1
|
Systems biology analysis of osteogenic differentiation behavior by canine mesenchymal stem cells derived from bone marrow and dental pulp. Sci Rep 2020; 10:20703. [PMID: 33244029 PMCID: PMC7692528 DOI: 10.1038/s41598-020-77656-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Utilization of canine mesenchymal stem cells (cMSCs) for regenerating incorrigible bone diseases has been introduced. However, cMSCs harvested from different sources showed distinct osteogenicity. To clarify this, comparative proteomics-based systems biology analysis was used to analyze osteogenic differentiation behavior by cMSCs harvested from bone marrow and dental pulp. The results illustrated that canine dental pulp stem cells (cDPSCs) contained superior osteogenicity comparing with canine bone marrow-derived MSCs (cBM-MSCs) regarding alkaline phosphatase activity, matrix mineralization, and osteogenic marker expression. Global analyses by proteomics platform showed distinct protein clustering and expression pattern upon an in vitro osteogenic induction between them. Database annotation using Reactome and DAVID revealed contrast and unique expression profile of osteogenesis-related proteins, particularly on signaling pathways, cellular components and processes, and cellular metabolisms. Functional assay and hierarchical clustering for tracking protein dynamic change confirmed that cBM-MSCs required the presences of Wnt, transforming growth factor (TGF)-beta, and bone-morphogenetic protein (BMP) signaling, while cDPSCs mainly relied on BMP signaling presentation during osteogenic differentiation in vitro. Therefore, these findings illustrated the comprehensive data regarding an in vitro osteogenic differentiation behavior by cBM-MSCs and cDPSCs which is crucial for further mechanism study and the establishment of cMSC-based bone tissue engineering (BTE) for veterinary practice.
Collapse
|
2
|
Sjöqvist M, Antfolk D, Suarez-Rodriguez F, Sahlgren C. From structural resilience to cell specification - Intermediate filaments as regulators of cell fate. FASEB J 2020; 35:e21182. [PMID: 33205514 PMCID: PMC7839487 DOI: 10.1096/fj.202001627r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 12/18/2022]
Abstract
During the last decades intermediate filaments (IFs) have emerged as important regulators of cellular signaling events, ascribing IFs with functions beyond the structural support they provide. The organ and developmental stage‐specific expression of IFs regulate cell differentiation within developing or remodeling tissues. Lack of IFs causes perturbed stem cell differentiation in vasculature, intestine, nervous system, and mammary gland, in transgenic mouse models. The aberrant cell fate decisions are caused by deregulation of different stem cell signaling pathways, such as Notch, Wnt, YAP/TAZ, and TGFβ. Mutations in genes coding for IFs cause an array of different diseases, many related to stem cell dysfunction, but the molecular mechanisms remain unresolved. Here, we provide a comprehensive overview of how IFs interact with and regulate the activity, localization and function of different signaling proteins in stem cells, and how the assembly state and PTM profile of IFs may affect these processes. Identifying when, where and how IFs and cell signaling congregate, will expand our understanding of IF‐linked stem cell dysfunction during development and disease.
Collapse
Affiliation(s)
- Marika Sjöqvist
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Daniel Antfolk
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Freddy Suarez-Rodriguez
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Cecilia Sahlgren
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
3
|
Sahu I, Nanaware P, Mane M, Mulla SW, Roy S, Venkatraman P. Role of a 19S Proteasome Subunit- PSMD10 Gankyrin in Neurogenesis of Human Neural Progenitor Cells. Int J Stem Cells 2019; 12:463-473. [PMID: 31474027 PMCID: PMC6881037 DOI: 10.15283/ijsc19007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 06/21/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022] Open
Abstract
PSMD10Gankyrin, a proteasome assembly chaperone, is a widely known oncoprotein which aspects many hall mark properties of cancer. However, except proteasome assembly chaperon function its role in normal cell function remains unknown. To address this issue, we induced PSMD10Gankyrin overexpression in HEK293 cells and the resultant large-scale changes in gene expression profile were analyzed. We constituted networks from microarray data of these differentially expressed genes and carried out extensive topological analyses. The overrecurring yet consistent theme that appeared throughout analysis using varied network metrics is that all genes and interactions identified as important would be involved in neurogenesis and neuronal development. Intrigued we tested the possibility that PSMD10Gankyrin may be strongly associated with cell fate decisions that commit neural stem cells to differentiate into neurons. Overexpression of PSMD10Gankyrin in human neural progenitor cells facilitated neuronal differentiation via β-catenin Ngn1 pathway. Here for the first time we provide preliminary and yet compelling experimental evidence for the involvement of a potential oncoprotein – PSMD10Gankyrin, in neuronal differentiation.
Collapse
Affiliation(s)
- Indrajit Sahu
- Advance Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India.,Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel.,Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India
| | - Padma Nanaware
- Advance Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India.,Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India.,Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Minal Mane
- Advance Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Saim Wasi Mulla
- Advance Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India.,Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India
| | - Soumen Roy
- Department of Physics, Bose Institute, Kolkata, India
| | - Prasanna Venkatraman
- Advance Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India.,Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India
| |
Collapse
|
4
|
Hodges H, Pollock K, Stroemer P, Patel S, Stevanato L, Reuter I, Sinden J. Making Stem Cell Lines Suitable for Transplantation. Cell Transplant 2017. [DOI: 10.3727/000000007783464605] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human stem cells, progenitor cells, and cell lines have been derived from embryonic, fetal, and adult sources in the search for graft tissue suitable for the treatment of CNS disorders. An increasing number of experimental studies have shown that grafts from several sources survive, differentiate into distinct cell types, and exert positive functional effects in experimental animal models, but little attention has been given to developing cells under conditions of good manufacturing practice (GMP) that can be scaled up for mass treatment. The capacity for continued division of stem cells in culture offers the opportunity to expand their production to meet the widespread clinical demands posed by neurodegenerative diseases. However, maintaining stem cell division in culture long term, while ensuring differentiation after transplantation, requires genetic and/or oncogenetic manipulations, which may affect the genetic stability and in vivo survival of cells. This review outlines the stages, selection criteria, problems, and ultimately the successes arising in the development of conditionally immortal clinical grade stem cell lines, which divide in vitro, differentiate in vivo, and exert positive functional effects. These processes are specifically exemplified by the murine MHP36 cell line, conditionally immortalized by a temperature-sensitive mutant of the SV40 large T antigen, and cell lines transfected with the c-myc protein fused with a mutated estrogen receptor (c-mycERTAM), regulated by a tamoxifen metabolite, but the issues raised are common to all routes for the development of effective clinical grade cells.
Collapse
Affiliation(s)
- Helen Hodges
- Department of Psychology, Institute of Psychiatry, Kings College, London, UK
- ReNeuron Ltd., Guildford, Surrey, UK
| | | | | | | | | | - Iris Reuter
- Department of Psychology, Institute of Psychiatry, Kings College, London, UK
- Department of Neurology, University of Giessen and Marburg, Germany
| | | |
Collapse
|
5
|
Keratins regulate colonic epithelial cell differentiation through the Notch1 signalling pathway. Cell Death Differ 2017; 24:984-996. [PMID: 28475172 PMCID: PMC5442467 DOI: 10.1038/cdd.2017.28] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 12/30/2016] [Accepted: 02/14/2017] [Indexed: 12/22/2022] Open
Abstract
Keratins (K) are intermediate filament proteins important in stress protection and mechanical support of epithelial tissues. K8, K18 and K19 are the main colonic keratins, and K8-knockout (K8−/−) mice display a keratin dose-dependent hyperproliferation of colonic crypts and a colitis-phenotype. However, the impact of the loss of K8 on intestinal cell differentiation has so far been unknown. Here we show that K8 regulates Notch1 signalling activity and differentiation in the epithelium of the large intestine. Proximity ligation and immunoprecipitation assays demonstrate that K8 and Notch1 co-localize and interact in cell cultures, and in vivo in the colonic epithelial cells. K8 with its heteropolymeric partner K18 enhance Notch1 protein levels and activity in a dose dependent manner. The levels of the full-length Notch1 receptor (FLN), the Notch1 intracellular domain (NICD) and expression of Notch1 downstream target genes are reduced in the absence of K8, and the K8-dependent loss of Notch1 activity can be rescued with re-expression of K8/K18 in K8-knockout CRISPR/Cas9 Caco-2 cells protein levels. In vivo, K8 deletion with subsequent Notch1 downregulation leads to a shift in differentiation towards a goblet cell and enteroendocrine phenotype from an enterocyte cell fate. Furthermore, the K8−/− colonic hyperproliferation results from an increased number of transit amplifying progenitor cells in these mice. K8/K18 thus interact with Notch1 and regulate Notch1 signalling activity during differentiation of the colonic epithelium.
