1
|
Fukumoto-Inukai AK, Bermeo K, Arenas I, Rosendo-Pineda MJ, Pimentel-Cabrera JA, Garcia DE. AMPK inhibits voltage-gated calcium channel-current in rat chromaffin cells. Mol Cell Endocrinol 2024; 591:112275. [PMID: 38777212 DOI: 10.1016/j.mce.2024.112275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/08/2024] [Accepted: 05/18/2024] [Indexed: 05/25/2024]
Abstract
Metabolic changes are critical in the regulation of Ca2+ influx in central and peripheral neuroendocrine cells. To study the regulation of L-type Ca2+ channels by AMPK we used biochemical reagents and ATP/glucose-concentration manipulations in rat chromaffin cells. AICAR and Compound-C, at low concentration, significantly induce changes in L-type Ca2+ channel-current amplitude and voltage dependence. Remarkably, an overlasting decrease in the channel-current density can be induced by lowering the intracellular level of ATP. Accordingly, Ca2+ channel-current density gradually diminishes by decreasing the extracellular glucose concentration. By using immunofluorescence, a decrease in the expression of CaV1.2 is observed while decreasing extracellular glucose, suggesting that AMPK reduces the number of functional Ca2+ channels into the plasma membrane. Together, these results support for the first time the dependence of metabolic changes in the maintenance of Ca2+ channel-current by AMPK. They reveal a key step in Ca2+ influx in secretory cells.
Collapse
Affiliation(s)
- A K Fukumoto-Inukai
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, UNAM, Circuito Exterior S/N, Ciudad Universitaria, Coyoacán, 04510, Ciudad de México, Mexico
| | - K Bermeo
- Licenciatura en Neurociencias, Facultad de Medicina, Universidad Nacional Autónoma de México, UNAM, Circuito Exterior S/N, Ciudad Universitaria, Coyoacán, 04510, Ciudad de México, Mexico
| | - I Arenas
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, UNAM, Circuito Exterior S/N, Ciudad Universitaria, Coyoacán, 04510, Ciudad de México, Mexico
| | - M J Rosendo-Pineda
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, UNAM, Circuito Exterior S/N, Ciudad Universitaria, Coyoacán, 04510, Ciudad de México, Mexico
| | - J A Pimentel-Cabrera
- Laboratorio Nacional de Microscopía Avanzada, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - D E Garcia
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, UNAM, Circuito Exterior S/N, Ciudad Universitaria, Coyoacán, 04510, Ciudad de México, Mexico.
| |
Collapse
|
2
|
Abe SI. Behavior and Functional Roles of CD34 + Mesenchymal Cells in Mammalian Testes. Int J Mol Sci 2022; 23:9585. [PMID: 36076981 PMCID: PMC9455925 DOI: 10.3390/ijms23179585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/19/2022] Open
Abstract
Mammalian testes consist of seminiferous tubules within which Sertoli cells line up at the periphery and nurse germ cells, and of interstitia that harbor various cells such as peritubular myoid cells (PMCs), Leydig cells (LCs), vascular endothelial cells, immune cells such as macrophages, and mesenchymal (stromal) cells. Morphological studies have recently reported the presence of telocytes with telopodes in the interstitium of adult mouse, rat, and human testes. CD34+PDGFRα+ telocytes with long and moniliform telopodes form reticular networks with various cell types such as LCs, PMCs, and vessels, indicating their potential functions in cell-cell communications and tissue homeostasis. Functional studies have recently been performed on testicular interstitial cells and CD34+ cells, using 3D re-aggregate cultures of dissociated testicular cells, and cell cultures. Direct observation of CD34+ cells and adult LCs (ALCs) revealed that CD34+ cells extend thin cytoplasmic processes (telopodes), move toward the LC-CD34+ cell-re-aggregates, and finally enter into the re-aggregates, indicating the chemotactic behavior of CD34+ telocytes toward ALCs. In mammalian testes, important roles of mesenchymal interstitial cells as stem/progenitors in the differentiation and regeneration of LCs have been reported. Here, reports on testicular telocytes so far obtained are reviewed, and future perspectives on the studies of testicular telocytes are noted.
Collapse
Affiliation(s)
- Shin-Ichi Abe
- Faculty of Health Science, Kumamoto Health Science University, Kumamoto 861-5598, Japan
| |
Collapse
|
3
|
Moriarty N, Kauhausen JA, Pavan C, Hunt CPJ, de Luzy IR, Penna V, Ermine CM, Thompson LH, Parish CL. Understanding the Influence of Target Acquisition on Survival, Integration, and Phenotypic Maturation of Dopamine Neurons within Stem Cell-Derived Neural Grafts in a Parkinson's Disease Model. J Neurosci 2022; 42:4995-5006. [PMID: 35610045 PMCID: PMC9233443 DOI: 10.1523/jneurosci.2431-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022] Open
Abstract
Midbrain dopaminergic (DA) neurons include many subtypes characterized by their location, connectivity and function. Surprisingly, mechanisms underpinning the specification of A9 neurons [responsible for motor function, including within ventral midbrain (VM) grafts for treating Parkinson's disease (PD)] over adjacent A10, remains largely speculated. We assessed the impact of synaptic targeting on survival, integration, and phenotype acquisition of dopaminergic neurons within VM grafts generated from fetal tissue or human pluripotent stem cells (PSCs). VM progenitors were grafted into female mice with 6OHDA-lesions of host midbrain dopamine neurons, with some animals also receiving intrastriatal quinolinic acid (QA) injections to ablate medium spiny neurons (MSN), the A9 neuron primary target. While loss of MSNs variably affected graft survival, it significantly reduced striatal yet increased cortical innervation. Consequently, grafts showed reduced A9 and increased A10 specification, with more DA neurons failing to mature into either subtype. These findings highlight the importance of target acquisition on DA subtype specification during development and repair.SIGNIFICANCE STATEMENT Parish and colleagues highlight, in a rodent model of Parkinson's disease (PD), the importance of synaptic target acquisition in the survival, integration and phenotypic specification of grafted dopamine neurons derived from fetal tissue and human stem cells. Ablation of host striatal neurons resulted in reduced dopamine neuron survival within grafts, re-routing of dopamine fibers from striatal to alternate cortical targets and a consequential reduced specification of A9 dopamine neurons (the subpopulation critical for restoration of motor function) and increase in A10 DA neurons.
Collapse
Affiliation(s)
- Niamh Moriarty
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jessica A Kauhausen
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Chiara Pavan
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Cameron P J Hunt
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Isabelle R de Luzy
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Vanessa Penna
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Charlotte M Ermine
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Lachlan H Thompson
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Clare L Parish
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
4
|
Winick-Ng W, Kukalev A, Harabula I, Zea-Redondo L, Szabó D, Meijer M, Serebreni L, Zhang Y, Bianco S, Chiariello AM, Irastorza-Azcarate I, Thieme CJ, Sparks TM, Carvalho S, Fiorillo L, Musella F, Irani E, Torlai Triglia E, Kolodziejczyk AA, Abentung A, Apostolova G, Paul EJ, Franke V, Kempfer R, Akalin A, Teichmann SA, Dechant G, Ungless MA, Nicodemi M, Welch L, Castelo-Branco G, Pombo A. Cell-type specialization is encoded by specific chromatin topologies. Nature 2021; 599:684-691. [PMID: 34789882 PMCID: PMC8612935 DOI: 10.1038/s41586-021-04081-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/30/2021] [Indexed: 11/09/2022]
Abstract
The three-dimensional (3D) structure of chromatin is intrinsically associated with gene regulation and cell function1-3. Methods based on chromatin conformation capture have mapped chromatin structures in neuronal systems such as in vitro differentiated neurons, neurons isolated through fluorescence-activated cell sorting from cortical tissues pooled from different animals and from dissociated whole hippocampi4-6. However, changes in chromatin organization captured by imaging, such as the relocation of Bdnf away from the nuclear periphery after activation7, are invisible with such approaches8. Here we developed immunoGAM, an extension of genome architecture mapping (GAM)2,9, to map 3D chromatin topology genome-wide in specific brain cell types, without tissue disruption, from single animals. GAM is a ligation-free technology that maps genome topology by sequencing the DNA content from thin (about 220 nm) nuclear cryosections. Chromatin interactions are identified from the increased probability of co-segregation of contacting loci across a collection of nuclear slices. ImmunoGAM expands the scope of GAM to enable the selection of specific cell types using low cell numbers (approximately 1,000 cells) within a complex tissue and avoids tissue dissociation2,10. We report cell-type specialized 3D chromatin structures at multiple genomic scales that relate to patterns of gene expression. We discover extensive 'melting' of long genes when they are highly expressed and/or have high chromatin accessibility. The contacts most specific of neuron subtypes contain genes associated with specialized processes, such as addiction and synaptic plasticity, which harbour putative binding sites for neuronal transcription factors within accessible chromatin regions. Moreover, sensory receptor genes are preferentially found in heterochromatic compartments in brain cells, which establish strong contacts across tens of megabases. Our results demonstrate that highly specific chromatin conformations in brain cells are tightly related to gene regulation mechanisms and specialized functions.
Collapse
Affiliation(s)
- Warren Winick-Ng
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany.
| | - Alexander Kukalev
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
| | - Izabela Harabula
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Luna Zea-Redondo
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dominik Szabó
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mandy Meijer
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Leonid Serebreni
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Yingnan Zhang
- School of Electrical Engineering and Computer Science, Ohio University, Athens, OH, USA
| | - Simona Bianco
- Dipartimentio di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant'Angelo, Naples, Italy
| | - Andrea M Chiariello
- Dipartimentio di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant'Angelo, Naples, Italy
| | - Ibai Irastorza-Azcarate
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
| | - Christoph J Thieme
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
| | - Thomas M Sparks
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
| | - Sílvia Carvalho
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
- UCIBIO, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Graduate Program in Areas of Basic and Applied Biology, Universidade do Porto, Porto, Portugal
| | - Luca Fiorillo
- Dipartimentio di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant'Angelo, Naples, Italy
| | - Francesco Musella
- Dipartimentio di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant'Angelo, Naples, Italy
| | - Ehsan Irani
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Elena Torlai Triglia
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aleksandra A Kolodziejczyk
- Cavendish Laboratory, University of Cambridge, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Andreas Abentung
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Galina Apostolova
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Eleanor J Paul
- Institute of Clinical Sciences, Imperial College London, London, UK
- Center for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Center for Neurodevelopmental Disorders, King's College London, London, UK
| | - Vedran Franke
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Bioinformatics and Omics Data Science Platform, Berlin, Germany
| | - Rieke Kempfer
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Altuna Akalin
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Bioinformatics and Omics Data Science Platform, Berlin, Germany
| | - Sarah A Teichmann
- Cavendish Laboratory, University of Cambridge, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Georg Dechant
- Institute for Neuroscience, Medical University of Innsbruck, Innsbruck, Austria
| | - Mark A Ungless
- Institute of Clinical Sciences, Imperial College London, London, UK
| | - Mario Nicodemi
- Dipartimentio di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant'Angelo, Naples, Italy
- Berlin Institute of Health, Berlin, Germany
| | - Lonnie Welch
- School of Electrical Engineering and Computer Science, Ohio University, Athens, OH, USA
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Ana Pombo
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, Berlin, Germany.
