1
|
Potdar C, Jagtap S, Singh K, Yadav R, Pal PK, Datta I. Impaired Sonic Hedgehog Responsiveness of Induced Pluripotent Stem Cell-Derived Floor Plate Cells Carrying the LRRK2-I1371V Mutation Contributes to the Ontogenic Origin of Lower Dopaminergic Neuron Yield. Stem Cells Dev 2024; 33:306-320. [PMID: 38753688 DOI: 10.1089/scd.2023.0283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Abstract
Lower population of dopaminergic (DA) neurons is known to increase susceptibility to Parkinson's disease (PD), and our earlier study showed a lower yield of DA neurons in Leucine-Rich Repeat Kinase Isoleucine 1371 Valine (LRRK2-I1371V) mutation-carrying PD patient-derived induced Pluripotent Stem Cells (iPSCs). Although the role of Sonic Hedgehog (SHH) in DA neurogenesis of floor plate cells (FPCs) is known, the effect of LRRK2 mutations on SHH responsiveness of FPCs impacting DA neuronal yield has not been studied. We investigated SHH responsiveness of FPCs derived from LRRK2-I1371V PD patient iPSCs with regard to the expression of SHH receptors Patched1 (Ptch1) and Smoothened (Smo), in conjunction with nuclear Gli1 (glioma-associated oncogene 1) expression, intracellular Ca2+ rise, and cytosolic cyclic adenosine monophosphate (cAMP) levels upon SHH induction. In addition, we examined the mechanistic link with LRRK2-I1371V gain-of-function by assessing membrane fluidity and Rab8A and Rab10 phosphorylation in SH-SY5Y cells and healthy control (HC) FPCs overexpressing LRRK2-I1371V as well as FPCs. Although total expression of Ptch1 and Smo was comparable, receptor expression on cell surface was significantly lower in LRRK2-I1371V FPCs than in HC FPCs, with distinctly lower nuclear expression of the downstream transcription factor Gli1. HC-FPCs transfected with LRRK2-I1371V exhibited a similarly reduced cell surface expression of Ptch1 and Smo. Intracellular Ca2+ response was significantly lower with corresponding elevated cAMP levels in LRRK2-I1371V FPCs compared with HC FPCs upon SHH stimulation. The LRRK2-I1371V mutant FPCs and LRRK2-I1371V-transfected SH-SY5Y and HC FPCs too exhibited higher autophosphorylation of phospho LRRK2 (pLRRK2) serine1292 and serine935, as well as substrate phosphorylation of Rab8A and Rab10. Concurrent increase in membrane fluidity, accompanied by a decrease in membrane cholesterol, and lower expression of lipid raft marker caveolin 1 were also observed in them. These findings suggest that impaired SHH responsiveness of LRRK2-I1371V PD FPCs indeed leads to lower yield of DA neurons during ontogeny. Reduced cell surface expression of SHH receptors is influenced by alteration in membrane fluidity owing to the increased substrate phosphorylation of Rab8A and reduced membrane protein trafficking due to pRab10, both results of the LRRK2-I1371V mutation.
Collapse
Affiliation(s)
- Chandrakanta Potdar
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Soham Jagtap
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Khushboo Singh
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| |
Collapse
|
2
|
Mohd Rafiq N, Fujise K, Rosenfeld MS, Xu P, De Camilli P. Parkinsonism Sac domain mutation in Synaptojanin-1 affects ciliary properties in iPSC-derived dopaminergic neurons. Proc Natl Acad Sci U S A 2024; 121:e2318943121. [PMID: 38635628 PMCID: PMC11047088 DOI: 10.1073/pnas.2318943121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/04/2024] [Indexed: 04/20/2024] Open
Abstract
Synaptojanin-1 (SJ1) is a major neuronal-enriched PI(4, 5)P2 4- and 5-phosphatase implicated in the shedding of endocytic factors during endocytosis. A mutation (R258Q) that impairs selectively its 4-phosphatase activity causes Parkinsonism in humans and neurological defects in mice (SJ1RQKI mice). Studies of these mice showed, besides an abnormal assembly state of endocytic factors at synapses, the presence of dystrophic nerve terminals selectively in a subset of nigro-striatal dopamine (DA)-ergic axons, suggesting a special lability of DA neurons to the impairment of SJ1 function. Here we have further investigated the impact of SJ1 on DA neurons using iPSC-derived SJ1 KO and SJ1RQKI DA neurons and their isogenic controls. In addition to the expected enhanced clustering of endocytic factors in nerve terminals, we observed in both SJ1 mutant neuronal lines increased cilia length. Further analysis of cilia of SJ1RQDA neurons revealed abnormal accumulation of the Ca2+ channel Cav1.3 and of ubiquitin chains, suggesting a defect in the clearing of ubiquitinated proteins at the ciliary base, where a focal concentration of SJ1 was observed. We suggest that SJ1 may contribute to the control of ciliary protein dynamics in DA neurons, with implications on cilia-mediated signaling.
Collapse
Affiliation(s)
- Nisha Mohd Rafiq
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Department of Cell biology, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Kenshiro Fujise
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Department of Cell biology, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Martin Shaun Rosenfeld
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Department of Cell biology, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Peng Xu
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Department of Cell biology, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Department of Cell biology, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| |
Collapse
|
3
|
Baddenhausen S, Lutz B, Hofmann C. Cannabinoid type-1 receptor signaling in dopaminergic Engrailed-1 expressing neurons modulates motivation and depressive-like behavior. Front Mol Neurosci 2024; 17:1379889. [PMID: 38660383 PMCID: PMC11042029 DOI: 10.3389/fnmol.2024.1379889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024] Open
Abstract
The endocannabinoid system comprises highly versatile signaling functions within the nervous system. It is reported to modulate the release of several neurotransmitters, consequently affecting the activity of neuronal circuits. Investigations have highlighted its roles in numerous processes, including appetite-stimulating characteristics, particularly for palatable food. Moreover, endocannabinoids are shown to fine-tune dopamine-signaled processes governing motivated behavior. Specifically, it has been demonstrated that excitatory and inhibitory inputs controlled by the cannabinoid type 1 receptor (CB1) regulate dopaminergic neurons in the mesocorticolimbic pathway. In the present study, we show that mesencephalic dopaminergic (mesDA) neurons in the ventral tegmental area (VTA) express CB1, and we investigated the consequences of specific deletion of CB1 in cells expressing the transcription factor Engrailed-1 (En1). To this end, we validated a new genetic mouse line EN1-CB1-KO, which displays a CB1 knockout in mesDA neurons beginning from their differentiation, as a tool to elucidate the functional contribution of CB1 in mesDA neurons. We revealed that EN1-CB1-KO mice display a significantly increased immobility time and shortened latency to the first immobility in the forced swim test of adult mice. Moreover, the maximal effort exerted to obtain access to chocolate-flavored pellets was significantly reduced under a progressive ratio schedule. In contrast, these mice do not differ in motor skills, anhedonia- or anxiety-like behavior compared to wild-type littermates. Taken together, these findings suggest a depressive-like or despair behavior in an inevitable situation and a lack of motivation to seek palatable food in EN1-CB1-KO mice, leading us to propose that CB1 plays an important role in the physiological functions of mesDA neurons. In particular, our data suggest that CB1 directly modifies the mesocorticolimbic pathway implicated in depressive-like/despair behavior and motivation. In contrast, the nigrostriatal pathway controlling voluntary movement seems to be unaffected.
Collapse
Affiliation(s)
- Sarah Baddenhausen
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
| | - Clementine Hofmann
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
4
|
Sasai N, Tada S, Ohshiro J, Kogiso C, Shinozuka T. Regulation of progenitor cell survival by a novel chromatin remodeling factor during neural tube development. Dev Growth Differ 2024; 66:89-100. [PMID: 38014908 DOI: 10.1111/dgd.12905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
During development, progenitor cell survival is essential for proper tissue functions, but the underlying mechanisms are not fully understood. Here we show that ERCC6L2, a member of the Snf2 family of helicase-like proteins, plays an essential role in the survival of developing chick neural cells. ERCC6L2 expression is induced by the Sonic Hedgehog (Shh) signaling molecule by a mechanism similar to that of the known Shh target genes Ptch1 and Gli1. ERCC6L2 blocks programmed cell death induced by Shh inhibition and this inhibition is independent of neural tube patterning. ERCC6L2 knockdown by siRNA resulted in the aberrant appearance of apoptotic cells. Furthermore, ERCC6L2 cooperates with the Shh signal and plays an essential role in the induction of the anti-apoptotic factor Bcl-2. Taken together, ERCC6L2 acts as a key factor in ensuring the survival of neural progenitor cells.
