1
|
García-Montaño LA, Licón-Muñoz Y, Martinez FJ, Keddari YR, Ziemke MK, Chohan MO, Piccirillo SG. Dissecting Intra-tumor Heterogeneity in the Glioblastoma Microenvironment Using Fluorescence-Guided Multiple Sampling. Mol Cancer Res 2023; 21:755-767. [PMID: 37255362 PMCID: PMC10390891 DOI: 10.1158/1541-7786.mcr-23-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/25/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
The treatment of the most aggressive primary brain tumor in adults, glioblastoma (GBM), is challenging due to its heterogeneous nature, invasive potential, and poor response to chemo- and radiotherapy. As a result, GBM inevitably recurs and only a few patients survive 5 years post-diagnosis. GBM is characterized by extensive phenotypic and genetic heterogeneity, creating a diversified genetic landscape and a network of biological interactions between subclones, ultimately promoting tumor growth and therapeutic resistance. This includes spatial and temporal changes in the tumor microenvironment, which influence cellular and molecular programs in GBM and therapeutic responses. However, dissecting phenotypic and genetic heterogeneity at spatial and temporal levels is extremely challenging, and the dynamics of the GBM microenvironment cannot be captured by analysis of a single tumor sample. In this review, we discuss the current research on GBM heterogeneity, in particular, the utility and potential applications of fluorescence-guided multiple sampling to dissect phenotypic and genetic intra-tumor heterogeneity in the GBM microenvironment, identify tumor and non-tumor cell interactions and novel therapeutic targets in areas that are key for tumor growth and recurrence, and improve the molecular classification of GBM.
Collapse
Affiliation(s)
- Leopoldo A. García-Montaño
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yamhilette Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Frank J. Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yasine R. Keddari
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of California, Merced, California
| | - Michael K. Ziemke
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Muhammad O. Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sara G.M. Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| |
Collapse
|
2
|
Huang Y, Sun W, Gao F, Ma H, Yuan T, Liu Z, Liu H, Hu J, Bai J, Zhang X, Wang R. Brain-Derived Estrogen Regulates Neurogenesis, Learning and Memory with Aging in Female Rats. BIOLOGY 2023; 12:760. [PMID: 37372046 DOI: 10.3390/biology12060760] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/18/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023]
Abstract
Although 17β-estradiol (E2) can be locally synthesized in the brain, whether and how brain-derived E2 (BDE2) impacts neurogenesis with aging is largely unclear. In this study, we examined the hippocampal neural stem cells, neurogenesis, and gliogenesis of 1, 3, 6, 14, and 18-month (Mon) female rats. Female forebrain neuronal aromatase knockout (FBN-ARO-KO) rats and letrozole-treated rats were also employed. We demonstraed that (1) the number of neural stem cells declined over 14-Mon age, and the differentiation of astrocytes and microglia markedly elevated and exhibited excessive activation. KO rats showed declines in astrocyte A2 subtype and elevation in A1 subtype at 18 Mon; (2) neurogenesis sharply dropped from 1-Mon age; (3) KO suppressed dentate gyrus (DG) neurogenesis at 1, 6 and 18 Mon. Additionally, KO and letrozole treatment led to declined neurogenesis at 1-Mon age, compared to age-matched WT controls; (4) FBN-ARO-KO inhibited CREB-BDNF activation, and decreased protein levels of neurofilament, spinophilin and PSD95. Notably, hippocampal-dependent spatial learning and memory was impaired in juvenile (1 Mon) and adulthood (6 Mon) KO rats. Taken together, we demonstrated that BDE2 plays a pivotal role for hippocampal neurogenesis, as well as learning and memory during female aging, especially in juvenile and middle age.
Collapse
Affiliation(s)
- Yuanyuan Huang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Wuxiang Sun
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Fujia Gao
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Haoran Ma
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Tao Yuan
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Zixuan Liu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Huiyu Liu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Jiewei Hu
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Jing Bai
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| | - Xin Zhang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Ruimin Wang
- Neurobiology Institute, School of Public Health, North China University of Science and Technology, Tangshan 063210, China
- School of Basic Medical Science, North China University of Science and Technology, Tangshan 063210, China
| |
Collapse
|
3
|
Dittmann NL, Torabi P, Watson AES, Yuzwa SA, Voronova A. Culture Protocol and Transcriptomic Analysis of Murine SVZ NPCs and OPCs. Stem Cell Rev Rep 2023; 19:983-1000. [PMID: 36617597 DOI: 10.1007/s12015-022-10492-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 01/10/2023]
Abstract
The mammalian adult brain contains two neural stem and precursor (NPC) niches: the subventricular zone [SVZ] lining the lateral ventricles and the subgranular zone [SGZ] in the hippocampus. From these, SVZ NPCs represent the largest NPC pool. While SGZ NPCs typically only produce neurons and astrocytes, SVZ NPCs produce neurons, astrocytes and oligodendrocytes throughout life. Of particular importance is the generation and replacement of oligodendrocytes, the only myelinating cells of the central nervous system (CNS). SVZ NPCs contribute to myelination by regenerating the parenchymal oligodendrocyte precursor cell (OPC) pool and by differentiating into oligodendrocytes in the developing and demyelinated brain. The neurosphere assay has been widely adopted by the scientific community to facilitate the study of NPCs in vitro. Here, we present a streamlined protocol for culturing postnatal and adult SVZ NPCs and OPCs from primary neurosphere cells. We characterize the purity and differentiation potential as well as provide RNA-sequencing profiles of postnatal SVZ NPCs, postnatal SVZ OPCs and adult SVZ NPCs. We show that primary neurospheres cells generated from postnatal and adult SVZ differentiate into neurons, astrocytes and oligodendrocytes concurrently and at comparable levels. SVZ OPCs are generated by subjecting primary neurosphere cells to OPC growth factors fibroblast growth factor (FGF) and platelet-derived growth factor-AA (PDGF-AA). We further show SVZ OPCs can differentiate into oligodendrocytes in the absence and presence of thyroid hormone T3. Transcriptomic analysis confirmed the identities of each cell population and revealed novel immune and signalling pathways expressed in an age and cell type specific manner.
Collapse
Affiliation(s)
- Nicole L Dittmann
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.,Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Pouria Torabi
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Adrianne E S Watson
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada. .,Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada. .,Women and Children's Health Research Institute5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, Alberta, T6G 1C9, Canada. .,Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada. .,Multiple Sclerosis Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
4
|
Cellerino M, Schiavi S, Lapucci C, Sbragia E, Boffa G, Rolla-Bigliani C, Tonelli S, Boccia D, Bruschi N, Tazza F, Franciotta D, Inglese M. In-vivo characterization of macro- and microstructural injury of the subventricular zone in relapsing-remitting and progressive multiple sclerosis. Front Neurosci 2023; 17:1112199. [PMID: 37113155 PMCID: PMC10126477 DOI: 10.3389/fnins.2023.1112199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/15/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction The subventricular zone (SVZ) represents one of the main adult brain neurogenesis niche. In-vivo imaging of SVZ is very challenging and little is known about MRI correlates of SVZ macro- and micro-structural injury in multiple sclerosis (MS) patients. Methods The aim of the present study is to evaluate differences in terms of volume and microstructural changes [as assessed with the novel Spherical Mean Technique (SMT) model, evaluating: Neurite Signal fraction (INTRA); Extra-neurite transverse (EXTRATRANS) and mean diffusivity (EXTRAMD)] in SVZ between relapsing-remitting (RR) or progressive (P) MS patients and healthy controls (HC). We are also going to explore whether SVZ microstructural injury correlate with caudate (a nucleus that is in the vicinity of the SVZ) or thalamus (another well-defined grey matter area which is further from SVZ than caudate) volume and clinical disability. Clinical and brain MRI data were prospectively acquired from 20 HC, 101 RRMS, and 50 PMS patients. Structural and diffusion metrics inside the global SVZ, normal appearing (NA-) SVZ, caudate and thalamus were collected. Results We found a statistically significant difference between groups in terms of NA-SVZ EXTRAMD (PMS>RRMS>HC; p = 0.002), EXTRATRANS (PMS>RRMS>HC; p<0.0001), and INTRA (HC>RRMS>PMS; p = 0.009). Multivariable models showed that NA-SVZ metrics significantly predicted caudate (R 2 = 0.21, p < 0.0001), but not thalamus, atrophy. A statistically significant correlation between EXTRAMD and EXTRATRANS of the NA-SVZ and EDSS (r=0.25, p=0.003 and r=0.24, p = 0.003, respectively) was found. These findings were confirmed in analyses restricted to RRMS, but not to PMS patients. Discussion In conclusion, the microstructural damage we observed within the NA-SVZ of MS patients - reflecting higher free water content (higher EXTRAMD), cytoarchitecture disruption and astrogliosis (higher EXTRATRANS and lower INTRA) - was more evident in the progressive as compared to the relapsing phases of MS. These abnormalities were significantly associated with a more pronounced caudate atrophy and higher clinical disability scores. Our findings may support the neuroprotective role of SVZ in MS patients.
Collapse
Affiliation(s)
- Maria Cellerino
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Simona Schiavi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Caterina Lapucci
- Laboratory of Experimental Neurosciences, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Elvira Sbragia
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Giacomo Boffa
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | | | - Serena Tonelli
- Department of Neuroradiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Daniele Boccia
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Nicolò Bruschi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Francesco Tazza
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Diego Franciotta
- Neuroimmunology Research Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Matilde Inglese
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
- Department of Neurology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- *Correspondence: Matilde Inglese,
| |
Collapse
|
5
|
Ahmed T. Neural stem cell engineering for the treatment of multiple sclerosis. BIOMEDICAL ENGINEERING ADVANCES 2022. [DOI: 10.1016/j.bea.2022.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
6
|
Emamnejad R, Dass M, Mahlis M, Bozkurt S, Ye S, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. Thyroid hormone-dependent oligodendroglial cell lineage genomic and non-genomic signaling through integrin receptors. Front Pharmacol 2022; 13:934971. [PMID: 36133808 PMCID: PMC9483185 DOI: 10.3389/fphar.2022.934971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous autoimmune disease whereby the pathological sequelae evolve from oligodendrocytes (OLs) within the central nervous system and are targeted by the immune system, which causes widespread white matter pathology and results in neuronal dysfunction and neurological impairment. The progression of this disease is facilitated by a failure in remyelination following chronic demyelination. One mediator of remyelination is thyroid hormone (TH), whose reliance on monocarboxylate transporter 8 (MCT8) was recently defined. MCT8 facilitates the entry of THs into oligodendrocyte progenitor cell (OPC) and pre-myelinating oligodendrocytes (pre-OLs). Patients with MS may exhibit downregulated MCT8 near inflammatory lesions, which emphasizes an inhibition of TH signaling and subsequent downstream targeted pathways such as phosphoinositide 3-kinase (PI3K)-Akt. However, the role of the closely related mammalian target of rapamycin (mTOR) in pre-OLs during neuroinflammation may also be central to the remyelination process and is governed by various growth promoting signals. Recent research indicates that this may be reliant on TH-dependent signaling through β1-integrins. This review identifies genomic and non-genomic signaling that is regulated through mTOR in TH-responsive pre-OLs and mature OLs in mouse models of MS. This review critiques data that implicates non-genomic Akt and mTOR signaling in response to TH-dependent integrin receptor activation in pre-OLs. We have also examined whether this can drive remyelination in the context of neuroinflammation and associated sequelae. Importantly, we outline how novel therapeutic small molecules are being designed to target integrin receptors on oligodendroglial lineage cells and whether these are viable therapeutic options for future use in clinical trials for MS.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Michael Mahlis
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Paschalis Theotokis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
- *Correspondence: Steven Petratos,
| |
Collapse
|
7
|
Fang M, Tang T, Qiu M, Xu X. Hedgehog Signaling in CNS Remyelination. Cells 2022; 11:cells11142260. [PMID: 35883703 PMCID: PMC9320235 DOI: 10.3390/cells11142260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 12/14/2022] Open
Abstract
Remyelination is a fundamental repair process in the central nervous system (CNS) that is triggered by demyelinating events. In demyelinating diseases, oligodendrocytes (OLs) are targeted, leading to myelin loss, axonal damage, and severe functional impairment. While spontaneous remyelination often fails in the progression of demyelinating diseases, increased understanding of the mechanisms and identification of targets that regulate myelin regeneration becomes crucial. To date, several signaling pathways have been implicated in the remyelination process, including the Hedgehog (Hh) signaling pathway. This review summarizes the current data concerning the complicated roles of the Hh signaling pathway in the context of remyelination. We will highlight the open issues that have to be clarified prior to bringing molecules targeting the Hh signaling to demyelinating therapy.
