative behavior may be important for Capsaspora 's ability to colonize the intermediate host of parasitic schistosomes and potentially prevent the spread of schistosomiasis. Both applications demand elucidation of the molecular mechanism of Capsaspora aggregation. Toward this goal, we first determined the necessary chemical properties of lipid cues that activate aggregation. We found that a wide range of abundant zwitterionic lipids induced aggregation, revealing that the aggregative behavior could be activated by diverse lipid-rich conditions. Furthermore, we demonstrated that aggregation in Capsaspora requires clathrin-mediated endocytosis, highlighting the potential significance of endocytosis-linked cellular signaling in recent animal ancestors. Finally, we found that aggregation was initiated by post-translational activation of cell-cell adhesion-not transcriptional regulation of cellular adhesion machinery. Our findings illuminate the chemical, molecular and cellular mechanisms that regulate Capsaspora aggregative behavior-with implications for the evolution of animal multicellularity and the transmission of parasites.
Collapse
|
3
|
Knittel LM, Swanson TL, Lee HJ, Copenhaver PF. Fasciclin 2 plays multiple roles in promoting cell migration within the developing nervous system of Manduca sexta. Dev Biol 2023; 499:31-46. [PMID: 37121309 PMCID: PMC10247491 DOI: 10.1016/j.ydbio.2023.04.009] [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: 02/07/2023] [Revised: 04/07/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
The coordination of neuronal and glial migration is essential to the formation of most nervous systems, requiring a complex interplay of cell-intrinsic responses and intercellular guidance cues. During the development of the enteric nervous system (ENS) in Manduca sexta (tobacco hornworm), the IgCAM Fasciclin 2 (Fas2) serves several distinct functions to regulate these processes. As the ENS forms, a population of 300 neurons (EP cells) undergoes sequential phases of migration along well-defined muscle pathways on the visceral mesoderm to form a branching Enteric Plexus, closely followed by a trailing wave of proliferating glial cells that enwrap the neurons. Initially, both the neurons and glial cells express a GPI-linked form of Fas2 (GPI-Fas2), which helps maintain cell-cell contact among the pre-migratory neurons and later promotes glial ensheathment. The neurons then switch isoforms, predominantly expressing a combination of transmembrane isoforms lacking an intracellular PEST domain (TM-Fas2 PEST-), while their muscle band pathways on the midgut transiently express transmembrane isoforms containing this domain (TM-Fas2 PEST+). Using intracellular injection protocols to manipulate Fas2 expression in cultured embryos, we found that TM-Fas2 promotes the directed migration and outgrowth of individual neurons in the developing ENS. Concurrently, TM-Fas2 expression by the underlying muscle bands is also required as a substrate cue to support normal migration, while glial expression of GPI-Fas2 helps support their ensheathment of the migratory neurons. These results demonstrate how a specific IgCAM can play multiple roles that help coordinate neuronal and glial migration in the developing nervous system.
Collapse
Affiliation(s)
- Laura M Knittel
- Department of Cell, Developmental and Cancer Biology L-215, Oregon Health & Sciences University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Tracy L Swanson
- Department of Cell, Developmental and Cancer Biology L-215, Oregon Health & Sciences University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Hun Joo Lee
- Department of Cell, Developmental and Cancer Biology L-215, Oregon Health & Sciences University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Philip F Copenhaver
- Department of Cell, Developmental and Cancer Biology L-215, Oregon Health & Sciences University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| |
Collapse
|
4
|
Howard AGA, Uribe RA. Hox proteins as regulators of extracellular matrix interactions during neural crest migration. Differentiation 2022; 128:26-32. [PMID: 36228422 PMCID: PMC10802151 DOI: 10.1016/j.diff.2022.09.003] [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: 08/02/2022] [Revised: 09/23/2022] [Accepted: 09/25/2022] [Indexed: 01/19/2023]
Abstract
Emerging during embryogenesis, the neural crest are a migratory, transient population of multipotent stem cell that differentiates into various cell types in vertebrates. Neural crest cells arise along the anterior-posterior extent of the neural tube, delaminate and migrate along routes to their final destinations. The factors that orchestrate how neural crest cells undergo delamination and their subsequent sustained migration is not fully understood. This review provides a primer about neural crest epithelial-to-mesenchymal transition (EMT), with a special emphasis on the role of the Extracellular matrix (ECM), cellular effector proteins of EMT, and subsequent migration. We also summarize published findings that link the expression of Hox transcription factors to EMT and ECM modification, thereby implicating Hox factors in regulation of EMT and ECM remodeling during neural crest cell ontogenesis.
Collapse
Affiliation(s)
- Aubrey G A Howard
- BioSciences Department, Rice University, Houston, TX, 77005, USA; Biochemistry and Cell Biology Program, Rice University, Houston, TX, 77005, USA
| | - Rosa A Uribe
- BioSciences Department, Rice University, Houston, TX, 77005, USA; Biochemistry and Cell Biology Program, Rice University, Houston, TX, 77005, USA.
| |
Collapse
|
5
|
Cell Junction and Vesicle Trafficking-Mediated Melanosome/Melanin Transfer Are Involved in the Dynamic Transformation of Goldfish Carassius auratus Skin Color. Int J Mol Sci 2022; 23:ijms232012214. [PMID: 36293071 PMCID: PMC9603685 DOI: 10.3390/ijms232012214] [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: 09/20/2022] [Revised: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 11/21/2022] Open
Abstract
Goldfish are one of the most popular models for studying the genetic diversity of skin color. Transcriptome sequencing (RNA-seq) and whole genome bisulfate sequencing (WGBS) of skin tissues from the third filial (F3) cyan (CN), black (BK), and white (WH) goldfish were conducted to analyze the molecular mechanism of color transformation in fish. The RNA-seq yielded 56 Gb of clean data and 56,627 transcripts from nine skin samples. The DEGs (differentially expressed genes) were enriched in cell junction cellular components and the tight junction pathway. Ninety-five homologs of the claudin family were predicted and 16 claudins were identified in correlation with skin color transformation. WGBS yielded 1079 Gb of clean data from 15 samples. Both the DEGs and the DMRs (differentially methylated regions) in the BK_CN group were found to be enriched in cytoskeleton reorganization and vesicle trafficking. Masson staining and TEM (transmission electron microscopy) confirmed the varied distribution and processes of melanosome/melanin in skin tissues. Our results suggested that cytoskeleton reorganization, cell junction, and the vesicle trafficking system played key roles in the transfer of the melanosome/melanin, and it was the extracellular translocation rather than the biosynthesis or metabolism of the melanin process that resulted in the color transformation of cyan goldfish. The data will facilitate the understanding of the molecular mechanisms underlying dynamic skin color transformation in goldfish.
Collapse
|
6
|
Candido-Ferreira IL, Lukoseviciute M, Sauka-Spengler T. Multi-layered transcriptional control of cranial neural crest development. Semin Cell Dev Biol 2022; 138:1-14. [PMID: 35941042 DOI: 10.1016/j.semcdb.2022.07.010] [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: 01/27/2022] [Revised: 07/23/2022] [Accepted: 07/23/2022] [Indexed: 11/28/2022]
Abstract
The neural crest (NC) is an emblematic population of embryonic stem-like cells with remarkable migratory ability. These distinctive attributes have inspired the curiosity of developmental biologists for over 150 years, however only recently the regulatory mechanisms controlling the complex features of the NC have started to become elucidated at genomic scales. Regulatory control of NC development is achieved through combinatorial transcription factor binding and recruitment of associated transcriptional complexes to distal cis-regulatory elements. Together, they regulate when, where and to what extent transcriptional programmes are actively deployed, ultimately shaping ontogenetic processes. Here, we discuss how transcriptional networks control NC ontogeny, with a special emphasis on the molecular mechanisms underlying specification of the cephalic NC. We also cover emerging properties of transcriptional regulation revealed in diverse developmental systems, such as the role of three-dimensional conformation of chromatin, and how they are involved in the regulation of NC ontogeny. Finally, we highlight how advances in deciphering the NC transcriptional network have afforded new insights into the molecular basis of human diseases.
Collapse
Affiliation(s)
- Ivan L Candido-Ferreira
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Martyna Lukoseviciute
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Tatjana Sauka-Spengler
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK.
| |
Collapse
|
7
|
Inubushi T, Nakanishi Y, Abe M, Takahata Y, Nishimura R, Kurosaka H, Irie F, Yamashiro T, Yamaguchi Y. The cell surface hyaluronidase TMEM2 plays an essential role in mouse neural crest cell development and survival. PLoS Genet 2022; 18:e1009765. [PMID: 35839257 PMCID: PMC9328550 DOI: 10.1371/journal.pgen.1009765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 07/27/2022] [Accepted: 06/29/2022] [Indexed: 11/30/2022] Open
Abstract
Hyaluronan (HA) is a major extracellular matrix component whose tissue levels are dynamically regulated during embryonic development. Although the synthesis of HA has been shown to exert a substantial influence on embryonic morphogenesis, the functional importance of the catabolic aspect of HA turnover is poorly understood. Here, we demonstrate that the transmembrane hyaluronidase TMEM2 plays an essential role in neural crest development and the morphogenesis of neural crest derivatives, as evidenced by the presence of severe craniofacial abnormalities in Wnt1-Cre–mediated Tmem2 knockout (Tmem2CKO) mice. Neural crest cells (NCCs) are a migratory population of cells that gives rise to diverse cell lineages, including the craniofacial complex, the peripheral nervous system, and part of the heart. Analysis of Tmem2 expression during NCC formation and migration reveals that Tmem2 is expressed at the site of NCC delamination and in emigrating Sox9-positive NCCs. In Tmem2CKO embryos, the number of NCCs emigrating from the neural tube is greatly reduced. Furthermore, linage tracing reveals that the number of NCCs traversing the ventral migration pathway and the number of post-migratory neural crest derivatives are both significantly reduced in a Tmem2CKO background. In vitro studies using Tmem2-depleted mouse O9-1 neural crest cells demonstrate that Tmem2 expression is essential for the ability of these cells to form focal adhesions on and to migrate into HA-containing substrates. Additionally, we show that Tmem2-deficient NCCs exhibit increased apoptotic cell death in NCC-derived tissues, an observation that is corroborated by in vitro experiments using O9-1 cells. Collectively, our data demonstrate that TMEM2-mediated HA degradation plays an essential role in normal neural crest development. This study reveals the hitherto unrecognized functional importance of HA degradation in embryonic development and highlights the pivotal role of Tmem2 in the developmental process. As a major component of the extracellular matrix, hyaluronan is particularly abundant in the extracellular matrix of embryonic tissues, where its expression is dynamically regulated during tissue morphogenetic processes. Tissue levels of hyaluronan are regulated not only by its synthesis but also by its degradation. Curiously, however, mice lacking known hyaluronidase molecules, including HYAL1 and HYAL2, exhibit minimal embryonic phenotypes. As a result, our understanding of the role of the catabolic aspect of hyaluronan metabolism in embryonic development is quite limited. Here, we show that TMEM2, a recently identified hyaluronidase that degrades hyaluronan on the cell surface, plays a critical role in the development of neural crest cells and their derivatives. Our analyses of Tmem2 conditional knockout mice, Tmem2 knock-in reporter mice, and in vitro cell cultures demonstrate that TMEM2 is essential for generating a tissue environment needed for efficient migration of neural crest cells from the neural tube. Our paper reveals for the first time that the degradation of hyaluronan plays a specific regulatory role in embryonic morphogenesis, and that dysregulation of hyaluronan degradation leads to severe developmental defects.
