1
|
Pivoňková H, Sitnikov S, Kamen Y, Vanhaesebrouck A, Matthey M, Spitzer SO, Ng YT, Tao C, de Faria O, Varga BV, Káradóttir RT. Heterogeneity in oligodendrocyte precursor cell proliferation is dynamic and driven by passive bioelectrical properties. Cell Rep 2024; 43:114873. [PMID: 39423130 PMCID: PMC11602547 DOI: 10.1016/j.celrep.2024.114873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/12/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) generate myelinating oligodendrocytes and are the main proliferative cells in the adult central nervous system. OPCs are a heterogeneous population, with proliferation and differentiation capacity varying with brain region and age. We demonstrate that during early postnatal maturation, cortical, but not callosal, OPCs begin to show altered passive bioelectrical properties, particularly increased inward potassium (K+) conductance, which correlates with G1 cell cycle stage and affects their proliferation potential. Neuronal activity-evoked transient K+ currents in OPCs with high inward K+ conductance potentially release OPCs from cell cycle arrest. Eventually, OPCs in all regions acquire high inward K+ conductance, the magnitude of which may underlie differences in OPC proliferation between regions, with cells being pushed into a dormant state as they acquire high inward K+ conductance and released from dormancy by synchronous neuronal activity. Age-related accumulation of OPCs with high inward K+ conductance might contribute to differentiation failure.
Collapse
Affiliation(s)
- Helena Pivoňková
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Sergey Sitnikov
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Yasmine Kamen
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - An Vanhaesebrouck
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Moritz Matthey
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Sonia Olivia Spitzer
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Yan Ting Ng
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Chenyue Tao
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Omar de Faria
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Balazs Viktor Varga
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Ragnhildur Thóra Káradóttir
- Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge CB2 0AW, UK; Department of Physiology, BioMedical Center, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland.
| |
Collapse
|
2
|
Rao R, Mohammed C, Alschuler L, Pomeranz Krummel DA, Sengupta S. Phytochemical Modulation of Ion Channels in Oncologic Symptomatology and Treatment. Cancers (Basel) 2024; 16:1786. [PMID: 38730738 PMCID: PMC11083444 DOI: 10.3390/cancers16091786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/24/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024] Open
Abstract
Modern chemotherapies offer a broad approach to cancer treatment but eliminate both cancer and non-cancer cells indiscriminately and, thus, are associated with a host of side effects. Advances in precision oncology have brought about new targeted therapeutics, albeit mostly limited to a subset of patients with an actionable mutation. They too come with side effects and, ultimately, 'self-resistance' to the treatment. There is recent interest in the modulation of ion channels, transmembrane proteins that regulate the flow of electrically charged molecules in and out of cells, as an approach to aid treatment of cancer. Phytochemicals have been shown to act on ion channels with high specificity regardless of the tumor's genetic profile. This paper explores the use of phytochemicals in cancer symptom management and treatment.
Collapse
Affiliation(s)
- Rohan Rao
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Caroline Mohammed
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Lise Alschuler
- Andrew Weil Center for Integrative Medicine, University of Arizona College of Medicine, Tucson, AZ 85719, USA
| | - Daniel A. Pomeranz Krummel
- Department of Neurosurgery, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Soma Sengupta
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Department of Neurosurgery, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
3
|
Ion Channel Drugs Suppress Cancer Phenotype in NG108-15 and U87 Cells: Toward Novel Electroceuticals for Glioblastoma. Cancers (Basel) 2022; 14:cancers14061499. [PMID: 35326650 PMCID: PMC8946312 DOI: 10.3390/cancers14061499] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma is a lethal brain cancer that commonly recurs after tumor resection and chemotherapy treatment. Depolarized resting membrane potentials and an acidic intertumoral extracellular pH have been associated with a proliferative state and drug resistance, suggesting that forced hyperpolarization and disruption of proton pumps in the plasma membrane could be a successful strategy for targeting glioblastoma overgrowth. We screened 47 compounds and compound combinations, most of which were ion-modulating, at different concentrations in the NG108-15 rodent neuroblastoma/glioma cell line. A subset of these were tested in the U87 human glioblastoma cell line. A FUCCI cell cycle reporter was stably integrated into both cell lines to monitor proliferation and cell cycle response. Immunocytochemistry, electrophysiology, and a panel of physiological dyes reporting voltage, calcium, and pH were used to characterize responses. The most effective treatments on proliferation in U87 cells were combinations of NS1643 and pantoprazole; retigabine and pantoprazole; and pantoprazole or NS1643 with temozolomide. Marker analysis and physiological dye signatures suggest that exposure to bioelectric drugs significantly reduces proliferation, makes the cells senescent, and promotes differentiation. These results, along with the observed low toxicity in human neurons, show the high efficacy of electroceuticals utilizing combinations of repurposed FDA approved drugs.
Collapse
|
4
|
Dansu DK, Sauma S, Casaccia P. Oligodendrocyte progenitors as environmental biosensors. Semin Cell Dev Biol 2021; 116:38-44. [PMID: 33092959 PMCID: PMC8053729 DOI: 10.1016/j.semcdb.2020.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 01/10/2023]
Abstract
The past decade has seen an important revision of the traditional concept of the role and function of glial cells. From "passive support" for neurons, oligodendrocyte lineage cells are now recognized as metabolic exchangers with neurons, a cellular interface with blood vessels and responders to gut-derived metabolites or changes in the social environment. In the developing brain, the differentiation of neonatal oligodendrocyte progenitors (nOPCs) is required for normal brain function. In adulthood, the differentiation of adult OPCs (aOPCs) serves an important role in learning, behavioral adaptation and response to myelin injury. Here, we propose the concept of OPCs as environmental biosensors, which "sense" chemical and physical stimuli over time and adjust to the new challenges by modifying their epigenome and consequent transcriptome. Because epigenetics defines the ability of the cell to "adapt" gene expression to changes in the environment, we propose a model of OPC differentiation resulting from time-dependent changes of the epigenomic landscape in response to declining mitogens, raising hormone levels, neuronal activity, changes in space constraints or stiffness of the extracellular matrix. We propose that the intrinsically different functional properties of aOPCs compared to nOPCs result from the accrual of "epigenetic memories" of distinct events, which are "recorded" in the nuclei of OPCs as histone and DNA marks, defining a "unique epigenomic landscape" over time.
Collapse
Affiliation(s)
- David K Dansu
- Graduate Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA
| | - Sami Sauma
- Graduate Program in Biology, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA
| | - Patrizia Casaccia
- Graduate Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA; Graduate Program in Biology, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA.
| |
Collapse
|
5
|
Kamen Y, Pivonkova H, Evans KA, Káradóttir RT. A Matter of State: Diversity in Oligodendrocyte Lineage Cells. Neuroscientist 2021; 28:144-162. [PMID: 33567971 DOI: 10.1177/1073858420987208] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) give rise to oligodendrocytes which myelinate axons in the central nervous system. Although classically thought to be a homogeneous population, OPCs are reported to have different developmental origins and display regional and temporal diversity in their transcriptome, response to growth factors, and physiological properties. Similarly, evidence is accumulating that myelinating oligodendrocytes display transcriptional heterogeneity. Analyzing this reported heterogeneity suggests that OPCs, and perhaps also myelinating oligodendrocytes, may exist in different functional cell states. Here, we review the evidence indicating that OPCs and oligodendrocytes are diverse, and we discuss the implications of functional OPC states for myelination in the adult brain and for myelin repair.
Collapse
Affiliation(s)
- Yasmine Kamen
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Helena Pivonkova
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Kimberley A Evans
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Ragnhildur T Káradóttir
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.,Department of Physiology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
6
|
Rosendo-Pineda MJ, Vicente JJ, Vivas O, Pacheco J, Loza-Huerta A, Sampieri A, Wordeman L, Moreno C, Vaca L. Phosphorylation of NMDA receptors by cyclin B/CDK1 modulates calcium dynamics and mitosis. Commun Biol 2020; 3:665. [PMID: 33184446 PMCID: PMC7665045 DOI: 10.1038/s42003-020-01393-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/15/2020] [Indexed: 12/02/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDAR) are glutamate-gated calcium channels named after their artificial agonist. NMDAR are implicated in cell proliferation under normal and pathophysiological conditions. However, the role of NMDAR during mitosis has not yet been explored in individual cells. We found that neurotransmitter-evoked calcium entry via endogenous NMDAR in cortical astrocytes was transient during mitosis. The same occurred in HEK293 cells transfected with the NR1/NR2A subunits of NMDAR. This transient calcium entry during mitosis was due to phosphorylation of the first intracellular loop of NMDAR (S584 of NR1 and S580 of NR2A) by cyclin B/CDK1. Expression of phosphomimetic mutants resulted in transient calcium influx and enhanced NMDAR inactivation independent of the cell cycle phase. Phosphomimetic mutants increased entry of calcium in interphase and generated several alterations during mitosis: increased mitotic index, increased number of cells with lagging chromosomes and fragmentation of pericentriolar material. In summary, by controlling cytosolic calcium, NMDAR modulate mitosis and probably cell differentiation/proliferation. Our results suggest that phosphorylation of NMDAR by cyclin B/CDK1 during mitosis is required to preserve mitotic fidelity. Altering the modulation of the NMDAR by cyclin B/CDK1 may conduct to aneuploidy and cancer.
Collapse
Affiliation(s)
| | - Juan Jesus Vicente
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Oscar Vivas
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Jonathan Pacheco
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, DF, 04510, Mexico
| | - Arlet Loza-Huerta
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, DF, 04510, Mexico
| | - Alicia Sampieri
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, DF, 04510, Mexico
| | - Linda Wordeman
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Claudia Moreno
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Luis Vaca
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, DF, 04510, Mexico.
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
7
|
Rosendo-Pineda MJ, Moreno CM, Vaca L. Role of ion channels during cell division. Cell Calcium 2020; 91:102258. [PMID: 32736154 DOI: 10.1016/j.ceca.2020.102258] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
Ion channels are transmembrane proteins whose canonical function is the transport of ions across the plasma membrane to regulate cell membrane potential and play an essential role in neural communication, nerve conduction, and muscle contraction. However, over the last few years, non-canonical functions have been identified for many channels, having active roles in phagocytosis, invasiveness, proliferation, among others. The participation of some channels in cell proliferation has raised the question of whether they may play an active role in mitosis. There are several reports showing the participation of channels during interphase, however, the direct participation of ion channels in mitosis has received less attention. In this article, we summarize the current evidence on the participation of ion channels in mitosis. We also summarize some tools that would allow the study of ion channels and cell cycle regulatory molecules in individual cells during mitosis.