Collapse
Affiliation(s)
- Minxi Fang
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China;
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tao Tang
- Department of Anatomy, Cell Biology & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
| | - Mengsheng Qiu
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China;
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- School of Basic Medicial Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (M.Q.); (X.X.)
| | - Xiaofeng Xu
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China;
- Correspondence: (M.Q.); (X.X.)
| |
Collapse
|
8
|
Beiriger J, Habib A, Jovanovich N, Kodavali CV, Edwards L, Amankulor N, Zinn PO. The Subventricular Zone in Glioblastoma: Genesis, Maintenance, and Modeling. Front Oncol 2022; 12:790976. [PMID: 35359410 PMCID: PMC8960165 DOI: 10.3389/fonc.2022.790976] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is a malignant tumor with a median survival rate of 15-16 months with standard care; however, cases of successful treatment offer hope that an enhanced understanding of the pathology will improve the prognosis. The cell of origin in GBM remains controversial. Recent evidence has implicated stem cells as cells of origin in many cancers. Neural stem/precursor cells (NSCs) are being evaluated as potential initiators of GBM tumorigenesis. The NSCs in the subventricular zone (SVZ) have demonstrated similar molecular profiles and share several distinctive characteristics to proliferative glioblastoma stem cells (GSCs) in GBM. Genomic and proteomic studies comparing the SVZ and GBM support the hypothesis that the tumor cells and SVZ cells are related. Animal models corroborate this connection, demonstrating migratory patterns from the SVZ to the tumor. Along with laboratory and animal research, clinical studies have demonstrated improved progression-free survival in patients with GBM after radiation to the ipsilateral SVZ. Additionally, key genetic mutations in GBM for the most part carry regulatory roles in the SVZ as well. An exciting avenue towards SVZ modeling and determining its role in gliomagenesis in the human context is human brain organoids. Here we comprehensively discuss and review the role of the SVZ in GBM genesis, maintenance, and modeling.
Collapse
Affiliation(s)
- Jamison Beiriger
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Ahmed Habib
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nicolina Jovanovich
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Chowdari V. Kodavali
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Lincoln Edwards
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nduka Amankulor
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Pascal O. Zinn
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| |
Collapse
|
9
|
Del Giovane A, Russo M, Tirou L, Faure H, Ruat M, Balestri S, Sposato C, Basoli F, Rainer A, Kassoussi A, Traiffort E, Ragnini-Wilson A. Smoothened/AMP-Activated Protein Kinase Signaling in Oligodendroglial Cell Maturation. Front Cell Neurosci 2022; 15:801704. [PMID: 35082605 PMCID: PMC8784884 DOI: 10.3389/fncel.2021.801704] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
The regeneration of myelin is known to restore axonal conduction velocity after a demyelinating event. Remyelination failure in the central nervous system contributes to the severity and progression of demyelinating diseases such as multiple sclerosis. Remyelination is controlled by many signaling pathways, such as the Sonic hedgehog (Shh) pathway, as shown by the canonical activation of its key effector Smoothened (Smo), which increases the proliferation of oligodendrocyte precursor cells via the upregulation of the transcription factor Gli1. On the other hand, the inhibition of Gli1 was also found to promote the recruitment of a subset of adult neural stem cells and their subsequent differentiation into oligodendrocytes. Since Smo is also able to transduce Shh signals via various non-canonical pathways such as the blockade of Gli1, we addressed the potential of non-canonical Smo signaling to contribute to oligodendroglial cell maturation in myelinating cells using the non-canonical Smo agonist GSA-10, which downregulates Gli1. Using the Oli-neuM cell line, we show that GSA-10 promotes Gli2 upregulation, MBP and MAL/OPALIN expression via Smo/AMP-activated Protein Kinase (AMPK) signaling, and efficiently increases the number of axonal contact/ensheathment for each oligodendroglial cell. Moreover, GSA-10 promotes the recruitment and differentiation of oligodendroglial progenitors into the demyelinated corpus callosum in vivo. Altogether, our data indicate that non-canonical signaling involving Smo/AMPK modulation and Gli1 downregulation promotes oligodendroglia maturation until axon engagement. Thus, GSA-10, by activation of this signaling pathway, represents a novel potential remyelinating agent.
Collapse
Affiliation(s)
- Alice Del Giovane
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Mariagiovanna Russo
- CNRS, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Saclay, France
| | - Linda Tirou
- CNRS, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Saclay, France
| | - Hélène Faure
- CNRS, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Saclay, France
| | - Martial Ruat
- CNRS, Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, Saclay, France
| | - Sonia Balestri
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Carola Sposato
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Francesco Basoli
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Alberto Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
- Institute of Nanotechnology (NANOTEC), National Research Council, Lecce, Italy
| | | | - Elisabeth Traiffort
- INSERM, U1195, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- *Correspondence: Elisabeth Traiffort,
| | | |
Collapse
|
10
|
Lespay-Rebolledo C, Tapia-Bustos A, Perez-Lobos R, Vio V, Casanova-Ortiz E, Farfan-Troncoso N, Zamorano-Cataldo M, Redel-Villarroel M, Ezquer F, Quintanilla ME, Israel Y, Morales P, Herrera-Marschitz M. Sustained Energy Deficit Following Perinatal Asphyxia: A Shift towards the Fructose-2,6-bisphosphatase (TIGAR)-Dependent Pentose Phosphate Pathway and Postnatal Development. Antioxidants (Basel) 2021; 11:74. [PMID: 35052577 PMCID: PMC8773255 DOI: 10.3390/antiox11010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Labor and delivery entail a complex and sequential metabolic and physiologic cascade, culminating in most circumstances in successful childbirth, although delivery can be a risky episode if oxygen supply is interrupted, resulting in perinatal asphyxia (PA). PA causes an energy failure, leading to cell dysfunction and death if re-oxygenation is not promptly restored. PA is associated with long-term effects, challenging the ability of the brain to cope with stressors occurring along with life. We review here relevant targets responsible for metabolic cascades linked to neurodevelopmental impairments, that we have identified with a model of global PA in rats. Severe PA induces a sustained effect on redox homeostasis, increasing oxidative stress, decreasing metabolic and tissue antioxidant capacity in vulnerable brain regions, which remains weeks after the insult. Catalase activity is decreased in mesencephalon and hippocampus from PA-exposed (AS), compared to control neonates (CS), in parallel with increased cleaved caspase-3 levels, associated with decreased glutathione reductase and glutathione peroxidase activity, a shift towards the TIGAR-dependent pentose phosphate pathway, and delayed calpain-dependent cell death. The brain damage continues long after the re-oxygenation period, extending for weeks after PA, affecting neurons and glial cells, including myelination in grey and white matter. The resulting vulnerability was investigated with organotypic cultures built from AS and CS rat newborns, showing that substantia nigra TH-dopamine-positive cells from AS were more vulnerable to 1 mM of H2O2 than those from CS animals. Several therapeutic strategies are discussed, including hypothermia; N-acetylcysteine; memantine; nicotinamide, and intranasally administered mesenchymal stem cell secretomes, promising clinical translation.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Andrea Tapia-Bustos
- School of Pharmacy, Faculty of Medicine, Universidad Andres Bello, Santiago 8370149, Chile;
| | - Ronald Perez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Valentina Vio
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Nancy Farfan-Troncoso
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Marta Zamorano-Cataldo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Martina Redel-Villarroel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Maria Elena Quintanilla
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Yedy Israel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| |
Collapse
|
11
|
Tapia-Bustos A, Lespay-Rebolledo C, Vío V, Pérez-Lobos R, Casanova-Ortiz E, Ezquer F, Herrera-Marschitz M, Morales P. Neonatal Mesenchymal Stem Cell Treatment Improves Myelination Impaired by Global Perinatal Asphyxia in Rats. Int J Mol Sci 2021; 22:ijms22063275. [PMID: 33806988 PMCID: PMC8004671 DOI: 10.3390/ijms22063275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/07/2021] [Accepted: 03/15/2021] [Indexed: 01/09/2023] Open
Abstract
The effect of perinatal asphyxia (PA) on oligodendrocyte (OL), neuroinflammation, and cell viability was evaluated in telencephalon of rats at postnatal day (P)1, 7, and 14, a period characterized by a spur of neuronal networking, evaluating the effect of mesenchymal stem cell (MSCs)-treatment. The issue was investigated with a rat model of global PA, mimicking a clinical risk occurring under labor. PA was induced by immersing fetus-containing uterine horns into a water bath for 21 min (AS), using sibling-caesarean-delivered fetuses (CS) as controls. Two hours after delivery, AS and CS neonates were injected with either 5 μL of vehicle (10% plasma) or 5 × 104 MSCs into the lateral ventricle. Samples were assayed for myelin-basic protein (MBP) levels; Olig-1/Olig-2 transcriptional factors; Gglial phenotype; neuroinflammation, and delayed cell death. The main effects were observed at P7, including: (i) A decrease of MBP-immunoreactivity in external capsule, corpus callosum, cingulum, but not in fimbriae of hippocampus; (ii) an increase of Olig-1-mRNA levels; (iii) an increase of IL-6-mRNA, but not in protein levels; (iv) an increase in cell death, including OLs; and (v) MSCs treatment prevented the effect of PA on myelination, OLs number, and cell death. The present findings show that PA induces regional- and developmental-dependent changes on myelination and OLs maturation. Neonatal MSCs treatment improves survival of mature OLs and myelination in telencephalic white matter.
Collapse
Affiliation(s)
- Andrea Tapia-Bustos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
- Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago 8370149, Chile
| | - Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Valentina Vío
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Ronald Pérez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago 7710162, Chile;
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
- Correspondence: (M.H.-M.); (P.M.); Tel.: +56-229786788 (M.H.-M. & P.M.)
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Correspondence: (M.H.-M.); (P.M.); Tel.: +56-229786788 (M.H.-M. & P.M.)
| |
Collapse
|
12
|
Bobinger T, Roeder SS, Spruegel MI, Froehlich K, Beuscher VD, Hoelter P, Lücking H, Corbeil D, Huttner HB. Variation of membrane particle-bound CD133 in cerebrospinal fluid of patients with subarachnoid and intracerebral hemorrhage. J Neurosurg 2021; 134:600-607. [PMID: 31978876 DOI: 10.3171/2019.11.jns191861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/25/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Previous studies have demonstrated that human CSF contains membrane particles carrying the stem cell antigenic marker CD133 (prominin-1). Here, the authors analyzed the variation of the amount of these CD133-positive particles in the CSF of patients with subarachnoid hemorrhage (SAH) and intracerebral hemorrhage (ICH). METHODS Consecutive CSF samples from 47 patients with SAH or ICH were compared to 14 healthy control patients. After differential ultracentrifugation of CSF, the membrane particle fraction was separated on gel electrophoresis and its CD133 content was probed by immunoblotting using the mouse monoclonal antibody 80B258 directed against human CD133. The antigen-antibody complexes were detected by chemiluminescence reagents and quantified using human Caco-2 cell extract as positive control with a standardized curve. RESULTS As compared to healthy controls (6.3 ± 0.5 ng of bound CD133 antibody; n = 14), the amount of membrane particle-associated CD133 immunoreactivities was significantly elevated in patients with SAH and ICH (38.2 ± 6.6 ng and 61.3 ± 11.0 ng [p < 0.001] for SAH [n = 18] and ICH [n = 29], respectively). In both groups the CD133 level dropped during the first 7 days (i.e., day 5-7: SAH group, 24.6 ± 10.1 ng [p = 0.06]; ICH group, 25.0 ± 4.8 ng [p = 0.002]). Whereas changes in the amount of CD133-positive membrane particles between admission and day 5-7 were not associated with clinical outcomes in patients with ICH (modified Rankin Scale [mRS] scores 0-3, -30.9 ± 12.8 ng vs mRS scores 4-6, -21.8 ± 10.7 ng; p = 0.239), persistent elevation of CD133 in patients with SAH was related to impaired functional outcome 3 months after ictus (mRS scores 0-2, -29.9 ± 8.1 ng vs mRS scores 3-6, 7.6 ± 20.3 ng; p = 0.027). These data are expressed as the mean ± standard error of the mean (SEM). CONCLUSIONS Levels of membrane particle-associated CD133 in the CSF of patients with SAH and ICH are significantly increased in comparison to healthy patients, and they decline during the hospital stay. Specifically, the persistent elevation of CD133-positive membrane particles within the first week may represent a possible surrogate measure for impaired functional outcome in patients with SAH.