Collapse
Affiliation(s)
- Toshihiro Inubushi
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, Osaka, Japan
- * E-mail:
| | - Yuichiro Nakanishi
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Makoto Abe
- Department of Oral Anatomy and Developmental Biology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yoshifumi Takahata
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Hiroshi Kurosaka
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Fumitoshi Irie
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Takashi Yamashiro
- Department of Orthodontics and Dentofacial Orthopedics, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yu Yamaguchi
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| |
Collapse
|
8
|
Kratochwil CF, Kautt AF, Nater A, Härer A, Liang Y, Henning F, Meyer A. An intronic transposon insertion associates with a trans-species color polymorphism in Midas cichlid fishes. Nat Commun 2022; 13:296. [PMID: 35027541 PMCID: PMC8758764 DOI: 10.1038/s41467-021-27685-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 12/01/2021] [Indexed: 11/09/2022] Open
Abstract
Polymorphisms have fascinated biologists for a long time, but their genetic underpinnings often remain elusive. Here, we aim to uncover the genetic basis of the gold/dark polymorphism that is eponymous of Midas cichlid fish (Amphilophus spp.) adaptive radiations in Nicaraguan crater lakes. While most Midas cichlids are of the melanic "dark morph", about 10% of individuals lose their melanic pigmentation during their ontogeny and transition into a conspicuous "gold morph". Using a new haplotype-resolved long-read assembly we discover an 8.2 kb, transposon-derived inverted repeat in an intron of an undescribed gene, which we term goldentouch in reference to the Greek myth of King Midas. The gene goldentouch is differentially expressed between morphs, presumably due to structural implications of inverted repeats in both DNA and/or RNA (cruciform and hairpin formation). The near-perfect association of the insertion with the phenotype across independent populations suggests that it likely underlies this trans-specific, stable polymorphism.
Collapse
Affiliation(s)
- Claudius F Kratochwil
- Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78457, Konstanz, Germany.
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Andreas F Kautt
- Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78457, Konstanz, Germany
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Alexander Nater
- Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78457, Konstanz, Germany
| | - Andreas Härer
- Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78457, Konstanz, Germany
- Division of Biological Sciences, Section of Ecology, Behavior & Evolution, University of California San Diego, La Jolla, CA, USA
| | - Yipeng Liang
- Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78457, Konstanz, Germany
- Department of Biology, University of Virginia, Charlottesville, VA, 22903, USA
| | - Frederico Henning
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Axel Meyer
- Zoology and Evolutionary Biology, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78457, Konstanz, Germany.
| |
Collapse
|
9
|
Eom DS, Patterson LB, Bostic RR, Parichy DM. Immunoglobulin superfamily receptor Junctional adhesion molecule 3 (Jam3) requirement for melanophore survival and patterning during formation of zebrafish stripes. Dev Biol 2021; 476:314-327. [PMID: 33933422 PMCID: PMC10069301 DOI: 10.1016/j.ydbio.2021.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/03/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022]
Abstract
Adhesive interactions are essential for tissue patterning and morphogenesis yet difficult to study owing to functional redundancies across genes and gene families. A useful system in which to dissect roles for cell adhesion and adhesion-dependent signaling is the pattern formed by pigment cells in skin of adult zebrafish, in which stripes represent the arrangement of neural crest derived melanophores, cells homologous to melanocytes. In a forward genetic screen for adult pattern defects, we isolated the pissarro (psr) mutant, having a variegated phenotype of spots, as well as defects in adult fin and lens. We show that psr corresponds to junctional adhesion protein 3b (jam3b) encoding a zebrafish orthologue of the two immunoglobulin-like domain receptor JAM3 (JAM-C), known for roles in adhesion and signaling in other developing tissues, and for promoting metastatic behavior of human and murine melanoma cells. We found that zebrafish jam3b is expressed post-embryonically in a variety of cells including melanophores, and that jam3b mutants have defects in melanophore survival. Jam3b supported aggregation of cells in vitro and was required autonomously by melanophores for an adherent phenotype in vivo. Genetic analyses further indicated both overlapping and non-overlapping functions with the related receptor, Immunoglobulin superfamily 11 (Igsf11) and Kit receptor tyrosine kinase. These findings suggest a model for Jam3b function in zebrafish melanophores and hint at the complexity of adhesive interactions underlying pattern formation.
Collapse
Affiliation(s)
- Dae Seok Eom
- Department of Biology, University of Virginia, Charlottesville, VA, USA.
| | | | - Raegan R Bostic
- Department of Biology, University of Virginia, Charlottesville, VA, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - David M Parichy
- Department of Biology, University of Virginia, Charlottesville, VA, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
10
|
Wang Y, Zhang Y, Sang B, Zhu X, Yu R, Zhou X. Human giant larvae-1 promotes migration and invasion of malignant glioma cells by regulating N-cadherin. Oncol Lett 2021; 21:167. [PMID: 33552285 PMCID: PMC7798033 DOI: 10.3892/ol.2021.12428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Human giant larvae-1 (Hugl-1) is a human homologue of Drosophila tumor suppressor lethal (2)-giant larvae and has been reported to be involved in the development of human malignancies. Previous studies performed by our group demonstrated that Hugl-1 inhibits glioma cell proliferation in an intracranial model of nude mice. However, the exact molecular mechanisms underlying the participation of Hugl-1 in glioma invasion and migration, and in the depolarizing process remain largely unknown. Utilizing the U251-MG cells with stable expression of Hugl-1, the present study used wound healing, Transwell invasion and western blot assays to explore the role and specific mechanism of Hugl-1 in glioma invasion and migration. The results of the present study demonstrated that overexpression of Hugl-1 decreased cell-cell adhesion and increased cell-cell extracellular matrix adhesion. In addition, overexpression of Hugl-1 promoted pseudopodia formation, glioma cell migration and invasion. The molecular mechanism of action involved the negative regulation of N-cadherin protein levels by Hugl-1. Overexpression or knockdown of N-cadherin partially suppressed or enhanced the effects of Hugl-1 on glioma cell migration and invasion, respectively. Furthermore, Hugl-1 inhibited cell proliferation, while promoting cell migration, which suggests that it may serve a two-sided biological role in cellular processes. Taken together, these results suggest that Hugl-1 promotes the migration and invasion of malignant glioma cells by decreasing N-cadherin expression. Thus, Hugl-1 may be applied in the development of targeted and personalized treatment.
Collapse
Affiliation(s)
- Yan Wang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Yu Zhang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Ben Sang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xianlong Zhu
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
11
|
Liu W, Zheng Y, Zhang F, Zhu M, Guo Q, Xu H, Liu C, Chen H, Wang X, Hu Y, Zhang T, Lin Z, Zhang C, Li G, Jiang K, Liu X. A Preliminary Investigation on Plasma Cell Adhesion Molecules Levels by Protein Microarray Technology in Major Depressive Disorder. Front Psychiatry 2021; 12:627469. [PMID: 33912082 PMCID: PMC8071998 DOI: 10.3389/fpsyt.2021.627469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/15/2021] [Indexed: 12/19/2022] Open
Abstract
Objectives: Major depressive disorder (MDD) is a serious mental disorder, and there is a great difficulty to diagnose and treat. Hitherto, relatively few studies have explored the correlation between the levels of plasma cell adhesion molecules and MDD. Methods: Thirty outpatients with acute episodes of MDD in Shanghai Mental Health Center and 34 healthy volunteers from the community were recruited as subjects. Protein microarray technology was applied to compared the differences in plasma levels of 17 kinds of adhesion molecular proteins between the two groups. Meanwhile, the diagnostic value of different proteins in depression was discussed by using the receiver operating characteristic curve. Results: The levels of Carcinoembryonic Antigen Related Cell Adhesion Molecule-1(CEACAM-1) and Neural Cell Adhesion Molecule (NrCAM) in MDD patients were significantly higher than those in healthy controls (P < 0.05). The area under ROC curve of CEACAM-1 combined with NrCAM was 0.723, with the sensitivity 0.800 and the specificity 0.676. Conclusion: The plasma levels of CEACAM-1 and NrCAM were significantly up-regulated in MDD, and their combined application was of potential diagnostic value, deserving to expand the sample size for further verification.