Collapse
Affiliation(s)
| | - Claudia M Moreno
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Luis Vaca
- Instituto de Fisiología Celular. Universidad Nacional Autónoma de México. Ciudad Universitaria, Coyoacán, DF, 04510, Mexico; Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
8
|
Goyal R, Spencer KA, Borodinsky LN. From Neural Tube Formation Through the Differentiation of Spinal Cord Neurons: Ion Channels in Action During Neural Development. Front Mol Neurosci 2020; 13:62. [PMID: 32390800 PMCID: PMC7193536 DOI: 10.3389/fnmol.2020.00062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022] Open
Abstract
Ion channels are expressed throughout nervous system development. The type and diversity of conductances and gating mechanisms vary at different developmental stages and with the progressive maturational status of neural cells. The variety of ion channels allows for distinct signaling mechanisms in developing neural cells that in turn regulate the needed cellular processes taking place during each developmental period. These include neural cell proliferation and neuronal differentiation, which are crucial for developmental events ranging from the earliest steps of morphogenesis of the neural tube through the establishment of neuronal circuits. Here, we compile studies assessing the ontogeny of ionic currents in the developing nervous system. We then review work demonstrating a role for ion channels in neural tube formation, to underscore the necessity of the signaling downstream ion channels even at the earliest stages of neural development. We discuss the function of ion channels in neural cell proliferation and neuronal differentiation and conclude with how the regulation of all these morphogenetic and cellular processes by electrical activity enables the appropriate development of the nervous system and the establishment of functional circuits adapted to respond to a changing environment.
Collapse
Affiliation(s)
- Raman Goyal
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Kira A Spencer
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Laura N Borodinsky
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
9
|
George K, Thomas NS, Malathi R. 4,4'-Diisothiocyanatostilbene-2,2'-disulfonate modulates voltage-gated K + current and influences cell cycle arrest in androgen sensitive and insensitive human prostate cancer cell lines. Toxicol Mech Methods 2020; 30:358-369. [PMID: 32193973 DOI: 10.1080/15376516.2020.1745343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The stilbene derivative, 4,4'-diisothiocyanatostilbene-2,2'-disulphonic acid (DIDS), an anion channel blocker is used in the present study to evaluate its modulatory effect on voltage-gated K+ current (IK) in human prostate cancer cell lines (LNCaP and PC-3). Voltage-gated K+ (KV) channels in the plasma membrane are critically involved in the proliferation of tumor cells. Therefore, KV channels are considered as a novel potential target for cancer treatment. The results of the present study show that the external perfusion of DIDS activates IK in a concentration-dependent manner, although the known K+ channel blocker TEA failed to block the DIDS activated IK in PC-3 cells. Whereas, in LNCaP cells, the higher concentration of DIDS blocked IK, though this effect was not completely recovered after washout. The difference in function of DIDS might be due to the expression of different Kv channel isoforms in LNCaP and PC-3 cells. Further, the anticancer studies show that treatment of DIDS significantly induced G2/M phase cell cycle arrest and induced moderate and low level of cell death in LNCaP and PC-3 cells respectively. This finding reveals that DIDS modulates IK and exerts cell cycle arrest and cell death in LNCaP and PC-3 cells.
Collapse
Affiliation(s)
- Kiran George
- Department of Biomedical Engineering, Chennai Institute of Technology, Chennai, India.,Bio Engineering Lab, Department of Electronics and Instrumentation Engineering, Annamalai University, Annamalai Nagar, India
| | - Nisha Susan Thomas
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, India
| | - Raman Malathi
- Bio Engineering Lab, Department of Electronics and Instrumentation Engineering, Annamalai University, Annamalai Nagar, India
| |
Collapse
|
10
|
Extrinsic Factors Driving Oligodendrocyte Lineage Cell Progression in CNS Development and Injury. Neurochem Res 2020; 45:630-642. [PMID: 31997102 PMCID: PMC7058689 DOI: 10.1007/s11064-020-02967-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OLs) generate myelin membranes for the rapid propagation of electrical signals along axons in the central nervous system (CNS) and provide metabolites to support axonal integrity and function. Differentiation of OLs from oligodendrocyte progenitor cells (OPCs) is orchestrated by a multitude of intrinsic and extrinsic factors in the CNS. Disruption of this process, or OL loss in the developing or adult brain, as observed in various neurological conditions including hypoxia/ischemia, stroke, and demyelination, results in axonal dystrophy, neuronal dysfunction, and severe neurological impairments. While much is known regarding the intrinsic regulatory signals required for OL lineage cell progression in development, studies from pathological conditions highlight the importance of the CNS environment and external signals in regulating OL genesis and maturation. Here, we review the recent findings in OL biology in the context of the CNS physiological and pathological conditions, focusing on extrinsic factors that facilitate OL development and regeneration.
Collapse
|
11
|
Chew LJ, Ming X, McEllin B, Dupree J, Hong E, Catron M, Fauveau M, Nait-Oumesmar B, Gallo V. Sox17 Regulates a Program of Oligodendrocyte Progenitor Cell Expansion and Differentiation during Development and Repair. Cell Rep 2019; 29:3173-3186.e7. [PMID: 31801081 PMCID: PMC7191642 DOI: 10.1016/j.celrep.2019.10.121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 05/07/2019] [Accepted: 10/29/2019] [Indexed: 11/30/2022] Open
Abstract
Sox17, a SoxF family member transiently upregulated during postnatal oligodendrocyte (OL) development, promotes OL cell differentiation, but its function in white matter development and pathology in vivo is unknown. Our analysis of oligodendroglial- and OL-progenitor-cell-targeted ablation in vivo using a floxed Sox17 mouse establishes a dependence of postnatal oligodendrogenesis on Sox17 and reveals Notch signaling as a mediator of Sox17 function. Following Sox17 ablation, reduced numbers of Olig2-expressing cells and mature OLs led to developmental hypomyelination and motor dysfunction. After demyelination, Sox17 deficiency inhibited OL regeneration. OL decline was unexpectedly preceded by transiently increased differentiation and a reduction of OL progenitor cells. Evidence of a dual role for Sox17 in progenitor cell expansion by Notch and differentiation involving TCF7L2 expression were found. A program of progenitor expansion and differentiation promoted by Sox17 through Notch thus contributes to OL production and determines the outcome of white matter repair.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA.
| | - Xiaotian Ming
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA
| | - Brian McEllin
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA
| | - Jeffrey Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, VA, USA; Research Service, Hunter Holmes McGuire VA Medical Center, Richmond, VA 23249, USA
| | - Elim Hong
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA
| | - Mackenzie Catron
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA
| | - Melissa Fauveau
- Institut du Cerveau et de la Moelle Épinière, ICM, INSERM U1127, CNRS UMR7225, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Brahim Nait-Oumesmar
- Institut du Cerveau et de la Moelle Épinière, ICM, INSERM U1127, CNRS UMR7225, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Hospital, Washington, DC 20010, USA.
| |
Collapse
|
12
|
Wang S, Du L, Peng GH. Optogenetic stimulation inhibits the self-renewal of mouse embryonic stem cells. Cell Biosci 2019; 9:73. [PMID: 31497278 PMCID: PMC6719367 DOI: 10.1186/s13578-019-0335-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 08/21/2019] [Indexed: 12/31/2022] Open
Abstract
Modulation of the embryonic stem cell state is beneficial for elucidating the innate mechanisms of development and regenerative medicine. Ion flux plays important roles in modulating the transition between stemness and differentiation in mouse embryonic stem cells (mESCs). Optogenetics is a novel tool for manipulating ion flux. To investigate the impact of optical stimulation on embryonic stem cells, optogenetically engineered V6.5 mESCs were used to measure the depolarization mediated by ChR2 on the proliferation, self-renewal, and differentiation of mESCs. Blue light stimulation significantly inhibited ChR2-GFP-V6.5 ESC proliferation and disrupted the cell cycle progression, reducing the proportion of cells in the S phase. Interestingly, optical stimulation could inhibit ChR2-GFP-V6.5 ESC self-renewal and trigger differentiation by activating the extracellular regulated protein kinase (ERK) signaling pathway. Our data suggest that membrane potential changes play pivotal roles in regulating the proliferation, self-renewal and initiation of differentiation of mESCs.
Collapse
Affiliation(s)
- Shaojun Wang
- 1Department of Ophthalmology, General Hospital of Chinese People's Liberation Army, Beijing, 100853 China.,2Department of Ophthalmology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, 100071 China.,3Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Lu Du
- 1Department of Ophthalmology, General Hospital of Chinese People's Liberation Army, Beijing, 100853 China
| | - Guang-Hua Peng
- 1Department of Ophthalmology, General Hospital of Chinese People's Liberation Army, Beijing, 100853 China.,3Department of Pathophysiology, Basic Medical College, Zhengzhou University, Zhengzhou, 450052 Henan China
| |
Collapse
|
13
|
George K, Thomas NS, Malathi R. Piperine blocks voltage gated K+ current and inhibits proliferation in androgen sensitive and insensitive human prostate cancer cell lines. Arch Biochem Biophys 2019; 667:36-48. [DOI: 10.1016/j.abb.2019.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 01/15/2023]
|
14
|
Tsata V, Kroehne V, Reinhardt S, El-Armouche A, Brand M, Wagner M, Reimer MM. Electrophysiological Properties of Adult Zebrafish Oligodendrocyte Progenitor Cells. Front Cell Neurosci 2019; 13:102. [PMID: 31031593 PMCID: PMC6473327 DOI: 10.3389/fncel.2019.00102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/28/2019] [Indexed: 11/13/2022] Open
Abstract
Low remyelination efficiency after spinal cord injury (SCI) is a major restraint to successful axonal and functional regeneration in mammals. In contrast, adult zebrafish can: (i) regenerate oligodendrocytes and myelin sheaths within 2 weeks post lesion; (ii) re-grow axonal projections across the lesion site and (iii) recover locomotor function within 6 weeks after spinal cord transection. However, little is known about the intrinsic properties of oligodendrocyte progenitor cells (OPCs), the remyelinating cells of the central nervous system (CNS). Here, we demonstrate that purified OPCs from the adult zebrafish spinal cord are electrically active. They functionally express voltage-gated K+ and Na+ channels, glutamate receptors and exhibit depolarizing, tetrodotoxin (TTX)-sensitive spikes, as previously seen in rodent and human OPCs. Furthermore, we show that the percentage of zebrafish OPCs exhibiting depolarizing spikes and Nav-mediated currents is lower as compared to rodent white matter OPCs, where these membrane characteristics have been shown to underlie OPC injury susceptibility. These findings imply that adult zebrafish OPCs resemble electrical properties found in mammals and represent a relevant cell type towards understanding the biology of the primary cells targeted in remyelination therapies for non-regenerative species. The in vitro platform introduced in this study could be used in the future to: (i) elucidate how membrane characteristics of zebrafish OPCs change upon injury and (ii) identify potential signaling components underlying OPC injury recognition.