Collapse
Affiliation(s)
| | | | | | | | | | - Philip Hoelter
- 2Neuroradiology, Friedrich-Alexander University Erlangen (FAU); and
| | - Hannes Lücking
- 2Neuroradiology, Friedrich-Alexander University Erlangen (FAU); and
| | - Denis Corbeil
- 3Biotechnology Center (BIOTEC), Technische Universität Dresden, Germany
| | | |
Collapse
|
13
|
Voshart DC, Wiedemann J, van Luijk P, Barazzuol L. Regional Responses in Radiation-Induced Normal Tissue Damage. Cancers (Basel) 2021; 13:cancers13030367. [PMID: 33498403 PMCID: PMC7864176 DOI: 10.3390/cancers13030367] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Normal tissue side effects remain a major concern in radiotherapy. The improved precision of radiation dose delivery of recent technological developments in radiotherapy has the potential to reduce the radiation dose to organ regions that contribute the most to the development of side effects. This review discusses the contribution of regional variation in radiation responses in several organs. In the brain, various regions were found to contribute to radiation-induced neurocognitive dysfunction. In the parotid gland, the region containing the major ducts was found to be critical in hyposalivation. The heart and lung were each found to exhibit regional responses while also mutually affecting each other's response to radiation. Sub-structures critical for the development of side effects were identified in the pancreas and bladder. The presence of these regional responses is based on a non-uniform distribution of target cells or sub-structures critical for organ function. These characteristics are common to most organs in the body and we therefore hypothesize that regional responses in radiation-induced normal tissue damage may be a shared occurrence. Further investigations will offer new opportunities to reduce normal tissue side effects of radiotherapy using modern and high-precision technologies.
Collapse
Affiliation(s)
- Daniëlle C. Voshart
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Julia Wiedemann
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Peter van Luijk
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Correspondence: (P.v.L.); (L.B.)
| | - Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (D.C.V.); (J.W.)
- Department of Biomedical Sciences of Cells & Systems–Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Correspondence: (P.v.L.); (L.B.)
| |
Collapse
|
14
|
Melià-Sorolla M, Castaño C, DeGregorio-Rocasolano N, Rodríguez-Esparragoza L, Dávalos A, Martí-Sistac O, Gasull T. Relevance of Porcine Stroke Models to Bridge the Gap from Pre-Clinical Findings to Clinical Implementation. Int J Mol Sci 2020; 21:ijms21186568. [PMID: 32911769 PMCID: PMC7555414 DOI: 10.3390/ijms21186568] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/18/2022] Open
Abstract
In the search of animal stroke models providing translational advantages for biomedical research, pigs are large mammals with interesting brain characteristics and wide social acceptance. Compared to rodents, pigs have human-like highly gyrencephalic brains. In addition, increasingly through phylogeny, animals have more sophisticated white matter connectivity; thus, ratios of white-to-gray matter in humans and pigs are higher than in rodents. Swine models provide the opportunity to study the effect of stroke with emphasis on white matter damage and neuroanatomical changes in connectivity, and their pathophysiological correlate. In addition, the subarachnoid space surrounding the swine brain resembles that of humans. This allows the accumulation of blood and clots in subarachnoid hemorrhage models mimicking the clinical condition. The clot accumulation has been reported to mediate pathological mechanisms known to contribute to infarct progression and final damage in stroke patients. Importantly, swine allows trustworthy tracking of brain damage evolution using the same non-invasive multimodal imaging sequences used in the clinical practice. Moreover, several models of comorbidities and pathologies usually found in stroke patients have recently been established in swine. We review here ischemic and hemorrhagic stroke models reported so far in pigs. The advantages and limitations of each model are also discussed.
Collapse
Affiliation(s)
- Marc Melià-Sorolla
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute, 08916 Badalona, Catalonia, Spain; (M.M.-S.); (N.D.-R.)
| | - Carlos Castaño
- Neurointerventional Radiology Unit, Department of Neurosciences, Hospital Germans Trias i Pujol, 08916 Badalona, Catalonia, Spain;
| | - Núria DeGregorio-Rocasolano
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute, 08916 Badalona, Catalonia, Spain; (M.M.-S.); (N.D.-R.)
| | - Luis Rodríguez-Esparragoza
- Stroke Unit, Department of Neurology, Hospital Germans Trias i Pujol, 08916 Badalona, Catalonia, Spain; (L.R.-E.); (A.D.)
| | - Antoni Dávalos
- Stroke Unit, Department of Neurology, Hospital Germans Trias i Pujol, 08916 Badalona, Catalonia, Spain; (L.R.-E.); (A.D.)
| | - Octavi Martí-Sistac
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute, 08916 Badalona, Catalonia, Spain; (M.M.-S.); (N.D.-R.)
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08916 Bellaterra, Catalonia, Spain
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Carretera del Canyet, Camí de les Escoles s/n, Edifici Mar, 08916 Badalona, Catalonia, Spain
- Correspondence: (O.M.-S.); (T.G.); Tel.: +34-930330531 (O.M.-S.)
| | - Teresa Gasull
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute, 08916 Badalona, Catalonia, Spain; (M.M.-S.); (N.D.-R.)
- Fundació Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Carretera del Canyet, Camí de les Escoles s/n, Edifici Mar, 08916 Badalona, Catalonia, Spain
- Correspondence: (O.M.-S.); (T.G.); Tel.: +34-930330531 (O.M.-S.)
| |
Collapse
|
15
|
Deboux C, Spigoni G, Caillava C, Garcia-Diaz B, Ypsilanti A, Sarrazin N, Bachelin C, Chédotal A, Baron-Van Evercooren A. Slit1 Protein Regulates SVZ-Derived Precursor Mobilization in the Adult Demyelinated CNS. Front Cell Neurosci 2020; 14:168. [PMID: 32670024 PMCID: PMC7332780 DOI: 10.3389/fncel.2020.00168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/19/2020] [Indexed: 01/03/2023] Open
Abstract
Slit1 is a secreted axon guidance molecule, also involved in adult neurogenesis. In physiological conditions, Slit1 loss promotes ectopic dispersal of SVZ-derived neural precursors (SVZ-NPCs) into periventricular structures such as the corpus callosum. Demyelination of the corpus callosum triggers SVZ-NPC migration to ectopic locations and their recruitment by the lesion, suggesting a possible role for Slit1 in SVZ-NPCs ectopic dispersal regulation in pathological conditions. Here, we have investigated the function of Slit1 protein in the recruitment of SVZ-NPCs after CNS demyelination. We find that the dynamics of oligodendrogenesis and temporal profile of developmental myelination in Slit1–/– mice are similar to Slit1+/− controls. SVZ micro-dissection and RT-PCR from wild-type mice, show that Slits and Robos are physiologically regulated at the transcriptional level in response to corpus callosum demyelination suggesting their role in the process of SVZ-NPC ectopic migration in demyelinating conditions. Moreover, we find that the number of SVZ-NPCs recruited by the lesion increases in Sli1–/– mice compared to Slit1+/− mice, leading to higher numbers of Olig2+ cells within the lesion. Time-lapse video-microscopy of immuno-purified NPCs shows that Slit1-deficient cells migrate faster and make more frequent directional changes than control NPCs, supporting a cell-autonomous mechanism of action of Slit1 in NPC migration. In conclusion, while Slit1 does not affect the normal developmental process of oligodendrogenesis and myelination, it regulates adult SVZ-NPC ectopic migration in response to demyelination, and consequently oligodendrocyte renewal within the lesion.
Collapse
Affiliation(s)
- C Deboux
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - G Spigoni
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - C Caillava
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - B Garcia-Diaz
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - A Ypsilanti
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - N Sarrazin
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - C Bachelin
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - A Chédotal
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - A Baron-Van Evercooren
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| |
Collapse
|
16
|
Secretome Analysis of Mesenchymal Stem Cell Factors Fostering Oligodendroglial Differentiation of Neural Stem Cells In Vivo. Int J Mol Sci 2020; 21:ijms21124350. [PMID: 32570968 PMCID: PMC7352621 DOI: 10.3390/ijms21124350] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cell (MSC)-secreted factors have been shown to significantly promote oligodendrogenesis from cultured primary adult neural stem cells (aNSCs) and oligodendroglial precursor cells (OPCs). Revealing underlying mechanisms of how aNSCs can be fostered to differentiate into a specific cell lineage could provide important insights for the establishment of novel neuroregenerative treatment approaches aiming at myelin repair. However, the nature of MSC-derived differentiation and maturation factors acting on the oligodendroglial lineage has not been identified thus far. In addition to missing information on active ingredients, the degree to which MSC-dependent lineage instruction is functional in vivo also remains to be established. We here demonstrate that MSC-derived factors can indeed stimulate oligodendrogenesis and myelin sheath generation of aNSCs transplanted into different rodent central nervous system (CNS) regions, and furthermore, we provide insights into the underlying mechanism on the basis of a comparative mass spectrometry secretome analysis. We identified a number of secreted proteins known to act on oligodendroglia lineage differentiation. Among them, the tissue inhibitor of metalloproteinase type 1 (TIMP-1) was revealed to be an active component of the MSC-conditioned medium, thus validating our chosen secretome approach.
Collapse
|
17
|
Zilkha-Falb R, Kaushansky N, Ben-Nun A. The Median Eminence, A New Oligodendrogenic Niche in the Adult Mouse Brain. Stem Cell Reports 2020; 14:1076-1092. [PMID: 32413277 PMCID: PMC7355143 DOI: 10.1016/j.stemcr.2020.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/17/2022] Open
Abstract
The subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus in the hippocampus are known as neurogenic niches. We show that the median eminence (ME) of the hypothalamus comprises BrdU+ newly proliferating cells co-expressing NG2 (oligodendrocyte progenitors) and RIP (pre-myelinating oligodendrocytes), suggesting their differentiation toward mature oligodendrocytes (OLs). ME cells can generate neurospheres (NS) in vitro, which differentiate mostly to OLs compared with SVZ-NS that typically generate neurons. Interestingly, this population of oligodendrocyte progenitors is increased in the ME from experimental autoimmune encephalomyelitis (EAE)-affected mice. Notably, the thrombospondin 1 (TSP1) expressed by astrocytes, acts as negative regulator of oligodendrogenesis in vitro and is downregulated in the ME of EAE mice. Importantly, transplanted ME-NS preferentially differentiate to MBP+ OLs compared with SVZ-NS in Shiverer mice. Hence, discovering the ME as a new site for myelin-producing cells has a great importance for advising future therapy for demyelinating diseases and spinal cord injury.
Collapse
Affiliation(s)
- Rina Zilkha-Falb
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| | - Nathali Kaushansky
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Avraham Ben-Nun
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
18
|
Güven A, Kalebic N, Long KR, Florio M, Vaid S, Brandl H, Stenzel D, Huttner WB. Extracellular matrix-inducing Sox9 promotes both basal progenitor proliferation and gliogenesis in developing neocortex. eLife 2020; 9:49808. [PMID: 32191207 PMCID: PMC7105383 DOI: 10.7554/elife.49808] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
Neocortex expansion is largely based on the proliferative capacity of basal progenitors (BPs), which is increased by extracellular matrix (ECM) components via integrin signaling. Here we show that the transcription factor Sox9 drives expression of ECM components and that laminin 211 increases BP proliferation in embryonic mouse neocortex. We show that Sox9 is expressed in human and ferret BPs and is required for BP proliferation in embryonic ferret neocortex. Conditional Sox9 expression in the mouse BP lineage, where it normally is not expressed, increases BP proliferation, reduces Tbr2 levels and induces Olig2 expression, indicative of premature gliogenesis. Conditional Sox9 expression also results in cell-non-autonomous stimulation of BP proliferation followed by increased upper-layer neuron production. Our findings demonstrate that Sox9 exerts concerted effects on transcription, BP proliferation, neuron production, and neurogenic vs. gliogenic BP cell fate, suggesting that Sox9 may have contributed to promote neocortical expansion.
