1
|
McLaughlin D, Keane A, Quinn J, Tipping N. The chick embryo model as an educational tool to explore the effect of alcohol on cardiovascular development. ADVANCES IN PHYSIOLOGY EDUCATION 2025; 49:438-460. [PMID: 40059860 DOI: 10.1152/advan.00195.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/03/2024] [Accepted: 03/04/2025] [Indexed: 04/08/2025]
Abstract
In development, the interrelationship between physiology and anatomy is challenging as learners must appreciate how physiological processes and anatomical structures change over time. In addition, the dynamic relationships between structure and function are often concealed largely due to the inaccessibility of the embryo in higher-order organisms. This makes it difficult for students to appreciate normal intricate balances or interpret the physiological consequences of developmental disruptions to normal embryological development. In this paper, the applicability of the chick embryo model for use in practical classes is explored as students can observe developmental processes firsthand within a controlled in ovo environment. Practical approaches involved in developing the chick embryo model are described and then expanded to demonstrate how the model can be utilized to showcase cardiovascular system development as an example. The model is further adapted to explore the effect of teratogenic disruptors such as ethanol on normal cardiovascular processes and highlights how prenatal alcohol exposure results in cardiovascular anomalies associated with fetal alcohol syndrome, such as septal defects and altered cardiac physiology. In class, students can directly observe chick development from 0 to 8 days postfertilization. Measurable outcomes, such as comparisons in septal thickness, are calculated, while questions and answers to stimulate student discussion around functional changes and the impact of maternal consumption of alcohol are provided as resource material. The method outlined uses relatively inexpensive materials and requires little space, making it a cost-effective educational tool to support student learning of embryology.NEW & NOTEWORTHY This study explores the use of the chick embryo model as a teaching aid to illustrate connections between anatomy and physiology during development. Providing direct observation opportunities, the model allows students to witness organ formation and the impact of teratogens, focusing on cardiovascular abnormalities associated with fetal alcohol syndrome. The paper outlines practical methodologies to assess developmental outcomes. Its adaptability, affordability, and ability to spark discussions make the model a valuable resource for diverse educational environments.
Collapse
Affiliation(s)
- Declan McLaughlin
- Centre for Biomedical Sciences Education, Queen's University Belfast, Belfast, United Kingdom
| | - Aisling Keane
- Centre for Biomedical Sciences Education, Queen's University Belfast, Belfast, United Kingdom
| | - Joe Quinn
- Centre for Biomedical Sciences Education, Queen's University Belfast, Belfast, United Kingdom
| | - Nuala Tipping
- Centre for Biomedical Sciences Education, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
2
|
Han X, Li Y, Wang E, Zhu H, Huang X, Pu W, Zhang M, Liu K, Zhao H, Liu Z, Zhao Y, Shen L, Li Y, Yang X, Wang QD, Ma X, Shen R, O Lui K, Wang L, He B, Zhou B. Exploring Origin-Dependent Susceptibility of Smooth Muscle Cells to Aortic Diseases Through Intersectional Genetics. Circulation 2025; 151:1248-1267. [PMID: 39925267 DOI: 10.1161/circulationaha.124.070782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/16/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND The developmental diversity among smooth muscle cells (SMCs) plays a crucial role in segment-specific aortic diseases. However, traditional genetic approaches are inadequate for enabling in vivo analysis of disease susceptibility associated with cellular origin. There is an urgent need to build genetic technologies that target different developmental origins to investigate the mechanisms of aortopathies, thereby facilitating the development of effective therapeutics. METHODS To address this challenge, we developed an advanced dual recombinase-mediated intersectional genetic system, specifically designed to precisely target SMCs from various developmental origins in mice. Specifically, we used Isl1-Dre, Wnt1-Dre, Meox1-DreER, and Upk3b-Dre to target SMC progenitors from the second heart field, cardiac neural crest, somites, and mesothelium, respectively. This system was combined with single-cell RNA sequencing to investigate the impact of TGF-β (transforming growth factor-β) signaling in different segments of the aorta by selectively knocking out Tgfbr2 in the ascending aorta and Smad4 in the aortic arch, respectively. RESULTS Through intersectional genetic approaches, we use the Myh11-Cre(ER) driver along with origin-specific Dre drivers to trace cells of diverse developmental origins within the SMC population. We found that a deficiency of Tgfbr2 in SMCs of the ascending aorta leads to aneurysm formation in this specific region. We also demonstrate the critical role of Smad4 in preserving aortic wall integrity and homeostasis in SMCs of the aortic arch. CONCLUSIONS Our approach to genetically targeting SMC subtypes provides a novel platform for exploring origin-dependent or location-specific aortic vascular diseases. This genetic system enables comprehensive analysis of contributions from different cell lineages to SMC behavior and pathology, thereby paving the way for targeted research and therapeutic interventions in the future.
Collapse
MESH Headings
- Animals
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Receptor, Transforming Growth Factor-beta Type II/metabolism
- Receptor, Transforming Growth Factor-beta Type II/deficiency
- Aortic Diseases/genetics
- Aortic Diseases/pathology
- Aortic Diseases/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Knockout
- Genetic Predisposition to Disease
- Aorta/metabolism
- Aorta/pathology
- Signal Transduction
Collapse
Affiliation(s)
- Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
| | - Enci Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (E.W., Y.Z., L.W.)
| | - Huan Zhu
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Xiuzhen Huang
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Wenjuan Pu
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Mingjun Zhang
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Kuo Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou (K.L., B.Z.)
| | - Huan Zhao
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Zixin Liu
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Yufei Zhao
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (E.W., Y.Z., L.W.)
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
| | - Yan Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China (Y.L.)
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China (Y.L.)
| | - Xiao Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, China (X.Y.)
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Q.-D.W.)
| | - Xin Ma
- Department of Pharmacology, Wuxi School of Medicine, Jiangnan University, China (X.M.)
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, China (R.S.)
| | - Kathy O Lui
- CAS CEMCS-CUHK Joint Laboratories, Department of Chemical Pathology; Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, Chinese University of Hong Kong, China (K.O.L.)
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (E.W., Y.Z., L.W.)
- Department of Vascular Surgery (Xiamen), Zhongshan Hospital, Fudan University, Xiamen, China (L.W.)
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
| | - Bin Zhou
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou (K.L., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (B.Z.)
| |
Collapse
|
3
|
Villaseca S, Leal JI, Tovar LM, Ruiz MJ, Guajardo J, Morales-Navarrete H, Mayor R, Torrejón M. Interaction of Gαi2 with EB1 controls microtubule dynamics and Rac1 activity in Xenopus neural crest cell migration. Development 2025; 152:dev204235. [PMID: 40136014 DOI: 10.1242/dev.204235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Cell migration is crucial in embryonic development, tissue repair and cancer metastasis, driven by the actin and tubulin cytoskeletons that control cell shape, polarity, adhesion and movement in response to various cues. Although heterotrimeric G proteins are known to be involved in cell migration, the specific mechanisms, especially during development, remain elusive. This study examines the role of Gαi2, a heterotrimeric G-protein subunit, in cranial neural crest (NC) cell migration during Xenopus embryonic development. Our research reveals that Gαi2 interacts directly with the microtubule-associated protein EB1, regulating microtubule dynamics. We show that Gαi2 knockdown stabilizes microtubules, disrupts cell polarity and morphology, increases Rac1-GTP at the leading edge and cell-cell contacts, and impairs actin localization and focal adhesion disassembly. Additionally, RhoA-GTP is reduced at cell-cell contacts and concentrated at the leading edge in Gαi2 knockdown cells, providing evidence of a role for Gαi2 in polarity. Treatment with nocodazole, a microtubule-depolymerizing agent, reduces Rac1 activity, restoring cranial NC cell morphology, actin distribution and overall migration. Our findings highlight a crucial role for Gαi2 in cranial NC cell migration by modulating microtubule dynamics through EB1 and Rac1 activity.
Collapse
Affiliation(s)
- Soraya Villaseca
- Laboratory of Signaling and Development, Group for the Study of Developmental Processes, Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Universidad de Concepción, Casilla 160-C, Concepción, Chile
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Juan Ignacio Leal
- Laboratory of Signaling and Development, Group for the Study of Developmental Processes, Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Lina Mariana Tovar
- Laboratory of Signaling and Development, Group for the Study of Developmental Processes, Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - María José Ruiz
- Laboratory of Signaling and Development, Group for the Study of Developmental Processes, Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Jossef Guajardo
- Laboratory of Signaling and Development, Group for the Study of Developmental Processes, Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | | | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Marcela Torrejón
- Laboratory of Signaling and Development, Group for the Study of Developmental Processes, Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| |
Collapse
|
4
|
Chen Z, Zhao C, Fu R, Yuan C, Zhang K, Zhang X. Wnt signaling pathway and retinoic acid signaling pathway involved in delamination and migration of chicken trunk NCCs and contributing to HVP phenotype. Poult Sci 2025; 104:105114. [PMID: 40209469 PMCID: PMC12005351 DOI: 10.1016/j.psj.2025.105114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/21/2025] [Accepted: 03/30/2025] [Indexed: 04/12/2025] Open
Abstract
Hyperpigmentation of the visceral peritoneum (HVP) is a hereditary trait that significantly affects the carcass quality in bearded chickens, yet its molecular mechanisms remain unclear. This study utilized data-independent acquisition proteomics to analyze the protein expression profiles of black peritoneum (B), faded peritoneum (F), and normal peritoneum (N) in bearded chickens at 40 and 120 d of age. Combined with histopathological and functional enrichment analyses, we revealed the regulatory network underlying HVP formation. Results indicated that the melanin content was significantly elevated in HVP samples, without accompanying inflammatory responses or tumor characteristics, suggesting that its formation is driven by developmental abnormalities. A total of 9,375 high-confidence proteins were identified through proteomics, with differentially abundant proteins at 40 d of age (219 proteins) primarily enriched in ribosomal function, tyrosine metabolism, and melanin synthesis pathways. In comparison, at 120 d of age (246 proteins), they were enriched in transcription regulation and chromatin remodeling pathways. The abnormal expression of key co-expressed proteins DHRS3 and DACT1 suggests that the dysregulation of retinoic acid (RA) and the Wnt signaling pathway may promote the directed differentiation of melanocytes by regulating neural crest cells (NCCs). The reduced abundance of the chondroitin sulfate proteoglycan, VCAN, weakened the peritoneal barrier function, whereas estradiol accelerated melanin synthesis via hormonal microenvironmental regulation. Furthermore, the formation of HVP led to a reprogramming of energy metabolism, reduced fat deposition, and a downregulation of immune-related molecules, implying that pigment deposition may weaken the chicken immune response. This study systematically elucidates the molecular mechanisms of HVP and provides potential targets for molecular breeding of HVP.
Collapse
Affiliation(s)
- Zhengyang Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China
| | - Changbin Zhao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China
| | - Rong Fu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China
| | - Chengyue Yuan
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China
| | - Ke Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China
| | - Xiquan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affair, Guangzhou, Guangdong, PR China; State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou, PR China.
| |
Collapse
|
5
|
Moore Zajic EL, Zhao R, McKinney MC, Yi K, Wood C, Trainor PA. Cell extrusion drives neural crest cell delamination. Proc Natl Acad Sci U S A 2025; 122:e2416566122. [PMID: 40063802 PMCID: PMC11929498 DOI: 10.1073/pnas.2416566122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/17/2025] [Indexed: 03/15/2025] Open
Abstract
Neural crest cells (NCC) comprise a heterogeneous population of cells with variable potency that contribute to nearly every tissue and organ throughout the body. Considered unique to vertebrates, NCC are transiently generated within the dorsolateral region of the neural plate or neural tube during neurulation. Their delamination and migration are crucial for embryo development as NCC differentiation is influenced by their final resting locations. Previous work in avian and aquatic species revealed that NCC delaminate via an epithelial-mesenchymal transition (EMT), which transforms these progenitor cells from static polarized epithelial cells into migratory mesenchymal cells with fluid front and back polarity. However, the cellular and molecular mechanisms facilitating NCC delamination in mammals are poorly understood. Through time-lapse imaging of NCC delamination in mouse embryos, we identified a subset of cells that exit the neuroepithelium as isolated round cells, which then halt for a short period prior to acquiring the mesenchymal migratory morphology classically associated with delaminating NCC. High-magnification imaging and protein localization analyses of the cytoskeleton, together with measurements of pressure and tension of delaminating NCC and neighboring neuroepithelial cells, revealed that round NCC are extruded from the neuroepithelium prior to completion of EMT. Furthermore, cranial NCC are extruded through activation of the mechanosensitive ion channel, PIEZO1. Our results support a model in which cell density, pressure, and tension in the neuroepithelium result in activation of the live cell extrusion pathway and delamination of a subpopulation of NCC in parallel with EMT, which has implications for cell delamination in development and disease.