Collapse
Affiliation(s)
- Noriaki Sasai
- Developmental Biomedical Science, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Shogo Tada
- Developmental Biomedical Science, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Jumi Ohshiro
- Developmental Biomedical Science, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Chikara Kogiso
- Developmental Biomedical Science, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Takuma Shinozuka
- Developmental Biomedical Science, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| |
Collapse
|
5
|
Maimaitili M, Chen M, Febbraro F, Ucuncu E, Kelly R, Niclis JC, Christiansen JR, Mermet-Joret N, Niculescu D, Lauritsen J, Iannielli A, Klæstrup IH, Jensen UB, Qvist P, Nabavi S, Broccoli V, Nykjær A, Romero-Ramos M, Denham M. Enhanced production of mesencephalic dopaminergic neurons from lineage-restricted human undifferentiated stem cells. Nat Commun 2023; 14:7871. [PMID: 38052784 DOI: 10.1038/s41467-023-43471-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 11/10/2023] [Indexed: 12/07/2023] Open
Abstract
Current differentiation protocols for generating mesencephalic dopaminergic (mesDA) neurons from human pluripotent stem cells result in grafts containing only a small proportion of mesDA neurons when transplanted in vivo. In this study, we develop lineage-restricted undifferentiated stem cells (LR-USCs) from pluripotent stem cells, which enhances their potential for differentiating into caudal midbrain floor plate progenitors and mesDA neurons. Using a ventral midbrain protocol, 69% of LR-USCs become bona fide caudal midbrain floor plate progenitors, compared to only 25% of human embryonic stem cells (hESCs). Importantly, LR-USCs generate significantly more mesDA neurons under midbrain and hindbrain conditions in vitro and in vivo. We demonstrate that midbrain-patterned LR-USC progenitors transplanted into 6-hydroxydopamine-lesioned rats restore function in a clinically relevant non-pharmacological behavioral test, whereas midbrain-patterned hESC-derived progenitors do not. This strategy demonstrates how lineage restriction can prevent the development of undesirable lineages and enhance the conditions necessary for mesDA neuron generation.
Collapse
Affiliation(s)
- Muyesier Maimaitili
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
| | - Muwan Chen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
| | - Fabia Febbraro
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Clinical Genetics, Aarhus University Hospital, 8200, Aarhus, Denmark
| | - Ekin Ucuncu
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
| | - Rachel Kelly
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
| | | | | | - Noëmie Mermet-Joret
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, 8000C, Aarhus, Denmark
- Center of Excellence PROMEMO, Aarhus University, Aarhus, Denmark
| | - Dragos Niculescu
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
- Center of Excellence PROMEMO, Aarhus University, Aarhus, Denmark
| | - Johanne Lauritsen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
| | - Angelo Iannielli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- CNR Institute of Neuroscience, 20129, Milan, Italy
| | - Ida H Klæstrup
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
| | - Uffe Birk Jensen
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Clinical Genetics, Aarhus University Hospital, 8200, Aarhus, Denmark
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
- Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8000C, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, 8000C, Aarhus, Denmark
- Centre for Genomics and Personalized Medicine, CGPM, Aarhus University, 8000C, Aarhus, Denmark
| | - Sadegh Nabavi
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, 8000C, Aarhus, Denmark
- Center of Excellence PROMEMO, Aarhus University, Aarhus, Denmark
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
- CNR Institute of Neuroscience, 20129, Milan, Italy
| | - Anders Nykjær
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
- Center of Excellence PROMEMO, Aarhus University, Aarhus, Denmark
| | - Marina Romero-Ramos
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark
| | - Mark Denham
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, 8000C, Aarhus, Denmark.
- Department of Biomedicine, Aarhus University, 8000C, Aarhus, Denmark.
| |
Collapse
|
6
|
Rafiq NM, Fujise K, Rosenfeld MS, Xu P, Wu Y, De Camilli P. Parkinsonism Sac domain mutation in Synaptojanin-1 affects ciliary properties in iPSC-derived dopaminergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.12.562142. [PMID: 37873399 PMCID: PMC10592818 DOI: 10.1101/2023.10.12.562142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Synaptojanin-1 (SJ1) is a major neuronal-enriched PI(4,5)P2 4- and 5-phosphatase implicated in the shedding of endocytic factors during endocytosis. A mutation (R258Q) that impairs selectively its 4-phosphatase activity causes Parkinsonism in humans and neurological defects in mice (SJ1RQKI mice). Studies of these mice showed, besides an abnormal assembly state of endocytic factors at synapses, the presence of dystrophic nerve terminals selectively in a subset of nigro-striatal dopamine (DA)-ergic axons, suggesting a special lability of DA neurons to the impairment of SJ1 function. Here we have further investigated the impact of SJ1 on DA neurons using iPSC-derived SJ1 KO and SJ1RQKI DA neurons and their isogenic controls. In addition to the expected enhanced clustering of endocytic factors in nerve terminals, we observed in both SJ1 mutant neuronal lines increased cilia length. Further analysis of cilia of SJ1RQDA neurons revealed abnormal accumulation of the Ca2+ channel Cav1.3 and of ubiquitin chains, suggesting an impaired clearing of proteins from cilia which may result from an endocytic defect at the ciliary base, where a focal concentration of SJ1 was observed. We suggest that SJ1 may contribute to the control of ciliary protein dynamics in DA neurons, with implications on cilia-mediated signaling.
Collapse
Affiliation(s)
- Nisha Mohd Rafiq
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair. Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Kenshiro Fujise
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair. Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Martin Shaun Rosenfeld
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair. Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Peng Xu
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair. Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Yumei Wu
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair. Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair. Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
7
|
Mendoza-Torreblanca JG, Cárdenas-Rodríguez N, Carro-Rodríguez J, Contreras-García IJ, Garciadiego-Cázares D, Ortega-Cuellar D, Martínez-López V, Alfaro-Rodríguez A, Evia-Ramírez AN, Ignacio-Mejía I, Vargas-Hernández MA, Bandala C. Antiangiogenic Effect of Dopamine and Dopaminergic Agonists as an Adjuvant Therapeutic Option in the Treatment of Cancer, Endometriosis, and Osteoarthritis. Int J Mol Sci 2023; 24:10199. [PMID: 37373348 DOI: 10.3390/ijms241210199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Dopamine (DA) and dopamine agonists (DA-Ag) have shown antiangiogenic potential through the vascular endothelial growth factor (VEGF) pathway. They inhibit VEGF and VEGF receptor 2 (VEGFR 2) functions through the dopamine receptor D2 (D2R), preventing important angiogenesis-related processes such as proliferation, migration, and vascular permeability. However, few studies have demonstrated the antiangiogenic mechanism and efficacy of DA and DA-Ag in diseases such as cancer, endometriosis, and osteoarthritis (OA). Therefore, the objective of this review was to describe the mechanisms of the antiangiogenic action of the DA-D2R/VEGF-VEGFR 2 system and to compile related findings from experimental studies and clinical trials on cancer, endometriosis, and OA. Advanced searches were performed in PubMed, Web of Science, SciFinder, ProQuest, EBSCO, Scopus, Science Direct, Google Scholar, PubChem, NCBI Bookshelf, DrugBank, livertox, and Clinical Trials. Articles explaining the antiangiogenic effect of DA and DA-Ag in research articles, meta-analyses, books, reviews, databases, and clinical trials were considered. DA and DA-Ag have an antiangiogenic effect that could reinforce the treatment of diseases that do not yet have a fully curative treatment, such as cancer, endometriosis, and OA. In addition, DA and DA-Ag could present advantages over other angiogenic inhibitors, such as monoclonal antibodies.