Collapse
Affiliation(s)
- Ayse Güven
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Human Technopole, Milan, Italy
| | - Katherine R Long
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marta Florio
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Holger Brandl
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Denise Stenzel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
19
|
Jensen BK, Roth LM, Grinspan JB, Jordan-Sciutto KL. White matter loss and oligodendrocyte dysfunction in HIV: A consequence of the infection, the antiretroviral therapy or both? Brain Res 2019; 1724:146397. [PMID: 31442414 DOI: 10.1016/j.brainres.2019.146397] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 01/13/2023]
Abstract
While the severe cognitive effects of HIV-associated dementia have been reduced by combined antiretroviral therapy (cART), nearly half of HIV-positive (HIV+) patients still suffer from some form of HIV-Associated Neurocognitive Disorders (HAND). While frank neuronal loss has been dramatically reduced in HAND patients, white matter loss, including dramatic thinning of the corpus callosum, and loss of volume and structural integrity of myelin persists despite viral control by cART. It remains unclear whether changes in white matter underlie the clinical manifestation seen in patients or whether they are the result of persistent viral reservoirs, remnant damage from the acute infection, the antiretroviral compounds used to treat HIV, secondary effects due to peripheral toxicities or other associated comorbid conditions. Both HIV infection itself and its treatment with antiretroviral drugs can induce metabolic syndrome, lipodystrophy, atherosclerosis and peripheral neuropathies by increased oxidative stress, induction of the unfolded protein response and dysregulation of lipid metabolism. These virally and/or cART-induced processes can also cause myelin loss in the CNS. This review aims to highlight existing data on the contribution of white matter damage to HAND and explore the mechanisms by which HIV infection and its treatment contribute to persistence of white matter changes in people living with HIV currently on cART.
Collapse
Affiliation(s)
- Brigid K Jensen
- Vickie and Jack Farber Institute for Neuroscience, Jefferson Weinberg ALS Center, Thomas Jefferson University, United States; Department of Neurology, The Children's Hospital of Philadelphia, United States; Department of Pathology, School of Dental Medicine, University of Pennsylvania, United States
| | - Lindsay M Roth
- Department of Neurology, The Children's Hospital of Philadelphia, United States; Department of Pathology, School of Dental Medicine, University of Pennsylvania, United States
| | - Judith B Grinspan
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, United States
| | | |
Collapse
|
20
|
Biswas S, Chung SH, Jiang P, Dehghan S, Deng W. Development of glial restricted human neural stem cells for oligodendrocyte differentiation in vitro and in vivo. Sci Rep 2019; 9:9013. [PMID: 31227736 PMCID: PMC6588721 DOI: 10.1038/s41598-019-45247-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 06/04/2019] [Indexed: 11/11/2022] Open
Abstract
In this study, we have developed highly expandable neural stem cells (NSCs) from HESCs and iPSCs that artificially express the oligodendrocyte (OL) specific transcription factor gene Zfp488. This is enough to restrict them to an exclusive oligodendrocyte progenitor cell (OPC) fate during differentiation in vitro and in vivo. During CNS development, Zfp488 is induced during the early stages of OL generation, and then again during terminal differentiation of OLs. Interestingly, the human ortholog Znf488, crucial for OL development in human, has been recently identified to function as a dorsoventral pattering regulator in the ventral spinal cord for the generation of P1, P2/pMN, and P2 neural progenitor domains. Forced expression of Zfp488 gene in human NSCs led to the robust generation of OLs and suppression of neuronal and astrocyte fate in vitro and in vivo. Zfp488 expressing NSC derived oligodendrocytes are functional and can myelinate rat dorsal root ganglion neurons in vitro, and form myelin in Shiverer mice brain in vivo. After transplantation near a site of demyelination, Zfp488 expressing hNSCs migrated to the lesion and differentiated into premyelinating OLs. A certain fraction also homed in the subventricular zone (SVZ). Zfp488-ZsGreen1-hNSC derived OLs formed compact myelin in Shiverer mice brain seen under the electron microscope. Transplanted human neural stem cells (NSC) that have the potential to differentiate into functional oligodendrocytes in response to remyelinating signals can be a powerful therapeutic intervention for disorders where oligodendrocyte (OL) replacement is beneficial.
Collapse
Affiliation(s)
- Sangita Biswas
- Department of Biochemistry and Molecular Medicine, School of Medicine, The University of California at Davis, Sacramento, California, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, 95817, USA.
- Department of Pharmaceutical Sciences, Sun Yat-Sen University, Shenzhen, China.
| | - Seung Hyuk Chung
- Department of Biochemistry and Molecular Medicine, School of Medicine, The University of California at Davis, Sacramento, California, 95817, USA
- Department of Oral Biology, College of Dentistry, The University of Illinois at Chicago, Chicago, Illinois, 60612, USA
| | - Peng Jiang
- Department of Biochemistry and Molecular Medicine, School of Medicine, The University of California at Davis, Sacramento, California, 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, 95817, USA
| | - Samaneh Dehghan
- Department of Biochemistry and Molecular Medicine, School of Medicine, The University of California at Davis, Sacramento, California, 95817, USA
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, School of Medicine, The University of California at Davis, Sacramento, California, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, 95817, USA.
| |
Collapse
|
21
|
Rodrigues RS, Lourenço DM, Paulo SL, Mateus JM, Ferreira MF, Mouro FM, Moreira JB, Ribeiro FF, Sebastião AM, Xapelli S. Cannabinoid Actions on Neural Stem Cells: Implications for Pathophysiology. Molecules 2019; 24:E1350. [PMID: 30959794 PMCID: PMC6480122 DOI: 10.3390/molecules24071350] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 02/06/2023] Open
Abstract
With the increase of life expectancy, neurodegenerative disorders are becoming not only a health but also a social burden worldwide. However, due to the multitude of pathophysiological disease states, current treatments fail to meet the desired outcomes. Therefore, there is a need for new therapeutic strategies focusing on more integrated, personalized and effective approaches. The prospect of using neural stem cells (NSC) as regenerative therapies is very promising, however several issues still need to be addressed. In particular, the potential actions of pharmacological agents used to modulate NSC activity are highly relevant. With the ongoing discussion of cannabinoid usage for medical purposes and reports drawing attention to the effects of cannabinoids on NSC regulation, there is an enormous, and yet, uncovered potential for cannabinoids as treatment options for several neurological disorders, specifically when combined with stem cell therapy. In this manuscript, we review in detail how cannabinoids act as potent regulators of NSC biology and their potential to modulate several neurogenic features in the context of pathophysiology.
Collapse
Affiliation(s)
- Rui S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Diogo M Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Sara L Paulo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Joana M Mateus
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Miguel F Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Francisco M Mouro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - João B Moreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Filipa F Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| |
Collapse
|
22
|
The Role of SVZ Stem Cells in Glioblastoma. Cancers (Basel) 2019; 11:cancers11040448. [PMID: 30934929 PMCID: PMC6521108 DOI: 10.3390/cancers11040448] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/27/2022] Open
Abstract
As most common primary brain cancer, glioblastoma is also the most aggressive and malignant form of cancer in the adult central nervous system. Glioblastomas are genetic and transcriptional heterogeneous tumors, which in spite of intensive research are poorly understood. Over the years conventional therapies failed to affect a cure, resulting in low survival rates of affected patients. To improve the clinical outcome, an important approach is to identify the cells of origin. One potential source for these are neural stem cells (NSCs) located in the subventricular zone, which is one of two niches in the adult nervous system where NSCs with the capacity of self-renewal and proliferation reside. These cells normally give rise to neuronal as well as glial progenitor cells. This review summarizes current findings about links between NSCs and cancer stem cells in glioblastoma and discusses current therapeutic approaches, which arise as a result of identifying the cell of origin in glioblastoma.
Collapse
|
23
|
Decoding epigenetic cell signaling in neuronal differentiation. Semin Cell Dev Biol 2019; 95:12-24. [PMID: 30578863 DOI: 10.1016/j.semcdb.2018.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022]
Abstract
Neurogenesis is the process by which new neurons are generated in the brain. Neural stem cells (NSCs) are differentiated into neurons, which are integrated into the neural network. Nowadays, pluripotent stem cells, multipotent stem cells, and induced pluripotent stem cells can be artificially differentiated into neurons utilizing several techniques. Specific transcriptional profiles from NSCs during differentiation are frequently used to approach and observe phenotype alteration and functional determination of neurons. In this context, the role of non-coding RNA, transcription factors and epigenetic changes in neuronal development and differentiation has gained importance. Epigenetic elucidation has become a field of intense research due to distinct patterns of normal conditions and different neurodegenerative disorders, which can be explored to develop new diagnostic methods or gene therapies. In this review, we discuss the complexity of transcription factors, non-coding RNAs, and extracellular vesicles that are responsible for guiding and coordinating neural development.
Collapse
|
24
|
Morris DC, Zhang ZG, Chopp M. Thymosin β4 for the treatment of acute stroke: neurorestorative or neuroprotective? Expert Opin Biol Ther 2019; 18:149-158. [PMID: 30063858 DOI: 10.1080/14712598.2018.1484100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Thymosin β4 (Tβ4) is a 5K peptide which influences cellular migration by inhibiting organization of the actin-cytoskeleton. Treatment of acute stroke presently involves use of rt-PA and/or endovascular treatment with thrombectomy, both of which have time limitations. Therefore, development of a treatment beyond these times is necessary as most stroke patients present beyond these time limits. A drug which could be administered within 24 h from symptom onset would provide substantial benefit. AREAS COVERED This review summarizes the data and results of two in-vivo studies testing Tβ4 in an embolic stroke model of young and aged rats. In addition, we describe in-vitro investigations of the neurorestorative and neuroprotective properties of Tβ4 in a variety of neuroprogenitor and oligoprogenitor cell models. EXPERT OPINION Tβ4 acts as a neurorestorative agent when employed in a young male rat model of embolic stroke while in an aged model it acts a neuroprotectant. However evaluation of Tβ4 as a treatment of stroke requires further preclinical evaluation in females and in males and females with comorbidities such as, hypertension and diabetes in models of embolic stroke to further define the mechanism of action and potential as a treatment of stroke in humans.
Collapse
Affiliation(s)
- Daniel C Morris
- a Department of Emergency Medicine , Henry Ford Health Systems , Detroit , MI , USA
| | - Zheng G Zhang
- b Department of Neurology , Henry Ford Health Systems , Detroit , MI , USA
| | - Michael Chopp
- b Department of Neurology , Henry Ford Health Systems , Detroit , MI , USA.,c Department of Physics , Oakland University , Rochester , MI , USA
| |
Collapse
|
25
|
Abstract
Current multiple sclerosis (MS) therapies are effective in reducing relapse rate, short-term measures of disability, and magnetic resonance imaging (MRI) measures of inflammation in relapsing remitting MS (RRMS), whereas in progressive/degenerative disease phases these medications are of little or no benefit. Therefore, the development of new therapies aimed at reversing neurodegeneration is of great interest. Remyelination, which is usually a spontaneous endogenous process, is achieved when myelin-producing oligodendrocytes are generated from oligodendrocyte precursor cells (OPCs). Even though these precursor cells are abundant in MS brains, their regeneration capacity is limited. Enhancing the generation of myelin-producing cells is therefore a major focus of MS research. Here we present an overview of the different advancements in the field of remyelination, including suitable animal models for testing remyelination therapies, approved medications with a proposed role in regeneration, myelin repair treatments under investigation in clinical trials, as well as future therapeutics aimed at facilitating myelin repair.
