1
|
Lou M, Zhang S, Yang W, Li S, Cao H, Zhang Z, Ling Y. Transcriptome analysis revealed the mechanism of skeletal muscle growth and development in different hybrid sheep. Anim Biosci 2025; 38:408-418. [PMID: 39483021 PMCID: PMC11917423 DOI: 10.5713/ab.24.0269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/15/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE This study aimed to analyze the molecular mechanism of heterosis in East Friesian sheep×Hu sheep (EH) hybrid sheep and Suffolk×EH (SHE) hybrid sheep (Ovis aries). METHODS In this research, the growth performance data of Hu sheep (H), EH and SHE from birth to 8 months of age were analyzed. Three 8-month-old sheep of each of the three strains (9 sheep in total) were chosen and their longissimus dorsi muscles were collected for transcriptome sequencing. We verified the expression of seven differentially expressed genes (DEGs) by real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR). RESULTS The results showed: (1) body weight and chest circumference of EH were significantly greater than H (p<0.05), except at 4 months of age. Body weight and chest circumference of SHE was significantly higher than EH (p<0.05), except at 6 months of age. (2) 310 DEGs were screened in the EH and H, gene ontology and Kyoto encyclopedia of genes and genomes showed DEGs were mainly concentrate on the categories of actin cytoskeleton, calcium binding, cGMP-PKG and mitogen-activated protein kinase (MAPK) signaling pathway, which correlating the development of skeletal muscle and energy metabolism. 329 DEGs were screened in the SHE and EH. DEGs were mainly enriched in extracellular matrix-receptor interactions and cell adhesion molecules. (3) Protein-protein interaction screening yielded five (MYL2, TNNI1, TNNI3, MYH11, TNNC1) and three (SOX10, COL2A1, MPZ) pivotal DEGs regulating muscle development in EH and SHE. (4) RT-qPCR test results were consistent with transcriptome sequencing. CONCLUSION This study provides candidate genes for improving sheep growth traits. It provides a theoretical basis for analyzing the mechanism of muscle development in crossbred sheep.
Collapse
Affiliation(s)
- Mengyu Lou
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Provincial Key Laboratory of Local Livestock and Poultry Genetic Resources Protection and Biological Breeding, Hefei 230036, China
| | - Sihuan Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Provincial Key Laboratory of Local Livestock and Poultry Genetic Resources Protection and Biological Breeding, Hefei 230036, China
| | - Wangxin Yang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Provincial Key Laboratory of Local Livestock and Poultry Genetic Resources Protection and Biological Breeding, Hefei 230036, China
| | - Shuang Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Provincial Key Laboratory of Local Livestock and Poultry Genetic Resources Protection and Biological Breeding, Hefei 230036, China
| | - Hongguo Cao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Provincial Key Laboratory of Local Livestock and Poultry Genetic Resources Protection and Biological Breeding, Hefei 230036, China
| | - Zijun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Provincial Key Laboratory of Local Livestock and Poultry Genetic Resources Protection and Biological Breeding, Hefei 230036, China
| | - Yinghui Ling
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Provincial Key Laboratory of Local Livestock and Poultry Genetic Resources Protection and Biological Breeding, Hefei 230036, China
| |
Collapse
|
2
|
Wang B, Guo H, Han Z, Wu S, Liu J, Lin Z, An F, Zhu J, Li M. NRG1 Regulates Proliferation, Migration and Differentiation of Human Limbal Epithelial Stem Cells. Curr Issues Mol Biol 2023; 45:10121-10130. [PMID: 38132478 PMCID: PMC10742012 DOI: 10.3390/cimb45120632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/10/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Limbal epithelial stem/progenitor cells (LESCs) proliferate, migrate and differentiate into mature corneal epithelium cells (CECs) that cover the ocular surface. LESCs play a crucial role in the maintenance and regeneration of the corneal epithelium, and their dysfunction can lead to various corneal diseases. Neuregulin 1 (NRG1) is a member of the epidermal growth factor family that regulates the growth and differentiation of epithelial tissues. Here, we depicted the dynamic transcriptomic profiles during human CEC differentiation, identifying six gene co-expression modules that were specific to different differentiation stages. We found that the expression of NRG1 was high in human LESCs and decreased dramatically upon differentiation. Knockdown of NRG1 significantly inhibited LESC proliferation and upregulated the expression of the terminal differentiation marker genes KRT3, KRT12 and CLU. In addition, the scratch wound closure assay showed that knockdown of NRG1 attenuated wound closure of LESCs over 24 h. Together, we dissected the transcriptional regulatory dynamics during CEC differentiation and identified NRG1 as a key regulator that promoted LESC proliferation and migration and maintained the undifferentiated state.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mingsen Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; (B.W.); (Z.H.)
| |
Collapse
|
3
|
Ninfali C, Siles L, Esteve-Codina A, Postigo A. The mesodermal and myogenic specification of hESCs depend on ZEB1 and are inhibited by ZEB2. Cell Rep 2023; 42:113222. [PMID: 37819755 DOI: 10.1016/j.celrep.2023.113222] [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: 03/12/2022] [Revised: 08/02/2023] [Accepted: 09/20/2023] [Indexed: 10/13/2023] Open
Abstract
Human embryonic stem cells (hESCs) can differentiate into any cell lineage. Here, we report that ZEB1 and ZEB2 promote and inhibit mesodermal-to-myogenic specification of hESCs, respectively. Knockdown and/or overexpression experiments of ZEB1, ZEB2, or PAX7 in hESCs indicate that ZEB1 is required for hESC Nodal/Activin-mediated mesodermal specification and PAX7+ human myogenic progenitor (hMuP) generation, while ZEB2 inhibits these processes. ZEB1 downregulation induces neural markers, while ZEB2 downregulation induces mesodermal/myogenic markers. Mechanistically, ZEB1 binds to and transcriptionally activates the PAX7 promoter, while ZEB2 binds to and activates the promoter of the neural OTX2 marker. Transplanting ZEB1 or ZEB2 knocked down hMuPs into the muscles of a muscular dystrophy mouse model, showing that hMuP engraftment and generation of dystrophin-positive myofibers depend on ZEB1 and are inhibited by ZEB2. The mouse model results suggest that ZEB1 expression and/or downregulating ZEB2 in hESCs may also enhance hESC regenerative capacity for human muscular dystrophy therapy.
Collapse
Affiliation(s)
- Chiara Ninfali
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036 Barcelona, Spain
| | - Laura Siles
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036 Barcelona, Spain
| | | | - Antonio Postigo
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036 Barcelona, Spain; Molecular Targets Program, J.G. Brown Center, Louisville University Healthcare Campus, Louisville, KY 40202, USA; ICREA, 08010 Barcelona, Spain.
| |
Collapse
|
4
|
Chi S, Li S, Cao G, Guo J, Han Y, Zhou Y, Zhang X, Li Y, Luo Z, Li X, Rong L, Zhang M, Li L, Tang S. The interplay of common genetic variants NRG1 rs2439302 and RET rs2435357 increases the risk of developing Hirschsprung's disease. Front Cell Dev Biol 2023; 11:1184799. [PMID: 37484916 PMCID: PMC10361661 DOI: 10.3389/fcell.2023.1184799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/14/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction: As a congenital and genetically related disease, many single nucleotide polymorphisms (SNPs) have been reported to be associated with the risk of HSCR. Our previous research showed that SNP rs2439302 (NRG1) interacted with rs2435357 (RET) to increase the risk of HSCR development. However, the underlying molecular mechanism is still not well understood. Methods: SNP rs2439302 (NRG1) and rs2435357 (RET) were genotyped in 470 HSCR cases. The expression of NRG1 and RET was investigated in the colon of HSCR patients. Knockdown of the NRG1 and RET homologs was performed in zebrafish to investigate their synergistic effect on ENS development. The effect of SNP rs2439302 and rs2435357 polymorphism on neuron proliferation, migration, and differentiation were investigated in SHSY-5Y cells and IPSCs. Results: Significant downregulation of NRG1 and RET expression was noticed in the aganglionic segment of HSCR patients and SHSY-5Y cells with rs2439302 GG/rs2435357 TT genotype. NRG1 and RET double mutants caused the most severe reduction in enteric neuron numbers than NRG1 single mutant or RET single mutant in the hindgut of zebrafish. SHSY-5Y cells and IPSCs with rs2439302 GG/rs2435357 TT genotype exhibited a decreased proliferative, migration, and differentiative capacity. CTCF showed a considerably higher binding ability to SNP rs2439302 CC than GG. NRG1 reduction caused a further decrease in SOX10 expression via the PI3K/Akt pathway, which regulates RET expression by directly binding to rs2435357. Discussion: SNP rs2439302 (NRG1) GG increases the risk of developing HSCR by affecting the binding of transcription factor CTCF and interacting with rs2435357 (RET) to regulate RET expression via the PI3K/Akt/SOX10 pathway.
Collapse
Affiliation(s)
- Shuiqing Chi
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoqing Cao
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jialing Guo
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunqiao Han
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Zhou
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yibo Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhibin Luo
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyang Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liying Rong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengxin Zhang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linglu Li
- China Zebrafish Resource Center, National Aquatic Biological Resource Center, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shaotao Tang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Hicks MR, Pyle AD. The emergence of the stem cell niche. Trends Cell Biol 2023; 33:112-123. [PMID: 35934562 PMCID: PMC9868094 DOI: 10.1016/j.tcb.2022.07.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 02/03/2023]
Abstract
Stem cell niches are composed of dynamic microenvironments that support stem cells over a lifetime. The emerging niche is distinct from the adult because its main role is to support the progenitors that build organ systems in development. Emerging niches mature through distinct stages to form the adult niche and enable proper stem cell support. As a model of emerging niches, this review highlights how differences in the skeletal muscle microenvironment influence emerging versus satellite cell (SC) niche formation in skeletal muscle, which is among the most regenerative tissue systems. We contrast how stem cell niches regulate intrinsic properties between progenitor and stem cells throughout development to adulthood. We describe new applications for generating emerging niches from human pluripotent stem cells (hPSCs) using developmental principles and highlight potential applications for regeneration and therapeutics.