Collapse
Affiliation(s)
| | - Noemi Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | - Jazmín Carro-Rodríguez
- Laboratorio de Medicina Traslacional Aplicada a Neurociencias, Enfermedades Crónicas y Emergentes, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Itzel Jatziri Contreras-García
- Laboratorio de Biología de la Reproducción, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | - David Garciadiego-Cázares
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico
| | - Daniel Ortega-Cuellar
- Laboratorio Nutrición Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Valentín Martínez-López
- Unidad de Ingeniería de Tejidos, Terapia Celular y Medicina Regenerativa, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico
| | - Alfonso Alfaro-Rodríguez
- Neurociencias Básicas, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Mexico City 14389, Mexico
| | - Alberto Nayib Evia-Ramírez
- Servicio de Reconstrucción Articular, Cadera y Rodilla, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico
| | - Iván Ignacio-Mejía
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, Mexico City 11200, Mexico
| | | | - Cindy Bandala
- Laboratorio de Medicina Traslacional Aplicada a Neurociencias, Enfermedades Crónicas y Emergentes, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| |
Collapse
|
8
|
Almeida AS, Nunes F, Marques DM, Machado ACL, Oliveira CB, Porciuncula LO. Sex differences in maternal odor preferences and brain levels of GAP-43 and sonic hedgehog proteins in infant SHR and Wistar Kyoto rats. Behav Brain Res 2023; 436:114102. [DOI: 10.1016/j.bbr.2022.114102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022]
|
9
|
Gupta R, Mehan S, Chhabra S, Giri A, Sherawat K. Role of Sonic Hedgehog Signaling Activation in the Prevention of Neurological Abnormalities Associated with Obsessive-Compulsive Disorder. Neurotox Res 2022; 40:1718-1738. [PMID: 36272053 DOI: 10.1007/s12640-022-00586-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 09/15/2022] [Accepted: 10/07/2022] [Indexed: 12/31/2022]
Abstract
The smoothened sonic hedgehog (Smo-Shh) pathway is one mechanism that influences neurogenesis, including brain cell differentiation and development during childhood. Shh signaling dysregulation leads to decreased target gene transcription, which contributes to increased neuronal excitation, apoptosis, and neurodegeneration, eventually leading to neurological deficits. Neuropsychiatric disorders such as OCD and related neurological dysfunctions are characterized by neurotransmitter imbalance, neuroinflammation, oxidative stress, and impaired neurogenesis, disturbing the cortico-striato-thalamo-cortical (CSTC) link neuronal network. Despite the availability of several treatments, such as selective serotonin reuptake inhibitors, some individuals may not benefit much from them. Several trials on the use of antipsychotics in the treatment of OCD have also produced inadequate findings. This evidence-based review focuses on a potential pharmacological approach to alleviating OCD and associated neuronal deficits by preventing neurochemical alterations, in which sonic hedgehog activators are neuroprotective, lowering neuronal damage while increasing neuronal maintenance and survival. As a result, stimulating SMO-Shh via its potential activators may have neuroprotective effects on neurological impairment associated with OCD. This review investigates the link between SMO-Shh signaling and the neurochemical abnormalities associated with the progression of OCD and associated neurological dysfunctions. Role of Smo-Shh signaling in serotonergic neurogenesis and in maintaining their neuronal identity. The Shh ligand activates two main transcriptional factors known as Foxa2 and Nkx2.2, which again activates another transcriptional factor, GATA (GATA2 and GATA3), in post mitotic precursor cells of serotonergic neurons-following increased expression of Pet-1 and Lmx1b after GATA regulates the expression of many serotonergic enzymes such as TPH2, SERT, VMAT, slc6a4, Htr1a, Htr1b (Serotonin receptor enzymes), and MAO that regulate and control the release of serotonin and maintain their neuronal identity after their maturation. Abbreviation: Foxa2: Forkhead box; GATA: Globin transcription factor; Lmx1b: LIM homeobox transcription factor 1 beta; TPH2: Tryptophan hydroxylase 2; Htr1a: Serotonin receptor 1a; Htr1b: Serotonin receptor 1b; SERT: Serotonin transporter; VMAT: Vesicular monoamine transporter; MAO: Monoamine oxidase.
Collapse
Affiliation(s)
- Ria Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| | - Swesha Chhabra
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Aditi Giri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Kajal Sherawat
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| |
Collapse
|
10
|
Turcato FC, Wegman E, Lu T, Ferguson N, Luo Y. Dopaminergic neurons are not a major Sonic hedgehog ligand source for striatal cholinergic or PV interneurons. iScience 2022; 25:105278. [PMID: 36281454 PMCID: PMC9587326 DOI: 10.1016/j.isci.2022.105278] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/05/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
A model was previously proposed that DA neurons provide SHH ligand to striatal interneurons, which in turn support the survival of DA neurons through the release of trophic factors such as Glial cell-derived neurotrophic factor (GDNF). However, some key clinical observations do not support this proposed model, and a recent independent study shows that striatal cholinergic neuron survival does not rely on intact DA neuron projections. To resolve this discrepancy, we generated several independent mouse lines to examine the exact role of DA neuron-derived Shh signaling in the maintenance of the basal ganglia circuit and to identify the Shh-producing cells in the adult brain. Our data suggest that the deletion of Shh in DA neurons does not affect DA neuron survival or locomotive function in cKO mice during aging, nor does it affect the long-term survival of cholinergic or FS PV + interneurons in the striatum (STR).
Collapse
Affiliation(s)
- Flavia Correa Turcato
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Elliot Wegman
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Tao Lu
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Nathan Ferguson
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yu Luo
- Department of Molecular Genetics, Biochemistry and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
11
|
Chalazonitis A, Rao M, Sulzer D. Similarities and differences between nigral and enteric dopaminergic neurons unravel distinctive involvement in Parkinson's disease. NPJ Parkinsons Dis 2022; 8:50. [PMID: 35459867 PMCID: PMC9033791 DOI: 10.1038/s41531-022-00308-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 03/14/2022] [Indexed: 11/09/2022] Open
Abstract
In addition to the well-known degeneration of midbrain dopaminergic neurons, enteric neurons can also be affected in neurodegenerative disorders such as Parkinson's disease (PD). Dopaminergic neurons have recently been identified in the enteric nervous system (ENS). While ENS dopaminergic neurons have been shown to degenerate in genetic mouse models of PD, analyses of their survival in enteric biopsies of PD patients have provided inconsistent results to date. In this context, this review seeks to highlight the distinctive and shared factors and properties that control the evolution of these two sets of dopaminergic neurons from neuronal precursors to aging neurons. Although their cellular sources and developmental times of origin differ, midbrain and ENS dopaminergic neurons express many transcription factors in common and their respective environments express similar neurotrophic molecules. For example, Foxa2 and Sox6 are expressed by both populations to promote the specification, differentiation, and long-term maintenance of the dopaminergic phenotype. Both populations exhibit sustained patterns of excitability that drive intrinsic vulnerability over time. In disorders such as PD, colon biopsies have revealed aggregation of alpha-synuclein in the submucosal plexus where dopaminergic neurons reside and lack blood barrier protection. Thus, these enteric neurons may be more susceptible to neurotoxic insults and aggregation of α-synuclein that spreads from gut to midbrain. Under sustained stress, inefficient autophagy leads to neurodegeneration, GI motility dysfunction, and PD symptoms. Recent findings suggest that novel neurotrophic factors such as CDNF have the potential to be used as neuroprotective agents to prevent and treat ENS symptoms of PD.
Collapse
Affiliation(s)
- Alcmène Chalazonitis
- Department of Pathology & Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| | - Meenakshi Rao
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Sulzer
- Departments of Psychiatry, Neurology, and Pharmacology, Division of Molecular Therapeutics, New York State Psychiatry Institute, Columbia University, New York, NY, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
12
|
Ma R, Kutchy NA, Chen L, Meigs DD, Hu G. Primary cilia and ciliary signaling pathways in aging and age-related brain disorders. Neurobiol Dis 2022; 163:105607. [PMID: 34979259 PMCID: PMC9280856 DOI: 10.1016/j.nbd.2021.105607] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 12/08/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Brain disorders are characterized by the progressive loss of structure and function of the brain as a consequence of progressive degeneration and/or death of nerve cells. Aging is a major risk factor for brain disorders such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and stroke. Various cellular and molecular events have been shown to play a role in the progress of neurodegenerative diseases. Emerging studies suggest that primary cilia could be a key regulator in brain diseases. The primary cilium is a singular cellular organelle expressed on the surface of many cell types, such as astrocytes and neurons in the mature brain. Primary cilia detect extracellular cues, such as Sonic Hedgehog (SHH) protein, and transduce these signals into cells to regulate various signaling pathways. Abnormalities in ciliary length and frequency (ratio of ciliated cells) have been implicated in various human diseases, including brain disorders. This review summarizes current findings and thoughts on the role of primary cilia and ciliary signaling pathways in aging and age-related brain disorders.
Collapse
Affiliation(s)
- Rong Ma
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Naseer A Kutchy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; Department of Anatomy, Physiology and Pharmacology, School of Veterinary Medicine, St. George's University, Grenada
| | - Liang Chen
- Department of Computer Science, College of Engineering, Shantou University, Shantou, Guangdong 515063, China; Key Laboratory of Intelligent Manufacturing Technology, Ministry of Education, Shantou University, Shantou, Guangdong 515063, China
| | - Douglas D Meigs
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA.
| |
Collapse
|
13
|
Sharma S, Jeyaraman M, Muthu S. Role of stem cell therapy in neurosciences. ESSENTIALS OF EVIDENCE-BASED PRACTICE OF NEUROANESTHESIA AND NEUROCRITICAL CARE 2022:163-179. [DOI: 10.1016/b978-0-12-821776-4.00012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
14
|
Spatiotemporal expression of sonic hedgehog signalling molecules in the embryonic mesencephalic dopaminergic neurons. Gene Expr Patterns 2021; 42:119217. [PMID: 34767969 DOI: 10.1016/j.gep.2021.119217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/09/2021] [Accepted: 11/07/2021] [Indexed: 11/22/2022]
Abstract
Midbrain dopaminergic neurons (mDA) play an important role in controlling the voluntary motor movement, reward, and emotion-based behaviour. Differentiation of mDA neurons from progenitors depends on several secreted proteins, such as sonic hedgehog (SHH). The present study attempted to elucidate the possible role(s) of some SHH signaling components (Ptch1, Gli1, Gli2 and Gli3) in the spatiotemporal development of mDA neurons along the rostrocaudal axis of the midbrain and their possible roles in differentiation and survival of mDA neurons and the significance of using in vitro models for studying the development of mDA neurons. At E12 and E14, only Ptch1 and Gli1 were expressed in ventrolateral midbrain domains. All examined SHH signalling molecules were not detected in mDA area. Whereas, in MN9D cells, many SHH signalling molecules were expressed and co-localized with the dopaminergic marker; tyrosine hydroxylase (TH), and their expression were upregulated with SHH treatment of the MN9D cells. These results suggest that mDA neurons differentiation and survival might be independent of SHH in the late developmental stages (E12-18). Besides, MN9D cell line is not the ideal in vitro model for investigating the differentiation of mDA and hence, the ventral midbrain primary culture might be favored over MN9D line.