Collapse
|
6
|
Lindqvist J, Wistbacka N, Eriksson JE. Studying Nestin and its Interrelationship with Cdk5. Methods Enzymol 2015; 568:509-35. [PMID: 26795482 DOI: 10.1016/bs.mie.2015.09.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Current research utilizes the specific expression pattern of intermediate filaments (IF) for identifying cellular state and origin, as well as for the purpose of disease diagnosis. Nestin is commonly utilized as a specific marker and driver for CNS progenitor cell types, but in addition, nestin can be found in several mesenchymal progenitor cells, and it is constitutively expressed in a few restricted locations, such as muscle neuromuscular junctions and kidney podocytes. Alike most other members of the IF protein family, nestin filaments are dynamic, constantly being remodeled through posttranslational modifications, which alter the solubility, protein levels, and signaling capacity of the nestin filaments. Through its interactions with kinases and other signaling executors, resulting in a complex and bidirectional regulation of cell signaling events, nestin has the potential to determine whether cells divide, differentiate, migrate, or stay in place. In this review, the broad and similar roles of IFs as dynamic signaling scaffolds, is exemplified by observations of nestin functions and its interaction with the cyclin- dependent kinase 5, the atypical kinase in the family of cyclin-dependent kinases.
Collapse
Affiliation(s)
- Julia Lindqvist
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Num Wistbacka
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - John E Eriksson
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
| |
Collapse
|
7
|
Zaky AZ, Moftah MZ. Neurogenesis and growth factors expression after complete spinal cord transection in Pleurodeles waltlii. Front Cell Neurosci 2015; 8:458. [PMID: 25628538 PMCID: PMC4292736 DOI: 10.3389/fncel.2014.00458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 12/16/2014] [Indexed: 01/24/2023] Open
Abstract
Following spinal lesion, connections between the supra-spinal centers and spinal neuronal networks can be disturbed, which causes the deterioration or even the complete absence of sublesional locomotor activity. In mammals, possibilities of locomotion restoration are much reduced since descending tracts either have very poor regenerative ability or do not regenerate at all. However, in lower vertebrates, there is spontaneous locomotion recuperation after complete spinal cord transection at the mid-trunk level. This phenomenon depends on a translesional descending axon re-growth originating from the brainstem. On the other hand, cellular and molecular mechanisms underlying spinal cord regeneration and in parallel, locomotion restoration of the animal, are not well known. Fibroblast growth factor 2 (FGF-2) plays an important role in different processes such as neural induction, neuronal progenitor proliferation and their differentiation. Studies had shown an over expression of this growth factor after tail amputation. Nestin, a protein specific for intermediate filaments, is considered an early marker for neuronal precursors. It has been recently shown that its expression increases after tail transection in urodeles. Using this marker and western blots, our results show that the number of FGF-2 and FGFR2 mRNAs increases and is correlated with an increase in neurogenesis especially in the central canal lining cells immediately after lesion. This study also confirms that spinal cord re-growth through the lesion site initially follows a rostrocaudal direction. In addition to its role known in neuronal differentiation, FGF-2 could be implicated in the differentiation of ependymal cells into neuronal progenitors.
Collapse
Affiliation(s)
- Amira Z Zaky
- Biochemistry Department, Faculty of Science, Alexandria University Alexandria, Egypt
| | - Marie Z Moftah
- Zoology Department, Faculty of Science, Alexandria University Alexandria, Egypt
| |
Collapse
|
8
|
Differentiation of human umbilical cord matrix mesenchymal stem cells into neural-like progenitor cells and maturation into an oligodendroglial-like lineage. PLoS One 2014; 9:e111059. [PMID: 25357129 PMCID: PMC4214693 DOI: 10.1371/journal.pone.0111059] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 09/18/2014] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are viewed as safe, readily available and promising adult stem cells, which are currently used in several clinical trials. Additionally, their soluble-factor secretion and multi-lineage differentiation capacities place MSCs in the forefront of stem cell types with expected near-future clinical applications. In the present work MSCs were isolated from the umbilical cord matrix (Wharton's jelly) of human umbilical cord samples. The cells were thoroughly characterized and confirmed as bona-fide MSCs, presenting in vitro low generation time, high proliferative and colony-forming unit-fibroblast (CFU-F) capacity, typical MSC immunophenotype and osteogenic, chondrogenic and adipogenic differentiation capacity. The cells were additionally subjected to an oligodendroglial-oriented step-wise differentiation protocol in order to test their neural- and oligodendroglial-like differentiation capacity. The results confirmed the neural-like plasticity of MSCs, and suggested that the cells presented an oligodendroglial-like phenotype throughout the differentiation protocol, in several aspects sharing characteristics common to those of bona-fide oligodendrocyte precursor cells and differentiated oligodendrocytes.
Collapse
|
9
|
Iacopino F, Angelucci C, Piacentini R, Biamonte F, Mangiola A, Maira G, Grassi C, Sica G. Isolation of cancer stem cells from three human glioblastoma cell lines: characterization of two selected clones. PLoS One 2014; 9:e105166. [PMID: 25121761 PMCID: PMC4133365 DOI: 10.1371/journal.pone.0105166] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/21/2014] [Indexed: 11/21/2022] Open
Abstract
Cancer stem cells (CSC) were isolated via a non-adherent neurosphere assay from three glioma cell lines: LI, U87, and U373. Using a clonal assay, two clones (D2 and F11) were selected from spheres derived from LI cells and were characterized for the: expression of stem cell markers (CD133, Nestin, Musashi-1 and Sox2); proliferation; differentiation capability (determined by the expression of GalC, βIII-Tubulin and GFAP); Ca2+ signaling and tumorigenicity in nude mice. Both D2 and F11 clones expressed higher levels of all stem cell markers with respect to the parental cell line. Clones grew more slowly than LI cells with a two-fold increase in duplication time. Markers of differentiation (βIII-Tubulin and GFAP) were expressed at high levels in both LI cells and in neurospheres. The expression of Nestin, Sox2, and βIII-Tubulin was down-regulated in D2 and F11 when cultured in serum-containing medium, whereas Musashi-1 was increased. In this condition, duplication time of D2 and F11 increased without reaching that of LI cells. D2, F11 and parental cells did not express voltage-dependent Ca2+-channels but they exhibited increased intracellular Ca2+ levels in response to ATP. These Ca2+ signals were larger in LI cells and in spheres cultured in serum-containing medium, while they were smaller in serum-free medium. The ATP treatment did not affect cell proliferation. Both D2 and F11 induced the appearance of tumors when ortotopically injected in athymic nude mice at a density 50-fold lower than that of LI cells. All these data indicate that both clones have characteristics of CSC and share the same stemness properties. The findings regarding the expression of differentiation markers and Ca2+-channels show that both clones are unable to reach the terminal differentiation. Both D2 and F11 might represent a good model to improve the knowledge on CSC in glioblastoma and to identify new therapeutic approaches.
Collapse
Affiliation(s)
- Fortunata Iacopino
- Institute of Histology and Embryology, Medical School, Catholic University of the Sacred Heart, Rome, Italy
- * E-mail:
| | - Cristiana Angelucci
- Institute of Histology and Embryology, Medical School, Catholic University of the Sacred Heart, Rome, Italy
| | - Roberto Piacentini
- Institute of Human Physiology, Medical School, Catholic University of the Sacred Heart, Rome, Italy
| | - Filippo Biamonte
- Institute of Histology and Embryology, Medical School, Catholic University of the Sacred Heart, Rome, Italy
| | - Annunziato Mangiola
- Institute of Neurosurgery, Medical School, Catholic University of the Sacred Heart, Rome, Italy
| | - Giulio Maira
- Institute of Neurosurgery, Medical School, Catholic University of the Sacred Heart, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Medical School, Catholic University of the Sacred Heart, Rome, Italy
| | - Gigliola Sica
- Institute of Histology and Embryology, Medical School, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
10
|
Wang W, Wang P, Li S, Yang J, Liang X, Tang Y, Li Y, Yang R, Wu Y, Shen H. Methylprednisolone inhibits the proliferation and affects the differentiation of rat spinal cord-derived neural progenitor cells cultured in low oxygen conditions by inhibiting HIF-1α and Hes1 in vitro. Int J Mol Med 2014; 34:788-95. [PMID: 24992925 DOI: 10.3892/ijmm.2014.1835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 06/05/2014] [Indexed: 11/05/2022] Open
Abstract
Although there is much controversy over the use of methylprednisolone (MP), it is one of the main drugs used in the treatment of acute spinal cord injury (SCI). The induction of the proliferation and differentiation of endogenous neural progenitor cells (NPCs) is considered a promising mode of treatment for SCI. However, the effects of MP on spinal cord-derived endogenous NPCs in a low oxygen enviroment remain to be delineated. Thus, the aim of this study was to investigate the potential effects of MP on NPCs cultured under low oxygen conditions in vitro and to elucidate the molecular mechanisms involved. Fetal rat spinal cord-derived NPCs were harvested and divided into 4 groups: 2 groups of cells cultured under normal oxygen conditions and treated with or without MP, and 2 groups incubated in 3% O2 (low oxygen) treated in a similar manner. We found that MP induced suppressive effects on NPC proliferation even under low oxygen conditions (3% O2). The proportion of nestin-positive NPCs decreased from 51.8±2.46% to 36.17±3.55% following the addition of MP and decreased more significantly to 27.20±2.68% in the cells cultured in 3% O2. In addition, a smaller number of glial fibrillary acidic protein (GFAP)-positive cells and a greater number of microtubule-associated protein 2 (MAP2)-positive cells was observed following the addition of MP under both normal (normoxic) and low oxygen (hypoxic) conditions. In response to MP treatment, hypoxia-inducible factor-1α (HIF-1α) and the Notch signaling pathway downstream protein, Hes1, but not the upstream Notch-1 intracelluar domain (NICD), were inhibited. After blocking NICD with a γ-secretase inhibitor (DAPT) MP still inhibited the expression of Hes1. Our results provide insight into the molecular mechanisms responsible for the MP-induced inhibition of proliferation and its effects on differentiation and suggest that HIF-1α and Hes1 play an important role in this effect.