Collapse
Affiliation(s)
- Michael R Hicks
- Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - April D Pyle
- Microbiology, Immunology, and Molecular Genetics, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Cai S, Hu B, Wang X, Liu T, Lin Z, Tong X, Xu R, Chen M, Duo T, Zhu Q, Liang Z, Li E, Chen Y, Li J, Liu X, Mo D. Integrative single-cell RNA-seq and ATAC-seq analysis of myogenic differentiation in pig. BMC Biol 2023; 21:19. [PMID: 36726129 PMCID: PMC9893630 DOI: 10.1186/s12915-023-01519-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Skeletal muscle development is a multistep process whose understanding is central in a broad range of fields and applications, from the potential medical value to human society, to its economic value associated with improvement of agricultural animals. Skeletal muscle initiates in the somites, with muscle precursor cells generated in the dermomyotome and dermomyotome-derived myotome before muscle differentiation ensues, a developmentally regulated process that is well characterized in model organisms. However, the regulation of skeletal muscle ontogeny during embryonic development remains poorly defined in farm animals, for instance in pig. Here, we profiled gene expression and chromatin accessibility in developing pig somites and myotomes at single-cell resolution. RESULTS We identified myogenic cells and other cell types and constructed a differentiation trajectory of pig skeletal muscle ontogeny. Along this trajectory, the dynamic changes in gene expression and chromatin accessibility coincided with the activities of distinct cell type-specific transcription factors. Some novel genes upregulated along the differentiation trajectory showed higher expression levels in muscular dystrophy mice than that in healthy mice, suggesting their involvement in myogenesis. Integrative analysis of chromatin accessibility, gene expression data, and in vitro experiments identified EGR1 and RHOB as critical regulators of pig embryonic myogenesis. CONCLUSIONS Collectively, our results enhance our understanding of the molecular and cellular dynamics in pig embryonic myogenesis and offer a high-quality resource for the further study of pig skeletal muscle development and human muscle disease.
Collapse
Affiliation(s)
- Shufang Cai
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640 Guangdong China
| | - Bin Hu
- Guangdong Key Laboratory of Animal Breeding and Nutrition, State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640 Guangdong China
| | - Xiaoyu Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Tongni Liu
- Faculty of Forestry, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Zhuhu Lin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Xian Tong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Rong Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Meilin Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Tianqi Duo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Qi Zhu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Ziyun Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Enru Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Jianhao Li
- Guangdong Key Laboratory of Animal Breeding and Nutrition, State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640 Guangdong China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| |
Collapse
|
7
|
Li J, Cao X, Chu T, Lin K, Chen L, Lv J, Tan Y, Chen M, Li M, Wang K, Zheng Q, Li D. The circHMGCS1-miR-205-5p-ErBB3 axis mediated the Sanggenon C-induced anti-proliferation effects on human prostate cancer. Pharmacol Res 2023; 187:106584. [PMID: 36462326 DOI: 10.1016/j.phrs.2022.106584] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022]
Abstract
Prostate cancer (PCa) is associated with a high incidence and metastasis rate globally, resulting in an unsatisfactory prognosis and a huge economic burden due to the current deficient of therapeutic strategies. As the most abundant component of Cortex Mori, Sanggenon C (SC) is well known to possess bioactivities in tumors, but its mechanism is poorly understood. Consequently, we attempted to investigate whether SC could modulate circular RNA(s) levels and hence anti-PCa development. We found that SC dramatically promoted cell apoptosis and induced G0/G1 phase arrest in PCa cell lines via the circHMGCS1-miR-205-5p-ErBB3 axis. In brief, circHMGCS1 is highly expressed in PCa and is positively correlated with the degree of malignancy. Over-expression of circHMGCS1 is not only associated with the proliferation of PCa cells but also blocks SC-induced pro-apoptotic effects. As a verified sponge of circHMGCS1, miR-205-5p is down-regulated in PCa tumors, which negatively regulates PCa cell proliferation by modulating ErBB3 expression. After miR-205-5p mimics or inhibitors were used to transfect PCa cells, the effects of circHMGCS1 OE and SC on PCa cells were completely diminished. Similar to miR-205-5p inhibitors, siErBB3 could oppose SC-triggered pro-apoptotic effects on PCa cells. All these results were confirmed in vivo. Together, SC exerts its anti-tumor effects on PCa by inhibiting circHMGCS1 expression and results in the latter losing the ability to sponge miR-205-5p. Subsequently, unfettered miR-205-5p could mostly down-regulate ErBB3 expression by binding to the 5'UTR of ErBB3 mRNA, which eventually resulted in PCa cell cycle arrest and pro-apoptosis.
Collapse
Affiliation(s)
- Jie Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Xinyue Cao
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Ting Chu
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Kehao Lin
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Lei Chen
- School of Nursing, Binzhou Medical University, 264003, Yantai, China
| | - Junlin Lv
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Yujun Tan
- Center for New Drug Safety Evaluation of Lunan Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., 273400, Linyi, China
| | - Miaomiao Chen
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Minjing Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Kejun Wang
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Qiusheng Zheng
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China.
| | - Defang Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China.
| |
Collapse
|
8
|
Li LK, Huang WC, Hsueh YY, Yamauchi K, Olivares N, Davila R, Fang J, Ding X, Zhao W, Soto J, Hasani M, Novitch B, Li S. Intramuscular delivery of neural crest stem cell spheroids enhances neuromuscular regeneration after denervation injury. Stem Cell Res Ther 2022; 13:205. [PMID: 35578348 PMCID: PMC9109326 DOI: 10.1186/s13287-022-02877-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 03/28/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Muscle denervation from trauma and motor neuron disease causes disabling morbidities. A limiting step in functional recovery is the regeneration of neuromuscular junctions (NMJs) for reinnervation. Stem cells have the potential to promote these regenerative processes, but current approaches have limited success, and the optimal types of stem cells remain to be determined. Neural crest stem cells (NCSCs), as the developmental precursors of the peripheral nervous system, are uniquely advantageous, but the role of NCSCs in neuromuscular regeneration is not clear. Furthermore, a cell delivery approach that can maintain NCSC survival upon transplantation is critical. METHODS We established a streamlined protocol to derive, isolate, and characterize functional p75+ NCSCs from human iPSCs without genome integration of reprogramming factors. To enhance survival rate upon delivery in vivo, NCSCs were centrifuged in microwell plates to form spheroids of desirable size by controlling suspension cell density. Human bone marrow mesenchymal stem cells (MSCs) were also studied for comparison. NCSC or MSC spheroids were injected into the gastrocnemius muscle with denervation injury, and the effects on NMJ formation and functional recovery were investigated. The spheroids were also co-cultured with engineered neuromuscular tissue to assess effects on NMJ formation in vitro. RESULTS NCSCs cultured in spheroids displayed enhanced secretion of soluble factors involved in neuromuscular regeneration. Intramuscular transplantation of spheroids enabled long-term survival and retention of NCSCs, in contrast to the transplantation of single-cell suspensions. Furthermore, NCSC spheroids significantly improved functional recovery after four weeks as shown by gait analysis, electrophysiology, and the rate of NMJ innervation. MSC spheroids, on the other hand, had insignificant effect. In vitro co-culture of NCSC or MSC spheroids with engineered myotubes and motor neurons further evidenced improved innervated NMJ formation with NCSC spheroids. CONCLUSIONS We demonstrate that stem cell type is critical for neuromuscular regeneration and that NCSCs have a distinct advantage and therapeutic potential to promote reinnervation following peripheral nerve injury. Biophysical effects of spheroidal culture, in particular, enable long-term NCSC survival following in vivo delivery. Furthermore, synthetic neuromuscular tissue, or "tissues-on-a-chip," may offer a platform to evaluate stem cells for neuromuscular regeneration.
Collapse
Affiliation(s)
- LeeAnn K Li
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
- David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Wen-Chin Huang
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Yuan-Yu Hsueh
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ken Yamauchi
- Department of Neurobiology, University of California, Los Angeles, USA
| | - Natalie Olivares
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Raul Davila
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Jun Fang
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Xili Ding
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Weikang Zhao
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Jennifer Soto
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Mahdi Hasani
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Bennett Novitch
- Department of Neurobiology, University of California, Los Angeles, USA
| | - Song Li
- Departments of Bioengineering and Department of Medicine, University of California, Los Angeles, USA.
| |
Collapse
|
9
|
Galindo CL, Nguyen VT, Hill B, Easterday E, Cleator JH, Sawyer DB. Neuregulin (NRG-1β) Is Pro-Myogenic and Anti-Cachectic in Respiratory Muscles of Post-Myocardial Infarcted Swine. BIOLOGY 2022; 11:682. [PMID: 35625411 PMCID: PMC9137990 DOI: 10.3390/biology11050682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/30/2022]
Abstract
Neuregulin-1β (NRG-1β) is a growth and differentiation factor with pleiotropic systemic effects. Because NRG-1β has therapeutic potential for heart failure and has known growth effects in skeletal muscle, we hypothesized that it might affect heart failure-associated cachexia, a severe co-morbidity characterized by a loss of muscle mass. We therefore assessed NRG-1β's effect on intercostal skeletal muscle gene expression in a swine model of heart failure using recombinant glial growth factor 2 (USAN-cimaglermin alfa), a version of NRG-1β that has been tested in humans with systolic heart failure. Animals received one of two intravenous doses (0.67 or 2 mg/kg) of NRG-1β bi-weekly for 4 weeks, beginning one week after infarct. Based on paired-end RNA sequencing, NRG-1β treatment altered the intercostal muscle gene expression of 581 transcripts, including genes required for myofiber growth, maintenance and survival, such as MYH3, MYHC, MYL6B, KY and HES1. Importantly, NRG-1β altered the directionality of at least 85 genes associated with cachexia, including myostatin, which negatively regulates myoblast differentiation by down-regulating MyoD expression. Consistent with this, MyoD was increased in NRG-1β-treated animals. In vitro experiments with myoblast cell lines confirmed that NRG-1β induces ERBB-dependent differentiation. These findings suggest a NRG-1β-mediated anti-atrophic, anti-cachexia effect that may provide additional benefits to this potential therapy in heart failure.