Collapse
|
15
|
Kalyn M, Ekker M. Cerebroventricular Microinjections of MPTP on Adult Zebrafish Induces Dopaminergic Neuronal Death, Mitochondrial Fragmentation, and Sensorimotor Impairments. Front Neurosci 2021; 15:718244. [PMID: 34512252 PMCID: PMC8432913 DOI: 10.3389/fnins.2021.718244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/26/2021] [Indexed: 11/21/2022] Open
Abstract
Mitochondria are dynamic organelles that mediate the energetic supply to cells and mitigate oxidative stress through the intricate balance of fission and fusion. Mitochondrial dysfunction is a prominent feature within Parkinson disease (PD) etiologies. To date, there have been conflicting studies of neurotoxin impact on dopaminergic cell death, mitochondrial function and behavioral impairment using adult zebrafish. Here, we performed cerebroventricular microinjections (CVMIs) of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on adult transgenic zebrafish that resulted in significant reductions in dopaminergic neurons within the telencephalon and olfactory bulbs (OB) of Tg(dat:eGFP) fish. Visualization of mCherry and mitochondrial gene expression analysis in Tg(dat:tom20 MLS:mCherry) fish reveal that MPTP induces mitochondrial fragmentation in dopaminergic neurons and the activation of the pink1/parkin pathway involved mitophagy. Moreover, the loss of dopaminergic neurons translated into a transient locomotor and olfactory phenotype. Taken together, these data can contribute to a better understanding of the mitochondrial impact on dopaminergic survivability.
Collapse
Affiliation(s)
- Michael Kalyn
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON, Canada
| | - Marc Ekker
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
16
|
Cai E, Zhang J, Ge X. Control of the Hedgehog pathway by compartmentalized PKA in the primary cilium. SCIENCE CHINA-LIFE SCIENCES 2021; 65:500-514. [PMID: 34505970 DOI: 10.1007/s11427-021-1975-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/01/2021] [Indexed: 01/20/2023]
Abstract
The Hedgehog (Hh) signaling is one of the essential signaling pathways during embryogenesis and in adults. Hh signal transduction relies on primary cilium, a specialized cell surface organelle viewed as the hub of cell signaling. Protein kinase A (PKA) has been recognized as a potent negative regulator of the Hh pathway, raising the question of how such a ubiquitous kinase specifically regulates one signaling pathway. We reviewed recent genetic, molecular and biochemical studies that have advanced our mechanistic understanding of PKA's role in Hh signaling in vertebrates, focusing on the compartmentalized PKA at the centrosome and in the primary cilium. We outlined the recently developed genetic and optical tools that can be harvested to study PKA activities during the course of Hh signal transduction.
Collapse
Affiliation(s)
- Eva Cai
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, 95340, USA
| | - Jingyi Zhang
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, 95340, USA
| | - Xuecai Ge
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, 95340, USA.
| |
Collapse
|
17
|
Bell S, McCarty V, Peng H, Jefri M, Hettige N, Antonyan L, Crapper L, O'Leary LA, Zhang X, Zhang Y, Wu H, Sutcliffe D, Kolobova I, Rosenberger TA, Moquin L, Gratton A, Popic J, Gantois I, Stumpf PS, Schuppert AA, Mechawar N, Sonenberg N, Tremblay ML, Jinnah HA, Ernst C. Lesch-Nyhan disease causes impaired energy metabolism and reduced developmental potential in midbrain dopaminergic cells. Stem Cell Reports 2021; 16:1749-1762. [PMID: 34214487 PMCID: PMC8282463 DOI: 10.1016/j.stemcr.2021.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 10/30/2022] Open
Abstract
Mutations in HPRT1, a gene encoding a rate-limiting enzyme for purine salvage, cause Lesch-Nyhan disease which is characterized by self-injury and motor impairments. We leveraged stem cell and genetic engineering technologies to model the disease in isogenic and patient-derived forebrain and midbrain cell types. Dopaminergic progenitor cells deficient in HPRT showed decreased intensity of all developmental cell-fate markers measured. Metabolic analyses revealed significant loss of all purine derivatives, except hypoxanthine, and impaired glycolysis and oxidative phosphorylation. real-time glucose tracing demonstrated increased shunting to the pentose phosphate pathway for de novo purine synthesis at the expense of ATP production. Purine depletion in dopaminergic progenitor cells resulted in loss of RHEB, impairing mTORC1 activation. These data demonstrate dopaminergic-specific effects of purine salvage deficiency and unexpectedly reveal that dopaminergic progenitor cells are programmed to a high-energy state prior to higher energy demands of terminally differentiated cells.
Collapse
Affiliation(s)
- Scott Bell
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Vincent McCarty
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Huashan Peng
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Malvin Jefri
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Nuwan Hettige
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Lilit Antonyan
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Liam Crapper
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Liam A O'Leary
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Xin Zhang
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Ying Zhang
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Hanrong Wu
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Diane Sutcliffe
- Department of Neurology, Emory University, Atlanta, GA, USA; Department of Human Genetics, Emory University, Atlanta, GA, USA; Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Ilaria Kolobova
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Thad A Rosenberger
- Department of Pharmacology, Physiology, and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Luc Moquin
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Alain Gratton
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Jelena Popic
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Ilse Gantois
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Patrick S Stumpf
- Joint Research Center for Computational Biomedicine, RWTH Aachen University, Aachen, Germany
| | - Andreas A Schuppert
- Joint Research Center for Computational Biomedicine, RWTH Aachen University, Aachen, Germany
| | - Naguib Mechawar
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Michel L Tremblay
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Hyder A Jinnah
- Department of Neurology, Emory University, Atlanta, GA, USA; Department of Human Genetics, Emory University, Atlanta, GA, USA; Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Carl Ernst
- Psychiatric Genetics Group, McGill University, Montreal, QC, Canada; Department of Psychiatry, McGill University and Douglas Hospital Research Institute, 6875 LaSalle Boulevard, Frank Common Building, Room 2101.2, Montreal, QC H4H 1R3, Canada.
| |
Collapse
|
18
|
Brady MV, Vaccarino FM. Role of SHH in Patterning Human Pluripotent Cells towards Ventral Forebrain Fates. Cells 2021; 10:cells10040914. [PMID: 33923415 PMCID: PMC8073580 DOI: 10.3390/cells10040914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 12/03/2022] Open
Abstract
The complexities of human neurodevelopment have historically been challenging to decipher but continue to be of great interest in the contexts of healthy neurobiology and disease. The classic animal models and monolayer in vitro systems have limited the types of questions scientists can strive to answer in addition to the technical ability to answer them. However, the tridimensional human stem cell-derived organoid system provides the unique opportunity to model human development and mimic the diverse cellular composition of human organs. This strategy is adaptable and malleable, and these neural organoids possess the morphogenic sensitivity to be patterned in various ways to generate the different regions of the human brain. Furthermore, recapitulating human development provides a platform for disease modeling. One master regulator of human neurodevelopment in many regions of the human brain is sonic hedgehog (SHH), whose expression gradient and pathway activation are responsible for conferring ventral identity and shaping cellular phenotypes throughout the neural axis. This review first discusses the benefits, challenges, and limitations of using organoids for studying human neurodevelopment and disease, comparing advantages and disadvantages with other in vivo and in vitro model systems. Next, we explore the range of control that SHH exhibits on human neurodevelopment, and the application of SHH to various stem cell methodologies, including organoids, to expand our understanding of human development and disease. We outline how this strategy will eventually bring us much closer to uncovering the intricacies of human neurodevelopment and biology.