Collapse
Affiliation(s)
| | - Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| | | | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO64110
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
6
|
Pally D, Kapoor N, Naba A. The novel ECM protein SNED1 mediates cell adhesion via the RGD-binding integrins α5β1 and αvβ3. J Cell Sci 2025; 138:JCS263479. [PMID: 39713860 PMCID: PMC11828466 DOI: 10.1242/jcs.263479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
The extracellular matrix (ECM) is a complex meshwork comprising over 100 proteins. It serves as an adhesive substrate for cells and, hence, plays crucial roles in health and disease. We have recently identified a novel ECM protein, SNED1, and have found that it is required for neural crest cell migration and craniofacial morphogenesis during development and in breast cancer, where it is necessary for the metastatic dissemination of tumor cells. Interestingly, both processes involve the dynamic remodeling of cell-ECM adhesions via cell surface receptors. Sequence analysis revealed that SNED1 contains two amino acid motifs, RGD and LDV, known to bind integrins, the largest class of ECM receptors. We thus sought to investigate the role of SNED1 in cell adhesion. Here, we report that SNED1 mediates breast cancer and neural crest cell adhesion via its RGD motif. We further demonstrate that cell adhesion to SNED1 is mediated by the RGD integrins α5β1 and αvβ3. These findings are a first step toward identifying the signaling pathways activated downstream of the SNED1-integrin interactions guiding craniofacial morphogenesis and breast cancer metastasis.
Collapse
Affiliation(s)
- Dharma Pally
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Nandini Kapoor
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| |
Collapse
|
7
|
Salinas E, Ruano-Rivadeneira F, Leal JI, Caprile T, Torrejón M, Arriagada C. Polarity and migration of cranial and cardiac neural crest cells: underlying molecular mechanisms and disease implications. Front Cell Dev Biol 2025; 12:1457506. [PMID: 39834387 PMCID: PMC11743681 DOI: 10.3389/fcell.2024.1457506] [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: 06/30/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
The Neural Crest cells are multipotent progenitor cells formed at the neural plate border that differentiate and give rise to a wide range of cell types and organs. Directional migration of NC cells and their correct positioning at target sites are essential during embryonic development, and defects in these processes results in congenital diseases. The NC migration begins with the epithelial-mesenchymal transition and extracellular matrix remodeling. The main cellular mechanisms that sustain this migration include contact inhibition of locomotion, co-attraction, chemotaxis and mechanical cues from the surrounding environment, all regulated by proteins that orchestrate cell polarity and motility. In this review we highlight the molecular mechanisms involved in neural crest cell migration and polarity, focusing on the role of small GTPases, Heterotrimeric G proteins and planar cell polarity complex. Here, we also discuss different congenital diseases caused by altered NC cell migration.
Collapse
Affiliation(s)
- Esteban Salinas
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Francis Ruano-Rivadeneira
- Developmental Biology Laboratory 116, School of Biological Sciences, Faculty of Exact and Natural Sciences, Pontificia Universidad Católica del Ecuador, Quito, Ecuador
| | - Juan Ignacio Leal
- Laboratory of Signaling and Development (LSD), Group for the Study of Developmental Processes (GDeP), Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Teresa Caprile
- Laboratory of Axonal Guidance, Group for the Study of Developmental Processes (GDeP), Department of Cellular Biology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Marcela Torrejón
- Laboratory of Signaling and Development (LSD), Group for the Study of Developmental Processes (GDeP), Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Concepción, Concepción, Chile
| | - Cecilia Arriagada
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
8
|
Pally D, Kapoor N, Naba A. The novel ECM protein SNED1 mediates cell adhesion via the RGD-binding integrins α5β1 and αvβ3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.606706. [PMID: 39149327 PMCID: PMC11326288 DOI: 10.1101/2024.08.07.606706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The extracellular matrix (ECM) is a complex meshwork comprising over 100 proteins. It serves as an adhesive substrate for cells and, hence, plays critical roles in health and disease. We have recently identified a novel ECM protein, SNED1, and have found that it is required for neural crest cell migration and craniofacial morphogenesis during development and in breast cancer, where it is necessary for the metastatic dissemination of tumor cells. Interestingly, both processes involve the dynamic remodeling of cell-ECM adhesions via cell surface receptors. Sequence analysis revealed that SNED1 contains two amino acid motifs, RGD and LDV, known to bind integrins, the largest class of ECM receptors. We thus sought to investigate the role of SNED1 in cell adhesion. Here, we report that SNED1 mediates breast cancer and neural crest cell adhesion via its RGD motif. We further demonstrate that cell adhesion to SNED1 is mediated by the RGD integrins α5β1 and αvβ3. These findings are a first step toward identifying the signaling pathways activated downstream of the SNED1-integrin interactions guiding craniofacial morphogenesis and breast cancer metastasis.
Collapse
Affiliation(s)
- Dharma Pally
- Department of Physiology and Biophysics, University of Illinois Chicago, Illinois, 60612, USA
| | - Nandini Kapoor
- Department of Physiology and Biophysics, University of Illinois Chicago, Illinois, 60612, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Illinois, 60612, USA
- University of Illinois Cancer Center, Chicago, Illinois, 60612, USA
| |
Collapse
|
9
|
Li Z, Lin J, Wu J, Suo J, Wang Z. The Hippo signalling pathway in bone homeostasis: Under the regulation of mechanics and aging. Cell Prolif 2024; 57:e13652. [PMID: 38700015 PMCID: PMC11471399 DOI: 10.1111/cpr.13652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
The Hippo signalling pathway is a conserved kinase cascade that orchestrates diverse cellular processes, such as proliferation, apoptosis, lineage commitment and stemness. With the onset of society ages, research on skeletal aging-mechanics-bone homeostasis has exploded. In recent years, aging and mechanical force in the skeletal system have gained groundbreaking research progress. Under the regulation of mechanics and aging, the Hippo signalling pathway has a crucial role in the development and homeostasis of bone. We synthesize the current knowledge on the role of the Hippo signalling pathway, particularly its downstream effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), in bone homeostasis. We discuss the regulation of the lineage specification and function of different skeletal cell types by the Hippo signalling pathway. The interactions of the Hippo signalling pathway with other pathways, such as Wnt, transforming growth factor beta and nuclear factor kappa-B, are also mentioned because of their importance for modulating bone homeostasis. Furthermore, YAP/TAZ have been extensively studied as mechanotransducers. Due to space limitations, we focus on reviewing how mechanical forces and aging influence cell fate, communications and homeostasis through a dysregulated Hippo signalling pathway.
Collapse
Affiliation(s)
- Zhengda Li
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Junqing Lin
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Jing Wu
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, and Department of Orthopedic SurgeryShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine ShanghaiShanghaiChina
| | - Zuoyun Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences and Shanghai Jing'an District Central HospitalFudan UniversityShanghaiChina
| |
Collapse
|
10
|
Wang H, Hu W, Xiang F, Lei Z, Zhang X, Zhang J, Ding Y, Kang X. Differentiation of cultured hair follicle neural crest stem cells into functional melanocytes. Heliyon 2024; 10:e35295. [PMID: 39170163 PMCID: PMC11336637 DOI: 10.1016/j.heliyon.2024.e35295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/02/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024] Open
Abstract
Many autologous melanocytes are required for surgical treatment of depigmentation diseases such as vitiligo. However, primary cultured melanocytes have a limited number of in vitro passages. The production of functional epidermal melanocytes from stem cells provides an unprecedented source of cell therapy for vitiligo. This study explores the clinical application of melanocytes induced by hair follicle neural crest stem cells (HFNCSCs). This study established an in vitro differentiation model of HFNCSCs into melanocytes. Results demonstrate that most differentiated melanocytes expressed the proteins C-KIT, MITF, S-100B, TYRP1, TYRP2, and tyrosinase. The HFNCSC-derived melanocytes were successfully transplanted onto the dorsal skin of mice and survived in the local tissues, expressing marker protein of melanocytes. In conclusion, HFNCSCs in mice can be induced to differentiate into melanocytes under specific conditions. These induced melanocytes exhibit the potential to facilitate repigmentation in the lesion areas of vitiligo-affected mice, suggesting a promising avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Hongjuan Wang
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Dermatologic Diseases, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, 830000, Xinjiang, China
| | - Wen Hu
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Dermatologic Diseases, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, 830000, Xinjiang, China
| | - Fang Xiang
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Dermatologic Diseases, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, 830000, Xinjiang, China
| | - Zixian Lei
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Dermatologic Diseases, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, 830000, Xinjiang, China
| | - Xiangyue Zhang
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Dermatologic Diseases, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, 830000, Xinjiang, China
| | - Jingzhan Zhang
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Dermatologic Diseases, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, 830000, Xinjiang, China
| | - Yuan Ding
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Dermatologic Diseases, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, 830000, Xinjiang, China
| | - Xiaojing Kang
- Department of Dermatology and Venereology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center for Dermatologic Diseases, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Dermatology Research, Urumqi, 830000, Xinjiang, China
| |
Collapse
|
11
|
Ventriglia S, Kalcheim C. From neural tube to spinal cord: The dynamic journey of the dorsal neuroepithelium. Dev Biol 2024; 511:26-38. [PMID: 38580174 DOI: 10.1016/j.ydbio.2024.04.001] [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: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
In a developing embryo, formation of tissues and organs is remarkably precise in both time and space. Through cell-cell interactions, neighboring progenitors coordinate their activities, sequentially generating distinct types of cells. At present, we only have limited knowledge, rather than a systematic understanding, of the underlying logic and mechanisms responsible for cell fate transitions. The formation of the dorsal aspect of the spinal cord is an outstanding model to tackle these dynamics, as it first generates the peripheral nervous system and is later responsible for transmitting sensory information from the periphery to the brain and for coordinating local reflexes. This is reflected first by the ontogeny of neural crest cells, progenitors of the peripheral nervous system, followed by formation of the definitive roof plate of the central nervous system and specification of adjacent interneurons, then a transformation of roof plate into dorsal radial glia and ependyma lining the forming central canal. How do these peripheral and central neural branches segregate from common progenitors? How are dorsal radial glia established concomitant with transformation of the neural tube lumen into a central canal? How do the dorsal radial glia influence neighboring cells? This is only a partial list of questions whose clarification requires the implementation of experimental paradigms in which precise control of timing is crucial. Here, we outline some available answers and still open issues, while highlighting the contributions of avian models and their potential to address mechanisms of neural patterning and function.
Collapse
Affiliation(s)
- Susanna Ventriglia
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| |
Collapse
|
12
|
Soliman O, Acharya Y, Gilard M, Duffy G, Wijns W, Kannan V, Sultan S. Systematic review of cardiovascular neurocristopathy-contemporary insights and future perspectives. Front Cardiovasc Med 2024; 11:1333265. [PMID: 38660479 PMCID: PMC11040563 DOI: 10.3389/fcvm.2024.1333265] [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: 12/02/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Neural crest cells (NCCs) are multipotent and are attributed to the combination of complex multimodal gene regulatory mechanisms. Cardiac neural crest (CNC) cells, originating from the dorsal neural tube, are pivotal architects of the cardio-neuro-vascular domain, which orchestrates the embryogenesis of critical cardiac and vascular structures. Remarkably, while the scientific community compiled a comprehensive inventory of neural crest derivatives by the early 1980s, our understanding of the CNC's role in various cardiovascular disease processes still needs to be explored. This review delves into the differentiation of NCC, specifically the CNC cells, and explores the diverse facets of non-syndromic cardiovascular neurocristopathies. Methods A systematic review was conducted as per the PRISMA Statement. Three prominent databases, PubMed, Scopus, and Embase, were searched, which yielded 1,840 studies. We excluded 1,796 studies, and the final selection of 44 studies formed the basis of this comprehensive review. Results Neurocristopathies are a group of genetic disorders that affect the development of cells derived from the NC. Cardiovascular neurocristopathy, i.e., cardiopathy and vasculopathy, associated with the NCC could occur in the form of (1) cardiac septation disorders, mainly the aortico-pulmonary septum; (2) great vessels and vascular disorders; (3) myocardial dysfunction; and (4) a combination of all three phenotypes. This could result from abnormalities in NCC migration, differentiation, or proliferation leading to structural abnormalities and are attributed to genetic, familial, sporadic or acquired causes. Discussion Phenotypic characteristics of cardiovascular neurocristopathies, such as bicuspid aortic valve and thoracic aortic aneurysm, share a common embryonic origin and are surprisingly prevalent in the general population, necessitating further research to identify the underlying pathogenic and genetic factors responsible for these cardiac anomalies. Such discoveries are essential for enhancing diagnostic screening and refining therapeutic interventions, ultimately improving the lives of individuals affected by these conditions.
