51
|
Basement Membranes in the Worm: A Dynamic Scaffolding that Instructs Cellular Behaviors and Shapes Tissues. CURRENT TOPICS IN MEMBRANES 2015; 76:337-71. [PMID: 26610919 DOI: 10.1016/bs.ctm.2015.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The nematode worm Caenorhabditis elegans has all the major basement membrane proteins found in vertebrates, usually with a smaller gene family encoding each component. With its powerful forward genetics, optical clarity, simple tissue organization, and the capability to functionally tag most basement membrane components with fluorescent proteins, C. elegans has facilitated novel insights into the assembly and function of basement membranes. Although basement membranes are generally thought of as static structures, studies in C. elegans have revealed their active properties and essential functions in tissue formation and maintenance. Here, we review discoveries from C. elegans development that highlight dynamic aspects of basement membrane assembly, function, and regulation during organ growth, tissue polarity, cell migration, cell invasion, and tissue attachment. These studies have helped transform our view of basement membranes from static support structures to dynamic scaffoldings that play broad roles in regulating tissue organization and cellular behavior that are essential for development and have important implications in human diseases.
Collapse
|
52
|
Polisetti N, Zenkel M, Menzel-Severing J, Kruse FE, Schlötzer-Schrehardt U. Cell Adhesion Molecules and Stem Cell-Niche-Interactions in the Limbal Stem Cell Niche. Stem Cells 2015; 34:203-19. [PMID: 26349477 DOI: 10.1002/stem.2191] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/02/2015] [Indexed: 12/19/2022]
Abstract
Interactions between stem cells and their microenvironment are critical for regulation and maintenance of stem cell function. To elucidate the molecular interactions within the human limbal epithelial stem/progenitor cell (LEPC) niche, which is essential for maintaining corneal transparency and vision, we performed a comprehensive expression analysis of cell adhesion molecules (CAMs) using custom-made quantitative real-time polymerase chain reaction (qRT-PCR) arrays and laser capture-microdissected LEPC clusters, comprising LEPCs, melanocytes, mesenchymal cells, and transmigrating immune cells. We show that LEPCs are anchored to their supporting basement membrane by the laminin receptors α3β1 and α6β4 integrin and the dystroglycan complex, while intercellular contacts between LEPCs and melanocytes are mediated by N-, P-, and E-cadherin together with L1-CAM, a member of the immunoglobulin superfamily (Ig)CAMs. In addition to the LEPC-associated heparan sulfate proteoglycans syndecan-2, glypican-3, and glypican-4, the IgCAM members ICAM-1 and VCAM-1 were found to be variably expressed on LEPCs and associated niche cells and to be dynamically regulated in response to chemokines such as interferon-γ to enhance interactions with immune cells. Moreover, junctional adhesion molecule JAM-C accumulating in the subepithelial limbal matrix, appeared to be involved in recruitment of immune cells, while mesenchymal stromal cells appeared to use the nephronectin receptor integrin α8 for approaching the limbal basement membrane. In summary, we identified a novel combination of cell surface receptors that may regulate both stable and dynamic cell-matrix and cell-cell interactions within the limbal niche. The findings provide a solid foundation for further functional studies and for advancement of our current therapeutic strategies for ocular surface reconstruction.
Collapse
Affiliation(s)
- Naresh Polisetti
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Zenkel
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Menzel-Severing
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Friedrich E Kruse
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ursula Schlötzer-Schrehardt
- Department of Ophthalmology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
53
|
Quantifying Molecular-Level Cell Adhesion on Electroactive Conducting Polymers using Electrochemical-Single Cell Force Spectroscopy. Sci Rep 2015; 5:13334. [PMID: 26335299 PMCID: PMC4558606 DOI: 10.1038/srep13334] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 07/21/2015] [Indexed: 01/28/2023] Open
Abstract
Single Cell Force Spectroscopy was combined with Electrochemical-AFM to quantify the adhesion between live single cells and conducting polymers whilst simultaneously applying a voltage to electrically switch the polymer from oxidized to reduced states. The cell-conducting polymer adhesion represents the non-specific interaction between cell surface glycocalyx molecules and polymer groups such as sulfonate and dodecylbenzene groups, which rearrange their orientation during electrical switching. Single cell adhesion significantly increases as the polymer is switched from an oxidized to fully reduced state, indicating stronger cell binding to sulfonate groups as opposed to hydrophobic groups. This increase in single cell adhesion is concomitant with an increase in surface hydrophilicity and uptake of cell media, driven by cation movement, into the polymer film during electrochemical reduction. Binding forces between the glycocalyx and polymer surface are indicative of molecular-level interactions and during electrical stimulation there is a decrease in both the binding force and stiffness of the adhesive bonds. The study provides insight into the effects of electrochemical switching on cell adhesion at the cell-conducting polymer interface and is more broadly applicable to elucidating the binding of cell adhesion molecules in the presence of electrical fields and directly at electrode interfaces.
Collapse
|
54
|
Wong FK, Fei JF, Mora-Bermúdez F, Taverna E, Haffner C, Fu J, Anastassiadis K, Stewart AF, Huttner WB. Sustained Pax6 Expression Generates Primate-like Basal Radial Glia in Developing Mouse Neocortex. PLoS Biol 2015; 13:e1002217. [PMID: 26252244 PMCID: PMC4529158 DOI: 10.1371/journal.pbio.1002217] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 06/30/2015] [Indexed: 11/21/2022] Open
Abstract
The evolutionary expansion of the neocortex in mammals has been linked to enlargement of the subventricular zone (SVZ) and increased proliferative capacity of basal progenitors (BPs), notably basal radial glia (bRG). The transcription factor Pax6 is known to be highly expressed in primate, but not mouse, BPs. Here, we demonstrate that sustaining Pax6 expression selectively in BP-genic apical radial glia (aRG) and their BP progeny of embryonic mouse neocortex suffices to induce primate-like progenitor behaviour. Specifically, we conditionally expressed Pax6 by in utero electroporation using a novel, Tis21–CreERT2 mouse line. This expression altered aRG cleavage plane orientation to promote bRG generation, increased cell-cycle re-entry of BPs, and ultimately increased upper-layer neuron production. Upper-layer neuron production was also increased in double-transgenic mouse embryos with sustained Pax6 expression in the neurogenic lineage. Strikingly, increased BPs existed not only in the SVZ but also in the intermediate zone of the neocortex of these double-transgenic mouse embryos. In mutant mouse embryos lacking functional Pax6, the proportion of bRG among BPs was reduced. Our data identify specific Pax6 effects in BPs and imply that sustaining this Pax6 function in BPs could be a key aspect of SVZ enlargement and, consequently, the evolutionary expansion of the neocortex. "Humanizing" the expression of the transcription factor Pax6 in cortical progenitors in the developing mouse brain is sufficient to endow these progenitors with a primate-like proliferative capacity. During development, neural progenitors generate all cells that make up the mammalian brain. Differences in brain size among the various mammalian species are attributed to differences in the abundance and proliferative capacity of a specific class of neural progenitors called basal progenitors. Among these, a specific progenitor type called basal radial glia is thought to have played an important role during evolution in the expansion of the neocortex, the part of the brain associated with higher cognitive functions like conscious thought and language. In the neocortex, the expression of the transcription factor Pax6 in basal progenitors is low in rodents, but high in primates, including humans. In this study, we aimed to mimic the elevated expression pattern of Pax6 seen in humans in basal progenitors of the embryonic mouse neocortex. To this end, we generated a novel, transgenic mouse line that allows sustained expression of the Pax6 gene in basal progenitors. This elevated expression resulted in an increase in the generation of basal radial glia, in the proliferative capacity of basal progenitors, and, ultimately, in the number of neurons produced. Our findings demonstrate that altering the expression of a single transcription factor from a mouse to a human-like pattern suffices to induce a primate-like proliferative behaviour in neural progenitors, which is thought to underlie the evolutionary expansion of the neocortex.
Collapse
Affiliation(s)
- Fong Kuan Wong
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ji-Feng Fei
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Elena Taverna
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Christiane Haffner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Jun Fu
- Biotechnology Center of the Technische Universität Dresden, Dresden, Germany
| | | | - A. Francis Stewart
- Biotechnology Center of the Technische Universität Dresden, Dresden, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail:
| |
Collapse
|
55
|
De Juan Romero C, Borrell V. Coevolution of radial glial cells and the cerebral cortex. Glia 2015; 63:1303-19. [PMID: 25808466 PMCID: PMC5008138 DOI: 10.1002/glia.22827] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/09/2015] [Indexed: 12/14/2022]
Abstract
Radial glia cells play fundamental roles in the development of the cerebral cortex, acting both as the primary stem and progenitor cells, as well as the guides for neuronal migration and lamination. These critical functions of radial glia cells in cortical development have been discovered mostly during the last 15 years and, more recently, seminal studies have demonstrated the existence of a remarkable diversity of additional cortical progenitor cell types, including a variety of basal radial glia cells with key roles in cortical expansion and folding, both in ontogeny and phylogeny. In this review, we summarize the main cellular and molecular mechanisms known to be involved in cerebral cortex development in mouse, as the currently preferred animal model, and then compare these with known mechanisms in other vertebrates, both mammal and nonmammal, including human. This allows us to present a global picture of how radial glia cells and the cerebral cortex seem to have coevolved, from reptiles to primates, leading to the remarkable diversity of vertebrate cortical phenotypes.