Collapse
Affiliation(s)
| | - Flora M. Vaccarino
- Child Study Center, Yale University, New Haven, CT 06520, USA;
- Department of Neuroscience, Yale University, New Haven, CT 06520, USA
- Yale Kavli Institute for Neuroscience, New Haven, CT 06520, USA
- Correspondence:
| |
Collapse
|
19
|
Diaz C, Puelles L. Developmental Genes and Malformations in the Hypothalamus. Front Neuroanat 2020; 14:607111. [PMID: 33324176 PMCID: PMC7726113 DOI: 10.3389/fnana.2020.607111] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
The hypothalamus is a heterogeneous rostral forebrain region that regulates physiological processes essential for survival, energy metabolism, and reproduction, mainly mediated by the pituitary gland. In the updated prosomeric model, the hypothalamus represents the rostralmost forebrain, composed of two segmental regions (terminal and peduncular hypothalamus), which extend respectively into the non-evaginated preoptic telencephalon and the evaginated pallio-subpallial telencephalon. Complex genetic cascades of transcription factors and signaling molecules rule their development. Alterations of some of these molecular mechanisms acting during forebrain development are associated with more or less severe hypothalamic and pituitary dysfunctions, which may be associated with brain malformations such as holoprosencephaly or septo-optic dysplasia. Studies on transgenic mice with mutated genes encoding critical transcription factors implicated in hypothalamic-pituitary development are contributing to understanding the high clinical complexity of these pathologies. In this review article, we will analyze first the complex molecular genoarchitecture of the hypothalamus resulting from the activity of previous morphogenetic signaling centers and secondly some malformations related to alterations in genes implicated in the development of the hypothalamus.
Collapse
Affiliation(s)
- Carmen Diaz
- Department of Medical Sciences, School of Medicine and Institute for Research in Neurological Disabilities, University of Castilla-La Mancha, Albacete, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| |
Collapse
|
20
|
Rahi S, Mehan S. Understanding Abnormal SMO-SHH Signaling in Autism Spectrum Disorder: Potential Drug Target and Therapeutic Goals. Cell Mol Neurobiol 2020; 42:931-953. [PMID: 33206287 DOI: 10.1007/s10571-020-01010-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Autism is a multifactorial neurodevelopmental condition; it demonstrates some main characteristics, such as impaired social relationships and increased repetitive behavior. The initiation of autism spectrum disorder is mostly triggered during brain development by the deregulation of signaling pathways. Sonic hedgehog (SHH) signaling is one such mechanism that influences neurogenesis and neural processes during the development of the central nervous system. SMO-SHH signaling is also an important part of a broad variety of neurological processes, including neuronal cell differentiation, proliferation, and survival. Dysregulation of SMO-SHH signaling leads to many physiological changes that lead to neurological disorders such as ASD and contribute to cognitive decline. The aberrant downregulation of SMO-SHH signals contributes to the proteolytic cleavage of GLI (glioma-associated homolog) into GLI3 (repressor), which increases oxidative stress, neuronal excitotoxicity, neuroinflammation, and apoptosis by suppressing target gene expression. We outlined in this review that SMO-SHH deregulation plays a crucial role in the pathogenesis of autism and addresses the current status of SMO-SHH pathway modulators. Additionally, a greater understanding of the SHH signaling pathway is an effort to improve successful treatment for autism and other neurological disorders.
Collapse
Affiliation(s)
- Saloni Rahi
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
21
|
Groves I, Placzek M, Fletcher AG. Of mitogens and morphogens: modelling Sonic Hedgehog mechanisms in vertebrate development. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190660. [PMID: 32829689 PMCID: PMC7482217 DOI: 10.1098/rstb.2019.0660] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2020] [Indexed: 12/22/2022] Open
Abstract
Sonic Hedgehog (Shh) Is a critical protein in vertebrate development, orchestrating patterning and growth in many developing systems. First described as a classic morphogen that patterns tissues through a spatial concentration gradient, subsequent studies have revealed a more complex mechanism, in which Shh can also regulate proliferation and differentiation. While the mechanism of action of Shh as a morphogen is well understood, it remains less clear how Shh might integrate patterning, proliferation and differentiation in a given tissue, to ultimately direct its morphogenesis. In tandem with experimental studies, mathematical modelling can help gain mechanistic insights into these processes and bridge the gap between Shh-regulated patterning and growth, by integrating these processes into a common theoretical framework. Here, we briefly review the roles of Shh in vertebrate development, focusing on its functions as a morphogen, mitogen and regulator of differentiation. We then discuss the contributions that modelling has made to our understanding of the action of Shh and highlight current challenges in using mathematical models in a quantitative and predictive way. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Ian Groves
- School of Mathematics and Statistics, University of Sheffield, Hicks Building, Hounsfield Road, Sheffield S3 7RH, UK
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Marysia Placzek
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Alexander G. Fletcher
- School of Mathematics and Statistics, University of Sheffield, Hicks Building, Hounsfield Road, Sheffield S3 7RH, UK
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| |
Collapse
|
22
|
Li X, Li Y, Li S, Li H, Yang C, Lin J. The role of Shh signalling pathway in central nervous system development and related diseases. Cell Biochem Funct 2020; 39:180-189. [PMID: 32840890 DOI: 10.1002/cbf.3582] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/07/2020] [Accepted: 08/01/2020] [Indexed: 12/15/2022]
Abstract
Sonic hedgehog (Shh) plays important roles in developmental of vertebrate animal central nervous system (CNS), and Gli is its downstream signal molecule. Shh signalling is essential for pattern formation, cell-fate specification, axon guidance, proliferation, survival and differentiation of neurons in CNS development. The abnormal signalling pathway of Shh leads to the occurrence of many nervous system diseases. The mechanism of Shh signalling is complex and remains incompletely understood. Nevertheless, studies have revealed that Shh signalling pathway is classified into canonical and non-canonical pathways. Here we review the role of the Shh signalling pathway and its impact in CNS development and related diseases. Specifically, we discuss the role of Shh in the spinal cord and brain development, cell differentiation and proliferation in CNS and related diseases such as brain tumour, Parkinson's diseases, epilepsy, autism, depression and traumatic brain injury. We also highlight future directions of research that could help to clarify the mechanisms and consequences of Shh signalling in the process of CNS development and related diseases. SIGNIFICANCE OF THE STUDY: This review summarized the role of Shh signalling pathway in CNS development and related diseases such as brain tumour, Parkinson's diseases, epilepsy, autism, depression and traumatic brain injury. It also presented the author's opinions on the future research direction of Shh signalling pathway.
Collapse
Affiliation(s)
- Xiaoying Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Yunxiao Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Shuanqing Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Han Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Ciqing Yang
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
23
|
Boa-Amponsem O, Zhang C, Burton D, Williams KP, Cole GJ. Ethanol and Cannabinoids Regulate Zebrafish GABAergic Neuron Development and Behavior in a Sonic Hedgehog and Fibroblast Growth Factor-Dependent Mechanism. Alcohol Clin Exp Res 2020; 44:1366-1377. [PMID: 32472575 DOI: 10.1111/acer.14383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/19/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND Ethanol (EtOH) has diverse effects on nervous system development, which includes development and survival of GABAergic neurons in a sonic hedgehog (Shh) and fibroblast growth factor (Fgf)-dependent mechanism. Cannabinoids also function as inhibitors of Shh signaling, raising the possibility that EtOH and cannabinoids may interact to broadly disrupt neuronal function during brain development. METHODS Zebrafish embryos were exposed to a range of EtOH and/or cannabinoid receptor 1 (CB1R) agonist concentrations at specific developmental stages, in the absence or presence of morpholino oligonucleotides that disrupt shh expression. In situ hybridization was employed to analyze glutamic acid decarboxylase (gad1) gene expression as a marker of GABAergic neuron differentiation, and zebrafish behavior was analyzed using the novel tank diving test as a measure of risk-taking behavior. RESULTS Combined acute subthreshold EtOH and CB1R agonist exposure results in a marked reduction in gad1 mRNA expression in zebrafish forebrain. Consistent with the EtOH and cannabinoid effects on Shh signaling, fgf8 mRNA overexpression rescues the EtOH- and cannabinoid-induced decrease in gad1 gene expression and also prevents the changes in behavior induced by EtOH and cannabinoids. CONCLUSIONS These studies provide evidence that forebrain GABAergic neuron development and zebrafish risk-taking behavior are sensitive to both EtOH and cannabinoid exposure in a Shh- and Fgf-dependent mechanism, and provide additional evidence that a signaling pathway involving Shh and Fgf crosstalk is a critical target of EtOH and cannabinoids in FASD.