Collapse
Affiliation(s)
- Osama Soliman
- Department of Cardiology, Galway University Hospital, Galway, Ireland
- CORRIB-CURAM-Vascular Group Collaborators, University of Galway, Galway, Ireland
| | - Yogesh Acharya
- CORRIB-CURAM-Vascular Group Collaborators, University of Galway, Galway, Ireland
- Western Vascular Institute, Department of Vascular and Endovascular Surgery, University Hospital Galway, University of Galway, Galway, Ireland
| | - Martine Gilard
- CORRIB-CURAM-Vascular Group Collaborators, University of Galway, Galway, Ireland
- Department of Cardiology, La Cavale Blanche Hospital, Brest, France
| | - Garry Duffy
- CORRIB-CURAM-Vascular Group Collaborators, University of Galway, Galway, Ireland
- Anatomy and Regenerative Medicine Institute (REMEDI), School of Medicine, University of Galway, Galway, Ireland
| | - William Wijns
- Department of Cardiology, Galway University Hospital, Galway, Ireland
- CORRIB-CURAM-Vascular Group Collaborators, University of Galway, Galway, Ireland
| | - Venkatesh Kannan
- CORRIB-CURAM-Vascular Group Collaborators, University of Galway, Galway, Ireland
- Irish Centre for High-End Computing (ICHEC), University of Galway, Galway, Ireland
| | - Sherif Sultan
- CORRIB-CURAM-Vascular Group Collaborators, University of Galway, Galway, Ireland
- Western Vascular Institute, Department of Vascular and Endovascular Surgery, University Hospital Galway, University of Galway, Galway, Ireland
- Department of Vascular Surgery and Endovascular Surgery, Galway Clinic, Doughiska, Royal College of Surgeons in Ireland and University of Galway Affiliated Hospital, Galway, Ireland
| |
Collapse
|
13
|
Moore E, Zhao R, McKinney MC, Yi K, Wood C, Trainor P. Cell extrusion - a novel mechanism driving neural crest cell delamination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.09.584232. [PMID: 38559094 PMCID: PMC10979875 DOI: 10.1101/2024.03.09.584232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Neural crest cells (NCC) comprise a heterogeneous population of cells with variable potency, that contribute to nearly every tissue and organ system throughout the body. Considered unique to vertebrates, NCC are transiently generated within the dorsolateral region of the neural plate or neural tube, during neurulation. Their delamination and migration are crucial events in embryo development as the differentiation of NCC is heavily influenced by their final resting locations. Previous work in avian and aquatic species has shown that NCC delaminate via an epithelial-mesenchymal transition (EMT), which transforms these stem and progenitor cells from static polarized epithelial cells into migratory mesenchymal cells with fluid front and back polarity. However, the cellular and molecular drivers facilitating NCC delamination in mammals are poorly understood. We performed live timelapse imaging of NCC delamination in mouse embryos and discovered a group of cells that exit the neuroepithelium as isolated round cells, which then halt for a short period prior to acquiring the mesenchymal migratory morphology classically associated with most delaminating NCC. High magnification imaging and protein localization analyses of the cytoskeleton, together with measurements of pressure and tension of delaminating NCC and neighboring neuroepithelial cells, revealed these round NCC are extruded from the neuroepithelium prior to completion of EMT. Furthermore, we demonstrate that cranial NCC are extruded through activation of the mechanosensitive ion channel, PIEZO1, a key regulator of the live cell extrusion pathway, revealing a new role for PIEZO1 in neural crest cell development. Our results elucidating the cellular and molecular dynamics orchestrating NCC delamination support a model in which high pressure and tension in the neuroepithelium results in activation of the live cell extrusion pathway and delamination of a subpopulation of NCC in parallel with EMT. This model has broad implications for our understanding of cell delamination in development and disease.
Collapse
Affiliation(s)
- Emma Moore
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Ruonan Zhao
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Mary C McKinney
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Paul Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
14
|
Crossley RM, Johnson S, Tsingos E, Bell Z, Berardi M, Botticelli M, Braat QJS, Metzcar J, Ruscone M, Yin Y, Shuttleworth R. Modeling the extracellular matrix in cell migration and morphogenesis: a guide for the curious biologist. Front Cell Dev Biol 2024; 12:1354132. [PMID: 38495620 PMCID: PMC10940354 DOI: 10.3389/fcell.2024.1354132] [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: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The extracellular matrix (ECM) is a highly complex structure through which biochemical and mechanical signals are transmitted. In processes of cell migration, the ECM also acts as a scaffold, providing structural support to cells as well as points of potential attachment. Although the ECM is a well-studied structure, its role in many biological processes remains difficult to investigate comprehensively due to its complexity and structural variation within an organism. In tandem with experiments, mathematical models are helpful in refining and testing hypotheses, generating predictions, and exploring conditions outside the scope of experiments. Such models can be combined and calibrated with in vivo and in vitro data to identify critical cell-ECM interactions that drive developmental and homeostatic processes, or the progression of diseases. In this review, we focus on mathematical and computational models of the ECM in processes such as cell migration including cancer metastasis, and in tissue structure and morphogenesis. By highlighting the predictive power of these models, we aim to help bridge the gap between experimental and computational approaches to studying the ECM and to provide guidance on selecting an appropriate model framework to complement corresponding experimental studies.
Collapse
Affiliation(s)
- Rebecca M. Crossley
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Samuel Johnson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Erika Tsingos
- Computational Developmental Biology Group, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Zoe Bell
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Optics11 life, Amsterdam, Netherlands
| | | | - Quirine J. S. Braat
- Department of Applied Physics and Science Education, Eindhoven University of Technology, Eindhoven, Netherlands
| | - John Metzcar
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
- Department of Informatics, Indiana University, Bloomington, IN, United States
| | | | - Yuan Yin
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
15
|
Huber PB, LaBonne C. Small molecule-mediated reprogramming of Xenopus blastula stem cells to a neural crest state. Dev Biol 2024; 505:34-41. [PMID: 37890713 PMCID: PMC11541498 DOI: 10.1016/j.ydbio.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023]
Abstract
Neural crest cells are a stem cell population unique to vertebrates that give rise to a diverse array of derivatives, including much of the peripheral nervous system, pigment cells, cartilage, mesenchyme, and bone. Acquisition of these cells drove the evolution of vertebrates and defects in their development underlies a broad set of neurocristopathies. Moreover, studies of neural crest can inform differentiation protocols for pluripotent stem cells and regenerative medicine applications. Xenopus embryos are an important system for studies of the neural crest and have provided numerous insights into the signals and transcription factors that control the formation and later lineage diversification of these stem cells. Pluripotent animal pole explants are a particularly powerful tool in this system as they can be cultured in simple salt solution and instructed to give rise to any cell type including the neural crest. Here we report a protocol for small molecule-mediated induction of the neural crest state from blastula stem cells and validate it using transcriptome analysis and grafting experiments. This is an powerful new tool for generating this important cell type that will facilitate future studies of neural crest development and mutations and variants linked to neurocristopathies.
Collapse
Affiliation(s)
- Paul B Huber
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA; NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA; NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
16
|
Song F, Zheng D, Yang Z, Shi L, Lu X, Yao F, Liang H, Wang L, Wang X, Chen H, Sun J, Luo J. Weighted correlation network analysis of the genes in the eyes of juvenile Plectropomus leopardus provide novel insights into the molecular mechanisms of the adaptation to the background color. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 48:101123. [PMID: 37604728 DOI: 10.1016/j.cbd.2023.101123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 08/23/2023]
Abstract
Plectropomus leopardus is a valuable marine fish whose skin color is strongly affected by the background color. However, the influence of the visual sense on the skin color variation of P. leopardus remains unknown. In the present study, transcriptome analysis was used to examine the visual response mechanism under different background colors. Paraffin sections of the eyes showed that the background color caused morphological changes in the pigment cells (PCs) and outer nuclear layer (ONL) and the darkening of the iris color. The transcriptome analysis results indicated that the gene expressions in the eyes of P. leopardus were significantly different for different background colors. We identified 4845, 3069, 5874, and 6309 differentially expressed genes (DEGs) in the pairwise comparisons of white vs. initial, blue vs. initial, red vs. initial, and black vs. initial groups, respectively. Some hub genes and key pathways regulating the adaptive mechanism of P. leopardus's eyes to the background color were identified, i.e., the JAK-STAT, mTOR, and Ras signaling pathways, and the ndufb7, slc6a13, and novel.3553 gene. This adaptation was achieved through the synthesis of stress proteins and energy balance supply mediated by hub genes and key pathways. In addition, the phenylalanine metabolism, tyrosine metabolism, and actin cytoskeleton-related processes or pathways and genes were responsible for iris and skin color adaptation. In summary, we inferred that stress protein synthesis, phenylalanine metabolism, and energy homeostasis were critical stress pathways for P. leopardus to adapt its skin color to the environment. These new findings indicate that the P. leopardus skin color variation may have been caused by the environmental adaption of the eyes. The results provide new insights into the molecular mechanisms underlying the skin color adaptation of P. leopardus.
Collapse
Affiliation(s)
- Feibiao Song
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Da Zheng
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Zihang Yang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Liping Shi
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Xingyu Lu
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Fucheng Yao
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Huan Liang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Lei Wang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Xinxin Wang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Huapeng Chen
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Junlong Sun
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China
| | - Jian Luo
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan Aquaculture Breeding Engineering Research Center, Hainan Academician Team Innovation Center, Sanya Nanfan Research Institute of Hainan University, College of Marine Sciences, Hainan University, Haikou 570228, China.
| |
Collapse
|
17
|
Li Z, Jia K, Chen X, Guo J, Zheng Z, Chen W, Peng Y, Yang Y, Lu H, Yang J. Exposure to Butylparaben Induces Craniofacial Bone Developmental Toxicity in Zebrafish (Danio rerio) Embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 265:115523. [PMID: 37776822 DOI: 10.1016/j.ecoenv.2023.115523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/30/2023] [Accepted: 09/23/2023] [Indexed: 10/02/2023]
Abstract
Butylparaben (BuP) is a common antibacterial preservative utilized extensively in food, medical supplies, cosmetics, and personal care products. The current study reports the use of Zebrafish (Danio rerio) embryos to investigate potential developmental toxicity caused by exposure to BuP. The development of Neural crest cells (NCCs) is highly active during gastrulation in Zebrafish embryos. Thus, we utilized 0.5 mg/L, 0.75 mg/L, and 1 mg/L BuP solutions, respectively, in accordance with the international safety standard dosage. We observed severe craniofacial cartilage deformities, periocular edema, cardiac dysplasia, and delayed otolith development in the Zebrafish larvae 5 days after exposure. The oxidative stress response was significantly enhanced. In addition, the biochemical analysis revealed that the activities of catalase (CAT) and superoxide dismutase (SOD) were significantly reduced relative to the control group, whereas the concentration of malondialdehyde (MDA) was significantly elevated. Furthermore, ALP activity, a marker of osteoblast activity, was also reduced. Moreover, the RT-qPCR results indicated that the expression of chondrocyte marker genes sox9a, sox9b, and col2a1a was down-regulated. In addition, the morphology of maxillofacial chondrocytes was altered in Zebrafish larvae, and the proliferation of cranial NCCs was inhibited. Accordingly, our findings indicate that strong oxidative stress induced by BuP inhibits the proliferation of NCCs in larval Zebrafish, leading to craniofacial deformities.