Collapse
Affiliation(s)
- Camino De Juan Romero
- Instituto De Neurociencias, Consejo Superior De Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan D'alacant, Spain
| | - Víctor Borrell
- Instituto De Neurociencias, Consejo Superior De Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan D'alacant, Spain
| |
Collapse
|
56
|
Tsimbouri PM. Adult Stem Cell Responses to Nanostimuli. J Funct Biomater 2015; 6:598-622. [PMID: 26193326 PMCID: PMC4598673 DOI: 10.3390/jfb6030598] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/29/2015] [Accepted: 07/08/2015] [Indexed: 12/31/2022] Open
Abstract
Adult or mesenchymal stem cells (MSCs) have been found in different tissues in the body, residing in stem cell microenvironments called "stem cell niches". They play different roles but their main activity is to maintain tissue homeostasis and repair throughout the lifetime of an organism. Their ability to differentiate into different cell types makes them an ideal tool to study tissue development and to use them in cell-based therapies. This differentiation process is subject to both internal and external forces at the nanoscale level and this response of stem cells to nanostimuli is the focus of this review.
Collapse
Affiliation(s)
- Penelope M Tsimbouri
- Centre for Cell Engineering, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, Joseph Black Building, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
57
|
Chu CC, Zavala JA, Spencer JL, Curzi MJ, Fields CJ, Drnevich J, Siegfried BD, Seufferheld MJ. Patterns of differential gene expression in adult rotation-resistant and wild-type western corn rootworm digestive tracts. Evol Appl 2015; 8:692-704. [PMID: 26240606 PMCID: PMC4516421 DOI: 10.1111/eva.12278] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/21/2015] [Indexed: 01/03/2023] Open
Abstract
The western corn rootworm (WCR,Diabrotica virgifera virgifera LeConte) is an important pest of corn. Annual crop rotation between corn and soybean disrupts the corn-dependent WCR life cycle and is widely adopted to manage this pest. This strategy selected for rotation-resistant (RR) WCR with reduced ovipositional fidelity to corn. Previous studies revealed that RR-WCR adults exhibit greater tolerance of soybean diets, different gut physiology, and host-microbe interactions compared to rotation-susceptible wild types (WT). To identify the genetic mechanisms underlying these phenotypic changes, a de novo assembly of the WCR adult gut transcriptome was constructed and used for RNA-sequencing analyses of RNA libraries from different WCR phenotypes fed with corn or soybean diets. Global gene expression profiles of WT- and RR-WCR were similar when feeding on corn diets, but different when feeding on soybean. Using network-based methods, we identified gene modules transcriptionally correlated with the RR phenotype. Gene ontology enrichment analyses indicated that the functions of these modules were related to metabolic processes, immune responses, biological adhesion, and other functions/processes that appear to correlate to documented traits in RR populations. These results suggest that gut transcriptomic divergence correlated with brief soybean feeding and other physiological traits may exist between RR- and WT-WCR adults.
Collapse
Affiliation(s)
- Chia-Ching Chu
- Department of Crop Sciences, University of Illinois Urbana, IL, USA
| | - Jorge A Zavala
- Facultad de Agronomía, Cátedra de Bioquímica INBA-CONICET, University of Buenos Aires-CONICET Buenos Aires, Argentina
| | - Joseph L Spencer
- Illinois Natural History Survey, University of Illinois Champaign, IL, USA
| | | | - Christopher J Fields
- High-Performance Biological Computing, Roy J. Carver Biotechnology Center Urbana, IL, USA
| | - Jenny Drnevich
- High-Performance Biological Computing, Roy J. Carver Biotechnology Center Urbana, IL, USA
| | | | - Manfredo J Seufferheld
- Department of Entomology, University of Illinois Urbana, IL, USA ; Illinois Natural History Survey, University of Illinois Champaign, IL, USA
| |
Collapse
|
58
|
Tsai Y, Cutts J, Kimura A, Varun D, Brafman DA. A chemically defined substrate for the expansion and neuronal differentiation of human pluripotent stem cell-derived neural progenitor cells. Stem Cell Res 2015; 15:75-87. [DOI: 10.1016/j.scr.2015.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 01/27/2023] Open
|
59
|
Yang Z, Dong P, Fu X, Li Q, Ma S, Wu D, Kang N, Liu X, Yan L, Xiao R. CD49f Acts as an Inflammation Sensor to Regulate Differentiation, Adhesion, and Migration of Human Mesenchymal Stem Cells. Stem Cells 2015; 33:2798-810. [PMID: 26013602 DOI: 10.1002/stem.2063] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022]
Abstract
The advent of mesenchymal stem cell (MSC)-based therapies has been an exciting innovation for the treatment of degenerative and inflammatory diseases. However, the surface markers that accurately reflect the self-renewal and differentiation potential of MSCs and their sensitivity to environmental cues remain poorly defined. Here, we studied the role of CD49f in bone marrow MSCs (BMSCs) and the mechanism by which it regulates the behavior of BMSCs under inflammatory conditions. We found that CD49f is preferentially expressed in fetal cells rather than adult cells, CD49f-positive BMSCs possess higher CFU-F formation ability and differentiation potential than CD49f negative cells, and the CD49f expression of BMSCs gradually decreases during in vitro passaging. CD49f knockdown dramatically decreased the differentiation of BMSCs and isoform A was demonstrated to be the main functional form that enhanced the differentiation ability of BMSCs. The influences of inflammatory cytokines on BMSCs revealed that TNF-α downregulated CD49f in BMSCs with impaired differentiation, decreased adhesion to laminins, and increased migration. Moreover, tissue transglutaminase was found to work together with CD49f to regulate the behavior of BMSCs. Finally, we showed that mTOR signaling rather than NF-κB activation mediated CD49f downregulation induced by TNF-α and maintained CD49f homeostasis in BMSCs. Our findings suggest that CD49f is a stemness marker of BMSCs and is tightly correlated with the behavioral changes of BMSCs under inflammatory conditions. These data demonstrate a novel role for CD49f in sensing inflammation through mTOR pathway to further modulate the behavior of MSCs to fulfill the requirements of the body.
Collapse
Affiliation(s)
- Zhigang Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union of Medical College, Beijing, People's Republic of China
| | - Ping Dong
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union of Medical College, Beijing, People's Republic of China
| | - Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union of Medical College, Beijing, People's Republic of China
| | - Qiuchen Li
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union of Medical College, Beijing, People's Republic of China
| | - Shize Ma
- 307-Ivy Translational Medicine Center, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Dongying Wu
- 307-Ivy Translational Medicine Center, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Ning Kang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union of Medical College, Beijing, People's Republic of China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union of Medical College, Beijing, People's Republic of China
| | - Li Yan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union of Medical College, Beijing, People's Republic of China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union of Medical College, Beijing, People's Republic of China
| |
Collapse
|
60
|
Syva SH, Ampon K, Lasimbang H, Fatimah SS. Microenvironmental factors involved in human amnion mesenchymal stem cells fate decisions. J Tissue Eng Regen Med 2015; 11:311-320. [PMID: 26073746 DOI: 10.1002/term.2043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 04/12/2015] [Accepted: 04/29/2015] [Indexed: 12/28/2022]
Abstract
Human amnion mesenchymal stem cells (HAMCs) show great differentiation and proliferation potential and also other remarkable features that could serve as an outstanding alternative source of stem cells in regenerative medicine. Recent reports have demonstrated various kinds of effective artificial niche that mimic the microenvironment of different types of stem cell to maintain and control their fate and function. The components of the stem cell microenvironment consist mainly of soluble and insoluble factors responsible for regulating stem cell differentiation and self-renewal. Extensive studies have been made on regulating HAMCs differentiation into specific phenotypes; however, the understanding of relevant factors in directing stem cell fate decisions in HAMCs remain underexplored. In this review, we have therefore identified soluble and insoluble factors, including mechanical stimuli and cues from the other supporting cells that are involved in directing HAMCs fate decisions. In order to strengthen the significance of understanding on the relevant factors involved in stem cell fate decisions, recent technologies developed to specifically mimic the microenvironments of specific cell lineages are also reviewed. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | - Kamaruzaman Ampon
- Biotechnology Research Institute, Universiti Malaysia Sabah, Malaysia
| | - Helen Lasimbang
- Faculty of Medicine and Health Science, Universiti Malaysia Sabah, Malaysia
| | | |
Collapse
|
61
|
Nelson DA, Larsen M. Heterotypic control of basement membrane dynamics during branching morphogenesis. Dev Biol 2015; 401:103-9. [PMID: 25527075 PMCID: PMC4465071 DOI: 10.1016/j.ydbio.2014.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/24/2014] [Accepted: 12/09/2014] [Indexed: 02/06/2023]
Abstract
Many mammalian organs undergo branching morphogenesis to create highly arborized structures with maximized surface area for specialized organ function. Cooperative cell-cell and cell-matrix adhesions that sculpt the emerging tissue architecture are guided by dynamic basement membranes. Properties of the basement membrane are reciprocally controlled by the interacting epithelial and mesenchymal cell populations. Here we discuss how basement membrane remodeling is required for branching morphogenesis to regulate cell-matrix and cell-cell adhesions that are required for cell patterning during morphogenesis and how basement membrane impacts morphogenesis by stimulation of cell patterning, force generation, and mechanotransduction. We suggest that in addition to creating mature epithelial architecture, remodeling of the epithelial basement membrane during branching morphogenesis is also essential to promote maturation of the stromal mesenchyme to create mature organ structure. Recapitulation of developmental cell-matrix and cell-cell interactions are of critical importance in tissue engineering and regeneration strategies that seek to restore organ function.