Collapse
Affiliation(s)
- Oswald Boa-Amponsem
- From the, Integrated Biosciences Program, (OB-A), North Carolina Central University, Durham, North Carolina.,Julius L. Chambers Biomedical/Biotechnology Research Institute, (OB-A, C-Z, GJC), North Carolina Central University, Durham, North Carolina
| | - Chengjin Zhang
- Julius L. Chambers Biomedical/Biotechnology Research Institute, (OB-A, C-Z, GJC), North Carolina Central University, Durham, North Carolina
| | - Derek Burton
- Biomanufacturing Research Institute and Technology Enterprise, (DB, KPW), North Carolina Central University, Durham, North Carolina
| | - Kevin P Williams
- Biomanufacturing Research Institute and Technology Enterprise, (DB, KPW), North Carolina Central University, Durham, North Carolina
| | - Gregory J Cole
- Julius L. Chambers Biomedical/Biotechnology Research Institute, (OB-A, C-Z, GJC), North Carolina Central University, Durham, North Carolina.,Department of Biological and Biomedical Sciences, (GJC), North Carolina Central University, Durham, North Carolina
| |
Collapse
|
24
|
Molecular Regulation in Dopaminergic Neuron Development. Cues to Unveil Molecular Pathogenesis and Pharmacological Targets of Neurodegeneration. Int J Mol Sci 2020; 21:ijms21113995. [PMID: 32503161 PMCID: PMC7312927 DOI: 10.3390/ijms21113995] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
The relatively few dopaminergic neurons in the mammalian brain are mostly located in the midbrain and regulate many important neural functions, including motor integration, cognition, emotive behaviors and reward. Therefore, alteration of their function or degeneration leads to severe neurological and neuropsychiatric diseases. Unraveling the mechanisms of midbrain dopaminergic (mDA) phenotype induction and maturation and elucidating the role of the gene network involved in the development and maintenance of these neurons is of pivotal importance to rescue or substitute these cells in order to restore dopaminergic functions. Recently, in addition to morphogens and transcription factors, microRNAs have been identified as critical players to confer mDA identity. The elucidation of the gene network involved in mDA neuron development and function will be crucial to identify early changes of mDA neurons that occur in pre-symptomatic pathological conditions, such as Parkinson’s disease. In addition, it can help to identify targets for new therapies and for cell reprogramming into mDA neurons. In this essay, we review the cascade of transcriptional and posttranscriptional regulation that confers mDA identity and regulates their functions. Additionally, we highlight certain mechanisms that offer important clues to unveil molecular pathogenesis of mDA neuron dysfunction and potential pharmacological targets for the treatment of mDA neuron dysfunction.
Collapse
|
25
|
Fan Y, Winanto, Ng SY. Replacing what's lost: a new era of stem cell therapy for Parkinson's disease. Transl Neurodegener 2020; 9:2. [PMID: 31911835 PMCID: PMC6945567 DOI: 10.1186/s40035-019-0180-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022] Open
Abstract
Background Stem cells hold tremendous promise for regenerative medicine because they can be expanded infinitely, giving rise to large numbers of differentiated cells required for transplantation. Stem cells can be derived from fetal sources, embryonic origins (embryonic stem cells or ESCs) or reprogrammed from adult cell types (induced pluripotent stem cells or iPSCs). One unique property of stem cells is their ability to be directed towards specific cell types of clinical interest, and can mature into functional cell types in vivo. While transplantations of fetal or ESC-derived tissues are known to illicit a host immunogenic response, autologous transplantations using cell types derived from one’s own iPSCs eliminate risks of tissue rejection and reduce the need for immunosuppressants. However, even with these benefits, cell therapy comes with significant hurdles that researchers are starting to overcome. In this review, we will discuss the various steps to ensure safety, efficacy and clinical practicality of cell replacement therapy in neurodegenerative diseases, in particular, Parkinson’s disease. Main body Parkinson’s disease (PD) results from a loss of dopaminergic neurons from the substantia nigra and is an ideal target for cell replacement therapy. Early trials using fetal midbrain material in the late 1980s have resulted in long term benefit for some patients, but there were multiple shortcomings including the non-standardization and quality control of the transplanted fetal material, and graft-induced dyskinesia that some patients experience as a result. On the other hand, pluripotent stem cells such as ESCs and iPSCs serve as an attractive source of cells because they can be indefinitely cultured and is an unlimited source of cells. Stem cell technologies and our understanding of the developmental potential of ESCs and iPSCs have deepened in recent years and a clinical trial for iPSC-derived dopaminergic cells is currently undergoing for PD patients in Japan. In this focused review, we will first provide a historical aspect of cell therapies in PD, and then discuss the various challenges pertaining to the safety and efficacy of stem cell-based cell transplantations, and how these hurdles were eventually overcome. Conclusion With the maturity of the iPSC technology, cell transplantation appears to be a safe and effective therapy. Grafts in non-human primates survive and remain functional for more than 2 years after transplantation, with no signs of tumorigenesis, indicating safety and efficacy of the treatment. However, immunosuppressants are still required because of the lack of “universal stem cells” that would not evoke an immune response. The results of ongoing and upcoming trials by a global consortium known as GForce-PD would be highly anticipated because the success of these trials would open up possibilities for using cell therapy for the treatment of PD and other degenerative diseases.
Collapse
Affiliation(s)
- Yong Fan
- 1The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150 China
| | - Winanto
- 2Institute of Molecular and Cell Biology, ASTAR Research Entities, Singapore, 138673 Singapore
| | - Shi-Yan Ng
- 1The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150 China.,2Institute of Molecular and Cell Biology, ASTAR Research Entities, Singapore, 138673 Singapore.,3Yong Loo Lin School of Medicine (Physiology), National University of Singapore, Singapore, 117456 Singapore.,4National Neuroscience Institute, Singapore, 308433 Singapore
| |
Collapse
|
26
|
Adachi C, Kakinuma N, Jo SH, Ishii T, Arai Y, Arai S, Kitaguchi T, Takeda S, Inoue T. Sonic hedgehog enhances calcium oscillations in hippocampal astrocytes. J Biol Chem 2019; 294:16034-16048. [PMID: 31506300 DOI: 10.1074/jbc.ra119.007883] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 09/04/2019] [Indexed: 01/05/2023] Open
Abstract
Sonic hedgehog (SHH) is important for organogenesis during development. Recent studies have indicated that SHH is also involved in the proliferation and transformation of astrocytes to the reactive phenotype. However, the mechanisms underlying these are unknown. Involvement of SHH signaling in calcium (Ca) signaling has not been extensively studied. Here, we report that SHH and Smoothened agonist (SAG), an activator of the signaling receptor Smoothened (SMO) in the SHH pathway, activate Ca oscillations in cultured murine hippocampal astrocytes. The response was rapid, on a minute time scale, indicating a noncanonical pathway activity. Pertussis toxin blocked the SAG effect, indicating an involvement of a Gi coupled to SMO. Depletion of extracellular ATP by apyrase, an ATP-degrading enzyme, inhibited the SAG-mediated activation of Ca oscillations. These results indicate that SAG increases extracellular ATP levels by activating ATP release from astrocytes, resulting in Ca oscillation activation. We hypothesize that SHH activates SMO-coupled Gi in astrocytes, causing ATP release and activation of Gq/11-coupled P2 receptors on the same cell or surrounding astrocytes. Transcription factor activities are often modulated by Ca patterns; therefore, SHH signaling may trigger changes in astrocytes by activating Ca oscillations. This enhancement of Ca oscillations by SHH signaling may occur in astrocytes in the brain in vivo because we also observed it in hippocampal brain slices. In summary, SHH and SAG enhance Ca oscillations in hippocampal astrocytes, Gi mediates SAG-induced Ca oscillations downstream of SMO, and ATP-permeable channels may promote the ATP release that activates Ca oscillations in astrocytes.
Collapse
Affiliation(s)
- Chihiro Adachi
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo 1628480, Japan
| | - Naoto Kakinuma
- Department of Anatomy and Cell Biology, Interdisciplinary School of Medicine & Engineering, University of Yamanashi, Yamanashi 4093898, Japan
| | - Soo Hyun Jo
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo 1628480, Japan
| | - Takayuki Ishii
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo 1628480, Japan
| | - Yusuke Arai
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo 1628480, Japan
| | - Satoshi Arai
- Cell Signaling Group, Waseda Bioscience Research Institute in Singapore (WABIOS), Singapore 138667.,Research Institute for Science and Engineering, Waseda University, Tokyo 1698555, Japan
| | - Tetsuya Kitaguchi
- Cell Signaling Group, Waseda Bioscience Research Institute in Singapore (WABIOS), Singapore 138667.,Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Tokyo 2268503, Japan
| | - Sen Takeda
- Department of Anatomy and Cell Biology, Interdisciplinary School of Medicine & Engineering, University of Yamanashi, Yamanashi 4093898, Japan
| | - Takafumi Inoue
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo 1628480, Japan
| |
Collapse
|
27
|
Li G, Chen K, You D, Xia M, Li W, Fan S, Chai R, Zhang Y, Li H, Sun S. Laminin-Coated Electrospun Regenerated Silk Fibroin Mats Promote Neural Progenitor Cell Proliferation, Differentiation, and Survival in vitro. Front Bioeng Biotechnol 2019; 7:190. [PMID: 31448271 PMCID: PMC6691020 DOI: 10.3389/fbioe.2019.00190] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/23/2019] [Indexed: 12/16/2022] Open
Abstract
Neural progenitor cell (NPC) transplantation is a promising technique for central nervous system (CNS) reconstruction and regeneration. Biomaterial scaffolds, frameworks, and platforms can support NPC proliferation and differentiation in vitro as well as serve as a temporary extracellular matrix after transplantation. However, further applications of biomaterials require improved biological attributes. Silk fibroin (SF), which is produced by Bombyx mori, is a widely used and studied protein polymer for biomaterial application. Here, we prepared aligned and random eletrospun regenerated SF (RSF) scaffolds, and evaluated their impact on the growth of NPCs. First, we isolated NPCs and then cultured them on either laminin-coated RSF mats or conventional laminin-coated coverslips for cell assays. We found that aligned and random RSF led to increases in NPC proliferation of 143.8 ± 13.3% and 156.3 ± 14.7%, respectively, compared to controls. Next, we investigated neuron differentiation and found that the aligned and the random RSF led to increases in increase in neuron differentiation of about 93.2 ± 6.4%, and 3167.1 ± 4.8%, respectively, compared to controls. Furthermore, we measured the survival of NPCs and found that RSF promoted NPC survival, and found there was no difference among those three groups. Finally, signaling pathways in cells cultured on RSF mats were studied for their contributions in neural cell differentiation. Our results indicate that RSF mats provide a functional microenvironment and represent a useful scaffold for the development of new strategies in neural engineering research.