Collapse
Affiliation(s)
- Wanying Liu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanqun Zheng
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuxu Zhang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mo Zhu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Guo
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Xu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caiping Liu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiying Chen
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoliang Wang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Hu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianhong Zhang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiguang Lin
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanjun Li
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaida Jiang
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohua Liu
- Department of Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Muppirala AN, Limbach LE, Bradford EF, Petersen SC. Schwann cell development: From neural crest to myelin sheath. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e398. [PMID: 33145925 DOI: 10.1002/wdev.398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022]
Abstract
Vertebrate nervous system function requires glial cells, including myelinating glia that insulate axons and provide trophic support that allows for efficient signal propagation by neurons. In vertebrate peripheral nervous systems, neural crest-derived glial cells known as Schwann cells (SCs) generate myelin by encompassing and iteratively wrapping membrane around single axon segments. SC gliogenesis and neurogenesis are intimately linked and governed by a complex molecular environment that shapes their developmental trajectory. Changes in this external milieu drive developing SCs through a series of distinct morphological and transcriptional stages from the neural crest to a variety of glial derivatives, including the myelinating sublineage. Cues originate from the extracellular matrix, adjacent axons, and the developing SC basal lamina to trigger intracellular signaling cascades and gene expression changes that specify stages and transitions in SC development. Here, we integrate the findings from in vitro neuron-glia co-culture experiments with in vivo studies investigating SC development, particularly in zebrafish and mouse, to highlight critical factors that specify SC fate. Ultimately, we connect classic biochemical and mutant studies with modern genetic and visualization tools that have elucidated the dynamics of SC development. This article is categorized under: Signaling Pathways > Cell Fate Signaling Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Anoohya N Muppirala
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neuroscience, Kenyon College, Gambier, Ohio, USA
| | | | | | - Sarah C Petersen
- Department of Neuroscience, Kenyon College, Gambier, Ohio, USA.,Department of Biology, Kenyon College, Gambier, Ohio, USA
| |
Collapse
|
13
|
Barqué A, Jan K, De La Fuente E, Nicholas CL, Hynes RO, Naba A. Knockout of the gene encoding the extracellular matrix protein SNED1 results in early neonatal lethality and craniofacial malformations. Dev Dyn 2020; 250:274-294. [PMID: 33012048 DOI: 10.1002/dvdy.258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/10/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The extracellular matrix (ECM) is a fundamental component of multicellular organisms that orchestrates developmental processes and controls cell and tissue organization. We previously identified the novel ECM protein SNED1 as a promoter of breast cancer metastasis and showed that its level of expression negatively correlated with breast cancer patient survival. Here, we sought to identify the roles of SNED1 during murine development. RESULTS We generated two novel Sned1 knockout mouse strains and showed that Sned1 is essential since homozygous ablation of the gene led to early neonatal lethality. Phenotypic analysis of the surviving knockout mice revealed a role for SNED1 in the development of craniofacial and skeletal structures since Sned1 knockout resulted in growth defects, nasal cavity occlusion, and craniofacial malformations. Sned1 is widely expressed in embryos, notably by cell populations undergoing epithelial-to-mesenchymal transition, such as the neural crest cells. We further show that mice with a neural-crest-cell-specific deletion of Sned1 survive, but display facial anomalies partly phenocopying the global knockout mice. CONCLUSIONS Our results demonstrate requisite roles for SNED1 during development and neonatal survival. Importantly, the deletion of 2q37.3 in humans, a region that includes the SNED1 locus, has been associated with facial dysmorphism and short stature.
Collapse
Affiliation(s)
- Anna Barqué
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Kyleen Jan
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Emanuel De La Fuente
- Department of Orthodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Christina L Nicholas
- Department of Orthodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Anthropology, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard O Hynes
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
14
|
Kim CW, Lee SM, Ko EB, Go RE, Jeung EB, Kim MS, Choi KC. Inhibitory effects of cigarette smoke extracts on neural differentiation of mouse embryonic stem cells. Reprod Toxicol 2020; 95:75-85. [PMID: 32454085 DOI: 10.1016/j.reprotox.2020.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/13/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022]
Abstract
Maternal smoking during the perinatal period is linked to adverse neonatal outcomes such as low birth weight and birth defects. Numerous studies have shown that cigarette smoke or nicotine exposure has a widespread effect on fetal nerve development. However, there exists a lack of understanding of what specific changes occur at the cellular level on persistent exposure to cigarette smoke during the differentiation of embryonic stem cells (ESCs) into neural cells. We previously investigated the effects of cigarette smoke extract (CSE) and its major component, nicotine, on the neural differentiation of mouse embryonic stem cells (mESCs). Differentiation of mESCs into neural progenitor cells (NPCs) or neural crest cells (NCCs) was induced with chemically defined media, and the cells were continuously exposed to CSE or nicotine during neural differentiation and development. Disturbed balance of the pluripotency state was observed in the NPCs, with consequent inhibition of neurite outgrowth and glial fibrillary acidic protein (Gfap) expression. These inhibitions correlated with the altered expression of proteins involved in the Notch-1 signaling pathways. The migration ability of NCCs was significantly decreased by CSE or nicotine exposure, which was associated with reduced protein expression of migration-related proteins. Taken together, we concluded that CSE and nicotine inhibit differentiation of mESCs into NPCs or NCCs, and may disrupt functional development of neural cells. These results imply that cigarette smoking during the perinatal period potentially inhibits neural differentiation and development of ESCs cells, leading to neonatal abnormal brain development and behavioral abnormalities.
Collapse
Affiliation(s)
- Cho-Won Kim
- Laboratory of Biochemistry and Immunology, Republic of Korea
| | - Sung-Moo Lee
- Laboratory of Biochemistry and Immunology, Republic of Korea
| | - Eul-Bee Ko
- Laboratory of Biochemistry and Immunology, Republic of Korea
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Min-Seok Kim
- Inhalation Toxicology Research Group, Jeonbuk Department of Inhalation Research, Jeongeup, Korea Institute of Toxicology, Jeonbuk, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, Republic of Korea.
| |
Collapse
|
15
|
Ray H, Chang C. The transcription factor Hypermethylated in Cancer 1 (Hic1) regulates neural crest migration via interaction with Wnt signaling. Dev Biol 2020; 463:169-181. [PMID: 32502469 DOI: 10.1016/j.ydbio.2020.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 01/20/2023]
Abstract
The transcription factor Hypermethylated in Cancer 1 (HIC1) is associated with both tumorigenesis and the complex human developmental disorder Miller-Dieker Syndrome. While many studies have characterized HIC1 as a tumor suppressor, HIC1 function in development is less understood. Loss-of-function mouse alleles show embryonic lethality accompanied with developmental defects, including craniofacial abnormalities that are reminiscent of human Miller-Dieker Syndrome patients. However, the tissue origin of the defects has not been reported. In this study, we use the power of the Xenopus laevis model system to explore Hic1 function in early development. We show that hic1 mRNA is expressed throughout early Xenopus development and has a spatial distribution within the neural plate border and in migrating neural crest cells in branchial arches. Targeted manipulation of hic1 levels in the dorsal ectoderm that gives rise to neural and neural crest tissues reveals that both overexpression and knockdown of hic1 result in craniofacial defects with malformations of the craniofacial cartilages. Neural crest specification is not affected by altered hic1 levels, but migration of the cranial neural crest is impaired both in vivo and in tissue explants. Mechanistically, we find that Hic1 regulates cadherin expression profiles and canonical Wnt signaling. Taken together, these results identify Hic1 as a novel regulator of the canonical Wnt pathway during neural crest migration.
Collapse
Affiliation(s)
- Heather Ray
- Dept. of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, MCLM 338, 1918 University Dr. Birmingham, AL, 35294, USA.
| | - Chenbei Chang
- Dept. of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, MCLM 338, 1918 University Dr. Birmingham, AL, 35294, USA
| |
Collapse
|
16
|
Leonard CE, Taneyhill LA. The road best traveled: Neural crest migration upon the extracellular matrix. Semin Cell Dev Biol 2020; 100:177-185. [PMID: 31727473 PMCID: PMC7071992 DOI: 10.1016/j.semcdb.2019.10.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/29/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022]
Abstract
Neural crest cells have the extraordinary task of building much of the vertebrate body plan, including the craniofacial cartilage and skeleton, melanocytes, portions of the heart, and the peripheral nervous system. To execute these developmental programs, stationary premigratory neural crest cells first acquire the capacity to migrate through an extensive process known as the epithelial-to-mesenchymal transition. Once motile, neural crest cells must traverse a complex environment consisting of other cells and the protein-rich extracellular matrix in order to get to their final destinations. Herein, we will highlight some of the main molecular machinery that allow neural crest cells to first exit the neuroepithelium and then later successfully navigate this intricate in vivo milieu. Collectively, these extracellular and intracellular factors mediate the appropriate migration of neural crest cells and allow for the proper development of the vertebrate embryo.
Collapse
Affiliation(s)
- Carrie E Leonard
- 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
|
17
|
Kalev-Altman R, Hanael E, Zelinger E, Blum M, Monsonego-Ornan E, Sela-Donenfeld D. Conserved role of matrix metalloproteases 2 and 9 in promoting the migration of neural crest cells in avian and mammalian embryos. FASEB J 2020; 34:5240-5261. [PMID: 32067275 DOI: 10.1096/fj.201901217rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 01/28/2020] [Accepted: 02/02/2020] [Indexed: 01/10/2023]
Abstract
Neural crest cells (NCCs) are a unique embryonic cell population that initially reside at the dorsal neural tube but later migrate in the embryo and differentiate into multiple types of derivatives. To acquire motility, NCCs undergo epithelial-to-mesenchymal transition and invade the surrounding extracellular matrix (ECM). Matrix metalloproteases (MMPs) are a large family of proteases which regulate migration of various embryonic and adult cells via ECM remodeling. The gelatinase's subgroup of MMPs is the most studied one due to its key role in metastasis. As it is composed of only two proteases, MMP2 and MMP9, it is important to understand whether each is indispensable or redundant in its biological function. Here we explored the role of the gelatinases in executing NCC migration, by determining whether MMP2 and/or MMP9 regulate migration across species in singular, combined, or redundant manners. Chick and mouse embryos were utilized to compare expression and activity of both MMPs using genetic and pharmacological approaches in multiple in vivo and ex vivo assays. Both MMPs were found to be expressed and active in mouse and chick NCCs. Inhibition of each MMP was sufficient to prevent NCC migration in both species. Yet, NCC migration was maintained in MMP2-/- or MMP9-/- mouse mutants due to compensation between the gelatinases, but reciprocal pharmacological inhibition in each mutant prevented NCC migration. This study reveals for the first time that both gelatinases are expressed in avian and mammalian NCCs, and demonstrates their fundamental and conserved role in promoting embryonic cell migration.
