1
|
Sun Y, Ikeuchi Y, Guo F, Hyun I, Ming GL, Fu J. Bioengineering innovations for neural organoids with enhanced fidelity and function. Cell Stem Cell 2025; 32:689-709. [PMID: 40315834 PMCID: PMC12052258 DOI: 10.1016/j.stem.2025.03.014] [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: 08/05/2024] [Revised: 02/19/2025] [Accepted: 03/31/2025] [Indexed: 05/04/2025]
Abstract
Neural organoids have been utilized to recapitulate different aspects of the developing nervous system. While hailed as promising experimental tools for studying human neural development and neuropathology, current neural organoids do not fully recapitulate the anatomy or microcircuitry-level functionality of the developing brain, spinal cord, or peripheral nervous system. In this review, we discuss emerging bioengineering approaches that control morphogen signals and biophysical microenvironments, which have improved the efficiency, fidelity, and utility of neural organoids. Furthermore, advancements in bioengineered tools have facilitated more sophisticated analyses of neural organoid functions and applications, including improved neural-bioelectronic interfaces and organoid-based information processing. Emerging bioethical issues associated with advanced neural organoids are also discussed. Future opportunities of neural organoid research lie in enhancing their fidelity, maturity, and complexity and expanding their applications in a scalable manner.
Collapse
Affiliation(s)
- Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA.
| | - Yoshiho Ikeuchi
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-8654, Japan
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, IN 47408, USA
| | - Insoo Hyun
- Center for Life Sciences and Public Learning, Museum of Science, Boston, MA 02114, USA; Center for Bioethics, Harvard Medical School, Boston, MA 02115, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School of Medicine, Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
2
|
Descoteaux AE, Radulovic M, Alburi D, Bradham CA. CMTM4 is an adhesion modulator that regulates skeletal patterning and primary mesenchyme cell migration in sea urchin embryos. Dev Biol 2025; 521:85-95. [PMID: 39947420 PMCID: PMC11909501 DOI: 10.1016/j.ydbio.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/19/2025]
Abstract
MARVEL proteins, including those of the CMTM gene family, are multi-pass transmembrane proteins that play important roles in vesicular trafficking and cell migration; however, little is understood about their role in development, and their role in skeletal patterning is unexplored. CMTM4 is the only CMTM family member found in the developmental transcriptome of the sea urchin Lytechinus variegatus. Here, we validate that LvCMTM4 is a transmembrane protein and show that perturbation of CMTM4 expression via zygotic morpholino or mRNA injection perturbs skeletal patterning, resulting in loss of secondary skeletal elements and rotational defects. We also demonstrate that normal levels of CMTM4 are required for normal PMC migration and filopodial organization, and that these effects are not due to gross mis-specification of the ectoderm. Finally, we show that CMTM4 is sufficient to mediate mesenchymal cell-cell adhesion. Taken together, these data suggest that CMTM4 controls PMC migration and biomineralization via adhesive regulation during sea urchin skeletogenesis. This is the first discovery of a functionally required adhesive gene in this skeletal patterning system.
Collapse
Affiliation(s)
- Abigail E Descoteaux
- Department of Biology, Boston University, Boston, MA, 02215, United States; Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, MA, 02215, United States; Biological Design Center, Boston University, Boston, MA, 02215, United States
| | - Marko Radulovic
- Department of Biology, Boston University, Boston, MA, 02215, United States; Biological Design Center, Boston University, Boston, MA, 02215, United States
| | - Dona Alburi
- Department of Biology, Boston University, Boston, MA, 02215, United States
| | - Cynthia A Bradham
- Department of Biology, Boston University, Boston, MA, 02215, United States; Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, MA, 02215, United States; Biological Design Center, Boston University, Boston, MA, 02215, United States; Program in Bioinformatics, Boston University, Boston, MA, 02215, United States.
| |
Collapse
|
3
|
van Bree N, Oppelt AS, Lindström S, Zhou L, Boutin L, Coyle B, Swartling FJ, Johnsen JI, Bräutigam L, Wilhelm M. Development of an orthotopic medulloblastoma zebrafish model for rapid drug testing. Neuro Oncol 2025; 27:779-794. [PMID: 39383211 PMCID: PMC11889719 DOI: 10.1093/neuonc/noae210] [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: 03/22/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Medulloblastoma (MB) is one of the most common malignant brain tumors in children. Current preclinical in vivo model systems for MB have increased our understanding of molecular mechanisms regulating MB development. However, they may not be suitable for large-scale studies. The aim of this study was to investigate if a zebrafish-based xenograft model can recapitulate MB growth and enable rapid drug testing. METHODS Nine different MB cell lines or patient-derived cells were transplanted into blastula-stage zebrafish embryos. Tumor development and migration were then monitored using live imaging. RNA sequencing was performed to investigate transcriptome changes after conditioning cells in a neural stem cell-like medium. Furthermore, drug treatments were tested in a 96-well format. RESULTS We demonstrate here that transplantation of MB cells into the blastula stage of zebrafish embryos leads to orthotopic tumor growth that can be observed within 24 h after transplantation. Importantly, the homing of transplanted cells to the hindbrain region and the aggressiveness of tumor growth are enhanced by pre-culturing cells in a neural stem cell-like medium. The change in culture conditions rewires the transcriptome toward a more migratory and neuronal phenotype, including the expression of guidance molecules SEMA3A and EFNB1, both of which correlate with lower overall survival in MB patients. Furthermore, we highlight that the orthotopic zebrafish MB model has the potential to be used for rapid drug testing. CONCLUSIONS Blastula-stage zebrafish MB xenografts present an alternative to current MB mouse xenograft models, enabling quick evaluation of tumor cell growth, neurotropism, and drug efficacy.
Collapse
Affiliation(s)
- Niek van Bree
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Ann-Sophie Oppelt
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | | | - Leilei Zhou
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Lola Boutin
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Beth Coyle
- Children’s Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University Park, University of Nottingham, Nottingham, UK
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - John Inge Johnsen
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Lars Bräutigam
- Comparative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Margareta Wilhelm
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Zhang L, Wei X. The Lego hypothesis of tissue morphogenesis: stereotypic organization of parallel orientational cell adhesions for epithelial self-assembly. Biol Rev Camb Philos Soc 2025; 100:445-460. [PMID: 39308450 PMCID: PMC11718597 DOI: 10.1111/brv.13147] [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: 01/13/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 01/11/2025]
Abstract
How tissues develop distinct structures remains poorly understood. We propose herein the Lego hypothesis of tissue morphogenesis, which states that during tissue morphogenesis, the topographical properties of cell surface adhesion molecules can be dynamically altered and polarised by regulating the spatiotemporal expression and localization of orientational cell adhesion (OCA) molecules cell-autonomously and non-cell-autonomously, thus modulating cells into unique Lego pieces for self-assembling into distinct cytoarchitectures. This concept can be exemplified by epithelial morphogenesis, in which cells are coalesced into a sheet by many types of adhesions. Among them, parallel OCAs (pOCAs) at the lateral cell membranes are essential for configuring cells in parallel. Major pOCAs include Na+/K+-ATPase-mediated adhesions, Crumbs-mediated adhesions, tight junctions, adherens junctions, and desmosomes. These pOCAs align in stereotypical orders along the apical-to-basal axis, and their absolute positioning is also regulated. Such spatial organization of pOCAs underlies proper epithelial morphogenesis. Thus, a key open question about tissue morphogenesis is how to regulate OCAs to make compatible adhesive cellular Lego pieces for tissue construction.
Collapse
Affiliation(s)
- Lili Zhang
- Department of PsychologyDalian Medical University9 Lvshun South Road WestDalian116044Liaoning ProvinceChina
| | - Xiangyun Wei
- Departments of Ophthalmology and Microbiology & Molecular GeneticsUniversity of Pittsburgh1622 Locust StreetPittsburgh15219PAUSA
| |
Collapse
|
5
|
Cho SH, Kim JH, Kim S. Perturbed cell cycle phase-dependent positioning and nuclear migration of retinal progenitors along the apico-basal axis underlie global retinal disorganization in the LCA8-like mouse model. Dev Biol 2025; 517:39-54. [PMID: 39284539 DOI: 10.1016/j.ydbio.2024.09.003] [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: 09/26/2023] [Revised: 08/16/2024] [Accepted: 09/08/2024] [Indexed: 09/29/2024]
Abstract
Combined removal of Crb1 and Crb2 from the developing optic vesicle evokes cellular and laminar disorganization by disrupting the apical cell-cell adhesion in developing retinal epithelium. As a result, at postnatal stages, affected mouse retinas show temporarily thickened, coarsely laminated retinas in addition to functional deficits, including a severely abnormal electroretinogram and decreased visual acuity. These features are reminiscent of Leber congenital amaurosis 8, which is caused in humans by subsets of Crb1 mutations. However, the cellular basis of the abnormalities in retinal progenitor cells (RPCs) that lead to retinal disorganization is largely unknown. In this study, we analyze specific features of RPCs in mutant retinas, including maintenance of the progenitor pool, cell cycle progression, cell cycle phase-dependent nuclear positioning, cell survival, and generation of mature retinal cell types. We find crucial defects in the mutant RPCs. Upon removal of CRB1 and CRB2, apical structures of the RPCs, determined by markers of cilia and centrosomes, are basally shifted. In addition, the positioning of the somata of the M-phase cells, normally localized at the apical surface of the retinal epithelium, is basally shifted in a nearly randomized pattern along the apico-basal axis. Consequently, we propose that positioning of RPCs is desynchronized from cell cycle phase and largely randomized during embryonic development at E17.5. Because the resultant postmitotic cells inevitably lose positional information, the outer and inner nuclear layers (ONL and INL) fail to form from ONBL during neonatal development and retinal cells become mixed locally and globally. Additional results of the lost tissue polarity in Crb1/Crb2 dKO retinas include atypical formation of heterotopic cell patches containing photoreceptor cells in the ganglion cell layer and acellular patches filled with neural processes. Collectively, these changes lead to a mouse model of LCA8-like pathology. LCA8-like pathology differs substantially from the well-characterized, broad range of degeneration phenotypes that arise during the differentiation of photoreceptor and Muller glial cells in retinitis pigmentosa 12, a closely related disease caused by mutated human Crb1. Importantly, the present results suggest that Crb1/Crb2 serve indispensable functions in maintaining cell-cycle phase-dependent positioning of RPCs along the apico-basal axis, regulating cell cycle progression, and maintaining structural laminar integrity without significantly affecting the size of the RPC pools, generation of the subsets of the retinal cell types, or the distribution of cell cycle phases during RPC division. Taken together, these findings provide the crucial cellular basis of the thickening and severely disorganized lamination that are the unique features of the retinal abnormalities in LCA8 patients.