Collapse
Affiliation(s)
- Vasiliki Tsata
- Center for Regenerative Therapies TU Dresden (CRTD) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universitaet, Dresden, Germany
| | - Volker Kroehne
- Center for Regenerative Therapies TU Dresden (CRTD) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universitaet, Dresden, Germany
| | - Susanne Reinhardt
- Dresden Genome Center, Center for Regenerative Therapies TU Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universitaet Dresden, Dresden, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Technische Universitaet Dresden, Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies TU Dresden (CRTD) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universitaet, Dresden, Germany
| | - Michael Wagner
- Department of Pharmacology and Toxicology, Technische Universitaet Dresden, Dresden, Germany.,Department of Rhythmology, Heart Center Dresden, Technische Universitaet Dresden, Dresden, Germany
| | - Michell M Reimer
- Center for Regenerative Therapies TU Dresden (CRTD) and Center for Molecular and Cellular Bioengineering (CMCB), Technische Universitaet, Dresden, Germany
| |
Collapse
|
15
|
Implication of Voltage-Gated Potassium Channels in Neoplastic Cell Proliferation. Cancers (Basel) 2019; 11:cancers11030287. [PMID: 30823672 PMCID: PMC6468671 DOI: 10.3390/cancers11030287] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 02/21/2019] [Accepted: 02/24/2019] [Indexed: 12/12/2022] Open
Abstract
Voltage-gated potassium channels (Kv) are the largest group of ion channels. Kv are involved in controlling the resting potential and action potential duration in the heart and brain. Additionally, these proteins participate in cell cycle progression as well as in several other important features in mammalian cell physiology, such as activation, differentiation, apoptosis, and cell volume control. Therefore, Kv remarkably participate in the cell function by balancing responses. The implication of Kv in physiological and pathophysiological cell growth is the subject of study, as Kv are proposed as therapeutic targets for tumor regression. Though it is widely accepted that Kv channels control proliferation by allowing cell cycle progression, their role is controversial. Kv expression is altered in many cancers, and their participation, as well as their use as tumor markers, is worthy of effort. There is an ever-growing list of Kv that remodel during tumorigenesis. This review focuses on the actual knowledge of Kv channel expression and their relationship with neoplastic proliferation. In this work, we provide an update of what is currently known about these proteins, thereby paving the way for a more precise understanding of the participation of Kv during cancer development.
Collapse
|
16
|
Abstract
Modern stem cell research has mainly focused on protein expression and transcriptional networks. However, transmembrane voltage gradients generated by ion channels and transporters have demonstrated to be powerful regulators of cellular processes. These physiological cues exert influence on cell behaviors ranging from differentiation and proliferation to migration and polarity. Bioelectric signaling is a fundamental element of living systems and an untapped reservoir for new discoveries. Dissecting these mechanisms will allow for novel methods of controlling cell fate and open up new opportunities in biomedicine. This review focuses on the role of ion channels and the resting membrane potential in the proliferation and differentiation of skeletal muscle progenitor cells. In addition, findings relevant to this topic are presented and potential implications for tissue engineering and regenerative medicine are discussed.
Collapse
Affiliation(s)
- Colin Fennelly
- Department of Neuroscience, Novartis Institutes for BioMedical Research, Inc., Cambridge, Massachusetts
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina
- Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
17
|
Scaglione A, Patzig J, Liang J, Frawley R, Bok J, Mela A, Yattah C, Zhang J, Teo SX, Zhou T, Chen S, Bernstein E, Canoll P, Guccione E, Casaccia P. PRMT5-mediated regulation of developmental myelination. Nat Commun 2018; 9:2840. [PMID: 30026560 PMCID: PMC6053423 DOI: 10.1038/s41467-018-04863-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/01/2018] [Indexed: 12/16/2022] Open
Abstract
Oligodendrocytes (OLs) are the myelin-forming cells of the central nervous system. They are derived from differentiation of oligodendrocyte progenitors through a process requiring cell cycle exit and histone modifications. Here we identify the histone arginine methyl-transferase PRMT5, a molecule catalyzing symmetric methylation of histone H4R3, as critical for developmental myelination. PRMT5 pharmacological inhibition, CRISPR/cas9 targeting, or genetic ablation decrease p53-dependent survival and impair differentiation without affecting proliferation. Conditional ablation of Prmt5 in progenitors results in hypomyelination, reduced survival and differentiation. Decreased histone H4R3 symmetric methylation is followed by increased nuclear acetylation of H4K5, and is rescued by pharmacological inhibition of histone acetyltransferases. Data obtained using purified histones further validate the results obtained in mice and in cultured oligodendrocyte progenitors. Together, these results identify PRMT5 as critical for oligodendrocyte differentiation and developmental myelination by modulating the cross-talk between histone arginine methylation and lysine acetylation. Myelin-forming cells derive from oligodendrocyte progenitors. Here the authors identify histone arginine methyl-transferase PRMT5 as critical for developmental myelination by modulating the cross-talk between histone arginine methylation and lysine acetylation, to favor differentiation.
Collapse
Affiliation(s)
- Antonella Scaglione
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Julia Patzig
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Jialiang Liang
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Rebecca Frawley
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Jabez Bok
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore
| | - Angeliki Mela
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Camila Yattah
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA.,Graduate Program in Biochemistry, The Graduate Center of The City University of New York, 365 5th Avenue, New York, NY, 10016, USA
| | - Jingxian Zhang
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore
| | - Shun Xie Teo
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore
| | - Ting Zhou
- Room A-829, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Shuibing Chen
- Room A-829, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA
| | - Emily Bernstein
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, New York, NY, 10032, USA
| | - Ernesto Guccione
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.,Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos Building #3-06, Singapore, 138673, Singapore.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Patrizia Casaccia
- Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, 85 St. Nicholas Terrace, New York, NY, 10031, USA. .,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA. .,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA. .,Graduate Program in Biochemistry, The Graduate Center of The City University of New York, 365 5th Avenue, New York, NY, 10016, USA.
| |
Collapse
|
18
|
Spinal Cord Stem Cells In Their Microenvironment: The Ependyma as a Stem Cell Niche. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:55-79. [PMID: 29204829 DOI: 10.1007/978-3-319-69194-7_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The ependyma of the spinal cord is currently proposed as a latent neural stem cell niche. This chapter discusses recent knowledge on the developmental origin and nature of the heterogeneous population of cells that compose this stem cell microenviroment, their diverse physiological properties and regulation. The chapter also reviews relevant data on the ependymal cells as a source of plasticity for spinal cord repair.
Collapse
|
19
|
Larson VA, Mironova Y, Vanderpool KG, Waisman A, Rash JE, Agarwal A, Bergles DE. Oligodendrocytes control potassium accumulation in white matter and seizure susceptibility. eLife 2018; 7:34829. [PMID: 29596047 PMCID: PMC5903864 DOI: 10.7554/elife.34829] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/28/2018] [Indexed: 12/19/2022] Open
Abstract
The inwardly rectifying K+ channel Kir4.1 is broadly expressed by CNS glia and deficits in Kir4.1 lead to seizures and myelin vacuolization. However, the role of oligodendrocyte Kir4.1 channels in controlling myelination and K+ clearance in white matter has not been defined. Here, we show that selective deletion of Kir4.1 from oligodendrocyte progenitors (OPCs) or mature oligodendrocytes did not impair their development or disrupt the structure of myelin. However, mice lacking oligodendrocyte Kir4.1 channels exhibited profound functional impairments, including slower clearance of extracellular K+ and delayed recovery of axons from repetitive stimulation in white matter, as well as spontaneous seizures, a lower seizure threshold, and activity-dependent motor deficits. These results indicate that Kir4.1 channels in oligodendrocytes play an important role in extracellular K+ homeostasis in white matter, and that selective loss of this channel from oligodendrocytes is sufficient to impair K+ clearance and promote seizures.
Collapse
Affiliation(s)
- Valerie A Larson
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Yevgeniya Mironova
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Kimberly G Vanderpool
- Department of Biomedical Sciences, Colorado State University, Fort Collins, United States
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - John E Rash
- Department of Biomedical Sciences, Colorado State University, Fort Collins, United States
| | - Amit Agarwal
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
20
|
Marichal N, Reali C, Rehermann MI, Trujillo-Cenóz O, Russo RE. Progenitors in the Ependyma of the Spinal Cord: A Potential Resource for Self-Repair After Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1015:241-264. [DOI: 10.1007/978-3-319-62817-2_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
21
|
Peruzzo R, Mattarei A, Romio M, Paradisi C, Zoratti M, Szabò I, Leanza L. Regulation of Proliferation by a Mitochondrial Potassium Channel in Pancreatic Ductal Adenocarcinoma Cells. Front Oncol 2017; 7:239. [PMID: 29034212 PMCID: PMC5626813 DOI: 10.3389/fonc.2017.00239] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/19/2017] [Indexed: 12/21/2022] Open
Abstract
Previous results link the mitochondrial potassium channel Kv1.3 (mitoKv1.3) to the regulation of apoptosis. By synthesizing new, mitochondria-targeted derivatives (PAPTP and PCARBTP) of PAP-1, a specific membrane-permeant Kv1.3 inhibitor, we have recently provided evidence that both drugs acting on mitoKv1.3 are able to induce apoptosis and reduce tumor growth in vivo without affecting healthy tissues and cells. In the present article, by exploiting these new drugs, we addressed the question whether mitoKv1.3 contributes to the regulation of cell proliferation as well. When used at low concentrations, which do not compromise cell survival, both drugs slightly increased the percentage of cells in S phase while decreased the population at G0/G1 stage of cells from two different pancreatic ductal adenocarcinoma lines. Our data suggest that the observed modulation is related to ROS levels within the cells, opening the way to link mitochondrial ion channel function to downstream, ROS-related signaling events that might be important for cell cycle progression.