Collapse
Affiliation(s)
- Guangfei Li
- NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology, Shanghai Engineering Research Centre of Cochlear Implant, Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Ear, Nose & Throat Institute, Fudan University, Shanghai, China
| | - Kai Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Materials Science and Engineering, Donghua University, Shanghai, China
| | - Dan You
- NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology, Shanghai Engineering Research Centre of Cochlear Implant, Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Ear, Nose & Throat Institute, Fudan University, Shanghai, China
| | - Mingyu Xia
- NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology, Shanghai Engineering Research Centre of Cochlear Implant, Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Ear, Nose & Throat Institute, Fudan University, Shanghai, China
| | - Wen Li
- NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology, Shanghai Engineering Research Centre of Cochlear Implant, Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Ear, Nose & Throat Institute, Fudan University, Shanghai, China
| | - Suna Fan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Materials Science and Engineering, Donghua University, Shanghai, China
| | - Renjie Chai
- NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology, Shanghai Engineering Research Centre of Cochlear Implant, Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Ear, Nose & Throat Institute, Fudan University, Shanghai, China.,Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Yaopeng Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-Dimension Materials, College of Materials Science and Engineering, Donghua University, Shanghai, China
| | - Huawei Li
- NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology, Shanghai Engineering Research Centre of Cochlear Implant, Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Ear, Nose & Throat Institute, Fudan University, Shanghai, China.,Collaborative Innovation Center for Brain Science, Institute of Biomedical Sciences, Institute of Brain Science, Fudan University, Shanghai, China
| | - Shan Sun
- NHC Key Laboratory of Hearing Medicine, State Key Laboratory of Medical Neurobiology, Shanghai Engineering Research Centre of Cochlear Implant, Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Ear, Nose & Throat Institute, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Abstract
Signaling pathways that mediate cell-cell communication are essential for collective cell behaviors in multicellular systems. The hedgehog (HH) pathway, first discovered and elucidated in Drosophila, is one of these iconic signaling systems that plays many roles during embryogenesis and in adults; abnormal HH signaling can lead to birth defects and cancer. We review recent structural and biochemical studies that have advanced our understanding of the vertebrate HH pathway, focusing on the mechanisms by which the HH signal is received by patched on target cells, transduced across the cell membrane by smoothened, and transmitted to the nucleus by GLI proteins to influence gene-expression programs.
Collapse
Affiliation(s)
- Jennifer H Kong
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
29
|
Chen W, Huang Q, Ma S, Li M. Progress in Dopaminergic Cell Replacement and Regenerative Strategies for Parkinson's Disease. ACS Chem Neurosci 2019; 10:839-851. [PMID: 30346716 DOI: 10.1021/acschemneuro.8b00389] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is a chronic progressive neurodegenerative disorder symptomatically characterized by resting tremor, rigidity, bradykinesia, and gait impairment. These motor deficits suffered by PD patients primarily result from selective dysfunction or loss of dopaminergic neurons of the substantia nigra pars compacta (SNpc). Most of the existing therapies for PD are based on the replacement of dopamine, which is symptomatically effective in the early stage but becomes increasingly less effective and is accompanied by serious side effects in the advanced stages of the disease. Currently, there are no strategies to slow neuronal degeneration or prevent the progression of PD. Thus, the prospect of regenerating functional dopaminergic neurons is very attractive. Over the last few decades, significant progress has been made in the development of dopaminergic regenerative strategies for curing PD. The most promising approach seems to be cell-replacement therapy (CRT) using human embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs), which are unlimitedly available and have gained much success in preclinical trials. Despite the challenges, stem cell-based CRT will make significant steps toward the clinic in the coming decade. Alternatively, direct lineage reprogramming, especially in situ direct conversion of glia cells to induced neurons, which exhibits some advantages including no ethical concerns, no risk of tumor formation, and even no need for transplantation, has gained much attention recently. Evoking the endogenous regeneration ability of neural stem cells (NSCs) is an idyllic method of dopaminergic neuroregeneration which remains highly controversial. Here, we review many of these advances, highlighting areas and strategies that might be particularly suited to the development of regenerative approaches that restore dopaminergic function in PD.
Collapse
Affiliation(s)
- Weizhao Chen
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Qiaoying Huang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Shanshan Ma
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Mingtao Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| |
Collapse
|
30
|
Brodski C, Blaess S, Partanen J, Prakash N. Crosstalk of Intercellular Signaling Pathways in the Generation of Midbrain Dopaminergic Neurons In Vivo and from Stem Cells. J Dev Biol 2019; 7:jdb7010003. [PMID: 30650592 PMCID: PMC6473842 DOI: 10.3390/jdb7010003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Dopamine-synthesizing neurons located in the mammalian ventral midbrain are at the center stage of biomedical research due to their involvement in severe human neuropsychiatric and neurodegenerative disorders, most prominently Parkinson’s Disease (PD). The induction of midbrain dopaminergic (mDA) neurons depends on two important signaling centers of the mammalian embryo: the ventral midline or floor plate (FP) of the neural tube, and the isthmic organizer (IsO) at the mid-/hindbrain boundary (MHB). Cells located within and close to the FP secrete sonic hedgehog (SHH), and members of the wingless-type MMTV integration site family (WNT1/5A), as well as bone morphogenetic protein (BMP) family. The IsO cells secrete WNT1 and the fibroblast growth factor 8 (FGF8). Accordingly, the FGF8, SHH, WNT, and BMP signaling pathways play crucial roles during the development of the mDA neurons in the mammalian embryo. Moreover, these morphogens are essential for the generation of stem cell-derived mDA neurons, which are critical for the modeling, drug screening, and cell replacement therapy of PD. This review summarizes our current knowledge about the functions and crosstalk of these signaling pathways in mammalian mDA neuron development in vivo and their applications in stem cell-based paradigms for the efficient derivation of these neurons in vitro.
Collapse
Affiliation(s)
- Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel.
| | - Sandra Blaess
- Institute of Reconstructive Neurobiology, University of Bonn Medical Center, 53127 Bonn, Germany.
| | - Juha Partanen
- Faculty of Biological and Environmental Sciences, FIN00014-University of Helsinki, P.O. Box 56, Viikinkaari 9, FIN-00014 Helsinki, Finland.
| | - Nilima Prakash
- Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany.
| |
Collapse
|
31
|
Chen SD, Yang JL, Hwang WC, Yang DI. Emerging Roles of Sonic Hedgehog in Adult Neurological Diseases: Neurogenesis and Beyond. Int J Mol Sci 2018; 19:ijms19082423. [PMID: 30115884 PMCID: PMC6121355 DOI: 10.3390/ijms19082423] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 12/14/2022] Open
Abstract
Sonic hedgehog (Shh), a member of the hedgehog (Hh) family, was originally recognized as a morphogen possessing critical characters for neural development during embryogenesis. Recently, however, Shh has emerged as an important modulator in adult neural tissues through different mechanisms such as neurogenesis, anti-oxidation, anti-inflammation, and autophagy. Therefore, Shh may potentially have clinical application in neurodegenerative diseases and brain injuries. In this article, we present some examples, including ours, to show different aspects of Shh signaling and how Shh agonists or mimetics are used to alter the neuronal fates in various disease models, both in vitro and in vivo. Other potential mechanisms that are discussed include alteration of mitochondrial function and anti-aging effect; both are critical for age-related neurodegenerative diseases. A thorough understanding of the protective mechanisms elicited by Shh may provide a rationale to design innovative therapeutic regimens for various neurodegenerative diseases.
Collapse
Affiliation(s)
- Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 83301, Taiwan.
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 83301, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan.
| | - Jenq-Lin Yang
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung City 83301, Taiwan.
| | - Wei-Chao Hwang
- Department of Neurology, Taipei City Hospital, Taipei 11556, Taiwan.
| | - Ding-I Yang
- Institute of Brain Science, National Yang-Ming University, Taipei 11221, Taiwan.