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany.
- Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
5
|
Tossell K, Dodhia RA, Galet B, Tkachuk O, Ungless MA. Tonic GABAergic inhibition, via GABA A receptors containing αβƐ subunits, regulates excitability of ventral tegmental area dopamine neurons. Eur J Neurosci 2021; 53:1722-1737. [PMID: 33522050 PMCID: PMC8651010 DOI: 10.1111/ejn.15133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/15/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022]
Abstract
The activity of midbrain dopamine neurons is strongly regulated by fast synaptic inhibitory γ‐Aminobutyric acid (GABA)ergic inputs. There is growing evidence in other brain regions that low concentrations of ambient GABA can persistently activate certain subtypes of GABAA receptor to generate a tonic current. However, evidence for a tonic GABAergic current in midbrain dopamine neurons is limited. To address this, we conducted whole‐cell recordings from ventral tegmental area (VTA) dopamine neurons in brain slices from mice. We found that application of GABAA receptor antagonists decreased the holding current, indicating the presence of a tonic GABAergic input. Global increases in GABA release, induced by either a nitric oxide donor or inhibition of GABA uptake, further increased this tonic current. Importantly, prolonged inhibition of the firing activity of local GABAergic neurons abolished the tonic current. A combination of pharmacology and immunohistochemistry experiments suggested that, unlike common examples of tonic inhibition, this current may be mediated by a relatively unusual combination of α4βƐ subunits. Lastly, we found that the tonic current reduced excitability in dopamine neurons suggesting a subtractive effect on firing activity.
Collapse
Affiliation(s)
- Kyoko Tossell
- MRC London Institute of Medical Sciences (LMS), London, UK.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Rakesh A Dodhia
- MRC London Institute of Medical Sciences (LMS), London, UK.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Benjamin Galet
- MRC London Institute of Medical Sciences (LMS), London, UK.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Olga Tkachuk
- MRC London Institute of Medical Sciences (LMS), London, UK.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Mark A Ungless
- MRC London Institute of Medical Sciences (LMS), London, UK.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
6
|
Harischandra DS, Rokad D, Ghaisas S, Verma S, Robertson A, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Enhanced differentiation of human dopaminergic neuronal cell model for preclinical translational research in Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165533. [PMID: 31442530 PMCID: PMC7010568 DOI: 10.1016/j.bbadis.2019.165533] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022]
Abstract
Human-derived neuronal cell lines are progressively being utilized in understanding neurobiology and preclinical translational research as they are biologically more relevant than rodent-derived cells lines. The Lund human mesencephalic (LUHMES) cell line comprises human neuronal cells that can be differentiated to post-mitotic neurons and is increasingly being used as an in vitro model for various neurodegenerative diseases. A previously published 2-step differentiation procedure leads to the generation of post-mitotic neurons within 5-days, but only a small proportion (10%) of the total cell population tests positive for tyrosine hydroxylase (TH). Here we report on a novel differentiation protocol that we optimized by using a cocktail of neurotrophic factors, pleiotropic cytokines, and antioxidants to effectively generate proportionately more dopaminergic neurons within the same time period. Visualization and quantification of TH-positive cells revealed that under our new protocol, 25% of the total cell population expressed markers of dopaminergic neurons with the TH-positive neuron count peaking on day 5. These neurons showed spontaneous electrical activity and responded to known Parkinsonian toxins as expected by showing decreased cell viability and dopamine uptake and a concomitant increase in apoptotic cell death. Together, our results outline an improved method for generating a higher proportion of dopaminergic neurons, thus making these cells an ideal neuronal culture model of Parkinson's disease (PD) for translational research.
Collapse
Affiliation(s)
- Dilshan S Harischandra
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Dharmin Rokad
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Shivani Ghaisas
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Saurabh Verma
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Alan Robertson
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Huajun Jin
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Vellareddy Anantharam
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Arthi Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, Parkinson's Disorder Research Program, Iowa State University, Ames, IA, USA.
| |
Collapse
|
7
|
Yamakawa M, Santosa SM, Chawla N, Ivakhnitskaia E, Del Pino M, Giakas S, Nadel A, Bontu S, Tambe A, Guo K, Han KY, Cortina MS, Yu C, Rosenblatt MI, Chang JH, Azar DT. Transgenic models for investigating the nervous system: Currently available neurofluorescent reporters and potential neuronal markers. Biochim Biophys Acta Gen Subj 2020; 1864:129595. [PMID: 32173376 DOI: 10.1016/j.bbagen.2020.129595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
Recombinant DNA technologies have enabled the development of transgenic animal models for use in studying a myriad of diseases and biological states. By placing fluorescent reporters under the direct regulation of the promoter region of specific marker proteins, these models can localize and characterize very specific cell types. One important application of transgenic species is the study of the cytoarchitecture of the nervous system. Neurofluorescent reporters can be used to study the structural patterns of nerves in the central or peripheral nervous system in vivo, as well as phenomena involving embryologic or adult neurogenesis, injury, degeneration, and recovery. Furthermore, crucial molecular factors can also be screened via the transgenic approach, which may eventually play a major role in the development of therapeutic strategies against diseases like Alzheimer's or Parkinson's. This review describes currently available reporters and their uses in the literature as well as potential neural markers that can be leveraged to create additional, robust transgenic models for future studies.
Collapse
Affiliation(s)
- Michael Yamakawa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Samuel M Santosa
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Neeraj Chawla
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Evguenia Ivakhnitskaia
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Matthew Del Pino
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Sebastian Giakas
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Arnold Nadel
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Sneha Bontu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Arjun Tambe
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Kai Guo
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Maria Soledad Cortina
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Charles Yu
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Jin-Hong Chang
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America.
| | - Dimitri T Azar
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America.
| |
Collapse
|
8
|
Mine Y, Momiyama T, Hayashi T, Kawase T. Grafted Miniature-Swine Neural Stem Cells of Early Embryonic Mesencephalic Neuroepithelial Origin can Repair the Damaged Neural Circuitry of Parkinson's Disease Model Rats. Neuroscience 2018; 386:51-67. [PMID: 29932984 DOI: 10.1016/j.neuroscience.2018.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 06/02/2018] [Accepted: 06/04/2018] [Indexed: 12/21/2022]
Abstract
Although recent progress in the use of human iPS cell-derived midbrain dopaminergic progenitors is remarkable, alternatives are essential in the strategies of treatment of basal-ganglia-related diseases. Attention has been focused on neural stem cells (NSCs) as one of the possible candidates of donor material for neural transplantation, because of their multipotency and self-renewal characteristics. In the present study, miniature-swine (mini-swine) mesencephalic neuroepithelial stem cells (M-NESCs) of embryonic 17 and 18 days grafted in the parkinsonian rat striatum were assessed immunohistochemically, behaviorally and electrophysiologically to confirm their feasibility for the neural xenografting as a donor material. Grafted mini-swine M-NESCs survived in parkinsonian rat striatum at 8 weeks after transplantation and many of them differentiated into tyrosine hydroxylase (TH)-positive cells. The parkinsonian model rats grafted with mini-swine M-NESCs exhibited a functional recovery from their parkinsonian behavioral defects. The majority of donor-derived TH-positive cells exhibited a matured morphology at 8 weeks. Whole-cell recordings from donor-derived neurons in the host rat brain slices incorporating the graft revealed the presence of multiple types of neurons including dopaminergic. Glutamatergic and GABAergic post-synaptic currents were evoked in the donor-derived cells by stimulation of the host site, suggesting they receive both excitatory and inhibitory synaptic inputs from host area. The present study shows that non-rodent mammalian M-NESCs can differentiate into functionally active neurons in the diseased xenogeneic environment and could improve the parkinsonian behavioral defects over the species. Neuroepithelial stem cells could be an attractive candidate as a source of donor material for neural transplantation.
Collapse
Affiliation(s)
- Yutaka Mine
- Department of Neurosurgery and Endovascular Surgery, Brain Nerve Center, Saiseikai Yokohamashi Tobu Hospital, Yokohama 230-8765, Japan; Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Clinical Research, Tochigi Medical Center, National Hospital Organization, Utsunomiya 320-8580, Japan
| | - Toshihiko Momiyama
- Division of Cerebral Structure, National Institute for Physiological Sciences, Okazaki 444-8787, Japan; Department of Pharmacology, Jikei University School of Medicine, Tokyo 105-8461, Japan.