Collapse
Affiliation(s)
- David Kremer
- Department of Neurology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Rainer Akkermann
- Department of Neurology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio-44195
| |
Collapse
|
26
|
Pérès EA, Etienne O, Grigis A, Boumezbeur F, Boussin FD, Le Bihan D. Longitudinal Study of Irradiation-Induced Brain Microstructural Alterations With S-Index, a Diffusion MRI Biomarker, and MR Spectroscopy. Int J Radiat Oncol Biol Phys 2018; 102:1244-1254. [DOI: 10.1016/j.ijrobp.2018.01.070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 12/19/2017] [Accepted: 01/22/2018] [Indexed: 01/19/2023]
|
27
|
Gothié JD, Demeneix B, Remaud S. Comparative approaches to understanding thyroid hormone regulation of neurogenesis. Mol Cell Endocrinol 2017; 459:104-115. [PMID: 28545819 DOI: 10.1016/j.mce.2017.05.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/11/2017] [Accepted: 05/19/2017] [Indexed: 12/12/2022]
Abstract
Thyroid hormone (TH) signalling, an evolutionary conserved pathway, is crucial for brain function and cognition throughout life, from early development to ageing. In humans, TH deficiency during pregnancy alters offspring brain development, increasing the risk of cognitive disorders. How TH regulates neurogenesis and subsequent behaviour and cognitive functions remains a major research challenge. Cellular and molecular mechanisms underlying TH signalling on proliferation, survival, determination, migration, differentiation and maturation have been studied in mammalian animal models for over a century. However, recent data show that THs also influence embryonic and adult neurogenesis throughout vertebrates (from mammals to teleosts). These latest observations raise the question of how TH availability is controlled during neurogenesis and particularly in specific neural stem cell populations. This review deals with the role of TH in regulating neurogenesis in the developing and the adult brain across different vertebrate species. Such evo-devo approaches can shed new light on (i) the evolution of the nervous system and (ii) the evolutionary control of neurogenesis by TH across animal phyla. We also discuss the role of thyroid disruptors on brain development in an evolutionary context.
Collapse
Affiliation(s)
- Jean-David Gothié
- CNRS, UMR 7221, Muséum National d'Histoire Naturelle, F-75005 Paris France
| | - Barbara Demeneix
- CNRS, UMR 7221, Muséum National d'Histoire Naturelle, F-75005 Paris France.
| | - Sylvie Remaud
- CNRS, UMR 7221, Muséum National d'Histoire Naturelle, F-75005 Paris France.
| |
Collapse
|
28
|
Gómez Pinto LI, Rodríguez D, Adamo AM, Mathieu PA. TGF-β pro-oligodendrogenic effects on adult SVZ progenitor cultures and its interaction with the Notch signaling pathway. Glia 2017; 66:396-412. [PMID: 29076551 DOI: 10.1002/glia.23253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 09/27/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
Abstract
Adult neural progenitor cells (NPCs) are capable of differentiating into neurons, astrocytes, and oligodendrocytes throughout life. Notch and transforming growth factor β1 (TGF-β) signaling pathways play critical roles in controlling these cell fate decisions. TGF-β has been previously shown to exert pro-neurogenic effects on hippocampal and subventricular zone (SVZ) NPCs in vitro and to interact with Notch in different cellular types. Therefore, the aim of our work was to study the effect of TGF-β on adult rat brain SVZ NPC glial commitment and its interaction with Notch signaling. Initial cell characterization revealed a large proportion of Olig2+, Nestin+, and glial fibrillary acidic protein (GFAP+) cells, a low percentage of platelet-derived growth factor receptor α (PDGFRα+) or NG2+ cells, and <1% Tuj1+ cells. Immunocytochemical analyses showed a significant increase in the percentage of PDGFRα+, NG2+, and GFAP+ cells upon four-day TGF-β treatment, which demonstrates the pro-gliogenic effect of this growth factor on adult brain SVZ NPCs. Real-time polymerase chain reaction analyses showed that TGF-β induced the expression of Notch ligand Jagged1 and downstream gene Hes1. Notch signaling inhibition in cultures treated with TGF-β produced a decrease in the proportion of PDGFRα+ cells, while TGF-β receptor II (TβRII) inhibition also rendered a decrease in the proportion of PDGFRα+ cells, concomitantly with a decrease in Jagged1 levels. These findings demonstrate the participation of Notch signaling in TGF-β effects and illustrate the impact of TGF-β on glial cell fate decisions of adult brain SVZ NPCs, as well as on oligodendroglial progenitor cell proliferation and maturation.
Collapse
Affiliation(s)
- Laura I Gómez Pinto
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, CABA, C1113AAD, Argentina
| | - Debora Rodríguez
- Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 and Av Constitución, 6700, Luján, Buenos Aires, Argentina
| | - Ana M Adamo
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, CABA, C1113AAD, Argentina
| | - Patricia A Mathieu
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, CABA, C1113AAD, Argentina
| |
Collapse
|
29
|
Jadasz JJ, Tepe L, Beyer F, Samper Agrelo I, Akkermann R, Spitzhorn LS, Silva ME, Oreffo ROC, Hartung HP, Prigione A, Rivera FJ, Adjaye J, Küry P. Human mesenchymal factors induce rat hippocampal- and human neural stem cell dependent oligodendrogenesis. Glia 2017; 66:145-160. [PMID: 28940767 DOI: 10.1002/glia.23233] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 08/04/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022]
Abstract
The generation of new oligodendrocytes is essential for adult brain repair in diseases such as multiple sclerosis. We previously identified the multifunctional p57kip2 protein as a negative regulator of myelinating glial cell differentiation and as an intrinsic switch of glial fate decision in adult neural stem cells (aNSCs). In oligodendroglial precursor cells (OPCs), p57kip2 protein nuclear exclusion was recently found to be rate limiting for differentiation to proceed. Furthermore, stimulation with mesenchymal stem cell (MSC)-derived factors enhanced oligodendrogenesis by yet unknown mechanisms. To elucidate this instructive interaction, we investigated to what degree MSC secreted factors are species dependent, whether hippocampal aNSCs respond equally well to such stimuli, whether apart from oligodendroglial differentiation also tissue integration and axonal wrapping can be promoted and whether the oligodendrogenic effect involved subcellular translocation of p57kip2. We found that CC1 positive oligodendrocytes within the hilus express nuclear p57kip2 protein and that MSC dependent stimulation of cultured hippocampal aNSCs was not accompanied by nuclear p57kip2 exclusion as observed for parenchymal OPCs after spontaneous differentiation. Stimulation with human MSC factors was observed to equally promote rat stem cell oligodendrogenesis, axonal wrapping and tissue integration. As forced nuclear shuttling of p57kip2 led to decreased CNPase- but elevated GFAP expression levels, this indicates heterogenic oligodendroglial mechanisms occurring between OPCs and aNSCs. We also show for the first time that dominant pro-oligodendroglial factors derived from human fetal MSCs can instruct human induced pluripotent stem cell-derived NSCs to differentiate into O4 positive oligodendrocytes.
Collapse
Affiliation(s)
- Janusz J Jadasz
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Lena Tepe
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Felix Beyer
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Iria Samper Agrelo
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Rainer Akkermann
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | - Lucas-Sebastian Spitzhorn
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, D-40225, Germany
| | - Maria Elena Silva
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Institute of Pharmacy, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Hans-Peter Hartung
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| | | | - Francisco J Rivera
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Institute of Molecular Regenerative Medicine & Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, D-40225, Germany
| | - Patrick Küry
- Department of Neurology, Neuroregeneration, Medical Faculty, Heinrich-Heine-University, Düsseldorf, D-40225, Germany
| |
Collapse
|
30
|
Inhibition of neurogenesis in a case of Marburg variant multiple sclerosis. Mult Scler Relat Disord 2017; 18:71-76. [PMID: 29141824 DOI: 10.1016/j.msard.2017.09.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/30/2017] [Accepted: 09/20/2017] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Neural stem cells (NSC) are located essentially in the subventricular zone (SVZ), subgranular zone (SGZ), and along the central canal of the spinal cord. These cells can proliferate in vitro and differentiate into neurons, oligodendrocytes, and astroglia, thus contributing to repair in multiple sclerosis (MS). We conducted a pathological study to analyse neurogenic response in a patient with Marburg variant MS. METHODS We present the case of a 27-year-old immunocompetent patient with Marburg variant MS, a fulminant form of the disease. The condition lasted 20 days. Diagnosis was based on clinical symptoms and MRI showed demyelinating lesions located in subependymal areas and histopathological findings. Neurogenic niches (SVZ and dentate gyrus) were analysed by confocal microscopy using markers of proliferation (Ki-67, PCNA), neuroblasts (PSA-NCAM, DCX, Tuj1), stem cells (Nestin, GFAPδ, SOX2, PAX6, Musashi), astrocytes (GFAP, AQ4), oligodendrocytes (NG2, Olig), microglia and cell infiltrates (IBA-1, CD68, MHCII), and cell death (TUNEL). RESULTS Expression of the markers GFAPδ, SOX2, and PAX6 in NSC was found to be very low. Likewise, markers of proliferation (Ki-67) and intermediate precursors (NG2) were also reduced. This lack of markers of the first stages of cell differentiation means that neurogenesis is inhibited even in very early stages of the disease. CONCLUSION Inhibition of neurogenesis in our patient, which cannot be explained by the fulminant nature of his symptoms, may be related to inflammation and immune response. This finding may further our knowledge of repair mechanisms in MS.
Collapse
|
31
|
Cole KLH, Early JJ, Lyons DA. Drug discovery for remyelination and treatment of MS. Glia 2017; 65:1565-1589. [PMID: 28618073 DOI: 10.1002/glia.23166] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 12/19/2022]
Abstract
Glia constitute the majority of the cells in our nervous system, yet there are currently no drugs that target glia for the treatment of disease. Given ongoing discoveries of the many roles of glia in numerous diseases of the nervous system, this is likely to change in years to come. Here we focus on the possibility that targeting the oligodendrocyte lineage to promote regeneration of myelin (remyelination) represents a therapeutic strategy for the treatment of the demyelinating disease multiple sclerosis, MS. We discuss how hypothesis driven studies have identified multiple targets and pathways that can be manipulated to promote remyelination in vivo, and how this work has led to the first ever remyelination clinical trials. We also highlight how recent chemical discovery screens have identified a host of small molecule compounds that promote oligodendrocyte differentiation in vitro. Some of these compounds have also been shown to promote myelin regeneration in vivo, with one already being trialled in humans. Promoting oligodendrocyte differentiation and remyelination represents just one potential strategy for the treatment of MS. The pathology of MS is complex, and its complete amelioration may require targeting multiple biological processes in parallel. Therefore, we present an overview of new technologies and models for phenotypic analyses and screening that can be exploited to study complex cell-cell interactions in in vitro and in vivo systems. Such technological platforms will provide insight into fundamental mechanisms and increase capacities for drug-discovery of relevance to glia and currently intractable disorders of the CNS.
Collapse
Affiliation(s)
- Katy L H Cole
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - Jason J Early
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - David A Lyons
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| |
Collapse
|
32
|
Xiao J, Yang R, Biswas S, Zhu Y, Qin X, Zhang M, Zhai L, Luo Y, He X, Mao C, Deng W. Neural Stem Cell-Based Regenerative Approaches for the Treatment of Multiple Sclerosis. Mol Neurobiol 2017; 55:3152-3171. [PMID: 28466274 DOI: 10.1007/s12035-017-0566-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/19/2017] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune, inflammatory, and demyelinating disorder of the central nervous system (CNS), which ultimately leads to axonal loss and permanent neurological disability. Current treatments for MS are largely comprised of medications that are either immunomodulatory or immunosuppressive and are aimed at reducing the frequency and intensity of relapses. Neural stem cells (NSCs) in the adult brain can differentiate into oligodendrocytes in a context-specific manner and are shown to be involved in the remyelination in these patients. NSCs may exert their beneficial effects not only through oligodendrocyte replacement but also by providing trophic support and immunomodulation, a phenomenon now known as "therapeutic plasticity." In this review, we first provided an update on the current knowledge regarding MS pathogenesis and the role of immune cells, microglia, and oligodendrocytes in MS disease progression. Next, we reviewed the current progress on research aimed toward stimulating endogenous NSC proliferation and differentiation to oligodendrocytes in vivo and in animal models of demyelination. In addition, we explored the neuroprotective and immunomodulatory effects of transplanted exogenous NSCs on T cell activation, microglial activation, and endogenous remyelination and their effects on the pathological process and prognosis in animal models of MS. Finally, we examined various protocols to generate genetically engineered NSCs as a potential therapy for MS. Overall, this review highlights the studies involving the immunomodulatory, neurotrophic, and regenerative effects of NSCs and novel methods aiming at stimulating the potential of NSCs for the treatment of MS.