Collapse
Affiliation(s)
- Seo-Hee Cho
- Center for Translational Medicine, Department of Medicine, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Ji Hyang Kim
- Center for Translational Medicine, Department of Medicine, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Seonhee Kim
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
6
|
Jiang N, Hu Z, Wang Q, Hao J, Yang R, Jiang J, Wang H. Fibroblast growth factor 2 enhances BMSC stemness through ITGA2-dependent PI3K/AKT pathway activation. J Cell Physiol 2024; 239:e31423. [PMID: 39188080 DOI: 10.1002/jcp.31423] [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: 03/29/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSC) are promising cellular reservoirs for treating degenerative diseases, tissue injuries, and immune system disorders. However, the stemness of BMSCs tends to decrease during in vitro cultivation, thereby restricting their efficacy in clinical applications. Consequently, investigating strategies that bolster the preservation of BMSC stemness and maximize therapeutic potential is necessary. Transcriptomic and single-cell sequencing methodologies were used to perform a comprehensive examination of BMSCs with the objective of substantiating the pivotal involvement of fibroblast growth factor 2 (FGF2) and integrin alpha 2 (ITGA2) in stemness regulation. To investigate the impact of these genes on the BMSC stemness in vitro, experimental approaches involving loss and gain of function were implemented. These approaches encompassed the modulation of FGF2 and ITGA2 expression levels via small interfering RNA and overexpression plasmids. Furthermore, we examined their influence on the proliferation and differentiation capacities of BMSCs, along with the expression of stemness markers, including octamer-binding transcription factor 4, Nanog homeobox, and sex determining region Y-box 2. Transcriptomic analyzes successfully identified FGF2 and ITGA2 as pivotal genes responsible for regulating the stemness of BMSCs. Subsequent single-cell sequencing revealed that elevated FGF2 and ITGA2 expression levels within specific stem cell subpopulations are closely associated with stemness maintenance. Moreover, additional in vitro experiments have convincingly demonstrated that FGF2 effectively enhances the BMSC stemness by upregulating ITGA2 expression, a process mediated by the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway. This conclusion was supported by the observed upregulation of stemness markers following the induction of FGF2 and ITGA2. Moreover, administration of the BEZ235 pathway inhibitor resulted in the repression of stemness transcription factors, suggesting the substantial involvement of the PI3K/AKT pathway in stemness preservation facilitated by FGF2 and ITGA2. This study elucidates the involvement of FGF2 in augmenting BMSC stemness by modulating ITGA2 and activating the PI3K/AKT pathway. These findings offer valuable contributions to stem cell biology and emphasize the potential of manipulating FGF2 and ITGA2 to optimize BMSCs for therapeutic purposes.
Collapse
Affiliation(s)
- Nizhou Jiang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
- Department of Spine Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhenxin Hu
- Department of Spine Surgery, Peking University Fourth School of Clinical Medicine, Beijing Jishuitan Hospital, Beijing, China
| | - Quanxiang Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Jiayu Hao
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Rui Yang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Jian Jiang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Hong Wang
- Department of Spine Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| |
Collapse
|
7
|
Viola V, Chinnappa K, Francis F. Radial glia progenitor polarity in health and disease. Front Cell Dev Biol 2024; 12:1478283. [PMID: 39416687 PMCID: PMC11479994 DOI: 10.3389/fcell.2024.1478283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Radial glia (RG) are the main progenitor cell type in the developing cortex. These cells are highly polarized, with a long basal process spanning the entire thickness of the cortex and acting as a support for neuronal migration. The RG cell terminates by an endfoot that contacts the pial (basal) surface. A shorter apical process also terminates with an endfoot that faces the ventricle, with a primary cilium protruding in the cerebrospinal fluid. These cell domains have particular subcellular compositions that are critical for the correct functioning of RG. When altered, this can affect proper development of the cortex, ultimately leading to cortical malformations, associated with different pathological outcomes. In this review, we focus on the current knowledge concerning the cell biology of these bipolar stem cells and discuss the role of their polarity in health and disease.
Collapse
Affiliation(s)
- Valeria Viola
- Institut du Fer à Moulin, Paris, France
- Institut National de Santé et de Recherche Médicale (INSERM, UMR-S 1270), Paris, France
- Faculty of Science and Engineering, Sorbonne University, Paris, France
| | - Kaviya Chinnappa
- Institut du Fer à Moulin, Paris, France
- Institut National de Santé et de Recherche Médicale (INSERM, UMR-S 1270), Paris, France
- Faculty of Science and Engineering, Sorbonne University, Paris, France
| | - Fiona Francis
- Institut du Fer à Moulin, Paris, France
- Institut National de Santé et de Recherche Médicale (INSERM, UMR-S 1270), Paris, France
- Faculty of Science and Engineering, Sorbonne University, Paris, France
| |
Collapse
|
8
|
Cui Y, Rolova T, Fagerholm SC. The role of integrins in brain health and neurodegenerative diseases. Eur J Cell Biol 2024; 103:151441. [PMID: 39002282 DOI: 10.1016/j.ejcb.2024.151441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024] Open
Abstract
Integrins are heterodimeric membrane proteins expressed on the surface of most cells. They mediate adhesion and signaling processes relevant for a wealth of physiological processes, including nervous system development and function. Interestingly, integrins are also recognized therapeutic targets for inflammatory diseases, such as multiple sclerosis. Here, we discuss the role of integrins in brain development and function, as well as in neurodegenerative diseases affecting the brain (Alzheimer's disease, multiple sclerosis, stroke). Furthermore, we discuss therapeutic targeting of these adhesion receptors in inflammatory diseases of the brain.
Collapse
Affiliation(s)
- Yunhao Cui
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki 00290, Finland
| | - Susanna C Fagerholm
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland.
| |
Collapse
|
9
|
Thor S. Indirect neurogenesis in space and time. Nat Rev Neurosci 2024; 25:519-534. [PMID: 38951687 DOI: 10.1038/s41583-024-00833-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2024] [Indexed: 07/03/2024]
Abstract
During central nervous system (CNS) development, neural progenitor cells (NPCs) generate neurons and glia in two different ways. In direct neurogenesis, daughter cells differentiate directly into neurons or glia, whereas in indirect neurogenesis, neurons or glia are generated after one or more daughter cell divisions. Intriguingly, indirect neurogenesis is not stochastically deployed and plays instructive roles during CNS development: increased generation of cells from specific lineages; increased generation of early or late-born cell types within a lineage; and increased cell diversification. Increased indirect neurogenesis might contribute to the anterior CNS expansion evident throughout the Bilateria and help to modify brain-region size without requiring increased NPC numbers or extended neurogenesis. Increased indirect neurogenesis could be an evolutionary driver of the gyrencephalic (that is, folded) cortex that emerged during mammalian evolution and might even have increased during hominid evolution. Thus, selection of indirect versus direct neurogenesis provides a powerful developmental and evolutionary instrument that drives not only the evolution of CNS complexity but also brain expansion and modulation of brain-region size, and thereby the evolution of increasingly advanced cognitive abilities. This Review describes indirect neurogenesis in several model species and humans, and highlights some of the molecular genetic mechanisms that control this important process.
Collapse
Affiliation(s)
- Stefan Thor
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
10
|
Sinha S, Huang MS, Mikos G, Bedi Y, Soto L, Lensch S, Ayushman M, Bintu L, Bhutani N, Heilshorn SC, Yang F. Laminin-associated integrins mediate Diffuse Intrinsic Pontine Glioma infiltration and therapy response within a neural assembloid model. Acta Neuropathol Commun 2024; 12:71. [PMID: 38706008 PMCID: PMC11070088 DOI: 10.1186/s40478-024-01765-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/24/2024] [Indexed: 05/07/2024] Open
Abstract
Diffuse Intrinsic Pontine Glioma (DIPG) is a highly aggressive and fatal pediatric brain cancer. One pre-requisite for tumor cells to infiltrate is adhesion to extracellular matrix (ECM) components. However, it remains largely unknown which ECM proteins are critical in enabling DIPG adhesion and migration and which integrin receptors mediate these processes. Here, we identify laminin as a key ECM protein that supports robust DIPG cell adhesion and migration. To study DIPG infiltration, we developed a DIPG-neural assembloid model, which is composed of a DIPG spheroid fused to a human induced pluripotent stem cell-derived neural organoid. Using this assembloid model, we demonstrate that knockdown of laminin-associated integrins significantly impedes DIPG infiltration. Moreover, laminin-associated integrin knockdown improves DIPG response to radiation and HDAC inhibitor treatment within the DIPG-neural assembloids. These findings reveal the critical role of laminin-associated integrins in mediating DIPG progression and drug response. The results also provide evidence that disrupting integrin receptors may offer a novel therapeutic strategy to enhance DIPG treatment outcomes. Finally, these results establish DIPG-neural assembloid models as a powerful tool to study DIPG disease progression and enable drug discovery.
Collapse
Affiliation(s)
- Sauradeep Sinha
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Georgios Mikos
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Yudhishtar Bedi
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, 240 Pasteur Dr., Biomedical Innovation Building 1254, Palo Alto, CA, 94305, USA
| | - Luis Soto
- Department of Radiation Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Sarah Lensch
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Manish Ayushman
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Lacramioara Bintu
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Nidhi Bhutani
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, 240 Pasteur Dr., Biomedical Innovation Building 1254, Palo Alto, CA, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, 476 Lomita Mall, McCullough Building, Room 246, Palo Alto, CA, 94305, USA.
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA.
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, 240 Pasteur Dr., Biomedical Innovation Building 1254, Palo Alto, CA, 94305, USA.
| |
Collapse
|
11
|
Barresi M, Hickmott RA, Bosakhar A, Quezada S, Quigley A, Kawasaki H, Walker D, Tolcos M. Toward a better understanding of how a gyrified brain develops. Cereb Cortex 2024; 34:bhae055. [PMID: 38425213 DOI: 10.1093/cercor/bhae055] [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: 04/24/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 03/02/2024] Open
Abstract
The size and shape of the cerebral cortex have changed dramatically across evolution. For some species, the cortex remains smooth (lissencephalic) throughout their lifetime, while for other species, including humans and other primates, the cortex increases substantially in size and becomes folded (gyrencephalic). A folded cortex boasts substantially increased surface area, cortical thickness, and neuronal density, and it is therefore associated with higher-order cognitive abilities. The mechanisms that drive gyrification in some species, while others remain lissencephalic despite many shared neurodevelopmental features, have been a topic of investigation for many decades, giving rise to multiple perspectives of how the gyrified cerebral cortex acquires its unique shape. Recently, a structurally unique germinal layer, known as the outer subventricular zone, and the specialized cell type that populates it, called basal radial glial cells, were identified, and these have been shown to be indispensable for cortical expansion and folding. Transcriptional analyses and gene manipulation models have provided an invaluable insight into many of the key cellular and genetic drivers of gyrification. However, the degree to which certain biomechanical, genetic, and cellular processes drive gyrification remains under investigation. This review considers the key aspects of cerebral expansion and folding that have been identified to date and how theories of gyrification have evolved to incorporate this new knowledge.
Collapse
Affiliation(s)
- Mikaela Barresi
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
- ACMD, St Vincent's Hospital Melbourne, Regent Street, Fitzroy, VIC 3065, Australia
| | - Ryan Alexander Hickmott
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
- ACMD, St Vincent's Hospital Melbourne, Regent Street, Fitzroy, VIC 3065, Australia
| | - Abdulhameed Bosakhar
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| | - Sebastian Quezada
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| | - Anita Quigley
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
- ACMD, St Vincent's Hospital Melbourne, Regent Street, Fitzroy, VIC 3065, Australia
- School of Engineering, RMIT University, La Trobe Street, Melbourne, VIC 3000, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital, Regent Street, Fitzroy, VIC 3065, Australia
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa 920-8640, Japan
| | - David Walker
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| |
Collapse
|
12
|
Abstract
Gliomas are a diverse group of primary central nervous system tumors that affect both children and adults. Recent studies have revealed a dynamic cross talk that occurs between glioma cells and components of their microenvironment, including neurons, astrocytes, immune cells, and the extracellular matrix. This cross talk regulates fundamental aspects of glioma development and growth. In this review, we discuss recent discoveries about the impact of these interactions on gliomas and highlight how tumor cells actively remodel their microenvironment to promote disease. These studies provide a better understanding of the interactions in the microenvironment that are important in gliomas, offer insight into the cross talk that occurs, and identify potential therapeutic vulnerabilities that can be utilized to improve clinical outcomes.