Collapse
Affiliation(s)
- Cristi L. Galindo
- Department of Biology, Ogden College of Science & Engineering, Western Kentucky University, Bowling Green, KY 42101, USA; (V.T.N.); (B.H.); (E.E.)
| | - Van Thuan Nguyen
- Department of Biology, Ogden College of Science & Engineering, Western Kentucky University, Bowling Green, KY 42101, USA; (V.T.N.); (B.H.); (E.E.)
| | - Braxton Hill
- Department of Biology, Ogden College of Science & Engineering, Western Kentucky University, Bowling Green, KY 42101, USA; (V.T.N.); (B.H.); (E.E.)
| | - Ethan Easterday
- Department of Biology, Ogden College of Science & Engineering, Western Kentucky University, Bowling Green, KY 42101, USA; (V.T.N.); (B.H.); (E.E.)
| | - John H. Cleator
- Centennial Heart at Skyline, 3443 Dickerson Pike, Suite 430, Nashville, TN 37207, USA;
| | - Douglas B. Sawyer
- Department of Cardiac Services, Maine Medical Center, Scarborough, ME 04074, USA
| |
Collapse
|
10
|
Vergnol A, Traoré M, Pietri-Rouxel F, Falcone S. New Insights in CaVβ Subunits: Role in the Regulation of Gene Expression and Cellular Homeostasis. Front Cell Dev Biol 2022; 10:880441. [PMID: 35465309 PMCID: PMC9019481 DOI: 10.3389/fcell.2022.880441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
The voltage-gated calcium channels (CaVs or VGCCs) are fundamental regulators of intracellular calcium homeostasis. When electrical activity induces their activation, the influx of calcium that they mediate or their interaction with intracellular players leads to changes in intracellular Ca2+ levels which regulate many processes such as contraction, secretion and gene expression, depending on the cell type. The essential component of the pore channel is the CaVα1 subunit. However, the fine-tuning of Ca2+-dependent signals is guaranteed by the modulatory role of the auxiliary subunits β, α2δ, and γ of the CaVs. In particular, four different CaVβ proteins (CaVβ1, CaVβ2, CaVβ3, and CaVβ4) are encoded by four different genes in mammalians, each of them displaying several splice variants. Some of these isoforms have been described in regulating CaVα1 docking and stability at the membrane and controlling the channel complex’s conformational changes. In addition, emerging evidences have highlighted other properties of the CaVβ subunits, independently of α1 and non-correlated to its channel or voltage sensing functions. This review summarizes the recent findings reporting novel roles of the auxiliary CaVβ subunits and in particular their direct or indirect implication in regulating gene expression in different cellular contexts.
Collapse
|
11
|
Fong C, Kushner BH, Di Giannatale A, Gundem G, Li S, Roberts SS, Basu EM, Price A, Cheung NKV, Modak S. Skeletal muscle metastases in neuroblastoma share common progenitors with primary tumor and biologically resemble stage MS disease. Front Oncol 2022; 12:1106597. [PMID: 36686814 PMCID: PMC9849794 DOI: 10.3389/fonc.2022.1106597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction While subcutaneous metastases are often observed with stage MS neuroblastoma, an entity that usually resolves spontaneously, skeletal muscle metastases (SMM) have been rarely described. The purpose of this retrospective study was to investigate the significance of SMM in neuroblastoma. Patients and methods Seventeen patients with neuroblastoma SMM were diagnosed at a median age of 4.3 (0.1-15.6) months. All had SMM at diagnosis and metastases at other sites. Fifteen (88%) had ≥ 2 SMM in disparate muscle groups. One, 14, and 2 patients had low, intermediate, and high-risk disease respectively. Fifteen tumors had favorable histology without MYCN amplification, and 2 were MYCN-amplified. Most SMM (80%; n=12/15 evaluated) were MIBG-avid. Results Only 1 patient (with MYCN-non-amplified neuroblastoma) had disease progression. All survive at median follow-up of 47.9 (16.9-318.9) months post-diagnosis. Biological markers (histology, chromosomal and genetic aberrations) were not prognostic. Whole genome sequencing of 3 matched primary and SMM lesions suggested that both primary and metastatic tumors arose from the same progenitor. SMM completely resolved in 10 patients by 12 months post-diagnosis. Of 4 patients managed with watchful observation alone without any cytotoxic therapy, 3 maintain complete remission with SMM resolving by 5, 13, and 21 months post-diagnosis respectively. Conclusions Children with neuroblastoma SMM have an excellent prognosis, with a clinical course suggestive of stage MS disease. Based on these results, the initial management of infants with non-MYCN-amplified NB with SMM could be watchful observation, which could eliminate or reduce exposure to genotoxic therapy.
Collapse
Affiliation(s)
- Christina Fong
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Angela Di Giannatale
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, United States
| | - Gunes Gundem
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Shanita Li
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Anita Price
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
12
|
From Bipotent Neuromesodermal Progenitors to Neural-Mesodermal Interactions during Embryonic Development. Int J Mol Sci 2021; 22:ijms22179141. [PMID: 34502050 PMCID: PMC8431582 DOI: 10.3390/ijms22179141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
To ensure the formation of a properly patterned embryo, multiple processes must operate harmoniously at sequential phases of development. This is implemented by mutual interactions between cells and tissues that together regulate the segregation and specification of cells, their growth and morphogenesis. The formation of the spinal cord and paraxial mesoderm derivatives exquisitely illustrate these processes. Following early gastrulation, while the vertebrate body elongates, a population of bipotent neuromesodermal progenitors resident in the posterior region of the embryo generate both neural and mesodermal lineages. At later stages, the somitic mesoderm regulates aspects of neural patterning and differentiation of both central and peripheral neural progenitors. Reciprocally, neural precursors influence the paraxial mesoderm to regulate somite-derived myogenesis and additional processes by distinct mechanisms. Central to this crosstalk is the activity of the axial notochord, which, via sonic hedgehog signaling, plays pivotal roles in neural, skeletal muscle and cartilage ontogeny. Here, we discuss the cellular and molecular basis underlying this complex developmental plan, with a focus on the logic of sonic hedgehog activities in the coordination of the neural-mesodermal axis.
Collapse
|
13
|
Larouche JA, Mohiuddin M, Choi JJ, Ulintz PJ, Fraczek P, Sabin K, Pitchiaya S, Kurpiers SJ, Castor-Macias J, Liu W, Hastings RL, Brown LA, Markworth JF, De Silva K, Levi B, Merajver SD, Valdez G, Chakkalakal JV, Jang YC, Brooks SV, Aguilar CA. Murine muscle stem cell response to perturbations of the neuromuscular junction are attenuated with aging. eLife 2021; 10:e66749. [PMID: 34323217 PMCID: PMC8360658 DOI: 10.7554/elife.66749] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/28/2021] [Indexed: 01/29/2023] Open
Abstract
During aging and neuromuscular diseases, there is a progressive loss of skeletal muscle volume and function impacting mobility and quality of life. Muscle loss is often associated with denervation and a loss of resident muscle stem cells (satellite cells or MuSCs); however, the relationship between MuSCs and innervation has not been established. Herein, we administered severe neuromuscular trauma to a transgenic murine model that permits MuSC lineage tracing. We show that a subset of MuSCs specifically engraft in a position proximal to the neuromuscular junction (NMJ), the synapse between myofibers and motor neurons, in healthy young adult muscles. In aging and in a mouse model of neuromuscular degeneration (Cu/Zn superoxide dismutase knockout - Sod1-/-), this localized engraftment behavior was reduced. Genetic rescue of motor neurons in Sod1-/- mice reestablished integrity of the NMJ in a manner akin to young muscle and partially restored MuSC ability to engraft into positions proximal to the NMJ. Using single cell RNA-sequencing of MuSCs isolated from aged muscle, we demonstrate that a subset of MuSCs are molecularly distinguishable from MuSCs responding to myofiber injury and share similarity to synaptic myonuclei. Collectively, these data reveal unique features of MuSCs that respond to synaptic perturbations caused by aging and other stressors.
Collapse
Affiliation(s)
- Jacqueline A Larouche
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Mahir Mohiuddin
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaUnited States
- School of Biological Sciences, Georgia Institute of TechnologyAtlantaUnited States
- Wallace Coulter Departmentof Biomedical Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Jeongmoon J Choi
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaUnited States
- School of Biological Sciences, Georgia Institute of TechnologyAtlantaUnited States
- Wallace Coulter Departmentof Biomedical Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Peter J Ulintz
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
- Internal Medicine-Hematology/Oncology, University of MichiganAnn ArborUnited States
| | - Paula Fraczek
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Kaitlyn Sabin
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | | | - Sarah J Kurpiers
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Jesus Castor-Macias
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Wenxuan Liu
- Department of Pharmacology and Physiology, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of Rochester Medical CenterRochesterUnited States
- Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical CenterRochesterUnited States
| | - Robert Louis Hastings
- Departmentof Molecular Biology, Cell Biology and Biochemistry, Brown UniversityProvidenceUnited States
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown UniversityProvidenceUnited States
| | - Lemuel A Brown
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - James F Markworth
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Kanishka De Silva
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
| | - Benjamin Levi
- Department of Surgery, University of Texas SouthwesternDallasUnited States
- Childrens Research Institute and Center for Mineral MetabolismDallasUnited States
- Program in Cellular and Molecular Biology, University of MichiganAnn ArborUnited States
| | - Sofia D Merajver
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Internal Medicine-Hematology/Oncology, University of MichiganAnn ArborUnited States
| | - Gregorio Valdez
- Departmentof Molecular Biology, Cell Biology and Biochemistry, Brown UniversityProvidenceUnited States
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown UniversityProvidenceUnited States
| | - Joe V Chakkalakal
- Department of Pharmacology and Physiology, University of Rochester Medical CenterRochesterUnited States
- Department of Biomedical Engineering, University of Rochester Medical CenterRochesterUnited States
- Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical CenterRochesterUnited States
| | - Young C Jang
- Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaUnited States
- School of Biological Sciences, Georgia Institute of TechnologyAtlantaUnited States
- Wallace Coulter Departmentof Biomedical Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Susan V Brooks
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Department of Molecular & Integrative Physiology, University of MichiganAnn ArborUnited States
| | - Carlos A Aguilar
- Department of Biomedical Engineering, University of MichiganAnn ArborUnited States
- Biointerfaces Institute, University of MichiganAnn ArborUnited States
- Childrens Research Institute and Center for Mineral MetabolismDallasUnited States
- Program in Cellular and Molecular Biology, University of MichiganAnn ArborUnited States
| |
Collapse
|
14
|
Le TL, Galmiche L, Levy J, Suwannarat P, Hellebrekers DM, Morarach K, Boismoreau F, Theunissen TE, Lefebvre M, Pelet A, Martinovic J, Gelot A, Guimiot F, Calleroz A, Gitiaux C, Hully M, Goulet O, Chardot C, Drunat S, Capri Y, Bole-Feysot C, Nitschké P, Whalen S, Mouthon L, Babcock HE, Hofstra R, de Coo IF, Tabet AC, Molina TJ, Keren B, Brooks A, Smeets HJ, Marklund U, Gordon CT, Lyonnet S, Amiel J, Bondurand N. Dysregulation of the NRG1/ERBB pathway causes a developmental disorder with gastrointestinal dysmotility in humans. J Clin Invest 2021; 131:145837. [PMID: 33497358 DOI: 10.1172/jci145837] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/14/2021] [Indexed: 02/06/2023] Open
Abstract
Hirschsprung disease (HSCR) is the most frequent developmental anomaly of the enteric nervous system, with an incidence of 1 in 5000 live births. Chronic intestinal pseudo-obstruction (CIPO) is less frequent and classified as neurogenic or myogenic. Isolated HSCR has an oligogenic inheritance with RET as the major disease-causing gene, while CIPO is genetically heterogeneous, caused by mutations in smooth muscle-specific genes. Here, we describe a series of patients with developmental disorders including gastrointestinal dysmotility, and investigate the underlying molecular bases. Trio-exome sequencing led to the identification of biallelic variants in ERBB3 and ERBB2 in 8 individuals variably associating HSCR, CIPO, peripheral neuropathy, and arthrogryposis. Thorough gut histology revealed aganglionosis, hypoganglionosis, and intestinal smooth muscle abnormalities. The cell type-specific ErbB3 and ErbB2 function was further analyzed in mouse single-cell RNA sequencing data and in a conditional ErbB3-deficient mouse model, revealing a primary role for ERBB3 in enteric progenitors. The consequences of the identified variants were evaluated using quantitative real-time PCR (RT-qPCR) on patient-derived fibroblasts or immunoblot assays on Neuro-2a cells overexpressing WT or mutant proteins, revealing either decreased expression or altered phosphorylation of the mutant receptors. Our results demonstrate that dysregulation of ERBB3 or ERBB2 leads to a broad spectrum of developmental anomalies, including intestinal dysmotility.