Collapse
Affiliation(s)
- Rotem Kalev-Altman
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel.,The Institute of Biochemistry and Nutrition, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Erez Hanael
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Einat Zelinger
- Core Facility Unit, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Martin Blum
- Institute of Zoology, University of Hohenheim, Stuttgart, Germany
| | - Efrat Monsonego-Ornan
- The Institute of Biochemistry and Nutrition, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| |
Collapse
|
18
|
Ceylan D, Tufekci KU, Keskinoglu P, Genc S, Özerdem A. Circulating exosomal microRNAs in bipolar disorder. J Affect Disord 2020; 262:99-107. [PMID: 31726266 DOI: 10.1016/j.jad.2019.10.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/23/2019] [Accepted: 10/27/2019] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Emerging evidence suggests central roles of miRNAs in the pathogenesis of bipolar disorder (BD). Exosomes are membrane-bound vesicles acting as "biological cargo carriers" of various types of molecules including microRNAs. In this study, we aimed to investigate circulating exosomal microRNAs as potential diagnostic biomarkers for BD. METHODS The exosomes were precipitated from plasma samples of patients with BD (n = 69; 15 depressed, 27 manic, 27 euthymic) and healthy controls (n = 41). Total RNA was extracted from the exosomes and the levels of miRNAs were assayed by qPCR. Dysregulated miRNAs were subjected to Kyoto Encyclopedia of Genes and Genomes" (KEGG) pathway analysis by DIANA-miRPath v3.0 to identify the predicted targets and the related pathways. RESULTS Thirteen miRNAs showed significant differences between patients with BD and healthy individuals; among these, MiR-484, -652-3p, -142-3p remained significantly downregulated and miR-185-5p remained significantly upregulated after accounting for multiple comparisons and adjustments for potential confounders. There were no significant alterations among different states of BD. The KEEG analysis of four dysregulated miRNAs highlighted several target pathways including PI3K/Akt signaling, fatty acid biosynthesis/metabolism, extracellular matrix and adhesion pathways. CONCLUSION Our findings suggest that dysregulation of miRNAs might be involved in the underlying pathophysiology of BD through several biological pathways; and highlight the importance of the exosomal miRNAs for biomarker research in BD. Further longitudinal studies may clarify the roles of exosomal miRNAs and their targets in the neurobiology of BD.
Collapse
Affiliation(s)
- Deniz Ceylan
- Izmir University of Economics, Faculty of Medicine, Department of Psychiatry, Izmir, Turkey
| | - Kemal Ugur Tufekci
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Pembe Keskinoglu
- Department of Biostatistics and Medical Informatics, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey; Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Ayşegül Özerdem
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University Health Campus, Izmir, Turkey; Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey.
| |
Collapse
|
19
|
Wang Y, Liu H, Zhang N, Luo E. Partial duplication of the jaw: case reports and review of relevant publications. Br J Oral Maxillofac Surg 2019; 58:34-42. [PMID: 31735399 DOI: 10.1016/j.bjoms.2019.10.311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 10/21/2019] [Indexed: 02/05/2023]
Abstract
Craniofacial duplication is a rare congenital malformation with a wide phenotypic range. The signs and symptoms range from partial craniofacial duplication to bicephalus. We describe two cases of partial duplication of jaw: a girl with a duplication of the maxilla, and a boy with duplication of the mandible. We review the relevant publications and discuss the pathogenesis.
Collapse
Affiliation(s)
- Y Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University.
| | - H Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University.
| | - N Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University.
| | - E Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University.
| |
Collapse
|
20
|
Saito-Diaz K, Zeltner N. Induced pluripotent stem cells for disease modeling, cell therapy and drug discovery in genetic autonomic disorders: a review. Clin Auton Res 2019; 29:367-384. [PMID: 30631982 DOI: 10.1007/s10286-018-00587-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/26/2018] [Indexed: 12/19/2022]
Abstract
The autonomic nervous system (ANS) regulates all organs in the body independent of consciousness, and is thus essential for maintaining homeostasis of the entire organism. Diseases of the ANS can arise due to environmental insults such as injury, toxins/drugs and infections or due to genetic lesions. Human studies and animal models have been instrumental to understanding connectivity and regulation of the ANS and its disorders. However, research into cellular pathologies and molecular mechanisms of ANS disorders has been hampered by the difficulties in accessing human patient-derived ANS cells in large numbers to conduct meaningful research, mainly because patient neurons cannot be easily biopsied and primary human neuronal cultures cannot be expanded.Human-induced pluripotent stem cell (hiPSC) technology can elegantly bridge these issues, allowing unlimited access of patient-derived ANS cell types for cellular, molecular and biochemical analysis, facilitating the discovery of novel therapeutic targets, and eventually leading to drug discovery. Additionally, such cells may provide a source for cell replacement therapy to replenish lost or injured ANS tissue in patients.Here, we first review the anatomy and embryonic development of the ANS, as this knowledge is crucial for understanding disease modeling approaches. We then review the current advances in human stem cell technology for modeling diseases of the ANS, recent strides toward cell replacement therapy and drug discovery initiatives.
Collapse
Affiliation(s)
- Kenyi Saito-Diaz
- Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - Nadja Zeltner
- Center for Molecular Medicine, University of Georgia, Athens, GA, USA. .,Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA. .,Department of Cellular Biology, University of Georgia, Athens, GA, USA.
| |
Collapse
|
21
|
Chen J, Tan X, Wang Z, Liu Y, Zhou J, Rong X, Lu L, Li Y. The ribosome biogenesis protein Esf1 is essential for pharyngeal cartilage formation in zebrafish. FEBS J 2018; 285:3464-3484. [DOI: 10.1111/febs.14622] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 06/10/2018] [Accepted: 08/01/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Jian‐Yang Chen
- Key Laboratory of Marine Drugs (Ocean University of China) Chinese Ministry of Education Qingdao China
- School of Medicine and Pharmacy Ocean University of China Qingdao China
- Laboratory for Marine Drugs and Biological Products Qingdao National Laboratory for Marine Science and Technology China
| | - Xungang Tan
- CAS Key Laboratory of Experimental Marine Biology Institute of Oceanology Chinese Academy of Sciences Qingdao China
| | - Zheng‐Hua Wang
- Key Laboratory of Marine Drugs (Ocean University of China) Chinese Ministry of Education Qingdao China
- School of Medicine and Pharmacy Ocean University of China Qingdao China
- Laboratory for Marine Drugs and Biological Products Qingdao National Laboratory for Marine Science and Technology China
- CAS Key Laboratory of Experimental Marine Biology Institute of Oceanology Chinese Academy of Sciences Qingdao China
| | - Yun‐Zhang Liu
- Key Laboratory of Marine Drugs (Ocean University of China) Chinese Ministry of Education Qingdao China
- School of Medicine and Pharmacy Ocean University of China Qingdao China
- Laboratory for Marine Drugs and Biological Products Qingdao National Laboratory for Marine Science and Technology China
| | - Jian‐Feng Zhou
- Key Laboratory of Marine Drugs (Ocean University of China) Chinese Ministry of Education Qingdao China
- School of Medicine and Pharmacy Ocean University of China Qingdao China
- Laboratory for Marine Drugs and Biological Products Qingdao National Laboratory for Marine Science and Technology China
| | - Xiao‐Zhi Rong
- Key Laboratory of Marine Drugs (Ocean University of China) Chinese Ministry of Education Qingdao China
- School of Medicine and Pharmacy Ocean University of China Qingdao China
- Laboratory for Marine Drugs and Biological Products Qingdao National Laboratory for Marine Science and Technology China
| | - Ling Lu
- Key Laboratory of Marine Drugs (Ocean University of China) Chinese Ministry of Education Qingdao China
- School of Medicine and Pharmacy Ocean University of China Qingdao China
- Laboratory for Marine Drugs and Biological Products Qingdao National Laboratory for Marine Science and Technology China
| | - Yun Li
- Key Laboratory of Marine Drugs (Ocean University of China) Chinese Ministry of Education Qingdao China
- School of Medicine and Pharmacy Ocean University of China Qingdao China
- Laboratory for Marine Drugs and Biological Products Qingdao National Laboratory for Marine Science and Technology China
| |
Collapse
|
22
|
Rasmussen KK, Falkesgaard MH, Winther M, Roed NK, Quistgaard CL, Teisen MN, Edslev SM, Petersen DL, Aljubouri A, Christensen C, Thulstrup PW, Lo Leggio L, Teilum K, Walmod PS. NCAM2 Fibronectin type-III domains form a rigid structure that binds and activates the Fibroblast Growth Factor Receptor. Sci Rep 2018; 8:8957. [PMID: 29895898 PMCID: PMC5997747 DOI: 10.1038/s41598-018-27089-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/24/2018] [Indexed: 11/17/2022] Open
Abstract
NCAM1 and NCAM2 have ectodomains consisting of 5 Ig domains followed by 2 membrane-proximal FnIII domains. In this study we investigate and compare the structures and functions of these FnIII domains. The NCAM1 and -2 FnIII2 domains both contain a Walker A motif. In NCAM1 binding of ATP to this motif interferes with NCAM1 binding to FGFR. We obtained a structural model of the NCAM2 FnIII2 domain by NMR spectroscopy, and by titration with an ATP analogue we show that the NCAM2 Walker A motif does not bind ATP. Small angle X-ray scattering (SAXS) data revealed that the NCAM2 FnIII1-2 double domain exhibits a very low degree of flexibility. Moreover, recombinant NCAM2 FnIII domains bind FGFR in vitro, and the FnIII1-2 double domain induces neurite outgrowth in a concentration-dependent manner through activation of FGFR. Several synthetic NCAM1-derived peptides induce neurite outgrowth via FGFR. Only 2 of 5 peptides derived from similar regions in NCAM2 induce neurite outgrowth, but the most potent of these peptides stimulates neurite outgrowth through FGFR-dependent activation of the Ras-MAPK pathway. These results reveal that the NCAM2 FnIII domains form a rigid structure that binds and activates FGFR in a manner related to, but different from NCAM1.
Collapse
Affiliation(s)
- Kim Krighaar Rasmussen
- Biological Chemistry, Department of Chemistry, University of Copenhagen, Copenhagen, Denmark.