Collapse
Affiliation(s)
- Maya Anjali Jayaram
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, California, USA;
| | - Joanna J Phillips
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, California, USA;
- Division of Neuropathology, Department of Pathology, University of California, San Francisco, California, USA
| |
Collapse
|
13
|
Valamparamban GF, Spéder P. Homemade: building the structure of the neurogenic niche. Front Cell Dev Biol 2023; 11:1275963. [PMID: 38107074 PMCID: PMC10722289 DOI: 10.3389/fcell.2023.1275963] [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: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 12/19/2023] Open
Abstract
Neural stem/progenitor cells live in an intricate cellular environment, the neurogenic niche, which supports their function and enables neurogenesis. The niche is made of a diversity of cell types, including neurons, glia and the vasculature, which are able to signal to and are structurally organised around neural stem/progenitor cells. While the focus has been on how individual cell types signal to and influence the behaviour of neural stem/progenitor cells, very little is actually known on how the niche is assembled during development from multiple cellular origins, and on the role of the resulting topology on these cells. This review proposes to draw a state-of-the art picture of this emerging field of research, with the aim to expose our knowledge on niche architecture and formation from different animal models (mouse, zebrafish and fruit fly). We will span its multiple aspects, from the existence and importance of local, adhesive interactions to the potential emergence of larger-scale topological properties through the careful assembly of diverse cellular and acellular components.
Collapse
Affiliation(s)
| | - Pauline Spéder
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Structure and Signals in the Neurogenic Niche, Paris, France
| |
Collapse
|
14
|
Zhang L, Wei X. Stepwise modulation of apical orientational cell adhesions for vertebrate neurulation. Biol Rev Camb Philos Soc 2023; 98:2271-2283. [PMID: 37534608 DOI: 10.1111/brv.13006] [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: 10/01/2022] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
Neurulation transforms the neuroectoderm into the neural tube. This transformation relies on reorganising the configurational relationships between the orientations of intrinsic polarities of neighbouring cells. These orientational intercellular relationships are established, maintained, and modulated by orientational cell adhesions (OCAs). Here, using zebrafish (Danio rerio) neurulation as a major model, we propose a new perspective on how OCAs contribute to the parallel, antiparallel, and opposing intercellular relationships that underlie the neural plate-keel-rod-tube transformation, a stepwise process of cell aggregation followed by cord hollowing. We also discuss how OCAs in neurulation may be regulated by various adhesion molecules, including cadherins, Eph/Ephrins, Claudins, Occludins, Crumbs, Na+ /K+ -ATPase, and integrins. By comparing neurulation among species, we reveal that antiparallel OCAs represent a conserved mechanism for the fusion of the neural tube. Throughout, we highlight some outstanding questions regarding OCAs in neurulation. Answers to these questions will help us understand better the mechanisms of tubulogenesis of many tissues.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Psychology, Dalian Medical University, 9 South LvShun Road, Dalian, 116044, China
| | - Xiangyun Wei
- Departments of Ophthalmology, Developmental Biology, and Microbiology & Molecular Genetics, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA
| |
Collapse
|
15
|
Espinosa-Medina I, Feliciano D, Belmonte-Mateos C, Linda Miyares R, Garcia-Marques J, Foster B, Lindo S, Pujades C, Koyama M, Lee T. TEMPO enables sequential genetic labeling and manipulation of vertebrate cell lineages. Neuron 2023; 111:345-361.e10. [PMID: 36417906 DOI: 10.1016/j.neuron.2022.10.035] [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: 01/31/2022] [Revised: 08/15/2022] [Accepted: 10/26/2022] [Indexed: 11/24/2022]
Abstract
During development, regulatory factors appear in a precise order to determine cell fates over time. Consequently, to investigate complex tissue development, it is necessary to visualize and manipulate cell lineages with temporal control. Current strategies for tracing vertebrate cell lineages lack genetic access to sequentially produced cells. Here, we present TEMPO (Temporal Encoding and Manipulation in a Predefined Order), an imaging-readable genetic tool allowing differential labeling and manipulation of consecutive cell generations in vertebrates. TEMPO is based on CRISPR and powered by a cascade of gRNAs that drive orderly activation and inactivation of reporters and/or effectors. Using TEMPO to visualize zebrafish and mouse neurogenesis, we recapitulated birth-order-dependent neuronal fates. Temporally manipulating cell-cycle regulators in mouse cortex progenitors altered the proportion and distribution of neurons and glia, revealing the effects of temporal gene perturbation on serial cell fates. Thus, TEMPO enables sequential manipulation of molecular factors, crucial to study cell-type specification.
Collapse
Affiliation(s)
| | - Daniel Feliciano
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Carla Belmonte-Mateos
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, PRBB, Barcelona 08003, Spain
| | - Rosa Linda Miyares
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Jorge Garcia-Marques
- Centro Nacional de Biotecnologia, Consejo Superior de Investigaciones Cientificas, Madrid 28049, Spain
| | - Benjamin Foster
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Sarah Lindo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Cristina Pujades
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, PRBB, Barcelona 08003, Spain
| | - Minoru Koyama
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON M1C 1A4, Canada
| | - Tzumin Lee
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| |
Collapse
|
16
|
Gerstmann K, Kindbeiter K, Telley L, Bozon M, Reynaud F, Théoulle E, Charoy C, Jabaudon D, Moret F, Castellani V. A balance of noncanonical Semaphorin signaling from the cerebrospinal fluid regulates apical cell dynamics during corticogenesis. SCIENCE ADVANCES 2022; 8:eabo4552. [PMID: 36399562 PMCID: PMC9674300 DOI: 10.1126/sciadv.abo4552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 10/03/2022] [Indexed: 06/01/2023]
Abstract
During corticogenesis, dynamic regulation of apical adhesion is fundamental to generate correct numbers and cell identities. While radial glial cells (RGCs) maintain basal and apical anchors, basal progenitors and neurons detach and settle at distal positions from the apical border. Whether diffusible signals delivered from the cerebrospinal fluid (CSF) contribute to the regulation of apical adhesion dynamics remains fully unknown. Secreted class 3 Semaphorins (Semas) trigger cell responses via Plexin-Neuropilin (Nrp) membrane receptor complexes. Here, we report that unconventional Sema3-Nrp preformed complexes are delivered by the CSF from sources including the choroid plexus to Plexin-expressing RGCs via their apical endfeet. Through analysis of mutant mouse models and various ex vivo assays mimicking ventricular delivery to RGCs, we found that two different complexes, Sema3B/Nrp2 and Sema3F/Nrp1, exert dual effects on apical endfeet dynamics, nuclei positioning, and RGC progeny. This reveals unexpected balance of CSF-delivered guidance molecules during cortical development.
Collapse
Affiliation(s)
- Katrin Gerstmann
- MeLis, CNRS UMR 5284, INSERM U1314, University of Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| | - Karine Kindbeiter
- MeLis, CNRS UMR 5284, INSERM U1314, University of Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| | - Ludovic Telley
- Department of Basic Neuroscience, University of Geneva, 1211 Geneva 4, Switzerland
| | - Muriel Bozon
- MeLis, CNRS UMR 5284, INSERM U1314, University of Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| | - Florie Reynaud
- MeLis, CNRS UMR 5284, INSERM U1314, University of Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| | - Emy Théoulle
- MeLis, CNRS UMR 5284, INSERM U1314, University of Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| | - Camille Charoy
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Denis Jabaudon
- Department of Basic Neuroscience, University of Geneva, 1211 Geneva 4, Switzerland
| | - Frédéric Moret
- MeLis, CNRS UMR 5284, INSERM U1314, University of Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| | - Valerie Castellani
- MeLis, CNRS UMR 5284, INSERM U1314, University of Lyon, Université Claude Bernard Lyon 1, Institut NeuroMyoGène, 8 avenue Rockefeller, 69008 Lyon, France
| |
Collapse
|
17
|
McMillan N, Kirschen GW, Desai S, Xia E, Tsirka SE, Aguirre A. ADAM10 facilitates rapid neural stem cell cycling and proper positioning within the subventricular zone niche via JAMC/RAP1Gap signaling. Neural Regen Res 2022; 17:2472-2483. [PMID: 35535899 PMCID: PMC9120697 DOI: 10.4103/1673-5374.339007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/01/2021] [Accepted: 08/04/2021] [Indexed: 11/23/2022] Open
Abstract
The mechanisms that regulate neural stem cell (NSC) lineage progression and maintain NSCs within different domains of the adult neural stem cell niche, the subventricular zone are not well defined. Quiescent NSCs are arranged at the apical ventricular wall, while mitotically activated NSCs are found in the basal, vascular region of the subventricular zone. Here, we found that ADAM10 (a disintegrin and metalloproteinase 10) is essential in NSC association with the ventricular wall, and via this adhesion to the apical domain, ADAM10 regulates the switch from quiescent and undifferentiated NSC to an actively proliferative and differentiating cell state. Processing of JAMC (junctional adhesion molecule C) by ADAM10 increases Rap1GAP activity. This molecular machinery promotes NSC transit from the apical to the basal compartment and subsequent lineage progression. Understanding the molecular mechanisms responsible for regulating the proper positioning of NSCs within the subventricular zone niche and lineage progression of NSCs could provide new targets for drug development to enhance the regenerative properties of neural tissue.
Collapse
Affiliation(s)
- Nadia McMillan
- Program in Neuroscience and Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Department of Neurology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Gregory W. Kirschen
- Department of Gynecology and Obstetrics, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Sanket Desai
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Emma Xia
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Stella E. Tsirka
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Adan Aguirre
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
18
|
Jury M, Matthiesen I, Rasti Boroojeni F, Ludwig SL, Civitelli L, Winkler TE, Selegård R, Herland A, Aili D. Bioorthogonally Cross-Linked Hyaluronan-Laminin Hydrogels for 3D Neuronal Cell Culture and Biofabrication. Adv Healthc Mater 2022; 11:e2102097. [PMID: 35114074 PMCID: PMC11468931 DOI: 10.1002/adhm.202102097] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/31/2022] [Indexed: 12/13/2022]
Abstract
Laminins (LNs) are key components in the extracellular matrix of neuronal tissues in the developing brain and neural stem cell niches. LN-presenting hydrogels can provide a biologically relevant matrix for the 3D culture of neurons toward development of advanced tissue models and cell-based therapies for the treatment of neurological disorders. Biologically derived hydrogels are rich in fragmented LN and are poorly defined concerning composition, which hampers clinical translation. Engineered hydrogels require elaborate and often cytotoxic chemistries for cross-linking and LN conjugation and provide limited possibilities to tailor the properties of the materials. Here a modular hydrogel system for neural 3D cell cultures, based on hyaluronan and poly(ethylene glycol), that is cross-linked and functionalized with human recombinant LN-521 using bioorthogonal copper-free click chemistry, is shown. Encapsulated human neuroblastoma cells demonstrate high viability and grow into spheroids. Long-term neuroepithelial stem cells (lt-NES) cultured in the hydrogels can undergo spontaneous differentiation to neural fate and demonstrate significantly higher viability than cells cultured without LN. The hydrogels further support the structural integrity of 3D bioprinted structures and maintain high viability of bioprinted and syringe extruded lt-NES, which can facilitate biofabrication and development of cell-based therapies.