| | - Takuro Hayashi
- Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan; Department of Neurosurgery, Tokyo Medical Center, National Hospital Organization, Tokyo 152-8902, Japan
| | - Takeshi Kawase
- Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
9
|
Whoolery CW, Walker AK, Richardson DR, Lucero MJ, Reynolds RP, Beddow DH, Clark KL, Shih HY, LeBlanc JA, Cole MG, Amaral WZ, Mukherjee S, Zhang S, Ahn F, Bulin SE, DeCarolis NA, Rivera PD, Chen BPC, Yun S, Eisch AJ. Whole-Body Exposure to 28Si-Radiation Dose-Dependently Disrupts Dentate Gyrus Neurogenesis and Proliferation in the Short Term and New Neuron Survival and Contextual Fear Conditioning in the Long Term. Radiat Res 2017; 188:532-551. [PMID: 28945526 PMCID: PMC5901735 DOI: 10.1667/rr14797.1] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Astronauts traveling to Mars will be exposed to chronic low doses of galactic cosmic space radiation, which contains highly charged, high-energy (HZE) particles. 56Fe-HZE-particle exposure decreases hippocampal dentate gyrus (DG) neurogenesis and disrupts hippocampal function in young adult rodents, raising the possibility of impaired astronaut cognition and risk of mission failure. However, far less is known about how exposure to other HZE particles, such as 28Si, influences hippocampal neurogenesis and function. To compare the influence of 28Si exposure on indices of neurogenesis and hippocampal function with previous studies on 56Fe exposure, 9-week-old C57BL/6J and Nestin-GFP mice (NGFP; made and maintained for 10 or more generations on a C57BL/6J background) received whole-body 28Si-particle-radiation exposure (0, 0.2 and 1 Gy, 300 MeV/n, LET 67 KeV/μ, dose rate 1 Gy/min). For neurogenesis assessment, the NGFP mice were injected with the mitotic marker BrdU at 22 h postirradiation and brains were examined for indices of hippocampal proliferation and neurogenesis, including Ki67+, BrdU+, BrdU+NeuN+ and DCX+ cell numbers at short- and long-term time points (24 h and 3 months postirradiation, respectively). In the short-term group, stereology revealed fewer Ki67+, BrdU+ and DCX+ cells in 1-Gy-irradiated group relative to nonirradiated control mice, fewer Ki67+ and DCX+ cells in 0.2 Gy group relative to control group and fewer BrdU+ and DCX+ cells in 1 Gy group relative to 0.2 Gy group. In contrast to the clearly observed radiation-induced, dose-dependent reductions in the short-term group across all markers, only a few neurogenesis indices were changed in the long-term irradiated groups. Notably, there were fewer surviving BrdU+ cells in the 1 Gy group relative to 0- and 0.2-Gy-irradiated mice in the long-term group. When the short- and long-term groups were analyzed by sex, exposure to radiation had a similar effect on neurogenesis indices in male and female mice, although only male mice showed fewer surviving BrdU+ cells in the long-term group. Fluorescent immunolabeling and confocal phenotypic analysis revealed that most surviving BrdU+ cells in the long-term group expressed the neuronal marker NeuN, definitively confirming that exposure to 1 Gy 28Si radiation decreased the number of surviving adult-generated neurons in male mice relative to both 0- and 0.2-Gy-irradiated mice. For hippocampal function assessment, 9-week-old male C57BL/6J mice received whole-body 28Si-particle exposure and were then assessed long-term for performance on contextual and cued fear conditioning. In the context test the animals that received 0.2 Gy froze less relative to control animals, suggesting decreased hippocampal-dependent function. However, in the cued fear conditioning test, animals that received 1 Gy froze more during the pretone portion of the test, relative to controls and 0.2-Gy-irradiated mice, suggesting enhanced anxiety. Compared to previously reported studies, these data suggest that 28Si-radiation exposure damages neurogenesis, but to a lesser extent than 56Fe radiation and that low-dose 28Si exposure induces abnormalities in hippocampal function, disrupting fear memory but also inducing anxiety-like behavior. Furthermore, exposure to 28Si radiation decreased new neuron survival in long-term male groups but not females suggests that sex may be an important factor when performing brain health risk assessment for astronauts traveling in space.
Collapse
Affiliation(s)
- Cody W. Whoolery
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Angela K. Walker
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | | | - Melanie J. Lucero
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Ryan P. Reynolds
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David H. Beddow
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - K. Lyles Clark
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hung-Ying Shih
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Junie A. LeBlanc
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Mara G. Cole
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | | | - Shibani Mukherjee
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Shichuan Zhang
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Francisca Ahn
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Sarah E. Bulin
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | | | - Phillip D. Rivera
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
| | - Benjamin P. C. Chen
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, Texas
| | - Sanghee Yun
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Amelia J. Eisch
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, Texas
- Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Pires AO, Teixeira FG, Mendes-Pinheiro B, Serra SC, Sousa N, Salgado AJ. Old and new challenges in Parkinson's disease therapeutics. Prog Neurobiol 2017; 156:69-89. [PMID: 28457671 DOI: 10.1016/j.pneurobio.2017.04.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 03/15/2017] [Accepted: 04/20/2017] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic neurons and/or loss od neuronal projections, in several dopaminergic networks. Current treatments for idiopathic PD rely mainly on the use of pharmacologic agents to improve motor symptomatology of PD patients. Nevertheless, so far PD remains an incurable disease. Therefore, it is of utmost importance to establish new therapeutic strategies for PD treatment. Over the last 20 years, several molecular, gene and cell/stem-cell therapeutic approaches have been developed with the aim of counteracting or retarding PD progression. The scope of this review is to provide an overview of PD related therapies and major breakthroughs achieved within this field. In order to do so, this review will start by focusing on PD characterization and current treatment options covering thereafter molecular, gene and cell/stem cell-based therapies that are currently being studied in animal models of PD or have recently been tested in clinical trials. Among stem cell-based therapies, those using MSCs as possible disease modifying agents for PD therapy and, specifically, the MSCs secretome contribution to meet the clinical challenge of counteracting or retarding PD progression, will be more deeply explored.
Collapse
Affiliation(s)
- Ana O Pires
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - F G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - B Mendes-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Sofia C Serra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
11
|
Krashia P, Martini A, Nobili A, Aversa D, D'Amelio M, Berretta N, Guatteo E, Mercuri NB. On the properties of identified dopaminergic neurons in the mouse substantia nigra and ventral tegmental area. Eur J Neurosci 2016; 45:92-105. [PMID: 27519559 DOI: 10.1111/ejn.13364] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/29/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023]
Abstract
We studied the properties of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and ventral tegmental area (VTA) in mice expressing the enhanced green fluorescent protein (eGFP) under the control of the tyrosine hydroxylase promoter (TH-GFP). By using a practical map of cell positioning in distinct SNpc and VTA subregions in horizontal midbrain slices we saw that the spontaneous firing, membrane properties, cell body size and magnitude of the hyperpolarization-activated current (Ih ) in TH-GFP-positive neurons (TH-GFP+ ) vary significantly among subregions, following a mediolateral gradient. Block of Ih with Zd7288 inhibited firing in the most lateral subregions, but had little effect in the intermediate/medial VTA. In addition, TH-GFP+ cells were excited by Met5 -Enkephalin. Extracellular recordings from a large neuron number showed that all TH-GFP+ cells were inhibited by dopamine, suggesting that this is a reliable approach for identifying dopaminergic neurons in vitro. Simultaneous recordings from dopamine-sensitive and dopamine-insensitive neurons showed that dopamine-insensitive cells (putative non-dopaminergic neurons) are unaffected by Zd7288 but inhibited by Met5 -Enkephalin. Under patch-clamp, dopamine generated a quantitatively similar outward current in most TH-GFP+ neurons, although medial VTA cells showed reduced dopamine sensitivity. Pargyline prolonged the dopamine current, whereas cocaine enhanced dopamine-mediated responses in both the SNpc and the VTA. Our work provides new insights into the variability in mouse midbrain dopaminergic neurons along the medial-lateral axis and points to the necessity of a combination of different electrophysiological and pharmacological approaches for reliably identifying these cells to distinguish them from non-dopaminergic neurons in the midbrain.
Collapse
Affiliation(s)
- Paraskevi Krashia
- Department of Experimental Neurology, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Systems Medicine, Faculty of Medicine, University of Rome 'Tor Vergata', Via Montpellier 1, 00133, Rome, Italy
| | - Alessandro Martini
- Department of Experimental Neurology, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Systems Medicine, Faculty of Medicine, University of Rome 'Tor Vergata', Via Montpellier 1, 00133, Rome, Italy
| | - Annalisa Nobili
- Department of Experimental Neurology, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Medicine, University Campus-Biomedico, Rome, Italy
| | - Daniela Aversa
- Department of Experimental Neurology, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Systems Medicine, Faculty of Medicine, University of Rome 'Tor Vergata', Via Montpellier 1, 00133, Rome, Italy
| | - Marcello D'Amelio
- Department of Experimental Neurology, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Medicine, University Campus-Biomedico, Rome, Italy
| | - Nicola Berretta
- Department of Experimental Neurology, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Ezia Guatteo
- Department of Experimental Neurology, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Motor Science and Wellness, University of Naples Parthenope, Naples, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neurology, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143, Rome, Italy.,Department of Systems Medicine, Faculty of Medicine, University of Rome 'Tor Vergata', Via Montpellier 1, 00133, Rome, Italy
| |
Collapse
|
12
|
Shen Y, Huang J, Liu L, Xu X, Han C, Zhang G, Jiang H, Li J, Lin Z, Xiong N, Wang T. A Compendium of Preparation and Application of Stem Cells in Parkinson's Disease: Current Status and Future Prospects. Front Aging Neurosci 2016; 8:117. [PMID: 27303288 PMCID: PMC4885841 DOI: 10.3389/fnagi.2016.00117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Parkinson's Disease (PD) is a progressively neurodegenerative disorder, implicitly characterized by a stepwise loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and explicitly marked by bradykinesia, rigidity, resting tremor and postural instability. Currently, therapeutic approaches available are mainly palliative strategies, including L-3,4-dihydroxy-phenylalanine (L-DOPA) replacement therapy, DA receptor agonist and deep brain stimulation (DBS) procedures. As the disease proceeds, however, the pharmacotherapeutic efficacy is inevitably worn off, worse still, implicated by side effects of motor response oscillations as well as L-DOPA induced dyskinesia (LID). Therefore, the frustrating status above has propeled the shift to cell replacement therapy (CRT), a promising restorative therapy intending to secure a long-lasting relief of patients' symptoms. By far, stem cell lines of multifarious origins have been established, which can be further categorized into embryonic stem cells (ESCs), neural stem cells (NSCs), induced neural stem cells (iNSCs), mesenchymal stem cells (MSCs), and induced pluripotent stem cells (iPSCs). In this review, we intend to present a compendium of preparation and application of multifarious stem cells, especially in relation to PD research and therapy. In addition, the current status, potential challenges and future prospects for practical CRT in PD patients will be elaborated as well.
Collapse
Affiliation(s)
- Yan Shen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jinsha Huang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Ling Liu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Xiaoyun Xu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Chao Han
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Haiyang Jiang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jie Li
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Zhicheng Lin
- Department of Psychiatry, Harvard Medical School, Division of Alcohol and Drug Abuse, and Mailman Neuroscience Research Center, McLean Hospital Belmont, MA, USA
| | - Nian Xiong
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Tao Wang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| |
Collapse
|
13
|
Haobam R, Tripathy D, Kaidery NA, Mohanakumar KP. Embryonic stem cells derived neuron transplantation recovery in models of parkinsonism in relation to severity of the disorder in rats. Rejuvenation Res 2016; 18:173-84. [PMID: 25546608 DOI: 10.1089/rej.2014.1626] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
6-Hydroxydopamine (6-OHDA)- and 1-methyl-4-phenylpyridinium (MPP(+))-induced hemi-parkinsonism was investigated in relation to the severity of the disorder in terms of behavioral disability and nigral neuronal loss and recovery regarding the number of stem cell-derived neurons transplanted in the striatum. Intra-median forebrain bundle infusion of the parkinsonian neurotoxins and intra-striatal transplantation of differentiated embryonic stem cells (ESCs) were carried out by rat brain stereotaxic surgery. The severity of the disease was determined using the number of amphetamine- or apomorphine-induced rotations, striatal dopamine levels as estimated by high-performance liquid chromatography (HPLC)-electrochemistry, and the number of surviving tyrosine hydroxylase immunoreactive dopaminergic neurons in the substantia nigra pars compacta. Rats that received unilateral infusion of 6-OHDA or MPP(+) responded with dose-dependent, unilateral bias in turning behavior when amphetamine or apomorphine was administered. Rotational asymmetry in both models correlated significantly well with the loss in the number of nigral dopaminergic neurons and striatal dopamine depletion. Transplantation of 2×10(5) differentiated murine ESCs revealed remarkably similar kinds of recovery in both animal models. The survival of the grafted dopaminergic cells in the striatum was better in animals with low-severity parkinsonism, but poor in the animals with severe parkinsonism. Amphetamine-induced rotational recovery correlated positively with an increasing number of cells transplanted in animals with uniform nigral neuronal lesion. These results suggest that disease severity is an important factor for determining the number of cells to be transplanted in parkinsonian rats for desirable recovery, which may be true in clinical conditions too.