Collapse
Affiliation(s)
- Wenhao Wang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Peng Wang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Shiyuan Li
- Foshan Hospital of Sun Yat-sen University, Foshan, Guangdong 528000, P.R. China
| | - Jiewen Yang
- Biotherapy Center, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong 510120, P.R. China
| | - Xinjun Liang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yong Tang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yuxi Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Rui Yang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yanfeng Wu
- Biotherapy Center, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong 510120, P.R. China
| | - Huiyong Shen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
11
|
Kristoffersen K, Villingshøj M, Poulsen HS, Stockhausen MT. Level of Notch activation determines the effect on growth and stem cell-like features in glioblastoma multiforme neurosphere cultures. Cancer Biol Ther 2013; 14:625-37. [PMID: 23792644 PMCID: PMC3742492 DOI: 10.4161/cbt.24595] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 02/08/2013] [Accepted: 04/07/2013] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Brain cancer stem-like cells (bCSC) are cancer cells with neural stem cell (NSC)-like properties found in glioblastoma multiforme (GBM) and they are assigned a central role in tumor initiation, progression and relapse. The Notch pathway is important for maintenance and cell fate decisions in the normal NSC population. Notch signaling is often deregulated in GBM and recent results suggest that this pathway plays a significant role in bCSC as well. We therefore wished to further elucidate the role of Notch activation in GBM-derived bCSC. METHODS Human-derived GBM xenograft cells were cultured as NSC-like neurosphere cultures. Notch modulation was accomplished either by blocking the pathway using the γ-secretase inhibitor DAPT or by activating it by transfecting the cells with the constitutive active Notch-1 receptor. RESULTS GBM neurosphere cultures with high endogenous Notch activation displayed sensitivity toward Notch inhibition with regard to tumorigenic features as demonstrated by increased G0/G1 population and reduced colony formation capacity. Of the NSC-like characteristics, only the primary sphere forming potential was affected, while no effect was observed on self-renewal or differentiation. In contrast, when Notch signaling was activated a decrease in the G0/G1 population and an enhanced capability of colony formation was observed, along with increased self-renewal and de-differentiation. CONCLUSION Based on the presented results we propose that active Notch signaling plays a role for cell growth and stem cell-like features in GBM neurosphere cultures and that Notch-targeted anti-bCSC treatment could be feasible for GBM patients with high endogenous Notch pathway activation.
Collapse
Affiliation(s)
- Karina Kristoffersen
- Department of Radiation Biology; The Finsen Center, Section 6321; Copenhagen University Hospital; Copenhagen, Denmark
| | - Mette Villingshøj
- Department of Radiation Biology; The Finsen Center, Section 6321; Copenhagen University Hospital; Copenhagen, Denmark
| | - Hans Skovgaard Poulsen
- Department of Radiation Biology; The Finsen Center, Section 6321; Copenhagen University Hospital; Copenhagen, Denmark
| | - Marie-Thérése Stockhausen
- Department of Radiation Biology; The Finsen Center, Section 6321; Copenhagen University Hospital; Copenhagen, Denmark
| |
Collapse
|
12
|
Valamanesh F, Monnin J, Morand-Villeneuve N, Michel G, Zaher M, Miloudi S, Chemouni D, Jeanny JC, Versaux-Botteri C. Nestin expression in the retina of rats with inherited retinal degeneration. Exp Eye Res 2013; 110:26-34. [DOI: 10.1016/j.exer.2013.01.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 01/18/2013] [Accepted: 01/25/2013] [Indexed: 01/02/2023]
|
13
|
Mamber C, Kamphuis W, Haring NL, Peprah N, Middeldorp J, Hol EM. GFAPδ expression in glia of the developmental and adolescent mouse brain. PLoS One 2012; 7:e52659. [PMID: 23285135 PMCID: PMC3528700 DOI: 10.1371/journal.pone.0052659] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 11/20/2012] [Indexed: 11/19/2022] Open
Abstract
Glial fibrillary acidic protein (GFAP) is the major intermediate filament (IF) protein in astrocytes. In the human brain, GFAP isoforms have unique expression patterns, which indicate that they play distinct functional roles. One isoform, GFAPδ, is expressed by proliferative radial glia in the developing human brain. In the adult human, GFAPδ is a marker for neural stem cells. However, it is unknown whether GFAPδ marks the same population of radial glia and astrocytes in the developing mouse brain as it does in the developing human brain. This study characterizes the expression pattern of GFAPδ throughout mouse embryogenesis and into adolescence. Gfapδ transcripts are expressed from E12, but immunohistochemistry shows GFAPδ staining only from E18. This finding suggests a translational uncoupling. GFAPδ expression increases from E18 to P5 and then decreases until its expression plateaus around P25. During development, GFAPδ is expressed by radial glia, as denoted by the co-expression of markers like vimentin and nestin. GFAPδ is also expressed in other astrocytic populations during development. A similar pattern is observed in the adolescent mouse, where GFAPδ marks both neural stem cells and mature astrocytes. Interestingly, the Gfapδ/Gfapα transcript ratio remains stable throughout development as well as in primary astrocyte and neurosphere cultures. These data suggest that all astroglia cells in the developing and adolescent mouse brain express GFAPδ, regardless of their neurogenic capabilities. GFAPδ may be an integral component of all mouse astrocytes, but it is not a specific neural stem cell marker in mice as it is in humans.
Collapse
Affiliation(s)
- Carlyn Mamber
- Department of Astrocyte Biology & Neurodegeneration, Netherlands Institute for Neuroscience - an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
| | - Willem Kamphuis
- Department of Astrocyte Biology & Neurodegeneration, Netherlands Institute for Neuroscience - an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
| | - Nina L. Haring
- Department of Astrocyte Biology & Neurodegeneration, Netherlands Institute for Neuroscience - an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
| | - Nuzrat Peprah
- Department of Astrocyte Biology & Neurodegeneration, Netherlands Institute for Neuroscience - an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
| | - Jinte Middeldorp
- Department of Astrocyte Biology & Neurodegeneration, Netherlands Institute for Neuroscience - an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Elly M. Hol
- Department of Astrocyte Biology & Neurodegeneration, Netherlands Institute for Neuroscience - an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Stockhausen MT, Kristoffersen K, Poulsen HS. Notch signaling and brain tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 727:289-304. [PMID: 22399356 DOI: 10.1007/978-1-4614-0899-4_22] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human brain tumors are a heterogenous group of neoplasms occurring inside the cranium and the central spinal cord. In adults and children, astrocytic glioma and medulloblastoma are the most common subtypes of primary brain tumors. These tumor types are thought to arise from cells in which Notch signaling plays a fundamental role during development. Recent findings have shown that Notch signaling is dysregulated and contributes to the malignant potential of these tumors. Growing evidence point towards an important role for cancer stem cells in the initiation and maintenance of glioma and medulloblastoma. In this chapter we will cover the present findings of Notch signaling in human glioma and medulloblastoma and try to create an overall picture of its relevance in the pathogenesis of these tumors.
Collapse
|
15
|
Hyder CL, Isoniemi KO, Torvaldson ES, Eriksson JE. Insights into intermediate filament regulation from development to ageing. J Cell Sci 2011; 124:1363-72. [PMID: 21502133 DOI: 10.1242/jcs.041244] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intermediate filament (IF) proteins comprise a large family with more than 70 members. Initially, IFs were assumed to provide only structural reinforcement for the cell. However, IFs are now known to be dynamic structures that are involved in a wide range of cellular processes during all stages of life, from development to ageing, and during homeostasis and stress. This Commentary discusses some lesser-known functional and regulatory aspects of IFs. We specifically address the emerging roles of nestin in myogenesis and cancer cell migration, and examine exciting evidence on the regulation of nestin and lamin A by the notch signalling pathway, which could have repercussions for our understanding of the roles of IF proteins in development and ageing. In addition, we discuss the modulation of the post-translational modifications of neuronally expressed IFs and their protein-protein interactions, as well as IF glycosylation, which not only has a role in stress and ageing, but might also regulate IFs during development. Although many of these recent findings are still preliminary, they nevertheless open new doors to explore the functionality of the IF family of proteins.