Collapse
Affiliation(s)
- Juan Xiao
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China.,Department of Biological Treatment, Handan Central Hospital, Handan, Hebei, China
| | - Rongbing Yang
- Department of Biological Treatment, Handan Central Hospital, Handan, Hebei, China
| | - Sangita Biswas
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, Guangdong, China. .,Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
| | - Yunhua Zhu
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Xin Qin
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Min Zhang
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Lihong Zhai
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Yi Luo
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Xiaoming He
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Chun Mao
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, Guangdong, China. .,Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
| |
Collapse
|
33
|
Bobinger T, May L, Lücking H, Kloska SP, Burkardt P, Spitzer P, Maler JM, Corbeil D, Huttner HB. CD133-Positive Membrane Particles in Cerebrospinal Fluid of Patients with Inflammatory and Degenerative Neurological Diseases. Front Cell Neurosci 2017; 11:77. [PMID: 28396625 PMCID: PMC5366322 DOI: 10.3389/fncel.2017.00077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/03/2017] [Indexed: 01/10/2023] Open
Abstract
Background: Analysis of cerebrospinal fluid (CSF) is a frequently used diagnostic tool in a variety of neurological diseases. Recent studies suggested that investigating membrane particles enriched with the stem cell marker CD133 may offer new avenues for studying neurological disease. In this study, we evaluated the amount of membrane particle-associated CD133 in human CSF in neuroinflammatory and degenerative diseases. Methods: We compared the amount of membrane particle-associated CD133 in CSF samples collected from 45 patients with normal pressure hydrocephalus, parkinsonism, dementia, and cognitive impairment, chronic inflammatory diseases and 10 healthy adult individuals as controls. After ultracentrifugation of CSF, gel electrophoresis and immunoblotting using anti-CD133 monoclonal antibody 80B258 were performed. Antigen-antibody complexes were detected using chemiluminescence. Results: The amount of membrane particle-associated CD133 was significantly increased in patients with normal pressure hydrocephalus (p < 0.001), parkinsonism (p = 0.011) as well as in patients with chronic inflammatory disease (p = 0.008). Analysis of CSF of patients with dementia and cognitive impairment revealed no significant change compared with healthy individuals. Furthermore, subgroup analysis of patients with chronic inflammatory diseases demonstrated significantly elevated levels in individuals with relapsing-remitting multiple sclerosis (p = 0.023) and secondary progressive multiple sclerosis (SPMS; p = 0.010). Conclusion: Collectively, our study revealed elevated levels of membrane particle-associated CD133 in patients with normal pressure hydrocephalus, parkinsonism as well as relapsing-remitting and SPMS. Membrane glycoprotein CD133 may be of clinical value for several neurological diseases.
Collapse
Affiliation(s)
- Tobias Bobinger
- Department of Neurology, University Hospital Erlangen Erlangen, Germany
| | - Lisa May
- Department of Neurology, University Hospital Erlangen Erlangen, Germany
| | - Hannes Lücking
- Department of Neuroradiology, University Hospital Erlangen Erlangen, Germany
| | - Stephan P Kloska
- Department of Neuroradiology, University Hospital Erlangen Erlangen, Germany
| | - Petra Burkardt
- Department of Neurology, University Hospital Erlangen Erlangen, Germany
| | - Philipp Spitzer
- Department of Psychiatry, University Hospital Erlangen Erlangen, Germany
| | - Juan M Maler
- Department of Psychiatry, University Hospital Erlangen Erlangen, Germany
| | - Denis Corbeil
- Biotechnology Center, Technische Universität Dresden Dresden, Germany
| | - Hagen B Huttner
- Department of Neurology, University Hospital Erlangen Erlangen, Germany
| |
Collapse
|
34
|
Kazanis I, Evans KA, Andreopoulou E, Dimitriou C, Koutsakis C, Karadottir RT, Franklin RJM. Subependymal Zone-Derived Oligodendroblasts Respond to Focal Demyelination but Fail to Generate Myelin in Young and Aged Mice. Stem Cell Reports 2017; 8:685-700. [PMID: 28196689 PMCID: PMC5355571 DOI: 10.1016/j.stemcr.2017.01.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 01/10/2017] [Accepted: 01/10/2017] [Indexed: 01/05/2023] Open
Abstract
Two populations of oligodendrogenic progenitors co-exist within the corpus callosum (CC) of the adult mouse. Local, parenchymal oligodendrocyte progenitor cells (pOPCs) and progenitors generated in the subependymal zone (SEZ) cytogenic niche. pOPCs are committed perinatally and retain their numbers through self-renewing divisions, while SEZ-derived cells are relatively “young,” being constantly born from neural stem cells. We compared the behavior of these populations, labeling SEZ-derived cells using hGFAP:CreErt2 mice, within the homeostatic and regenerating CC of the young-adult and aging brain. We found that SEZ-derived oligodendroglial progenitors have limited self-renewing potential and are therefore not bona fide OPCs but rather “oligodendroblasts” more similar to the neuroblasts of the neurogenic output of the SEZ. In the aged CC their mitotic activity is much reduced, although they still act as a “fast-response element” to focal demyelination. In contrast to pOPCs, they fail to generate mature myelinating oligodendrocytes at all ages studied. SEZ-derived cells in the CC are oligodendroblasts and not OPCs Oligodendroblasts have limited self-renewal capacity and do not make myelin Oligodendroblasts respond rapidly after demyelination Aging does not affect the oligodendroblast-pOPC balance
Collapse
Affiliation(s)
- Ilias Kazanis
- Wellcome Trust-MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge CB2 0AH, UK; Lab of Developmental Biology, Department of Biology, University of Patras, Patras 26500, Greece.
| | - Kimberley A Evans
- Wellcome Trust-MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Evangelia Andreopoulou
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras 26500, Greece
| | - Christina Dimitriou
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras 26500, Greece
| | - Christos Koutsakis
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras 26500, Greece
| | | | - Robin J M Franklin
- Wellcome Trust-MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge CB2 0AH, UK.
| |
Collapse
|
35
|
Panaccione A, Guo Y, Yarbrough WG, Ivanov SV. Expression Profiling of Clinical Specimens Supports the Existence of Neural Progenitor-Like Stem Cells in Basal Breast Cancers. Clin Breast Cancer 2017; 17:298-306.e7. [PMID: 28216417 DOI: 10.1016/j.clbc.2017.01.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/09/2017] [Accepted: 01/20/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND We previously characterized in salivary adenoid cystic carcinoma (ACC) a novel population of cancer stem cells (CSCs) marked by coexpression of 2 stemness genes, sex-determining region Y (SRY)-related HMG box-containing factor 10 (SOX10) and CD133. We also reported that in ACC and basal-like breast carcinoma (BBC), a triple-negative breast cancer subtype, expression of SOX10 similarly demarcates a highly conserved gene signature enriched with neural stem cell genes. On the basis of these findings, we hypothesized that BBC might be likewise driven by SOX10-positive (SOX10+)/CD133+ cells with neural stem cell properties. MATERIALS AND METHODS To validate our hypothesis on clinical data, we used a novel approach to meta-analysis that merges gene expression data from independent breast cancer studies and ranks genes according to statistical significance of their coexpression with the gene of interest. Genes that showed strong association with CD133/PROM1 as well as SOX10 were validated across different platforms and data sets and analyzed for enrichment with genes involved in neurogenesis. RESULTS We identified in clinical breast cancer data sets a highly conserved SOX10/PROM1 gene signature that contains neural stem cell markers common for Schwann cells, ACC, BBC, and melanoma. Identification of tripartite motif-containing 2 (TRIM2), TRIM29, MPZL2, potassium calcium-activated channel subfamily N member 4 (KCNN4), and V-set domain containing T cell activation inhibitor 1 (VTCN1)/B7 homolog 4 (B7H4) within this signature provides insight into molecular mechanisms of CSC maintenance. CONCLUSION Our results suggest that BBC is driven by SOX10+/CD133+ cells that express neural stem cell-specific markers and share molecular similarities with CSCs of neural crest origin. Our study provides clinically relevant information on possible drivers of these cells that might facilitate development of CSC-targeting therapies against this cancer distinguished with poor prognosis and resistance to conventional therapies.
Collapse
Affiliation(s)
- Alex Panaccione
- Department of Surgery, Section of Otolaryngology, Yale School of Medicine, New Haven, CT
| | - Yan Guo
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Wendell G Yarbrough
- Department of Surgery, Section of Otolaryngology, Yale School of Medicine, New Haven, CT; Head and Neck Disease Center, Smilow Cancer Hospital, New Haven, CT; Molecular Virology Program, Yale Cancer Center, New Haven, CT
| | - Sergey V Ivanov
- Department of Surgery, Section of Otolaryngology, Yale School of Medicine, New Haven, CT.
| |
Collapse
|
36
|
Dulamea AO. Role of Oligodendrocyte Dysfunction in Demyelination, Remyelination and Neurodegeneration in Multiple Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 958:91-127. [PMID: 28093710 DOI: 10.1007/978-3-319-47861-6_7] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oligodendrocytes (OLs) are the myelinating cells of the central nervous system (CNS) during development and throughout adulthood. They result from a complex and well controlled process of activation, proliferation, migration and differentiation of oligodendrocyte progenitor cells (OPCs) from the germinative niches of the CNS. In multiple sclerosis (MS), the complex pathological process produces dysfunction and apoptosis of OLs leading to demyelination and neurodegeneration. This review attempts to describe the patterns of demyelination in MS, the steps involved in oligodendrogenesis and myelination in healthy CNS, the different pathways leading to OLs and myelin loss in MS, as well as principles involved in restoration of myelin sheaths. Environmental factors and their impact on OLs and pathological mechanisms of MS are also discussed. Finally, we will present evidence about the potential therapeutic targets in re-myelination processes that can be accessed in order to develop regenerative therapies for MS.
Collapse
Affiliation(s)
- Adriana Octaviana Dulamea
- Neurology Clinic, University of Medicine and Pharmacy "Carol Davila", Fundeni Clinical Institute, Building A, Neurology Clinic, Room 201, 022328, Bucharest, Romania.
| |
Collapse
|
37
|
Conover JC, Todd KL. Development and aging of a brain neural stem cell niche. Exp Gerontol 2016; 94:9-13. [PMID: 27867091 DOI: 10.1016/j.exger.2016.11.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/02/2016] [Accepted: 11/10/2016] [Indexed: 01/19/2023]
Abstract
In the anterior forebrain, along the lateral wall of the lateral ventricles, a neurogenic stem cell niche is found in a region referred to as the ventricular-subventricular zone (V-SVZ). In rodents, robust V-SVZ neurogenesis provides new neurons to the olfactory bulb throughout adulthood; however, with increasing age stem cell numbers are reduced and neurogenic capacity is significantly diminished, but new olfactory bulb neurons continue to be produced even in old age. Humans, in contrast, show little to no new neurogenesis after two years of age and whether V-SVZ neural stem cells persist in the adult human brain remains unclear. Here, we review functional and organizational differences in the V-SVZ stem cell niche of mice and humans, and examine how aging affects the V-SVZ niche and its associated functions.
Collapse
Affiliation(s)
- Joanne C Conover
- Department of Physiology and Neurobiology, Institute for Brain and Cognitive Sciences, Center for Aging, University of Connecticut, Storrs, CT 06269, United States.
| | - Krysti L Todd
- Department of Physiology and Neurobiology, Institute for Brain and Cognitive Sciences, Center for Aging, University of Connecticut, Storrs, CT 06269, United States
| |
Collapse
|
38
|
Takase H, Washida K, Hayakawa K, Arai K, Wang X, Lo EH, Lok J. Oligodendrogenesis after traumatic brain injury. Behav Brain Res 2016; 340:205-211. [PMID: 27829126 DOI: 10.1016/j.bbr.2016.10.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/20/2016] [Accepted: 10/21/2016] [Indexed: 01/14/2023]
Abstract
White matter injury is an important contributor to long term motor and cognitive dysfunction after traumatic brain injury. During brain trauma, acceleration, deceleration, torsion, and compression forces often cause direct damage to the axon tracts, and pathways that are triggered by the initial injury can trigger molecular events that result in secondary axon degeneration. White matter injury is often associated with altered mental status, memory deficits, motor or autonomic dysfunction, and contribute to the development of chronic neurodegenerative diseases. The presence and proper functioning of oligodendrocyte precursor cells offer the potential for repair and recovery of injured white matter. The process of the proliferation, maturation of oligodendrocyte precursor cells and their migration to the site of injury to replace injured or lost oligodendrocytes is know as oligodendrogenesis. The process of oligodendrogenesis, as well as the interaction of oligodendrocyte precursor cells with other elements of the neurovascular unit, will be discussed in this review.