Collapse
Affiliation(s)
- Deirdre A Nelson
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, 1400 Washington Ave, Albany, NY 12222, USA
| | - Melinda Larsen
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, 1400 Washington Ave, Albany, NY 12222, USA.
| |
Collapse
|
62
|
Dehay C, Kennedy H, Kosik KS. The outer subventricular zone and primate-specific cortical complexification. Neuron 2015; 85:683-94. [PMID: 25695268 DOI: 10.1016/j.neuron.2014.12.060] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Evolutionary expansion and complexification of the primate cerebral cortex are largely linked to the emergence of the outer subventricular zone (OSVZ), a uniquely structured germinal zone that generates the expanded primate supragranular layers. The primate OSVZ departs from rodent germinal zones in that it includes a higher diversity of precursor types, inter-related in bidirectional non-hierarchical lineages. In addition, primate-specific regulatory mechanisms are operating in primate cortical precursors via the occurrence of novel miRNAs. Here, we propose that the origin and evolutionary importance of the OSVZ is related to genetic changes in multiple regulatory loops and that cell-cycle regulation is a favored target for evolutionary adaptation of the cortex.
Collapse
Affiliation(s)
- Colette Dehay
- Stem Cell and Brain Research Institute, INSERM U846, 18 Avenue Doyen Lepine, 69500 Bron, France; Université de Lyon, Université Lyon I, 69003, Lyon, France.
| | - Henry Kennedy
- Stem Cell and Brain Research Institute, INSERM U846, 18 Avenue Doyen Lepine, 69500 Bron, France; Université de Lyon, Université Lyon I, 69003, Lyon, France.
| | - Kenneth S Kosik
- Neuroscience Research Institute and Dept Cellular Molecular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
63
|
Wan PX, Wang BW, Wang ZC. Importance of the stem cell microenvironment for ophthalmological cell-based therapy. World J Stem Cells 2015; 7:448-460. [PMID: 25815128 PMCID: PMC4369500 DOI: 10.4252/wjsc.v7.i2.448] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 09/17/2014] [Accepted: 10/29/2014] [Indexed: 02/06/2023] Open
Abstract
Cell therapy is a promising treatment for diseases that are caused by cell degeneration or death. The cells for clinical transplantation are usually obtained by culturing healthy allogeneic or exogenous tissue in vitro. However, for diseases of the eye, obtaining the adequate number of cells for clinical transplantation is difficult due to the small size of tissue donors and the frequent needs of long-term amplification of cells in vitro, which results in low cell viability after transplantation. In addition, the transplanted cells often develop fibrosis or degrade and have very low survival. Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPS) are also promising candidates for cell therapy. Unfortunately, the differentiation of ESCs can bring immune rejection, tumorigenicity and undesired differentiated cells, limiting its clinical application. Although iPS cells can avoid the risk of immune rejection caused by ES cell differentiation post-transplantation, the low conversion rate, the risk of tumor formation and the potentially unpredictable biological changes that could occur through genetic manipulation hinder its clinical application. Thus, the desired clinical effect of cell therapy is impaired by these factors. Recent research findings recognize that the reason for low survival of the implanted cells not only depends on the seeded cells, but also on the cell microenvironment, which determines the cell survival, proliferation and even reverse differentiation. When used for cell therapy, the transplanted cells need a specific three-dimensional structure to anchor and specific extra cellular matrix components in addition to relevant cytokine signaling to transfer the required information to support their growth. These structures present in the matrix in which the stem cells reside are known as the stem cell microenvironment. The microenvironment interaction with the stem cells provides the necessary homeostasis for cell maintenance and growth. A large number of studies suggest that to explore how to reconstruct the stem cell microenvironment and strengthen its combination with the transplanted cells are key steps to successful cell therapy. In this review, we will describe the interactions of the stem cell microenvironment with the stem cells, discuss the importance of the stem cell microenvironment for cell-based therapy in ocular diseases, and introduce the progress of stem cell-based therapy for ocular diseases.
Collapse
|
64
|
McMillan M, Andronicos N, Davey R, Stockwell S, Hinch G, Schmoelzl S. Claudin-8 expression in Sertoli cells and putative spermatogonial stem cells in the bovine testis. Reprod Fertil Dev 2015; 26:633-44. [PMID: 23673210 DOI: 10.1071/rd12259] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 04/16/2013] [Indexed: 12/25/2022] Open
Abstract
Adhesion molecules are expressed by both adult and embryonic stem cells, with different classes of adhesion molecules involved in cell-membrane and intercellular contacts. In this study the expression of the adhesion molecule claudin-8 (CLDN8), a tight-junction protein, was investigated as a potential marker for undifferentiated spermatogonia in the bovine testis. We found that CLDN8 was expressed by both spermatogonia and a subset of Sertoli cells in the bovine testis. We also showed co-expression of GFRα1 in testis cells with CLDN8 and with Dolichos biflorus agglutinin-fluorescein isothiocyanate (DBA-FITC) staining. We observed co-enrichment of spermatogonia and CLDN8-expressing Sertoli cells in DBA-FITC-assisted magnetic-activated cell sorting (MACS), an observation supported by results from fluorescence-activated cell sorting analysis, which showed CLDN8-expressing cells were over-represented in the MACS-positive cell fraction, leading to the hypothesis that CLDN8 may play a role in the spermatogonial stem-cell niche.
Collapse
Affiliation(s)
- Mary McMillan
- CSIRO Food Futures National Research Flagship, North Ryde, NSW 2113, Australia
| | - Nicholas Andronicos
- CSIRO Animal, Food and Health Sciences, F. D. McMaster Laboratory, Armidale, NSW 2350, Australia
| | - Rhonda Davey
- CSIRO Food Futures National Research Flagship, North Ryde, NSW 2113, Australia
| | - Sally Stockwell
- CSIRO Food Futures National Research Flagship, North Ryde, NSW 2113, Australia
| | - Geoff Hinch
- School of Environmental and Rural Science, University of New England, Armidale, NSW 2351, Australia
| | - Sabine Schmoelzl
- CSIRO Food Futures National Research Flagship, North Ryde, NSW 2113, Australia
| |
Collapse
|
65
|
Maartens AP, Brown NH. Anchors and signals: the diverse roles of integrins in development. Curr Top Dev Biol 2015; 112:233-72. [PMID: 25733142 DOI: 10.1016/bs.ctdb.2014.11.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Integrins mediate cell adhesion by providing a link between the actin cytoskeleton and the extracellular matrix. As well as acting to anchor cells, integrin adhesions provide sensory input via mechanotransduction and synergism with signaling pathways, and provide the cell with the conditions necessary for differentiation in a permissive manner. In this review, we explore how integrins contribute to development, and what this tells us about how they work. From a signaling perspective, the influence of integrins on cell viability and fate is muted in a developmental context as compared to cell culture. Integrin phenotypes tend to arise from a failure of normally specified cells to create tissues properly, due to defective adhesion. The diversity of integrin functions in development shows how cell adhesion is continuously adjusted, both within and between animals, to fit developmental purpose.
Collapse
Affiliation(s)
- Aidan P Maartens
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas H Brown
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
66
|
Rahman MM, Ghosh M, Subramani J, Fong GH, Carlson ME, Shapiro LH. CD13 regulates anchorage and differentiation of the skeletal muscle satellite stem cell population in ischemic injury. Stem Cells 2015; 32:1564-77. [PMID: 24307555 DOI: 10.1002/stem.1610] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 10/16/2013] [Accepted: 10/21/2013] [Indexed: 01/03/2023]
Abstract
CD13 is a multifunctional cell surface molecule that regulates inflammatory and angiogenic mechanisms in vitro, but its contribution to these processes in vivo or potential roles in stem cell biology remains unexplored. We investigated the impact of loss of CD13 on a model of ischemic skeletal muscle injury that involves angiogenesis, inflammation, and stem cell mobilization. Consistent with its role as an inflammatory adhesion molecule, lack of CD13 altered myeloid trafficking in the injured muscle, resulting in cytokine profiles skewed toward a prohealing environment. Despite this healing-favorable context, CD13(KO) animals showed significantly impaired limb perfusion with increased necrosis, fibrosis, and lipid accumulation. Capillary density was correspondingly decreased, implicating CD13 in skeletal muscle angiogenesis. The number of CD45-/Sca1-/α7-integrin+/β1-integrin+ satellite cells was markedly diminished in injured CD13(KO) muscles and adhesion of isolated CD13(KO) satellite cells was impaired while their differentiation was accelerated. Bone marrow transplantation studies showed contributions from both host and donor cells to wound healing. Importantly, CD13 was coexpressed with Pax7 on isolated muscle-resident satellite cells. Finally, phosphorylated-focal adhesion kinase and ERK levels were reduced in injured CD13(KO) muscles, consistent with CD13 regulating satellite cell adhesion, potentially contributing to the maintenance and renewal of the satellite stem cell pool and facilitating skeletal muscle regeneration.