Collapse
Affiliation(s)
- Claire L Hyder
- Turku Centre for Biotechnology, University of Turku, Turku, Finland
| | | | | | | |
Collapse
|
16
|
Patkar S, Tate R, Modo M, Plevin R, Carswell HVO. Conditionally immortalised neural stem cells promote functional recovery and brain plasticity after transient focal cerebral ischaemia in mice. Stem Cell Res 2011; 8:14-25. [PMID: 22099017 DOI: 10.1016/j.scr.2011.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 05/27/2011] [Accepted: 07/19/2011] [Indexed: 11/27/2022] Open
Abstract
Cell therapy has enormous potential to restore neurological function after stroke. The present study investigated effects of conditionally immortalised neural stem cells (ciNSCs), the Maudsley hippocampal murine neural stem cell line clone 36 (MHP36), on sensorimotor and histological outcome in mice subjected to transient middle cerebral artery occlusion (MCAO). Adult male C57BL/6 mice underwent MCAO by intraluminal thread or sham surgery and MHP36 cells or vehicle were implanted into ipsilateral cortex and caudate 2 days later. Functional recovery was assessed for 28 days using cylinder and ladder rung tests and tissue analysed for plasticity, differentiation and infarct size. MHP36-implanted animals showed accelerated and augmented functional recovery and an increase in neurons (MAP-2), synaptic plasticity (synaptophysin) and axonal projections (GAP-43) but no difference in astrocytes (GFAP), oligodendrocytes (CNPase), microglia (IBA-1) or lesion volumes when compared to vehicle group. This is the first study showing a potential functional benefit of the ciNSCs, MHP36, after focal MCAO in mice, which is probably mediated by promoting neuronal differentiation, synaptic plasticity and axonal projections and opens up opportunities for future exploitation of genetically altered mice for dissection of mechanisms of stem cell based therapy.
Collapse
Affiliation(s)
- Shalmali Patkar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | | | | | | | | |
Collapse
|
17
|
Albers AE, Chen C, Köberle B, Qian X, Klussmann JP, Wollenberg B, Kaufmann AM. Stem cells in squamous head and neck cancer. Crit Rev Oncol Hematol 2011; 81:224-40. [PMID: 21511490 DOI: 10.1016/j.critrevonc.2011.03.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 03/07/2011] [Accepted: 03/18/2011] [Indexed: 12/18/2022] Open
Abstract
The initiation and metastasis of head and neck squamous cell carcinomas (HNSCC) and other cancers have recently been related to the presence of cancer stem cells (CSC). CSC are cancer initiating, sustaining and are mostly quiescent. Specific markers that vary considerably depending on tumor type or tissue of origin characterize putative CSC. Compared to the bulk tumor mass, CSC are less sensitive to chemo- and radiotherapy and may also have low immunogenicity. Therapeutic targeting of CSC may improve clinical outcome of HNSCC which has two distinct etiologies: infection of epithelial stem cells by high-risk types of the human papillomavirus, or long-term tobacco and alcohol abuse. Recent knowledge on the role of CSC in HNSCC is reviewed and where necessary parallels to CSC of other origin are drawn to give a more comprehensive picture.
Collapse
Affiliation(s)
- Andreas E Albers
- Department of Otolaryngology and Head and Neck Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
18
|
Cardano M, Diaferia GR, Cattaneo M, Dessì SS, Long Q, Conti L, Deblasio P, Cattaneo E, Biunno I. mSEL-1L (Suppressor/enhancer Lin12-like) protein levels influence murine neural stem cell self-renewal and lineage commitment. J Biol Chem 2011; 286:18708-19. [PMID: 21454627 DOI: 10.1074/jbc.m110.210740] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Murine SEL-1L (mSEL-1L) is a key component of the endoplasmic reticulum-associated degradation pathway. It is essential during development as revealed by the multi-organ dysfunction and in uterus lethality occurring in homozygous mSEL-1L-deficient mice. Here we show that mSEL-1L is highly expressed in pluripotent embryonic stem cells and multipotent neural stem cells (NSCs) but silenced in all mature neural derivatives (i.e. astrocytes, oligodendrocytes, and neurons) by mmu-miR-183. NSCs derived from homozygous mSEL-1L-deficient embryos (mSEL-1L(-/-) NSCs) fail to proliferate in vitro, show a drastic reduction of the Notch effector HES-5, and reveal a significant down-modulation of the early neural progenitor markers PAX-6 and OLIG-2, when compared with the wild type (mSEL-1L(+/+) NSCs) counterpart. Furthermore, these cells are almost completely deprived of the neural marker Nestin, display a significant decrease of SOX-2 expression, and rapidly undergo premature astrocytic commitment and apoptosis. The data suggest severe self-renewal defects occurring in these cells probably mediated by misregulation of the Notch signaling. The results reported here denote mSEL-1L as a primitive marker with a possible involvement in the regulation of neural progenitor stemness maintenance and lineage determination.
Collapse
Affiliation(s)
- Marina Cardano
- Doctorate School of Molecular Medicine, Università degli Studi di Milano, 20100 Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Albers AE, Strauss L, Liao T, Hoffmann TK, Kaufmann AM. T cell-tumor interaction directs the development of immunotherapies in head and neck cancer. Clin Dev Immunol 2010; 2010:236378. [PMID: 21234340 PMCID: PMC3017942 DOI: 10.1155/2010/236378] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Accepted: 10/16/2010] [Indexed: 01/29/2023]
Abstract
The competent immune system controls disease effectively due to induction, function, and regulation of effector lymphocytes. Immunosurveillance is exerted mostly by cytotoxic T-lymphocytes (CTLs) while specific immune suppression is associated with tumor malignancy and progression. In squamous cell carcinoma of the head and neck, the presence, activity, but also suppression of tumor-specific CTL have been demonstrated. Functional CTL may exert a selection pressure on the tumor cells that consecutively escape by a combination of molecular and cellular evasion mechanisms. Certain of these mechanisms target antitumor effector cells directly or indirectly by affecting cells that regulate CTL function. This results in the dysfunction or apoptosis of lymphocytes and dysregulated lymphocyte homeostasis. Another important tumor-escape mechanism is to avoid recognition by dysregulation of antigen processing and presentation. Thus, both induction of functional CTL and susceptibility of the tumor and its microenvironment to become T cell targets should be considered in CTL-based immunotherapy.
Collapse
Affiliation(s)
- A. E. Albers
- Department of Otolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12200 Berlin, Germany
| | - L. Strauss
- Fondazione Humanitas per la Ricerca, 20089 Rozzano, Italy
| | - T. Liao
- Department of Otolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12200 Berlin, Germany
| | - T. K. Hoffmann
- Department of Otolaryngology, Head and Neck Surgery, Universität Essen, 45147 Essen, Germany
| | - A. M. Kaufmann
- Department of Gynecology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin and Campus Mitte, 12200 Berlin, Germany
| |
Collapse
|
20
|
Chen HC, Sytwu HK, Chang JL, Wang HW, Chen HK, Kang BH, Liu DW, Chen CH, Chao TT, Wang CH. Hypoxia enhances the stemness markers of cochlear stem/progenitor cells and expands sphere formation through activation of hypoxia-inducible factor-1 alpha. Hear Res 2010; 275:43-52. [PMID: 21147209 DOI: 10.1016/j.heares.2010.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 11/22/2010] [Accepted: 12/01/2010] [Indexed: 10/18/2022]
Abstract
Unlike neural stem cells that maintain populations in the adult brains of both rodents and humans, cochlear stem cells appear to diminish in number after birth and may become quiescent in adult mammalian cochleae. Hypoxia has been observed to promote an undifferentiated cell state in various stem cell populations; however, little is known about such an effect on cochlear stem/progenitor cells (SPCs). The aims of this study were to assess the effect of hypoxia on cochlear SPCs and to examine the impact of hypoxia-inducible factor-1 alpha (Hif-1a) on regulating such an effect. Our data demonstrate that hypoxic culturing for 24 h significantly increased sphere formation and viability of cochlear SPCs compared with those cultured under normoxic conditions. Concurrent with these proliferation promotion effects are changes in the expression of multiple stemness and cell-cycle quiescent associated gene targets, including Abcg2, nestin, p27(Kip1)and Vegf. Knockdown of Hif-1a expression by small-interfering RNA inhibited hypoxia-induced cochlear SPC expansion and resulted in downregulation of Vegf, Abcg2, and nestin and upregulation of p27(Kip1) gene expression. These results suggest that Hif-1a plays an important role in the stimulation of the proliferation of cochlear SPCs, which confers a great benefit of expanding cochlear SPCs via hypoxic conditions.