Collapse
Affiliation(s)
- Thuy-Linh Le
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Louise Galmiche
- INSERM UMR 1235, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, University of Nantes, Nantes, France.,Pathology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Necker-Enfants Malades Hospital, Paris, France
| | - Jonathan Levy
- Genetics Department, Robert Debré Hospital, AP-HP, Paris, France.,Université de Paris, NeuroDiderot, INSERM UMR 1141, Paris, France
| | - Pim Suwannarat
- Department of Genetics, Mid-Atlantic Permanente Medical Group, Suitland, Maryland, USA
| | - Debby Mei Hellebrekers
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, Netherlands
| | - Khomgrit Morarach
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Franck Boismoreau
- Institut de Biologie de l'ENS (IBENS), INSERM, CNRS, École Normale Supérieure, PSL Research University, Paris, France
| | - Tom Ej Theunissen
- Department of Genetics and Cell Biology, Maastricht University, Maastricht, Netherlands
| | - Mathilde Lefebvre
- Fetal Pathology Unit, Armand Trousseau Hospital, AP-HP, Paris, France
| | - Anna Pelet
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Jelena Martinovic
- Fetal Pathology Unit, Antoine Béclère Hospital, AP-HP, Paris-Saclay University, Clamart, France
| | - Antoinette Gelot
- Neuropathology, Pathology Department, Armand Trousseau Hospital, AP-HP, Paris, France.,Aix-Marseille University, INMED INSERM UMR1249, Campus de Luminy, Marseille, France
| | - Fabien Guimiot
- Université de Paris, NeuroDiderot, INSERM UMR 1141, Paris, France.,Fetal Pathology Unit, Robert Debré Hospital, AP-HP, Paris, France
| | - Amanda Calleroz
- Pathology and Laboratory Medicine Division, Children's National Hospital, Washington DC, USA
| | - Cyril Gitiaux
- Department of Pediatric Clinical Neurophysiology, Necker-Enfants Malades Hospital, AP-HP, Université de Paris, Paris, France
| | - Marie Hully
- Department of Pediatric Neurology and Rehabilitation, Necker-Enfants Malades Hospital, AP-HP, Université de Paris, Paris, France
| | - Olivier Goulet
- Department of Pediatric Gastroenterology-Hepatology-Nutrition, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Christophe Chardot
- Department of Pediatric Surgery, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Severine Drunat
- Genetics Department, Robert Debré Hospital, AP-HP, Paris, France.,Université de Paris, NeuroDiderot, INSERM UMR 1141, Paris, France
| | - Yline Capri
- Genetics Department, Robert Debré Hospital, AP-HP, Paris, France
| | - Christine Bole-Feysot
- Genomics Core Facility, Imagine Institute-Structure Federative de Recherche Necker, INSERM UMR 1163 and INSERM US24/CNRS UMS 3633, Université de Paris, Paris, France
| | | | - Sandra Whalen
- Clinical Genetics Unit and Reference Center, Anomalies du Développement et Syndromes Malformatifs, AP-HP, Sorbonne University, Armand Trousseau Hospital, Paris, France
| | - Linda Mouthon
- Department of Genetics, La Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Holly E Babcock
- Children's National Hospital, Rare Disease Institute, Washington, DC, USA
| | - Robert Hofstra
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Irenaeus Fm de Coo
- Department of Toxicogenomics, Unit Clinical Genomics, Maastricht University, MHeNs School for Mental Health and Neuroscience, Maastricht, Netherlands
| | - Anne-Claude Tabet
- Genetics Department, Robert Debré Hospital, AP-HP, Paris, France.,Human Genetics and Cognitive Functions, Institut Pasteur, UMR3571 CNRS, Université de Paris, Paris, France
| | - Thierry J Molina
- Pathology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Necker-Enfants Malades Hospital, Paris, France.,Université de Paris, Imagine Institute, Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM UMR 1163, Paris, France
| | - Boris Keren
- Department of Genetics, La Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Alice Brooks
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| | - Hubert Jm Smeets
- Department of Toxicogenomics, Unit Clinical Genomics, Maastricht University, MHeNs School for Mental Health and Neuroscience, Maastricht, Netherlands
| | - Ulrika Marklund
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Christopher T Gordon
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Stanislas Lyonnet
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France.,Fédération de Génétique, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Jeanne Amiel
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France.,Fédération de Génétique, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Nadège Bondurand
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| |
Collapse
|
15
|
Figliolini F, Ranghino A, Grange C, Cedrino M, Tapparo M, Cavallari C, Rossi A, Togliatto G, Femminò S, Gugliuzza MV, Camussi G, Brizzi MF. Extracellular Vesicles From Adipose Stem Cells Prevent Muscle Damage and Inflammation in a Mouse Model of Hind Limb Ischemia: Role of Neuregulin-1. Arterioscler Thromb Vasc Biol 2019; 40:239-254. [PMID: 31665908 DOI: 10.1161/atvbaha.119.313506] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Critical hindlimb ischemia is a severe consequence of peripheral artery disease. Surgical treatment does not prevent skeletal muscle impairment or improve long-term patient outcomes. The present study investigates the protective/regenerative potential and the mechanism of action of adipose stem cell-derived extracellular vesicles (ASC-EVs) in a mouse model of hindlimb ischemia. Approach and Results: We demonstrated that ASC-EVs exert a protective effect on muscle damage by acting both on tissue microvessels and muscle cells. The genes involved in muscle regeneration were up-regulated in the ischemic muscles of ASC-EV-treated animals. MyoD expression has also been confirmed in satellite cells. This was followed by a reduction in muscle function impairment in vivo. ASC-EVs drive myoblast proliferation and differentiation in the in vitro ischemia/reoxygenation model. Moreover, ASC-EVs have shown an anti-apoptotic effect both in vitro and in vivo. Transcriptomic analyses have revealed that ASC-EVs carry a variety of pro-angiogenic mRNAs, while proteomic analyses have demonstrated an enrichment of NRG1 (neuregulin 1). A NRG1 blocking antibody used in vivo demonstrated that NRG1 is relevant to ASC-EV-induced muscle protection, vascular growth, and recruitment of inflammatory cells. Finally, bioinformatic analyses on 18 molecules that were commonly detected in ASC-EVs, including mRNAs and proteins, confirmed the enrichment of pathways involved in vascular growth and muscle regeneration/protection. CONCLUSIONS This study demonstrates that ASC-EVs display pro-angiogenic and skeletal muscle protective properties that are associated with their NRG1/mRNA cargo. We, therefore, propose that ASC-EVs are a useful tool for therapeutic angiogenesis and muscle protection.
Collapse
Affiliation(s)
- Federico Figliolini
- From the 2i3T Scarl University of Turin (F.F., M.C., C.C.), University of Turin, Italy
| | - Andrea Ranghino
- Department of Medical Sciences (A. Ranghino, C.G., M.T., A. Rossi, G.T., S.F., M.V.G., G.C., M.F.B.), University of Turin, Italy
| | - Cristina Grange
- Department of Medical Sciences (A. Ranghino, C.G., M.T., A. Rossi, G.T., S.F., M.V.G., G.C., M.F.B.), University of Turin, Italy
| | - Massimo Cedrino
- From the 2i3T Scarl University of Turin (F.F., M.C., C.C.), University of Turin, Italy
| | - Marta Tapparo
- Department of Medical Sciences (A. Ranghino, C.G., M.T., A. Rossi, G.T., S.F., M.V.G., G.C., M.F.B.), University of Turin, Italy
| | - Claudia Cavallari
- From the 2i3T Scarl University of Turin (F.F., M.C., C.C.), University of Turin, Italy
| | - Andrea Rossi
- Department of Medical Sciences (A. Ranghino, C.G., M.T., A. Rossi, G.T., S.F., M.V.G., G.C., M.F.B.), University of Turin, Italy
| | - Gabriele Togliatto
- Department of Medical Sciences (A. Ranghino, C.G., M.T., A. Rossi, G.T., S.F., M.V.G., G.C., M.F.B.), University of Turin, Italy
| | - Saveria Femminò
- Department of Medical Sciences (A. Ranghino, C.G., M.T., A. Rossi, G.T., S.F., M.V.G., G.C., M.F.B.), University of Turin, Italy
| | - Maria Vittoria Gugliuzza
- Department of Medical Sciences (A. Ranghino, C.G., M.T., A. Rossi, G.T., S.F., M.V.G., G.C., M.F.B.), University of Turin, Italy
| | - Giovanni Camussi
- Department of Medical Sciences (A. Ranghino, C.G., M.T., A. Rossi, G.T., S.F., M.V.G., G.C., M.F.B.), University of Turin, Italy
| | - Maria Felice Brizzi
- Department of Medical Sciences (A. Ranghino, C.G., M.T., A. Rossi, G.T., S.F., M.V.G., G.C., M.F.B.), University of Turin, Italy
| |
Collapse
|
16
|
|
17
|
Abstract
Neural crest cells are the embryonic precursors of most neurons and all glia of the peripheral nervous system, pigment cells, some endocrine components, and connective tissue of the head, face, neck, and heart. Following induction, crest cells undergo an epithelial to mesenchymal transition that enables them to migrate along specific pathways culminating in their phenotypic differentiation. Researching this unique embryonic population has revealed important understandings of basic biological and developmental principles. These principles are likely to assist in clarifying the etiology and help in finding strategies for the treatment of neural crest diseases, collectively termed neurocristopathies. The progress achieved in neural crest research is made feasible thanks to the continuous development of species-specific in vivo and in vitro paradigms and more recently the possibility to produce neural crest cells and specific derivatives from embryonic or induced pluripotent stem cells. All of the above assist us in elucidating mechanisms that regulate neural crest development using state-of-the art cellular, molecular, and imaging approaches.