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Maria Hansen Falkesgaard
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Malene Winther
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Nikolaj Kulahin Roed
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Christine Louise Quistgaard
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Marie Nygaard Teisen
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Sofie Marie Edslev
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - David Leander Petersen
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ali Aljubouri
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Claus Christensen
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Peter Waaben Thulstrup
- Biological Chemistry, Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
| | - Leila Lo Leggio
- Biological Chemistry, Department of Chemistry, University of Copenhagen, Copenhagen, Denmark
| | - Kaare Teilum
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Peter Schledermann Walmod
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Ziermann JM, Diogo R, Noden DM. Neural crest and the patterning of vertebrate craniofacial muscles. Genesis 2018; 56:e23097. [PMID: 29659153 DOI: 10.1002/dvg.23097] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/22/2018] [Accepted: 02/25/2018] [Indexed: 12/17/2022]
Abstract
Patterning of craniofacial muscles overtly begins with the activation of lineage-specific markers at precise, evolutionarily conserved locations within prechordal, lateral, and both unsegmented and somitic paraxial mesoderm populations. Although these initial programming events occur without influence of neural crest cells, the subsequent movements and differentiation stages of most head muscles are neural crest-dependent. Incorporating both descriptive and experimental studies, this review examines each stage of myogenesis up through the formation of attachments to their skeletal partners. We present the similarities among developing muscle groups, including comparisons with trunk myogenesis, but emphasize the morphogenetic processes that are unique to each group and sometimes subsets of muscles within a group. These groups include branchial (pharyngeal) arches, which encompass both those with clear homologues in all vertebrate classes and those unique to one, for example, mammalian facial muscles, and also extraocular, laryngeal, tongue, and neck muscles. The presence of several distinct processes underlying neural crest:myoblast/myocyte interactions and behaviors is not surprising, given the wide range of both quantitative and qualitative variations in craniofacial muscle organization achieved during vertebrate evolution.
Collapse
Affiliation(s)
- Janine M Ziermann
- Department of Anatomy, Howard University College of Medicine, Washington, DC
| | - Rui Diogo
- Department of Anatomy, Howard University College of Medicine, Washington, DC
| | - Drew M Noden
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY
| |
Collapse
|
24
|
Nagy N, Goldstein AM. Enteric nervous system development: A crest cell's journey from neural tube to colon. Semin Cell Dev Biol 2017; 66:94-106. [PMID: 28087321 DOI: 10.1016/j.semcdb.2017.01.006] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 12/31/2022]
Abstract
The enteric nervous system (ENS) is comprised of a network of neurons and glial cells that are responsible for coordinating many aspects of gastrointestinal (GI) function. These cells arise from the neural crest, migrate to the gut, and then continue their journey to colonize the entire length of the GI tract. Our understanding of the molecular and cellular events that regulate these processes has advanced significantly over the past several decades, in large part facilitated by the use of rodents, avians, and zebrafish as model systems to dissect the signals and pathways involved. These studies have highlighted the highly dynamic nature of ENS development and the importance of carefully balancing migration, proliferation, and differentiation of enteric neural crest-derived cells (ENCCs). Proliferation, in particular, is critically important as it drives cell density and speed of migration, both of which are important for ensuring complete colonization of the gut. However, proliferation must be tempered by differentiation among cells that have reached their final destination and are ready to send axonal extensions, connect to effector cells, and begin to produce neurotransmitters or other signals. Abnormalities in the normal processes guiding ENCC development can lead to failure of ENS formation, as occurs in Hirschsprung disease, in which the distal intestine remains aganglionic. This review summarizes our current understanding of the factors involved in early development of the ENS and discusses areas in need of further investigation.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; Center for Neurointestinal Health, Massachusetts General Hospital, Boston, MA, United States; Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; Center for Neurointestinal Health, Massachusetts General Hospital, Boston, MA, United States.
| |
Collapse
|
25
|
Gennarini G, Bizzoca A, Picocci S, Puzzo D, Corsi P, Furley AJW. The role of Gpi-anchored axonal glycoproteins in neural development and neurological disorders. Mol Cell Neurosci 2016; 81:49-63. [PMID: 27871938 DOI: 10.1016/j.mcn.2016.11.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/10/2016] [Accepted: 11/14/2016] [Indexed: 01/06/2023] Open
Abstract
This review article focuses on the Contactin (CNTN) subset of the Immunoglobulin supergene family (IgC2/FNIII molecules), whose components share structural properties (the association of Immunoglobulin type C2 with Fibronectin type III domains), as well as a general role in cell contact formation and axonal growth control. IgC2/FNIII molecules include 6 highly related components (CNTN 1-6), associated with the cell membrane via a Glycosyl Phosphatidyl Inositol (GPI)-containing lipid tail. Contactin 1 and Contactin 2 share ~50 (49.38)% identity at the aminoacid level. They are components of the cell surface, from which they may be released in soluble forms. They bind heterophilically to multiple partners in cis and in trans, including members of the related L1CAM family and of the Neurexin family Contactin-associated proteins (CNTNAPs or Casprs). Such interactions are important for organising the neuronal membrane, as well as for modulating the growth and pathfinding of axon tracts. In addition, they also mediate the functional maturation of axons by promoting their interactions with myelinating cells at the nodal, paranodal and juxtaparanodal regions. Such interactions also mediate differential ionic channels (both Na+ and K+) distribution, which is of critical relevance in the generation of the peak-shaped action potential. Indeed, thanks to their interactions with Ankyrin G, Na+ channels map within the nodal regions, where they drive axonal depolarization. However, no ionic channels are found in the flanking Contactin1-containing paranodal regions, where CNTN1 interactions with Caspr1 and with the Ig superfamily component Neurofascin 155 in cis and in trans, respectively, build a molecular barrier between the node and the juxtaparanode. In this region K+ channels are clustered, depending upon molecular interactions with Contactin 2 and with Caspr2. In addition to these functions, the Contactins appear to have also a role in degenerative and inflammatory disorders: indeed Contactin 2 is involved in neurodegenerative disorders with a special reference to the Alzheimer disease, given its ability to work as a ligand of the Alzheimer Precursor Protein (APP), which results in increased Alzheimer Intracellular Domain (AICD) release in a γ-secretase-dependent manner. On the other hand Contactin 1 drives Notch signalling activation via the Hes pathway, which could be consistent with its ability to modulate neuroinflammation events, and with the possibility that Contactin 1-dependent interactions may participate to the pathogenesis of the Multiple Sclerosis and of other inflammatory disorders.
Collapse
Affiliation(s)
- Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy.
| | - Antonella Bizzoca
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Sabrina Picocci
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - Patrizia Corsi
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Andrew J W Furley
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2NT, UK
| |
Collapse
|
26
|
Regulators of gene expression in Enteric Neural Crest Cells are putative Hirschsprung disease genes. Dev Biol 2016; 416:255-265. [DOI: 10.1016/j.ydbio.2016.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 05/17/2016] [Accepted: 06/02/2016] [Indexed: 11/21/2022]
|
27
|
Control of the collective migration of enteric neural crest cells by the Complement anaphylatoxin C3a and N-cadherin. Dev Biol 2016; 414:85-99. [PMID: 27041467 PMCID: PMC4937886 DOI: 10.1016/j.ydbio.2016.03.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 03/09/2016] [Accepted: 03/09/2016] [Indexed: 12/25/2022]
Abstract
We analyzed the cellular and molecular mechanisms governing the adhesive and migratory behavior of enteric neural crest cells (ENCCs) during their collective migration within the developing mouse gut. We aimed to decipher the role of the complement anaphylatoxin C3a during this process, because this well-known immune system attractant has been implicated in cephalic NCC co-attraction, a process controlling directional migration. We used the conditional Ht-PA-cre transgenic mouse model allowing a specific ablation of the N-cadherin gene and the expression of a fluorescent reporter in migratory ENCCs without affecting the central nervous system. We performed time-lapse videomicroscopy of ENCCs from control and N-cad-herin mutant gut explants cultured on fibronectin (FN) and micropatterned FN-stripes with C3a or C3aR antagonist, and studied cell migration behavior with the use of triangulation analysis to quantify cell dispersion. We performed ex vivo gut cultures with or without C3aR antagonist to determine the effect on ENCC behavior. Confocal microscopy was used to analyze the cell-matrix adhesion properties. We provide the first demonstration of the localization of the complement anaphylatoxin C3a and its receptor on ENCCs during their migration in the embryonic gut. C3aR receptor inhibition alters ENCC adhesion and migration, perturbing directionality and increasing cell dispersion both in vitro and ex vivo. N-cad-herin-null ENCCs do not respond to C3a co-attraction. These findings indicate that C3a regulates cell migration in a N-cadherin-dependent process. Our results shed light on the role of C3a in regulating collective and directional cell migration, and in ganglia network organization during enteric nervous system ontogenesis. The detection of an immune system chemokine in ENCCs during ENS development may also shed light on new mechanisms for gastrointestinal disorders.
Collapse
|
28
|
McLennan R, Schumacher LJ, Morrison JA, Teddy JM, Ridenour DA, Box AC, Semerad CL, Li H, McDowell W, Kay D, Maini PK, Baker RE, Kulesa PM. VEGF signals induce trailblazer cell identity that drives neural crest migration. Dev Biol 2015; 407:12-25. [PMID: 26278036 DOI: 10.1016/j.ydbio.2015.08.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 10/23/2022]
Abstract
Embryonic neural crest cells travel in discrete streams to precise locations throughout the head and body. We previously showed that cranial neural crest cells respond chemotactically to vascular endothelial growth factor (VEGF) and that cells within the migratory front have distinct behaviors and gene expression. We proposed a cell-induced gradient model in which lead neural crest cells read out directional information from a chemoattractant profile and instruct trailers to follow. In this study, we show that migrating chick neural crest cells do not display distinct lead and trailer gene expression profiles in culture. However, exposure to VEGF in vitro results in the upregulation of a small subset of genes associated with an in vivo lead cell signature. Timed addition and removal of VEGF in culture reveals the changes in neural crest cell gene expression are rapid. A computational model incorporating an integrate-and-switch mechanism between cellular phenotypes predicts migration efficiency is influenced by the timescale of cell behavior switching. To test the model hypothesis that neural crest cellular phenotypes respond to changes in the VEGF chemoattractant profile, we presented ectopic sources of VEGF to the trailer neural crest cell subpopulation and show diverted cell trajectories and stream alterations consistent with model predictions. Gene profiling of trailer cells that diverted and encountered VEGF revealed upregulation of a subset of 'lead' genes. Injection of neuropilin1 (Np1)-Fc into the trailer subpopulation or electroporation of VEGF morpholino to reduce VEGF signaling failed to alter trailer neural crest cell trajectories, suggesting trailers do not require VEGF to maintain coordinated migration. These results indicate that VEGF is one of the signals that establishes lead cell identity and its chemoattractant profile is critical to neural crest cell migration.