Collapse
Affiliation(s)
- Hsin-Chien Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC; Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
A quest for initiating cells of head and neck cancer and their treatment. Cancers (Basel) 2010; 2:1528-54. [PMID: 24281171 PMCID: PMC3837320 DOI: 10.3390/cancers2031528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 07/12/2010] [Accepted: 07/13/2010] [Indexed: 12/12/2022] Open
Abstract
The biology of head and neck squamous cell carcinomas (HNSCC) and other cancers have been related to cancer stem-like cells (CSC). Specific markers, which vary considerably depending on tumor type or tissue of origin, characterize CSC. CSC are cancer initiating, sustaining and mostly quiescent. Compared to bulk tumors, CSC are less sensitive to chemo- and radiotherapy and may have low immunogenicity. Therapeutic targeting of CSC may improve clinical outcome. HNSCC has two main etiologies: human papillomavirus, a virus infecting epithelial stem cells, and tobacco and alcohol abuse. Here, current knowledge of HNSCC-CSC biology is reviewed and parallels to CSC of other origin are drawn where necessary for a comprehensive picture.
Collapse
|
22
|
Morgan SC, Yasin S, Uwanogho D, Jeffries A, Price J. Positional Specification in a Neural Stem Cell Line Involves Modulation of Musashi1 Expression. Stem Cells Dev 2010; 19:579-92. [DOI: 10.1089/scd.2009.0108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Sarah C. Morgan
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| | - Shireena Yasin
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| | - Dafe Uwanogho
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| | - Aaron Jeffries
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| | - Jack Price
- Centre for the Cellular Basis of Behaviour, King’s College London, Institute of Psychiatry, Denmark Hill, London, United Kingdom
| |
Collapse
|
23
|
Lee JM, Bae JS, Jin HK. Intracerebellar transplantation of neural stem cells into mice with neurodegeneration improves neuronal networks with functional synaptic transmission. J Vet Med Sci 2010; 72:999-1009. [PMID: 20339259 DOI: 10.1292/jvms.09-0514] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent studies have shown that many kinds of stem cells are beneficial for patients suffering with neurodegenerative diseases. We investigated the effects of neural stem cell (NSC), Maudsley hippocampal clone 36 (MHP36) in the Niemann-Pick disease type C (NP-C) model mice. Herein, we demonstrate that MHP36 transplantation improves the neuropathological features without acute immune response and promotes neuronal networks with functional synaptic transmission. The number of surviving Purkinje neurons substantially increased in MHP36 transplanted NP-C mice compared with sham-transplanted NP-C mice. MHP36 significantly reduced both of astrocytic and microglial activations. We also found that these surviving Purkinje neurons have normal functional synapses with parallel fibers that have normal glutamate release probability in MHP36 transplanted NP-C mice. Furthermore, real-time PCR analysis revealed up-regulation of genes involved in both excitatory and inhibitory neurotransmission encoding subunits of the ionotropic glutamate receptors GluR2, 3 and GABAA receptor beta2. These findings suggest that NSC, MHP36 transplantation may have therapeutic effects in the treatment of NP-C and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ji Min Lee
- Stem Cell Neuroplasticity Research Group, Department of Laboratory Animal Medicine, Kyungpook National University, Daegu, Korea
| | | | | |
Collapse
|
24
|
Yu L, Baxter PA, Voicu H, Gurusiddappa S, Zhao Y, Adesina A, Man TK, Shu Q, Zhang YJ, Zhao XM, Su JM, Perlaky L, Dauser R, Chintagumpala M, Lau CC, Blaney SM, Rao PH, Leung HCE, Li XN. A clinically relevant orthotopic xenograft model of ependymoma that maintains the genomic signature of the primary tumor and preserves cancer stem cells in vivo. Neuro Oncol 2010; 12:580-94. [PMID: 20511191 DOI: 10.1093/neuonc/nop056] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Limited availability of in vitro and in vivo model systems has hampered efforts to understand tumor biology and test novel therapies for ependymoma, the third most common malignant brain tumor that occurs in children. To develop clinically relevant animal models of ependymoma, we directly injected a fresh surgical specimen from a 9-year-old patient into the right cerebrum of RAG2/severe complex immune deficiency (SCID) mice. All five mice receiving the initial transplantation of the patient tumor developed intracerebral xenografts, which have since been serially subtransplanted in vivo in mouse brains for 4 generations and can be cryopreserved for long-term maintenance of tumorigenicity. The xenograft tumors shared nearly identical histopathological features with the original tumors, harbored 8 structural chromosomal abnormalities as detected with spectral karyotyping, maintained gene expression profiles resembling that of the original patient tumor with the preservation of multiple key genetic abnormalities commonly found in human ependymomas, and contained a small population (<2.2%) of CD133(+) stem cells that can form neurospheres and display multipotent capabilities in vitro. The permanent cell line (BXD-1425EPN), which was derived from a passage II xenograft tumor and has been passaged in vitro more than 70 times, expressed similar differentiation markers of the xenograft tumors, maintained identical chromosomal abnormalities, and formed tumors in the brains of SCID mice. In conclusion, direct injection of primary ependymoma tumor cells played an important role in the generation of a clinically relevant mouse model IC-1425EPN and a novel cell line, BXD-1425EPN. This cell line and model will facilitate the biological studies and preclinical drug screenings for pediatric ependymomas.
Collapse
Affiliation(s)
- Litian Yu
- Laboratory of Molecular Neuro-Oncology, Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, 6621 Fannin Street, MC 3-3320, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bai Y, Meng Z, Cui M, Zhang X, Chen F, Xiao J, Shen L, Zhang Y. An Ang1-Tie2-PI3K axis in neural progenitor cells initiates survival responses against oxygen and glucose deprivation. Neuroscience 2009; 160:371-81. [PMID: 19409199 DOI: 10.1016/j.neuroscience.2009.01.076] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 01/23/2009] [Accepted: 01/23/2009] [Indexed: 12/25/2022]
Abstract
Neural progenitor cells (NPCs) have the potential to survive brain ischemia and participate in neurogenesis after stroke. However, it is not clear how survival responses are initiated in NPCs. Using embryonic mouse NPCs and the in vitro oxygen and glucose deprivation (OGD) model, we found that angiopoietin-1 (Ang1) could prevent NPCs from OGD-induced apoptosis, as evidenced by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and annexin V labeling. Ang1 significantly elevated tunica intima endothelial kinase 2 (Tie2) autophosphorylation level, suggesting the existence of functional Tie2 receptors on NPCs. NPCs under OGD conditions exhibited reduction of Akt phosphorylation, decrease of the Bcl-2/Bax ratio, activation of caspase-3, cleavage of PARP, and downregulation of beta-catenin and nestin. Ang1 reversed the above changes concomitantly with significant rising of survival rates of NPCs under OGD, but all these effects of Ang1 could be blocked by either soluble extracellular domain of Tie2 Fc fusion protein (sTie2Fc) or the phosphoinositide 3-kinase (PI3K) inhibitor 2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one (LY294002). Our findings suggest the existence of an Ang1-Tie2-PI3K signaling axis that is essential in initiation of survival responses in NPCs against cerebral ischemia and hypoxia.
Collapse
Affiliation(s)
- Y Bai
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, 38 XueYuan Road, Beijing, 100191, China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Uwanogho DA, Yasin SA, Starling B, Price J. The intergenic region between the Mouse Recql4 and Lrrc14 genes functions as an evolutionary conserved bidirectional promoter. Gene 2009; 449:103-17. [PMID: 19720120 DOI: 10.1016/j.gene.2009.08.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 08/13/2009] [Accepted: 08/17/2009] [Indexed: 11/25/2022]
Abstract
Mammalian genomes are highly complex, with neighbouring genes arranged in divergent, convergent, tandem, antisense, and interleaving fashions. Despite the vast genomic space, a substantial portion of human genes (approximately 10%) are arranged in a divergent, head-to-head fashion and controlled by bidirectional promoters. Here we define a small core bidirectional promoter that drives expression of the mouse genes Recql4, on one strand, and Lrrc14; a novel member of the LRR gene family, on the opposite strand. Regulation of Lrrc14 expression is highly complex, involving multiple promoters' and alternative splicing. Expression of this gene is predominately restricted to neural tissue during embryogenesis and is expressed in a wide range of tissues in the adult.