Collapse
Affiliation(s)
- Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel.
- Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
18
|
Magli A, Perlingeiro RRC. Myogenic progenitor specification from pluripotent stem cells. Semin Cell Dev Biol 2018; 72:87-98. [PMID: 29107681 DOI: 10.1016/j.semcdb.2017.10.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 12/21/2022]
Abstract
Pluripotent stem cells represent important tools for both basic and translational science as they enable to study mechanisms of development, model diseases in vitro and provide a potential source of tissue-specific progenitors for cell therapy. Concomitantly with the increasing knowledge of the molecular mechanisms behind activation of the skeletal myogenic program during embryonic development, novel findings in the stem cell field provided the opportunity to begin recapitulating in vitro the events occurring during specification of the myogenic lineage. In this review, we will provide a perspective of the molecular mechanisms responsible for skeletal myogenic commitment in the embryo and how this knowledge was instrumental for specifying this lineage from pluripotent stem cells. In addition, we will discuss the current limitations for properly recapitulating skeletal myogenesis in the petri dish, and we will provide insights about future applications of pluripotent stem cell-derived myogenic cells.
Collapse
Affiliation(s)
- Alessandro Magli
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rita R C Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
19
|
Chang CN, Kioussi C. Location, Location, Location: Signals in Muscle Specification. J Dev Biol 2018; 6:E11. [PMID: 29783715 PMCID: PMC6027348 DOI: 10.3390/jdb6020011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 12/15/2022] Open
Abstract
Muscles control body movement and locomotion, posture and body position and soft tissue support. Mesoderm derived cells gives rise to 700 unique muscles in humans as a result of well-orchestrated signaling and transcriptional networks in specific time and space. Although the anatomical structure of skeletal muscles is similar, their functions and locations are specialized. This is the result of specific signaling as the embryo grows and cells migrate to form different structures and organs. As cells progress to their next state, they suppress current sequence specific transcription factors (SSTF) and construct new networks to establish new myogenic features. In this review, we provide an overview of signaling pathways and gene regulatory networks during formation of the craniofacial, cardiac, vascular, trunk, and limb skeletal muscles.
Collapse
Affiliation(s)
- Chih-Ning Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
- Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA.
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
- Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
20
|
Hicks MR, Hiserodt J, Paras K, Fujiwara W, Eskin A, Jan M, Xi H, Young CS, Evseenko D, Nelson SF, Spencer MJ, Handel BV, Pyle AD. ERBB3 and NGFR mark a distinct skeletal muscle progenitor cell in human development and hPSCs. Nat Cell Biol 2018; 20:46-57. [PMID: 29255171 PMCID: PMC5962356 DOI: 10.1038/s41556-017-0010-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/16/2017] [Indexed: 12/24/2022]
Abstract
Human pluripotent stem cells (hPSCs) can be directed to differentiate into skeletal muscle progenitor cells (SMPCs). However, the myogenicity of hPSC-SMPCs relative to human fetal or adult satellite cells remains unclear. We observed that hPSC-SMPCs derived by directed differentiation are less functional in vitro and in vivo compared to human satellite cells. Using RNA sequencing, we found that the cell surface receptors ERBB3 and NGFR demarcate myogenic populations, including PAX7 progenitors in human fetal development and hPSC-SMPCs. We demonstrated that hPSC skeletal muscle is immature, but inhibition of transforming growth factor-β signalling during differentiation improved fusion efficiency, ultrastructural organization and the expression of adult myosins. This enrichment and maturation strategy restored dystrophin in hundreds of dystrophin-deficient myofibres after engraftment of CRISPR-Cas9-corrected Duchenne muscular dystrophy human induced pluripotent stem cell-SMPCs. The work provides an in-depth characterization of human myogenesis, and identifies candidates that improve the in vivo myogenic potential of hPSC-SMPCs to levels that are equal to directly isolated human fetal muscle cells.
Collapse
MESH Headings
- Adult
- Aged
- CRISPR-Cas Systems
- Cell Differentiation
- Dystrophin/genetics
- Dystrophin/metabolism
- Female
- Gene Editing
- Gene Expression Regulation, Developmental
- Humans
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/metabolism
- Male
- Middle Aged
- Muscle Development/genetics
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/therapy
- Myoblasts/cytology
- Myoblasts/metabolism
- Myosins/genetics
- Myosins/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/metabolism
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Signal Transduction
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Michael R Hicks
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Julia Hiserodt
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Katrina Paras
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Wakana Fujiwara
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Ascia Eskin
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Majib Jan
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Haibin Xi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Courtney S Young
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stanley F Nelson
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Melissa J Spencer
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
- Department of Neurology, University of California, Los Angeles, CA, USA
| | - Ben Van Handel
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - April D Pyle
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA.
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA, USA.
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA.
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA.
| |
Collapse
|
21
|
Sato S, Furuta Y, Kawakami K. Regulation of continuous but complex expression pattern of Six1 during early sensory development. Dev Dyn 2017; 247:250-261. [PMID: 29106072 DOI: 10.1002/dvdy.24603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/17/2017] [Accepted: 10/31/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In vertebrates, cranial sensory placodes give rise to neurosensory and endocrine structures, such as the olfactory epithelium, inner ear, and anterior pituitary. We report here the establishment of a transgenic mouse line that expresses Cre recombinase under the control of Six1-21, a major placodal enhancer of the homeobox gene Six1. RESULTS In the new Cre-expressing line, mSix1-21-NLSCre, the earliest Cre-mediated recombination was induced at embryonic day 8.5 in the region overlapping with the otic-epibranchial progenitor domain (OEPD), a transient, common precursor domain for the otic and epibranchial placodes. Recombination was later observed in the OEPD-derived structures (the entire inner ear and the VIIth-Xth cranial sensory ganglia), olfactory epithelium, anterior pituitary, pharyngeal ectoderm and pouches. Other Six1-positive structures, such as salivary/lacrimal glands and limb buds, were also positive for recombination. Moreover, comparison with another mouse line expressing Cre under the control of the sensory neuron enhancer, Six1-8, indicated that the continuous and complex expression pattern of Six1 during sensory organ formation is pieced together by separate enhancers. CONCLUSIONS mSix1-21-NLSCre has several unique characteristics to make it suitable for analysis of cell lineage and gene function in sensory placodes as well as nonplacodal Six1-positive structures. Developmental Dynamics 247:250-261, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shigeru Sato
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yasuhide Furuta
- Animal Resource Development Unit and Genetic Engineering Team, Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), Kobe, Hyogo, Japan
| | - Kiyoshi Kawakami
- Division of Biology, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
22
|
Deries M, Thorsteinsdóttir S. Axial and limb muscle development: dialogue with the neighbourhood. Cell Mol Life Sci 2016; 73:4415-4431. [PMID: 27344602 PMCID: PMC11108464 DOI: 10.1007/s00018-016-2298-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/03/2016] [Accepted: 06/21/2016] [Indexed: 11/29/2022]
Abstract
Skeletal muscles are part of the musculoskeletal system which also includes nerves, tendons, connective tissue, bones and blood vessels. Here we review the development of axial and limb muscles in amniotes within the context of their surrounding tissues in vivo. We highlight the reciprocal dialogue mediated by signalling factors between cells of these adjacent tissues and developing muscles and also demonstrate its importance from the onset of muscle cell differentiation well into foetal development. Early embryonic tissues secrete factors which are important regulators of myogenesis. However, later muscle development relies on other tissue collaborators, such as developing nerves and connective tissue, which are in turn influenced by the developing muscles themselves. We conclude that skeletal muscle development in vivo is a compelling example of the importance of reciprocal interactions between developing tissues for the complete and coordinated development of a functional system.
Collapse
Affiliation(s)
- Marianne Deries
- Centro de Ecologia, Evolução e Alterações Ambientais, Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal.
| | - Sólveig Thorsteinsdóttir
- Centro de Ecologia, Evolução e Alterações Ambientais, Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
23
|
Hatakeyama J, Wald JH, Rafidi H, Cuevas A, Sweeney C, Carraway KL. The ER structural protein Rtn4A stabilizes and enhances signaling through the receptor tyrosine kinase ErbB3. Sci Signal 2016; 9:ra65. [PMID: 27353365 DOI: 10.1126/scisignal.aaf1604] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ErbB3 and ErbB4 are receptor tyrosine kinases that are activated by the neuregulin (NRG) family of growth factors. These receptors govern various developmental processes, and their dysregulation contributes to several human disease states. The abundance of ErbB3 and ErbB4, and thus signaling through these receptors, is limited by the E3 ubiquitin ligase Nrdp1, which targets ErbB3 and ErbB4 for degradation. Reticulons are proteins that influence the morphology of the endoplasmic reticulum (ER) by promoting the formation of tubules, a response of cells to some stressors. We found that the ER structural protein reticulon 4A (Rtn4A, also known as Nogo-A) increased ErbB3 abundance and proliferative signaling by suppressing Nrdp1 function. Rtn4A interacted with Nrdp1 and stabilized ErbB3 in an Nrdp1-dependent manner. Rtn4A overexpression induced the redistribution of Nrdp1 from a cytosolic or perinuclear localization to ER tubules. Rtn4A knockdown in human breast tumor cells decreased ErbB3 abundance, NRG-stimulated signaling, and cellular proliferation and migration. Because proteins destined for the plasma membrane are primarily synthesized in the sheet portions of the ER, our observations suggest that Rtn4A counteracts the Nrdp1-mediated degradation of ErbB3 by sequestering the ubiquitin ligase into ER tubules. The involvement of a reticulon suggests a molecular link between ER structure and the sensitivity of cells to receptor tyrosine kinase-mediated survival signals at the cell surface.