Collapse
Affiliation(s)
- Rebecca McLennan
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Linus J Schumacher
- University of Oxford, Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK; Department of Computer Science, University of Oxford, Oxford OX1 3QD, UK
| | - Jason A Morrison
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Jessica M Teddy
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Dennis A Ridenour
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Andrew C Box
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Craig L Semerad
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - Hua Li
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - William McDowell
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | - David Kay
- Department of Computer Science, University of Oxford, Oxford OX1 3QD, UK
| | - Philip K Maini
- University of Oxford, Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK
| | - Ruth E Baker
- University of Oxford, Wolfson Centre for Mathematical Biology, Mathematical Institute, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK
| | - Paul M Kulesa
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA; Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS 66160, USA.
| |
Collapse
|
29
|
Coste C, Neirinckx V, Gothot A, Wislet S, Rogister B. Are neural crest stem cells the missing link between hematopoietic and neurogenic niches? Front Cell Neurosci 2015; 9:218. [PMID: 26136659 PMCID: PMC4469833 DOI: 10.3389/fncel.2015.00218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 05/22/2015] [Indexed: 12/24/2022] Open
Abstract
Hematopoietic niches are defined as cellular and molecular microenvironments that regulate hematopoietic stem cell (HSC) function together with stem cell autonomous mechanisms. Many different cell types have been characterized as contributors to the formation of HSC niches, such as osteoblasts, endothelial cells, Schwann cells, and mesenchymal progenitors. These mesenchymal progenitors have themselves been classified as CXC chemokine ligand (CXCL) 12-abundant reticular (CAR) cells, stem cell factor expressing cells, or nestin-positive mesenchymal stem cells (MSCs), which have been recently identified as neural crest-derived cells (NCSCs). Together, these cells are spatially associated with HSCs and believed to provide appropriate microenvironments for HSC self-renewal, differentiation, mobilization and hibernation both by cell-cell contact and soluble factors. Interestingly, it appears that regulatory pathways governing the hematopoietic niche homeostasis are operating in the neurogenic niche as well. Therefore, this review paper aims to compare both the regulation of hematopoietic and neurogenic niches, in order to highlight the role of NCSCs and nervous system components in the development and the regulation of the hematopoietic system.
Collapse
Affiliation(s)
- Cécile Coste
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Virginie Neirinckx
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - André Gothot
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Cardiovascular Sciences, University of Liège Liège, Belgium ; Hematology Department, University Hospital Liège, Belgium
| | - Sabine Wislet
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium
| | - Bernard Rogister
- Groupe Interdisciplinaire de Génoprotéomique Appliquée-Neurosciences, Unit of Nervous System Disorders and Treatment, University of Liège Liège, Belgium ; Groupe Interdisciplinaire de Génoprotéomique Appliquée-Development, Stem Cells and Regenerative Medicine, University of Liège Liège, Belgium ; Neurology Department, University Hospital Liège, Belgium
| |
Collapse
|
30
|
Kashef J, Franz CM. Quantitative methods for analyzing cell–cell adhesion in development. Dev Biol 2015; 401:165-74. [DOI: 10.1016/j.ydbio.2014.11.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/07/2014] [Accepted: 11/08/2014] [Indexed: 11/26/2022]
|
31
|
Pentimento: Neural Crest and the origin of mesectoderm. Dev Biol 2015; 401:37-61. [DOI: 10.1016/j.ydbio.2014.12.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/28/2014] [Accepted: 12/30/2014] [Indexed: 11/17/2022]
|
32
|
Rahman MS, Akhtar N, Jamil HM, Banik RS, Asaduzzaman SM. TGF-β/BMP signaling and other molecular events: regulation of osteoblastogenesis and bone formation. Bone Res 2015; 3:15005. [PMID: 26273537 PMCID: PMC4472151 DOI: 10.1038/boneres.2015.5] [Citation(s) in RCA: 398] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/26/2015] [Accepted: 02/27/2015] [Indexed: 02/08/2023] Open
Abstract
Transforming growth factor-beta (TGF-β)/bone morphogenetic protein (BMP) plays a fundamental role in the regulation of bone organogenesis through the activation of receptor serine/threonine kinases. Perturbations of TGF-β/BMP activity are almost invariably linked to a wide variety of clinical outcomes, i.e., skeletal, extra skeletal anomalies, autoimmune, cancer, and cardiovascular diseases. Phosphorylation of TGF-β (I/II) or BMP receptors activates intracellular downstream Smads, the transducer of TGF-β/BMP signals. This signaling is modulated by various factors and pathways, including transcription factor Runx2. The signaling network in skeletal development and bone formation is overwhelmingly complex and highly time and space specific. Additive, positive, negative, or synergistic effects are observed when TGF-β/BMP interacts with the pathways of MAPK, Wnt, Hedgehog (Hh), Notch, Akt/mTOR, and miRNA to regulate the effects of BMP-induced signaling in bone dynamics. Accumulating evidence indicates that Runx2 is the key integrator, whereas Hh is a possible modulator, miRNAs are regulators, and β-catenin is a mediator/regulator within the extensive intracellular network. This review focuses on the activation of BMP signaling and interaction with other regulatory components and pathways highlighting the molecular mechanisms regarding TGF-β/BMP function and regulation that could allow understanding the complexity of bone tissue dynamics.
Collapse
Affiliation(s)
- Md Shaifur Rahman
- Tissue Banking and Biomaterial Research Unit, Atomic Energy Research Establishment , Dhaka 1349, Bangladesh
| | - Naznin Akhtar
- Tissue Banking and Biomaterial Research Unit, Atomic Energy Research Establishment , Dhaka 1349, Bangladesh
| | - Hossen Mohammad Jamil
- Tissue Banking and Biomaterial Research Unit, Atomic Energy Research Establishment , Dhaka 1349, Bangladesh
| | - Rajat Suvra Banik
- Lab of Network Biology, Biotechnology and Genetic Engineering Discipline, Khulna University , Khulna 9208, Bangladesh
| | - Sikder M Asaduzzaman
- Tissue Banking and Biomaterial Research Unit, Atomic Energy Research Establishment , Dhaka 1349, Bangladesh
| |
Collapse
|
33
|
Barriga EH, Mayor R. Embryonic cell-cell adhesion: a key player in collective neural crest migration. Curr Top Dev Biol 2015; 112:301-23. [PMID: 25733144 DOI: 10.1016/bs.ctdb.2014.11.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell migration is essential for morphogenesis, adult tissue remodeling, wound healing, and cancer cell migration. Cells can migrate as individuals or groups. When cells migrate in groups, cell-cell interactions are crucial in order to promote the coordinated behavior, essential for collective migration. Interestingly, recent evidence has shown that cell-cell interactions are also important for establishing and maintaining the directionality of these migratory events. We focus on neural crest cells, as they possess extraordinary migratory capabilities that allow them to migrate and colonize tissues all over the embryo. Neural crest cells undergo an epithelial-to-mesenchymal transition at the same time than perform directional collective migration. Cell-cell adhesion has been shown to be an important source of planar cell polarity and cell coordination during collective movement. We also review molecular mechanisms underlying cadherin turnover, showing how the modulation and dynamics of cell-cell adhesions are crucial in order to maintain tissue integrity and collective migration in vivo. We conclude that cell-cell adhesion during embryo development cannot be considered as simple passive resistance to force, but rather participates in signaling events that determine important cell behaviors required for cell migration.
Collapse
Affiliation(s)
- Elias H Barriga
- Cell and Developmental Biology Department, University College London, London, United Kingdom
| | - Roberto Mayor
- Cell and Developmental Biology Department, University College London, London, United Kingdom.
| |
Collapse
|
34
|
Shamloo A, Heibatollahi M, Mofrad MRK. Directional migration and differentiation of neural stem cells within three-dimensional microenvironments. Integr Biol (Camb) 2015; 7:335-44. [PMID: 25633746 DOI: 10.1039/c4ib00144c] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Harnessing neural stem cells to repair neuronal damage is a promising potential treatment for neuronal diseases. To enable future therapeutic efficacy, the survival, proliferation, migration and differentiation of neural stem/progenitor cells (NPCs) should be accurately studied and optimized in in vitro platforms before transplanting these cells into the body for treatment purposes. Such studies can determine the appropriate quantities of the biochemical and biomechanical factors needed to control and optimize NPC behavior in vivo. In this study, NPCs were cultured within a microfluidic device while being encapsulated within the collagen matrix. The migration and differentiation of NPCs were studied in response to varying concentrations of nerve growth factor (NGF) and within varying densities of collagen matrices. It was shown that the migration and differentiation of NPCs can be significantly improved by providing the appropriate range of NGF concentrations while encapsulating the cells within the collagen matrix of optimal density. In particular, it was observed that within collagen matrices of intermediate density (0.9 mg ml(-1)), NPCs have a higher ability to migrate farther and in a collective manner while their differentiation into neurons is significantly higher and the cells can form protrusions and connections with their neighboring cells. Within collagen matrices with higher densities (1.8 mg ml(-1)), the cells did not migrate significantly as compared to the ones within lower matrix densities; within the matrices with lower collagen densities (0.45 mg ml(-1)) most of the cells migrated in an individual manner. However, no significant differentiation into neurons was observed for these two cases. It was also found that depending on the collagen matrix density, a minimum concentration of NGF caused a collective migration of NPCs, and a minimum concentration gradient of this factor stimulated the directional navigation of the cells. The results of this study can be implemented in designing platforms appropriate for regeneration of damaged neuronal systems.
Collapse
Affiliation(s)
- Amir Shamloo
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA 94720, USA.
| | | | | |
Collapse
|
35
|
Abstract
Embryonic cell migration patterns are amazingly complex in the timing and spatial distribution of cells throughout the vertebrate landscape. However, advances in in vivo visualization, cell interrogation, and computational modeling are extracting critical features that underlie the mechanistic nature of these patterns. The focus of this review highlights recent advances in the study of the highly invasive neural crest cells and their migratory patterns during embryonic development. We discuss these advances within three major themes and include a description of computational models that have emerged to more rapidly integrate and test hypothetical mechanisms of neural crest migration. We conclude with technological advances that promise to reveal new insights and help translate results to human neural crest-related birth defects and metastatic cancer.