Collapse
Affiliation(s)
- D A Uwanogho
- Department of Neuroscience, Centre for the Cellular Basis of Behaviour & MRC Centre for Neurodegeneration Research, Institute of Psychiatry, King's College London, Denmark Hill, London SE5 9NU, UK.
| | | | | | | |
Collapse
|
27
|
El-Helou V, Proulx C, Béguin P, Assimakopoulos J, Gosselin H, Clement R, Villeneuve L, Huot-Marchand JÉ, DeBlois D, Lajoie C, Calderone A. The cardiac neural stem cell phenotype is compromised in streptozotocin-induced diabetic cardiomyopathy. J Cell Physiol 2009; 220:440-9. [DOI: 10.1002/jcp.21785] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
28
|
Wang Z, Li Y, Banerjee S, Sarkar FH. Emerging role of Notch in stem cells and cancer. Cancer Lett 2009; 279:8-12. [PMID: 19022563 PMCID: PMC2699045 DOI: 10.1016/j.canlet.2008.09.030] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 09/21/2008] [Accepted: 09/12/2008] [Indexed: 12/21/2022]
Abstract
The Notch signaling pathway is known to be responsible for maintaining a balance between cell proliferation and death and, as such, plays important roles in the formation of many types of human tumors. Recently, Notch signaling pathway has been shown to control stem cell self-renewal and multi-potency. As many cancers are thought to be developed from a number of cancer stem-like cells, which are also known to be linked with the acquisition of epithelial-mesenchymal transition (EMT); and thus suggesting an expanding role of Notch signaling in human tumor progression.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of Pathology, Karmanos Cancer Institute, Wayne State University School of Medicine, 9374 Scott Hall, 540 E Canfield, Detroit, MI 48201, United States
| | | | | | | |
Collapse
|
29
|
Role of ubiquitin ligases in neural stem and progenitor cells. Arch Immunol Ther Exp (Warsz) 2009; 57:177-88. [DOI: 10.1007/s00005-009-0019-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 01/30/2009] [Indexed: 01/18/2023]
|
30
|
Stachowiak EK, Roy I, Lee YW, Capacchietti M, Aletta JM, Prasad PN, Stachowiak MK. Targeting novel integrative nuclear FGFR1 signaling by nanoparticle-mediated gene transfer stimulates neurogenesis in the adult brain. Integr Biol (Camb) 2009; 1:394-403. [PMID: 20023746 DOI: 10.1039/b902617g] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neurogenesis, the process of differentiation of neuronal stem/progenitor cells (NS/PC) into mature neurons, holds the key to the treatment of various neurodegenerative disorders, which are a major health issue for the world's aging population. We report that targeting the novel integrative nuclear FGF Receptor 1 signaling (INFS) pathway enhances the latent potential of NS/PCs to undergo neuronal differentiation, thus promoting neurogenesis in the adult brain. Employing organically modified silica (ORMOSIL)-DNA nanoplexes to efficiently transfect recombinant nuclear forms of FGFR1 and its FGF-2 ligand into the brain subventricular zone, we find that INFS stimulates the NS/PC to withdraw from the cell cycle, differentiate into doublecortin expressing migratory neuroblasts and neurons that migrate to the olfactory bulb, subcortical brain regions and in the brain cortex. Thus, nanoparticle-mediated non-viral gene transfer may be used to induce selective differentiation of NS/PCs, providing a potentially significant impact on the treatment of a broad range of neurological disorders.
Collapse
Affiliation(s)
- Ewa K Stachowiak
- Molecular and Structural Neurobiology and Gene Therapy Program, Department of Pathology and Anatomical Sciences, State University of New York, Buffalo, NY 14214, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Vlashi E, Kim K, Lagadec C, Donna LD, McDonald JT, Eghbali M, Sayre JW, Stefani E, McBride W, Pajonk F. In vivo imaging, tracking, and targeting of cancer stem cells. J Natl Cancer Inst 2009; 101:350-9. [PMID: 19244169 DOI: 10.1093/jnci/djn509] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND There is increasing evidence that solid cancers contain cancer-initiating cells (CICs) that are capable of regenerating a tumor that has been surgically removed and/or treated with chemotherapy and/or radiation therapy. Currently, cell surface markers, like CD133 or CD44, are used to identify CICs in vitro; however, these markers cannot be used to identify and track CICs in vivo. The 26S proteasome is the main regulator of many processes within a proliferating cell, and its activity may be altered depending on the phenotype of a cell. METHODS Human glioma and breast cancer cells were engineered to stably express ZsGreen fused to the carboxyl-terminal degron of ornithine decarboxylase, resulting in a fluorescent fusion protein that accumulates in cells in the absence of 26S proteasome activity; activities of individual proteases were monitored in a plate reader by detecting the cleavage of fluorogenic peptide substrates. Proteasome subunit expression in cells expressing the fusion protein was assessed by quantitative reverse transcription-polymerase chain reaction, and the stem cell phenotype of CICs was assessed by a sphere formation assay, by immunohistochemical staining for known stem cell markers in vitro, and by analyzing their tumorigenicity in vivo. CICs were tracked by in vivo fluorescence imaging after radiation treatment of tumor-bearing mice and targeted specifically via a thymidine kinase-degron fusion construct. All P values were derived from two-sided tests. RESULTS Cancer cells grown as sphere cultures in conditions, which enrich for cancer stem cells (CSCs), had decreased proteasome activity relative to the respective monolayers (percent decrease in chymotryptic-like activity of sphere cultures relative to monolayers--U87MG: 26.64%, 95% confidence interval [CI] = 10.19 to 43.10, GL261, 52.91%, 95% CI = 28.38 to 77.43). The cancer cells with low proteasome activity can thus be monitored in vitro and in vivo by the accumulation of a fluorescent protein (ZsGreen) fused to a degron that targets it for 26S proteasome degradation. In vitro, ZsGreen-positive cells had increased sphere-forming capacity, expressed CSC markers, and lacked differentiation markers compared with ZsGreen-negative cells. In vivo, ZsGreen-positive cells were approximately 100-fold more tumorigenic than ZsGreen-negative cells when injected into nude mice (ZsGreen positive, 30 mice per group; ZsGreen negative, 31 mice per group), and the number of CICs in tumors increased after 72 hours post radiation treatment. CICs were selectively targeted via a proteasome-dependent suicide gene, and their elimination in vivo led to tumor regression. CONCLUSION Our results demonstrate that reduced 26S proteasome activity is a general feature of CICs that can easily be exploited to identify, track, and target them in vitro and in vivo.
Collapse
Affiliation(s)
- Erina Vlashi
- Division of Molecular and Cellular Oncology, Department of Radiation Oncology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1714, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Dell'Albani P. Stem cell markers in gliomas. Neurochem Res 2008; 33:2407-15. [PMID: 18493853 DOI: 10.1007/s11064-008-9723-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Accepted: 04/21/2008] [Indexed: 01/15/2023]
Abstract
Gliomas are the most common tumours of the central nervous system (CNS) and a frequent cause of mental impairment and death. Treatment of malignant gliomas is often palliative because of their infiltrating nature and high recurrence. Genetic events that lead to brain tumours are mostly unknown. A growing body of evidence suggests that gliomas may rise from cancer stem cells (CSC) sharing with neural stem cells (NSC) the capacity of cell renewal and multipotency. Accordingly, a population of cells called "side population" (SP), which has been isolated from gliomas on the basis of their ability to extrude fluorescent dyes, behaves as stem cells and is resistant to chemotherapeutic treatments. This review will focus on the expression of the stem cell markers nestin and CD133 in glioma cancer stem cells. In addition, the possible role of Platelet Derived Growth Factor receptor type alpha (PDGFR-alpha) and Notch signalling in normal development and tumourigenesis of gliomas are also discussed. Future work elucidating the mechanisms that control normal development will help to identify new cancer stem cell-related genes. The identification of important markers and the elucidation of signalling pathways involved in survival, proliferation and differentiation of CSCs appear to be fundamental for developing an effective therapy of brain tumours.