Collapse
Affiliation(s)
- Reena Haobam
- 1 Division of Cell Biology & Physiology, Laboratory of Clinical and Experimental Neuroscience, CSIR-Indian Institute of Chemical Biology , Jadavpur, Kolkata, India
| | | | | | | |
Collapse
|
14
|
Characterization of Nestin-positive stem Leydig cells as a potential source for the treatment of testicular Leydig cell dysfunction. Cell Res 2014; 24:1466-85. [PMID: 25418539 PMCID: PMC4260348 DOI: 10.1038/cr.2014.149] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 01/19/2014] [Accepted: 09/01/2014] [Indexed: 11/23/2022] Open
Abstract
The ability to identify and isolate lineage-specific stem cells from adult tissues could facilitate cell replacement therapy. Leydig cells (LCs) are the primary source of androgen in the mammalian testis, and the prospective identification of stem Leydig cells (SLCs) may offer new opportunities for treating testosterone deficiency. Here, in a transgenic mouse model expressing GFP driven by the Nestin (Nes) promoter, we observed Nes-GFP+ cells located in the testicular interstitial compartment where SLCs normally reside. We showed that these Nes-GFP+ cells expressed LIFR and PDGFR-α, but not LC lineage markers. We further observed that these cells were capable of clonogenic self-renewal and extensive proliferation in vitro and could differentiate into neural or mesenchymal cell lineages, as well as LCs, with the ability to produce testosterone, under defined conditions. Moreover, when transplanted into the testes of LC-disrupted or aging models, the Nes-GFP+ cells colonized the interstitium and partially increased testosterone production, and then accelerated meiotic and post-meiotic germ cell recovery. In addition, we further demonstrated that CD51 might be a putative cell surface marker for SLCs, similar with Nestin. Taken together, these results suggest that Nes-GFP+ cells from the testis have the characteristics of SLCs, and our study would shed new light on developing stem cell replacement therapy for testosterone deficiency.
Collapse
|
15
|
Wei PC, Chao A, Peng HH, Chao AS, Chang YL, Chang SD, Wang HS, Chang YJ, Tsai MS, Sieber M, Chen HC, Chen SJ, Lee YS, Hwang SM, Wang TH. SOX9 as a Predictor for Neurogenesis Potentiality of Amniotic Fluid Stem Cells. Stem Cells Transl Med 2014; 3:1138-47. [PMID: 25154783 DOI: 10.5966/sctm.2014-0019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Preclinical studies of amniotic fluid-derived cell therapy have been successful in the research of neurodegenerative diseases, peripheral nerve injury, spinal cord injury, and brain ischemia. Transplantation of human amniotic fluid stem cells (AFSCs) into rat brain ventricles has shown improvement in symptoms of Parkinson's disease and also highlighted the minimal immune rejection risk of AFSCs, even between species. Although AFSCs appeared to be a promising resource for cell-based regenerative therapy, AFSCs contain a heterogeneous pool of distinct cell types, rendering each preparation of AFSCs unique. Identification of predictive markers for neuron-prone AFSCs is necessary before such stem cell-based therapeutics can become a reality. In an attempt to identify markers of AFSCs to predict their ability for neurogenesis, we performed a two-phase study. In the discovery phase of 23 AFSCs, we tested ZNF521/Zfp521, OCT6, SOX1, SOX2, SOX3, and SOX9 as predictive markers of AFSCs for neural differentiation. In the validation phase, the efficacy of these predictive markers was tested in independent sets of 18 AFSCs and 14 dental pulp stem cells (DPSCs). We found that high expression of SOX9 in AFSCs is associated with good neurogenetic ability, and these positive correlations were confirmed in independent sets of AFSCs and DPSCs. Furthermore, knockdown of SOX9 in AFSCs inhibited their neuronal differentiation. In conclusion, the discovery of SOX9 as a predictive marker for neuron-prone AFSCs could expedite the selection of useful clones for regenerative medicine, in particular, in neurological diseases and injuries.
Collapse
Affiliation(s)
- Pei-Cih Wei
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Angel Chao
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Hsiu-Huei Peng
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - An-Shine Chao
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Yao-Lung Chang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Shuenn-Dyh Chang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Hsin-Shih Wang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Yu-Jen Chang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Ming-Song Tsai
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Martin Sieber
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Hua-Chien Chen
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Shu-Jen Chen
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Yun-Shien Lee
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Shiaw-Min Hwang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| | - Tzu-Hao Wang
- Graduate Institute of Biomedical Sciences, Department of Biomedical Sciences, and School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan, Republic of China; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan, Republic of China; Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan, Republic of China; Prenatal Diagnosis Center, Cathay General Hospital, Taipei, Taiwan, Republic of China; Bionet Corp., Taipei, Taiwan, Republic of China; Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan, Republic of China; Department of Biotechnology, Ming-Chuan University, Taoyuan, Taiwan, Republic of China
| |
Collapse
|
16
|
Brazel CY, Alaythan AA, Felling RJ, Calderon F, Levison SW. Molecular features of neural stem cells enable their enrichment using pharmacological inhibitors of survival-promoting kinases. J Neurochem 2013; 128:376-90. [PMID: 24032666 DOI: 10.1111/jnc.12447] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 09/05/2013] [Accepted: 09/10/2013] [Indexed: 11/28/2022]
Abstract
Isolating a pure population of neural stem cells (NSCs) has been difficult since no exclusive surface markers have been identified for panning or FACS purification. Moreover, additional refinements for maintaining NSCs in culture are required, since NSCs generate a variety of neural precursors (NPs) as they proliferate. Here, we demonstrate that post-natal rat NPs express low levels of pro-apoptotic molecules and resist phosphatidylinositol 3'OH kinase and extracellular regulated kinase 1/2 inhibition as compared to late oligodendrocyte progenitors. Furthermore, maintaining subventricular zone precursors in LY294002 and PD98059, inhibitors of PI3K and ERK1/2 signaling, eliminated lineage-restricted precursors as revealed by enrichment for Nestin(+)/SOX-2(+) cells. The cells that survived formed neurospheres and 89% of these neurospheres were tripotential, generating neurons, astrocytes, and oligodendrocytes. Without this enrichment step, less than 50% of the NPs were Nestin(+)/SOX-2(+) and 42% of the neurospheres were tripotential. In addition, neurospheres enriched using this procedure produced 3-times more secondary neurospheres, supporting the conclusion that this procedure enriches for NSCs. A number of genes that enhance survival were more highly expressed in neurospheres compared to late oligodendrocyte progenitors. Altogether, these studies demonstrate that primitive neural precursors can be enriched using a relatively simple and inexpensive means that will facilitate cell replacement strategies using stem cells as well as other studies whose goal is to reveal the fundamental properties of primitive neural precursors.
Collapse
Affiliation(s)
- Christine Y Brazel
- Department of Neurology and Neurosciences, Rutgers University-New Jersey Medical School, Newark, New Jersey, USA
| | | | | | | | | |
Collapse
|
17
|
Lin CH, Lee HT, Lee SD, Lee W, Cho CWC, Lin SZ, Wang HJ, Okano H, Su CY, Yu YL, Hsu CY, Shyu WC. Role of HIF-1α-activated Epac1 on HSC-mediated neuroplasticity in stroke model. Neurobiol Dis 2013; 58:76-91. [PMID: 23702312 DOI: 10.1016/j.nbd.2013.05.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/02/2013] [Accepted: 05/10/2013] [Indexed: 12/27/2022] Open
Abstract
Exchange protein activated by cAMP-1 (Epac1) plays an important role in cell proliferation, cell survival and neuronal signaling, and activation of Epac1 in endothelial progenitor cells increases their homing to ischemic muscles and promotes neovascularization in a model of hind limb ischemia. Moreover, upregulation of Epac1 occurs during organ development and in diseases such as myocardial hypertrophy, diabetes, and Alzheimer's disease. We report here that hypoxia upregulated Epac1 through HIF-1α induction in the CD34-immunosorted human umbilical cord blood hematopoietic stem cells (hUCB(34)). Importantly, implantation of hUCB(34) subjected to hypoxia-preconditioning (HP-hUCB(34)) improved stroke outcome, more than did implantation of untreated hUCB(34), in rodents subjected to cerebral ischemia, and this required Epac1-to-matrix metalloprotease (MMP) signaling. This improved therapeutic efficacy correlated with better engraftment and differentiation of these cells in the ischemic host brain. In addition, more than did implantation of untreated HP-hUCB(34), implantation of HP-hUCB(34) improved cerebral blood flow into the ischemic brain via induction of angiogenesis, facilitated proliferation/recruitment of endogenous neural progenitor cells in the ischemic brain, and promoted neurite outgrowth following cerebral ischemia. Consistent with our proposed role of Epac1-to-MMP signaling in hypoxia-preconditioning, the above mentioned effects of implanting HP-hUCB(34) could be abolished by pharmacological inhibition and genetic disruption/deletion of Epac1 or MMPs. We have discovered a HIF-1α-to-Epac1-to-MMP signaling pathway that is required for the improved therapeutic efficacy resulting from hypoxia preconditioning of hUCB(34) in vitro prior to their implantation into the host brain in vivo.