Collapse
Affiliation(s)
- M Mamunur Rahman
- Center for Vascular Biology and University of Connecticut Health Center, Farmington, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
67
|
Suh HN, Han HJ. Sonic hedgehog increases the skin wound-healing ability of mouse embryonic stem cells through the microRNA 200 family. Br J Pharmacol 2014; 172:815-28. [PMID: 25257936 DOI: 10.1111/bph.12947] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 09/03/2014] [Accepted: 09/13/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE To use stem cell therapy effectively, it is important to enhance the therapeutic potential of stem cells with soluble factors. Although sonic hedgehog (shh) is important in maintaining the stem cell, the recovery effect of mouse embryonic stem cells (mESCs) with shh has not yet been elucidated. The present study investigated the effect of mESCs with shh in skin recovery in vivo as well as the related intracellular signal pathways in vitro. EXPERIMENTAL APPROACH The healing effect of mESCs with shh on skin wounds was examined in vivo in ICR mice. The involvement of Smads, the microRNA (miR)-200 family, zinc finger E-box-binding homeobox (ZEBs) and E-cadherin on shh-induced mESC migration and self-renewal was determined in vitro. KEY RESULTS The mESCs with shh increased re-epithelialization and VEGF expression in skin wounds. Shh-treated mESCs increased both secreted and intracellular levels of VEGF. Shh induced dephosphorylation of glycogen synthase kinase 3β through the Smoothened receptor and increased the phosphorylation of Smad1 and Smad2/3 in mESCs. Shh-induced decrease of the mmu-miR-141, -200c, -200a, -200b and -429 expression levels was significantly reversed by Smad4 siRNA. Shh increased nuclear expression of ZEB1/ZEB2 and decreased E-cadherin expression while increasing cell migration and skin wound healing. Both these effects were reversed by mmu-miR-141 and -200b mimics. CONCLUSIONS AND IMPLICATIONS Mouse ESCs accelerated skin wound healing by shh through down-regulating E-cadherin, an effect dependent on mmu-miR-141 and -200b. Our data provides evidence for the effectiveness of shh in stem cell-based therapy in vivo.
Collapse
Affiliation(s)
- Han Na Suh
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | | |
Collapse
|
68
|
McMurray RJ, Dalby MJ, Tsimbouri PM. Using biomaterials to study stem cell mechanotransduction, growth and differentiation. J Tissue Eng Regen Med 2014; 9:528-39. [DOI: 10.1002/term.1957] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 06/11/2014] [Accepted: 08/28/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Rebecca J. McMurray
- Mrksich Research Group, Department of Biomedical Engineering; Northwestern University; Evanston IL USA
| | - Matthew J. Dalby
- Centre for Cell Engineering, Institute of Molecular, Cell and Systems Biology; University of Glasgow; UK
| | - P. Monica Tsimbouri
- Centre for Cell Engineering, Institute of Molecular, Cell and Systems Biology; University of Glasgow; UK
| |
Collapse
|
69
|
Martin-Lannerée S, Hirsch TZ, Hernandez-Rapp J, Halliez S, Vilotte JL, Launay JM, Mouillet-Richard S. PrP(C) from stem cells to cancer. Front Cell Dev Biol 2014; 2:55. [PMID: 25364760 PMCID: PMC4207012 DOI: 10.3389/fcell.2014.00055] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/11/2014] [Indexed: 12/23/2022] Open
Abstract
The cellular prion protein PrP(C) was initially discovered as the normal counterpart of the pathological scrapie prion protein PrP(Sc), the main component of the infectious agent of Transmissible Spongiform Encephalopathies. While clues as to the physiological function of this ubiquitous protein were greatly anticipated from the development of knockout animals, PrP-null mice turned out to be viable and to develop without major phenotypic abnormalities. Notwithstanding, the discovery that hematopoietic stem cells from PrP-null mice have impaired long-term repopulating potential has set the stage for investigating into the role of PrP(C) in stem cell biology. A wealth of data have now exemplified that PrP(C) is expressed in distinct types of stem cells and regulates their self-renewal as well as their differentiation potential. A role for PrP(C) in the fate restriction of embryonic stem cells has further been proposed. Paralleling these observations, an overexpression of PrP(C) has been documented in various types of tumors. In line with the contribution of PrP(C) to stemness and to the proliferation of cancer cells, PrP(C) was recently found to be enriched in subpopulations of tumor-initiating cells. In the present review, we summarize the current knowledge of the role played by PrP(C) in stem cell biology and discuss how the subversion of its function may contribute to cancer progression.
Collapse
Affiliation(s)
- Séverine Martin-Lannerée
- Toxicology, Pharmacology and Cellular Signaling, INSERM UMR-S1124 Paris, France ; Toxicology, Pharmacology and Cellular Signaling, Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France
| | - Théo Z Hirsch
- Toxicology, Pharmacology and Cellular Signaling, INSERM UMR-S1124 Paris, France ; Toxicology, Pharmacology and Cellular Signaling, Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France
| | - Julia Hernandez-Rapp
- Toxicology, Pharmacology and Cellular Signaling, INSERM UMR-S1124 Paris, France ; Toxicology, Pharmacology and Cellular Signaling, Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France ; Université Paris Sud 11, ED419 Biosigne Orsay, France
| | - Sophie Halliez
- U892 Virologie et Immunologie Moléculaires, INRA Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- UMR1313 Génétique Animale et Biologie Intégrative, INRA Jouy-en-Josas, France
| | - Jean-Marie Launay
- AP-HP Service de Biochimie, Fondation FondaMental, INSERM U942 Hôpital Lariboisière Paris, France ; Pharma Research Department, F. Hoffmann-La-Roche Ltd. Basel, Switzerland
| | - Sophie Mouillet-Richard
- Toxicology, Pharmacology and Cellular Signaling, INSERM UMR-S1124 Paris, France ; Toxicology, Pharmacology and Cellular Signaling, Université Paris Descartes, Sorbonne Paris Cité, UMR-S1124 Paris, France
| |
Collapse
|
70
|
Taverna E, Götz M, Huttner WB. The cell biology of neurogenesis: toward an understanding of the development and evolution of the neocortex. Annu Rev Cell Dev Biol 2014; 30:465-502. [PMID: 25000993 DOI: 10.1146/annurev-cellbio-101011-155801] [Citation(s) in RCA: 535] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neural stem and progenitor cells have a central role in the development and evolution of the mammalian neocortex. In this review, we first provide a set of criteria to classify the various types of cortical stem and progenitor cells. We then discuss the issue of cell polarity, as well as specific subcellular features of these cells that are relevant for their modes of division and daughter cell fate. In addition, cortical stem and progenitor cell behavior is placed into a tissue context, with consideration of extracellular signals and cell-cell interactions. Finally, the differences across species regarding cortical stem and progenitor cells are dissected to gain insight into key developmental and evolutionary mechanisms underlying neocortex expansion.
Collapse
Affiliation(s)
- Elena Taverna
- Max-Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany;
| | | | | |
Collapse
|
71
|
Porlan E, Martí-Prado B, Morante-Redolat JM, Consiglio A, Delgado AC, Kypta R, López-Otín C, Kirstein M, Fariñas I. MT5-MMP regulates adult neural stem cell functional quiescence through the cleavage of N-cadherin. Nat Cell Biol 2014; 16:629-38. [DOI: 10.1038/ncb2993] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 05/20/2014] [Indexed: 12/14/2022]
|
72
|
Herrera A, Saade M, Menendez A, Marti E, Pons S. Sustained Wnt/β-catenin signalling causes neuroepithelial aberrations through the accumulation of aPKC at the apical pole. Nat Commun 2014; 5:4168. [PMID: 24942669 DOI: 10.1038/ncomms5168] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 05/19/2014] [Indexed: 11/09/2022] Open
Abstract
β-Catenin mediates the canonical Wnt pathway by stimulating Tcf-dependent transcription and also associates to N-cadherin at the apical complex (AC) of neuroblasts. Here, we show that while β-catenin activity is required to form the AC and to maintain the cell polarity, oncogenic mutations that render stable forms of β-catenin (sβ-catenin) maintain the stemness of neuroblasts, inhibiting their differentiation and provoking aberrant growth. In examining the transcriptional and structural roles of β-catenin, we find that while β-catenin/Tcf transcriptional activity induces atypical protein kinase C (aPKC) expression, an alternative effect of β-catenin restricts aPKC to the apical pole of neuroepithelial cells. In agreement, we show that a constitutively active form of aPKC reproduces the neuroepithelial aberrations induced by β-catenin. Therefore, we conclude that β-catenin controls the cell fate and polarity of the neuroblasts through the expression and localization of aPKC.