Collapse
Affiliation(s)
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Matteo Romio
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Cristina Paradisi
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Mario Zoratti
- Institute of Neuroscience, CNR, Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ildikò Szabò
- Department of Biology, University of Padova, Padova, Italy.,Institute of Neuroscience, CNR, Padova, Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
22
|
Minovi A, Aguado A, Brunert D, Kurtenbach S, Dazert S, Hatt H, Conrad H. Isolation, culture optimization and functional characterization of stem cell neurospheres from mouse neonatal olfactory bulb and epithelium. Eur Arch Otorhinolaryngol 2017; 274:3071-3085. [PMID: 28478501 DOI: 10.1007/s00405-017-4590-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/25/2017] [Indexed: 10/19/2022]
Abstract
The olfactory epithelium contains basal cells with stem cell characteristics, which have the capacity to differentiate throughout life into olfactory receptor neurons (ORNs). Here we investigate the in vitro characteristics of stem cells taken from the olfactory bulb (OB) and the olfactory epithelium (OE) of neonatal TIS21 knock-in mice. The major aim of the study was the generation of olfactory neurospheres (ONS) derived from OB and OE of neonatal mice as a tool to further analyze the elementary processes of ORN development. Our data showed that the presence of epidermal growth factor (EGF) and fibroblast growth factor (FGF) leads to a significant increase in number of ONS derived from OB but not from OE. The differentiation of ONSs led to the formation of different neuronal cell types, in particular to bipolar-shaped cells as well as putative pyramidal-neurons, astrocytes and oligodendrocytes. Immunohistochemical staining confirmed the presence of astrocytes and neurons in both types of ONSs. In order to investigate the functionality of the neurons we performed calcium imaging and patch-clamp experiments. Calcium imaging experiments revealed that the application of high potassium concentration provokes calcium transients. No excitable properties, neither sodium currents nor action potentials, were observed for the bipolar-shaped cells derived from OB and OE neurospheres, which means that these types of cells morphologically defined as putative neuronal cells, were not physiologically active. Interestingly, patch-clamp recordings performed in the pyramidal-shaped cells of OB neurospheres showed sodium and potassium currents as well as action potentials. Our study will help to establish further models in the field of olfactology.
Collapse
Affiliation(s)
- Amir Minovi
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany.
| | - Ainhara Aguado
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany.,Department of Cell Physiology, Ruhr-University Bochum, Universitätstrasse 150, 44801, Bochum, Germany
| | - Daniela Brunert
- Department of Chemosensation, Institute for Biology II, RWTH Aachen University, 52074, Aachen, Germany
| | - Stefan Kurtenbach
- Department of Cell Physiology, Ruhr-University Bochum, Universitätstrasse 150, 44801, Bochum, Germany
| | - Stefan Dazert
- Department of Otorhinolaryngology, Head and Neck Surgery, St. Elisabeth Hospital, Ruhr-University Bochum, Bleichstr. 15, 44787, Bochum, Germany
| | - Hanns Hatt
- Department of Cell Physiology, Ruhr-University Bochum, Universitätstrasse 150, 44801, Bochum, Germany
| | - Heike Conrad
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| |
Collapse
|
23
|
Sustained Depolarization of the Resting Membrane Potential Regulates Muscle Progenitor Cell Growth and Maintains Stem Cell Properties In Vitro. Stem Cell Rev Rep 2016; 12:634-644. [DOI: 10.1007/s12015-016-9687-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
24
|
Sun W, Matthews EA, Nicolas V, Schoch S, Dietrich D. NG2 glial cells integrate synaptic input in global and dendritic calcium signals. eLife 2016; 5. [PMID: 27644104 PMCID: PMC5052029 DOI: 10.7554/elife.16262] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 09/17/2016] [Indexed: 12/30/2022] Open
Abstract
Synaptic signaling to NG2-expressing oligodendrocyte precursor cells (NG2 cells) could be key to rendering myelination of axons dependent on neuronal activity, but it has remained unclear whether NG2 glial cells integrate and respond to synaptic input. Here we show that NG2 cells perform linear integration of glutamatergic synaptic inputs and respond with increasing dendritic calcium elevations. Synaptic activity induces rapid Ca2+ signals mediated by low-voltage activated Ca2+ channels under strict inhibitory control of voltage-gated A-type K+ channels. Ca2+ signals can be global and originate throughout the cell. However, voltage-gated channels are also found in thin dendrites which act as compartmentalized processing units and generate local calcium transients. Taken together, the activity-dependent control of Ca2+ signals by A-type channels and the global versus local signaling domains make intracellular Ca2+ in NG2 cells a prime signaling molecule to transform neurotransmitter release into activity-dependent myelination. DOI:http://dx.doi.org/10.7554/eLife.16262.001
Collapse
Affiliation(s)
- Wenjing Sun
- Department of Neurosurgery, University Clinic Bonn, Bonn, Germany
| | | | - Vicky Nicolas
- Department of Neurosurgery, University Clinic Bonn, Bonn, Germany
| | - Susanne Schoch
- Department of Neuropathology, University Clinic Bonn, Bonn, Germany
| | - Dirk Dietrich
- Department of Neurosurgery, University Clinic Bonn, Bonn, Germany
| |
Collapse
|
25
|
Abstract
Ion channels have emerged as regulators of developmental processes. In model organisms and in people with mutations in ion channels, disruption of ion channel function can affect cell proliferation, cell migration, and craniofacial and limb patterning. Alterations of ion channel function affect morphogenesis in fish, frogs, mammals, and flies, demonstrating that ion channels have conserved roles in developmental processes. One model suggests that ion channels affect proliferation and migration through changes in cell volume. However, ion channels have not explicitly been placed in canonical developmental signaling cascades until recently. This review gives examples of ion channels that influence developmental processes, offers a potential underlying molecular mechanism involving bone morphogenetic protein (BMP) signaling, and finally explores exciting possibilities for manipulating ion channels to influence cell fate for regenerative medicine and to impact disease.
Collapse
Affiliation(s)
- Emily Bates
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045;
| |
Collapse
|
26
|
Pai VP, Martyniuk CJ, Echeverri K, Sundelacruz S, Kaplan DL, Levin M. Genome-wide analysis reveals conserved transcriptional responses downstream of resting potential change in Xenopus embryos, axolotl regeneration, and human mesenchymal cell differentiation. ACTA ACUST UNITED AC 2015; 3:3-25. [PMID: 27499876 PMCID: PMC4857752 DOI: 10.1002/reg2.48] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 08/20/2015] [Accepted: 08/25/2015] [Indexed: 12/14/2022]
Abstract
Endogenous bioelectric signaling via changes in cellular resting potential (Vmem) is a key regulator of patterning during regeneration and embryogenesis in numerous model systems. Depolarization of Vmem has been functionally implicated in dedifferentiation, tumorigenesis, anatomical re‐specification, and appendage regeneration. However, no unbiased analyses have been performed to understand genome‐wide transcriptional responses to Vmem change in vivo. Moreover, it is unknown which genes or gene networks represent conserved targets of bioelectrical signaling across different patterning contexts and species. Here, we use microarray analysis to comparatively analyze transcriptional responses to Vmem depolarization. We compare the response of the transcriptome during embryogenesis (Xenopus development), regeneration (axolotl regeneration), and stem cell differentiation (human mesenchymal stem cells in culture) to identify common networks across model species that are associated with depolarization. Both subnetwork enrichment and PANTHER analyses identified a number of key genetic modules as targets of Vmem change, and also revealed important (well‐conserved) commonalities in bioelectric signal transduction, despite highly diverse experimental contexts and species. Depolarization regulates specific transcriptional networks across all three germ layers (ectoderm, mesoderm, and endoderm) such as cell differentiation and apoptosis, and this information will be used for developing mechanistic models of bioelectric regulation of patterning. Moreover, our analysis reveals that Vmem change regulates transcripts related to important disease pathways such as cancer and neurodegeneration, which may represent novel targets for emerging electroceutical therapies.
Collapse
Affiliation(s)
- Vaibhav P Pai
- Biology Department and Center for Regenerative and Developmental Biology Tufts University Medford Massachusetts 02155 USA
| | - Christopher J Martyniuk
- Center for Environmental and Human Toxicology and Department of Physiological Sciences UF Genetics Institute, University of Florida Gainesville Florida 32611 USA
| | - Karen Echeverri
- Department of Genetics, Cell Biology and Development University of Minnesota Minneapolis Minnesota 55455 USA
| | - Sarah Sundelacruz
- Department of Biomedical Engineering Tufts University Medford Massachusetts 02155 USA
| | - David L Kaplan
- Department of Biomedical Engineering Tufts University Medford Massachusetts 02155 USA
| | - Michael Levin
- Biology Department and Center for Regenerative and Developmental Biology Tufts University Medford Massachusetts 02155 USA
| |
Collapse
|
27
|
Silencing of Kv1.5 Gene Inhibits Proliferation and Induces Apoptosis of Osteosarcoma Cells. Int J Mol Sci 2015; 16:26914-26. [PMID: 26569226 PMCID: PMC4661860 DOI: 10.3390/ijms161126002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/08/2015] [Accepted: 10/30/2015] [Indexed: 11/17/2022] Open
Abstract
Kv1.5 (also known as KCNA5) is a protein encoded by the KCNA5 gene, which belongs to the voltage-gated potassium channel, shaker-related subfamily. Recently, a number of studies have suggested that Kv1.5 is overexpressed in numerous cancers and plays crucial roles in cancer development. However, until now, the expression and functions of Kv1.5 in osteosarcoma are still unclear. To characterize the potential biological functions of Kv1.5 in osteosarcoma, herein, we examined the expression levels of Kv1.5 in osteosarcoma cells and tissues using quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and immunohistochemistry assays. Four short hairpin RNAs (shRNAs) targeting Kv1.5 were designed and homologous recombination technology was used to construct pGeneSil-Kv1.5 vectors. In addition, the vectors were transfected into osteosarcoma MG63 cells and Kv1.5 mRNA level was measured by qRT-PCR and the Kv1.5 protein level was examined by western blot. We also examined the effects of Kv1.5 silencing on proliferation, cell cycle and apoptosis of the osteosarcoma cells using CCK-8, colony formation, flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays. Our results showed that Kv1.5 was aberrantly expressed in osteosarcoma and that the synthesized shRNA targeting Kv1.5 reduced Kv1.5 mRNA and protein expression effectively. Silencing Kv1.5 expression in the osteosarcoma cells significantly inhibited the proliferation of osteosarcoma cells, induced cell cycle arrest at G0/G1 phase, and induced cell apoptosis through up-regulation of p21, p27, Bax, Bcl-XL and caspase-3 and down-regulation of cyclins A, cyclins D1, cyclins E, Bcl-2 and Bik. In summary, our results indicate that Kv1.5 silencing could suppress osteosarcoma progression through multiple signaling pathways and suggest that Kv1.5 may be a novel target for osteosarcoma therapeutics.