Collapse
Affiliation(s)
- Zekun Li
- Department of Endodontics, Affiliated Stomatological Hospital, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory of Oral Biomedicine, Jiangxi Clinical Medical Research Center of Oral Diseases, Nanchang 330006, Jiangxi,China
| | - Kun Jia
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, Jiangxi, China; School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Xiaomei Chen
- Department of Endodontics, Affiliated Stomatological Hospital, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory of Oral Biomedicine, Jiangxi Clinical Medical Research Center of Oral Diseases, Nanchang 330006, Jiangxi,China
| | - Jun Guo
- Department of Endodontics, Affiliated Stomatological Hospital, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory of Oral Biomedicine, Jiangxi Clinical Medical Research Center of Oral Diseases, Nanchang 330006, Jiangxi,China
| | - Zhiguo Zheng
- Department of Endodontics, Affiliated Stomatological Hospital, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory of Oral Biomedicine, Jiangxi Clinical Medical Research Center of Oral Diseases, Nanchang 330006, Jiangxi,China
| | - Weihua Chen
- Jiangxi Key Laboratory of Oral Biomedicine, Jiangxi Clinical Medical Research Center of Oral Diseases, Nanchang 330006, Jiangxi,China
| | - Yuan Peng
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, Jiangxi, China
| | - Yuhao Yang
- Department of Endodontics, Affiliated Stomatological Hospital, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory of Oral Biomedicine, Jiangxi Clinical Medical Research Center of Oral Diseases, Nanchang 330006, Jiangxi,China
| | - Huiqiang Lu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, Jiangxi, China; Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an 343009, Jiangxi, China
| | - Jian Yang
- Department of Endodontics, Affiliated Stomatological Hospital, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Key Laboratory of Oral Biomedicine, Jiangxi Clinical Medical Research Center of Oral Diseases, Nanchang 330006, Jiangxi,China.
| |
Collapse
|
18
|
Bai X, Cao R, Wu D, Zhang H, Yang F, Wang L. Dental Pulp Stem Cells for Bone Tissue Engineering: A Literature Review. Stem Cells Int 2023; 2023:7357179. [PMID: 37868704 PMCID: PMC10586346 DOI: 10.1155/2023/7357179] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/03/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
Bone tissue engineering (BTE) is a promising approach for repairing and regenerating damaged bone tissue, using stem cells and scaffold structures. Among various stem cell sources, dental pulp stem cells (DPSCs) have emerged as a potential candidate due to their multipotential capabilities, ability to undergo osteogenic differentiation, low immunogenicity, and ease of isolation. This article reviews the biological characteristics of DPSCs, their potential for BTE, and the underlying transcription factors and signaling pathways involved in osteogenic differentiation; it also highlights the application of DPSCs in inducing scaffold tissues for bone regeneration and summarizes animal and clinical studies conducted in this field. This review demonstrates the potential of DPSC-based BTE for effective bone repair and regeneration, with implications for clinical translation.
Collapse
Affiliation(s)
- Xiaolei Bai
- Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Ruijue Cao
- Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Danni Wu
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Huicong Zhang
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Fan Yang
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| | - Linhong Wang
- Center for Plastic & Reconstructive Surgery, Department of Stomatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310018, Zhejiang, China
| |
Collapse
|
19
|
Kwartler CS, Pedroza AJ, Kaw A, Guan P, Ma S, Duan XY, Kernell C, Wang C, Pinelo JEE, Bowen MSB, Chen J, Zhong Y, Sinha S, Shen X, Fischbein MP, Milewicz DM. Nuclear Smooth Muscle α-actin Participates in Vascular Smooth Muscle Cell Differentiation. NATURE CARDIOVASCULAR RESEARCH 2023; 2:937-955. [PMID: 38919852 PMCID: PMC11198982 DOI: 10.1038/s44161-023-00337-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/23/2023] [Indexed: 06/27/2024]
Abstract
Missense variants throughout ACTA2, encoding smooth muscle α-actin (αSMA), predispose to adult-onset thoracic aortic disease, but variants disrupting arginine 179 (R179) lead to Smooth Muscle Dysfunction Syndrome (SMDS) characterized by diverse childhood-onset vascular diseases. Here we show that αSMA localizes to the nucleus in wildtype (WT) smooth muscle cells (SMCs), enriches in the nucleus with SMC differentiation, and associates with chromatin remodeling complexes and SMC contractile gene promotors. The ACTA2 p.R179 αSMA variant shows decreased nuclear localization. Primary SMCs from Acta2 SMC-R179C/+ mice are less differentiated than WT SMCs in vitro and in vivo and have global changes in chromatin accessibility. Induced pluripotent stem cells from patients with ACTA2 p.R179 variants fail to fully differentiate from neuroectodermal progenitor cells to SMCs, and single-cell transcriptomic analyses of an ACTA2 p.R179H patient's aortic tissue show increased SMC plasticity. Thus, nuclear αSMA participates in SMC differentiation, and loss of this nuclear activity occurs with ACTA2 p.R179 pathogenic variants.
Collapse
Affiliation(s)
- Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305
| | - Anita Kaw
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Pujun Guan
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Shuangtao Ma
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
- Current address: Department Medicine, Michigan State University, East Lansing, MI 48824
| | - Xue-yan Duan
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Caroline Kernell
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Charis Wang
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Jose Emiliano Esparza Pinelo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Mikayla S. Borthwick Bowen
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Jiyuan Chen
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yuan Zhong
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Xuetong Shen
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
| | | | - Dianna M. Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
20
|
Lemaître QIB, Bartsch N, Kouzel IU, Busengdal H, Richards GS, Steinmetz PRH, Rentzsch F. NvPrdm14d-expressing neural progenitor cells contribute to non-ectodermal neurogenesis in Nematostella vectensis. Nat Commun 2023; 14:4854. [PMID: 37563174 PMCID: PMC10415408 DOI: 10.1038/s41467-023-39789-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 06/29/2023] [Indexed: 08/12/2023] Open
Abstract
Neurogenesis has been studied extensively in the ectoderm, from which most animals generate the majority of their neurons. Neurogenesis from non-ectodermal tissue is, in contrast, poorly understood. Here we use the cnidarian Nematostella vectensis as a model to provide new insights into the molecular regulation of non-ectodermal neurogenesis. We show that the transcription factor NvPrdm14d is expressed in a subpopulation of NvSoxB(2)-expressing endodermal progenitor cells and their NvPOU4-expressing progeny. Using a new transgenic reporter line, we show that NvPrdm14d-expressing cells give rise to neurons in the body wall and in close vicinity of the longitudinal retractor muscles. RNA-sequencing of NvPrdm14d::GFP-expressing cells and gene knockdown experiments provide candidate genes for the development and function of these neurons. Together, the identification of a population of endoderm-specific neural progenitor cells and of previously undescribed putative motoneurons in Nematostella provide new insights into the regulation of non-ectodermal neurogenesis.
Collapse
Affiliation(s)
- Quentin I B Lemaître
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway
| | - Natascha Bartsch
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway
- Department for Biological Sciences, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway
| | - Ian U Kouzel
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway
| | - Henriette Busengdal
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway
| | - Gemma Sian Richards
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway
| | | | - Fabian Rentzsch
- Michael Sars Centre, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway.
- Department for Biological Sciences, University of Bergen, Thormøhlensgate 55, 5006, Bergen, Norway.
| |
Collapse
|
21
|
Gouignard N, Bibonne A, Mata JF, Bajanca F, Berki B, Barriga EH, Saint-Jeannet JP, Theveneau E. Paracrine regulation of neural crest EMT by placodal MMP28. PLoS Biol 2023; 21:e3002261. [PMID: 37590318 PMCID: PMC10479893 DOI: 10.1371/journal.pbio.3002261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 09/05/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is an early event in cell dissemination from epithelial tissues. EMT endows cells with migratory, and sometimes invasive, capabilities and is thus a key process in embryo morphogenesis and cancer progression. So far, matrix metalloproteinases (MMPs) have not been considered as key players in EMT but rather studied for their role in matrix remodelling in later events such as cell migration per se. Here, we used Xenopus neural crest cells to assess the role of MMP28 in EMT and migration in vivo. We show that a catalytically active MMP28, expressed by neighbouring placodal cells, is required for neural crest EMT and cell migration. We provide strong evidence indicating that MMP28 is imported in the nucleus of neural crest cells where it is required for normal Twist expression. Our data demonstrate that MMP28 can act as an upstream regulator of EMT in vivo raising the possibility that other MMPs might have similar early roles in various EMT-related contexts such as cancer, fibrosis, and wound healing.
Collapse
Affiliation(s)
- Nadège Gouignard
- Molecular Cellular and Developmental Biology department (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
- New York University, College of Dentistry, Department of Molecular Pathobiology, New York, New York, United States of America
| | - Anne Bibonne
- Molecular Cellular and Developmental Biology department (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - João F. Mata
- Instituto Gulbenkian de Ciência, Mechanisms of Morphogenesis Lab, Oeiras, Portugal
| | - Fernanda Bajanca
- Molecular Cellular and Developmental Biology department (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Bianka Berki
- Molecular Cellular and Developmental Biology department (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Elias H. Barriga
- Instituto Gulbenkian de Ciência, Mechanisms of Morphogenesis Lab, Oeiras, Portugal
| | - Jean-Pierre Saint-Jeannet
- New York University, College of Dentistry, Department of Molecular Pathobiology, New York, New York, United States of America
| | - Eric Theveneau
- Molecular Cellular and Developmental Biology department (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
22
|
Morita K, Shinzato T, Endo Y, Suzuki M, Yoshida H, Sone J, Nagai K. A case of unusual renal manifestation in a patient with neuronal intranuclear inclusion disease treated with steroids. Clin Case Rep 2023; 11:e7730. [PMID: 37564608 PMCID: PMC10410123 DOI: 10.1002/ccr3.7730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/12/2023] [Accepted: 07/06/2023] [Indexed: 08/12/2023] Open
Abstract
Neuronal intranuclear inclusion disease (NIID) is a progressive neurodegenerative disorder characterized by intranuclear inclusions. Kidney injury involvement and successful treatment for NIID have rarely been reported. A NIID patient developed crescentic IgA nephropathy. Steroid therapy resolved digestive symptoms and recovered renal function. Steroids are considered for concomitant symptoms of NIID.
Collapse
Affiliation(s)
- Keisuke Morita
- Department of Nephrology Shizuoka General Hospital Shizuoka Japan
| | | | - Yuzo Endo
- Department of Diagnostic Pathology Shizuoka General Hospital Shizuoka Japan
| | - Makoto Suzuki
- Department of Diagnostic Pathology Shizuoka General Hospital Shizuoka Japan
| | - Hidefumi Yoshida
- Department of Neurology Shizuoka General Hospital Shizuoka Japan
| | - Jun Sone
- Department of Neuropathology, Institute for Medical Science of Aging Aichi Medical University Aichi Japan
| | - Kojiro Nagai
- Department of Nephrology Shizuoka General Hospital Shizuoka Japan
| |
Collapse
|
23
|
Parast SM, Yu D, Chen C, Dickinson AJ, Chang C, Wang H. Recognition of H2AK119ub plays an important role in RSF1-regulated early Xenopus development. Front Cell Dev Biol 2023; 11:1168643. [PMID: 37529237 PMCID: PMC10389277 DOI: 10.3389/fcell.2023.1168643] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/07/2023] [Indexed: 08/03/2023] Open
Abstract
Polycomb group (PcG) proteins are key regulators of gene expression and developmental programs via covalent modification of histones, but the factors that interpret histone modification marks to regulate embryogenesis are less studied. We previously identified Remodeling and Spacing Factor 1 (RSF1) as a reader of histone H2A lysine 119 ubiquitination (H2AK119ub), the histone mark deposited by Polycomb Repressive Complex 1 (PRC1). In the current study, we used Xenopus laevis as a model to investigate how RSF1 affects early embryonic development and whether recognition of H2AK119ub is important for the function of RSF1. We showed that knockdown of Xenopus RSF1, rsf1, not only induced gastrulation defects as reported previously, but specific targeted knockdown in prospective neural precursors induced neural and neural crest defects, with reductions of marker genes. In addition, similar to knockdown of PRC1 components in Xenopus, the anterior-posterior neural patterning was affected in rsf1 knockdown embryos. Binding of H2AK119ub appeared to be crucial for rsf1 function, as a construct with deletion of the UAB domain, which is required for RSF1 to recognize the H2AK119ub nucleosomes, failed to rescue rsf1 morphant embryos and was less effective in interfering with early Xenopus development when ectopically expressed. Furthermore, ectopic deposition of H2AK119ub on the Smad2 target gene gsc using a ring1a-smad2 fusion protein led to ectopic recruitment of RSF1. The fusion protein was inefficient in inducing mesodermal markers in the animal region or a secondary axis when expressed in the ventral tissues. Taken together, our results reveal that rsf1 modulates similar developmental processes in early Xenopus embryos as components of PRC1 do, and that RSF1 acts at least partially through binding to the H2AK119ub mark via the UAB domain during development.