Collapse
Affiliation(s)
- Antonio Herrera
- Instituto de Investigaciones Biomédicas de Barcelona, CSIC-IDIBAPS, Rossellò 161, Barcelona 08036, Spain
| | - Murielle Saade
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Baldiri Reixac 20, Barcelona 08028, Spain
| | - Anghara Menendez
- Instituto de Investigaciones Biomédicas de Barcelona, CSIC-IDIBAPS, Rossellò 161, Barcelona 08036, Spain
| | - Elisa Marti
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Baldiri Reixac 20, Barcelona 08028, Spain
| | - Sebastian Pons
- 1] Instituto de Investigaciones Biomédicas de Barcelona, CSIC-IDIBAPS, Rossellò 161, Barcelona 08036, Spain [2] Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, Baldiri Reixac 20, Barcelona 08028, Spain
| |
Collapse
|
73
|
Zeng XXI, Yelon D. Cadm4 restricts the production of cardiac outflow tract progenitor cells. Cell Rep 2014; 7:951-60. [PMID: 24813897 DOI: 10.1016/j.celrep.2014.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 03/11/2014] [Accepted: 04/08/2014] [Indexed: 11/29/2022] Open
Abstract
Heart assembly requires input from two populations of progenitor cells, the first and second heart fields (FHF and SHF), that differentiate at distinct times and create different cardiac components. The cardiac outflow tract (OFT) is built through recruitment of late-differentiating, SHF-derived cardiomyocytes to the arterial pole of the heart. The mechanisms responsible for selection of an appropriate number of OFT cells from the SHF remain unclear. Here, we find that cell adhesion molecule 4 (cadm4) is essential for restricting the size of the zebrafish OFT. Knockdown of cadm4 causes dramatic OFT expansion, and overexpression of cadm4 results in a greatly diminished OFT. Moreover, cadm4 activity limits the production of OFT progenitor cells and the duration of their accumulation at the arterial pole. Together, our data suggest a role for cell adhesion in restraining SHF deployment to the OFT, perturbation of which could cause congenital OFT defects.
Collapse
Affiliation(s)
- Xin-Xin I Zeng
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
74
|
Drosophila perlecan regulates intestinal stem cell activity via cell-matrix attachment. Stem Cell Reports 2014; 2:761-9. [PMID: 24936464 PMCID: PMC4050351 DOI: 10.1016/j.stemcr.2014.04.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 01/15/2023] Open
Abstract
Stem cells require specialized local microenvironments, termed niches, for normal retention, proliferation, and multipotency. Niches are composed of cells together with their associated extracellular matrix (ECM). Currently, the roles of ECM in regulating niche functions are poorly understood. Here, we demonstrate that Perlecan (Pcan), a highly conserved ECM component, controls intestinal stem cell (ISC) activities and ISC-ECM attachment in Drosophila adult posterior midgut. Loss of Pcan from ISCs, but not other surrounding cells, causes ISCs to detach from underlying ECM, lose their identity, and fail to proliferate. These defects are not a result of a loss of epidermal growth factor receptor (EGFR) or Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling activity but partially depend on integrin signaling activity. We propose that Pcan secreted by ISCs confers niche properties to the adjacent ECM that is required for ISC maintenance of stem cell identity, activity, and anchorage to the niche.
Collapse
|
75
|
Nomura T, Murakami Y, Gotoh H, Ono K. Reconstruction of ancestral brains: exploring the evolutionary process of encephalization in amniotes. Neurosci Res 2014; 86:25-36. [PMID: 24671134 DOI: 10.1016/j.neures.2014.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 02/20/2014] [Accepted: 03/03/2014] [Indexed: 11/24/2022]
Abstract
There is huge divergence in the size and complexity of vertebrate brains. Notably, mammals and birds have bigger brains than other vertebrates, largely because these animal groups established larger dorsal telencephali. Fossil evidence suggests that this anatomical trait could have evolved independently. However, recent comparative developmental analyses demonstrate surprising commonalities in neuronal subtypes among species, although this interpretation is highly controversial. In this review, we introduce intriguing evidence regarding brain evolution collected from recent studies in paleontology and developmental biology, and we discuss possible evolutionary changes in the cortical developmental programs that led to the encephalization and structural complexity of amniote brains. New research concepts and approaches will shed light on the origin and evolutionary processes of amniote brains, particularly the mammalian cerebral cortex.
Collapse
Affiliation(s)
- Tadashi Nomura
- Developmental Neurobiology, Kyoto Prefectural University of Medicine, Nishitakatsukasa-cho 13, Taishogun, Kita-ku, Kyoto 603-8334, Japan; Japan Science and Technology Agency, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| | - Yasunori Murakami
- Graduate School of Science and Engineering, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Hitoshi Gotoh
- Developmental Neurobiology, Kyoto Prefectural University of Medicine, Nishitakatsukasa-cho 13, Taishogun, Kita-ku, Kyoto 603-8334, Japan
| | - Katsuhiko Ono
- Developmental Neurobiology, Kyoto Prefectural University of Medicine, Nishitakatsukasa-cho 13, Taishogun, Kita-ku, Kyoto 603-8334, Japan
| |
Collapse
|
76
|
Rojas-Ríos P, González-Reyes A. Concise Review: The Plasticity of Stem Cell Niches: A General Property Behind Tissue Homeostasis and Repair. Stem Cells 2014; 32:852-9. [DOI: 10.1002/stem.1621] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/30/2013] [Accepted: 11/09/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Patricia Rojas-Ríos
- Centro Andaluz de Biología del Desarrollo; CSIC/Universidad Pablo de Olavide/JA; Carretera de Utrera km 1 Sevilla Spain
| | - Acaimo González-Reyes
- Centro Andaluz de Biología del Desarrollo; CSIC/Universidad Pablo de Olavide/JA; Carretera de Utrera km 1 Sevilla Spain
| |
Collapse
|
77
|
Farahani E, Patra HK, Jangamreddy JR, Rashedi I, Kawalec M, Rao Pariti RK, Batakis P, Wiechec E. Cell adhesion molecules and their relation to (cancer) cell stemness. Carcinogenesis 2014; 35:747-59. [PMID: 24531939 DOI: 10.1093/carcin/bgu045] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite decades of search for anticancer drugs targeting solid tumors, this group of diseases remains largely incurable, especially if in advanced, metastatic stage. In this review, we draw comparison between reprogramming and carcinogenesis, as well as between stem cells (SCs) and cancer stem cells (CSCs), focusing on changing garniture of adhesion molecules. Furthermore, we elaborate on the role of adhesion molecules in the regulation of (cancer) SCs division (symmetric or asymmetric), and in evolving interactions between CSCs and extracellular matrix. Among other aspects, we analyze the role and changes of expression of key adhesion molecules as cancer progresses and metastases develop. Here, the role of cadherins, integrins, as well as selected transcription factors like Twist and Snail is highlighted, not only in the regulation of epithelial-to-mesenchymal transition but also in the avoidance of anoikis. Finally, we briefly discuss recent developments and new strategies targeting CSCs, which focus on adhesion molecules or targeting tumor vasculature.
Collapse
Affiliation(s)
- Ensieh Farahani
- Department of Clinical and Experimental Medicine, Division of Cell Biology and Integrative Regenerative Medicine Center (IGEN) and
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Gattazzo F, Urciuolo A, Bonaldo P. Extracellular matrix: a dynamic microenvironment for stem cell niche. Biochim Biophys Acta Gen Subj 2014; 1840:2506-19. [PMID: 24418517 PMCID: PMC4081568 DOI: 10.1016/j.bbagen.2014.01.010] [Citation(s) in RCA: 896] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 01/05/2014] [Accepted: 01/06/2014] [Indexed: 02/08/2023]
Abstract
Background Extracellular matrix (ECM) is a dynamic and complex environment characterized by biophysical, mechanical and biochemical properties specific for each tissue and able to regulate cell behavior. Stem cells have a key role in the maintenance and regeneration of tissues and they are located in a specific microenvironment, defined as niche. Scope of review We overview the progresses that have been made in elucidating stem cell niches and discuss the mechanisms by which ECM affects stem cell behavior. We also summarize the current tools and experimental models for studying ECM–stem cell interactions. Major conclusions ECM represents an essential player in stem cell niche, since it can directly or indirectly modulate the maintenance, proliferation, self-renewal and differentiation of stem cells. Several ECM molecules play regulatory functions for different types of stem cells, and based on its molecular composition the ECM can be deposited and finely tuned for providing the most appropriate niche for stem cells in the various tissues. Engineered biomaterials able to mimic the in vivo characteristics of stem cell niche provide suitable in vitro tools for dissecting the different roles exerted by the ECM and its molecular components on stem cell behavior. General significance ECM is a key component of stem cell niches and is involved in various aspects of stem cell behavior, thus having a major impact on tissue homeostasis and regeneration under physiological and pathological conditions. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties. Stem cells have a key role in the maintenance and regeneration of tissues. The extracellular matrix is a critical regulator of stem cell function. Stem cells reside in a dynamic and specialized microenvironment denoted as niche. The extracellular matrix represents an essential component of stem cell niches. Bioengineered niches can be used for investigating stem cell–matrix interactions.
Collapse
Affiliation(s)
- Francesca Gattazzo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Anna Urciuolo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy.
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
79
|
Betizeau M, Cortay V, Patti D, Pfister S, Gautier E, Bellemin-Ménard A, Afanassieff M, Huissoud C, Douglas RJ, Kennedy H, Dehay C. Precursor diversity and complexity of lineage relationships in the outer subventricular zone of the primate. Neuron 2014; 80:442-57. [PMID: 24139044 DOI: 10.1016/j.neuron.2013.09.032] [Citation(s) in RCA: 336] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2013] [Indexed: 01/09/2023]
Abstract
Long-term ex vivo live imaging combined with unbiased sampling of cycling precursors shows that macaque outer subventricular zone (OSVZ) includes four distinct basal radial glial (bRG) cell morphotypes, bearing apical and/or basal processes in addition to nonpolar intermediate progenitors (IPs). Each of the five precursor types exhibits extensive self-renewal and proliferative capacities as well as the ability to directly generate neurons, albeit with different frequencies. Cell-cycle parameters exhibited an unusual stage-specific regulation with short cell-cycle duration and increased rates of proliferative divisions during supragranular layer production at late corticogenesis. State transition analysis of an extensive clonal database reveals bidirectional transitions between OSVZ precursor types as well as stage-specific differences in their progeny and topology of the lineage relationships. These results explore rodent-primate differences and show that primate cortical neurons are generated through complex lineages by a mosaic of precursors, thereby providing an innovative framework for understanding specific features of primate corticogenesis.