Collapse
Affiliation(s)
- Chen-Huan Lin
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Krause P, Unthan-Fechner K, Probst I, Koenig S. Cultured hepatocytes adopt progenitor characteristics and display bipotent capacity to repopulate the liver. Cell Transplant 2013; 23:805-17. [PMID: 23485196 DOI: 10.3727/096368913x664856] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Clinical studies have proved the therapeutic potential of hepatocyte transplantation as a promising alternative to whole organ liver transplantation in the treatment of hereditary or end-stage liver disease. However, donor shortage seriously restricts cell availability, and the lack of appropriate cell culture protocols for the storage and maintenance of donor cells constitutes a significant obstacle. The aim of this study was to stimulate mature hepatocytes in culture to multiply in vitro and track their fate on transplantation. Rat hepatocytes isolated nonenzymatically were cultured serum free for up to 10 days. They were stimulated into proliferation in the presence of growth factors and conditioned media from nonparenchymal and hepatocyte culture supernatants, as well as 10 mM lithium chloride (LiCl). Cell proliferation was assessed by determining DNA content. Additionally, the extent of cell differentiation was estimated using immunofluorescence staining of hepatic, biliary, progenitor, and mesenchymal markers and gene expression analyses. Transplantation studies were performed on the Fischer CD26-mutant rat following pretreatment with retrorsine and partial hepatectomy. Proliferating hepatocytes increasingly adopted precursor characteristics, expressing progenitor (OV6, CD133), hepatic lineage (CK18), biliary (CD49f, CK7, CK19), and mesenchymal (vimentin) markers. The supplement of LiCl further enhanced the proliferative capacity by 30%. Transplantation studies revealed extensive repopulation by large donor hepatocyte clusters. Furthermore, bile duct-like structures deriving from donor cells proved to be immunoreactive to ductular markers and formed in close proximity to endogenous bile ducts. Mature hepatocytes reveal their potential to "switch" between phenotypes, adopting progenitor characteristics during proliferation in vitro. Following transplantation, these "retrodifferentiated" cells further expanded in vivo, thereby generating bipotentially differentiated progenies (hepatocytes and bile duct-like structures). This apparent plasticity of mature hepatocytes may open new approaches for cell-based strategies to treat liver disease.
Collapse
Affiliation(s)
- Petra Krause
- Department of General and Visceral Surgery, University Medical Centre Goettingen, Goettingen, Germany
| | | | | | | |
Collapse
|
19
|
Pignatelli A, Borin M, Fogli Iseppe A, Gambardella C, Belluzzi O. The h-current in periglomerular dopaminergic neurons of the mouse olfactory bulb. PLoS One 2013; 8:e56571. [PMID: 23418585 PMCID: PMC3572079 DOI: 10.1371/journal.pone.0056571] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 01/11/2013] [Indexed: 12/31/2022] Open
Abstract
The properties of the hyperpolarization-activated cation current (I(h)) were investigated in rat periglomerular dopaminergic neurons using patch-clamp recordings in thin slices. A reliable identification of single dopaminergic neurons was made possible by use of a transgenic line of mice expressing eGFP under the tyrosine hydroxylase promoter. At 37 °C and minimizing the disturbance of the intracellular milieu with perforated patches, this current shows a midpoint of activation around -82.7 mV, with a significant level of opening already at rest, thereby giving a substantial contribution to the resting potential, and ultimately playing a relevant function in the control of the cell excitability. The blockage of I(h) has a profound influence on the spontaneous firing of these neurons, which result as strongly depressed. However the effect is not due to a direct role of the current in the pacemaker process, but to the I(h) influence on the resting membrane potential. I(h) kinetics is sensitive to the intracellular levels of cAMP, whose increase promotes a shift of the activation curve towards more positive potentials. The direct application of DA and 5-HT neurotransmitters, physiologically released onto bulbar dopaminergic neurons and known to act on metabotropic receptors coupled to the cAMP pathway, do not modifythe I(h) amplitude. On the contrary, noradrenaline almost halves the I(h) amplitude. Our data indicate that the HCN channels do not participate directly to the pacemaker activity of periglomerular dopaminergic neurons, but influence their resting membrane potential by controlling the excitability profile of these cells, and possibly affecting the processing of sensory information taking place at the entry of the bulbar circuitry.
Collapse
Affiliation(s)
- Angela Pignatelli
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
| | - Mirta Borin
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
| | - Alex Fogli Iseppe
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
| | - Cristina Gambardella
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
| | - Ottorino Belluzzi
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
- * E-mail:
| |
Collapse
|
20
|
Jin GZ, Cho SJ, Choi EG, Lee YS, Yu XF, Choi KS, Yee ST, Jeon JT, Kim MO, Kong IK. Rat mesenchymal stem cells increase tyrosine hydroxylase expression and dopamine content in ventral mesencephalic cells in vitro. Cell Biol Int 2013; 32:1433-8. [DOI: 10.1016/j.cellbi.2008.08.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Revised: 06/25/2008] [Accepted: 08/12/2008] [Indexed: 01/01/2023]
|
21
|
Gambardella C, Pignatelli A, Belluzzi O. The h-current in the substantia Nigra pars compacta neurons: a re-examination. PLoS One 2012; 7:e52329. [PMID: 23284989 PMCID: PMC3528748 DOI: 10.1371/journal.pone.0052329] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 11/12/2012] [Indexed: 01/15/2023] Open
Abstract
The properties of the hyperpolarization-activated cation current (Ih) were investigated in rat substantia nigra - pars compacta (SNc) principal neurons using patch-clamp recordings in thin slices. A reliable identification of single dopaminergic neurons was made possible by the use of a transgenic line of mice expressing eGFP under the tyrosine hydroxylase promoter. The effects of temperature and different protocols on the Ih kinetics showed that, at 37°C and minimizing the disturbance of the intracellular milieu with perforated patch, this current actually activates at potentials more positive than what is generally indicated, with a half-activation potential of −77.05 mV and with a significant level of opening already at rest, thereby substantially contributing to the control of membrane potential, and ultimately playing a relevant function in the regulation of the cell excitability. The implications of the known influence of intracellular cAMP levels on Ih amplitude and kinetics were examined. The direct application of neurotransmitters (DA, 5-HT and noradrenaline) physiologically released onto SNc neurons and known to act on metabotropic receptors coupled to the cAMP pathway modify the Ih amplitude. Here, we show that direct activation of dopaminergic and of 5-HT receptors results in Ih inhibition of SNc DA cells, whereas noradrenaline has the opposite effect. Together, these data suggest that the modulation of Ih by endogenously released neurotransmitters acting on metabotropic receptors –mainly but not exclusively linked to the cAMP pathway- could contribute significantly to the control of SNc neuron excitability.
Collapse
Affiliation(s)
- Cristina Gambardella
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
| | - Angela Pignatelli
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
| | - Ottorino Belluzzi
- Dipartimento di Scienze della Vita e Biotecnologie, University of Ferrara and Istituto Nazionale di Neuroscienze, Ferrara, Italy
- * E-mail:
| |
Collapse
|
22
|
Walton RM, Parmentier T, Wolfe JH. Postnatal neural precursor cell regions in the rostral subventricular zone, hippocampal subgranular zone and cerebellum of the dog (Canis lupus familiaris). Histochem Cell Biol 2012. [PMID: 23192285 DOI: 10.1007/s00418-012-1053-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Identification of neural stem and progenitor cells (NPCs) in vitro and in vivo is essential to the use of developmental and disease models of neurogenesis. The dog is a valuable large animal model for multiple neurodegenerative diseases and is more closely matched to humans than rodents with respect to brain organization and complexity. It is therefore important to determine whether immunohistochemical markers associated with NPCs in humans and rodents are also appropriate for the dog. The NPC markers CD15, CD133, nestin, GFAP and phosphacan (DSD-1) were evaluated in situ in the canine rostral telencephalon, hippocampal dentate gyrus, and cerebellum at different postnatal time-points. Positive staining results were interpreted in the context of region and cellular morphology. Our results showed that neurospheres and cells within the rostral subventricular zone (SVZ), dentate gyrus subgranular zone (SGZ), and white matter tracts of the cerebellum were immunopositive for CD15, nestin and GFAP. Neurospheres and the cerebellum were immunonegative for CD133, whereas CD133 staining was present in the postnatal rostral SVZ. Anti-phosphacan antibody staining delineated the neurogenic niches of the rostral lateral ventricle SVZ and the hippocampal SGZ. Positive staining for phosphacan was also noted in white matter tracts of the cerebellum and within the Purkinje layer. Our results showed that in the dog these markers were associated with regions shown to be neurogenic in rodents and primates.
Collapse
Affiliation(s)
- Raquel M Walton
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
23
|
Choi EY, Yang JW, Park MS, Sun W, Kim H, Kim SU, Lee MA. Transgenic mice expressing yellow fluorescent protein under control of the human tyrosine hydroxylase promoter. J Neurosci Res 2012; 90:1949-59. [PMID: 22714400 DOI: 10.1002/jnr.23085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 04/15/2012] [Indexed: 11/11/2022]
Abstract
Pathogenesis of Parkinson's disease and related catecholaminergic neurological disorders is closely associated with changes in the levels of tyrosine hydroxylase (TH). Therefore, investigation of the regulation of the TH gene system should assist in understanding the pathomechanisms involved in these neurological disorders. To identify regulatory domains that direct human TH expression in the central nervous system (CNS), we generated two transgenic mouse lines in which enhanced yellow fluorescent protein (EYFP) is expressed under the control of either 3.2-kb (hTHP-EYFP construct) human TH promoter or 3.2-kb promoter with 2-kb 3'-flanking regions (hTHP-ex3-EYFP construct) of the TH gene. In the adult transgenic mouse brain, the hTHP-EYFP construct directs neuron-specific EYFP expression in various CNS areas, such as olfactory bulb, striatum, interpeduncular nucleus, cerebral cortex, hippocampus, and particularly dentate gyrus. Although these EYFP-positive cells were identified as mature neurons, few EYFP-positive cells were TH-positive neurons. On the other hand, we could detect the EYFP mRNA expression in a subset of neurons in the olfactory bulb, midbrain, and cerebellum, in which expression of endogenous TH is enriched, with hTHP-ex3-EYFP transgenic mice. These results indicate that the 3.2-kb sequence upstream of the TH gene is not sufficient for proper expression and that the 2-kb sequence from the translation start site to exon 3 is necessary for expression of EYFP in a subset of catecholaminergic neurons.