Collapse
Affiliation(s)
- Michael Jury
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
| | - Isabelle Matthiesen
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyStockholm100 44Sweden
| | - Fatemeh Rasti Boroojeni
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
| | - Saskia L. Ludwig
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyStockholm100 44Sweden
| | - Livia Civitelli
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
- Nuffield Department of Clinical NeurosciencesJohn Radcliffe HospitalWest WingUniversity of OxfordOxfordOX3 9DUUK
| | - Thomas E. Winkler
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyStockholm100 44Sweden
- Institute of MicrotechnologyCenter of Pharmaceutical EngineeringTechnische Universität BraunschweigBraunschweig38106Germany
| | - Robert Selegård
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
| | - Anna Herland
- Division of Micro and NanosystemsKTH Royal Institute of TechnologyStockholm100 44Sweden
- AIMES, Center for Integrated Medical and Engineering ScienceDepartment of NeuroscienceKarolinska InstituteSolna171 65Sweden
- Division of NanobiotechnologyDepartment of Protein Science, Science for Life LaboratoryKTH Royal Institute of TechnologyStockholm17165Sweden
| | - Daniel Aili
- Laboratory of Molecular MaterialsDivision of Biophysics and BioengineeringDepartment of Physics, Chemistry and BiologyLinköping UniversityLinköping581 83Sweden
| |
Collapse
|
19
|
Divergence between Neuronal and Oligodendroglial Cell Fate, in Postnatal Brain Neural Stem Cells, Leads to Divergent Properties in Polymorphic In Vitro Assays. Cells 2022; 11:cells11111743. [PMID: 35681436 PMCID: PMC9179558 DOI: 10.3390/cells11111743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Two main stem cell pools exist in the postnatal mammalian brain that, although they share some “stemness” properties, also exhibit significant differences. Multipotent neural stem cells survive within specialized microenvironments, called niches, and they are vulnerable to ageing. Oligodendroglial lineage-restricted progenitor cells are widely distributed in the brain parenchyma and are more resistant to the effects of ageing. Here, we create polymorphic neural stem cell cultures and allow cells to progress towards the neuronal and the oligodendroglial lineage. We show that the divergence of cell fate is accompanied by a divergence in the properties of progenitors, which reflects their adaptation to life in the niche or the parenchyma. Neurogenesis shows significant spatial restrictions and a dependence on laminin, a major niche component, while oligodendrogenesis shows none of these constraints. Furthermore, the blocking of integrin-β1 leads to opposing effects, reducing neurogenesis and enhancing oligodendrogenesis. Therefore, polymorphic neural stem cell assays can be used to investigate the divergence of postnatal brain stem cells and also to predict the in vivo effects of potential therapeutic molecules targeting stem and progenitor cells, as we do for the microneurotrophin BNN-20.
Collapse
|
20
|
Schaberg E, Götz M, Faissner A. The extracellular matrix molecule tenascin-C modulates cell cycle progression and motility of adult neural stem/progenitor cells from the subependymal zone. Cell Mol Life Sci 2022; 79:244. [PMID: 35430697 PMCID: PMC9013340 DOI: 10.1007/s00018-022-04259-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 02/16/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022]
Abstract
Adult neurogenesis has been described in two canonical regions of the adult central nervous system (CNS) of rodents, the subgranular zone (SGZ) of the hippocampus and the subependymal zone (SEZ) of the lateral ventricles. The stem cell niche of the SEZ provides a privileged environment composed of a specialized extracellular matrix (ECM) that comprises the glycoproteins tenascin-C (Tnc) and laminin-1 (LN1). In the present study, we investigated the function of these ECM glycoproteins in the adult stem cell niche. Adult neural stem/progenitor cells (aNSPCs) of the SEZ were prepared from wild type (Tnc+/+) and Tnc knockout (Tnc−/−) mice and analyzed using molecular and cell biological approaches. A delayed maturation of aNSPCs in Tnc−/− tissue was reflected by a reduced capacity to form neurospheres in response to epidermal growth factor (EGF). To examine a potential influence of the ECM on cell proliferation, aNSPCs of both genotypes were studied by cell tracking using digital video microscopy. aNSPCs were cultivated on three different substrates, namely, poly-d-lysine (PDL) and PDL replenished with either LN1 or Tnc for up to 6 days in vitro. On each of the three substrates aNSPCs displayed lineage trees that could be investigated with regard to cell cycle length. The latter appeared reduced in Tnc−/− aNSPCs on PDL and LN1 substrates, less so on Tnc that seemed to compensate the absence of the ECM compound to some extent. Close inspection of the lineage trees revealed a subpopulation of late dividing aNSPCslate that engaged into cycling after a notable delay. aNSPCslate exhibited a clearly different morphology, with a larger cell body and conspicuous processes. aNSPCslate reiterated the reduction in cell cycle length on all substrates tested, which was not rescued on Tnc substrates. When the migratory activity of aNSPC-derived progeny was determined, Tnc−/− neuroblasts displayed significantly longer migration tracks. This was traced to an increased rate of migration episodes compared to the wild-type cells that rested for longer time periods. We conclude that Tnc intervenes in the proliferation of aNSPCs and modulates the motility of neuroblasts in the niche of the SEZ.
Collapse
Affiliation(s)
- Elena Schaberg
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, LMU, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Biomedical Center, LMU, Planegg-Martinsried, Germany
- Synergy, Excellence Cluster for Systems Neurology, BMC, LMU, Planegg-Martinsried, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
21
|
Quaresima S, Istiaq A, Jono H, Cacci E, Ohta K, Lupo G. Assessing the Role of Ependymal and Vascular Cells as Sources of Extracellular Cues Regulating the Mouse Ventricular-Subventricular Zone Neurogenic Niche. Front Cell Dev Biol 2022; 10:845567. [PMID: 35450289 PMCID: PMC9016221 DOI: 10.3389/fcell.2022.845567] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Neurogenesis persists in selected regions of the adult mouse brain; among them, the ventricular-subventricular zone (V-SVZ) of the lateral ventricles represents a major experimental paradigm due to its conspicuous neurogenic output. Postnatal V-SVZ neurogenesis is maintained by a resident population of neural stem cells (NSCs). Although V-SVZ NSCs are largely quiescent, they can be activated to enter the cell cycle, self-renew and generate progeny that gives rise to olfactory bulb interneurons. These adult-born neurons integrate into existing circuits to modify cognitive functions in response to external stimuli, but cells shed by V-SVZ NSCs can also reach injured brain regions, suggesting a latent regenerative potential. The V-SVZ is endowed with a specialized microenvironment, which is essential to maintain the proliferative and neurogenic potential of NSCs, and to preserve the NSC pool from exhaustion by finely tuning their quiescent and active states. Intercellular communication is paramount to the stem cell niche properties of the V-SVZ, and several extracellular signals acting in the niche milieu have been identified. An important part of these signals comes from non-neural cell types, such as local vascular cells, ependymal and glial cells. Understanding the crosstalk between NSCs and other niche components may aid therapeutic approaches for neuropathological conditions, since neurodevelopmental disorders, age-related cognitive decline and neurodegenerative diseases have been associated with dysfunctional neurogenic niches. Here, we review recent advances in the study of the complex interactions between V-SVZ NSCs and their cellular niche. We focus on the extracellular cues produced by ependymal and vascular cells that regulate NSC behavior in the mouse postnatal V-SVZ, and discuss the potential implication of these molecular signals in pathological conditions.
Collapse
Affiliation(s)
- Sabrina Quaresima
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Arif Istiaq
- Department of Stem Cell Biology, Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirofumi Jono
- Department of Pharmacy, Kumamoto University Hospital, Kumamoto, Japan
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Emanuele Cacci
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Kunimasa Ohta
- Department of Stem Cell Biology, Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
- *Correspondence: Kunimasa Ohta, ; Giuseppe Lupo,
| | - Giuseppe Lupo
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- *Correspondence: Kunimasa Ohta, ; Giuseppe Lupo,
| |
Collapse
|
22
|
Long KR, Huttner WB. The Role of the Extracellular Matrix in Neural Progenitor Cell Proliferation and Cortical Folding During Human Neocortex Development. Front Cell Neurosci 2022; 15:804649. [PMID: 35140590 PMCID: PMC8818730 DOI: 10.3389/fncel.2021.804649] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular matrix (ECM) has long been known to regulate many aspects of neural development in many different species. However, the role of the ECM in the development of the human neocortex is not yet fully understood. In this review we discuss the role of the ECM in human neocortex development and the different model systems that can be used to investigate this. In particular, we will focus on how the ECM regulates human neural stem and progenitor cell proliferation and differentiation, how the ECM regulates the architecture of the developing human neocortex and the effect of mutations in ECM and ECM-associated genes in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Katherine R. Long
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
23
|
McClenahan F, Dimitriou C, Koutsakis C, Dimitrakopoulos D, Arampatzis A, Kakouri P, Kourla M, Oikonomou S, Andreopoulou E, Patsonis M, Meri DK, Rasool RT, Franklin RJ, Kazanis I. Isolation of neural stem and oligodendrocyte progenitor cells from the brain of live rats. Stem Cell Reports 2021; 16:2534-2547. [PMID: 34560001 PMCID: PMC8514974 DOI: 10.1016/j.stemcr.2021.08.015] [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: 12/08/2019] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/09/2022] Open
Abstract
Postnatal brain neural stem and progenitor cells (NSPCs) cluster in anatomically inaccessible stem cell niches, such as the subependymal zone (SEZ). Here, we describe a method for the isolation of NSPCs from live animals, which we term “milking.” The intracerebroventricular injection of a release cocktail, containing neuraminidase, integrin-β1-blocking antibody, and fibroblast growth factor 2, induces the controlled flow of NSPCs in the cerebrospinal fluid, where they are collected via liquid biopsies. Isolated cells retain key in vivo self-renewal properties and their cell-type profile reflects the cell composition of their source area, while the function of the niche is sustained even 8 months post-milking. By changing the target area more caudally, we also isolate oligodendrocyte progenitor cells (OPCs) from the corpus callosum. This novel approach for sampling NSPCs and OPCs paves the way for performing longitudinal studies in experimental animals, for more in vivo relevant cell culture assays, and for future clinical neuro-regenerative applications. Isolation of brain neural stem and oligodendrocyte progenitor cells from live rats Cells are induced to flow from their niche into the cerebrospinal fluid Neurogenesis persists despite long-term ependymal damage/loss Collected cells retain the properties of endogenous neural stem/progenitor cells
Collapse
Affiliation(s)
- Freyja McClenahan
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK
| | - Christina Dimitriou
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Christos Koutsakis
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | | | - Asterios Arampatzis
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK
| | - Paraskevi Kakouri
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Michaela Kourla
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Sofia Oikonomou
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Evangelia Andreopoulou
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Melina Patsonis
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Danai-Kassandra Meri
- Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Rana-Tahir Rasool
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK
| | - Robin Jm Franklin
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK
| | - Ilias Kazanis
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK; Lab of Developmental Biology, Department of Biology, University of Patras, 26504 Patras, Greece.
| |
Collapse
|
24
|
Ros O, Baudet S, Zagar Y, Loulier K, Roche F, Couvet S, Aghaie A, Atkins M, Louail A, Petit C, Metin C, Mechulam Y, Nicol X. SpiCee: A Genetic Tool for Subcellular and Cell-Specific Calcium Manipulation. Cell Rep 2021; 32:107934. [PMID: 32697983 DOI: 10.1016/j.celrep.2020.107934] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/21/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
Calcium is a second messenger crucial to a myriad of cellular processes ranging from regulation of metabolism and cell survival to vesicle release and motility. Current strategies to directly manipulate endogenous calcium signals lack cellular and subcellular specificity. We introduce SpiCee, a versatile and genetically encoded chelator combining low- and high-affinity sites for calcium. This scavenger enables altering endogenous calcium signaling and functions in single cells in vitro and in vivo with biochemically controlled subcellular resolution. SpiCee paves the way to investigate local calcium signaling in vivo and directly manipulate this second messenger for therapeutic use.