Collapse
Affiliation(s)
- Jason Hatakeyama
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jessica H Wald
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Hanine Rafidi
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Antonio Cuevas
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Colleen Sweeney
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Kermit L Carraway
- Department of Biochemistry and Molecular Medicine, and UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
24
|
Applications of Mesenchymal Stem Cells and Neural Crest Cells in Craniofacial Skeletal Research. Stem Cells Int 2016; 2016:2849879. [PMID: 27006661 PMCID: PMC4783549 DOI: 10.1155/2016/2849879] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/02/2016] [Indexed: 12/28/2022] Open
Abstract
Craniofacial skeletal tissues are composed of tooth and bone, together with nerves and blood vessels. This composite material is mainly derived from neural crest cells (NCCs). The neural crest is transient embryonic tissue present during neural tube formation whose cells have high potential for migration and differentiation. Thus, NCCs are promising candidates for craniofacial tissue regeneration; however, the clinical application of NCCs is hindered by their limited accessibility. In contrast, mesenchymal stem cells (MSCs) are easily accessible in adults, have similar potential for self-renewal, and can differentiate into skeletal tissues, including bones and cartilage. Therefore, MSCs may represent good sources of stem cells for clinical use. MSCs are classically identified under adherent culture conditions, leading to contamination with other cell lineages. Previous studies have identified mouse- and human-specific MSC subsets using cell surface markers. Additionally, some studies have shown that a subset of MSCs is closely related to neural crest derivatives and endothelial cells. These MSCs may be promising candidates for regeneration of craniofacial tissues from the perspective of developmental fate. Here, we review the fundamental biology of MSCs in craniofacial research.
Collapse
|
25
|
Adameyko I, Fried K. The Nervous System Orchestrates and Integrates Craniofacial Development: A Review. Front Physiol 2016; 7:49. [PMID: 26924989 PMCID: PMC4759458 DOI: 10.3389/fphys.2016.00049] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/02/2016] [Indexed: 01/14/2023] Open
Abstract
Development of a head is a dazzlingly complex process: a number of distinct cellular sources including cranial ecto- and endoderm, mesoderm and neural crest contribute to facial and other structures. In the head, an extremely fine-tuned developmental coordination of CNS, peripheral neural components, sensory organs and a musculo-skeletal apparatus occurs, which provides protection and functional integration. The face can to a large extent be considered as an assembly of sensory systems encased and functionally fused with appendages represented by jaws. Here we review how the developing brain, neurogenic placodes and peripheral nerves influence the morphogenesis of surrounding tissues as a part of various general integrative processes in the head. The mechanisms of this impact, as we understand it now, span from the targeted release of the morphogens necessary for shaping to providing a niche for cellular sources required in later development. In this review we also discuss the most recent findings and ideas related to how peripheral nerves and nerve-associated cells contribute to craniofacial development, including teeth, during the post- neural crest period and potentially in regeneration.
Collapse
Affiliation(s)
- Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska InstitutetStockholm, Sweden; Department of Molecular Neurosciences, Center of Brain Research, Medical University of ViennaVienna, Austria
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| |
Collapse
|
26
|
Rapid and simple method for in vivo ex utero development of mouse embryo explants. Differentiation 2016; 91:57-67. [PMID: 26897458 DOI: 10.1016/j.diff.2015.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/04/2015] [Indexed: 01/17/2023]
Abstract
The in utero development of mammals drastically reduces the accessibility of the mammalian embryo and therefore limits the range of experimental manipulation that can be done to study functions of genes or signaling pathways during embryo development. Over the past decades, tissue and organ-like culture methods have been developed with the intention of reproducing in vivo situations. Developing accessible and simple techniques to study and manipulate embryos is an everlasting challenge. Herein, we describe a reliable and quick technique to culture mid-gestation explanted mouse embryos on top of a floating membrane filter in a defined medium. Viability of the cultured tissues was assessed by apoptosis and proliferation analysis showing that cell proliferation is normal and there is only a slight increase in apoptosis after 12h of culture compared to embryos developing in utero. Moreover, differentiation and morphogenesis proceed normally as assessed by 3D imaging of the transformation of the myotome into deep back muscles. Not only does muscle cell differentiation occur as expected, but so do extracellular matrix organization and the characteristic splitting of the myotome into the three epaxial muscle groups. Our culture method allows for the culture and manipulation of mammalian embryo explants in a very efficient way, and it permits the manipulation of in vivo developmental events in a controlled environment. Explants grown under these ex utero conditions simulate real developmental events that occur in utero.
Collapse
|
27
|
Endo T. Molecular mechanisms of skeletal muscle development, regeneration, and osteogenic conversion. Bone 2015; 80:2-13. [PMID: 26453493 DOI: 10.1016/j.bone.2015.02.028] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 02/18/2015] [Accepted: 02/28/2015] [Indexed: 12/21/2022]
Abstract
Both skeletal muscle and bone are of mesodermal origin and derived from somites during embryonic development. Somites differentiate into the dorsal dermomyotome and the ventral sclerotome, which give rise to skeletal muscle and bone, respectively. Extracellular signaling molecules, such as Wnt and Shh, secreted from the surrounding environment, determine the developmental fate of skeletal muscle. Dermomyotome cells are specified as trunk muscle progenitor cells by transcription factor networks involving Pax3. These progenitor cells delaminate and migrate to form the myotome, where they are determined as myoblasts that differentiate into myotubes or myofibers. The MyoD family of transcription factors plays pivotal roles in myogenic determination and differentiation. Adult skeletal muscle regenerates upon exercise, muscle injury, or degeneration. Satellite cells are muscle-resident stem cells and play essential roles in muscle growth and regeneration. Muscle regeneration recapitulates the process of muscle development in many aspects. In certain muscle diseases, ectopic calcification or heterotopic ossification, as well as fibrosis and adipogenesis, occurs in skeletal muscle. Muscle-resident mesenchymal progenitor cells, which may be derived from vascular endothelial cells, are responsible for the ectopic osteogenesis, fibrogenesis, and adipogenesis. The small GTPase M-Ras is likely to participate in the ectopic calcification and ossification, as well as in osteogenesis during development. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
28
|
Buckingham M, Relaix F. PAX3 and PAX7 as upstream regulators of myogenesis. Semin Cell Dev Biol 2015; 44:115-25. [PMID: 26424495 DOI: 10.1016/j.semcdb.2015.09.017] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/23/2015] [Indexed: 10/23/2022]
Abstract
Like other subclasses within the PAX transcription factor family, PAX3 and PAX7 play important roles in the emergence of a number of different tissues during development. PAX3 regulates neural crest and, together with its orthologue PAX7, is also expressed in parts of the central nervous system. In this chapter we will focus on their role in skeletal muscle. Both factors are key regulators of myogenesis where Pax3 plays a major role during early skeletal muscle formation in the embryo while Pax7 predominates during post-natal growth and muscle regeneration in the adult. We review the expression and functions of these factors in the myogenic context. We also discuss mechanistic aspects of PAX3/7 function and modulation of their activity by interaction with other proteins, as well as the post-transcriptional and transcriptional regulation of their expression.
Collapse
Affiliation(s)
- Margaret Buckingham
- Department of Developmental and Stem Cell Biology, CNRS URA 2578, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France.
| | - Frédéric Relaix
- INSERM U955 IMRB, Team 10, 94000 Creteil, France; UPEC Paris Est-Creteil University, Faculty of Medicine, F-94000 Creteil, France; Etablissement Français du Sang, 94017 Creteil, France; Université Paris Est, Ecole Nationale Veterinaire d'Alfort, 94700 Maison Alfort, France.
| |
Collapse
|
29
|
Abstract
Skeletal muscle development has been the focus of intensive study for many decades. Recent advances in genetic manipulation of the mouse have increased our understanding of the cell signalling involved in the development of muscle progenitors which give rise to adult skeletal muscles and their stem cell populations. However, the influence of a vital tissue type – the peripheral nerve—has largely been ignored since its earliest descriptions. Here we carefully describe the timing in which myogenic progenitors expressing Pax3 and Pax7 (the earliest markers of myogenic cells) enter the limb buds of rat and mouse embryos, as well as the spatiotemporal relationship between these progenitors and the ingrowing peripheral nerve. We show that progenitors expressing Pax3 enter the limb bud one full day ahead of the first neurites and that Pax7-expressing progenitors (associated with secondary myogenesis in the limb) are first seen in the limb bud at the time of nerve entry and in close proximity to the nerve. The initial entry of the nerve also coincides with the first expression of myosin heavy chain showing that the first contact between nerves and myogenic cells correlates with the onset of myogenic differentiation. Furthermore, as the nerve grows into the limb, Pax3 expression is progressively replaced by Pax7 expression in myogenic progenitors. These findings indicate that the ingrowing nerve enters the limb presumptive muscle masses earlier than what was generally described and raises the possibility that nerve may influence the differentiation of muscle progenitors in rodent limbs.