Collapse
Affiliation(s)
- Hajime Takase
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazuo Washida
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Division of Neurology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Ken Arai
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Xiaoying Wang
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Eng H Lo
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Josephine Lok
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Pediatrics, Massachusetts General Hospital, Boston, MA, United States.
| |
Collapse
|
39
|
Kalakh S, Mouihate A. Demyelination-Induced Inflammation Attracts Newly Born Neurons to the White Matter. Mol Neurobiol 2016; 54:5905-5918. [PMID: 27660277 DOI: 10.1007/s12035-016-0127-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 09/12/2016] [Indexed: 12/24/2022]
Abstract
There is compelling evidence that microglial activation negatively impacts neurogenesis. However, microglia have also been shown to promote recruitment of newly born neurons to injured areas of the gray matter. In the present study, we explored whether demyelination-triggered inflammation alters the process of neurogenesis in the white matter. A 2-μl solution of 0.04 % ethidium bromide was stereotaxically injected into the corpus callosum of adult male rats. Brain inflammation was dampened by daily injections of progesterone (5 mg/kg, s.c.) for 14 days. Control rats received oil (s.c.). Newly born neurons (DCX and Tbr2), microglia (Iba-1), astrocytes (vimentin or GFAP), oligodendrocyte progenitor cells (OPCs; NG2), and mature oligodendrocytes (CC-1) were monitored in the vicinity of demyelination site using immunofluorescent staining. Western blot was used to explore microglial polarization using M1 (iNOS) and M2 (arginase-1) markers. Focal demyelination elicited strong microglial and astroglial activation and reduced the number of OPCs at the site of demyelination. This inflammatory response was associated with enhanced number of newly born neurons in the white matter and the subventricular zone (SVZ). A proportion of newly born neurons within the white matter showed features of OPCs. Interestingly, blunting brain inflammation led to reduced neurogenesis around the demyelination area and in the SVZ. These data suggest that the white matter inflammation creates a conducive environment for the recruitment of newly born neurons. The fact that a sizable fraction of these newly born neurons adopt OPC features suggests that they could contribute to the remyelination process.
Collapse
Affiliation(s)
- Samah Kalakh
- Department of Physiology, Faculty of Medicine, Health Sciences Centre, Kuwait University, P.O. Box 24923, 13110, Safat, Kuwait
| | - Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Health Sciences Centre, Kuwait University, P.O. Box 24923, 13110, Safat, Kuwait.
| |
Collapse
|
40
|
Rusznák Z, Henskens W, Schofield E, Kim WS, Fu Y. Adult Neurogenesis and Gliogenesis: Possible Mechanisms for Neurorestoration. Exp Neurobiol 2016; 25:103-12. [PMID: 27358578 PMCID: PMC4923354 DOI: 10.5607/en.2016.25.3.103] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/08/2016] [Accepted: 06/08/2016] [Indexed: 12/22/2022] Open
Abstract
The subgranular zone (SGZ) and subventricular zone (SVZ) are developmental remnants of the germinal regions of the brain, hence they retain the ability to generate neuronal progenitor cells in adult life. Neurogenesis in adult brain has an adaptive function because newly produced neurons can integrate into and modify existing neuronal circuits. In contrast to the SGZ and SVZ, other brain regions have a lower capacity to produce new neurons, and this usually occurs via parenchymal and periventricular cell genesis. Compared to neurogenesis, gliogenesis occurs more prevalently in the adult mammalian brain. Under certain circumstances, interaction occurs between neurogenesis and gliogenesis, facilitating glial cells to transform into neuronal lineage. Therefore, modulating the balance between neurogenesis and gliogenesis may present a new perspective for neurorestoration, especially in diseases associated with altered neurogenesis and/or gliogenesis, cell loss, or disturbed homeostasis of cellular constitution. The present review discusses important neuroanatomical features of adult neurogenesis and gliogenesis, aiming to explore how these processes could be modulated toward functional repair of the adult brain.
Collapse
Affiliation(s)
- Zoltán Rusznák
- Neuroscience Research Australia, Sydney, NSW 2031, Australia
| | - Willem Henskens
- Neuroscience Research Australia, Sydney, NSW 2031, Australia.; Prince of Wales Clinical School, UNSW Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Emma Schofield
- Neuroscience Research Australia, Sydney, NSW 2031, Australia
| | - Woojin S Kim
- Neuroscience Research Australia, Sydney, NSW 2031, Australia.; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - YuHong Fu
- Neuroscience Research Australia, Sydney, NSW 2031, Australia.; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
41
|
Intraventricular injections of mesenchymal stem cells activate endogenous functional remyelination in a chronic demyelinating murine model. Cell Death Dis 2016; 7:e2223. [PMID: 27171265 PMCID: PMC4917663 DOI: 10.1038/cddis.2016.130] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 02/07/2023]
Abstract
Current treatments for demyelinating diseases are generally only capable of ameliorating the symptoms, with little to no effect in decreasing myelin loss nor promoting functional recovery. Mesenchymal stem cells (MSCs) have been shown by many researchers to be a potential therapeutic tool in treating various neurodegenerative diseases, including demyelinating disorders. However, in the majority of the cases, the effect was only observed locally, in the area surrounding the graft. Thus, in order to achieve general remyelination in various brain structures simultaneously, bone marrow-derived MSCs were transplanted into the lateral ventricles (LVs) of the cuprizone murine model. In this manner, the cells may secrete soluble factors into the cerebrospinal fluid (CSF) and boost the endogenous oligodendrogenic potential of the subventricular zone (SVZ). As a result, oligodendrocyte progenitor cells (OPCs) were recruited within the corpus callosum (CC) over time, correlating with an increased myelin content. Electrophysiological studies, together with electron microscopy (EM) analysis, indicated that the newly formed myelin correctly enveloped the demyelinated axons and increased signal transduction through the CC. Moreover, increased neural stem progenitor cell (NSPC) proliferation was observed in the SVZ, possibly due to the tropic factors released by the MSCs. In conclusion, the findings of this study revealed that intraventricular injections of MSCs is a feasible method to elicit a paracrine effect in the oligodendrogenic niche of the SVZ, which is prone to respond to the factors secreted into the CSF and therefore promoting oligodendrogenesis and functional remyelination.
Collapse
|
42
|
An F, Qu Y, Liu X, Zhong R, Luo Y. Organ-on-a-Chip: New Platform for Biological Analysis. ANALYTICAL CHEMISTRY INSIGHTS 2015; 10:39-45. [PMID: 26640364 PMCID: PMC4664205 DOI: 10.4137/aci.s28905] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/09/2015] [Accepted: 07/17/2015] [Indexed: 12/15/2022]
Abstract
Direct detection and analysis of biomolecules and cells in physiological microenvironment is urgently needed for fast evaluation of biology and pharmacy. The past several years have witnessed remarkable development opportunities in vitro organs and tissues models with multiple functions based on microfluidic devices, termed as “organ-on-a-chip”. Briefly speaking, it is a promising technology in rebuilding physiological functions of tissues and organs, featuring mammalian cell co-culture and artificial microenvironment created by microchannel networks. In this review, we summarized the advances in studies of heart-, vessel-, liver-, neuron-, kidney- and Multi-organs-on-a-chip, and discussed some noteworthy potential on-chip detection schemes.
Collapse
Affiliation(s)
- Fan An
- School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China. ; State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Yueyang Qu
- School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China. ; State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Xianming Liu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Runtao Zhong
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yong Luo
- School of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China. ; State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian, China
| |
Collapse
|
43
|
Galvez-Contreras AY, Gonzalez-Castaneda RE, Campos-Ordonez T, Luquin S, Gonzalez-Perez O. Phenytoin enhances the phosphorylation of epidermal growth factor receptor and fibroblast growth factor receptor in the subventricular zone and promotes the proliferation of neural precursor cells and oligodendrocyte differentiation. Eur J Neurosci 2015; 43:139-47. [PMID: 26370587 DOI: 10.1111/ejn.13079] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 09/07/2015] [Indexed: 11/30/2022]
Abstract
Phenytoin is a widely used antiepileptic drug that induces cell proliferation in several tissues, such as heart, bone, skin, oral mucosa and neural precursors. Some of these effects are mediated via fibroblast growth factor receptor (FGFR) and epidermal growth factor receptor (EGFR). These receptors are strongly expressed in the adult ventricular-subventricular zone (V-SVZ), the main neurogenic niche in the adult brain. The aim of this study was to determine the cell lineage and cell fate of V-SVZ neural progenitors expanded by phenytoin, as well as the effects of this drug on EGFR/FGFR phosphorylation. Male BALB/C mice received 10 mg/kg phenytoin by oral cannula for 30 days. We analysed the proliferation of V-SVZ neural progenitors by immunohistochemistry and western blot. Our findings indicate that phenytoin enhanced twofold the phosphorylation of EGFR and FGFR in the V-SVZ, increased the number of bromodeoxyuridine (BrdU)+/Sox2+ and BrdU+/doublecortin+ cells in the V-SVZ, and expanded the population of Olig2-expressing cells around the lateral ventricles. After phenytoin removal, a large number of BrdU+/Receptor interacting protein (RIP)+ cells were observed in the olfactory bulb. In conclusion, phenytoin enhanced the phosphorylation of FGFR and EGFR, and promoted the expression of neural precursor markers in the V-SVZ. In parallel, the number of oligodendrocytes increased significantly after phenytoin removal.
Collapse
Affiliation(s)
- Alma Y Galvez-Contreras
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, Mexico
| | - Rocio E Gonzalez-Castaneda
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, Mexico
| | - Tania Campos-Ordonez
- Laboratory of Neuroscience, Facultad de Psicologia, Universidad de Colima, Av. Universidad 333, Colima, COL, 28040, Mexico
| | - Sonia Luquin
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, Mexico
| | - Oscar Gonzalez-Perez
- Laboratory of Neuroscience, Facultad de Psicologia, Universidad de Colima, Av. Universidad 333, Colima, COL, 28040, Mexico
| |
Collapse
|
44
|
Itoh K, Maki T, Lok J, Arai K. Mechanisms of cell-cell interaction in oligodendrogenesis and remyelination after stroke. Brain Res 2015; 1623:135-49. [PMID: 25960351 PMCID: PMC4569526 DOI: 10.1016/j.brainres.2015.04.039] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/19/2015] [Accepted: 04/20/2015] [Indexed: 12/20/2022]
Abstract
White matter damage is a clinically important aspect of several central nervous system diseases, including stroke. Cerebral white matter primarily consists of axonal bundles ensheathed with myelin secreted by mature oligodendrocytes, which play an important role in neurotransmission between different areas of gray matter. During the acute phase of stroke, damage to oligodendrocytes leads to white matter dysfunction through the loss of myelin. On the contrary, during the chronic phase, white matter components promote an environment, which is favorable for neural repair, vascular remodeling, and remyelination. For effective remyelination to take place, oligodendrocyte precursor cells (OPCs) play critical roles by proliferating and differentiating into mature oligodendrocytes, which help to decrease the burden of axonal injury. Notably, other types of cells contribute to these OPC responses under the ischemic conditions. This mini-review summarizes the non-cell autonomous mechanisms in oligodendrogenesis and remyelination after white matter damage, focusing on how OPCs receive support from their neighboring cells. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Kanako Itoh
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
45
|
Qu X, Guo R, Zhang Z, Ma L, Wu X, Luo M, Dong F, Yao R. bFGF Protects Pre-oligodendrocytes from Oxygen/Glucose Deprivation Injury to Ameliorate Demyelination. Cell Mol Neurobiol 2015; 35:913-20. [PMID: 25833395 DOI: 10.1007/s10571-015-0186-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/19/2015] [Indexed: 02/05/2023]
Abstract
One of the pathological hallmarks of periventricular white matter injury is the vulnerability of pre-oligodendrocytes (preOLs) to hypoxia-ischemia (HI). There is increasing evidence that basic fibroblast growth factor (bFGF) is an important signaling molecule for neurogenesis and neuroprotection in the central nervous system. However, it is unknown whether bFGF protects preOLs from oxygen/glucose deprivation (OGD) damage in vitro and promotes remyelination in HI-induced rats. In this present study, bFGF exerted a protective effect on myelin by increasing the myelin thickness, the number of myelinated axons, and myelin basic protein expression in the HI-induced demyelinated neonatal rat corpus callosum. In vitro, bFGF ameliorated the impaired mitochondria and cell processes induced by OGD to promote the survival of isolated O4-positive preOLs. Additionally, the expression of fibroblast growth factor receptor 3 (FGFR3) was dramatically up-regulated in the preOLs after bFGF administration in vivo and in vitro. Thus, bFGF-stimulated remyelination in HI-induced rats by protecting the preOLs from hypoxic injury, and the mechanism involved may be mediated by FGFR3.