Collapse
Affiliation(s)
- Oriol Ros
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Sarah Baudet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Karine Loulier
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Fiona Roche
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Sandrine Couvet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Alain Aghaie
- INSERM, Sorbonne Université, Institut Pasteur, UMR_S 1120, 75012 Paris, France
| | - Melody Atkins
- INSERM, UMR-S839, Sorbonne Université, Institut du Fer à Moulin, 75005 Paris, France
| | - Alice Louail
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France
| | - Christine Petit
- INSERM, Sorbonne Université, Institut Pasteur, UMR_S 1120, 75012 Paris, France; Collège de France, 75005 Paris, France
| | - Christine Metin
- INSERM, UMR-S839, Sorbonne Université, Institut du Fer à Moulin, 75005 Paris, France
| | - Yves Mechulam
- Laboratoire de Biochimie, Ecole Polytechnique, CNRS UMR 7654, 91128 Palaiseau, France
| | - Xavier Nicol
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, 75012 Paris, France.
| |
Collapse
|
25
|
Guo R, Li J, Chen C, Xiao M, Liao M, Hu Y, Liu Y, Li D, Zou J, Sun D, Torre V, Zhang Q, Chai R, Tang M. Biomimetic 3D bacterial cellulose-graphene foam hybrid scaffold regulates neural stem cell proliferation and differentiation. Colloids Surf B Biointerfaces 2021; 200:111590. [PMID: 33529926 DOI: 10.1016/j.colsurfb.2021.111590] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/29/2020] [Accepted: 01/22/2021] [Indexed: 01/09/2023]
Abstract
Neural stem cell (NSC)-based therapy is a promising candidate for treating neurodegenerative diseases and the preclinical researches call an urgent need for regulating the growth and differentiation of such cells. The recognition that three-dimensional culture has the potential to be a biologically significant system has stimulated an extraordinary impetus for scientific researches in tissue engineering and regenerative medicine. Here, A novel scaffold for culturing NSCs, three-dimensional bacterial cellulose-graphene foam (3D-BC/G), which was prepared via in situ bacterial cellulose interfacial polymerization on the skeleton surface of porous graphene foam has been reported. 3D-BC/G not only supports NSC growth and adhesion, but also maintains NSC stemness and enhances their proliferative capacity. Further phenotypic analysis indicated that 3D-BC/G induces NSCs to selectively differentiate into neurons, forming a neural network in a short amount of time. The scaffold has good biocompatibility with primary cortical neurons enhancing the neuronal network activities. To explore the underlying mechanisms, RNA-Seq analysis to identify genes and signaling pathways was performed and it suggests that 3D-BC/G offers a more promising three-dimensional conductive substrate for NSC research and neural tissue engineering, and the repertoire of gene expression serves as a basis for further studies to better understand NSC biology.
Collapse
Affiliation(s)
- Rongrong Guo
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China; Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China; Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Jian Li
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China
| | - Chuntao Chen
- Institute of Chemicobiology and Functional Materials, Key Laboratory for Soft Chemistry and Functional Materials of Ministry Education, School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Miao Xiao
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, China; International School for Advanced Studies (SISSA), via Bonomea 265, Trieste, 34136, Italy
| | - Menghui Liao
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yangnan Hu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yun Liu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Dan Li
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China; Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China
| | - Jun Zou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu, 215006, China
| | - Dongping Sun
- Institute of Chemicobiology and Functional Materials, Key Laboratory for Soft Chemistry and Functional Materials of Ministry Education, School of Chemical Engineering, Nanjing University of Science and Technology, 200 Xiao Ling Wei Street, Nanjing, 210094, Jiangsu Province, China
| | - Vincent Torre
- International School for Advanced Studies (SISSA), via Bonomea 265, Trieste, 34136, Italy
| | - Qi Zhang
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China; Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China; Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China.
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, School of Life Sciences and Technology, Southeast University, Nanjing, 210096, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China; Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
26
|
McDonald WS, Miyamoto K, Rivera R, Kennedy G, Almeida BSV, Kingsbury MA, Chun J. Altered cleavage plane orientation with increased genomic aneuploidy produced by receptor-mediated lysophosphatidic acid (LPA) signaling in mouse cerebral cortical neural progenitor cells. Mol Brain 2020; 13:169. [PMID: 33317583 PMCID: PMC7734743 DOI: 10.1186/s13041-020-00709-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/02/2020] [Indexed: 01/03/2023] Open
Abstract
The brain is composed of cells having distinct genomic DNA sequences that arise post-zygotically, known as somatic genomic mosaicism (SGM). One form of SGM is aneuploidy-the gain and/or loss of chromosomes-which is associated with mitotic spindle defects. The mitotic spindle orientation determines cleavage plane positioning and, therefore, neural progenitor cell (NPC) fate during cerebral cortical development. Here we report receptor-mediated signaling by lysophosphatidic acid (LPA) as a novel extracellular signal that influences cleavage plane orientation and produces alterations in SGM by inducing aneuploidy during murine cortical neurogenesis. LPA is a bioactive lipid whose actions are mediated by six G protein-coupled receptors, LPA1-LPA6. RNAscope and qPCR assessment of all six LPA receptor genes, and exogenous LPA exposure in LPA receptor (Lpar)-null mice, revealed involvement of Lpar1 and Lpar2 in the orientation of the mitotic spindle. Lpar1 signaling increased non-vertical cleavage in vivo by disrupting cell-cell adhesion, leading to breakdown of the ependymal cell layer. In addition, genomic alterations were significantly increased after LPA exposure, through production of chromosomal aneuploidy in NPCs. These results identify LPA as a receptor-mediated signal that alters both NPC fate and genomes during cortical neurogenesis, thus representing an extracellular signaling mechanism that can produce stable genomic changes in NPCs and their progeny. Normal LPA signaling in early life could therefore influence both the developing and adult brain, whereas its pathological disruption could contribute to a range of neurological and psychiatric diseases, via long-lasting somatic genomic alterations.
Collapse
Affiliation(s)
- Whitney S McDonald
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA, 92037, USA.,The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Kyoko Miyamoto
- The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Richard Rivera
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA, 92037, USA.,The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Grace Kennedy
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA, 92037, USA.,The Scripps Research Institute, La Jolla, CA, 92037, USA
| | | | | | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA, 92037, USA. .,The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
27
|
Amin S, Borrell V. The Extracellular Matrix in the Evolution of Cortical Development and Folding. Front Cell Dev Biol 2020; 8:604448. [PMID: 33344456 PMCID: PMC7744631 DOI: 10.3389/fcell.2020.604448] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/12/2020] [Indexed: 02/02/2023] Open
Abstract
The evolution of the mammalian cerebral cortex leading to humans involved a remarkable sophistication of developmental mechanisms. Specific adaptations of progenitor cell proliferation and neuronal migration mechanisms have been proposed to play major roles in this evolution of neocortical development. One of the central elements influencing neocortex development is the extracellular matrix (ECM). The ECM provides both a structural framework during tissue formation and to present signaling molecules to cells, which directly influences cell behavior and movement. Here we review recent advances in the understanding of the role of ECM molecules on progenitor cell proliferation and neuronal migration, and how these contribute to cerebral cortex expansion and folding. We discuss how transcriptomic studies in human, ferret and mouse identify components of ECM as being candidate key players in cortex expansion during development and evolution. Then we focus on recent functional studies showing that ECM components regulate cortical progenitor cell proliferation, neuron migration and the mechanical properties of the developing cortex. Finally, we discuss how these features differ between lissencephalic and gyrencephalic species, and how the molecular evolution of ECM components and their expression profiles may have been fundamental in the emergence and evolution of cortex folding across mammalian phylogeny.
Collapse
Affiliation(s)
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d’Alacant, Spain
| |
Collapse
|
28
|
Ferent J, Zaidi D, Francis F. Extracellular Control of Radial Glia Proliferation and Scaffolding During Cortical Development and Pathology. Front Cell Dev Biol 2020; 8:578341. [PMID: 33178693 PMCID: PMC7596222 DOI: 10.3389/fcell.2020.578341] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/08/2020] [Indexed: 01/14/2023] Open
Abstract
During the development of the cortex, newly generated neurons migrate long-distances in the expanding tissue to reach their final positions. Pyramidal neurons are produced from dorsal progenitors, e.g., radial glia (RGs) in the ventricular zone, and then migrate along RG processes basally toward the cortex. These neurons are hence dependent upon RG extensions to support their migration from apical to basal regions. Several studies have investigated how intracellular determinants are required for RG polarity and subsequent formation and maintenance of their processes. Fewer studies have identified the influence of the extracellular environment on this architecture. This review will focus on extracellular factors which influence RG morphology and pyramidal neuronal migration during normal development and their perturbations in pathology. During cortical development, RGs are present in different strategic positions: apical RGs (aRGs) have their cell bodies located in the ventricular zone with an apical process contacting the ventricle, while they also have a basal process extending radially to reach the pial surface of the cortex. This particular conformation allows aRGs to be exposed to long range and short range signaling cues, whereas basal RGs (bRGs, also known as outer RGs, oRGs) have their cell bodies located throughout the cortical wall, limiting their access to ventricular factors. Long range signals impacting aRGs include secreted molecules present in the embryonic cerebrospinal fluid (e.g., Neuregulin, EGF, FGF, Wnt, BMP). Secreted molecules also contribute to the extracellular matrix (fibronectin, laminin, reelin). Classical short range factors include cell to cell signaling, adhesion molecules and mechano-transduction mechanisms (e.g., TAG1, Notch, cadherins, mechanical tension). Changes in one or several of these components influencing the RG extracellular environment can disrupt the development or maintenance of RG architecture on which neuronal migration relies, leading to a range of cortical malformations. First, we will detail the known long range signaling cues impacting RG. Then, we will review how short range cell contacts are also important to instruct the RG framework. Understanding how RG processes are structured by their environment to maintain and support radial migration is a critical part of the investigation of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Julien Ferent
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| | - Donia Zaidi
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| | - Fiona Francis
- Inserm, U 1270, Paris, France.,Sorbonne University, UMR-S 1270, IFM, Paris, France.,Institut du Fer á Moulin, Paris, France
| |
Collapse
|
29
|
SponGee: A Genetic Tool for Subcellular and Cell-Specific cGMP Manipulation. Cell Rep 2020; 27:4003-4012.e6. [PMID: 31242429 DOI: 10.1016/j.celrep.2019.05.102] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 03/22/2019] [Accepted: 05/29/2019] [Indexed: 12/22/2022] Open
Abstract
cGMP is critical to a variety of cellular processes, but the available tools to interfere with endogenous cGMP lack cellular and subcellular specificity. We introduce SponGee, a genetically encoded chelator of this cyclic nucleotide that enables in vitro and in vivo manipulations in single cells and in biochemically defined subcellular compartments. SponGee buffers physiological changes in cGMP concentration in various model systems while not affecting cAMP signals. We provide proof-of-concept strategies by using this tool to highlight the role of cGMP signaling in vivo and in discrete subcellular domains. SponGee enables the investigation of local cGMP signals in vivo and paves the way for therapeutic strategies that prevent downstream signaling activation.