Collapse
Affiliation(s)
- Marion Betizeau
- Stem Cell and Brain Research Institute, INSERM U846, 18 Avenue Doyen Lepine, 69500 Bron, France; Université de Lyon, Université Lyon I, 69003 Lyon, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Nono JK, Lutz MB, Brehm K. EmTIP, a T-Cell immunomodulatory protein secreted by the tapeworm Echinococcus multilocularis is important for early metacestode development. PLoS Negl Trop Dis 2014; 8:e2632. [PMID: 24392176 PMCID: PMC3879249 DOI: 10.1371/journal.pntd.0002632] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Accepted: 11/26/2013] [Indexed: 01/05/2023] Open
Abstract
Background Alveolar echinococcosis (AE), caused by the metacestode of the tapeworm Echinococcus multilocularis, is a lethal zoonosis associated with host immunomodulation. T helper cells are instrumental to control the disease in the host. Whereas Th1 cells can restrict parasite proliferation, Th2 immune responses are associated with parasite proliferation. Although the early phase of host colonization by E. multilocularis is dominated by a potentially parasitocidal Th1 immune response, the molecular basis of this response is unknown. Principal Findings We describe EmTIP, an E. multilocularis homologue of the human T-cell immunomodulatory protein, TIP. By immunohistochemistry we show EmTIP localization to the intercellular space within parasite larvae. Immunoprecipitation and Western blot experiments revealed the presence of EmTIP in the excretory/secretory (E/S) products of parasite primary cell cultures, representing the early developing metacestode, but not in those of mature metacestode vesicles. Using an in vitro T-cell stimulation assay, we found that primary cell E/S products promoted interferon (IFN)-γ release by murine CD4+ T-cells, whereas metacestode E/S products did not. IFN-γ release by T-cells exposed to parasite products was abrogated by an anti-EmTIP antibody. When recombinantly expressed, EmTIP promoted IFN-γ release by CD4+ T-cells in vitro. After incubation with anti-EmTIP antibody, primary cells showed an impaired ability to proliferate and to form metacestode vesicles in vitro. Conclusions We provide for the first time a possible explanation for the early Th1 response observed during E. multilocularis infections. Our data indicate that parasite primary cells release a T-cell immunomodulatory protein, EmTIP, capable of promoting IFN-γ release by CD4+ T-cells, which is probably driving or supporting the onset of the early Th1 response during AE. The impairment of primary cell proliferation and the inhibition of metacestode vesicle formation by anti-EmTIP antibodies suggest that this factor fulfills an important role in early E. multilocularis development within the intermediate host. E. multilocularis is a parasitic helminth causing the chronic human disease alveolar echinococcosis. Current disease control measures are very limited resulting in a high case-fatality rate. A transiently dominating Th1 immune response is mounted at the early phase of the infection, potentially limiting parasite proliferation and disease progression. Understanding the molecular basis of this early anti-Echinococcocus Th1 response would provide valuable information to improve disease control. The authors found that EmTIP, a T-cell immunomodulatory protein homologue, is secreted by the parasite early larva and promotes a Th1 response in host cells. Interestingly, EmTIP binding by antibodies impairs the development of the early parasite larva towards the chronic stage. Altogether the authors propose that E. multilocularis utilizes EmTIP for early larval development, but in the process, the factor is released by the parasite larva and influences host T-cells by directing a parasitocidal Th1 immune response. Therefore, the authors recommend EmTIP as a promising lead for future studies on the development of anti-Echinococcus intervention strategies.
Collapse
Affiliation(s)
- Justin Komguep Nono
- University of Würzburg, Institute for Hygiene and Microbiology, Würzburg, Germany
| | - Manfred B. Lutz
- University of Würzburg, Institute of Virology and Immunobiology, Würzburg, Germany
| | - Klaus Brehm
- University of Würzburg, Institute for Hygiene and Microbiology, Würzburg, Germany
- * E-mail:
| |
Collapse
|
81
|
Turner LA, J. Dalby M. Nanotopography – potential relevance in the stem cell niche. Biomater Sci 2014; 2:1574-1594. [DOI: 10.1039/c4bm00155a] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nanotopographical cues observed in vivo (such as in the sinusoid and bone) closely resemble nanotopographies that in vitro have been shown to promote niche relevant stem cells behaviours; specifically, retention of multipotency and osteogenic differentiation on ordered and disordered nano-pits respectively. These and other observations highlight a potential role for nano topography in the stem cell niche.
Collapse
Affiliation(s)
- Lesley-Anne Turner
- Centre for Cell Engineering
- Institute of Molecular
- Cell and Systems Biology
- College of Medical
- Veterinary and Life Sciences
| | - Matthew J. Dalby
- Centre for Cell Engineering
- Institute of Molecular
- Cell and Systems Biology
- College of Medical
- Veterinary and Life Sciences
| |
Collapse
|
82
|
Abstract
Morphogenesis is the remarkable process by which cells self-assemble into complex tissues and organs that exhibit specialized form and function during embryological development. Many of the genes and chemical cues that mediate tissue and organ formation have been identified; however, these signals alone are not sufficient to explain how tissues and organs are constructed that exhibit their unique material properties and three-dimensional forms. Here, we review work that has revealed the central role that physical forces and extracellular matrix mechanics play in the control of cell fate switching, pattern formation, and tissue development in the embryo and how these same mechanical signals contribute to tissue homeostasis and developmental control throughout adult life.
Collapse
Affiliation(s)
- Tadanori Mammoto
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115;
| | | | | |
Collapse
|
83
|
Freida D, Lecourt S, Cras A, Vanneaux V, Letort G, Gidrol X, Guyon L, Larghero J, Thery M. Human Bone Marrow Mesenchymal Stem Cells Regulate Biased DNA Segregation in Response to Cell Adhesion Asymmetry. Cell Rep 2013; 5:601-10. [DOI: 10.1016/j.celrep.2013.09.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 07/12/2013] [Accepted: 09/12/2013] [Indexed: 01/01/2023] Open
|
84
|
Abstract
Within the adult organism, stem cells reside in defined anatomical microenvironments called niches. These architecturally diverse microenvironments serve to balance stem cell self-renewal and differentiation. Proper regulation of this balance is instrumental to tissue repair and homeostasis, and any imbalance can potentially lead to diseases such as cancer. Within each of these microenvironments, a myriad of chemical and physical stimuli interact in a complex (synergistic or antagonistic) manner to tightly regulate stem cell fate. The in vitro replication of these in vivo microenvironments will be necessary for the application of stem cells for disease modeling, drug discovery, and regenerative medicine purposes. However, traditional reductionist approaches have only led to the generation of cell culture methods that poorly recapitulate the in vivo microenvironment. To that end, novel engineering and systems biology approaches have allowed for the investigation of the biological and mechanical stimuli that govern stem cell fate. In this review, the application of these technologies for the dissection of stem cell microenvironments will be analyzed. Moreover, the use of these engineering approaches to construct in vitro stem cell microenvironments that precisely control stem cell fate and function will be reviewed. Finally, the emerging trend of using high-throughput, combinatorial methods for the stepwise engineering of stem cell microenvironments will be explored.
Collapse
Affiliation(s)
- David A Brafman
- Department of Cellular and Molecular Medicine, Stem Cell Program, University of California at San Diego, La Jolla, California
| |
Collapse
|
85
|
Wong VW, Gurtner GC, Longaker MT. Wound healing: a paradigm for regeneration. Mayo Clin Proc 2013; 88:1022-31. [PMID: 24001495 DOI: 10.1016/j.mayocp.2013.04.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 04/12/2013] [Indexed: 02/07/2023]
Abstract
Human skin is a remarkably plastic organ that sustains insult and injury throughout life. Its ability to expeditiously repair wounds is paramount to survival and is thought to be regulated by wound components such as differentiated cells, stem cells, cytokine networks, extracellular matrix, and mechanical forces. These intrinsic regenerative pathways are integrated across different skin compartments and are being elucidated on the cellular and molecular levels. Recent advances in bioengineering and nanotechnology have allowed researchers to manipulate these microenvironments in increasingly precise spatial and temporal scales, recapitulating key homeostatic cues that may drive regeneration. The ultimate goal is to translate these bench achievements into viable bedside therapies that address the growing global burden of acute and chronic wounds. In this review, we highlight current concepts in cutaneous wound repair and propose that many of these evolving paradigms may underlie regenerative processes across diverse organ systems.