Collapse
Affiliation(s)
- Saeid Mohammad Parast
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Deli Yu
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Chunxu Chen
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Amanda J. Dickinson
- Department of Biology, College of Humanities and Sciences, Virginia Commonwealth University, Richmond, VA, United States
| | - Chenbei Chang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Department of Internal Medicine, Division of Hematology, Oncology and Palliative Care, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
24
|
Liu X, Jones WD, Quesnel-Vallières M, Devadiga SA, Lorent K, Valvezan AJ, Myers RL, Li N, Lengner CJ, Barash Y, Pack M, Klein PS. The Tumor Suppressor Adenomatous Polyposis Coli (apc) Is Required for Neural Crest-Dependent Craniofacial Development in Zebrafish. J Dev Biol 2023; 11:29. [PMID: 37489330 PMCID: PMC10366761 DOI: 10.3390/jdb11030029] [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: 11/29/2022] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 07/26/2023] Open
Abstract
Neural crest (NC) is a unique vertebrate cell type arising from the border of the neural plate and epidermis that gives rise to diverse tissues along the entire body axis. Roberto Mayor and colleagues have made major contributions to our understanding of NC induction, delamination, and migration. We report that a truncating mutation of the classical tumor suppressor Adenomatous Polyposis Coli (apc) disrupts craniofacial development in zebrafish larvae, with a marked reduction in the cranial neural crest (CNC) cells that contribute to mandibular and hyoid pharyngeal arches. While the mechanism is not yet clear, the altered expression of signaling molecules that guide CNC migration could underlie this phenotype. For example, apcmcr/mcr larvae express substantially higher levels of complement c3, which Mayor and colleagues showed impairs CNC cell migration when overexpressed. However, we also observe reduction in stroma-derived factor 1 (sdf1/cxcl12), which is required for CNC migration into the head. Consistent with our previous work showing that APC directly enhances the activity of glycogen synthase kinase 3 (GSK-3) and, independently, that GSK-3 phosphorylates multiple core mRNA splicing factors, we identify 340 mRNA splicing variations in apc mutant zebrafish, including a splice variant that deletes a conserved domain in semaphorin 3f (sema3f), an axonal guidance molecule and a known regulator of CNC migration. Here, we discuss potential roles for apc in CNC development in the context of some of the seminal findings of Mayor and colleagues.
Collapse
Affiliation(s)
- Xiaolei Liu
- Department of Medicine (Hematology-Oncology), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William D. Jones
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mathieu Quesnel-Vallières
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sudhish A. Devadiga
- Faculty of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristin Lorent
- Department of Medicine (Gastroenterology), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexander J. Valvezan
- Department of Medicine (Hematology-Oncology), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rebecca L. Myers
- Department of Medicine (Hematology-Oncology), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ning Li
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher J. Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yoseph Barash
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Pack
- Department of Medicine (Gastroenterology), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter S. Klein
- Department of Medicine (Hematology-Oncology), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
25
|
Hartmann J, Mayor R. Self-organized collective cell behaviors as design principles for synthetic developmental biology. Semin Cell Dev Biol 2023; 141:63-73. [PMID: 35450765 DOI: 10.1016/j.semcdb.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
Over the past two decades, molecular cell biology has graduated from a mostly analytic science to one with substantial synthetic capability. This success is built on a deep understanding of the structure and function of biomolecules and molecular mechanisms. For synthetic biology to achieve similar success at the scale of tissues and organs, an equally deep understanding of the principles of development is required. Here, we review some of the central concepts and recent progress in tissue patterning, morphogenesis and collective cell migration and discuss their value for synthetic developmental biology, emphasizing in particular the power of (guided) self-organization and the role of theoretical advances in making developmental insights applicable in synthesis.
Collapse
Affiliation(s)
- Jonas Hartmann
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
26
|
Alvizi L, Nani D, Brito LA, Kobayashi GS, Passos-Bueno MR, Mayor R. Neural crest E-cadherin loss drives cleft lip/palate by epigenetic modulation via pro-inflammatory gene-environment interaction. Nat Commun 2023; 14:2868. [PMID: 37225711 PMCID: PMC10209087 DOI: 10.1038/s41467-023-38526-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 05/05/2023] [Indexed: 05/26/2023] Open
Abstract
Gene-environment interactions are believed to play a role in multifactorial phenotypes, although poorly described mechanistically. Cleft lip/palate (CLP), the most common craniofacial malformation, has been associated with both genetic and environmental factors, with little gene-environment interaction experimentally demonstrated. Here, we study CLP families harbouring CDH1/E-Cadherin variants with incomplete penetrance and we explore the association of pro-inflammatory conditions to CLP. By studying neural crest (NC) from mouse, Xenopus and humans, we show that CLP can be explained by a 2-hit model, where NC migration is impaired by a combination of genetic (CDH1 loss-of-function) and environmental (pro-inflammatory activation) factors, leading to CLP. Finally, using in vivo targeted methylation assays, we demonstrate that CDH1 hypermethylation is the major target of the pro-inflammatory response, and a direct regulator of E-cadherin levels and NC migration. These results unveil a gene-environment interaction during craniofacial development and provide a 2-hit mechanism to explain cleft lip/palate aetiology.
Collapse
Affiliation(s)
- Lucas Alvizi
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Diogo Nani
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Instituto de Biociencias, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Luciano Abreu Brito
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Instituto de Biociencias, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Gerson Shigeru Kobayashi
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Instituto de Biociencias, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Maria Rita Passos-Bueno
- Centro de Estudos do Genoma Humano e Celulas-Tronco, Departamento de Genetica e Biologia Evolutiva, Instituto de Biociencias, Universidade de Sao Paulo, Sao Paulo, Brazil.
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.
| |
Collapse
|
27
|
Vöcking O, Van Der Meulen K, Patel MK, Famulski JK. Zebrafish anterior segment mesenchyme progenitors are defined by function of tfap2a but not sox10. Differentiation 2023; 130:32-42. [PMID: 36563566 PMCID: PMC10006344 DOI: 10.1016/j.diff.2022.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/18/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022]
Abstract
The anterior segment is a critical component of the visual system. Developing independent of the retina, the AS relies partially on cranial neural crest cells (cNCC) as its earliest progenitors. The cNCCs are thought to first adopt a periocular mesenchyme (POM) fate and subsequently target to the AS upon formation of the rudimentary retina. AS targeted POM is termed anterior segment mesenchyme (ASM). However, it remains unknown when and how the switch from cNCC to POM or POM to ASM takes place. As such, we sought to visualize the timing of these transitions and identify the regulators of this process using the zebrafish embryo model. Using two color fluorescence in situ hybridization, we tracked cNCC and ASM target gene expression from 12 to 24hpf. In doing so, we identified a tfap2a and foxC1a co-expression at 16hpf, identifying the earliest ASM to arrive at the AS. Interestingly, expression of two other key regulators of NCC, foxD3 and sox10 was not associated with early ASM. Functional analysis of tfap2a, foxD3 and sox10 revealed that tfap2a and foxD3 are both critical regulators of ASM specification and AS formation while sox10 was dispensable for either specification or development of the AS. Using genetic knockout lines, we show that in the absence of tfap2a or foxD3 function ASM cells are not specified, and subsequently the AS is malformed. Conversely, sox10 genetic mutants or CRISPR Cas9 injected embryos displayed no defects in ASM specification, migration or the AS. Lastly, using transcriptomic analysis, we show that GFP + cNCCs derived from Tg [foxD3:GFP] and Tg [foxC1b:GFP] share expression profiles consistent with ASM development whereas cNCCs isolated from Tg [sox10:GFP] exhibit expression profiles associated with vasculogenesis, muscle function and pigmentation. Taken together, we propose that the earliest stage of anterior segment mesenchyme (ASM) specification in zebrafish is approximately 16hpf and involves tfap2a/foxC1a positive cNCCs.
Collapse
Affiliation(s)
| | | | - M K Patel
- Department of Biology, University of Kentucky, USA
| | - J K Famulski
- Department of Biology, University of Kentucky, USA.
| |
Collapse
|
28
|
Hernández JA, Chifflet S, Justet C, Torriglia A. A mathematical model of wound healing in bovine corneal endothelium. J Theor Biol 2023; 559:111374. [PMID: 36460056 DOI: 10.1016/j.jtbi.2022.111374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022]
Abstract
We developed a mathematical model to describe healing processes in bovine corneal endothelial (BCE) cells in culture, triggered by mechanical wounds with parallel edges. Previous findings from our laboratory show that, in these cases, BCE monolayers exhibit an approximately constant healing velocity. Also, that caspase-dependent apoptosis occurs, with the fraction of apoptotic cells increasing with the distance traveled by the healing edge. In addition, in this study we report the novel findings that, for wound scratch assays performed preserving the basal extracellular matrix: i) the healing cells increase their en face surface area in a characteristic fashion, and ii) the average length of the segments of the cell columns actively participating in the healing process increases linearly with time. These latter observations preclude the utilization of standard traveling wave formalisms to model wound healing in BCE cells. Instead, we developed and studied a simple phenomenological model based on a plausible formula for the spreading dynamics of the individual healing cells, that incorporates original evidence about the process in BCE cells. The model can be simulated to: i) obtain an approximately constant healing velocity; ii) reproduce the profile of the healing cell areas, and iii) obtain approximately linear time dependences of the mean cell area and average length of the front active segments per column. In view of its accuracy to account for the experimental observations, the model can also be acceptably employed to quantify the appearance of apoptotic cells during BCE wound healing. The strategy utilized here could offer a novel formal framework to represent modifications undergone by some epithelial cell lines during wound healing.
Collapse
Affiliation(s)
- Julio A Hernández
- Sección Biofísica y Biología de Sistemas, Facultad de Ciencias, Universidad de la República, Iguá s/n esq. Mataojo, 11400 Montevideo, Uruguay.
| | - Silvia Chifflet
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, 11800 Montevideo, Uruguay
| | - Cristian Justet
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, 11800 Montevideo, Uruguay
| | - Alicia Torriglia
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, F-75006 Paris, France
| |
Collapse
|
29
|
He S, Wang J, Huang Y, Kong F, Yang R, Zhan Y, Li Z, Ye C, Meng L, Ren Y, Zhou Y, Chen G, Shen Z, Sun S, Zheng S, Dong R. Intestinal fibrosis in aganglionic segment of Hirschsprung's disease revealed by single-cell RNA sequencing. Clin Transl Med 2023; 13:e1193. [PMID: 36738110 PMCID: PMC9898741 DOI: 10.1002/ctm2.1193] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Hirschsprung's disease (HSCR) is a relatively common congenital disability. Accumulating extracellular matrix (ECM) prompts intestinal fibrosis remodelling in the aganglionic segments of HSCR. The contributions of various cellular subsets in the fibrogenesis of HSCR segments are poorly understood. METHODS Single-cell transcriptomics from 8 aganglionic segments and 5 normal segments of 7 HSCR subjects and 26 healthy segments of seven healthy donors were analysed. Fibrotic phenotype and alterations were explored using differential expression analysis and single-cell trajectory analysis. Fibrosis-related transcription factors were inferred through single-cell regulatory network inference. Bulk transcriptomic data, proteomic data, immunohistochemistry (IHC) and real-time polymerase chain reaction were used to validate the alterations in the HSCR intestine. RESULTS Various collagen, fibronectin and laminin protein-coding genes expression were up-regulated in the stromal and glial cells of the HSCR intestine. The number of fibroblasts and myofibroblasts in the aganglionic segments increased, and more myofibroblasts were activated at an earlier stage in HSCR segments, which infers that there is an intestinal fibrosis phenotype in HSCR segments. The fibrotic regulators POSTN, ANXA1 and HSP70 were highly expressed in the ECM-related cellular subsets in the transitional segments and aganglionic segments. The transcription factor regulatory network revealed that fibrosis-related and megacolon-related NR2F1 in the fibroblasts and glial subsets was up-regulated in the aganglionic segment. CONCLUSIONS This work identifies intestinal fibrosis and related regulators in aganglionic segments of HSCR; hence, anti-fibrotic therapy may be considered to prevent HSCR-associated enterocolitis (HAEC), relieve intestinal stricture and improve cell therapy.