Collapse
|
28
|
Dimou L, Gallo V. NG2-glia and their functions in the central nervous system. Glia 2015; 63:1429-51. [PMID: 26010717 DOI: 10.1002/glia.22859] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/04/2015] [Indexed: 12/12/2022]
Abstract
In the central nervous system, NG2-glia represent a neural cell population that is distinct from neurons, astrocytes, and oligodendrocytes. While in the past the main role ascribed to these cells was that of progenitors for oligodendrocytes, in the last years it has become more obvious that they have further functions in the brain. Here, we will discuss some of the most current and highly debated issues regarding NG2-glia: Do these cells represent a heterogeneous population? Can they give rise to different progenies, and does this change under pathological conditions? How do they respond to injury or pathology? What is the role of neurotransmitter signaling between neurons and NG2-glia? We will first give an overview on the developmental origin of NG2-glia, and then discuss whether their distinct properties in different brain regions are the result of environmental influences, or due to intrinsic differences. We will then review and discuss their in vitro differentiation potential and in vivo lineage under physiological and pathological conditions, together with their electrophysiological properties in distinct brain regions and at different developmental stages. Finally, we will focus on their potential to be used as therapeutic targets in demyelinating and neurodegenerative diseases. Therefore, this review article will highlight the importance of NG2-glia not only in the healthy, but also in the diseased brain.
Collapse
Affiliation(s)
- L Dimou
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University, Munich, 80336, Germany.,Institute for Stem Cell Research, Helmholtz Zentrum Munich, Neuherberg, 85764, Germany
| | - V Gallo
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, District of Columbia
| |
Collapse
|
29
|
Zonouzi M, Scafidi J, Li P, McEllin B, Edwards J, Dupree JL, Harvey L, Sun D, Hübner CA, Cull-Candy SG, Farrant M, Gallo V. GABAergic regulation of cerebellar NG2 cell development is altered in perinatal white matter injury. Nat Neurosci 2015; 18:674-82. [PMID: 25821912 PMCID: PMC4459267 DOI: 10.1038/nn.3990] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/06/2015] [Indexed: 01/11/2023]
Abstract
Diffuse white matter injury (DWMI), a leading cause of neurodevelopmental disabilities in preterm infants, is characterized by reduced oligodendrocyte formation. NG2-expressing oligodendrocyte precursor cells (NG2 cells) are exposed to various extrinsic regulatory signals, including the neurotransmitter GABA. We investigated GABAergic signaling to cerebellar white matter NG2 cells in a mouse model of DWMI (chronic neonatal hypoxia). We found that hypoxia caused a loss of GABAA receptor-mediated synaptic input to NG2 cells, extensive proliferation of these cells and delayed oligodendrocyte maturation, leading to dysmyelination. Treatment of control mice with a GABAA receptor antagonist or deletion of the chloride-accumulating transporter NKCC1 mimicked the effects of hypoxia. Conversely, blockade of GABA catabolism or GABA uptake reduced NG2 cell numbers and increased the formation of mature oligodendrocytes both in control and hypoxic mice. Our results indicate that GABAergic signaling regulates NG2 cell differentiation and proliferation in vivo, and suggest that its perturbation is a key factor in DWMI.
Collapse
MESH Headings
- Action Potentials/drug effects
- Animals
- Animals, Newborn
- Asphyxia Neonatorum/pathology
- Carbachol/pharmacology
- Cell Count
- Cells, Cultured
- Cerebellum/growth & development
- Cerebellum/pathology
- Demyelinating Diseases/chemically induced
- Demyelinating Diseases/etiology
- Disease Models, Animal
- Female
- GABA-A Receptor Antagonists/toxicity
- Hypoxia, Brain/pathology
- Hypoxia, Brain/physiopathology
- Interneurons/pathology
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Neural Stem Cells/cytology
- Neurogenesis/drug effects
- Neurogenesis/physiology
- Nipecotic Acids/pharmacology
- Nipecotic Acids/therapeutic use
- Oligodendroglia/cytology
- Purkinje Cells/pathology
- Receptors, GABA-A/physiology
- Solute Carrier Family 12, Member 2/deficiency
- Solute Carrier Family 12, Member 2/physiology
- Tiagabine
- Vigabatrin/pharmacology
- Vigabatrin/therapeutic use
- White Matter/injuries
- gamma-Aminobutyric Acid/physiology
Collapse
Affiliation(s)
- Marzieh Zonouzi
- 1] Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA. [2] Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Joseph Scafidi
- 1] Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA. [2] Department of Neurology, Children's National Medical Center, Washington, DC, USA
| | - Peijun Li
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA
| | - Brian McEllin
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA
| | - Jorge Edwards
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA
| | - Jeffrey L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Medical Center, Richmond, Virginia, USA
| | - Lloyd Harvey
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christian A Hübner
- Friedrich-Schiller-University Jena, Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Stuart G Cull-Candy
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Mark Farrant
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|
30
|
Litan A, Langhans SA. Cancer as a channelopathy: ion channels and pumps in tumor development and progression. Front Cell Neurosci 2015; 9:86. [PMID: 25852478 PMCID: PMC4362317 DOI: 10.3389/fncel.2015.00086] [Citation(s) in RCA: 210] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/23/2015] [Indexed: 12/23/2022] Open
Abstract
Increasing evidence suggests that ion channels and pumps not only regulate membrane potential, ion homeostasis, and electric signaling in excitable cells but also play important roles in cell proliferation, migration, apoptosis and differentiation. Consistent with a role in cell signaling, channel proteins and ion pumps can form macromolecular complexes with growth factors, and cell adhesion and other signaling molecules. And while cancer is still not being cataloged as a channelopathy, as the non-traditional roles of ion pumps and channels are being recognized, it is increasingly being suggested that ion channels and ion pumps contribute to cancer progression. Cancer cell migration requires the regulation of adhesion complexes between migrating cells and surrounding extracellular matrix (ECM) proteins. Cell movement along solid surfaces requires a sequence of cell protrusions and retractions that mainly depend on regulation of the actin cytoskeleton along with contribution of microtubules and molecular motor proteins such as mysoin. This process is triggered and modulated by a combination of environmental signals, which are sensed and integrated by membrane receptors, including integrins and cadherins. Membrane receptors transduce these signals into downstream signaling pathways, often involving the Rho GTPase protein family. These pathways regulate the cytoskeletal rearrangements necessary for proper timing of adhesion, contraction and detachment of cells in order to find their way through extracellular spaces. Migration and adhesion involve continuous modulation of cell motility, shape and volume, in which ion channels and pumps play major roles. Research on cancer cells suggests that certain ion channels may be involved in aberrant tumor growth and channel inhibitors often lead to growth arrest. This review will describe recent research into the role of ion pumps and ion channels in cell migration and adhesion, and how they may contribute to tumor development.
Collapse
Affiliation(s)
- Alisa Litan
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children Wilmington, DE, USA
| | - Sigrid A Langhans
- Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children Wilmington, DE, USA
| |
Collapse
|
31
|
Huang W, Liu M, Zhu L, Liu S, Luo H, Ma L, Wang H, Lu R, Sun X, Chen L, Wang L. Functional expression of chloride channels and their roles in the cell cycle and cell proliferation in highly differentiated nasopharyngeal carcinoma cells. Physiol Rep 2014; 2:2/9/e12137. [PMID: 25214521 PMCID: PMC4270222 DOI: 10.14814/phy2.12137] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We previously demonstrated that the growth of the poorly differentiated nasopharyngeal carcinoma cells (CNE‐2Z) was more dependent on the activities of volume‐activated chloride channels than that of the normal nasopharyngeal epithelial cells (NP69‐SV40T). However, the activities and roles of such volume‐activated chloride channels in highly differentiated nasopharyngeal carcinoma cells (CNE‐1) are not clarified. In this study, it was found that a volume‐activated chloride current and a regulatory volume decrease (RVD) were induced by 47% hypotonic challenges. The current density and the capacity of RVD in the highly differentiated CNE‐1 cells were lower than those in the poorly differentiated CNE‐2Z cells, and higher than those in the normal cells (NP69‐SV40T). The chloride channel blockers, 5‐nitro‐2‐(3‐phenylpropylamino) benzoic acid (NPPB) and tamoxifen inhibited the current and RVD. Depletion of intracellular Cl− abolished the RVD. The chloride channel blockers reversibly inhibited cell proliferation in a concentration‐ and time‐dependent manner, and arrested cells at the G0/G1 phases, but did not change cell viability. The sensitivity of the three cell lines to the chloride channel blockers was different, with the highest in poorly differentiated cells (CNE‐2Z) and the lowest in the normal cells (NP69‐SV40T). ClC‐3 proteins were expressed in the three cells and distributed inside the cells as well as on the cell membrane. In conclusion, the highly differentiated nasopharyngeal carcinoma CNE‐1 cells functionally expressed the volume‐activated chloride channels, which may play important roles in controlling cell proliferation through modulating the cell cycle, and may be associated with cell differentiation. Chloride channels may be a potential target of anticancer therapy. In this paper, we demonstrated that the volume‐activated chloride channels were involved in regulating CNE‐1 cells proliferation and cell cycle progress. Thus, volume‐activated chloride channels may be a potential target of anticancer therapy.