Collapse
Affiliation(s)
- Victor W Wong
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, Stanford, CA, USA
| | | | | |
Collapse
|
86
|
Cellular kinetics of perivascular MSC precursors. Stem Cells Int 2013; 2013:983059. [PMID: 24023546 PMCID: PMC3760099 DOI: 10.1155/2013/983059] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/13/2013] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) and MSC-like multipotent stem/progenitor cells have been widely investigated for regenerative medicine and deemed promising in clinical applications. In order to further improve MSC-based stem cell therapeutics, it is important to understand the cellular kinetics and functional roles of MSCs in the dynamic regenerative processes. However, due to the heterogeneous nature of typical MSC cultures, their native identity and anatomical localization in the body have remained unclear, making it difficult to decipher the existence of distinct cell subsets within the MSC entity. Recent studies have shown that several blood-vessel-derived precursor cell populations, purified by flow cytometry from multiple human organs, give rise to bona fide MSCs, suggesting that the vasculature serves as a systemic reservoir of MSC-like stem/progenitor cells. Using individually purified MSC-like precursor cell subsets, we and other researchers have been able to investigate the differential phenotypes and regenerative capacities of these contributing cellular constituents in the MSC pool. In this review, we will discuss the identification and characterization of perivascular MSC precursors, including pericytes and adventitial cells, and focus on their cellular kinetics: cell adhesion, migration, engraftment, homing, and intercellular cross-talk during tissue repair and regeneration.
Collapse
|
87
|
Abstract
Integrins are transmembrane receptors that mediate cell adhesion to neighboring cells and to the extracellular matrix. Here, the various modes in which integrin-mediated adhesion regulates intracellular signaling pathways impinging on cell survival, proliferation, and differentiation are considered. Subsequently, evidence that integrins also control crucial signaling cascades in cancer cells is discussed. Lastly, the important role of integrin signaling in tumor cells as well as in stromal cells that support cancer growth, metastasis, and therapy resistance indicates that integrin signaling may be an attractive target for (combined) cancer therapy strategies. Current approaches to target integrins in this context are reviewed.
Collapse
|
88
|
Wong BG, Paz A, Corrado MA, Ramos BR, Cinquin A, Cinquin O, Hui EE. Live imaging reveals active infiltration of mitotic zone by its stem cell niche. Integr Biol (Camb) 2013; 5:976-82. [PMID: 23695198 PMCID: PMC3708607 DOI: 10.1039/c3ib20291g] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Stem cells niches are increasingly recognized as dynamic environments that play a key role in transducing signals that allow an organism to exert control on its stem cells. Live imaging of stem cell niches in their in vivo setting is thus of high interest to dissect stem cell controls. Here we report a new microfluidic design that is highly amenable to dissemination in biology laboratories that have no microfluidics expertise. This design has allowed us to perform the first time lapse imaging of the C. elegans germline stem cell niche. Our results show that this niche is strikingly dynamic, and that morphological changes that take place during development are the result of a highly active process. These results lay the foundation for future studies to dissect molecular mechanisms by which stem cell niche morphology is modulated, and by which niche morphology controls stem cell behavior.
Collapse
Affiliation(s)
- Brandon G. Wong
- Department of Biomedical Engineering, University of California, Irvine, CA 92697-2715, USA. Fax: +1 949 824 1727; Tel: +1 949 824 8723
- Department of Developmental & Cell Biology, University of California, Irvine, CA 92697, USA. Tel: +1 949 257 2819
- Now at Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Adrian Paz
- Department of Developmental & Cell Biology, University of California, Irvine, CA 92697, USA. Tel: +1 949 257 2819
- Center for Complex Biological Systems, University of California, Irvine, CA 92697
| | - Michael A. Corrado
- Department of Biomedical Engineering, University of California, Irvine, CA 92697-2715, USA. Fax: +1 949 824 1727; Tel: +1 949 824 8723
- Center for Complex Biological Systems, University of California, Irvine, CA 92697
| | - Brian R. Ramos
- Department of Developmental & Cell Biology, University of California, Irvine, CA 92697, USA. Tel: +1 949 257 2819
- Center for Complex Biological Systems, University of California, Irvine, CA 92697
| | - Amanda Cinquin
- Department of Developmental & Cell Biology, University of California, Irvine, CA 92697, USA. Tel: +1 949 257 2819
- Center for Complex Biological Systems, University of California, Irvine, CA 92697
| | - Olivier Cinquin
- Department of Developmental & Cell Biology, University of California, Irvine, CA 92697, USA. Tel: +1 949 257 2819
- Center for Complex Biological Systems, University of California, Irvine, CA 92697
| | - Elliot E. Hui
- Department of Biomedical Engineering, University of California, Irvine, CA 92697-2715, USA. Fax: +1 949 824 1727; Tel: +1 949 824 8723
- Center for Complex Biological Systems, University of California, Irvine, CA 92697
| |
Collapse
|
89
|
Lavado A, He Y, Paré J, Neale G, Olson EN, Giovannini M, Cao X. Tumor suppressor Nf2 limits expansion of the neural progenitor pool by inhibiting Yap/Taz transcriptional coactivators. Development 2013; 140:3323-34. [PMID: 23863479 DOI: 10.1242/dev.096537] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Brain development requires a precise balance between expansion of the neural progenitor pool and the production of postmitotic neurons and glia. Disruption of this equilibrium results in a myriad of structural abnormalities and disorders of the nervous system. The molecular mechanism that restricts neural progenitor expansion is poorly understood. Here we show that the tumor suppressor neurofibromatosis 2 (Nf2; merlin) limits the expansion of neural progenitor cells (NPCs) in the mammalian dorsal telencephalon. Nf2 is localized at the apical region of NPCs. In the absence of Nf2, NPCs of the cortical hem, hippocampal primordium and neocortical primordium overexpand, while production of Cajal-Retzius cells and hippocampal neurons decreases, resulting in severe malformation of the hippocampus in adult mice. We further show that Nf2 functions by inhibiting the Yap/Taz transcriptional coactivators, probably through a mechanism that is distinct from the canonical Hippo pathway. Overexpressing human YAP in NPCs causes a hippocampal malformation phenotype that closely resembles that of Nf2 mutants and, importantly, deleting Yap in the Nf2 mutant background largely restores hippocampal development. Our studies uncover Nf2 as an important inhibitor of neural progenitor expansion and establish Yap/Taz as key downstream effectors of Nf2 during brain development.
Collapse
Affiliation(s)
- Alfonso Lavado
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | |
Collapse
|
90
|
Nelson BR, Hodge RD, Bedogni F, Hevner RF. Dynamic interactions between intermediate neurogenic progenitors and radial glia in embryonic mouse neocortex: potential role in Dll1-Notch signaling. J Neurosci 2013; 33:9122-39. [PMID: 23699523 PMCID: PMC3716275 DOI: 10.1523/jneurosci.0791-13.2013] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/08/2013] [Accepted: 04/11/2013] [Indexed: 11/21/2022] Open
Abstract
The mammalian neocortical progenitor cell niche is composed of a diverse repertoire of neuroepithelial cells, radial glia (RG), and intermediate neurogenic progenitors (INPs). Previously, live-cell imaging experiments have proved crucial in identifying these distinct progenitor populations, especially INPs, which amplify neural output by undergoing additional rounds of proliferation before differentiating into new neurons. INPs also provide feedback to the RG pool by serving as a source of Delta-like 1 (Dll1), a key ligand for activating Notch signaling in neighboring cells, a well-known mechanism for maintaining RG identity. While much is known about Dll1-Notch signaling at the molecular level, little is known about how this cell-cell contact dependent feedback is transmitted at the cellular level. To investigate how RG and INPs might interact to convey Notch signals, we used high-resolution live-cell multiphoton microscopy (MPM) to directly observe cellular interactions and dynamics, in conjunction with Notch-pathway specific reporters in the neocortical neural stem cell niche in organotypic brain slices from embryonic mice. We found that INPs and RG interact via dynamic and transient elongate processes, some apparently long-range (extending from the subventricular zone to the ventricular zone), and some short-range (filopodia-like). Gene expression profiling of RG and INPs revealed further progenitor cell diversification, including different subpopulations of Hes1+ and/or Hes5+ RG, and Dll1+ and/or Dll3+ INPs. Thus, the embryonic progenitor niche includes a network of dynamic cell-cell interactions, using different combinations of Notch signaling molecules to maintain and likely diversify progenitor pools.
Collapse
Affiliation(s)
- Branden R. Nelson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101, and
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
| | - Rebecca D. Hodge
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
| | - Francesco Bedogni
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
| | - Robert F. Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101, and
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
| |
Collapse
|
91
|
Abstract
Stem cell niches are special microenvironments that maintain stem cells and control their behavior to ensure tissue homeostasis and regeneration throughout life. The liver has a high regenerative capacity that involves stem/progenitor cells when the proliferation of hepatocytes is impaired. In recent years progress has been made in the identification of potential hepatic stem cell niches. There is evidence that hepatic progenitor cells can originate from niches in the canals of Hering; in addition, the space of Disse may also serve as a stem cell niche during fetal hematopoiesis and constitute a niche for stellate cells in adults.
Collapse
Affiliation(s)
- Claus Kordes
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | |
Collapse
|
92
|
Stem Cell Niche. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
93
|
Yajima M, Wessel GM. Autonomy in specification of primordial germ cells and their passive translocation in the sea urchin. Development 2012; 139:3786-94. [PMID: 22991443 PMCID: PMC3445309 DOI: 10.1242/dev.082230] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2012] [Indexed: 01/22/2023]
Abstract
The process of germ line determination involves many conserved genes, yet is highly variable. Echinoderms are positioned at the base of Deuterostomia and are crucial to understanding these evolutionary transitions, yet the mechanism of germ line specification is not known in any member of the phyla. Here we demonstrate that small micromeres (SMics), which are formed at the fifth cell division of the sea urchin embryo, illustrate many typical features of primordial germ cell (PGC) specification. SMics autonomously express germ line genes in isolated culture, including selective Vasa protein accumulation and transcriptional activation of nanos; their descendants are passively displaced towards the animal pole by secondary mesenchyme cells and the elongating archenteron during gastrulation; Cadherin (G form) has an important role in their development and clustering phenotype; and a left/right integration into the future adult anlagen appears to be controlled by a late developmental mechanism. These results suggest that sea urchin SMics share many more characteristics typical of PGCs than previously thought, and imply a more widely conserved system of germ line development among metazoans.