Collapse
|
30
|
Arreguin AJ, Colognato H. Brain Dysfunction in LAMA2-Related Congenital Muscular Dystrophy: Lessons From Human Case Reports and Mouse Models. Front Mol Neurosci 2020; 13:118. [PMID: 32792907 PMCID: PMC7390928 DOI: 10.3389/fnmol.2020.00118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/09/2020] [Indexed: 12/26/2022] Open
Abstract
Laminin α2 gene (LAMA2)-related Congenital Muscular Dystrophy (CMD) was distinguished by a defining central nervous system (CNS) abnormality—aberrant white matter signals by MRI—when first described in the 1990s. In the past 25 years, researchers and clinicians have expanded our knowledge of brain involvement in LAMA2-related CMD, also known as Congenital Muscular Dystrophy Type 1A (MDC1A). Neurological changes in MDC1A can be structural, including lissencephaly and agyria, as well as functional, including epilepsy and intellectual disability. Mouse models of MDC1A include both spontaneous and targeted LAMA2 mutations and range from a partial loss of LAMA2 function (e.g., dy2J/dy2J), to a complete loss of LAMA2 expression (dy3K/dy3K). Diverse cellular and molecular changes have been reported in the brains of MDC1A mouse models, including blood-brain barrier dysfunction, altered neuro- and gliogenesis, changes in synaptic plasticity, and decreased myelination, providing mechanistic insight into potential neurological dysfunction in MDC1A. In this review article, we discuss selected studies that illustrate the potential scope and complexity of disturbances in brain development in MDC1A, and as well as highlight mechanistic insights that are emerging from mouse models.
Collapse
Affiliation(s)
- Andrea J Arreguin
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States.,Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY, United States
| | - Holly Colognato
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
31
|
Glotzbach K, Stamm N, Weberskirch R, Faissner A. Hydrogels Derivatized With Cationic Moieties or Functional Peptides as Efficient Supports for Neural Stem Cells. Front Neurosci 2020; 14:475. [PMID: 32508574 PMCID: PMC7251306 DOI: 10.3389/fnins.2020.00475] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
The increasing incidence of neurodegenerative diseases such as Alzheimer's or Parkinson's disease represents a significant burden for patients and national health systems. The conditions are primarily caused by the death of neurons and other neural cell types. One important aim of current stem cell research is to find a way to replace the lost cells. In this perspective, neural stem cells (NSCs) have been considered as a promising tool in the field of regenerative medicine. The behavior of NSCs is modulated by environmental influences, for example hormones, growth factors, cytokines, and extracellular matrix molecules or biomechanics. These factors can be studied by using well-defined hydrogels, which are polymeric networks of synthetic or natural origin with the ability to swell in water. These gels can be modified with a variety of molecules and optimized with regard to their mechanical properties to mimic the natural extracellular environment. In particular modifications applying distinct units such as functional domains and peptides can modulate the development of NSCs with regard to proliferation, differentiation and migration. One well-known peptide sequence that affects the behavior of NSCs is the integrin recognition sequence RGD that has originally been derived from fibronectin. In the present review we provide an overview concerning the applications of modified hydrogels with an emphasis on synthetic hydrogels based on poly(acrylamides), as modified with either cationic moieties or the peptide sequence RGD. This knowledge might be used in tissue engineering and regenerative medicine for the therapy of spinal cord injuries, neurodegenerative diseases and traumata.
Collapse
Affiliation(s)
- Kristin Glotzbach
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Nils Stamm
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Ralf Weberskirch
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
32
|
Brązert M, Kranc W, Celichowski P, Jankowski M, Piotrowska-Kempisty H, Pawelczyk L, Bruska M, Zabel M, Nowicki M, Kempisty B. Expression of genes involved in neurogenesis, and neuronal precursor cell proliferation and development: Novel pathways of human ovarian granulosa cell differentiation and transdifferentiation capability in vitro. Mol Med Rep 2020; 21:1749-1760. [PMID: 32319615 PMCID: PMC7057781 DOI: 10.3892/mmr.2020.10972] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/10/2019] [Indexed: 01/17/2023] Open
Abstract
The process of neural tissue formation is associated primarily with the course of neurogenesis during embryonic life. The source of neural-like cells is stem cells, which, under the influence of appropriate differentiating factors, may differentiate/transdifferentiate towards a neural-like lineage. The present study suggested that, under long-term in vitro culture conditions, human ovarian granulosa cells (GCs), obtained from granulosa-rich follicular fluid, acquired new properties and expressed genes characteristic of the ontological groups ‘neurogenesis’ (GO:0022008), ‘neuronal precursor cell proliferation’ (GO:0061351) and ‘nervous system development’ (GO:0007399), which are closely related to the formation of neurons. The present study collected GCs from 20 women referred for the procedure of in vitro fertilization. Cells were maintained in long-term in vitro culture for 30 days, and RNA was isolated after 1, 7, 15 and 30 days of culture. The expression profile of individual genes was determined using the Affymetrix microarray method. The 131 genes with the highest expression change in relation to day 1 of culture were then selected; the 10 most affected genes found to be primarily involved in nerve cell formation processes were chosen for consideration in this study: CLDN11, OXTR, DFNA5, ATP8B1, ITGA3, CD9, FRY, NANOS1, CRIM1 and NTN4. The results of the present study revealed that these genes may be considered potential markers of the uninduced differentiation potential of GCs. In addition, it was suggested that GCs may be used to develop a cell line showing neuronal characteristics after 30 days of cultivation. In addition, due to their potential, these cells could possibly be used in the treatment of neurodegenerative diseases, not only in the form of ‘cultured neurons’ but also as producers of factors involved in the regeneration of the nervous system.
Collapse
Affiliation(s)
- Maciej Brązert
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznań University of Medical Sciences, 60‑535 Poznań, Poland
| | - Wiesława Kranc
- Department of Anatomy, Poznań University of Medical Sciences, 60‑781 Poznań, Poland
| | - Piotr Celichowski
- Department of Histology and Embryology, Poznań University of Medical Sciences, 60‑781 Poznań, Poland
| | - Maurycy Jankowski
- Department of Anatomy, Poznań University of Medical Sciences, 60‑781 Poznań, Poland
| | | | - Leszek Pawelczyk
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznań University of Medical Sciences, 60‑535 Poznań, Poland
| | - Małgorzata Bruska
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznań University of Medical Sciences, 60‑535 Poznań, Poland
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wrocław Medical University, 50‑368 Wrocław, Poland
| | - Michał Nowicki
- Department of Histology and Embryology, Poznań University of Medical Sciences, 60‑781 Poznań, Poland
| | - Bartosz Kempisty
- Department of Anatomy, Poznań University of Medical Sciences, 60‑781 Poznań, Poland
| |
Collapse
|
33
|
Güven A, Kalebic N, Long KR, Florio M, Vaid S, Brandl H, Stenzel D, Huttner WB. Extracellular matrix-inducing Sox9 promotes both basal progenitor proliferation and gliogenesis in developing neocortex. eLife 2020; 9:49808. [PMID: 32191207 PMCID: PMC7105383 DOI: 10.7554/elife.49808] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
Neocortex expansion is largely based on the proliferative capacity of basal progenitors (BPs), which is increased by extracellular matrix (ECM) components via integrin signaling. Here we show that the transcription factor Sox9 drives expression of ECM components and that laminin 211 increases BP proliferation in embryonic mouse neocortex. We show that Sox9 is expressed in human and ferret BPs and is required for BP proliferation in embryonic ferret neocortex. Conditional Sox9 expression in the mouse BP lineage, where it normally is not expressed, increases BP proliferation, reduces Tbr2 levels and induces Olig2 expression, indicative of premature gliogenesis. Conditional Sox9 expression also results in cell-non-autonomous stimulation of BP proliferation followed by increased upper-layer neuron production. Our findings demonstrate that Sox9 exerts concerted effects on transcription, BP proliferation, neuron production, and neurogenic vs. gliogenic BP cell fate, suggesting that Sox9 may have contributed to promote neocortical expansion.
Collapse
Affiliation(s)
- Ayse Güven
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Human Technopole, Milan, Italy
| | - Katherine R Long
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marta Florio
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Holger Brandl
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Denise Stenzel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
34
|
Veeraval L, O'Leary CJ, Cooper HM. Adherens Junctions: Guardians of Cortical Development. Front Cell Dev Biol 2020; 8:6. [PMID: 32117958 PMCID: PMC7025593 DOI: 10.3389/fcell.2020.00006] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/10/2020] [Indexed: 12/01/2022] Open
Abstract
Apical radial glia comprise the pseudostratified neuroepithelium lining the embryonic lateral ventricles and give rise to the extensive repertoire of pyramidal neuronal subtypes of the neocortex. The establishment of a highly apicobasally polarized radial glial morphology is a mandatory prerequisite for cortical development as it governs neurogenesis, neural migration and the integrity of the ventricular wall. As in all epithelia, cadherin-based adherens junctions (AJs) play an obligate role in the maintenance of radial glial apicobasal polarity and neuroepithelial cohesion. In addition, the assembly of resilient AJs is critical to the integrity of the neuroepithelium which must resist the tensile forces arising from increasing CSF volume and other mechanical stresses associated with the expansion of the ventricles in the embryo and neonate. Junctional instability leads to the collapse of radial glial morphology, disruption of the ventricular surface and cortical lamination defects due to failed neuronal migration. The fidelity of cortical development is therefore dependent on AJ assembly and stability. Mutations in genes known to control radial glial junction formation are causative for a subset of inherited cortical malformations (neuronal heterotopias) as well as perinatal hydrocephalus, reinforcing the concept that radial glial junctions are pivotal determinants of successful corticogenesis. In this review we explore the key animal studies that have revealed important insights into the role of AJs in maintaining apical radial glial morphology and function, and as such, have provided a deeper understanding of the aberrant molecular and cellular processes contributing to debilitating cortical malformations. We highlight the reciprocal interactions between AJs and the epithelial polarity complexes that impose radial glial apicobasal polarity. We also discuss the critical molecular networks promoting AJ assembly in apical radial glia and emphasize the role of the actin cytoskeleton in the stabilization of cadherin adhesion – a crucial factor in buffering the mechanical forces exerted as a consequence of cortical expansion.