Collapse
Affiliation(s)
- Weiyuan Huang
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Mei Liu
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Linyan Zhu
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Shanwen Liu
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Hai Luo
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Lianshun Ma
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Haibo Wang
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Ruiling Lu
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Xiaoxue Sun
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| | - Lixin Chen
- Department of Pharmacology, Medical College, Jinan University, Guangzhou, China
| | - Liwei Wang
- Department of Physiology, Medical College, Jinan University, Guangzhou, China
| |
Collapse
|
32
|
Effect of Optogenetic Stimulus on the Proliferation and Cell Cycle Progression of Neural Stem Cells. J Membr Biol 2014; 247:493-500. [DOI: 10.1007/s00232-014-9659-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 03/17/2014] [Indexed: 12/29/2022]
|
33
|
Back SA, Miller SP. Brain injury in premature neonates: A primary cerebral dysmaturation disorder? Ann Neurol 2014; 75:469-86. [PMID: 24615937 PMCID: PMC5989572 DOI: 10.1002/ana.24132] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 03/04/2014] [Accepted: 03/05/2014] [Indexed: 12/11/2022]
Abstract
With advances in neonatal care, preterm neonates are surviving with an evolving constellation of motor and cognitive disabilities that appear to be related to widespread cellular maturational disturbances that target cerebral gray and white matter. Whereas preterm infants were previously at high risk for destructive brain lesions that resulted in cystic white matter injury and secondary cortical and subcortical gray matter degeneration, contemporary cohorts of preterm survivors commonly display less severe injury that does not appear to involve pronounced glial or neuronal loss. Nevertheless, these milder forms of injury are also associated with reduced cerebral growth. Recent human and experimental studies support that impaired cerebral growth is related to disparate responses in gray and white matter. Myelination disturbances in cerebral white matter are related to aberrant regeneration and repair responses to acute death of premyelinating late oligodendrocyte progenitors (preOLs). In response to preOL death, early oligodendrocyte progenitors rapidly proliferate and differentiate, but the regenerated preOLs fail to normally mature to myelinating cells required for white matter growth. Although immature neurons appear to be more resistant to cell death from hypoxia-ischemia than glia, they display widespread disturbances in maturation of their dendritic arbors, which further contribute to impaired cerebral growth. These complex and disparate responses of neurons and preOLs thus result in large numbers of cells that fail to fully mature during a critical window in development of neural circuitry. These recently recognized forms of cerebral gray and white matter dysmaturation raise new diagnostic challenges and suggest new therapeutic directions centered on reversal of the processes that promote dysmaturation.
Collapse
Affiliation(s)
- Stephen A Back
- Departments of Pediatrics, Oregon Health and Science University, Portland; Departments of Neurology, Oregon Health and Science University, Portland
| | | |
Collapse
|
34
|
Back SA, Rosenberg PA. Pathophysiology of glia in perinatal white matter injury. Glia 2014; 62:1790-815. [PMID: 24687630 DOI: 10.1002/glia.22658] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/13/2014] [Accepted: 02/27/2014] [Indexed: 12/12/2022]
Abstract
Injury to the preterm brain has a particular predilection for cerebral white matter. White matter injury (WMI) is the most common cause of brain injury in preterm infants and a major cause of chronic neurological morbidity including cerebral palsy. Factors that predispose to WMI include cerebral oxygenation disturbances and maternal-fetal infection. During the acute phase of WMI, pronounced oxidative damage occurs that targets late oligodendrocyte progenitors (pre-OLs). The developmental predilection for WMI to occur during prematurity appears to be related to both the timing of appearance and regional distribution of susceptible pre-OLs that are vulnerable to a variety of chemical mediators including reactive oxygen species, glutamate, cytokines, and adenosine. During the chronic phase of WMI, the white matter displays abberant regeneration and repair responses. Early OL progenitors respond to WMI with a rapid robust proliferative response that results in a several fold regeneration of pre-OLs that fail to terminally differentiate along their normal developmental time course. Pre-OL maturation arrest appears to be related in part to inhibitory factors that derive from reactive astrocytes in chronic lesions. Recent high field magnetic resonance imaging (MRI) data support that three distinct forms of chronic WMI exist, each of which displays unique MRI and histopathological features. These findings suggest the possibility that therapies directed at myelin regeneration and repair could be initiated early after WMI and monitored over time. These new mechanisms of acute and chronic WMI provide access to a variety of new strategies to prevent or promote repair of WMI in premature infants.
Collapse
Affiliation(s)
- Stephen A Back
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon; Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
35
|
Urrego D, Tomczak AP, Zahed F, Stühmer W, Pardo LA. Potassium channels in cell cycle and cell proliferation. Philos Trans R Soc Lond B Biol Sci 2014; 369:20130094. [PMID: 24493742 PMCID: PMC3917348 DOI: 10.1098/rstb.2013.0094] [Citation(s) in RCA: 280] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Normal cell-cycle progression is a crucial task for every multicellular organism, as it determines body size and shape, tissue renewal and senescence, and is also crucial for reproduction. On the other hand, dysregulation of the cell-cycle progression leading to uncontrolled cell proliferation is the hallmark of cancer. Therefore, it is not surprising that it is a tightly regulated process, with multifaceted and very complex control mechanisms. It is now well established that one of those mechanisms relies on ion channels, and in many cases specifically on potassium channels. Here, we summarize the possible mechanisms underlying the importance of potassium channels in cell-cycle control and briefly review some of the identified channels that illustrate the multiple ways in which this group of proteins can influence cell proliferation and modulate cell-cycle progression.
Collapse
Affiliation(s)
- Diana Urrego
- Oncophysiology Group, Max Planck Institute of Experimental Medicine, , Hermann-Rein-Strasse 3, Göttingen 37075, Germany
| | | | | | | | | |
Collapse
|
36
|
Park KS, Han MH, Jang HK, Kim KA, Cha EJ, Kim WJ, Choi YH, Kim Y. The TREK2 Channel Is Involved in the Proliferation of 253J Cell, a Human Bladder Carcinoma Cell. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2013; 17:511-6. [PMID: 24381500 PMCID: PMC3874438 DOI: 10.4196/kjpp.2013.17.6.511] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/11/2013] [Accepted: 11/19/2013] [Indexed: 11/17/2022]
Abstract
Bladder cancer is the seventh most common cancer in men that smoke, and the incidence of disease increases with age. The mechanism of occurrence has not yet been established. Potassium channels have been linked with cell proliferation. Some two-pore domain K+ channels (K2P), such as TASK3 and TREK1, have recently been shown to be overexpressed in cancer cells. Here we focused on the relationship between cell growth and the mechanosensitive K2P channel, TREK2, in the human bladder cancer cell line, 253J. We confirmed that TREK2 was expressed in bladder cancer cell lines by Western blot and quantitative real-time PCR. Using the patch-clamp technique, the mechanosensitive TREK2 channel was recorded in the presence of symmetrical 150 mM KCl solutions. In 253J cells, the TREK2 channel was activated by polyunsaturated fatty acids, intracellular acidosis at -60 mV and mechanical stretch at -40 mV or 40 mV. Furthermore, small interfering RNA (siRNA)-mediated TREK2 knockdown resulted in a slight depolarization from -19.9 mV±0.8 (n=116) to -8.5 mV±1.4 (n=74) and decreased proliferation of 253J cells, compared to negative control siRNA. 253J cells treated with TREK2 siRNA showed a significant increase in the expression of cell cycle boundary proteins p21 and p53 and also a remarkable decrease in protein expression of cyclins D1 and D3. Taken together, the TREK2 channel is present in bladder cancer cell lines and may, at least in part, contribute to cell cycle-dependent growth.
Collapse
Affiliation(s)
- Kyung-Sun Park
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Min Ho Han
- Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan 614-714, Korea. ; Personalized Tumor Engineering Research Center, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | - Hee Kyung Jang
- Department of Physiology, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea. ; Personalized Tumor Engineering Research Center, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | - Kyung-A Kim
- Department of Biomedical Engineering, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea. ; Personalized Tumor Engineering Research Center, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | - Eun-Jong Cha
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea. ; Personalized Tumor Engineering Research Center, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | - Wun-Jae Kim
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea. ; Personalized Tumor Engineering Research Center, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan 614-714, Korea. ; Personalized Tumor Engineering Research Center, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea
| | - Yangmi Kim
- Department of Physiology, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea. ; Personalized Tumor Engineering Research Center, College of Medicine, Chungbuk National University, Cheongju 361-763, Korea
| |
Collapse
|
37
|
García Münzer D, Kostoglou M, Georgiadis M, Pistikopoulos E, Mantalaris A. Developing a cyclin blueprint as a tool for mapping the cell cycle in GS-NS0. Biochem Eng J 2013. [DOI: 10.1016/j.bej.2013.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
38
|
Light-controlled inhibition of malignant glioma by opsin gene transfer. Cell Death Dis 2013; 4:e893. [PMID: 24176851 PMCID: PMC3920933 DOI: 10.1038/cddis.2013.425] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 09/19/2013] [Accepted: 09/20/2013] [Indexed: 11/08/2022]
Abstract
Glioblastomas are aggressive cancers with low survival rates and poor prognosis because of their highly proliferative and invasive capacity. In the current study, we describe a new optogenetic strategy that selectively inhibits glioma cells through light-controlled membrane depolarization and cell death. Transfer of the engineered opsin ChETA (engineered Channelrhodopsin-2 variant) gene into primary human glioma cells or cell lines, but not normal astrocytes, unexpectedly decreased cell proliferation and increased mitochondria-dependent apoptosis, upon light stimulation. These optogenetic effects were mediated by membrane depolarization-induced reductions in cyclin expression and mitochondrial transmembrane potential. Importantly, the ChETA gene transfer and light illumination in mice significantly inhibited subcutaneous and intracranial glioma growth and increased the survival of the animals bearing the glioma. These results uncover an unexpected effect of opsin ion channels on glioma cells and offer the opportunity for the first time to treat glioma using a light-controllable optogenetic approach.