Collapse
Affiliation(s)
- Mamiko Yajima
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA
| | - Gary M. Wessel
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA
| |
Collapse
|
94
|
Bröhl D, Vasyutina E, Czajkowski M, Griger J, Rassek C, Rahn HP, Purfürst B, Wende H, Birchmeier C. Colonization of the Satellite Cell Niche by Skeletal Muscle Progenitor Cells Depends on Notch Signals. Dev Cell 2012; 23:469-81. [DOI: 10.1016/j.devcel.2012.07.014] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 06/19/2012] [Accepted: 07/17/2012] [Indexed: 10/28/2022]
|
95
|
McNulty MA, Virdi AS, Christopherson KW, Sena K, Frank RR, Sumner DR. Adult stem cell mobilization enhances intramembranous bone regeneration: a pilot study. Clin Orthop Relat Res 2012; 470:2503-12. [PMID: 22528386 PMCID: PMC3830081 DOI: 10.1007/s11999-012-2357-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Stem cell mobilization, which is defined as the forced egress of stem cells from the bone marrow to the peripheral blood (PB) using chemokine receptor agonists, is an emerging concept for enhancing tissue regeneration. However, the effect of stem cell mobilization by a single injection of the C-X-C chemokine receptor type 4 (CXCR4) antagonist AMD3100 on intramembranous bone regeneration is unclear. QUESTIONS/PURPOSES We therefore asked: Does AMD3100 mobilize adult stem cells in C57BL/6 mice? Are stem cells mobilized to the PB after marrow ablation? And does AMD3100 enhance bone regeneration? METHODS Female C57BL/6 mice underwent femoral marrow ablation surgery alone (n = 25), systemic injection of AMD3100 alone (n = 15), or surgery plus AMD3100 (n = 57). We used colony-forming unit assays, flow cytometry, and micro-CT to investigate mobilization of mesenchymal stem cells, endothelial progenitor cells, and hematopoietic stem cells to the PB and bone regeneration. RESULTS AMD3100 induced mobilization of stem cells to the PB, resulting in a 40-fold increase in mesenchymal stem cells. The marrow ablation injury mobilized all three cell types to the PB over time. Administration of AMD3100 led to a 60% increase in bone regeneration at Day 21. CONCLUSIONS A single injection of a CXCR4 antagonist lead to stem cell mobilization and enhanced bone volume in the mouse marrow ablation model of intramembranous bone regeneration.
Collapse
Affiliation(s)
- Margaret A. McNulty
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| | - Amarjit S. Virdi
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| | | | - Kotaro Sena
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| | - Robin R. Frank
- Division of Hematology & Oncology, Rush University Medical Center, Chicago, IL USA
| | - Dale R. Sumner
- Department of Anatomy & Cell Biology, Rush University Medical Center, 600 Paulina Street, Chicago, IL 60612 USA
| |
Collapse
|
96
|
Brizzi MF, Tarone G, Defilippi P. Extracellular matrix, integrins, and growth factors as tailors of the stem cell niche. Curr Opin Cell Biol 2012; 24:645-51. [PMID: 22898530 DOI: 10.1016/j.ceb.2012.07.001] [Citation(s) in RCA: 282] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/13/2012] [Accepted: 07/09/2012] [Indexed: 12/21/2022]
Abstract
It is widely acknowledged that integrins, the major receptors for the extracellular matrix (ECM) proteins, exert an extensive crosstalk with many growth factor and cytokine receptors. Among them, growth factor receptors, such as the EGFR, MET, PDGFR and VEGFR, and the IL-3 receptor have been shown to be physically and functionally associated to integrins. The connection between integrins and other transmembrane receptors is bidirectional, integrins being essential for receptor signalling, and receptors being involved in regulation of integrin expression or activation. Moreover, there is accumulating evidence for direct binding of specific growth factors and morphogens to the ECM proteins, suggesting that ECM might spatially integrate different types of signals in a specific microenvironment, facilitating integrin/transmembrane receptors connection. These interactions are crucial in controlling a variety of cell behaviours including proliferation, survival and differentiation. The increasing interest for cell therapy in regenerative medicine has recently emphasized the role of cell-ECM adhesion as stem cell determinant. The relevance of ECM, integrins and growth factor receptor network in the establishment of stem cell niche, in maintenance of stem cells and in their differentiation will be analyzed in the present review.
Collapse
Affiliation(s)
- Maria Felice Brizzi
- Università degli Studi di Torino, Department of Medical Sciences, Torino, Italy
| | | | | |
Collapse
|
97
|
Transcriptomes of germinal zones of human and mouse fetal neocortex suggest a role of extracellular matrix in progenitor self-renewal. Proc Natl Acad Sci U S A 2012; 109:11836-41. [PMID: 22753484 DOI: 10.1073/pnas.1209647109] [Citation(s) in RCA: 243] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The expansion of the neocortex during mammalian brain evolution results primarily from an increase in neural progenitor cell divisions in its two principal germinal zones during development, the ventricular zone (VZ) and the subventricular zone (SVZ). Using mRNA sequencing, we analyzed the transcriptomes of fetal human and embryonic mouse VZ, SVZ, and cortical plate. In mouse, the transcriptome of the SVZ was more similar to that of the cortical plate than that of the VZ, whereas in human the opposite was the case, with the inner and outer SVZ being highly related to each other despite their cytoarchitectonic differences. We describe sets of genes that are up- or down-regulated in each germinal zone. These data suggest that cell adhesion and cell-extracellular matrix interactions promote the proliferation and self-renewal of neural progenitors in the developing human neocortex. Notably, relevant extracellular matrix-associated genes include distinct sets of collagens, laminins, proteoglycans, and integrins, along with specific sets of growth factors and morphogens. Our data establish a basis for identifying novel cell-type markers and open up avenues to unravel the molecular basis of neocortex expansion during evolution.
Collapse
|
98
|
Abstract
Stem cell-based therapies offer tremendous potential for skin regeneration following injury and disease. Functional stem cell units have been described throughout all layers of human skin and the collective physical and chemical microenvironmental cues that enable this regenerative potential are known as the stem cell niche. Stem cells in the hair follicle bulge, interfollicular epidermis, dermal papillae, and perivascular space have been closely investigated as model systems for niche-driven regeneration. These studies suggest that stem cell strategies for skin engineering must consider the intricate molecular and biologic features of these niches. Innovative biomaterial systems that successfully recapitulate these microenvironments will facilitate progenitor cell-mediated skin repair and regeneration.
Collapse
|
99
|
Simon R, Brylka H, Schwegler H, Venkataramanappa S, Andratschke J, Wiegreffe C, Liu P, Fuchs E, Jenkins NA, Copeland NG, Birchmeier C, Britsch S. A dual function of Bcl11b/Ctip2 in hippocampal neurogenesis. EMBO J 2012; 31:2922-36. [PMID: 22588081 PMCID: PMC3395096 DOI: 10.1038/emboj.2012.142] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 04/17/2012] [Indexed: 11/17/2022] Open
Abstract
The transcription factor Bcl11b/Ctip2 promotes hippocampal progenitor proliferation and neural differentiation in a non-cell autonomous manner by regulating the expression of the cell adhesion molecule Desmoplakin. Forebrain-specific ablation causes defective spatial learning and memory. The development of the dentate gyrus is characterized by distinct phases establishing a durable stem-cell pool required for postnatal and adult neurogenesis. Here, we report that Bcl11b/Ctip2, a zinc finger transcription factor expressed in postmitotic neurons, plays a critical role during postnatal development of the dentate gyrus. Forebrain-specific ablation of Bcl11b uncovers dual phase-specific functions of Bcl11b demonstrated by feedback control of the progenitor cell compartment as well as regulation of granule cell differentiation, leading to impaired spatial learning and memory in mutants. Surprisingly, we identified Desmoplakin as a direct transcriptional target of Bcl11b. Similarly to Bcl11b, postnatal neurogenesis and granule cell differentiation are impaired in Desmoplakin mutants. Re-expression of Desmoplakin in Bcl11b mutants rescues impaired neurogenesis, suggesting Desmoplakin to be an essential downstream effector of Bcl11b in hippocampal development. Together, our data define an important novel regulatory pathway in hippocampal development, by linking transcriptional functions of Bcl11b to Desmoplakin, a molecule known to act on cell adhesion.
Collapse
Affiliation(s)
- Ruth Simon
- Institute of Molecular and Cellular Anatomy, Ulm University, Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
The attachment of stem cells to specialized functional niches instructs stem cell maintenance, with loss of adhesion associated with differentiation driven by cell-intrinsic programs. Id transcription factors are now shown to link cell-intrinsic maintenance programs and extrinsic cues by promoting adhesion of neural stem cells to the niche.
Collapse
|