Collapse
Affiliation(s)
- Shiwei He
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Junfeng Wang
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Yanlei Huang
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Fanyang Kong
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Ran Yang
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Yong Zhan
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Zifeng Li
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Chunjing Ye
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Lingdu Meng
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Yankang Ren
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Ying Zhou
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Gong Chen
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Zhen Shen
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Song Sun
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Shan Zheng
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| | - Rui Dong
- Department of Pediatric SurgeryShanghai Key Laboratory of Birth DefectChildren's Hospital of Fudan UniversityMinistry of HealthShanghaiChina
| |
Collapse
|
30
|
Margadant C. Cell Migration in Three Dimensions. Methods Mol Biol 2023; 2608:1-14. [PMID: 36653698 DOI: 10.1007/978-1-0716-2887-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cell migration plays an essential role in many pathophysiological processes, including embryonic development, wound healing, immunity, and cancer invasion, and is therefore a widely studied phenomenon in many different fields from basic cell biology to regenerative medicine. During the past decades, a multitude of increasingly complex methods have been developed to study cell migration. Here we compile a series of current state-of-the-art methods and protocols to investigate cell migration in a variety of model systems ranging from cells, organoids, tissue explants, and microfluidic systems to Drosophila, zebrafish, and mice. Together they cover processes as diverse as nuclear deformation, energy consumption, endocytic trafficking, and matrix degradation, as well as tumor vascularization and cancer cell invasion, sprouting angiogenesis, and leukocyte extravasation. Furthermore, methods to study developmental processes such as neural tube closure, germ layer specification, and branching morphogenesis are included, as well as scripts for the automated analysis of several aspects of cell migration. Together, this book constitutes a unique collection of methods of prime importance to those interested in the analysis of cell migration.
Collapse
Affiliation(s)
- Coert Margadant
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
Haage A, Tanentzapf G. Analysis of Integrin-Dependent Melanoblast Migration During Development. Methods Mol Biol 2023; 2608:207-221. [PMID: 36653710 DOI: 10.1007/978-1-0716-2887-4_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The neural crest is a transient embryonic structure that gives rise to a number of important cell types and tissues, including most of the peripheral and enteric nervous systems, pigment-producing skin cells known as melanocytes, and many craniofacial structures. Melanoblasts, the precursors of melanocytes, are derived from the so-called trunk neural crest cells. These cells delaminate and migrate along a dorsolateral pathway to colonize their final destination in the skin, and consequently, defects in melanoblast migration result in pigmentation defects. Studying melanocyte migration is a topic of great interest due to the involvement of melanocytes in highly metastatic skin cancer. A role for integrin-mediated adhesion is well established in neural crest migration, and our recent work has provided direct evidence for a key role for integrin-based adhesion in melanocyte migration. Imaging of melanoblast migration in the context of intact skin has proven to be a particularly powerful tool to study integrin-based adhesion during melanoblast migration. Here, we describe the use of skin explants combined with genetically encoded markers for melanocytes and high-resolution live imaging as a powerful and informative approach to analyze melanoblast migration in an ex vivo context.
Collapse
Affiliation(s)
- Amanda Haage
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA.
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
32
|
Lei F, Xu X, Huang J, Su D, Wan P. Drosophila RhoGAP18B regulates actin cytoskeleton during border cell migration. PLoS One 2023; 18:e0280652. [PMID: 36662713 PMCID: PMC9858088 DOI: 10.1371/journal.pone.0280652] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
Drosophila RhoGAP18B was identified as a negative regulator of small GTPase in the behavioral response to ethanol. However, the effect of RhoGAP18B on cell migration is unknown. Here, we report that RhoGAP18B regulates the migration of border cells in Drosophila ovary. The RhoGAP18B gene produces four transcripts and encodes three translation isoforms. We use different RNAi lines to knockdown each RhoGAP18B isoform, and find that knockdown of RhoGAP18B-PA, but not PC or PD isoform, blocks border cell migration. Knockdown of RhoGAP18B-PA disrupts the asymmetric distribution of F-actin in border cell cluster and increases F-actin level. Furthermore, RhoGAP18B-PA may act on Rac to regulate F-actin organization. Our data indicate that RhoGAP18B shows isoform-specific regulation of border cell migration.
Collapse
Affiliation(s)
- Fengyun Lei
- Laboratory of Molecular Biology, School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Xiaoqing Xu
- Laboratory of Molecular Biology, School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Jianhua Huang
- Laboratory of Molecular Biology, School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Dan Su
- Key Laboratory of Animal Model of TCM Syndromes of Depression, Jiangxi Administration of traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Ping Wan
- Laboratory of Molecular Biology, School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, China
| |
Collapse
|
33
|
Hayot G, Massonot M, Keime C, Faure E, Golzio C. Loss of autism-candidate CHD8 perturbs neural crest development and intestinal homeostatic balance. Life Sci Alliance 2023; 6:e202201456. [PMID: 36375841 PMCID: PMC9664244 DOI: 10.26508/lsa.202201456] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Individuals with mutations in CHD8 present with gastrointestinal complaints, yet the underlying mechanisms are understudied. Here, using a stable constitutive chd8 mutant zebrafish model, we found that the loss of chd8 leads to a reduced number of vagal neural crest cells (NCCs), enteric neural and glial progenitors, emigrating from the neural tube, and that their early migration capability was altered. At later stages, although the intestinal colonization by NCCs was complete, we found the decreased numbers of both serotonin-producing enterochromaffin cells and NCC-derived serotonergic neurons, suggesting an intestinal hyposerotonemia in the absence of chd8 Furthermore, transcriptomic analyses revealed an altered expression of key receptors and enzymes in serotonin and acetylcholine signaling pathways. The tissue examination of chd8 mutants revealed a thinner intestinal epithelium accompanied by an accumulation of neutrophils and the decreased numbers of goblet cells and eosinophils. Last, single-cell sequencing of whole intestines showed a global disruption of the immune balance with a perturbed expression of inflammatory interleukins and changes in immune cell clusters. Our findings propose a causal developmental link between chd8, NCC development, intestinal homeostasis, and autism-associated gastrointestinal complaints.
Collapse
Affiliation(s)
- Gaëlle Hayot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Mathieu Massonot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Elodie Faure
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Christelle Golzio
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| |
Collapse
|
34
|
Charbe NB, Tambuwala M, Palakurthi SS, Warokar A, Hromić‐Jahjefendić A, Bakshi H, Zacconi F, Mishra V, Khadse S, Aljabali AA, El‐Tanani M, Serrano‐Aroca Ã, Palakurthi S. Biomedical applications of three-dimensional bioprinted craniofacial tissue engineering. Bioeng Transl Med 2023; 8:e10333. [PMID: 36684092 PMCID: PMC9842068 DOI: 10.1002/btm2.10333] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 02/06/2023] Open
Abstract
Anatomical complications of the craniofacial regions often present considerable challenges to the surgical repair or replacement of the damaged tissues. Surgical repair has its own set of limitations, including scarcity of the donor tissues, immune rejection, use of immune suppressors followed by the surgery, and restriction in restoring the natural aesthetic appeal. Rapid advancement in the field of biomaterials, cell biology, and engineering has helped scientists to create cellularized skeletal muscle-like structures. However, the existing method still has limitations in building large, highly vascular tissue with clinical application. With the advance in the three-dimensional (3D) bioprinting technique, scientists and clinicians now can produce the functional implants of skeletal muscles and bones that are more patient-specific with the perfect match to the architecture of their craniofacial defects. Craniofacial tissue regeneration using 3D bioprinting can manage and eliminate the restrictions of the surgical transplant from the donor site. The concept of creating the new functional tissue, exactly mimicking the anatomical and physiological function of the damaged tissue, looks highly attractive. This is crucial to reduce the donor site morbidity and retain the esthetics. 3D bioprinting can integrate all three essential components of tissue engineering, that is, rehabilitation, reconstruction, and regeneration of the lost craniofacial tissues. Such integration essentially helps to develop the patient-specific treatment plans and damage site-driven creation of the functional implants for the craniofacial defects. This article is the bird's eye view on the latest development and application of 3D bioprinting in the regeneration of the skeletal muscle tissues and their application in restoring the functional abilities of the damaged craniofacial tissue. We also discussed current challenges in craniofacial bone vascularization and gave our view on the future direction, including establishing the interactions between tissue-engineered skeletal muscle and the peripheral nervous system.
Collapse
Affiliation(s)
- Nitin Bharat Charbe
- Irma Lerma Rangel College of PharmacyTexas A&M Health Science CenterKingsvilleTexasUSA
| | - Murtaza Tambuwala
- School of Pharmacy and Pharmaceutical ScienceUlster UniversityColeraineUK
| | | | - Amol Warokar
- Department of PharmacyDadasaheb Balpande College of PharmacyNagpurIndia
| | - Altijana Hromić‐Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural SciencesInternational University of SarajevoSarajevoBosnia and Herzegovina
| | - Hamid Bakshi
- School of Pharmacy and Pharmaceutical ScienceUlster UniversityColeraineUK
| | - Flavia Zacconi
- Departamento de Quimica Orgánica, Facultad de Química y de FarmaciaPontificia Universidad Católica de ChileSantiagoChile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| | - Vijay Mishra
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
| | - Saurabh Khadse
- Department of Pharmaceutical ChemistryR.C. Patel Institute of Pharmaceutical Education and ResearchDhuleIndia
| | - Alaa A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutical SciencesYarmouk UniversityIrbidJordan
| | - Mohamed El‐Tanani
- Pharmacological and Diagnostic Research Centre, Faculty of PharmacyAl‐Ahliyya Amman UniversityAmmanJordan
| | - Ãngel Serrano‐Aroca
- Biomaterials and Bioengineering Lab Translational Research Centre San Alberto MagnoCatholic University of Valencia San Vicente MártirValenciaSpain
| | - Srinath Palakurthi
- Irma Lerma Rangel College of PharmacyTexas A&M Health Science CenterKingsvilleTexasUSA
| |
Collapse
|
35
|
Zhou J, Yang YJ, Gan RH, Wang Y, Li Z, Zhang XJ, Gui JF, Zhou L. Foxl2a and Foxl2b are involved in midbrain-hindbrain boundary development in zebrafish. Gene Expr Patterns 2022; 46:119286. [PMID: 36341978 DOI: 10.1016/j.gep.2022.119286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/23/2022] [Accepted: 10/24/2022] [Indexed: 11/04/2022]
Abstract
Foxl2 plays conserved central function in ovarian differentiation and maintenance in several fish species. However, its expression pattern and function in fish embryogenesis are still largely unknown. In this study, we first presented a sequential expression pattern of zebrafish foxl2a and foxl2b during embryo development. They were predominantly expressed in the cranial paraxial mesoderm (CPM) and cranial venous vasculature (CVV) during somitogenesis and subsequently expressed in the pharyngeal arches after 48 h post-fertilization (hpf). Then, we compared the brain structures among zebrafish wildtype (WT) and three homozygous foxl2 mutants (foxl2a-/-, foxl2b-/- and foxl2a-/-;foxl2b-/-) and found the reduction of the fourth ventricle in the three foxl2 mutants, especially in foxl2a-/-;foxl2b-/- mutant. Finally, we detected several key transcription factors involved in the gene regulatory network of midbrain-hindbrain boundary (MHB) patterning, such as wnt1, en1b and pax2a. Their expression levels were obviously downregulated in MHB of foxl2a-/- and foxl2a-/-;foxl2b-/- mutants. Thus, we suggest that Foxl2a and Foxl2b are involved in MHB and the fourth ventricle development in zebrafish. The current study provides insights into the molecular mechanism underlying development of brain ventricular system.