Collapse
|
30
|
Klhl31 attenuates β-catenin dependent Wnt signaling and regulates embryo myogenesis. Dev Biol 2015; 402:61-71. [DOI: 10.1016/j.ydbio.2015.02.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 11/19/2022]
|
31
|
Burger NB, Bekker MN, de Groot CJM, Christoffels VM, Haak MC. Why increased nuchal translucency is associated with congenital heart disease: a systematic review on genetic mechanisms. Prenat Diagn 2015; 35:517-28. [DOI: 10.1002/pd.4586] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/09/2014] [Accepted: 02/21/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Nicole B. Burger
- Department of Obstetrics and Gynaecology; VU University Medical Center; Amsterdam The Netherlands
| | - Mireille N. Bekker
- Department of Obstetrics and Gynaecology; Radboud University Medical Center; Nijmegen The Netherlands
| | | | - Vincent M. Christoffels
- Department of Anatomy, Embryology & Physiology; Academic Medical Center; Amsterdam The Netherlands
| | - Monique C. Haak
- Department of Obstetrics; Leiden University Medical Center; Leiden The Netherlands
| |
Collapse
|
32
|
C-Nap1 mutation affects centriole cohesion and is associated with a Seckel-like syndrome in cattle. Nat Commun 2015; 6:6894. [PMID: 25902731 PMCID: PMC4423223 DOI: 10.1038/ncomms7894] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 03/11/2015] [Indexed: 12/11/2022] Open
Abstract
Caprine-like Generalized Hypoplasia Syndrome (SHGC) is an autosomal-recessive disorder in Montbéliarde cattle. Affected animals present a wide range of clinical features that include the following: delayed development with low birth weight, hind limb muscular hypoplasia, caprine-like thin head and partial coat depigmentation. Here we show that SHGC is caused by a truncating mutation in the CEP250 gene that encodes the centrosomal protein C-Nap1. This mutation results in centrosome splitting, which neither affects centriole ultrastructure and duplication in dividing cells nor centriole function in cilium assembly and mitotic spindle organization. Loss of C-Nap1-mediated centriole cohesion leads to an altered cell migration phenotype. This discovery extends the range of loci that constitute the spectrum of autosomal primary recessive microcephaly (MCPH) and Seckel-like syndromes.
Collapse
|
33
|
Zalc A, Rattenbach R, Auradé F, Cadot B, Relaix F. Pax3 and Pax7 play essential safeguard functions against environmental stress-induced birth defects. Dev Cell 2015; 33:56-66. [PMID: 25800090 DOI: 10.1016/j.devcel.2015.02.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 12/11/2014] [Accepted: 02/08/2015] [Indexed: 12/23/2022]
Abstract
Exposure to environmental teratogenic pollutant leads to severe birth defects. However, the biological events underlying these developmental abnormalities remain undefined. Here, we report a molecular link between an environmental stress response pathway and key developmental genes during craniofacial development. Strikingly, mutant mice with impaired Pax3/7 function display severe craniofacial defects. We show that these are associated with an upregulation of the signaling pathway mediated by the Aryl hydrocarbon receptor (AHR), the receptor to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), revealing a genetic interaction between Pax3 and AHR signaling. Activation of AHR signaling in Pax3-deficient embryos drives facial mesenchymal cells out of the cell cycle through the upregulation of p21 expression. Accordingly, inhibiting AHR activity rescues the cycling status of these cells and the facial closure of Pax3/7 mutants. Together, our findings demonstrate that the regulation of AHR signaling by Pax3/7 is required to protect against TCDD/AHR-mediated teratogenesis during craniofacial development.
Collapse
Affiliation(s)
- Antoine Zalc
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France; INSERM U955 IMRB, Team 10, 94000 Creteil, France; UPEC Paris Est-Creteil University, Faculty of Medicine, 94000 Creteil, France
| | - Revital Rattenbach
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France; INSERM U955 IMRB, Team 10, 94000 Creteil, France; UPEC Paris Est-Creteil University, Faculty of Medicine, 94000 Creteil, France
| | - Frédéric Auradé
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France
| | - Bruno Cadot
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France
| | - Frédéric Relaix
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center, INSERM U974, CNRS FRE 3617, Institut de Myologie, 75013 Paris, France; INSERM U955 IMRB, Team 10, 94000 Creteil, France; UPEC Paris Est-Creteil University, Faculty of Medicine, 94000 Creteil, France; Etablissement Français du Sang, 94017 Creteil, France; Université Paris Est, Ecole Nationale Veterinaire d'Alfort, 94700 Maison-Alfort, France.
| |
Collapse
|
34
|
Wang G, Li Y, Wang XY, Chuai M, Yeuk-Hon Chan J, Lei J, Münsterberg A, Lee KKH, Yang X. Misexpression of BRE gene in the developing chick neural tube affects neurulation and somitogenesis. Mol Biol Cell 2015; 26:978-92. [PMID: 25568339 PMCID: PMC4342032 DOI: 10.1091/mbc.e14-06-1144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
This is the first study of the role of BRE in embryonic development using early chick embryos. BRE is expressed in the developing neural tube, neural crest cells, and somites. BRE thus plays an important role in regulating neurogenesis and indirectly somitogenesis during early chick embryo development. The brain and reproductive expression (BRE) gene is expressed in numerous adult tissues and especially in the nervous and reproductive systems. However, little is known about BRE expression in the developing embryo or about its role in embryonic development. In this study, we used in situ hybridization to reveal the spatiotemporal expression pattern for BRE in chick embryo during development. To determine the importance of BRE in neurogenesis, we overexpressed BRE and also silenced BRE expression specifically in the neural tube. We established that overexpressing BRE in the neural tube indirectly accelerated Pax7+ somite development and directly increased HNK-1+ neural crest cell (NCC) migration and TuJ-1+ neurite outgrowth. These altered morphogenetic processes were associated with changes in the cell cycle of NCCs and neural tube cells. The inverse effect was obtained when BRE expression was silenced in the neural tube. We also determined that BMP4 and Shh expression in the neural tube was affected by misexpression of BRE. This provides a possible mechanism for how altering BRE expression was able to affect somitogenesis, neurogenesis, and NCC migration. In summary, our results demonstrate that BRE plays an important role in regulating neurogenesis and indirectly somite differentiation during early chick embryo development.
Collapse
Affiliation(s)
- Guang Wang
- Department of Histology and Embryology, School of Medicine, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Yan Li
- Department of Histology and Embryology, School of Medicine, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Xiao-Yu Wang
- Department of Histology and Embryology, School of Medicine, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Manli Chuai
- Division of Cell and Developmental Biology, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - John Yeuk-Hon Chan
- Department of Histology and Embryology, School of Medicine, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Jian Lei
- Department of Histology and Embryology, School of Medicine, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Andrea Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - Kenneth Ka Ho Lee
- Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xuesong Yang
- Department of Histology and Embryology, School of Medicine, Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| |
Collapse
|
35
|
Applebaum M, Kalcheim C. Mechanisms of myogenic specification and patterning. Results Probl Cell Differ 2015; 56:77-98. [PMID: 25344667 DOI: 10.1007/978-3-662-44608-9_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesodermal somites are initially composed of columnar cells arranged as a pseudostratified epithelium that undergoes sequential and spatially restricted changes to generate the sclerotome and dermomyotome, intermediate structures that develop into vertebrae, striated muscles of the body and limbs, dermis, smooth muscle, and endothelial cells. Regional cues were elucidated that impart differential traits upon the originally multipotent progenitors. How do somite cells and their intermediate progenitors interpret these extrinsic cues and translate them into various levels and/or modalities of intracellular signaling that lead to differential gene expression profiles remains a significant challenge. So is the understanding of how differential fate specification relates to complex cellular migrations prefiguring the formation of body muscles and vertebrae. Research in the past years has largely transited from a descriptive phase in which the lineages of distinct somite-derived progenitors and their cellular movements were traced to a more mechanistic understanding of the local function of genes and regulatory networks underlying lineage segregation and tissue organization. In this chapter, we focus on some major advances addressing the segregation of lineages from the dermomyotome, while discussing both cellular as well as molecular mechanisms, where possible.
Collapse
Affiliation(s)
- Mordechai Applebaum
- Department of Medical Neurobiology, IMRIC and ELSC-Hebrew University-Hadassah Medical School, Jerusalem, 9101201, 12272, Israel,
| | | |
Collapse
|
36
|
Abstract
This review will focus on the use of the chicken and quail as model systems to analyze myogenesis and as such will emphasize the experimental approaches that are strongest in these systems-the amenability of the avian embryo to manipulation and in ovo observation. During somite differentiation, a wide spectrum of developmental processes occur such as cellular differentiation, migration, and fusion. Cell lineage studies combined with recent advancements in cell imaging allow these biological phenomena to be readily observed and hypotheses tested extremely rapidly-a strength that is restricted to the avian system. A clear weakness of the chicken in the past has been genetic approaches to modulate gene function. Recent advances in the electroporation of expression vectors, siRNA constructs, and use of tissue specific reporters have opened the door to increasingly sophisticated experiments that address questions of interest not only to the somite/muscle field in particular but also fundamental to biology in general. Importantly, an ever-growing body of evidence indicates that somite differentiation in birds is indistinguishable to that of mammals; therefore, these avian studies complement the complex genetic models of the mouse.
Collapse
Affiliation(s)
- Claire E Hirst
- EMBL Australia, Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton, VIC, 3800, Australia,
| | | |
Collapse
|
37
|
Burger NB, Stuurman KE, Kok E, Konijn T, Schooneman D, Niederreither K, Coles M, Agace WW, Christoffels VM, Mebius RE, van de Pavert SA, Bekker MN. Involvement of neurons and retinoic acid in lymphatic development: new insights in increased nuchal translucency. Prenat Diagn 2014; 34:1312-9. [PMID: 25088217 DOI: 10.1002/pd.4473] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/28/2014] [Accepted: 07/28/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Increased nuchal translucency originates from disturbed lymphatic development. Abnormal neural crest cell (NCC) migration may be involved in lymphatic development. Because both neuronal and lymphatic development share retinoic acid (RA) as a common factor, this study investigated the involvement of NCCs and RA in specific steps in lymphatic endothelial cell (LEC) differentiation and nuchal edema, which is the morphological equivalent of increased nuchal translucency. METHODS Mouse embryos in which all NCCs were fluorescently labeled (Wnt1-Cre;Rosa26(eYfp) ), reporter embryos for in vivo RA activity (DR5-luciferase) and embryos with absent (Raldh2(-/-) ) or in utero inhibition of RA signaling (BMS493) were investigated. Immunofluorescence using markers for blood vessels, lymphatic endothelium and neurons was applied. Flow cytometry was performed to measure specific LEC populations. RESULTS Cranial nerves were consistently close to the jugular lymph sac (JLS), in which NCCs were identified. In the absence of RA synthesis, enlarged JLS and nuchal edema were observed. Inhibiting RA signaling in utero resulted in a significantly higher amount of precursor-LECs at the expense of mature LECs and caused nuchal edema. CONCLUSIONS Neural crest cells are involved in lymphatic development. RA is required for differentiation into mature LECs. Blocking RA signaling in mouse embryos results in abnormal lymphatic development and nuchal edema.