Collapse
Affiliation(s)
- Eun Yang Choi
- Brain Disease Research Center, and Institute for Medical Sciences, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
'Cancer stem cells' (CSCs) are tumor cells with stem cell properties hypothesized to be responsible for tumorigenesis, metastatis, and resistance to treatment, and have been identified in different tumors including cutaneous melanoma, using stem cell markers such as CD133. This study explored expression of CD133 and other putative stem cell markers in uveal melanoma. Eight uveal melanoma cell lines were subjected to flow-cytometric (fluorescence-activated cell sorting) analysis of CD133 and other stem cell markers. Eight paraffin-embedded tumors were analyzed by immunohistochemistry for CD133, Pax6, Musashi, nestin, Sox2, ABCB5, and CD68 expressions. Ocular, uveal melanoma, and hematopoietic stem cell distributions of C-terminal and N-terminal CD133 mRNA splice variants were compared by reverse-transcription PCR. Fluorescence-activated cell sorting analysis revealed a population of CD133-positive/nestin-positive cells in cell lines Mel270, OMM 2.3, and OMM2.5. All cell lines studied were positive for nestin, CXCR-4, CD44, and c-kit. Immunohistochemistry identified cells positive for CD133, Pax6, Musashi, nestin, Sox2, ABCB5, and CD68 predominantly at the invading tumor front. C-terminal primers interacting with CD133 splice variant s2 detected a novel variant lacking exon 27. Differential expression of CD133 splice variants was found in iris, ciliary body, retina, and retinal pigment epithelium/choroid as well as in uveal melanoma cell lines. mRNA for nestin, Sox2, and Musashi was present in all studied cell lines. Uveal melanoma such as cutaneous melanoma may therefore contain CSCs. Further experiments are needed to isolate stem cell marker-positive cells, to evaluate their functional properties and to explore therapeutical approaches to these putative CSCs in uveal melanoma.
Collapse
|
25
|
Transduction of E13 murine neural precursor cells by non-immunogenic recombinant adeno-associated viruses induces major changes in neuronal phenotype. Neuroscience 2012; 210:82-98. [PMID: 22406416 DOI: 10.1016/j.neuroscience.2012.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 02/03/2012] [Accepted: 02/06/2012] [Indexed: 11/21/2022]
Abstract
Neural precursor cells (NPCs) provide a cellular model to compare transduction efficiency and toxicity for a series of recombinant adeno-associated viruses (rAAVs). Results led to the choice of rAAV9 as a preferred candidate to transduce NPCs for in vivo transplantation. Importantly, transduction promoted a neuronal phenotype characterized by neurofilament M (NFM) with a concomitant decrease in the embryonic marker, nestin, without significant change in glial fibrillary acidic protein (GFAP). In marked contrast to recent studies for induced pluripotent stem cells (iPSCs), exposure to rAAVs is non-immunogenic and these do not result in genetic abnormalities, thus bolstering the earlier use of NPCs such as those isolated from E13 murine cells for clinical applications. Mechanisms of cellular interactions were explored by treatment with genistein, a pan-specific inhibitor of protein receptor tyrosine kinases (PRTKs) that blocked the transduction and differentiation, thus implying a central role for this pathway for inducing infectivity along with observed phenotypic changes and as a method for drug design. Implantation of transduced NPCs into adult mouse hippocampus survived up to 28 days producing a time line for targeting or migration to dentate gyrus and CA3-1 compatible with future clinical applications. Furthermore, a majority showed commitment to highly differentiated neuronal phenotypes. Lack of toxicity and immune response of rAAVs plus ability for expansion of NPCs in vitro auger well for their isolation and suggest potential therapeutic applications in repair or replacement of diseased neurons in neurodegeneration.
Collapse
|
26
|
Abstract
Steroidogenic factor 1 (SF-1; officially designated NR5a1) is a member of a nuclear receptor superfamily with important roles in the development of endocrine systems. Studies with global and tissue-specific (i.e. central nervous system) knockout mice have revealed several roles of SF-1 in brain. These include morphological effects on the development of the ventromedial nucleus of the hypothalamus and functional effects on body weight regulation through modulation of physical activity, anxiety-like behaviours and female sexual behaviours. Although such defects are almost certainly a result of the absence of SF-1 acting as a transcription factor in the hypothalamus, global SF-1 knockout mice also represent a model for studying the sex differences in the brain that develop in the absence of exposure to foetal sex steroid hormones as a result of the absence of gonads. In the present review, current knowledge of the roles of SF-1 protein in the central nervous system is discussed.
Collapse
Affiliation(s)
- T Büdefeld
- Centre for Animal Genomics, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | | | |
Collapse
|
27
|
Wakeman DR, Dodiya HB, Kordower JH. Cell transplantation and gene therapy in Parkinson's disease. ACTA ACUST UNITED AC 2011; 78:126-58. [PMID: 21259269 DOI: 10.1002/msj.20233] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Parkinson's disease is a progressive neurodegenerative disorder affecting, in part, dopaminergic motor neurons of the ventral midbrain and their terminal projections that course to the striatum. Symptomatic strategies focused on dopamine replacement have proven effective at remediating some motor symptoms during the course of disease but ultimately fail to deliver long-term disease modification and lose effectiveness due to the emergence of side effects. Several strategies have been experimentally tested as alternatives for Parkinson's disease, including direct cell replacement and gene transfer through viral vectors. Cellular transplantation of dopamine-secreting cells was hypothesized as a substitute for pharmacotherapy to directly provide dopamine, whereas gene therapy has primarily focused on restoration of dopamine synthesis or neuroprotection and restoration of spared host dopaminergic circuitry through trophic factors as a means to enhance sustained controlled dopamine transmission. This seems now to have been verified in numerous studies in rodents and nonhuman primates, which have shown that grafts of fetal dopamine neurons or gene transfer through viral vector delivery can lead to improvements in biochemical and behavioral indices of dopamine deficiency. However, in clinical studies, the improvements in parkinsonism have been rather modest and variable and have been plagued by graft-induced dyskinesias. New developments in stem-cell transplantation and induced patient-derived cells have opened the doors for the advancement of cell-based therapeutics. In addition, viral-vector-derived therapies have been developed preclinically with excellent safety and efficacy profiles, showing promise in clinical trials thus far. Further progress and optimization of these therapies will be necessary to ensure safety and efficacy before widespread clinical use is deemed appropriate.
Collapse
|
28
|
Cucchiaroni ML, Freestone PS, Berretta N, Viscomi MT, Bisicchia E, Okano H, Molinari M, Bernardi G, Lipski J, Mercuri NB, Guatteo E. Properties of dopaminergic neurons in organotypic mesencephalic-striatal co-cultures - evidence for a facilitatory effect of dopamine on the glutamatergic input mediated by α-1 adrenergic receptors. Eur J Neurosci 2011; 33:1622-36. [DOI: 10.1111/j.1460-9568.2011.07659.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
29
|
Thibault D, Albert PR, Pineyro G, Trudeau LÉ. Neurotensin triggers dopamine D2 receptor desensitization through a protein kinase C and beta-arrestin1-dependent mechanism. J Biol Chem 2011; 286:9174-84. [PMID: 21233215 PMCID: PMC3059057 DOI: 10.1074/jbc.m110.166454] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 01/12/2011] [Indexed: 11/06/2022] Open
Abstract
The peptide neurotensin (NT) is known to exert a potent excitatory effect on the dopaminergic system by inhibiting D2 dopamine (DA) receptor (D2R) function. This regulation is dependent on activation of PKC, a well known effector of the type 1 NT receptor (NTR1). Because PKC phosphorylation of the D2R has recently been shown to induce its internalization, we hypothesized that NT acts to reduce D2R function through heterologous desensitization of the D2R. In the present study, we first used HEK-293 cells to demonstrate that NT induces PKC-dependent D2R internalization. Furthermore, internalization displayed faster kinetics in cells expressing the D2R short isoform, known to act as an autoreceptor in DA neurons, than in cells expressing the long isoform, known to act as a postsynaptic D2R. In patch clamp experiments on cultured DA neurons, overexpression of a mutant D2S lacking three key PKC phosphorylation sites abrogated the ability of NT to reduce D2R-mediated cell firing inhibition. Short interfering RNA-mediated inhibition of β-arrestin1 and dynamin2, proteins important for receptor desensitization, reduced agonist-induced desensitization of D2R function, but only the inhibition of β-arrestin1 reduced the effect of NT on D2R function. Taken together, our data suggest that NT acutely regulates D2 autoreceptor function and DA neuron excitability through PKC-mediated phosphorylation of the D2R, leading to heterologous receptor desensitization.
Collapse
Affiliation(s)
- Dominic Thibault
- From the Department of Pharmacology
- Department of Physiology
- the Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Québec H3C 3J7, Canada
| | - Paul R. Albert
- the Ottawa Hospital Research Institute, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Graciela Pineyro
- From the Department of Pharmacology
- Department of Psychiatry, Faculty of Medicine, and
- the Centre de Recherche du Centre Hospitalier Universitaire Sainte Justine, Université de Montréal, Quebec H3T 1C5, Canada, and
| | - Louis-Éric Trudeau
- From the Department of Pharmacology
- Department of Physiology
- Department of Psychiatry, Faculty of Medicine, and
- the Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Québec H3C 3J7, Canada
| |
Collapse
|
30
|
Malmersjö S, Liste I, Dyachok O, Tengholm A, Arenas E, Uhlén P. Ca2+ and cAMP signaling in human embryonic stem cell-derived dopamine neurons. Stem Cells Dev 2011; 19:1355-64. [PMID: 20043754 DOI: 10.1089/scd.2009.0436] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Human embryonic stem (hES) cell differentiation into dopamine neurons is considered a promising strategy for cell replacement therapy in Parkinson's disease, yet the functional properties of hES cell-derived dopamine neurons remain poorly defined. The objective of this study was to characterize intracellular calcium (Ca(2+)) and sub-plasma membrane cyclic AMP-signaling properties in hES cell-derived dopamine neurons. We found that hES cell-derived dopamine neurons and neural progenitors raised Ca(2+) from intra- and extracellular compartments in response to depolarization, glutamate, ATP, and dopamine D(2) receptor activation, while undifferentiated hES cells only mobilized Ca(2+) from intracellular stores in response to ATP and D(2) receptor-induced activation. Interestingly, we also found that hES cell-derived dopamine neurons in addition to primary ventral midbrain dopamine neurons were more prone to release Ca(2+) from intracellular stores than non-dopamine neurons following treatment with the neuropeptide neurotensin. Furthermore, hES cell-derived dopamine neurons showed cAMP elevations in response to forskolin and 3-isobutyl-methylxanthine, similar to primary dopamine neurons. Taken together, these results unravel the temporal sequence by which hES cells acquire Ca(2+) and cAMP signaling competence during dopamine differentiation.
Collapse
Affiliation(s)
- Seth Malmersjö
- Department of Medical Biochemistry and Biophysics, Linnaeus Center in Developmental Biology for Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
31
|
Prospects and limitations of using endogenous neural stem cells for brain regeneration. Genes (Basel) 2011; 2:107-30. [PMID: 24710140 PMCID: PMC3924842 DOI: 10.3390/genes2010107] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Revised: 12/06/2010] [Accepted: 01/04/2011] [Indexed: 01/19/2023] Open
Abstract
Neural stem cells (NSCs) are capable of producing a variety of neural cell types, and are indispensable for the development of the mammalian brain. NSCs can be induced in vitro from pluripotent stem cells, including embryonic stem cells and induced-pluripotent stem cells. Although the transplantation of these exogenous NSCs is a potential strategy for improving presently untreatable neurological conditions, there are several obstacles to its implementation, including tumorigenic, immunological, and ethical problems. Recent studies have revealed that NSCs also reside in the adult brain. The endogenous NSCs are activated in response to disease or trauma, and produce new neurons and glia, suggesting they have the potential to regenerate damaged brain tissue while avoiding the above-mentioned problems. Here we present an overview of the possibility and limitations of using endogenous NSCs in regenerative medicine.