Collapse
Affiliation(s)
- Xuebin Qu
- Department of Neurobiology, Xuzhou Medical College, 209# Tongshan Road, Yunlong District, Xuzhou, 221000, Jiangsu, People's Republic of China.
| | - Rui Guo
- Department of Neurobiology, Xuzhou Medical College, 209# Tongshan Road, Yunlong District, Xuzhou, 221000, Jiangsu, People's Republic of China
| | - Zhenzhong Zhang
- Department of Neurobiology, Xuzhou Medical College, 209# Tongshan Road, Yunlong District, Xuzhou, 221000, Jiangsu, People's Republic of China
| | - Li Ma
- Department of Neurobiology, Xuzhou Medical College, 209# Tongshan Road, Yunlong District, Xuzhou, 221000, Jiangsu, People's Republic of China
| | - Xiuxiang Wu
- Department of Neurobiology, Xuzhou Medical College, 209# Tongshan Road, Yunlong District, Xuzhou, 221000, Jiangsu, People's Republic of China
| | - Mengjiao Luo
- Department of Neurobiology, Xuzhou Medical College, 209# Tongshan Road, Yunlong District, Xuzhou, 221000, Jiangsu, People's Republic of China
| | - Fuxing Dong
- Department of Neurobiology, Xuzhou Medical College, 209# Tongshan Road, Yunlong District, Xuzhou, 221000, Jiangsu, People's Republic of China
| | - Ruiqin Yao
- Department of Neurobiology, Xuzhou Medical College, 209# Tongshan Road, Yunlong District, Xuzhou, 221000, Jiangsu, People's Republic of China.
| |
Collapse
|
46
|
Franco PG, Pasquini LA, Pérez MJ, Rosato-Siri MV, Silvestroff L, Pasquini JM. Paving the way for adequate myelination: The contribution of galectin-3, transferrin and iron. FEBS Lett 2015; 589:3388-95. [PMID: 26296311 DOI: 10.1016/j.febslet.2015.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/29/2015] [Accepted: 08/11/2015] [Indexed: 12/24/2022]
Abstract
Considering the worldwide incidence of well characterized demyelinating disorders such as Multiple Sclerosis (MS) and the increasing number of pathologies recently found to involve hypomyelinating factors such as micronutrient deficits, elucidating the molecular basis of central nervous system (CNS) demyelination, remyelination and hypomyelination becomes essential to the development of future neuroregenerative therapies. In this context, this review discusses novel findings on the contribution of galectin-3 (Gal-3), transferrin (Tf) and iron to the processes of myelination and remyelination and their potentially positive regulation of oligodendroglial precursor cell (OPC) differentiation. Studies were conducted in cuprizone (CPZ)-induced demyelination and iron deficiency (ID)-induced hypomyelination, and the participation of glial and neural stem cells (NSC) in the remyelination process was evaluated by means of both in vivo and in vitro assays on primary cell cultures.
Collapse
Affiliation(s)
- Paula G Franco
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, IQUIFIB-CONICET, Universidad de Buenos Aires, Argentina
| | - Laura A Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, IQUIFIB-CONICET, Universidad de Buenos Aires, Argentina
| | - María J Pérez
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, IQUIFIB-CONICET, Universidad de Buenos Aires, Argentina
| | - María V Rosato-Siri
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, IQUIFIB-CONICET, Universidad de Buenos Aires, Argentina
| | - Lucas Silvestroff
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, IQUIFIB-CONICET, Universidad de Buenos Aires, Argentina
| | - Juana M Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, IQUIFIB-CONICET, Universidad de Buenos Aires, Argentina.
| |
Collapse
|
47
|
Maki T, Takahashi Y, Miyamoto N, Liang AC, Ihara M, Lo EH, Arai K. Adrenomedullin promotes differentiation of oligodendrocyte precursor cells into myelin-basic-protein expressing oligodendrocytes under pathological conditions in vitro. Stem Cell Res 2015; 15:68-74. [PMID: 26002630 DOI: 10.1016/j.scr.2015.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 04/25/2015] [Accepted: 05/03/2015] [Indexed: 12/28/2022] Open
Abstract
Oligodendrocytes, which are the main cell type in cerebral white matter, are generated from their precursor cells (oligodendrocyte precursor cells: OPCs). However, the differentiation from OPCs to oligodendrocytes is disturbed under stressed conditions. Therefore, drugs that can improve oligodendrocyte regeneration may be effective for white matter-related diseases. Here we show that a vasoactive peptide adrenomedullin (AM) promotes the in vitro differentiation of OPCs under pathological conditions. Primary OPCs were prepared from neonatal rat brains, and differentiated into myelin-basic-protein expressing oligodendrocytes over time. This in vitro OPC differentiation was inhibited by prolonged chemical hypoxic stress induced by non-lethal CoCl(2) treatment. However, AM promoted the OPC differentiation under the hypoxic stress conditions, and the AM receptor antagonist AM(22-52) canceled the AM-induced OPC differentiation. In addition, AM treatment increased the phosphorylation level of Akt in OPC cultures, and correspondingly, the PI3K/Akt inhibitor LY294002 blocked the AM-induced OPC differentiation. Taken together, AM treatment rescued OPC maturation under pathological conditions via an AM-receptor-PI3K/Akt pathway. Oligodendrocytes play critical roles in white matter by forming myelin sheath. Therefore, AM signaling may be a promising therapeutic target to boost oligodendrocyte regeneration in CNS disorders.
Collapse
Affiliation(s)
- Takakuni Maki
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, MA General Hospital, Harvard Medical School, USA
| | - Yoko Takahashi
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, MA General Hospital, Harvard Medical School, USA; Department of Developmental Neuroscience, United Centers for Advanced Research and Translational Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Nobukazu Miyamoto
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, MA General Hospital, Harvard Medical School, USA
| | - Anna C Liang
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, MA General Hospital, Harvard Medical School, USA
| | - Masafumi Ihara
- Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, MA General Hospital, Harvard Medical School, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, MA General Hospital, Harvard Medical School, USA.
| |
Collapse
|
48
|
Shamloo A, Heibatollahi M, Mofrad MRK. Directional migration and differentiation of neural stem cells within three-dimensional microenvironments. Integr Biol (Camb) 2015; 7:335-44. [PMID: 25633746 DOI: 10.1039/c4ib00144c] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Harnessing neural stem cells to repair neuronal damage is a promising potential treatment for neuronal diseases. To enable future therapeutic efficacy, the survival, proliferation, migration and differentiation of neural stem/progenitor cells (NPCs) should be accurately studied and optimized in in vitro platforms before transplanting these cells into the body for treatment purposes. Such studies can determine the appropriate quantities of the biochemical and biomechanical factors needed to control and optimize NPC behavior in vivo. In this study, NPCs were cultured within a microfluidic device while being encapsulated within the collagen matrix. The migration and differentiation of NPCs were studied in response to varying concentrations of nerve growth factor (NGF) and within varying densities of collagen matrices. It was shown that the migration and differentiation of NPCs can be significantly improved by providing the appropriate range of NGF concentrations while encapsulating the cells within the collagen matrix of optimal density. In particular, it was observed that within collagen matrices of intermediate density (0.9 mg ml(-1)), NPCs have a higher ability to migrate farther and in a collective manner while their differentiation into neurons is significantly higher and the cells can form protrusions and connections with their neighboring cells. Within collagen matrices with higher densities (1.8 mg ml(-1)), the cells did not migrate significantly as compared to the ones within lower matrix densities; within the matrices with lower collagen densities (0.45 mg ml(-1)) most of the cells migrated in an individual manner. However, no significant differentiation into neurons was observed for these two cases. It was also found that depending on the collagen matrix density, a minimum concentration of NGF caused a collective migration of NPCs, and a minimum concentration gradient of this factor stimulated the directional navigation of the cells. The results of this study can be implemented in designing platforms appropriate for regeneration of damaged neuronal systems.
Collapse
Affiliation(s)
- Amir Shamloo
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA 94720, USA.
| | | | | |
Collapse
|
49
|
Treatment with Anti-EGF Ab Ameliorates Experimental Autoimmune Encephalomyelitis via Induction of Neurogenesis and Oligodendrogenesis. Mult Scler Int 2014; 2014:926134. [PMID: 25610650 PMCID: PMC4295609 DOI: 10.1155/2014/926134] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 01/15/2023] Open
Abstract
Background. The neural stem cells (NSCs) migrate to the damaged sites in multiple sclerosis (MS) and in experimental autoimmune encephalomyelitis (EAE). However, the differentiation into neurons or oligodendrocytes is blocked. Epidermal growth factor (EGF) stimulates NSC proliferation and mobilization to demyelinated lesions but also induces astrogenesis and glial scar. Objective. To examine the clinical and histopathological effects of EGF neutralization on EAE. Methods. EAE-induced SJL mice were intravenously treated with either anti-EGF neutralizing antibody (Ab) or isotype control or PBS. On day 9 after immunization, 3 mice of each group were daily treated for 9 days with BrdU and then sacrificed for immunohistochemical analysis. Results. Treatment with anti-EGF Ab significantly ameliorated EAE symptoms during the second relapse. Anti-EGF Ab induced a shift from BrdU+GFAP+ NSCs to BrdU+DCX+ neuroblasts in the subventricular zone (SVZ), increased BrdU+NeuN+ neurons in the granular cell layer of the dentate gyrus, and increased BrdU+O4+ oligodendrocytes in the SVZ. There was no change in the inflammatory infiltrates in response to anti-EGF Ab. Conclusions. Therapy with anti-EGF Ab ameliorates EAE via induction of neurogenesis and oligodendrogenesis. No immunosuppressive effect was found. Further investigation is needed to support these notions of beneficial effect of anti-EGF Ab in MS.
Collapse
|
50
|
Multiple functional therapeutic effects of the estrogen receptor β agonist indazole-Cl in a mouse model of multiple sclerosis. Proc Natl Acad Sci U S A 2014; 111:18061-6. [PMID: 25453074 DOI: 10.1073/pnas.1411294111] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Currently available immunomodulatory therapies do not stop the pathogenesis underlying multiple sclerosis (MS) and are only partially effective in preventing the onset of permanent disability in patients with MS. Identifying a drug that stimulates endogenous remyelination and/or minimizes axonal degeneration would reduce the rate and degree of disease progression. Here, the effects of the highly selective estrogen receptor (ER) β agonist indazole chloride (Ind-Cl) on functional remyelination in chronic experimental autoimmune encephalomyelitis (EAE) mice were investigated by assessing pathologic, functional, and behavioral consequences of both prophylactic and therapeutic (peak EAE) treatment with Ind-Cl. Peripheral cytokines from autoantigen-stimulated splenocytes were measured, and central nervous system infiltration by immune cells, axon health, and myelination were assessed by immunohistochemistry and electron microscopy. Therapeutic Ind-Cl improved clinical disease and rotorod performance and also decreased peripheral Th1 cytokines and reactive astrocytes, activated microglia, and T cells in brains of EAE mice. Increased callosal myelination and mature oligodendrocytes correlated with improved callosal conduction and refractoriness. Therapeutic Ind-Cl-induced remyelination was independent of its effects on the immune system, as Ind-Cl increased remyelination within the cuprizone diet-induced demyelinating model. We conclude that Ind-Cl is a refined pharmacologic agent capable of stimulating functionally relevant endogenous myelination, with important implications for progressive MS treatment.
Collapse
|