Collapse
|
39
|
Morgan PJ, Hübner R, Rolfs A, Frech MJ. Spontaneous Calcium Transients in Human Neural Progenitor Cells Mediated by Transient Receptor Potential Channels. Stem Cells Dev 2013; 22:2477-86. [DOI: 10.1089/scd.2013.0061] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Peter J. Morgan
- University of Rostock, Albrecht-Kossel-Institute for Neuroregeneration, Rostock, Germany
| | - Rayk Hübner
- University of Rostock, Albrecht-Kossel-Institute for Neuroregeneration, Rostock, Germany
| | - Arndt Rolfs
- University of Rostock, Albrecht-Kossel-Institute for Neuroregeneration, Rostock, Germany
| | - Moritz J. Frech
- University of Rostock, Albrecht-Kossel-Institute for Neuroregeneration, Rostock, Germany
| |
Collapse
|
40
|
Neuron-NG2 cell synapses: novel functions for regulating NG2 cell proliferation and differentiation. BIOMED RESEARCH INTERNATIONAL 2013; 2013:402843. [PMID: 23984358 PMCID: PMC3747365 DOI: 10.1155/2013/402843] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 07/08/2013] [Indexed: 01/11/2023]
Abstract
NG2 cells are a population of CNS cells that are distinct from neurons, mature oligodendrocytes, astrocytes, and microglia. These cells can be identified by their NG2 proteoglycan expression. NG2 cells have a highly branched morphology, with abundant processes radiating from the cell body, and express a complex set of voltage-gated channels, AMPA/kainate, and GABA receptors. Neurons notably form classical and nonclassical synapses with NG2 cells, which have varied characteristics and functions. Neuron-NG2 cell synapses could fine-tune NG2 cell activities, including the NG2 cell cycle, differentiation, migration, and myelination, and may be a novel potential therapeutic target for NG2 cell-related diseases, such as hypoxia-ischemia injury and periventricular leukomalacia. Furthermore, neuron-NG2 cell synapses may be correlated with the plasticity of CNS in adulthood with the synaptic contacts passing onto their progenies during proliferation, and synaptic contacts decrease rapidly upon NG2 cell differentiation. In this review, we highlight the characteristics of classical and nonclassical neuron-NG2 cell synapses, the potential functions, and the fate of synaptic contacts during proliferation and differentiation, with the emphasis on the regulation of the NG2 cell cycle by neuron-NG2 cell synapses and their potential underlying mechanisms.
Collapse
|
41
|
Coppi E, Cellai L, Maraula G, Pugliese AM, Pedata F. Adenosine A₂A receptors inhibit delayed rectifier potassium currents and cell differentiation in primary purified oligodendrocyte cultures. Neuropharmacology 2013; 73:301-10. [PMID: 23770463 DOI: 10.1016/j.neuropharm.2013.05.035] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 05/07/2013] [Accepted: 05/19/2013] [Indexed: 11/24/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) are a population of cycling cells which persist in the adult central nervous system (CNS) where, under opportune stimuli, they differentiate into mature myelinating oligodendrocytes. Adenosine A(2A) receptors are Gs-coupled P1 purinergic receptors which are widely distributed throughout the CNS. It has been demonstrated that OPCs express A(2A) receptors, but their functional role in these cells remains elusive. Oligodendrocytes express distinct voltage-gated ion channels depending on their maturation. Here, by electrophysiological recordings coupled with immunocytochemical labeling, we studied the effects of adenosine A(2A) receptors on membrane currents and differentiation of purified primary OPCs isolated from the rat cortex. We found that the selective A(2A) agonist, CGS21680, inhibits sustained, delayed rectifier, K(+) currents (I(K)) without modifying transient (I(A)) conductances. The effect was observed in all cells tested, independently from time in culture. CGS21680 inhibition of I(K) current was concentration-dependent (10-200 nM) and blocked in the presence of the selective A(2A) antagonist SCH58261 (100 nM). It is known that I(K) currents play an important role during OPC development since their block decreases cell proliferation and differentiation. In light of these data, our further aim was to investigate whether A(2A) receptors modulate these processes. CGS21680, applied at 100 nM in the culture medium of oligodendrocyte cultures, inhibits OPC differentiation (an effect prevented by SCH58261) without affecting cell proliferation. Data demonstrate that cultured OPCs express functional A(2A) receptors whose activation negatively modulate I(K) currents. We propose that, by this mechanism, A(2A) adenosine receptors inhibit OPC differentiation.
Collapse
Affiliation(s)
- Elisabetta Coppi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | | | | | | | | |
Collapse
|
42
|
Coppi E, Maraula G, Fumagalli M, Failli P, Cellai L, Bonfanti E, Mazzoni L, Coppini R, Abbracchio MP, Pedata F, Pugliese AM. UDP-glucose enhances outward K(+) currents necessary for cell differentiation and stimulates cell migration by activating the GPR17 receptor in oligodendrocyte precursors. Glia 2013; 61:1155-71. [PMID: 23640798 DOI: 10.1002/glia.22506] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 03/13/2013] [Indexed: 01/30/2023]
Abstract
In the developing and mature central nervous system, NG2 expressing cells comprise a population of cycling oligodendrocyte progenitor cells (OPCs) that differentiate into mature, myelinating oligodendrocytes (OLGs). OPCs are also characterized by high motility and respond to injury by migrating into the lesioned area to support remyelination. K(+) currents in OPCs are developmentally regulated during differentiation. However, the mechanisms regulating these currents at different stages of oligodendrocyte lineage are poorly understood. Here we show that, in cultured primary OPCs, the purinergic G-protein coupled receptor GPR17, that has recently emerged as a key player in oligodendrogliogenesis, crucially regulates K(+) currents. Specifically, receptor stimulation by its agonist UDP-glucose enhances delayed rectifier K(+) currents without affecting transient K(+) conductances. This effect was observed in a subpopulation of OPCs and immature pre-OLGs whereas it was absent in mature OLGs, in line with GPR17 expression, that peaks at intermediate phases of oligodendrocyte differentiation and is thereafter downregulated to allow terminal maturation. The effect of UDP-glucose on K(+) currents is concentration-dependent, blocked by the GPR17 antagonists MRS2179 and cangrelor, and sensitive to the K(+) channel blocker tetraethyl-ammonium, which also inhibits oligodendrocyte maturation. We propose that stimulation of K(+) currents is responsible for GPR17-induced oligodendrocyte differentiation. Moreover, we demonstrate, for the first time, that GPR17 activation stimulates OPC migration, suggesting an important role for this receptor after brain injury. Our data indicate that modulation of GPR17 may represent a strategy to potentiate the post-traumatic response of OPCs under demyelinating conditions, such as multiple sclerosis, stroke, and brain trauma.
Collapse
Affiliation(s)
- Elisabetta Coppi
- Divi Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Marichal N, García G, Radmilovich M, Trujillo-Cenóz O, Russo RE. Spatial domains of progenitor-like cells and functional complexity of a stem cell niche in the neonatal rat spinal cord. Stem Cells 2013; 30:2020-31. [PMID: 22821702 DOI: 10.1002/stem.1175] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During spinal cord development, progenitors in the neural tube are arranged within spatial domains that generate specific cell types. The ependyma of the postnatal spinal cord seems to retain cells with properties of the primitive neural stem cells, some of which are able to react to injury with active proliferation. However, the functional complexity and organization of this stem cell niche in mammals remains poorly understood. Here, we combined immunohistochemistry for cell-specific markers with patch-clamp recordings to test the hypothesis that the ependyma of the neonatal rat spinal cord contains progenitor-like cells functionally segregated within specific domains. Cells on the lateral aspects of the ependyma combined morphological and molecular traits of ependymocytes and radial glia (RG) expressing S100β and vimentin, displayed passive membrane properties and were electrically coupled via Cx43. Cells contacting the ventral and dorsal poles expressed the neural stem cell markers nestin and/or vimentin, had the typical morphology of RG, and appeared uncoupled displaying various combinations of K(+) and Ca(2+) voltage-gated currents. Although progenitor-like cells were mitotically active around the entire ependyma, the proliferative capacity seemed higher on lateral domains. Our findings represent the first evidence that the ependyma of the rat harbors progenitor-like cells with heterogeneous electrophysiological phenotypes organized in spatial domains. The manipulation of specific functional properties in the heterogeneous population of progenitor-like cells contacting the ependyma may in future help to regulate their behavior and lineage potential, providing the cell types required for the endogenous repair of the injured spinal cord.
Collapse
Affiliation(s)
- Nicolás Marichal
- Neurofisiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable, Avenida Italia 3318, CP11600, Montevideo, Uruguay
| | | | | | | | | |
Collapse
|
44
|
Bioelectric state and cell cycle control of Mammalian neural stem cells. Stem Cells Int 2012; 2012:816049. [PMID: 23024660 PMCID: PMC3447385 DOI: 10.1155/2012/816049] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 07/22/2012] [Indexed: 12/25/2022] Open
Abstract
The concerted action of ion channels and pumps establishing a resting membrane potential has been most thoroughly studied in the context of excitable cells, most notably neurons, but emerging evidences indicate that they are also involved in controlling proliferation and differentiation of nonexcitable somatic stem cells. The importance of understanding stem cell contribution to tissue formation during embryonic development, adult homeostasis, and regeneration in disease has prompted many groups to study and manipulate the membrane potential of stem cells in a variety of systems. In this paper we aimed at summarizing the current knowledge on the role of ion channels and pumps in the context of mammalian corticogenesis with particular emphasis on their contribution to the switch of neural stem cells from proliferation to differentiation and generation of more committed progenitors and neurons, whose lineage during brain development has been recently elucidated.