Collapse
Affiliation(s)
- Jian Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yan-Jing Yang
- College of Fisheries, Tianjin Agricultural University, China
| | - Rui-Hai Gan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhi Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Juan Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jian-Fang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Hubei Hongshan Laboratory, The Innovation Academy of Seed Design, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
36
|
Comparative Assessment of Computed Tomography and Magnetic Resonance Imaging of Spider Morph and Wild Type Ball Pythons (Python regius) for Evaluation of the Morphological Correlate of Wobble Syndrome. J Comp Pathol 2022; 196:26-40. [DOI: 10.1016/j.jcpa.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/09/2022] [Accepted: 05/26/2022] [Indexed: 11/18/2022]
|
37
|
Candido-Ferreira IL, Lukoseviciute M, Sauka-Spengler T. Multi-layered transcriptional control of cranial neural crest development. Semin Cell Dev Biol 2022; 138:1-14. [PMID: 35941042 DOI: 10.1016/j.semcdb.2022.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/23/2022] [Accepted: 07/23/2022] [Indexed: 11/28/2022]
Abstract
The neural crest (NC) is an emblematic population of embryonic stem-like cells with remarkable migratory ability. These distinctive attributes have inspired the curiosity of developmental biologists for over 150 years, however only recently the regulatory mechanisms controlling the complex features of the NC have started to become elucidated at genomic scales. Regulatory control of NC development is achieved through combinatorial transcription factor binding and recruitment of associated transcriptional complexes to distal cis-regulatory elements. Together, they regulate when, where and to what extent transcriptional programmes are actively deployed, ultimately shaping ontogenetic processes. Here, we discuss how transcriptional networks control NC ontogeny, with a special emphasis on the molecular mechanisms underlying specification of the cephalic NC. We also cover emerging properties of transcriptional regulation revealed in diverse developmental systems, such as the role of three-dimensional conformation of chromatin, and how they are involved in the regulation of NC ontogeny. Finally, we highlight how advances in deciphering the NC transcriptional network have afforded new insights into the molecular basis of human diseases.
Collapse
Affiliation(s)
- Ivan L Candido-Ferreira
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Martyna Lukoseviciute
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK
| | - Tatjana Sauka-Spengler
- University of Oxford, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford OX3 9DS, UK.
| |
Collapse
|
38
|
Kobayashi Y, Nouet J, Baljinnyam E, Siddiqui Z, Fine DH, Fraidenraich D, Kumar VA, Shimizu E. iPSC-derived cranial neural crest-like cells can replicate dental pulp tissue with the aid of angiogenic hydrogel. Bioact Mater 2022; 14:290-301. [PMID: 35310357 PMCID: PMC8897656 DOI: 10.1016/j.bioactmat.2021.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 12/18/2022] Open
Abstract
The dental pulp has irreplaceable roles in maintaining healthy teeth and its regeneration is a primary aim of regenerative endodontics. This study aimed to replicate the characteristics of dental pulp tissue by using cranial neural crest (CNC)-like cells (CNCLCs); these cells were generated by modifying several steps of a previously established method for deriving NC-like cells from induced pluripotent stem cells (iPSCs). CNC is the anterior region of the neural crest in vertebrate embryos, which contains the primordium of dental pulp cells or odontoblasts. The produced CNCLCs showed approximately 2.5–12,000-fold upregulations of major CNC marker genes. Furthermore, the CNCLCs exhibited remarkable odontoblastic differentiation ability, especially when treated with a combination of the fibroblast growth factors (FGFs) FGF4 and FGF9. The FGFs induced odontoblast marker genes by 1.7–5.0-fold, as compared to bone morphogenetic protein 4 (BMP4) treatment. In a mouse subcutaneous implant model, the CNCLCs briefly fated with FGF4 + FGF9 replicated dental pulp tissue characteristics, such as harboring odontoblast-like cells, a dentin-like layer, and vast neovascularization, induced by the angiogenic self-assembling peptide hydrogel (SAPH), SLan. SLan acts as a versatile biocompatible scaffold in the canal space. This study demonstrated a successful collaboration between regenerative medicine and SAPH technology. Cranial neural crest like cells (CNCLCs) were generated by simplifying a previously established method for deriving neural crest-like cells from iPSCs. The produced CNCLCs showed approximately ∼12,000-fold upregulations of major CNC marker genes. The combination of fibroblast growth factors, FGF4 and FGF9, induced the CNCLCs toward odontoblastic differentiation more effectively than BMP4. In a mice subcutaneous implant model, the CNCLCs replicated the characteristics of dental pulp harboring vast neovascularization with the aid of the angiogenic hydrogel, SLan.
Collapse
|
39
|
Canales Coutiño B, Mayor R. Neural crest mechanosensors: Seeing old proteins in a new light. Dev Cell 2022; 57:1792-1801. [PMID: 35901790 DOI: 10.1016/j.devcel.2022.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/26/2022] [Accepted: 07/05/2022] [Indexed: 11/25/2022]
Abstract
Mechanical forces exerted on neural crest cells control their collective migration and differentiation. This perspective discusses our current understanding of neural crest mechanotransduction during cell migration and differentiation. Additionally, we describe proteins that have mechanosensitive functions in other systems, such as mechanosensitive G-protein-coupled receptors, mechanosensitive ion channels, cell-cell adhesion, and cell-matrix-interacting proteins, and highlight that these same proteins have in the past been studied in neural crest development from a purely signaling point of view. We propose that future studies elucidate the mechanosensitive functions these receptors may play in neural crest development and integrate this with their known molecular role.
Collapse
Affiliation(s)
- Brenda Canales Coutiño
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
40
|
Fu J, Zhang X, Zheng H, Yang G, Chen Z, Yuan G. A WWP2-PTEN-KLF5 signaling axis regulates odontoblast differentiation and dentinogenesis in mice. J Biol Chem 2022; 298:102220. [PMID: 35780838 PMCID: PMC9358474 DOI: 10.1016/j.jbc.2022.102220] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/28/2022] Open
Abstract
WW domain–containing E3 Ubiquitin-protein ligase 2 (WWP2) has been found to positively regulate odontoblastic differentiation by monoubiquitinating the transcription factor Kruppel-like factor 5 (KLF5) in a cell culture system. However, the in vivo role of WWP2 in mouse teeth remains unknown. To explore this, here we generated Wwp2 knockout (Wwp2 KO) mice. We found that molars in Wwp2 KO mice exhibited thinner dentin, widened predentin, and reduced numbers of dentinal tubules. In addition, expression of the odontoblast differentiation markers Dspp and Dmp1 was decreased in the odontoblast layers of Wwp2 KO mice. These findings demonstrate that WWP2 may facilitate odontoblast differentiation and dentinogenesis. Furthermore, we show for the first time that phosphatase and tensin homolog (PTEN), a tumor suppressor, is expressed in dental papilla cells and odontoblasts of mouse molars and acts as a negative regulator of odontoblastic differentiation. Further investigation indicated that PTEN is targeted by WWP2 for degradation during odontoblastic differentiation. We demonstrate PTEN physically interacts with and inhibits the transcriptional activity of KLF5 on Dspp and Dmp1. Finally, we found WWP2 was able to suppress the interaction between PTEN and KLF5, which diminished the inhibition effect of PTEN on KLF5. Taken together, this study confirms the essential role of WWP2 and the WWP2–PTEN–KLF5 signaling axis in odontoblast differentiation and dentinogenesis in vivo.
Collapse
Affiliation(s)
- Jing Fu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Xiaobo Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Huiwen Zheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Guobin Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guohua Yuan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
| |
Collapse
|
41
|
Single-cell transcriptomic analysis of neuroepithelial cells and other cell types of the gills of zebrafish (Danio rerio) exposed to hypoxia. Sci Rep 2022; 12:10144. [PMID: 35710785 PMCID: PMC9203529 DOI: 10.1038/s41598-022-13693-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/26/2022] [Indexed: 12/21/2022] Open
Abstract
The fish gill is a multifunctional organ involved in numerous physiological processes, such as gas exchange and sensing of hypoxia by respiratory chemoreceptors, called neuroepithelial cells (NECs). Many studies have focused on zebrafish (Danio rerio) to investigate the structure, function and development of the gills, yet the transcriptomic profile of most gill cells remains obscure. We present the results of a comprehensive transcriptomic analysis of the gills of zebrafish using single-cell RNA sequencing (scRNA‐seq). Gill cells from ETvmat2:EGFP zebrafish were individually labelled before scRNA‐seq library construction using 10× Genomics Chromium technology. 12,819 cells were sequenced with an average depth of over 27,000 reads per cell. We identified a median of 485 genes per cell and 16 cell clusters, including NECs, neurons, pavement cells, endothelial cells and mitochondrion-rich cells. The identity of NECs was confirmed by expression of slc18a2, encoding the vesicular monoamine transporter, Vmat2. Highly differentially-expressed genes in NECs included tph1a, encoding tryptophan hydroxylase, sv2 (synaptic vesicle protein), and proteins implicated in O2 sensing (ndufa4l2a, cox8al and epas1a). In addition, NECs and neurons expressed genes encoding transmembrane receptors for serotonergic, cholinergic or dopaminergic neurotransmission. Differential expression analysis showed a clear shift in the transcriptome of NECs following 14 days of acclimation to hypoxia. NECs in the hypoxia group showed high expression of genes involved in cell cycle control and proliferation. The present article provides a complete cell atlas for the zebrafish gill and serves as a platform for future studies investigating the molecular biology and physiology of this organ.
Collapse
|
42
|
Chen K, Gao H, Yao Y. Prospects of cell chemotactic factors in bone and cartilage tissue engineering. Expert Opin Biol Ther 2022; 22:883-893. [PMID: 35668707 DOI: 10.1080/14712598.2022.2087471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Ke Chen
- Department of Joint Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangdong Key Laboratory of Orthopedic Technology and Implant Materials
| | - Hui Gao
- Department of Joint Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangdong Key Laboratory of Orthopedic Technology and Implant Materials
| | - Yongchang Yao
- Department of Joint Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
- Guangdong Key Laboratory of Orthopedic Technology and Implant Materials
| |
Collapse
|
43
|
Furukawa Y, Odashima A, Hoshino T, Onodera S, Saito A, Ichinohe T, Azuma T. Effects of KnockOut Serum Replacement on Differentiation of Mouse-Induced Pluripotent Stem Cells into Odontoblasts. THE BULLETIN OF TOKYO DENTAL COLLEGE 2022; 63:75-83. [PMID: 35613864 DOI: 10.2209/tdcpublication.2021-0042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Serum serves as a source of rich nutrients during in vitro cell culture, facilitating cell adhesion, growth, and differentiation. When culturing stem cells for transplantation, however, it must be remembered that such culture medium may contain substances potentially harmful to the proposed recipient and may even induce cellular damage. The purpose of this study was to determine whether KnockOut Serum Replacement (KSR), a chemically defined medium supplement, enhanced in vitro differentiation of induced pluripotent stem cells into odontoblasts. Cranial neural crest cells, precursors of odontoblasts, were generated from mouse-induced pluripotent stem cells. They were then cultured in serum-free Dulbecco's modified Eagle's/F12 medium containing fibroblast growth factor 8 with or without KSR. The cells cultured with KSR showed strong proliferation, acquired a spindle-like morphology, and connected with the surrounding cells. KnockOut Serum Replacement also boosted expression of odontoblast markers as measured by qRT-PCR, and increased dentin sialoprotein as assessed by immunostaining. These results confirmed that mouse-induced pluripotent stem cells differentiated into odontoblasts under serum-free conditions, and that KSR enhanced the efficiency of this process.
Collapse
Affiliation(s)
- Yuki Furukawa
- Department of Dental Anesthesiology, Tokyo Dental College
| | - Ayano Odashima
- Department of Dental Anesthesiology, Tokyo Dental College.,Department of Oral Science Center, Tokyo Dental College
| | | | - Shoko Onodera
- Department of Dental Biochemistry, Tokyo Dental College
| | - Akiko Saito
- Department of Dental Biochemistry, Tokyo Dental College
| | | | | |
Collapse
|
44
|
The effect of BMP4, FGF8 and WNT3a on mouse iPS cells differentiating to odontoblast-like cells. Med Mol Morphol 2022; 55:199-209. [PMID: 35578118 DOI: 10.1007/s00795-022-00318-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/23/2022] [Indexed: 10/18/2022]
Abstract
We investigated whether BMP4, FGF8, and/or WNT3a on neural crest-like cells (NCLC) derived from mouse induced pluripotent stem (miPS) cells will promote differentiation of odontoblasts-like cells. After the miPS cells matured into embryonic body (EB) cells, they were cultured in a neural induction medium to produce NCLC. As the differentiation of NCLC were confirmed by RT-qPCR, they were then disassociated and cultured with a medium containing, BMP4, FGF8, and/or WNT3a for 7 and 14 days. The effect of these stimuli on NCLC were assessed by RT-qPCR, ALP staining, and immunocytochemistry. The cultured EB cells presented a significant increase of Snai1, Slug, and Sox 10 substantiating the differentiation of NCLC. NCLC stimulated with more than two stimuli significantly increased the odontoblast markers Dmp-1, Dspp, Nestin, Alp, and Runx2 expression compared to control with no stimulus. The expression of Dmp-1 and Dspp upregulated more when FGF8 was combined with WNT3a. ALP staining was positive in groups containing BMP4 and fluorescence was observed in immunocytochemistry of the common significant groups between Dmp-1 and Dspp. After stimulation, the cell morphology demonstrated a spindle-shaped cells with long projections resembling odontoblasts. Simultaneous BMP4, FGF8, and WNT3a stimuli significantly differentiated NCLC into odontoblast-like cells.