Collapse
Affiliation(s)
- Paul M. Kulesa
- Stowers Institute for Medical Research1000 E. 50 St, Kansas City, MO 64110USA
- Department of Anatomy and Cell Biology, University of Kansas School of MedicineKansas City, KS, 66160USA
| | - Rebecca McLennan
- Stowers Institute for Medical Research1000 E. 50 St, Kansas City, MO 64110USA
| |
Collapse
|
36
|
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: 1.8] [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
|
37
|
Abstract
The subdivision of the embryo into physically distinct regions is one of the most fundamental processes in development. General hypotheses for tissue separation based on differential adhesion or tension have been proposed in the past, but with little experimental support. During the last decade, the field has experienced a strong revival, largely driven by renewed interest in biophysical modeling of development. Here, I will discuss the various models of boundary formation and summarize recent studies that have shifted our understanding of the process from the simple juxtaposition of global tissue properties to the characterization of local cellular reactions. Current evidence favors a model whereby separation is controlled by cell surface cues, which, upon cell-cell contact, generate acute changes in cytoskeletal and adhesive properties to inhibit cell mixing, and whereby the integration of multiple local cues may dictate both the global morphogenetic properties of a tissue and its separation from adjacent cell populations.
Collapse
Affiliation(s)
- François Fagotto
- Department of Biology, McGill University, Montreal, Quebec H3G 0B1, Canada
| |
Collapse
|
38
|
Zhang D, Ighaniyan S, Stathopoulos L, Rollo B, Landman K, Hutson J, Newgreen D. The neural crest: a versatile organ system. ACTA ACUST UNITED AC 2014; 102:275-98. [PMID: 25227568 DOI: 10.1002/bdrc.21081] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 08/26/2014] [Indexed: 02/02/2023]
Abstract
The neural crest is the name given to the strip of cells at the junction between neural and epidermal ectoderm in neurula-stage vertebrate embryos, which is later brought to the dorsal neural tube as the neural folds elevate. The neural crest is a heterogeneous and multipotent progenitor cell population whose cells undergo EMT then extensively and accurately migrate throughout the embryo. Neural crest cells contribute to nearly every organ system in the body, with derivatives of neuronal, glial, neuroendocrine, pigment, and also mesodermal lineages. This breadth of developmental capacity has led to the neural crest being termed the fourth germ layer. The neural crest has occupied a prominent place in developmental biology, due to its exaggerated migratory morphogenesis and its remarkably wide developmental potential. As such, neural crest cells have become an attractive model for developmental biologists for studying these processes. Problems in neural crest development cause a number of human syndromes and birth defects known collectively as neurocristopathies; these include Treacher Collins syndrome, Hirschsprung disease, and 22q11.2 deletion syndromes. Tumors in the neural crest lineage are also of clinical importance, including the aggressive melanoma and neuroblastoma types. These clinical aspects have drawn attention to the selection or creation of neural crest progenitor cells, particularly of human origin, for studying pathologies of the neural crest at the cellular level, and also for possible cell therapeutics. The versatility of the neural crest lends itself to interlinked research, spanning basic developmental biology, birth defect research, oncology, and stem/progenitor cell biology and therapy.
Collapse
|
39
|
Wakamatsu Y, Nomura T, Osumi N, Suzuki K. Comparative gene expression analyses reveal heterochrony forSox9expression in the cranial neural crest during marsupial development. Evol Dev 2014; 16:197-206. [DOI: 10.1111/ede.12083] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yoshio Wakamatsu
- Department of Developmental Neuroscience, United Centers for Advanced Research and Translational Medicine (ART); Tohoku University Graduate School of Medicine; Sendai Miyagi 980-8575 Japan
| | - Tadashi Nomura
- Department of Biology; Kyoto Prefectural University of Medicine; Kyoto 603-8334 Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, United Centers for Advanced Research and Translational Medicine (ART); Tohoku University Graduate School of Medicine; Sendai Miyagi 980-8575 Japan
| | - Kunihiro Suzuki
- Department of Biology; Nihon University School of Dentistry at Matsudo; Chiba 271-8587 Japan
| |
Collapse
|
40
|
Gong SG. Cranial neural crest: migratory cell behavior and regulatory networks. Exp Cell Res 2014; 325:90-5. [PMID: 24680987 DOI: 10.1016/j.yexcr.2014.03.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 03/16/2014] [Accepted: 03/18/2014] [Indexed: 12/19/2022]
Abstract
Defects of the head and neck region account for a substantial portion of all human birth disorders. The high incidence of malformations in this region may be attributed in part to the intricate means by which the facial region is assembled during embryonic development. The starting constituent for the majority of skeletal and connective tissues in the face is a pluripotent population of cells, the cranial neural crest (CNC) cells. This population of cells exhibit remarkable migratory abilities and diversity of potential cell types. This review draws on extensive research that has been done in the field, focusing specifically on findings generated in the last decade on cell behavior and the gene regulatory networks of migratory CNC cells. In the later part of this review, the importance of the CNC cells in the overall development of the craniofacial region will be illustrated with a discussion of a craniofacial birth defect, the Treacher Collins syndrome. The next decade will most likely herald in an era of greater understanding of the integrative molecular networks at different stages of the development of the CNC cells. Such new information is essential towards a better understanding the etiology and pathogenesis of the many craniofacial birth defects and will ultimately lead to new therapeutic modalities.
Collapse
Affiliation(s)
- Siew-Ging Gong
- Department of Orthodontics, Dental Research Institute, Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, Ontario, Canada M5G 1G6.
| |
Collapse
|
41
|
Young HM, Bergner AJ, Simpson MJ, McKeown SJ, Hao MM, Anderson CR, Enomoto H. Colonizing while migrating: how do individual enteric neural crest cells behave? BMC Biol 2014; 12:23. [PMID: 24670214 PMCID: PMC4101823 DOI: 10.1186/1741-7007-12-23] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/21/2014] [Indexed: 12/15/2022] Open
Abstract
Background Directed cell migration is essential for normal development. In most of the migratory cell populations that have been analyzed in detail to date, all of the cells migrate as a collective from one location to another. However, there are also migratory cell populations that must populate the areas through which they migrate, and thus some cells get left behind while others advance. Very little is known about how individual cells behave to achieve concomitant directional migration and population of the migratory route. We examined the behavior of enteric neural crest-derived cells (ENCCs), which must both advance caudally to reach the anal end and populate each gut region. Results The behavior of individual ENCCs was examined using live imaging and mice in which ENCCs express a photoconvertible protein. We show that individual ENCCs exhibit very variable directionalities and speed; as the migratory wavefront of ENCCs advances caudally, each gut region is populated primarily by some ENCCs migrating non-directionally. After populating each region, ENCCs remain migratory for at least 24 hours. Endothelin receptor type B (EDNRB) signaling is known to be essential for the normal advance of the ENCC population. We now show that perturbation of EDNRB principally affects individual ENCC speed rather than directionality. The trajectories of solitary ENCCs, which occur transiently at the wavefront, were consistent with an unbiased random walk and so cell-cell contact is essential for directional migration. ENCCs migrate in close association with neurites. We showed that although ENCCs often use neurites as substrates, ENCCs lead the way, neurites are not required for chain formation and neurite growth is more directional than the migration of ENCCs as a whole. Conclusions Each gut region is initially populated by sub-populations of ENCCs migrating non-directionally, rather than stopping. This might provide a mechanism for ensuring a uniform density of ENCCs along the growing gut.
Collapse
Affiliation(s)
- Heather M Young
- Department of Anatomy & Neuroscience, University of Melbourne, Melbourne 3010 VIC, Australia.
| | | | | | | | | | | | | |
Collapse
|
42
|
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: 1.4] [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
|
43
|
Czarnobaj J, Bagnall KM, Bamforth JS, Milos NC. The different effects on cranial and trunk neural crest cell behaviour following exposure to a low concentration of alcohol in vitro. Arch Oral Biol 2014; 59:500-12. [PMID: 24631632 DOI: 10.1016/j.archoralbio.2014.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 12/17/2013] [Accepted: 02/10/2014] [Indexed: 11/19/2022]
Abstract
Embryonic neural crest cells give rise to large regions of the face and peripheral nervous system. Exposure of these cells to high alcohol concentrations leads to cell death in the craniofacial region resulting in facial defects. However, the effects of low concentrations of alcohol on neural crest cells are not clear. In this study, cranial neural crest cells from Xenopus laevis were cultured in an ethanol concentration approximately equivalent to one drink. Techniques were developed to study various aspects of neural crest cell behaviour and a number of cellular parameters were quantified. In the presence of alcohol, a significant number of cranial neural crest cells emigrated from the explant on fibronectin but the liberation of individual cells was delayed. The cells also remained close to the explant and their morphology changed. Cranial neural crest cells did not grow on Type 1 collagen. For the purposes of comparison, the behaviour of trunk neural crest cells was also studied. The presence of alcohol correlated with increased retention of single cells on fibronectin but left other parameters unchanged. The behaviour of trunk neural crest cells growing on Type 1 collagen in the presence of alcohol did not differ from controls. Low concentrations of alcohol therefore significantly affected both cranial and trunk neural crest cells, with a wider variety of effects on cells from the cranial as opposed to the trunk region. The results suggest that low concentrations of alcohol may be more detrimental to early events in organ formation than currently suspected.
Collapse
Affiliation(s)
- Joanna Czarnobaj
- Department of Dentistry, Faculty of Medicine and Dentistry, 7020 Katz Building University of Alberta, Edmonton, Alberta, Canada T6G 2E1.
| | - Keith M Bagnall
- Department of Anatomy, Faculty of Medicine and Health Sciences, United Arab Emirates University, Box 17666 Al Ain, United Arab Emirates.
| | - J Steven Bamforth
- Department of Medical Genetics, Faculty of Medicine and Dentistry, 8-53 Medical Science Building, University of Alberta, Edmonton, Alberta, Canada T6G 2H7.
| | - Nadine C Milos
- Department of Dentistry, Faculty of Medicine and Dentistry, 7020 Katz Building University of Alberta, Edmonton, Alberta, Canada T6G 2E1.
| |
Collapse
|
44
|
Tondeleir D, Noelanders R, Bakkali K, Ampe C. Beta-actin is required for proper mouse neural crest ontogeny. PLoS One 2014; 9:e85608. [PMID: 24409333 PMCID: PMC3883714 DOI: 10.1371/journal.pone.0085608] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/05/2013] [Indexed: 02/06/2023] Open
Abstract
The mouse genome consists of six functional actin genes of which the expression patterns are temporally and spatially regulated during development and in the adult organism. Deletion of beta-actin in mouse is lethal during embryonic development, although there is compensatory expression of other actin isoforms. This suggests different isoform specific functions and, more in particular, an important function for beta-actin during early mammalian development. We here report a role for beta-actin during neural crest ontogeny. Although beta-actin null neural crest cells show expression of neural crest markers, less cells delaminate and their migration arrests shortly after. These phenotypes were associated with elevated apoptosis levels in neural crest cells, whereas proliferation levels were unchanged. Specifically the pre-migratory neural crest cells displayed higher levels of apoptosis, suggesting increased apoptosis in the neural tube accounts for the decreased amount of migrating neural crest cells seen in the beta-actin null embryos. These cells additionally displayed a lack of membrane bound N-cadherin and dramatic decrease in cadherin-11 expression which was more pronounced in the pre-migratory neural crest population, potentially indicating linkage between the cadherin-11 expression and apoptosis. By inhibiting ROCK ex vivo, the knockout neural crest cells regained migratory capacity and cadherin-11 expression was upregulated. We conclude that the presence of beta-actin is vital for survival, specifically of pre-migratory neural crest cells, their proper emigration from the neural tube and their subsequent migration. Furthermore, the absence of beta-actin affects cadherin-11 and N-cadherin function, which could partly be alleviated by ROCK inhibition, situating the Rho-ROCK signaling in a feedback loop with cadherin-11.