Collapse
Affiliation(s)
- Lenin Veeraval
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Conor J O'Leary
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
35
|
Ahmed M, Marziali LN, Arenas E, Feltri ML, Ffrench-Constant C. Laminin α2 controls mouse and human stem cell behaviour during midbrain dopaminergic neuron development. Development 2019; 146:dev.172668. [PMID: 31371375 DOI: 10.1242/dev.172668] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 07/24/2019] [Indexed: 01/16/2023]
Abstract
Development of the central nervous system requires coordination of the proliferation and differentiation of neural stem cells. Here, we show that laminin alpha 2 (lm-α2) is a component of the midbrain dopaminergic neuron (mDA) progenitor niche in the ventral midbrain (VM) and identify a concentration-dependent role for laminin α2β1γ1 (lm211) in regulating mDA progenitor proliferation and survival via a distinct set of receptors. At high concentrations, lm211-rich environments maintain mDA progenitors in a proliferative state via integrins α6β1 and α7β1, whereas low concentrations of lm211 support mDA lineage survival via dystroglycan receptors. We confirmed our findings in vivo, demonstrating that the VM was smaller in the absence of lm-α2, with increased apoptosis; furthermore, the progenitor pool was depleted through premature differentiation, resulting in fewer mDA neurons. Examination of mDA neuron subtype composition showed a reduction in later-born mDA neurons of the ventral tegmental area, which control a range of cognitive behaviours. Our results identify a novel role for laminin in neural development and provide a possible mechanism for autism-like behaviours and the brainstem hypoplasia seen in some individuals with mutations of LAMA2.
Collapse
Affiliation(s)
- Maqsood Ahmed
- MRC Centre of Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Leandro N Marziali
- Departments of Biochemistry and Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Ernest Arenas
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm 17177, Sweden
| | - M Laura Feltri
- Departments of Biochemistry and Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | | |
Collapse
|
36
|
Penisson M, Ladewig J, Belvindrah R, Francis F. Genes and Mechanisms Involved in the Generation and Amplification of Basal Radial Glial Cells. Front Cell Neurosci 2019; 13:381. [PMID: 31481878 PMCID: PMC6710321 DOI: 10.3389/fncel.2019.00381] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/05/2019] [Indexed: 12/22/2022] Open
Abstract
The development of the cerebral cortex relies on different types of progenitor cell. Among them, the recently described basal radial glial cell (bRG) is suggested to be of critical importance for the development of the brain in gyrencephalic species. These cells are highly numerous in primate and ferret brains, compared to lissencephalic species such as the mouse in which they are few in number. Their somata are located in basal subventricular zones in gyrencephalic brains and they generally possess a basal process extending to the pial surface. They sometimes also have an apical process directed toward the ventricular surface, similar to apical radial glial cells (aRGs) from which they are derived, and whose somata are found more apically in the ventricular zone. bRGs share similarities with aRGs in terms of gene expression (SOX2, PAX6, and NESTIN), whilst also expressing a range of more specific genes (such as HOPX). In primate brains, bRGs can divide multiple times, self-renewing and/or generating intermediate progenitors and neurons. They display a highly specific cytokinesis behavior termed mitotic somal translocation. We focus here on recently identified molecular mechanisms associated with the generation and amplification of bRGs, including bRG-like cells in the rodent. These include signaling pathways such as the FGF-MAPK cascade, SHH, PTEN/AKT, PDGF pathways, and proteins such as INSM, GPSM2, ASPM, TRNP1, ARHGAP11B, PAX6, and HIF1α. A number of these proteins were identified through transcriptome comparisons in human aRGs vs. bRGs, and validated by modifying their activities or expression levels in the mouse. This latter experiment often revealed enhanced bRG-like cell production, even in some cases generating folds (gyri) on the surface of the mouse cortex. We compare the features of the identified cells and methods used to characterize them in each model. These important data converge to indicate pathways essential for the production and expansion of bRGs, which may help us understand cortical development in health and disease.
Collapse
Affiliation(s)
- Maxime Penisson
- Inserm, Institut du Fer à Moulin, Sorbonne Université, Paris, France.,Inserm UMR-S 1270, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Julia Ladewig
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research (gGmbH), Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Richard Belvindrah
- Inserm, Institut du Fer à Moulin, Sorbonne Université, Paris, France.,Inserm UMR-S 1270, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Fiona Francis
- Inserm, Institut du Fer à Moulin, Sorbonne Université, Paris, France.,Inserm UMR-S 1270, Paris, France.,Institut du Fer à Moulin, Paris, France
| |
Collapse
|
37
|
Morante-Redolat JM, Porlan E. Neural Stem Cell Regulation by Adhesion Molecules Within the Subependymal Niche. Front Cell Dev Biol 2019; 7:102. [PMID: 31245371 PMCID: PMC6581678 DOI: 10.3389/fcell.2019.00102] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/27/2019] [Indexed: 12/14/2022] Open
Abstract
In the mammalian adult brain, neural stem cells persist in neurogenic niches. The subependymal zone is the most prolific neurogenic niche in adult rodents, where residing stem cells generate large numbers of immature neurons that migrate into the olfactory bulb, where they differentiate into different types of interneurons. Subependymal neural stem cells derive from embryonic radial glia and retain some of their features like apico-basal polarity, with apical processes piercing the ependymal layer, and a basal process contacting blood vessels, constituting an epithelial niche. Conservation of the cytoarchitecture of the niche is of crucial importance for the maintenance of stem cells and for their neurogenic potential. In this minireview we will focus on extracellular matrix and adhesion molecules in the adult subependymal zone, showing their involvement not only as structural elements sustaining the niche architecture and topology, but also in the maintenance of stemness and regulation of the quiescence-proliferation balance.
Collapse
Affiliation(s)
- Jose Manuel Morante-Redolat
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universitat de València, Burjassot, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina, Universitat de València, Burjassot, Spain
| | - Eva Porlan
- Departamento de Neuropatología Molecular, Centro de Biología Molecular Severo Ochoa Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
38
|
Angiogenic potential of co-spheroids of neural stem cells and endothelial cells in injectable gelatin-based hydrogel. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 99:140-149. [DOI: 10.1016/j.msec.2019.01.089] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/12/2018] [Accepted: 01/18/2019] [Indexed: 12/20/2022]
|
39
|
Lee TJ, Nair M, Banasavadi-Siddegowda Y, Liu J, Nallanagulagari T, Jaime-Ramirez AC, Guo JY, Quadri H, Zhang J, Bockhorst KH, Aghi MK, Carbonell WS, Kaur B, Yoo JY. Enhancing Therapeutic Efficacy of Oncolytic Herpes Simplex Virus-1 with Integrin β1 Blocking Antibody OS2966. Mol Cancer Ther 2019; 18:1127-1136. [PMID: 30926634 DOI: 10.1158/1535-7163.mct-18-0953] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/01/2018] [Accepted: 03/20/2019] [Indexed: 02/06/2023]
Abstract
Integrin β1 receptor, expressed on the surface of tumor cells and macrophages in the tumor microenvironment (TME), has been implicated in both tumor progression and resistance to multiple modalities of therapy. OS2966 is the first clinical-ready humanized monoclonal antibody to block integrin β1 and was recently orphan designated by the FDA Office of Orphan Products Development. Here, we tested therapeutic potential of OS2966-mediated integrin β1 blockade to enhance the efficacy of oncolytic herpes simplex virus-1 (oHSV) through evaluation of virus replication, tumor cell killing efficiency, effect on the antiviral signaling pathway, co-culture assays of oHSV-infected cells with macrophages, and in vivo bioluminescence imaging on mammary fat pad triple-negative breast cancer xenograft and subcutaneous and intracranial glioma xenografts. OS2966 treatment decreased interferon signaling and proinflammatory cytokine induction in oHSV-treated tumor cells and inhibited migration of macrophages, resulting in enhanced oHSV replication and cytotoxicity. OS2966 treatment also significantly enhanced oHSV replication and oHSV-mediated antitumor efficacy in orthotopic xenograft models, including triple-negative breast cancer and glioblastoma. The results demonstrated the synergistic potential of the combinatory treatment approach with OS2966 to improve antitumor efficacy of conventional oHSV therapy.
Collapse
Affiliation(s)
- Tae Jin Lee
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Mitra Nair
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Yeshavanth Banasavadi-Siddegowda
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas.,Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, Maryland
| | - Joseph Liu
- Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Tejaswini Nallanagulagari
- Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Biochemistry and Microbiology Majors, The Ohio State University, Columbus, Ohio
| | - Alena Cristina Jaime-Ramirez
- Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Jeffrey Yunhua Guo
- Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Biology Major, The Ohio State University, Columbus, Ohio
| | | | - Jianying Zhang
- Center for Biostatistics, Department of Biomedical Informatics, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Kurt H Bockhorst
- Department of Diagnostic and Interventional Imaging, University of Texas Health Science Center at Houston, Houston, Texas
| | - Manish K Aghi
- University of California at San Francisco, California
| | | | - Balveen Kaur
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Ji Young Yoo
- The Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
40
|
Kalebic N, Gilardi C, Stepien B, Wilsch-Bräuninger M, Long KR, Namba T, Florio M, Langen B, Lombardot B, Shevchenko A, Kilimann MW, Kawasaki H, Wimberger P, Huttner WB. Neocortical Expansion Due to Increased Proliferation of Basal Progenitors Is Linked to Changes in Their Morphology. Cell Stem Cell 2019; 24:535-550.e9. [PMID: 30905618 DOI: 10.1016/j.stem.2019.02.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/14/2018] [Accepted: 02/22/2019] [Indexed: 11/24/2022]
Abstract
The evolutionary expansion of the mammalian neocortex (Ncx) is thought to be linked to increased proliferative capacity of basal progenitors (BPs) and their neurogenic capacity. Here, by quantifying BP morphology in the developing Ncx of mouse, ferret, and human, we show that increased BP proliferative capacity is linked to an increase in BP process number. We identify human membrane-bound PALMDELPHIN (PALMD-Caax) as an underlying factor, and we show that it drives BP process growth and proliferation when expressed in developing mouse and ferret Ncx. Conversely, CRISPR/Cas9-mediated disruption of PALMD or its binding partner ADDUCIN-γ in fetal human Ncx reduces BP process numbers and proliferation. We further show that PALMD-induced processes enable BPs to receive pro-proliferative integrin-dependent signals. These findings provide a link between BP morphology and proliferation, suggesting that changes in BP morphology may have contributed to the evolutionary expansion of the Ncx.
Collapse
Affiliation(s)
- Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Carlotta Gilardi
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Barbara Stepien
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Katherine R Long
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marta Florio
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Barbara Langen
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Benoit Lombardot
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Anna Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Pauline Wimberger
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
41
|
Cell migration promotes dynamic cellular interactions to control cerebral cortex morphogenesis. Nat Rev Neurosci 2019; 20:318-329. [DOI: 10.1038/s41583-019-0148-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
42
|
Nirwane A, Yao Y. Laminins and their receptors in the CNS. Biol Rev Camb Philos Soc 2019; 94:283-306. [PMID: 30073746 DOI: 10.1111/brv.12454] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 01/24/2023]
Abstract
Laminin, an extracellular matrix protein, is widely expressed in the central nervous system (CNS). By interacting with integrin and non-integrin receptors, laminin exerts a large variety of important functions in the CNS in both physiological and pathological conditions. Due to the existence of many laminin isoforms and their differential expression in various cell types in the CNS, the exact functions of each individual laminin molecule in CNS development and homeostasis remain largely unclear. In this review, we first briefly introduce the structure and biochemistry of laminins and their receptors. Next, the dynamic expression of laminins and their receptors in the CNS during both development and in adulthood is summarized in a cell-type-specific manner, which allows appreciation of their functional redundancy/compensation. Furthermore, we discuss the biological functions of laminins and their receptors in CNS development, blood-brain barrier (BBB) maintenance, neurodegeneration, stroke, and neuroinflammation. Last, key challenges and potential future research directions are summarized and discussed. Our goals are to provide a synthetic review to stimulate future studies and promote the formation of new ideas/hypotheses and new lines of research in this field.