Collapse
|
45
|
Antonaci M, Wheeler GN. MicroRNAs in neural crest development and neurocristopathies. Biochem Soc Trans 2022; 50:965-974. [PMID: 35383827 PMCID: PMC9162459 DOI: 10.1042/bst20210828] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022]
Abstract
The neural crest (NC) is a vertebrate-specific migratory population of multipotent stem cells that originate during late gastrulation in the region between the neural and non-neural ectoderm. This population of cells give rise to a range of derivatives, such as melanocytes, neurons, chondrocytes, chromaffin cells, and osteoblasts. Because of this, failure of NC development can cause a variety of pathologies, often syndromic, that are globally called neurocristopathies. Many genes are known to be involved in NC development, but not all of them have been identified. In recent years, attention has moved from protein-coding genes to non-coding genes, such as microRNAs (miRNA). There is increasing evidence that these non-coding RNAs are playing roles during embryogenesis by regulating the expression of protein-coding genes. In this review, we give an introduction to miRNAs in general and then focus on some miRNAs that may be involved in NC development and neurocristopathies. This new direction of research will give geneticists, clinicians, and molecular biologists more tools to help patients affected by neurocristopathies, as well as broadening our understanding of NC biology.
Collapse
Affiliation(s)
- Marco Antonaci
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR7 7TJ, U.K
| | - Grant N. Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR7 7TJ, U.K
| |
Collapse
|
46
|
Takizawa H, Karakawa A, Suzawa T, Chatani M, Ikeda M, Sakai N, Azetsu Y, Takahashi M, Urano E, Kamijo R, Maki K, Takami M. Neural crest-derived cells possess differentiation potential to keratinocytes in the process of wound healing. Biomed Pharmacother 2021; 146:112593. [PMID: 34968925 DOI: 10.1016/j.biopha.2021.112593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 11/02/2022] Open
Abstract
Neural crest-derived cells (NCDCs), which exist as neural crest cells during the fetal stage and differentiate into palate cells, also exist in adult palate tissues, though with unknown roles. In the present study, NCDCs were labeled with EGFP derived from P0-Cre/CAG-CAT-EGFP (P0-EGFP) double transgenic mice, then their function in palate mucosa wound healing was analyzed. As a palate wound healing model, left-side palate mucosa of P0-EGFP mice was resected, and stem cell markers and keratinocyte markers were detected in healed areas. NCDCs were extracted from normal palate mucosa and precultured with stem cell media for 14 days, then were differentiated into keratinocytes or osteoblasts to analyze pluripotency. The wound healing process started with marginal mucosal regeneration on day two and the entire wound area was lined by regenerated mucosa with EGFP-positive cells (NCDCs) on day 28. EGFP-positive cells comprised approximately 60% of cells in healed oral mucosa, and 65% of those expressed stem cell markers (Sca-1+, PDGFRα+) and 30% expressed a keratinocyte marker (CK13+). In tests of cultured palate mucosa cells, approximately 70% of EGFP-positive cells expressed stem cell markers (Sca-1+, PDGFRα+). Furthermore, under differentiation inducing conditions, cultured EGFP-positive cells were successfully induced to differentiate into keratinocytes and osteoblasts. We concluded that NCDCs exist in adult palate tissues as stem cells and have potential to differentiate into various cell types during the wound healing process.
Collapse
Affiliation(s)
- Hideomi Takizawa
- Department of Orthodontics, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo 145-8515, Japan; Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Akiko Karakawa
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| | - Tetsuo Suzawa
- Department of Biochemistry, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Masahiro Chatani
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Megumi Ikeda
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Division of Endodontology, Department of Conservative Dentistry, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Nobuhiro Sakai
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Yuki Azetsu
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Masahiro Takahashi
- Department of Orthodontics, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo 145-8515, Japan
| | - Eri Urano
- Department of Prosthodontics, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo 145-8515, Japan
| | - Ryutaro Kamijo
- Department of Biochemistry, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Koutaro Maki
- Department of Orthodontics, Showa University School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo 145-8515, Japan
| | - Masamichi Takami
- Department of Pharmacology, Showa University School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| |
Collapse
|
47
|
Čada Š, Bryja V. Local Wnt signalling in the asymmetric migrating vertebrate cells. Semin Cell Dev Biol 2021; 125:26-36. [PMID: 34896020 DOI: 10.1016/j.semcdb.2021.11.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 11/27/2022]
Abstract
Wnt signalling is known to generate cellular asymmetry via Wnt/planar cell polarity pathway (Wnt/PCP). Wnt/PCP acts locally (i) to orient membrane polarity and asymmetric establishment of intercellular junctions via conserved set of PCP proteins most specifically represented by Vangl and Prickle, and (ii) to asymmetrically rearrange cytoskeletal structures via downstream effectors of Dishevelled (Dvl). This process is best described on stable phenotypes of epithelial cells. Here, however, we review the activity of Wnt signalling in migratory cells which experience the extensive rearrangements of cytoskeleton and consequently dynamic asymmetry, making the localised effects of Wnt signalling easier to distinguish. Firstly, we focused on migration of neuronal axons, which allows to study how the pre-existent cellular asymmetry can influence Wnt signalling outcome. Then, we reviewed the role of Wnt signalling in models of mesenchymal migration including neural crest, melanoma, and breast cancer cells. Last, we collected evidence for local Wnt signalling in amoeboid cells, especially lymphocytes. As the outcome of this review, we identify blank spots in our current understanding of this topic, propose models that synthesise the current observations and allow formulation of testable hypotheses for the future research.
Collapse
Affiliation(s)
- Štěpán Čada
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic; Department of Cytokinetics, Institute of Biophysics CAS, Královopolská 135, 61265 Brno, Czech Republic.
| |
Collapse
|
48
|
Xia Z, Bi X, Yang S, Yang X, Song Z, Wei J, Xu P, Rink L, Min J, Wang F. Metal transporter Slc30a1 controls pharyngeal neural crest differentiation via the zinc-Snai2-Jag1 cascade. MedComm (Beijing) 2021; 2:778-797. [PMID: 34977877 PMCID: PMC8706747 DOI: 10.1002/mco2.91] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Abstract
The pharyngeal arch (PA) is a neural crest (NC)-derived organ that is transiently developed during embryogenesis and is required for the subsequent development of various tissues. However, the role of zinc during PA differentiation from NC progenitor cells is unknown. Here, we found that the metal transporters Slc30a1a and Slc30a1b mediate zinc homeostasis during PA differentiation. Slc30a1-deficient zebrafish develop zinc accumulation in NC cells, with increased expression of stemness markers and PA dorsal genes, and SMART-seq analyses revealed that the genes snai2 and jag1b may serve as downstream targets. Furthermore, functional studies showed that knocking down either snai2 or jag1b rescues PA development in Slc30a1-deficient zebrafish. Notably, we identified the double zinc-finger domain in the transcription factor Snai2 as a zinc-responsive element that regulates jag1b expression. Our findings indicate that the Slc30a1/zinc-snai2-jag1b axis is an essential regulatory network controlling PA differentiation, shedding new light on the function of zinc homeostasis in maintaining NC cell stemness and multipotency in vertebrates.
Collapse
Affiliation(s)
- Zhidan Xia
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Xinying Bi
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
- The First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangChina
| | - Sisi Yang
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Xiu Yang
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Zijun Song
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Jiayu Wei
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Pengfei Xu
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Lothar Rink
- Faculty of MedicineInstitute of ImmunologyRWTH Aachen UniversityAachenGermany
| | - Junxia Min
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Fudi Wang
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
- The First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangChina
| |
Collapse
|
49
|
Synthetic Heterocyclic Derivatives as Kinase Inhibitors Tested for the Treatment of Neuroblastoma. Molecules 2021; 26:molecules26237069. [PMID: 34885651 PMCID: PMC8658969 DOI: 10.3390/molecules26237069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/21/2022] Open
Abstract
In the last few years, small molecules endowed with different heterocyclic scaffolds have been developed as kinase inhibitors. Some of them are being tested at preclinical or clinical levels for the potential treatment of neuroblastoma (NB). This disease is the most common extracranial solid tumor in childhood and is responsible for 10% to 15% of pediatric cancer deaths. Despite the availability of some treatments, including the use of very toxic cytotoxic chemotherapeutic agents, high-risk (HR)-NB patients still have a poor prognosis and a survival rate below 50%. For these reasons, new pharmacological options are urgently needed. This review focuses on synthetic heterocyclic compounds published in the last five years, which showed at least some activity on this severe disease and act as kinase inhibitors. The specific mechanism of action, selectivity, and biological activity of these drug candidates are described, when established. Moreover, the most remarkable clinical trials are reported. Importantly, kinase inhibitors approved for other diseases have shown to be active and endowed with lower toxicity compared to conventional cytotoxic agents. The data collected in this article can be particularly useful for the researchers working in this area.
Collapse
|
50
|
Maccarana M, Tykesson E, Pera EM, Gouignard N, Fang J, Malmström A, Ghiselli G, Li JP. Inhibition of iduronic acid biosynthesis by ebselen reduces glycosaminoglycan accumulation in mucopolysaccharidosis type I fibroblasts. Glycobiology 2021; 31:1319-1329. [PMID: 34192316 PMCID: PMC8600295 DOI: 10.1093/glycob/cwab066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/08/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Mucopolysaccharidosis type I (MPS-I) is a rare lysosomal storage disorder caused by deficiency of the enzyme alpha-L-iduronidase, which removes iduronic acid in both chondroitin/dermatan sulfate (CS/DS) and heparan sulfate (HS) and thereby contributes to the catabolism of glycosaminoglycans (GAGs). To ameliorate this genetic defect, the patients are currently treated by enzyme replacement and bone marrow transplantation, which have a number of drawbacks. This study was designed to develop an alternative treatment by inhibition of iduronic acid formation. By screening the Prestwick drug library, we identified ebselen as a potent inhibitor of enzymes that produce iduronic acid in CS/DS and HS. Ebselen efficiently inhibited iduronic acid formation during CS/DS synthesis in cultured fibroblasts. Treatment of MPS-I fibroblasts with ebselen not only reduced accumulation of CS/DS but also promoted GAG degradation. In early Xenopus embryos, this drug phenocopied the effect of downregulation of DS-epimerase 1, the main enzyme responsible for iduronic production in CS/DS, suggesting that ebselen inhibits iduronic acid production in vivo. However, ebselen failed to ameliorate the CS/DS and GAG burden in MPS-I mice. Nevertheless, the results propose a potential of iduronic acid substrate reduction therapy for MPS-I patients.
Collapse
Affiliation(s)
- Marco Maccarana
- Department of Medical Biochemistry and Microbiology, BMC B11, Uppsala University, Husargatan 3 Box 582 751 23 Uppsala, Sweden
- Department of Experimental Medical Science, BMC C12, Lund University, BMC H11, 221 84 Lund, Sweden
| | - Emil Tykesson
- Department of Experimental Medical Science, BMC C12, Lund University, BMC H11, 221 84 Lund, Sweden
| | - Edgar M Pera
- Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, BMC H11, 221 84 Lund, Sweden
| | - Nadège Gouignard
- Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, BMC H11, 221 84 Lund, Sweden
| | - Jianping Fang
- GlycoNovo Technologies Co., Ltd., Shanghai 201203, China
| | - Anders Malmström
- Department of Experimental Medical Science, BMC C12, Lund University, BMC H11, 221 84 Lund, Sweden
| | - Giancarlo Ghiselli
- Glyconova Srl, Parco Scientifico Silvano Fumero, Bioindustry Park Silvano Fumero S.p.A Via Ribes, 5 - 10010 - Colleretto Giacosa (TO), Italy
| | - Jin-ping Li
- Department of Medical Biochemistry and Microbiology, BMC B11, Uppsala University, Husargatan 3 Box 582 751 23 Uppsala, Sweden
| |
Collapse
|