Collapse
Affiliation(s)
- Davina Tondeleir
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Rivka Noelanders
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Karima Bakkali
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Christophe Ampe
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|
45
|
Hua ZL, Smallwood PM, Nathans J. Frizzled3 controls axonal development in distinct populations of cranial and spinal motor neurons. eLife 2013; 2:e01482. [PMID: 24347548 PMCID: PMC3865743 DOI: 10.7554/elife.01482] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Disruption of the Frizzled3 (Fz3) gene leads to defects in axonal growth in the VIIth and XIIth cranial motor nerves, the phrenic nerve, and the dorsal motor nerve in fore- and hindlimbs. In Fz3−/− limbs, dorsal axons stall at a precise location in the nerve plexus, and, in contrast to the phenotypes of several other axon path-finding mutants, Fz3−/− dorsal axons do not reroute to other trajectories. Affected motor neurons undergo cell death 2 days prior to the normal wave of developmental cell death that coincides with innervation of muscle targets, providing in vivo evidence for the idea that developing neurons with long-range axons are programmed to die unless their axons arrive at intermediate targets on schedule. These experiments implicate planar cell polarity (PCP) signaling in motor axon growth and they highlight the question of how PCP proteins, which form cell–cell complexes in epithelia, function in the dynamic context of axonal growth. DOI:http://dx.doi.org/10.7554/eLife.01482.001 For the nervous system to become wired up correctly, neurons within the developing embryo must project over long distances to form connections with remote targets. They do this by lengthening their axons—the ‘cables’ along which electrical signals flow—and some axons in adult humans can grow to be more than 1 metre long. This type of long-range pathfinding activity is particularly common for neurons that control movement, as many of these neurons must establish connections with muscles that are some distance away from the brain. For example, motor neurons in the brainstem form connections with muscles in the face to control facial expressions, while motor neurons in parts of the spinal cord project to muscles in the limbs. Multiple signaling pathways tell the developing axons which direction to grow en route to their final targets. Now, Hua et al. have shown that an evolutionarily conserved protein called Frizzled3 is also involved in this process. In mouse embryos that lacked Frizzled3, the motor nerves that control breathing and limb movements were thinner than those in normal mice. In the mutant animals, many motor axons failed to form connections with their targets. Instead, these axons came to an abrupt halt midway along their intended paths and the neurons from which they originated died soon afterwards. These experiments support the idea that developing neurons are programmed to die unless their axons progress on the appropriate schedule. As well as increasing our knowledge of the networks of connections that form within the developing mammalian nervous system, the work of Hua et al. provides new insights into some of the molecular mechanisms by which these connections are established. DOI:http://dx.doi.org/10.7554/eLife.01482.002
Collapse
Affiliation(s)
- Zhong L Hua
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States
| | | | | |
Collapse
|
46
|
Christian L, Bahudhanapati H, Wei S. Extracellular metalloproteinases in neural crest development and craniofacial morphogenesis. Crit Rev Biochem Mol Biol 2013; 48:544-60. [PMID: 24066766 DOI: 10.3109/10409238.2013.838203] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The neural crest (NC) is a population of migratory stem/progenitor cells that is found in early vertebrate embryos. NC cells are induced during gastrulation, and later migrate to multiple destinations and contribute to many types of cells and tissues, such as craniofacial structures, cardiac tissues, pigment cells and the peripheral nervous system. Recently, accumulating evidence suggests that many extracellular metalloproteinases, including matrix metalloproteinases (MMPs), a disintegrin and metalloproteinases (ADAMs), and ADAMs with thrombospondin motifs (ADAMTSs), play important roles in various stages of NC development. Interference with metalloproteinase functions often causes defects in craniofacial structures, as well as in other cells and tissues that are contributed by NC cells, in humans and other vertebrates. In this review, we summarize the current state of the field concerning the roles of these three families of metalloproteinases in NC development and related tissue morphogenesis, with a special emphasis on craniofacial morphogenesis.
Collapse
Affiliation(s)
- Laura Christian
- Department of Biology, West Virginia University , Morgantown, WV , USA
| | | | | |
Collapse
|
47
|
Macara IG, McCaffrey L. Cell polarity in morphogenesis and metastasis. Philos Trans R Soc Lond B Biol Sci 2013; 368:20130012. [PMID: 24062582 DOI: 10.1098/rstb.2013.0012] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Most human cancers arise either from epithelial cells or their progenitors. Epithelial cells possess a distinctive apical-basal polarity and loss of polarity is frequently assumed to be a common feature of cancer progression. In particular, cancer cell dissemination to ectopic sites, and metastatic growth at those sites, is often considered to require a mesenchymal transition in which the transformed epithelial cells lose their apical-basal polarity. However, many cancers retain epithelial characteristics, and until recently there has been little conclusive evidence for an involvement of the cell polarity machinery in tumour growth and metastasis. In this article, we discuss evidence that polarity proteins can be potent invasion suppressors but that loss of epithelial character is not essential either for tumour growth and invasion, or metastatic colonization.
Collapse
Affiliation(s)
- Ian G Macara
- Department of Cell and Developmental Biology, Vanderbilt University, , Nashville, TN 37215, USA
| | | |
Collapse
|
48
|
Abstract
The neural crest (NC) is a highly migratory multipotent cell population that forms at the interface between the neuroepithelium and the prospective epidermis of a developing embryo. Following extensive migration throughout the embryo, NC cells eventually settle to differentiate into multiple cell types, ranging from neurons and glial cells of the peripheral nervous system to pigment cells, fibroblasts to smooth muscle cells, and odontoblasts to adipocytes. NC cells migrate in large numbers and their migration is regulated by multiple mechanisms, including chemotaxis, contact-inhibition of locomotion and cell sorting. Here, we provide an overview of NC formation, differentiation and migration, highlighting the molecular mechanisms governing NC migration.
Collapse
Affiliation(s)
- Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| | | |
Collapse
|
49
|
Watanabe Y, Broders-Bondon F, Baral V, Paul-Gilloteaux P, Pingault V, Dufour S, Bondurand N. Sox10 and Itgb1 interaction in enteric neural crest cell migration. Dev Biol 2013; 379:92-106. [PMID: 23608456 DOI: 10.1016/j.ydbio.2013.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 04/10/2013] [Accepted: 04/12/2013] [Indexed: 01/11/2023]
Abstract
SOX10 involvement in syndromic form of Hirschsprung disease (intestinal aganglionosis, HSCR) in humans as well as developmental defects in animal models highlight the importance of this transcription factor in control of the pool of enteric progenitors and their differentiation. Here, we characterized the role of SOX10 in cell migration and its interactions with β1-integrins. To this end, we crossed the Sox10(lacZ/+) mice with the conditional Ht-PA::Cre; beta1(neo/+) and beta1(fl/fl) mice and compared the phenotype of embryos of different genotypes during enteric nervous system (ENS) development. The Sox10(lacZ/+); Ht-PA::Cre; beta1(neo/fl) double mutant embryos presented with increased intestinal aganglionosis length and more severe neuronal network disorganization compared to single mutants. These defects, detected by E11.5, are not compensated after birth, showing that a coordinated and balanced interaction between these two genes is required for normal ENS development. Use of video-microscopy revealed that defects observed result from reduced migration speed and altered directionality of enteric neural crest cells. Expression of β1-integrins upon SOX10 overexpression or in Sox10(lacZ/+) mice was also analyzed. The modulation of SOX10 expression altered β1-integrins, suggesting that SOX10 levels are critical for proper expression and function of this adhesion molecule. Together with previous studies, our results strongly indicate that SOX10 mediates ENCC adhesion and migration, and contribute to the understanding of the molecular and cellular basis of ENS defects observed both in mutant mouse models and in patients carrying SOX10 mutations.
Collapse
Affiliation(s)
- Yuli Watanabe
- INSERM U955, Equipe 11, F-94000 Créteil, France; Université Paris-Est, UMR_S955, UPEC, F-94000 Créteil, France
| | | | | | | | | | | | | |
Collapse
|
50
|
Endo Y, Ishiwata-Endo H, Yamada KM. Cloning and characterization of chicken α5 integrin: endogenous and experimental expression in early chicken embryos. Matrix Biol 2013; 32:381-6. [PMID: 23587661 DOI: 10.1016/j.matbio.2013.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/08/2013] [Accepted: 04/08/2013] [Indexed: 11/19/2022]
Abstract
Key roles for fibronectin and its integrin receptors have been postulated in the multiple cell-matrix interactions essential for chick embryo morphogenesis. However, mechanistic studies of these processes have been hampered by the current absence of sequence data and chicken cDNA clones for the major fibronectin receptor subunit, integrin α5 (ITGA5). We report here the sequence, endogenous expression pattern, and transfection of full-length chicken integrin α5. During early chicken embryonic development, α5 is highly expressed in cranial neural folds and migrating neural crest cells, suggesting potential roles in neural crest formation and migration. In fact, over-expression of this integrin in early neural tube selectively induces BMP5, a growth factor recently implicated in neural crest formation. Availability of these α5 integrin tools should facilitate studies of its functions in early embryonic development.
Collapse
Affiliation(s)
- Yukinori Endo
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, United States.
| | | | | |
Collapse
|