Collapse
|
45
|
Min J, Singh S, Fitzgerald-Bocarsly P, Wood TL. Insulin-like growth factor I regulates G2/M progression through mammalian target of rapamycin signaling in oligodendrocyte progenitors. Glia 2012; 60:1684-95. [PMID: 22836368 DOI: 10.1002/glia.22387] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 06/17/2012] [Accepted: 06/18/2012] [Indexed: 12/20/2022]
Abstract
Extrinsic factors including growth factors influence decisions of oligodendrocyte progenitor cells (OPCs) to continue cell cycle progression or exit the cell cycle and terminally differentiate into oligodendrocytes capable of producing myelin. Multiple studies have elucidated how the G1/S transition is regulated in OPCs; however, little is known about how S phase progression and the G2/M transition are regulated in these cells. Herein, we report that insulin-like growth factor (IGF)-I coordinates with FGF-2 to promote S phase progression but regulates G2/M progression independently. During S phase, IGF-I/FGF-2 enhances protein expression of cyclin A and cdk2, and further increases effective complex formation resulting in enhanced cdk2 activity. Surprisingly, however, OPCs exposed to FGF-2 in the absence of IGF-I fail to traverse through G2/M. Consistent with this observation, OPCs exposed to IGF-I, but not FGF-2, increase cell number over 48 h. IGF-I enhances cdk1 kinase activity during G2/M by promoting nuclear localization of cyclin B/cdk1 as well as of Cdc25C, an activator of cdk1. IGF-I also induces phosphorylation of histone 3 indicating traverse of cells through mitosis. Finally, we demonstrate that IGF-I-mediated G2/M regulation requires mammalian target of rapamycin activity. These data support an important function for IGF-I in G2/M progression in OPCs.
Collapse
Affiliation(s)
- Jungsoo Min
- Department of Neurology and Neuroscience and NJMS Cancer Center, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07101-1709, USA
| | | | | | | |
Collapse
|
46
|
Kim BS, Im YB, Jung SJ, Park CH, Kang SK. Argonaute2 regulation for K+ channel-mediated human adipose tissue-derived stromal cells self-renewal and survival in nucleus. Stem Cells Dev 2012; 21:1736-48. [PMID: 22014067 DOI: 10.1089/scd.2011.0388] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Argonaute2 (Ago2) is a well-known factor that has intrinsic endonuclease activity and is a part of the fundamental gene regulatory machinery. Recently, we showed that nuclear Ago2 regulates voltage-gated potassium (Kv) channels and that Ago2/Kv1.3 has crucial functions in the self-renewal and cell de-aging processes in adipose tissue-derived stromal cells (ATSCs). In the nucleus, Ago2 bound to the promoter regions of calcium-activated potassium channel 3, potassium channel subfamily K member 1 (KCNK1), and voltage-gated potassium channel 2, and the expression of these genes was significantly upregulated at the level of transcription. We detected an active K+ channel that plays a critical role in Ago2-mediated ATSC self-renewal through the control of membrane potential during cell self-renewal and differentiation. Among the several regulatory subunits of voltage-dependent K+ (Kv) channels, Kv1.3 and Kv1.5 have been shown to impact tissue differentiation and cell growth in cultured ATSCs following their direct binding to the regulatory region of the Kv channel gene. In ATSCs, interference with Ago2 or K+ channel gene expression or treatment with tetraethylammonium significantly downregulated stemness gene expression, induced cell cycle arrest, and inhibited the ability of cells to transdifferentiate into neurons or β-cells via Oct4 knockdown. Blockage of the K+ channel significantly induced protein kinase C (PKC) α, β, and δ phosphorylation and negatively affected Ago2 and Oct4 expression. This K+ channel blockage also resulted in the upregulation of p53 and p21 expression and the inactivation of mitogen-activated protein kinase (MEK), extracellular signal-regulated kinase 1/2 (ERK 1/2), AKT, β-catenin, and STAT3. Our results suggest that the nuclear Ago2 regulation of the K+ channel or stemness-related gene expression plays a critical role in adult stem cell self-renewal and differentiation.
Collapse
Affiliation(s)
- Bong Sun Kim
- Department of Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
47
|
Rooj AK, McNicholas CM, Bartoszewski R, Bebok Z, Benos DJ, Fuller CM. Glioma-specific cation conductance regulates migration and cell cycle progression. J Biol Chem 2011; 287:4053-65. [PMID: 22130665 DOI: 10.1074/jbc.m111.311688] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In this study, we have investigated the role of a glioma-specific cation channel assembled from subunits of the Deg/epithelial sodium channel (ENaC) superfamily, in the regulation of migration and cell cycle progression in glioma cells. Channel inhibition by psalmotoxin-1 (PcTX-1) significantly inhibited migration and proliferation of D54-MG glioma cells. Both PcTX-1 and benzamil, an amiloride analog, caused cell cycle arrest of D54-MG cells in G(0)/G(1) phases (by 30 and 40%, respectively) and reduced cell accumulation in S and G(2)/M phases after 24 h of incubation. Both PcTX-1 and benzamil up-regulated expression of cyclin-dependent kinase inhibitor proteins p21(Cip1) and p27(Kip1). Similar results were obtained in U87MG and primary glioblastoma multiforme cells maintained in primary culture and following knockdown of one of the component subunits, ASIC1. In contrast, knocking down δENaC, which is not a component of the glioma cation channel complex, had no effect on cyclin-dependent kinase inhibitor expression. Phosphorylation of ERK1/2 was also inhibited by PcTX-1, benzamil, and knockdown of ASIC1 but not δENaC in D54MG cells. Our data suggest that a specific cation conductance composed of acid-sensing ion channels and ENaC subunits regulates migration and cell cycle progression in gliomas.
Collapse
Affiliation(s)
- Arun K Rooj
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
48
|
Fröhlich N, Nagy B, Hovhannisyan A, Kukley M. Fate of neuron-glia synapses during proliferation and differentiation of NG2 cells. J Anat 2011; 219:18-32. [PMID: 21592101 PMCID: PMC3130157 DOI: 10.1111/j.1469-7580.2011.01392.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2011] [Indexed: 11/30/2022] Open
Abstract
Progenitor cells expressing proteoglycan NG2 (also known as oligodendrocyte precursor cells or polydendrocytes) are widespread in the grey and white matter of the CNS; they comprise 8-9% of the total cell population in adult white matter, and 2-3% of total cells in adult grey matter. NG2 cells have a complex stellate morphology, with highly branched processes that may extend more than 100 μm around the cell body. NG2 cells express a complex set of voltage-gated channels, AMPA/kainate and/or γ-aminobutyric acid (GABA)(A) receptors, and receive glutamatergic and/or GABAergic synaptic input from neurons. In every region of the brain NG2 cells are found as proliferative cells, and the fraction of actively cycling NG2 cells is quite high in young as well as in adult animals. During development NG2 cells either differentiate into myelinating oligodendrocytes (and possibly also few astrocytes and neurons) or persist in the brain parenchyma as NG2 cells. This review highlights new findings related to the morphological and electrophysiological changes of NG2 cells, and the fate of synaptic input between neurons and NG2 cells during proliferation and differentiation of these cells in the neonatal and adult nervous system of rodents.
Collapse
Affiliation(s)
- Nicole Fröhlich
- Group of Neuron–Glia Interactions, Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingen, Germany
| | - Bálint Nagy
- Group of Neuron–Glia Interactions, Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingen, Germany
| | - Anahit Hovhannisyan
- Group of Neuron–Glia Interactions, Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingen, Germany
- Group of Retinal Circuits and Optogenetics, Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingen, Germany
| | - Maria Kukley
- Group of Neuron–Glia Interactions, Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingen, Germany
| |
Collapse
|
49
|
Dalfampridine in multiple sclerosis: from symptomatic treatment to immunomodulation. Clin Immunol 2011; 142:84-92. [PMID: 21742559 DOI: 10.1016/j.clim.2011.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 06/16/2011] [Accepted: 06/17/2011] [Indexed: 01/08/2023]
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease that is deemed to affect more than 2.1 million people worldwide, and for which there is no cure. Early symptoms of MS are believed to result from axonal demyelination leading to slowing or blockade of impulse conduction. The blockade of K+ channels has been proven to improve conduction deficiencies secondary to demyelination in patients with MS. Dalfampridine is a K+ channel blocker that was recently approved by FDA for the symptomatic treatment of ambulation hardship in MS. Understanding the mechanisms by which Dalfampridine exerts its therapeutic effects is a complex issue as it blocks a wide variety of K+ channels that are distributed across multiple cell types in the nervous system but also in the immune system, and because of their molecular identities remaining unknown. This review describes Dalfampridine potential roles at the cellular and molecular levels in MS pathogenesis.
Collapse
|
50
|
Paintlia AS, Paintlia MK, Singh AK, Orak JK, Singh I. Activation of PPAR-γ and PTEN cascade participates in lovastatin-mediated accelerated differentiation of oligodendrocyte progenitor cells. Glia 2011; 58:1669-85. [PMID: 20578043 DOI: 10.1002/glia.21039] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previously, we and others documented that statins including-lovastatin (LOV) promote the differentiation of oligodendrocyte progenitor cells (OPCs) and remyelination in experimental autoimmune encephalomyelitis (EAE), an multiple sclerosis (MS) model. Conversely, some recent studies demonstrated that statins negatively influence oligodendrocyte (OL) differentiation in vitro and remyelination in a cuprizone-CNS demyelinating model. Therefore, herein, we first investigated the cause of impaired differentiation of OLs by statins in vitro settings. Our observations indicated that the depletion of cholesterol was detrimental to LOV treated OPCs under cholesterol/serum-deprived culture conditions similar to that were used in conflicting studies. However, the depletion of geranylgeranyl-pp under normal cholesterol homeostasis conditions enhanced the phenotypic commitment and differentiation of LOV-treated OPCs ascribed to inhibition of RhoA-Rho kinase. Interestingly, this effect of LOV was associated with increased activation and expression of both PPAR-γ and PTEN in OPCs as confirmed by various pharmacological and molecular based approaches. Furthermore, PTEN was involved in an inhibition of OPCs proliferation via PI3K-Akt inhibition and induction of cell cycle arrest at G1 phase, but without affecting their cell survival. These effects of LOV on OPCs in vitro were absent in the CNS of normal rats chronically treated with LOV concentrations used in EAE indicating that PPAR-γ induction in normal brain may be tightly regulated-providing evidences that statins are therapeutically safe for humans. Collectively, these data provide initial evidence that statin-mediated activation of the PPAR-γ-PTEN cascade participates in OL differentiation, thus suggesting new therapeutic-interventions for MS or related CNS-demyelinating diseases.
Collapse
Affiliation(s)
- Ajaib S Paintlia
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, South Carolina, USA
| | | | | | | | | |
Collapse
|