Collapse
Affiliation(s)
- Nicole B Burger
- Department of Obstetrics and Gynecology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Taylor J, Pereyra A, Zhang T, Messi ML, Wang ZM, Hereñú C, Kuan PF, Delbono O. The Cavβ1a subunit regulates gene expression and suppresses myogenin in muscle progenitor cells. ACTA ACUST UNITED AC 2014; 205:829-46. [PMID: 24934157 PMCID: PMC4068134 DOI: 10.1083/jcb.201403021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cavβ1a acts as a voltage-gated calcium channel-independent regulator of gene expression in muscle progenitor cells and is required for their normal expansion during myogenic development. Voltage-gated calcium channel (Cav) β subunits are auxiliary subunits to Cavs. Recent reports show Cavβ subunits may enter the nucleus and suggest a role in transcriptional regulation, but the physiological relevance of this localization remains unclear. We sought to define the nuclear function of Cavβ in muscle progenitor cells (MPCs). We found that Cavβ1a is expressed in proliferating MPCs, before expression of the calcium conducting subunit Cav1.1, and enters the nucleus. Loss of Cavβ1a expression impaired MPC expansion in vitro and in vivo and caused widespread changes in global gene expression, including up-regulation of myogenin. Additionally, we found that Cavβ1a localizes to the promoter region of a number of genes, preferentially at noncanonical (NC) E-box sites. Cavβ1a binds to a region of the Myog promoter containing an NC E-box, suggesting a mechanism for inhibition of myogenin gene expression. This work indicates that Cavβ1a acts as a Cav-independent regulator of gene expression in MPCs, and is required for their normal expansion during myogenic development.
Collapse
Affiliation(s)
- Jackson Taylor
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Andrea Pereyra
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157 Biochemistry Research Institute of La Plata (INIBIOLP)/National Scientific and Technical Research Council (CONICET), School of Medicine, National University of La Plata, 1900 La Plata, BA, Argentina
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Claudia Hereñú
- Biochemistry Research Institute of La Plata (INIBIOLP)/National Scientific and Technical Research Council (CONICET), School of Medicine, National University of La Plata, 1900 La Plata, BA, Argentina
| | - Pei-Fen Kuan
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157Department of Internal Medicine-Gerontology, Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
39
|
Abstract
In amniotes, it is widely accepted that WNTs secreted by the dorsal neural tube form a concentration gradient that regulates early somite patterning and myotome organization. Here we demonstrate in the chicken embryo that WNT protein is not secreted to act at a distance, but rather loaded onto migrating neural crest cells that deliver it to somites. Inhibiting neural crest migration or ablating their population has a profound impact on the WNT response in somites. Furthermore, we show that a central player in the efficient delivery of WNT to somites is the heparan sulfate proteoglycan GPC4, expressed by neural crest. Together, our data describe a novel mode of signaling whereby WNT proteins hitch a ride on migratory neural crest cells to pattern the somites at a distance from its source.
Collapse
Affiliation(s)
- Olivier Serralbo
- EMBL Australia, Australian Regenerative Medicine Institute (ARMI), Monash University, Building 75, Clayton, Victoria 3800, Australia
| | - Christophe Marcelle
- EMBL Australia, Australian Regenerative Medicine Institute (ARMI), Monash University, Building 75, Clayton, Victoria 3800, Australia
| |
Collapse
|
40
|
Figeac N, Serralbo O, Marcelle C, Zammit PS. ErbB3 binding protein-1 (Ebp1) controls proliferation and myogenic differentiation of muscle stem cells. Dev Biol 2013; 386:135-51. [PMID: 24275324 DOI: 10.1016/j.ydbio.2013.11.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 10/21/2013] [Accepted: 11/15/2013] [Indexed: 12/16/2022]
Abstract
Satellite cells are resident stem cells of skeletal muscle, supplying myoblasts for post-natal muscle growth, hypertrophy and repair. Many regulatory networks control satellite cell function, which includes EGF signalling via the ErbB family of receptors. Here we investigated the role of ErbB3 binding protein-1 (Ebp1) in regulation of myogenic stem cell proliferation and differentiation. Ebp1 is a well-conserved DNA/RNA binding protein that is implicated in cell growth, apoptosis and differentiation in many cell types. Of the two main Ebp1 isoforms, only p48 was expressed in satellite cells and C2C12 myoblasts. Although not present in quiescent satellite cells, p48 was strongly induced during activation, remaining at high levels during proliferation and differentiation. While retroviral-mediated over-expression of Ebp1 had only minor effects, siRNA-mediated Ebp1 knockdown inhibited both proliferation and differentiation of satellite cells and C2C12 myoblasts, with a clear failure of myotube formation. Ebp1-knockdown significantly reduced ErbB3 receptor levels, yet over-expression of ErbB3 in Ebp1 knockdown cells did not rescue differentiation. Ebp1 was also expressed by muscle cells during developmental myogenesis in mouse. Since Ebp1 is well-conserved between mouse and chick, we switched to chick to examine its role in muscle formation. In chick embryo, Ebp1 was expressed in the dermomyotome, and myogenic differentiation of muscle progenitors was inhibited by specific Ebp1 down-regulation using shRNA electroporation. These observations demonstrate a conserved function of Ebp1 in the regulation of embryonic muscle progenitors and adult muscle stem cells, which likely operates independently of ErbB3 signaling.
Collapse
Affiliation(s)
- Nicolas Figeac
- King's College London, Randall Division of Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, England, UK
| | - Olivier Serralbo
- EMBL Australia, Australian Regenerative Medicine Institute (ARMI), Monash University, Building 75, Clayton, Victoria 3800, Australia
| | - Christophe Marcelle
- EMBL Australia, Australian Regenerative Medicine Institute (ARMI), Monash University, Building 75, Clayton, Victoria 3800, Australia
| | - Peter S Zammit
- King's College London, Randall Division of Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, England, UK.
| |
Collapse
|
41
|
Wilzén A, Krona C, Sveinbjörnsson B, Kristiansson E, Dalevi D, Øra I, De Preter K, Stallings RL, Maris J, Versteeg R, Nilsson S, Kogner P, Abel F. ERBB3 is a marker of a ganglioneuroblastoma/ganglioneuroma-like expression profile in neuroblastic tumours. Mol Cancer 2013; 12:70. [PMID: 23835063 PMCID: PMC3766266 DOI: 10.1186/1476-4598-12-70] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 06/25/2013] [Indexed: 12/21/2022] Open
Abstract
Background Neuroblastoma (NB) tumours are commonly divided into three cytogenetic subgroups. However, by unsupervised principal components analysis of gene expression profiles we recently identified four distinct subgroups, r1-r4. In the current study we characterized these different subgroups in more detail, with a specific focus on the fourth divergent tumour subgroup (r4). Methods Expression microarray data from four international studies corresponding to 148 neuroblastic tumour cases were subject to division into four expression subgroups using a previously described 6-gene signature. Differentially expressed genes between groups were identified using Significance Analysis of Microarray (SAM). Next, gene expression network modelling was performed to map signalling pathways and cellular processes representing each subgroup. Findings were validated at the protein level by immunohistochemistry and immunoblot analyses. Results We identified several significantly up-regulated genes in the r4 subgroup of which the tyrosine kinase receptor ERBB3 was most prominent (fold change: 132–240). By gene set enrichment analysis (GSEA) the constructed gene network of ERBB3 (n = 38 network partners) was significantly enriched in the r4 subgroup in all four independent data sets. ERBB3 was also positively correlated to the ErbB family members EGFR and ERBB2 in all data sets, and a concurrent overexpression was seen in the r4 subgroup. Further studies of histopathology categories using a fifth data set of 110 neuroblastic tumours, showed a striking similarity between the expression profile of r4 to ganglioneuroblastoma (GNB) and ganglioneuroma (GN) tumours. In contrast, the NB histopathological subtype was dominated by mitotic regulating genes, characterizing unfavourable NB subgroups in particular. The high ErbB3 expression in GN tumour types was verified at the protein level, and showed mainly expression in the mature ganglion cells. Conclusions Conclusively, this study demonstrates the importance of performing unsupervised clustering and subtype discovery of data sets prior to analyses to avoid a mixture of tumour subtypes, which may otherwise give distorted results and lead to incorrect conclusions. The current study identifies ERBB3 as a clear-cut marker of a GNB/GN-like expression profile, and we suggest a 7-gene expression signature (including ERBB3) as a complement to histopathology analysis of neuroblastic tumours. Further studies of ErbB3 and other ErbB family members and their role in neuroblastic differentiation and pathogenesis are warranted.
Collapse
Affiliation(s)
- Annica Wilzén
- Department of Clinical Genetics, Institution of Biomedicine, Box 413, S- 405 30, Gothenburg University, Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Theveneau E, Mayor R. Neural crest delamination and migration: from epithelium-to-mesenchyme transition to collective cell migration. Dev Biol 2012; 366:34-54. [PMID: 22261150 DOI: 10.1016/j.ydbio.2011.12.041] [Citation(s) in RCA: 374] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 12/26/2011] [Indexed: 10/25/2022]
Abstract
After induction and specification in the ectoderm, at the border of the neural plate, the neural crest (NC) population leaves its original territory through a delamination process. Soon afterwards, the NC cells migrate throughout the embryo and colonize a myriad of tissues and organs where they settle and differentiate. The delamination involves a partial or complete epithelium-to-mesenchyme transition (EMT) regulated by a complex network of transcription factors including several proto-oncogenes. Studying the relationship between these genes at the time of emigration, and their individual or collective impact on cell behavior, provides valuable information about their role in EMT in other contexts such as cancer metastasis. During migration, NC cells are exposed to large number of positive and negative regulators that control where they go by generating permissive and restricted areas and by modulating their motility and directionality. In addition, as most NC cells migrate collectively, cell-cell interactions play a crucial role in polarizing the cells and interpreting external cues. Cell cooperation eventually generates an overall polarity to the population, leading to directional collective cell migration. This review will summarize our current knowledge on delamination, EMT and migration of NC cells using key examples from chicken, Xenopus, zebrafish and mouse embryos. Given the similarities between neural crest migration and cancer invasion, these cells may represent a useful model for understanding the mechanisms of metastasis.
Collapse
Affiliation(s)
- Eric Theveneau
- Department of Cell and Developmental Biology, University College London, UK
| | | |
Collapse
|
43
|
Regulation of Trunk Myogenesis by the Neural Crest: A New Facet of Neural Crest-Somite Interactions. Dev Cell 2011; 21:187-8. [DOI: 10.1016/j.devcel.2011.07.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|