Collapse
|
32
|
Characterization and Classification of Stem Cells. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
33
|
Hamanoue M, Okano H. Cell surface N-glycans-mediated isolation of mouse neural stem cells. J Cell Physiol 2010; 226:1433-8. [PMID: 20945342 DOI: 10.1002/jcp.22436] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The isolation of neural stem cells (NSCs) has been hampered by the lack of valid cell-surface antigens on NSCs, and novel valuable markers have been proposed. Glycan (oligosaccharide chain) is a potential candidate as a marker to isolate NSCs, because the species and the combination order of saccharides in glycan generate remarkable structural diversity and specificity. At present, the expression of hundreds of glycoconjugates with glycans have been found in the NSCs; however, just a few glycan-epitopes have been identified as valuable cell-surface markers. This review focused on the isolation of NSC using glycoprotein, especially complex type N-glycans. The cell-surface N-glycan-mediated isolation of NSCs is therefore expected to provide a comprehensive understanding of the biologic characteristics of NSCs in the brain, and thereby help to develop novel strategies in the field of regenerative medicine.
Collapse
Affiliation(s)
- Makoto Hamanoue
- Department of Physiology, Toho University School of Medicine, Tokyo, Japan.
| | | |
Collapse
|
34
|
Higashida T, Jitsuki S, Kubo A, Mitsushima D, Kamiya Y, Kanno H. Skin-derived precursors differentiating into dopaminergic neuronal cells in the brains of Parkinson disease model rats. J Neurosurg 2010; 113:648-55. [PMID: 20302395 DOI: 10.3171/2010.2.jns091432] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECT In the authors' previous study, they observed that amino acids 157-171 of von Hippel-Lindau protein (VHL peptide) induced neuronal differentiation of skin-derived precursors. They also noted that transplantation of these differentiated cells into the striata of a Parkinson disease (PD) rat model reduced apomorphine-induced rotations. In the present study, they investigated if these cells produce dopamine in the striatum. METHODS Skin-derived precursors were differentiated into neurons using VHL peptide and transplanted into the striata of a PD model of rats. Four weeks after transplantation, a probe was inserted into rat striata and extracellular dopamine was extracted by microdialysis. Dopamine levels were measured by high-pressure liquid chromatography. Brain sections were assessed by immunohistochemical analysis for the presence of tyrosine hydroxylase and dopamine transporter. RESULTS Increased dopamine levels in the striata of the rats were observed after transplantation (p < 0.01), and these were correlated with a reduction in the number of apomorphine-induced rotations (p < 0.05). Skin-derived precursors observed along the tract of transplantation were positive for tyrosine hydroxylase and dopamine transporter. CONCLUSIONS This study suggests that transplantation of skin-derived precursors, differentiated into neuronal cells using VHL peptide, can improve PD-like symptoms by enabling production of dopamine in the striata in a PD model of rats.
Collapse
Affiliation(s)
- Tetsuhiro Higashida
- Department of Neurosurgery, Yokohama City University, School of Medicine, Yokohama, Kanagawa, Japan.
| | | | | | | | | | | |
Collapse
|
35
|
Sun J, Gao Q, Zhang J, Bao L, Dong H, Liang N, Li G, Li Z, Gao Y. Ephrinb3 induces mesostriatal dopaminergic projection to the striatum. Biochem Biophys Res Commun 2010; 400:194-9. [DOI: 10.1016/j.bbrc.2010.08.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 08/07/2010] [Indexed: 12/26/2022]
|
36
|
King FW, Ritner C, Liszewski W, Kwan HCK, Pedersen A, Leavitt AD, Bernstein HS. Subpopulations of human embryonic stem cells with distinct tissue-specific fates can be selected from pluripotent cultures. Stem Cells Dev 2010; 18:1441-50. [PMID: 19254177 DOI: 10.1089/scd.2009.0012] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Directed differentiation of human embryonic stem cells (hESCs) has generated much interest in the field of regenerative medicine. While subpopulations of hESCs within pluripotent cultures have been identified based on expression of specific surface antigens, their significance and fates are not well understood. To determine whether such subpopulations indicate specific tissue fates or represent stochastic antigen distributions within proliferating cultures, we isolated CD133(+) or CD135(+) hESCs from proliferating cultures constitutively expressing enhanced green fluorescent protein (GFP), and co-cultured these with unselected GFP(-) hESCs. After passage in culture, GFP(+) hESCs reanalyzed for the persistence of CD133 or CD135 expression, as well as other surface antigens (Tra-1-60, SSEA-4, FGFR-1), demonstrated that these two subpopulations continued to express CD133 or CD135 over serial passage, and that CD133(+) hESCs were enriched for SSEA-4 expression as well. Upon differentiation in vitro, CD133(+)GFP(+) hESCs gave rise solely to ectoderm, as detected by expression of nestin. Tissues representing endoderm (alpha-fetoprotein(+)) and mesoderm (smooth muscle actin(+)) were not seen among GFP(+) tissues. In contrast, selection against CD133 gave rise almost exclusively to mesoderm and endoderm. In contrast, CD135(+)GFP(+) hESCs gave rise to tissues representing all three embryonic germ layers, and were virtually indistinguishable from CD135(-)-derived tissues. Similar results were obtained by in vivo differentiation in teratomas. These data establish that subpopulations of proliferating hESCs whose tissue fate is predetermined exist, and challenge the notion that all cells within proliferating hESC cultures are truly "pluripotent." This co-culture approach also will enable identification of other distinct hESC subpopulations, and selection for these should prove valuable in generating tissue-specific reagents for cell-based therapy.
Collapse
Affiliation(s)
- Frank W King
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-1346, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Remy S, Tesson L, Usal C, Menoret S, Bonnamain V, Nerriere-Daguin V, Rossignol J, Boyer C, Nguyen TH, Naveilhan P, Lescaudron L, Anegon I. New lines of GFP transgenic rats relevant for regenerative medicine and gene therapy. Transgenic Res 2010; 19:745-63. [PMID: 20094912 DOI: 10.1007/s11248-009-9352-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 12/08/2009] [Indexed: 02/07/2023]
Abstract
Adoptive cell transfer studies in regenerative research and identification of genetically modified cells after gene therapy in vivo require unequivocally identifying and tracking the donor cells in the host tissues, ideally over several days or for up to several months. The use of reporter genes allows identifying the transferred cells but unfortunately most are immunogenic to wild-type hosts and thus trigger rejection in few days. The availability of transgenic animals from the same strain that would express either high levels of the transgene to identify the cells or low levels but that would be tolerant to the transgene would allow performing long-term analysis of labelled cells. Herein, using lentiviral vectors we develop two new lines of GFP-expressing transgenic rats displaying different levels and patterns of GFP-expression. The "high-expresser" line (GFP(high)) displayed high expression in most tissues, including adult neurons and neural precursors, mesenchymal stem cells and in all leukocytes subtypes analysed, including myeloid and plasmacytoid dendritic cells, cells that have not or only poorly characterized in previous GFP-transgenic rats. These GFP(high)-transgenic rats could be useful for transplantation and immunological studies using GFP-positive cells/tissue. The "low-expresser" line expressed very low levels of GFP only in the liver and in less than 5% of lymphoid cells. We demonstrate these animals did not develop detectable humoral and cellular immune responses against both transferred GFP-positive splenocytes and lentivirus-mediated GFP gene transfer. Thus, these GFP-transgenic rats represent useful tools for regenerative medicine and gene therapy.
Collapse
Affiliation(s)
- S Remy
- INSERM, U643, 30 Bd Jean Monnet, 44093, Nantes cedex 01, Nantes, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Takaoka T, Shiotani A, Saito K, Tomifuji M, Mori Y, Fujimine T, Okano H, Ogawa K. Neuronal re-juvenilization in the nucleus ambiguus after vagal nerve injury. Neurosci Res 2009; 65:353-9. [DOI: 10.1016/j.neures.2009.08.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 08/05/2009] [Accepted: 08/24/2009] [Indexed: 10/20/2022]
|
39
|
Hotta R, Natarajan D, Thapar N. Potential of cell therapy to treat pediatric motility disorders. Semin Pediatr Surg 2009; 18:263-73. [PMID: 19782309 DOI: 10.1053/j.sempedsurg.2009.07.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Gut motility disorders represent a significant challenge in clinical management with current palliative approaches failing to overcome disease and treatment-related morbidity. The recent progress with stem cells to restore missing or defective elements of the gut neuromusculature offers new hope for potential cure. Focusing on enteric neuropathies such as Hirschsprung's disease, the review discusses the progress that has been made in the sourcing of putative stem cells and the studies into their biology and therapeutic potential. It also explores the practical challenges that must be overcome before stem cell-based therapies can be applied in the clinical arena. Although many obstacles remain, the speed of advancement of the enteric stem cell field suggests that such therapies are on the horizon.
Collapse
Affiliation(s)
- Ryo Hotta
- Department of Anatomy & Cell Biology, University of Melbourne, Victoria, Australia
| | | | | |
Collapse
|
40
|
Amoh Y, Kanoh M, Niiyama S, Hamada Y, Kawahara K, Sato Y, Hoffman RM, Katsuoka K. Human hair follicle pluripotent stem (hfPS) cells promote regeneration of peripheral-nerve injury: an advantageous alternative to ES and iPS cells. J Cell Biochem 2009; 107:1016-20. [PMID: 19507228 DOI: 10.1002/jcb.22204] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The optimal source of stem cells for regenerative medicine is a major question. Embryonic stem (ES) cells have shown promise for pluripotency but have ethical issues and potential to form teratomas. Pluripotent stem cells have been produced from skin cells by either viral-, plasmid- or transposon-mediated gene transfer. These stem cells have been termed induced pluripotent stem cells or iPS cells. iPS cells may also have malignant potential and are inefficiently produced. Embryonic stem cells may not be suited for individualized therapy, since they can undergo immunologic rejection. To address these fundamental problems, our group is developing hair follicle pluripotent stem (hfPS) cells. Our previous studies have shown that mouse hfPS cells can differentiate to neurons, glial cells in vitro, and other cell types, and can promote nerve and spinal cord regeneration in vivo. hfPS cells are located above the hair follicle bulge in what we have termed the hfPS cell area (hfPSA) and are nestin positive and keratin 15 (K-15) negative. Human hfPS cells can also differentiate into neurons, glia, keratinocytes, smooth muscle cells, and melanocytes in vitro. In the present study, human hfPS cells were transplanted in the severed sciatic nerve of the mouse where they differentiated into glial fibrillary-acidic-protein (GFAP)-positive Schwann cells and promoted the recovery of pre-existing axons, leading to nerve generation. The regenerated nerve recovered function and, upon electrical stimulation, contracted the gastrocnemius muscle. The hfPS cells can be readily isolated from the human scalp, thereby providing an accessible, autologous and safe source of stem cells for regenerative medicine that have important advantages over ES or iPS cells.