Collapse
Affiliation(s)
- Paola Dell'Albani
- Institute of Neurological Sciences, National Research Council (CNR), V.le Regina Margherita, 6, 95123, Catania, Italy.
| |
Collapse
|
33
|
Naujokat C, Sarić T. Concise Review: Role and Function of the Ubiquitin-Proteasome System in Mammalian Stem and Progenitor Cells. Stem Cells 2007; 25:2408-18. [PMID: 17641241 DOI: 10.1634/stemcells.2007-0255] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Highly ordered degradation of cell proteins by the ubiquitin-proteasome system, a sophisticated cellular proteolytic machinery, has been identified as a key regulatory mechanism in many eukaryotic cells. Accumulating evidence reveals that the ubiquitin-proteasome system is involved in the regulation of fundamental processes in mammalian stem and progenitor cells of embryonic, neural, hematopoietic, and mesenchymal origin. Such processes, including development, survival, differentiation, lineage commitment, migration, and homing, are directly controlled by the ubiquitin-proteasome system, either via proteolytic degradation of key regulatory proteins of signaling and gene expression pathways or via nonproteolytic mechanisms involving the proteasome itself or posttranslational modifications of target proteins by ubiquitin or other ubiquitin-like modifiers. Future characterization of the precise roles and functions of the ubiquitin-proteasome system in mammalian stem and early progenitor cells will improve our understanding of stem cell biology and may provide an experimental basis for the development of novel therapeutic strategies in regenerative medicine. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Cord Naujokat
- Institute of Immunology, University of Heidelberg, Im Neuenheimer Feld 305, D-69120 Heidelberg, Germany.
| | | |
Collapse
|
34
|
Sakairi T, Hiromura K, Yamashita S, Takeuchi S, Tomioka M, Ideura H, Maeshima A, Kaneko Y, Kuroiwa T, Nangaku M, Takeuchi T, Nojima Y. Nestin expression in the kidney with an obstructed ureter. Kidney Int 2007; 72:307-18. [PMID: 17429339 DOI: 10.1038/sj.ki.5002277] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nestin is an intermediate filament protein originally identified in neuroepithelial stem cells. This cytoskeletal-associated protein is also expressed in some non-neuronal organs including renal tubular cells and glomerular endothelial cells during kidney development. Little is known, however, about nestin expression in the kidney during injury. In this study, we find nestin expression induced in renal tubular and interstitial myofibroblasts in the adult rat kidney following unilateral ureteral obstruction. The degree of nestin expression was well correlated with the degree of tubulointerstitial fibrosis. Immunohistochemical identification of specific nephron segments showed that nestin was primarily expressed by proximal tubules, partially by distal tubules and thick ascending limbs of Henle but not by collecting ducts. The nestin-positive tubular cells also expressed vimentin and heat-shock protein 47 (HSP47) suggesting these cells reverted to a mesenchymal phenotype. Not all vimentin- or HSP-expressing cells expressed nestin; however, suggesting that nestin is distinct from these conventional mesenchymal markers. Nestin expression was also found associated with phenotypical changes in cultured renal cells induced by hypoxia or transforming growth factor-beta. Nestin expression was located in hypoxic regions of the kidney with an obstructed ureter. Our results indicate that nestin could be a novel marker for tubulointerstitial injury.
Collapse
Affiliation(s)
- T Sakairi
- Department of Medicine and Clinical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Brekke C, Morgan SC, Lowe AS, Meade TJ, Price J, Williams SCR, Modo M. The in vitro effects of a bimodal contrast agent on cellular functions and relaxometry. NMR IN BIOMEDICINE 2007; 20:77-89. [PMID: 16952123 DOI: 10.1002/nbm.1077] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The in vivo monitoring of cell survival and migration will be essential to the translation of cell-based therapies from the laboratory to clinical studies. The pre-labeling of cells with magnetic resonance imaging (MRI) contrast agents renders them visible in vivo for serial cellular imaging. However, little is known about the impact of the presence of these metal particles inside transplanted cells. The use of the bimodal contrast agent GRID made it possible to demonstrate by means of fluorescent microscopy and inductively coupled plasma mass spectrometry (ICP-MS) that, after 16 h of incubation (without the use of a transfection agent), neural stem cells (NSCs) were saturated and no longer incorporated particles. With this maximal uptake, no significant effect on cell viability was observed. However, a significant decrease in proliferation was evident in cells that underwent 24 h of labeling. A significant increase in reactive oxygen species was observed for all GRID labeling, with a very significant increase with 24 h of labeling. GRID labeling did not affect cell motility in comparison with PKH26-labeled NSCs in a glioma-based migration assay and also allowed differentiation into all major cell types of the brain. GRID-labeled cells induced a signal change of 47% on T(2) measurements and allows a detection of cell clusters of approximately 220 cells/microl. Further in vivo testing will be required to ensure that cell labeling with gadolinium-based MRI contrast agents does not impair their ability to repair.
Collapse
Affiliation(s)
- Cecilie Brekke
- NeuroImaging Research Group - Department of Neurology, Institute of Psychiatry, King's College London, UK
| | | | | | | | | | | | | |
Collapse
|
36
|
Bentz K, Molcanyi M, Hess S, Schneider A, Hescheler J, Neugebauer E, Schaefer U. Neural differentiation of embryonic stem cells is induced by signalling from non-neural niche cells. Cell Physiol Biochem 2007; 18:275-86. [PMID: 17167232 DOI: 10.1159/000097674] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2006] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Embryonic stem cell (ESC) transplantation offers new therapeutic strategies for neurodegenerative diseases and injury. However, the mechanisms underlying integration and differentiation of engrafted ESCs are poorly understood. This study elucidates the influence of exogenous signals on ESC differentiation using in vitro modelling of non-stem/stem cell interactions. METHODS Murine ESCs were co-cultured with endothelial cells and astrocytes or conditioned medium obtained from endothelial or astrocyte cultures. After 7 days of co-culture isolated RNA was analysed using RT-PCR for the expression of pluripotency marker oct-4, neural progenitor marker nestin, and neurofilament (NFL), an early marker of neuronal lineage commitment. The presence of the glial cell surface marker A2B5 was determined in ESCs by flow cytometry. RESULTS Neuronal differentiation was inhibited in ESCs when grown in close vicinity to cerebral endothelial or glial cells. Under these conditions, ESC differentiation was predominantly directed towards a glial fate. However, treatment of ESCs with endothelial cell- or astrocyte-conditioned medium promoted neuronal as well as glial differentiation. CONCLUSION Our results indicate that ESC fate is determined by endothelial and glial cells that comprise the environmental niche of these stem cells in vivo. The direction of differentiation processes appears to be dependent on humoral factors secreted by adjacent cell lines.
Collapse
Affiliation(s)
- Kristine Bentz
- Institute of Developmental Genetics, GSF - National Research Centre for Environment and Health, Munich/Neuherberg, Germany
| | | | | | | | | | | | | |
Collapse
|
37
|
Greenway DJ, Street M, Jeffries A, Buckley NJ. RE1 Silencing transcription factor maintains a repressive chromatin environment in embryonic hippocampal neural stem cells. Stem Cells 2006; 25:354-63. [PMID: 17082226 DOI: 10.1634/stemcells.2006-0207] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The control of gene expression in neural stem cells is key to understanding their developmental and therapeutic potential, yet we know little of the transcriptional mechanisms that underlie their differentiation. Recent evidence has implicated the RE1 silencing transcription factor (REST) in neuronal differentiation. However, the means by which REST regulates transcription in neural stem cells remain unclear. Here, we show that REST recruits distinct corepressor platforms in neural stem cells. REST is able to both silence and repress neuronal genes in embryonic hippocampal neural stem cells by creating a chromatin environment that contains both repressive local epigenetic signature (characterized by low levels of histones H4 and H3K9 acetylation and elevated dimethylation of H3K9) and H3K4 methylation, which are characteristic of gene activation. Furthermore, inhibition of REST function leads to activation of several neuron-specific genes but does not lead to overt formation of mature neurons, supporting the notion that REST regulates part, but not all, of the neuronal differentiation program.
Collapse
Affiliation(s)
- Deborah J Greenway
- Institute of Membrane and Systems Biology, University of Leeds, Leeds, United Kingdom
| | | | | | | |
Collapse
|
38
|
Bakshi A, Shimizu S, Keck CA, Cho S, LeBold DG, Morales D, Arenas E, Snyder EY, Watson DJ, McIntosh TK. Neural progenitor cells engineered to secrete GDNF show enhanced survival, neuronal differentiation and improve cognitive function following traumatic brain injury. Eur J Neurosci 2006; 23:2119-34. [PMID: 16630059 DOI: 10.1111/j.1460-9568.2006.04743.x] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We sought to evaluate the potential of C17.2 neural progenitor cells (NPCs) engineered to secrete glial cell line-derived neurotrophic factor (GDNF) to survive, differentiate and promote functional recovery following engraftment into the brains of adult male Sprague-Dawley rats subjected to lateral fluid percussion brain injury. First, we demonstrated continued cortical expression of GDNF receptor components (GFRalpha-1, c-Ret), suggesting that GDNF could have a physiological effect in the immediate post-traumatic period. Second, we demonstrated that GDNF over-expression reduced apoptotic NPC death in vitro. Finally, we demonstrated that GDNF over-expression improved survival, promoted neuronal differentiation of GDNF-NPCs at 6 weeks, as compared with untransduced (MT) C17.2 cells, following transplantation into the perilesional cortex of rats at 24 h post-injury, and that brain-injured animals receiving GDNF-C17.2 transplants showed improved learning compared with those receiving vehicle or MT-C17.2 cells. Our results suggest that transplantation of GDNF-expressing NPCs in the acute post-traumatic period promotes graft survival, migration, neuronal differentiation and improves cognitive outcome following traumatic brain injury.