Collapse
Affiliation(s)
- Abhijit Nirwane
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA 30602, U.S.A
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA 30602, U.S.A
| |
Collapse
|
43
|
Long KR, Huttner WB. How the extracellular matrix shapes neural development. Open Biol 2019; 9:180216. [PMID: 30958121 PMCID: PMC6367132 DOI: 10.1098/rsob.180216] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/11/2018] [Indexed: 12/17/2022] Open
Abstract
During development, both cells and tissues must acquire the correct shape to allow their proper function. This is especially relevant in the nervous system, where the shape of individual cell processes, such as the axons and dendrites, and the shape of entire tissues, such as the folding of the neocortex, are highly specialized. While many aspects of neural development have been uncovered, there are still several open questions concerning the mechanisms governing cell and tissue shape. In this review, we discuss the role of the extracellular matrix (ECM) in these processes. In particular, we consider how the ECM regulates cell shape, proliferation, differentiation and migration, and more recent work highlighting a key role of ECM in the morphogenesis of neural tissues.
Collapse
Affiliation(s)
- Katherine R. Long
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| |
Collapse
|
44
|
Venkei ZG, Yamashita YM. Emerging mechanisms of asymmetric stem cell division. J Cell Biol 2018; 217:3785-3795. [PMID: 30232100 PMCID: PMC6219723 DOI: 10.1083/jcb.201807037] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/06/2018] [Accepted: 09/13/2018] [Indexed: 01/10/2023] Open
Abstract
Venkei and Yamashita summarize recent advances in our understanding of asymmetric stem cell division in tissue homeostasis. The asymmetric cell division of stem cells, which produces one stem cell and one differentiating cell, has emerged as a mechanism to balance stem cell self-renewal and differentiation. Elaborate cellular mechanisms that orchestrate the processes required for asymmetric cell divisions are often shared between stem cells and other asymmetrically dividing cells. During asymmetric cell division, cells must establish asymmetry/polarity, which is guided by varying degrees of intrinsic versus extrinsic cues, and use intracellular machineries to divide in a desired orientation in the context of the asymmetry/polarity. Recent studies have expanded our knowledge on the mechanisms of asymmetric cell divisions, revealing the previously unappreciated complexity in setting up the cellular and/or environmental asymmetry, ensuring binary outcomes of the fate determination. In this review, we summarize recent progress in understanding the mechanisms and regulations of asymmetric stem cell division.
Collapse
Affiliation(s)
- Zsolt G Venkei
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Yukiko M Yamashita
- Life Sciences Institute, University of Michigan, Ann Arbor, MI .,Department of Cell and Developmental Biology, Medical School, University of Michigan, Ann Arbor, MI.,Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI
| |
Collapse
|
45
|
Abstract
Distinct micro-environmental properties have been reported to be essential for maintenance of neural precursor cells (NPCs) within the adult brain. Due to high complexity and technical limitations, the natural niche can barely be studied systematically in vivo. By reconstituting selected environmental properties (adhesiveness, proteolytic degradability, and elasticity) in geldrop cultures, we show that NPCs can be maintained stably at high density over an extended period of time (up to 8 days). In both conventional systems, neurospheres and monolayer cultures, they would expand and (in the case of neurospheres) differentiate rapidly. Further, we report a critical dualism between matrix adhesiveness and degradability. Only if both features are functional NPCs stay proliferative. Lastly, Rho-associated protein kinase was identified as part of a pivotal intracellular signaling cascade controlling cell morphology in response to environmental cues inside geldrop cultures. Our findings demonstrate that simple manipulations of the microenvironment in vitro result in an important preservation of stemness features in the cultured precursor cells.
Collapse
|
46
|
Andreopoulou E, Arampatzis A, Patsoni M, Kazanis I. Being a Neural Stem Cell: A Matter of Character But Defined by the Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:81-118. [PMID: 29204830 DOI: 10.1007/978-3-319-69194-7_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cells that build the nervous system, either this is a small network of ganglia or a complicated primate brain, are called neural stem and progenitor cells. Even though the very primitive and the very recent neural stem cells (NSCs) share common basic characteristics that are hard-wired within their character, such as the expression of transcription factors of the SoxB family, their capacity to give rise to extremely different neural tissues depends significantly on instructions from the microenvironment. In this chapter we explore the nature of the NSC microenvironment, looking through evolution, embryonic development, maturity and even disease. Experimental work undertaken over the last 20 years has revealed exciting insight into the NSC microcosmos. NSCs are very capable in producing their own extracellular matrix and in regulating their behaviour in an autocrine and paracrine manner. Nevertheless, accumulating evidence indicates an important role for the vasculature, especially within the NSC niches of the postnatal brain; while novel results reveal direct links between the metabolic state of the organism and the function of NSCs.
Collapse
Affiliation(s)
- Evangelia Andreopoulou
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Asterios Arampatzis
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK
- School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Melina Patsoni
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Ilias Kazanis
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece.
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
47
|
Gong L, Cao L, Shen Z, Shao L, Gao S, Zhang C, Lu J, Li W. Materials for Neural Differentiation, Trans-Differentiation, and Modeling of Neurological Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1705684. [PMID: 29573284 DOI: 10.1002/adma.201705684] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/04/2017] [Indexed: 05/02/2023]
Abstract
Neuron regeneration from pluripotent stem cells (PSCs) differentiation or somatic cells trans-differentiation is a promising approach for cell replacement in neurodegenerative diseases and provides a powerful tool for investigating neural development, modeling neurological diseases, and uncovering the mechanisms that underlie diseases. Advancing the materials that are applied in neural differentiation and trans-differentiation promotes the safety, efficiency, and efficacy of neuron regeneration. In the neural differentiation process, matrix materials, either natural or synthetic, not only provide a structural and biochemical support for the monolayer or three-dimensional (3D) cultured cells but also assist in cell adhesion and cell-to-cell communication. They play important roles in directing the differentiation of PSCs into neural cells and modeling neurological diseases. For the trans-differentiation of neural cells, several materials have been used to make the conversion feasible for future therapy. Here, the most current applications of materials for neural differentiation for PSCs, neuronal trans-differentiation, and neurological disease modeling is summarized and discussed.
Collapse
Affiliation(s)
- Lulu Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Lining Cao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhenmin Shen
- The VIP Department, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Li Shao
- The VIP Department, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Shaorong Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jianfeng Lu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
48
|
Cross-talk between blood vessels and neural progenitors in the developing brain. Neuronal Signal 2018; 2:NS20170139. [PMID: 32714582 PMCID: PMC7371013 DOI: 10.1042/ns20170139] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/20/2018] [Accepted: 02/21/2018] [Indexed: 01/26/2023] Open
Abstract
The formation of the central nervous system (CNS) involves multiple cellular and molecular interactions between neural progenitor cells (NPCs) and blood vessels to establish extensive and complex neural networks and attract a vascular supply that support their function. In this review, we discuss studies that have performed genetic manipulations of chick, fish and mouse embryos to define the spatiotemporal roles of molecules that mediate the reciprocal regulation of NPCs and blood vessels. These experiments have highlighted core functions of NPC-expressed ligands in initiating vascular growth into and within the neural tube as well as establishing the blood-brain barrier. More recent findings have also revealed indispensable roles of blood vessels in regulating NPC expansion and eventual differentiation, and specific regional differences in the effect of angiocrine signals. Accordingly, NPCs initially stimulate blood vessel growth and maturation to nourish the brain, but blood vessels subsequently also regulate NPC behaviour to promote the formation of a sufficient number and diversity of neural cells. A greater understanding of the molecular cross-talk between NPCs and blood vessels will improve our knowledge of how the vertebrate nervous system forms and likely help in the design of novel therapies aimed at regenerating neurons and neural vasculature following CNS disease or injury.
Collapse
|
49
|
Valiente M, Ahluwalia MS, Boire A, Brastianos PK, Goldberg SB, Lee EQ, Le Rhun E, Preusser M, Winkler F, Soffietti R. The Evolving Landscape of Brain Metastasis. Trends Cancer 2018; 4:176-196. [PMID: 29506669 PMCID: PMC6602095 DOI: 10.1016/j.trecan.2018.01.003] [Citation(s) in RCA: 188] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 11/24/2022]
Abstract
Metastasis, involving the spread of systemic cancer to the brain, results in neurologic disability and death. Current treatments are largely palliative in nature; improved therapeutic approaches represent an unmet clinical need. However, recent experimental and clinical advances challenge the bleak long-term outcome of this disease. Encompassing key recent findings in epidemiology, genetics, microenvironment, leptomeningeal disease, neurocognition, targeted therapy, immunotherapy, and prophylaxis, we review preclinical and clinical studies to provide a comprehensive picture of contemporary research and the management of secondary brain tumors.
Collapse
Affiliation(s)
- Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Center (CNIO), Melchor Fernández Almagro 3, Madrid, Spain.
| | - Manmeet S Ahluwalia
- Brain Metastasis Research Program, Burkhardt Brain Tumor and Neuro-Oncology Center, Department of Medicine, Cleveland Clinic, Neurological Institute, 9500 Euclid Avenue, 44195 Cleveland, OH, USA
| | - Adrienne Boire
- Department of Neurology, Human Oncology and Pathogenesis Program, Brain Tumor Center, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, 10065 New York, NY, USA
| | - Priscilla K Brastianos
- Division of Hematology/Oncology, Department of Medicine; Division of Neuro-Oncology, Department of Neurology; Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street Boston, 02114 Boston, MA, USA
| | - Sarah B Goldberg
- Department of Medicine (Medical Oncology), Yale School of Medicine, 333 Cedar Street, New Haven, CT, USA
| | - Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, 02215 Boston, MA, USA
| | - Emilie Le Rhun
- Neuro-Oncology, Department of Neurosurgery, University Hospital Lille, Salengro Hospital, Rue Emile Laine, 59037 Lille, France; Neurology, Department of Medical Oncology, Oscar Lambret Center, 59020 Lille, France; Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1192, Villeneuve d'Ascq, France; Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | - Matthias Preusser
- Department of Medicine I, Comprehensive Cancer Center Vienna, CNS Unit (CCC-CNS), Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, and Clinical Cooperation Unit Neurooncology, German Cancer Research Center, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University Hospital Turin, Via Cherasco 15, 10126 Turin, Italy.
| |
Collapse
|
50
|
Agirman G, Broix L, Nguyen L. Cerebral cortex development: an outside‐in perspective. FEBS Lett 2017; 591:3978-3992. [DOI: 10.1002/1873-3468.12924] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Gulistan Agirman
- GIGA‐Neurosciences Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) Liège Belgium
| | - Loïc Broix
- GIGA‐Neurosciences Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) Liège Belgium
| | - Laurent Nguyen
- GIGA‐Neurosciences Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) Liège Belgium
| |
Collapse
|