Collapse
Affiliation(s)
- Yasuyuki Amoh
- Department of Dermatology, Kitasato University School of Medicine, Sagamihara, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Mine Y, Hayashi T, Yamada M, Okano H, Kawase T. ENVIRONMENTAL CUE-DEPENDENT DOPAMINERGIC NEURONAL DIFFERENTIATION AND FUNCTIONAL EFFECT OF GRAFTED NEUROEPITHELIAL STEM CELLS IN PARKINSONIAN BRAIN. Neurosurgery 2009; 65:741-53; discussion 753. [DOI: 10.1227/01.neu.0000351281.45986.76] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Yutaka Mine
- Department of Neurosurgery, School of Medicine, Keio University, Tokyo, Japan
| | - Takuro Hayashi
- Department of Neurosurgery, Eiju General Hospital, Tokyo, Japan
| | - Motoyuki Yamada
- Department of Neurosurgery, College of Medicine, University of South Florida, Tampa, Florida
| | - Hideyuki Okano
- Department of Neurosurgery, Tokyo-Kita Social Insurance Hospital, Tokyo, Japan
| | - Takeshi Kawase
- Department of Physiology, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
42
|
Wijeyekoon R, Barker RA. Cell replacement therapy for Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2009; 1792:688-702. [DOI: 10.1016/j.bbadis.2008.10.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 10/12/2008] [Accepted: 10/13/2008] [Indexed: 12/21/2022]
|
43
|
Hamanoue M, Matsuzaki Y, Sato KI, Okano HJ, Shibata S, Sato I, Suzuki S, Ogawara M, Takamatsu K, Okano H. Cell surface N-glycans mediated isolation of mouse neural stem cells. J Neurochem 2009; 110:1575-84. [PMID: 19573022 DOI: 10.1111/j.1471-4159.2009.06256.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The isolation of neural stem cells (NSCs) from the brain has been hampered by the lack of valid cell surface markers and the requirement for long-term in vitro cultivation that may lead to phenotype deterioration. However, few suitable specific cell surface antigens are available on NSCs that could be used for their prospective isolation. The present study demonstrated that the expression of complex type asparagine-linked oligosaccharide (N-glycans) was detected on brain cells dissociated from embryonic and adult brain using Phaseolus vulgaris erythroagglutinating lectin (E-PHA) which binds to biantennary complex type N-glycans, and demonstrated that E-PHA bound preferentially to purified NSCs, but not to neurons, microglia, or oligodendrocyte precursor cells. The labeling of dissociated mouse embryonic brain cells or adult brain cells with E-PHA enabled the enrichment of NSCs by 25-fold or 9-fold of the number of neurosphere-forming cells in comparison to that of unsorted cells, respectively. Furthermore, a lectin blot analysis revealed the presence of several glycoproteins which were recognized by E-PHA in the membrane fraction of the proliferating NSCs, but not in the differentiated cells. These results indicate that complex type N-glycans is a valuable cell surface marker for living mouse NSCs from both the embryonic and adult brain.
Collapse
Affiliation(s)
- Makoto Hamanoue
- Department of Physiology, Toho University School of Medicine, 5-21-16 Ohmori-nishi, Ohta-ku, Tokyo 143-8540, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Bosio A, Huppert V, Donath S, Hennemann P, Malchow M, Heinlein UAO. Isolation and enrichment of stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2009; 114:23-72. [PMID: 19347268 DOI: 10.1007/10_2008_38] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Stem cells have the potential to revolutionize tissue regeneration and engineering. Both general types of stem cells, those with pluripotent differentiation potential as well as those with multipotent differentiation potential, are of equal interest. They are important tools to further understanding of general cellular processes, to refine industrial applications for drug target discovery and predictive toxicology, and to gain more insights into their potential for tissue regeneration. This chapter provides an overview of existing sorting technologies and protocols, outlines the phenotypic characteristics of a number of different stem cells, and summarizes their potential clinical applications.
Collapse
Affiliation(s)
- Andreas Bosio
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429, Bergisch Gladbach, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Lectin panning method: the prospective isolation of mouse neural progenitor cells by the attachment of cell surface N-glycans to Phaseolus vulgaris erythroagglutinating lectin-coated dishes. Neuroscience 2008; 157:762-71. [PMID: 18952155 DOI: 10.1016/j.neuroscience.2008.09.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 09/24/2008] [Accepted: 09/29/2008] [Indexed: 02/05/2023]
Abstract
Retrospective isolation of neural progenitor cells (NPCs) may cause deterioration of the phenotype during the long-term cultivation. Therefore, prospective isolation is essential for understanding the exact characteristics of intact NPCs in the brain. However, few suitable specific cell surface antigens on NPCs that could be used for their prospective isolation are available. The present study demonstrated that within 60 min after initial plating, embryonic day 12 (E12) brain cells firmly attach to several types of lectin-coated culture wells, including Phaseolus vulgaris erythroagglutinating lectin (E-PHA), concanavalin A (Con A) and wheat germ agglutinin (WGA). Approximately 80% of the cells isolated from E-PHA-coated wells expressed the nestin antigen, which is a specific intracellular marker for NPCs and the ratio of 5-bromo-2'-deoxyuridine (BrdU)-positive/nestin-positive cells to the cells attached on the E-PHA-coated wells was significantly higher than that of the cells attached on the wells coated with other adhesive substrates. The cells that were isolated from the E-PHA-coated wells continued to attach to the well for 1 week, while those isolated from Con A- and WGA-coated wells lost their attachment after 6 days and 1 day, respectively. Furthermore, the cells isolated from the E-PHA-coated wells grew quite satisfactorily and formed numerous attached neurospheres. Their growth rate was almost equal to that observed in suspension cultures. These results indicate that the lectin panning method enables the prospective, quick and easy isolation of mouse NPCs without requiring a fluorescence-activated cell sorter (FACS) system.
Collapse
|
46
|
Maciaczyk J, Singec I, Maciaczyk D, Nikkhah G. Combined use of BDNF, ascorbic acid, low oxygen, and prolonged differentiation time generates tyrosine hydroxylase-expressing neurons after long-term in vitro expansion of human fetal midbrain precursor cells. Exp Neurol 2008; 213:354-62. [DOI: 10.1016/j.expneurol.2008.06.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Revised: 06/11/2008] [Accepted: 06/17/2008] [Indexed: 02/05/2023]
|
47
|
Okano H, Sawamoto K. Neural stem cells: involvement in adult neurogenesis and CNS repair. Philos Trans R Soc Lond B Biol Sci 2008; 363:2111-22. [PMID: 18339601 DOI: 10.1098/rstb.2008.2264] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Recent advances in stem cell research, including the selective expansion of neural stem cells (NSCs) in vitro, the induction of particular neural cells from embryonic stem cells in vitro, the identification of NSCs or NSC-like cells in the adult brain and the detection of neurogenesis in the adult brain (adult neurogenesis), have laid the groundwork for the development of novel therapies aimed at inducing regeneration in the damaged central nervous system (CNS). There are two major strategies for inducing regeneration in the damaged CNS: (i) activation of the endogenous regenerative capacity and (ii) cell transplantation therapy. In this review, we summarize the recent findings from our group and others on NSCs, with respect to their role in insult-induced neurogenesis (activation of adult NSCs, proliferation of transit-amplifying cells, migration of neuroblasts and survival and maturation of the newborn neurons), and implications for therapeutic interventions, together with tactics for using cell transplantation therapy to treat the damaged CNS.
Collapse
Affiliation(s)
- Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | | |
Collapse
|
48
|
Darabi R, Santos FNC, Perlingeiro RCR. The Therapeutic Potential of Embryonic and Adult Stem Cells for Skeletal Muscle Regeneration. ACTA ACUST UNITED AC 2008; 4:217-25. [DOI: 10.1007/s12015-008-9023-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2008] [Indexed: 11/28/2022]
|
49
|
Zhang H, Wang Y, Zhao Y, Yin Y, Xu Q, Xu Q. Immortalized human neural progenitor cells from the ventral telencephalon with the potential to differentiate into GABAergic neurons. J Neurosci Res 2008; 86:1217-26. [PMID: 18189314 DOI: 10.1002/jnr.21581] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Human neural progenitor cells (hNPCs) are believed to have important potential in clinical applications and basic neuroscience research. In the present study, we created a new immortalized human neural cell line, hSN12W-TERT, derived from human fetal ventral telencephalon, using IRES-based retroviral overexpression of human telomerase reverse transcriptase. We showed that after more than 40 passages, hSN12W-TERT cells possess high telomerase activity, maintain a normal diploid karyotype, and retain the characteristics of hNPCs. Under proliferative conditions, these cells remained undifferentiated, expressing the neural progenitor cell markers nestin, vimentin, and Sox2. The cells were able to differentiate into neurons, astrocytes, and oligodendrocytes after a significant decrease in the level of telomerase following withdrawal of growth factors. The neurons were postmitotic and achieved electrophysiologic competence. Furthermore, we showed that most neurons were GABAergic, especially on differentiation induced by bone morphogenetic protein-2 (BMP2). RT-PCR analysis also confirmed that hSN12W-TERT cells expressed mammalian achaete-scute homolog 1 (Mash1) and Dlx2, genes associated with the development of GABAergic cortical interneurons. BMP2 exposure may activate a positive-feedback loop of BMP signaling in hSN12W-TERT cells. Our data indicated that this hSN12W-TERT cell line could be a valuable experimental tool with which to study the regulatory roles of intrinsic and extrinsic factors in human neural stem cell biology and that it would be useful in basic research and in research seeking to discover novel drug targets for clinical candidates.
Collapse
Affiliation(s)
- Haiyan Zhang
- Department of Cell Biology, Capital Medical University, Beijing, China
| | | | | | | | | | | |
Collapse
|
50
|
Hassan HT, Zhai X, Goodacre JA. CD133 stem cells in adult human brain. J Neurooncol 2008; 89:247-8; author reply 249. [DOI: 10.1007/s11060-008-9620-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Accepted: 05/28/2008] [Indexed: 11/24/2022]
|