Collapse
Affiliation(s)
- Asha Bakshi
- Traumatic Brain Injury Laboratory, Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Steiner B, Klempin F, Wang L, Kott M, Kettenmann H, Kempermann G. Type-2 cells as link between glial and neuronal lineage in adult hippocampal neurogenesis. Glia 2006; 54:805-14. [PMID: 16958090 DOI: 10.1002/glia.20407] [Citation(s) in RCA: 262] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the course of adult hippocampal neurogenesis, new cells go through a series of stages associated with proliferative activity. The most highly proliferative cell type is an intermediate precursor cell, called type-2 cell. We here report that on the level of type-2 cells a transition takes place between features associated with the glial and the neuronal lineage. We show that stem-cell marker Sox2 and radial glia marker BLBP are expressed in type-2 cells but label only a small percentage of the proliferating cells. By and large, precursor cell marker Sox2 was found to be widely expressed in hippocampal astrocytes. Between 3 h and 1 week after a single injection of permanent S-phase marker bromodeoxyuridine (BrdU), the number of BrdU-labeled BLBP-positive cells did not change, consistent with the idea that both markers here are associated with the maintained precursor cell pool. Using reporter gene mice expressing the green fluorescent protein (GFP) under the promoter for nestin we found an overlap of GFP with markers of the neuronal lineage, doublecortin (DCX) and transcription factor NeuroD1 in type-2 cells, whereas in glial fibrillary acidic protein (GFAP)-GFP mice expression of GFP and NeuroD1 or DCX was mutually exclusive. Electrophysiologically, the group of type-2 cells fell into two subgroups: one with astrocytic properties and another with an early "complex" phenotype of neural progenitor cells. Our data further support the existence of proliferative precursor cells that mark the transition between glia-like states and neuronal differentiation.
Collapse
Affiliation(s)
- Barbara Steiner
- Max-Delbrück-Center for Molecular Medicine Berlin-Buch, 13125 Berlin-Buch, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Sun YM, Greenway DJ, Johnson R, Street M, Belyaev ND, Deuchars J, Bee T, Wilde S, Buckley NJ. Distinct profiles of REST interactions with its target genes at different stages of neuronal development. Mol Biol Cell 2005; 16:5630-8. [PMID: 16195345 PMCID: PMC1289408 DOI: 10.1091/mbc.e05-07-0687] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2005] [Revised: 09/15/2005] [Accepted: 09/20/2005] [Indexed: 02/07/2023] Open
Abstract
Differentiation of pluripotent embryonic stem (ES) cells through multipotent neural stem (NS) cells into differentiated neurons is accompanied by wholesale changes in transcriptional programs. One factor that is present at all three stages and a key to neuronal differentiation is the RE1-silencing transcription factor (REST/NRSF). Here, we have used a novel chromatin immunoprecipitation-based cloning strategy (SACHI) to identify 89 REST target genes in ES cells, embryonic hippocampal NS cells and mature hippocampus. The gene products are involved in all aspects of neuronal function, especially neuronal differentiation, axonal growth, vesicular transport and release, and ionic conductance. Most target genes are silent or expressed at low levels in ES and NS cells, but are expressed at much higher levels in hippocampus. These data indicate that the REST regulon is specific to each developmental stage and support the notion that REST plays distinct roles in regulating gene expression in pluripotent ES cells, multipotent NS cells, and mature neurons.
Collapse
Affiliation(s)
- Yuh-Man Sun
- School of Biochemistry and Microbiology, University of Leeds, Leeds LS2 9JT, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Wong AM, Hodges H, Horsburgh K. Neural stem cell grafts reduce the extent of neuronal damage in a mouse model of global ischaemia. Brain Res 2005; 1063:140-50. [PMID: 16289485 DOI: 10.1016/j.brainres.2005.09.049] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2005] [Revised: 09/13/2005] [Accepted: 09/25/2005] [Indexed: 12/27/2022]
Abstract
The therapeutic potential of neural stem cell transplantation has been well demonstrated in many models of focal brain damage. However, few studies have sought to determine whether neural stem cells are therapeutic in models of diffuse brain injury, such as observed in Alzheimer's disease and global ischaemia. The present study investigated the effects of transplanted MHP36 neural stem cells on the extent of ischaemic damage in a mouse model of global ischaemia and the effects of the immunosuppressive agent cyclosporin A (CsA). C57Bl/6J mice received an intrastriatal graft of MHP36 neural stem cells 3 days after selective neuronal damage had been induced by global ischaemia. The experimental group was subdivided into CsA or saline controls. We discovered that grafts of MHP36 neural stem cells were able to differentiate into neurons and reduce the extent of ischaemic neuronal damage. This reduction was particularly apparent at 4 week post-transplantation and is independent of CsA immunosuppression. MHP36 cells survived robustly in host ischaemic brain and migrated away from the injection tract towards the caudate nucleus and corpus callosum. Although MHP36 grafts were associated with an acute inflammatory response from reactive astrocytes and microglia at 1 week post-transplantation, this decreased markedly by 4 weeks post-transplantation even in the absence of CsA immunosuppression. This is the first study showing a therapeutic benefit of neural stem cells in a highly diffuse brain injury, further highlighting the possibilities of stem cell transplantation for all types of neurodegenerative disease.
Collapse
Affiliation(s)
- Andrew M Wong
- Centre for Neuroscience Research, University of Edinburgh, 1 George Square, Edinburgh EH8 9LS, UK.
| | | | | |
Collapse
|
42
|
Machka C, Kersten M, Zobawa M, Harder A, Horsch M, Halder T, Lottspeich F, Hrabé de Angelis M, Beckers J. Identification of Dll1 (Delta1) target genes during mouse embryogenesis using differential expression profiling. Gene Expr Patterns 2005; 6:94-101. [PMID: 15979417 DOI: 10.1016/j.modgep.2005.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Revised: 03/29/2005] [Accepted: 04/19/2005] [Indexed: 11/17/2022]
Abstract
The Notch signaling pathway has pleiotropic functions during mammalian embryogenesis. It is required for the patterning and differentiation of the presomitic and somitic paraxial mesoderm and of the neural tube. We used DNA-chip expression profiling and 2D-gel electrophoresis combined with peptide mass fingerprinting to identify genes and proteins differentially regulated in E10.5 Dll1 (delta-like 1, Delta1) mutant embryos. The differential expression profiling approach identified 47 regulated transcripts and 40 differentially expressed proteins. The majority of these genes has until now not been associated with Notch signaling. Subsequent whole-mount in situ hybridization confirmed that a subset of the identified transcripts has restricted and distinct patterns of expression in E10.5 mouse embryos. For most genes these expression patterns were affected in the presomitic mesoderm, in differentiating somites of Dll1 mutant embryos and in the neural tube and cells differentiating from it. Similar effects were observed in embryos homozygous for the Headturner (Htu) and pudgy (pu) mutations, which are alleles of the Notch ligands Jag1 and Dll3. The regulated expression of a subset of the proteins was validated by immunoblots. Remarkably six of the proteins down-regulated in Dll1 mutant embryos are proteasome subunits. The large set of regulated genes identified in this differential expression profiling approach is an important resource for further functional studies.
Collapse
Affiliation(s)
- C Machka
- Institute of Experimental Genetics, GSF-National Research Center, Ingolstädter Landstr.1, 85764 Neuherberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Couillard-Despres S, Winner B, Schaubeck S, Aigner R, Vroemen M, Weidner N, Bogdahn U, Winkler J, Kuhn HG, Aigner L. Doublecortin expression levels in adult brain reflect neurogenesis. Eur J Neurosci 2005; 21:1-14. [PMID: 15654838 DOI: 10.1111/j.1460-9568.2004.03813.x] [Citation(s) in RCA: 780] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Progress in the field of neurogenesis is currently limited by the lack of tools enabling fast and quantitative analysis of neurogenesis in the adult brain. Doublecortin (DCX) has recently been used as a marker for neurogenesis. However, it was not clear whether DCX could be used to assess modulations occurring in the rate of neurogenesis in the adult mammalian central nervous system following lesioning or stimulatory factors. Using two paradigms increasing neurogenesis levels (physical activity and epileptic seizures), we demonstrate that quantification of DCX-expressing cells allows for an accurate measurement of modulations in the rate of adult neurogenesis. Importantly, we excluded induction of DCX expression during physiological or reactive gliogenesis and excluded also DCX re-expression during regenerative axonal growth. Our data validate DCX as a reliable and specific marker that reflects levels of adult neurogenesis and its modulation. We demonstrate that DCX is a valuable alternative to techniques currently used to measure the levels of neurogenesis. Importantly, in contrast to conventional techniques, analysis of neurogenesis through the detection of DCX does not require in vivo labelling of proliferating cells, thereby opening new avenues for the study of human neurogenesis under normal and pathological conditions.
Collapse
Affiliation(s)
- Sebastien Couillard-Despres
- Volkswagen-Foundation Junior Group, University of Regensburg, Franz-Josef-Strauss Allee 11, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|