- Brain Research Center, National Yang-Ming University, Taipei 11221, Taiwan.
| |
Collapse
|
32
|
Cellular fate decisions in the developing female anteroventral periventricular nucleus are regulated by canonical Notch signaling. Dev Biol 2018; 442:87-100. [PMID: 29885287 DOI: 10.1016/j.ydbio.2018.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/05/2018] [Indexed: 01/20/2023]
Abstract
The hypothalamic anteroventral periventricular nucleus (AVPV) is the major regulator of reproductive function within the hypothalamic-pituitary-gonadal (HPG) axis. Despite an understanding of the function of neuronal subtypes within the AVPV, little is known about the molecular mechanisms regulating their development. Previous work from our laboratory has demonstrated that Notch signaling is required in progenitor cell maintenance and formation of kisspeptin neurons of the arcuate nucleus (ARC) while simultaneously restraining POMC neuron number. Based on these findings, we hypothesized that the Notch signaling pathway may act similarly in the AVPV by promoting development of kisspeptin neurons at the expense of other neuronal subtypes. To address this hypothesis, we utilized a genetic mouse model with a conditional loss of Rbpj in Nkx2.1 expressing cells (Rbpj cKO). We noted an increase in cellular proliferation, as marked by Ki-67, in the hypothalamic ventricular zone (HVZ) in Rbpj cKO mice at E13.5. This corresponded to an increase in general neurogenesis and more TH-positive neurons. Additionally, an increase in OLIG2-positive early oligodendrocytic precursor cells was observed at postnatal day 0 in Rbpj cKO mice. By 5 weeks of age in Rbpj cKO mice, TH-positive cells were readily detected in the AVPV but few kisspeptin neurons were present. To elucidate the direct effects of Notch signaling on neuron and glia differentiation, an in vitro primary hypothalamic neurosphere assay was employed. We demonstrated that treatment with the chemical Notch inhibitor DAPT increased mKi67 and Olig2 mRNA expression while decreasing astroglial Gfap expression, suggesting Notch signaling regulates both proliferation and early glial fate decisions. A modest increase in expression of TH in both the cell soma and neurite extensions was observed after extended culture, suggesting that inhibition of Notch signaling alone is enough to bias progenitors towards a dopaminergic fate. Together, these data suggest that Notch signaling restricts early cellular proliferation and differentiation of neurons and oligodendrocytes both in vivo and in vitro and acts as a fate selector of kisspeptin neurons.
Collapse
|
33
|
Srivastava A, Singh S, Pandey A, Kumar D, Rajpurohit CS, Khanna VK, Pant AB. Secretome of Differentiated PC12 Cells Enhances Neuronal Differentiation in Human Mesenchymal Stem Cells Via NGF-Like Mechanism. Mol Neurobiol 2018. [PMID: 29527653 DOI: 10.1007/s12035-018-0981-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The secretome-mediated responses over cellular physiology are well documented. Stem cells have been ruling the field of secretomics and its role in regenerative medicine since the past few years. However, the mechanistic aspects of secretome-mediated responses and the role of other cells in this area remain somewhat elusive. Here, we investigate the effects of secretome-enriched conditioned medium (CM) of neuronally differentiated PC12 cells on the neuronal differentiation of human mesenchymal stem cells (hMSCs). The exposure to CM at a ratio of 1:1 (CM: conditioned medium of PC12 cells) led to neuronal induction in hMSCs. This neuronal induction was compared with a parallel group of cells exposed to nerve growth factor (NGF). There was a marked increase in neurite length and expression of neuronal markers (β-III tubulin, neurofilament-M (NF-M), synaptophysin, NeuN in exposed hMSCs). Experimental group co-exposed to NGF and CM showed an additive response via MAPK signaling and directed the cells particularly towards cholinergic lineage. The ability of CM to enhance the neuronal properties of stem cells could aid in their rapid differentiation into neuronal subtypes in case of stem cell transplantation for neuronal injuries, thus broadening the scope of non-stem cell-based applications in the area of secretomics.
Collapse
Affiliation(s)
- A Srivastava
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India
| | - S Singh
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - A Pandey
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India
| | - D Kumar
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - C S Rajpurohit
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - V K Khanna
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - A B Pant
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India. .,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India.
| |
Collapse
|
34
|
Shall G, Menosky M, Decker S, Nethala P, Welchko R, Leveque X, Lu M, Sandstrom M, Hochgeschwender U, Rossignol J, Dunbar G. Effects of Passage Number and Differentiation Protocol on the Generation of Dopaminergic Neurons from Rat Bone Marrow-Derived Mesenchymal Stem Cells. Int J Mol Sci 2018; 19:ijms19030720. [PMID: 29498713 PMCID: PMC5877581 DOI: 10.3390/ijms19030720] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/09/2018] [Accepted: 02/28/2018] [Indexed: 01/01/2023] Open
Abstract
Multiple studies have demonstrated the ability of mesenchymal stem cells (MSCs) to differentiate into dopamine-producing cells, in vitro and in vivo, indicating their potential to be used in the treatment of Parkinson’s disease (PD). However, there are discrepancies among studies regarding the optimal time (i.e., passage number) and method for dopaminergic induction, in vitro. In the current study, we compared the ability of early (P4) and later (P40) passaged bone marrow-derived MSCs to differentiate into dopaminergic neurons using two growth-factor-based approaches. A direct dopaminergic induction (DDI) was used to directly convert MSCs into dopaminergic neurons, and an indirect dopaminergic induction (IDI) was used to direct MSCs toward a neuronal lineage prior to terminal dopaminergic differentiation. Results indicate that both early and later passaged MSCs exhibited positive expression of neuronal and dopaminergic markers following either the DDI or IDI protocols. Additionally, both early and later passaged MSCs released dopamine and exhibited spontaneous neuronal activity following either the DDI or IDI. Still, P4 MSCs exhibited significantly higher spiking and bursting frequencies as compared to P40 MSCs. Findings from this study provide evidence that early passaged MSCs, which have undergone the DDI, are more efficient at generating dopaminergic-like cells in vitro, as compared to later passaged MSCs or MSCs that have undergone the IDI.
Collapse
Affiliation(s)
- Gabrielle Shall
- Field Neurosciences Institute Laboratory for Restorative Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Megan Menosky
- Field Neurosciences Institute Laboratory for Restorative Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Sarah Decker
- Field Neurosciences Institute Laboratory for Restorative Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Priya Nethala
- Field Neurosciences Institute Laboratory for Restorative Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Ryan Welchko
- Field Neurosciences Institute Laboratory for Restorative Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Xavier Leveque
- Field Neurosciences Institute Laboratory for Restorative Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Ming Lu
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Michael Sandstrom
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
- College of Humanities and Social and Behavioral Sciences, Psychology Department, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Ute Hochgeschwender
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
- College of Medicine, Central Michigan University, Mount Pleasant, MI 48859 USA.
- Field Neurosciences Institute, 4677 Towne Centre Rd. Suite 101, Saginaw, MI 48604, USA.
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
- College of Medicine, Central Michigan University, Mount Pleasant, MI 48859 USA.
| | - Gary Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
- Neuroscience Program, Central Michigan University, Mount Pleasant, MI 48859, USA.
- College of Humanities and Social and Behavioral Sciences, Psychology Department, Central Michigan University, Mount Pleasant, MI 48859, USA.
- College of Medicine, Central Michigan University, Mount Pleasant, MI 48859 USA.
| |
Collapse
|
35
|
Zammit V, Baron B, Ayers D. MiRNA Influences in Neuroblast Modulation: An Introspective Analysis. Genes (Basel) 2018; 9:genes9010026. [PMID: 29315268 PMCID: PMC5793179 DOI: 10.3390/genes9010026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/22/2017] [Accepted: 12/29/2017] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma (NB) is the most common occurring solid paediatric cancer in children under the age of five years. Whether of familial or sporadic origin, chromosome abnormalities contribute to the development of NB and cause dysregulation of microRNAs (miRNAs). MiRNAs are small non-coding, single stranded RNAs that target messenger RNAs at the post-transcriptional levels by repressing translation within all facets of human physiology. Such gene 'silencing' activities by miRNAs allows the development of regulatory feedback loops affecting multiple functions within the cell, including the possible differentiation of neural stem cell (NSC) lineage selection. Neurogenesis includes stages of self-renewal and fate specification of NSCs, migration and maturation of young neurones, and functional integration of new neurones into the neural circuitry, all of which are regulated by miRNAs. The role of miRNAs and their interaction in cellular processes are recognised aspects of cancer genetics, and miRNAs are currently employed as biomarkers for prognosis and tumour characterisation in multiple cancer models. Consequently, thorough understanding of the mechanisms of how these miRNAs interplay at the transcriptomic level will definitely lead to the development of novel, bespoke and efficient therapeutic measures, with this review focusing on the influences of miRNAs on neuroblast modulations leading to neuroblastoma.
Collapse
Affiliation(s)
- Vanessa Zammit
- National Blood Transfusion Service, St. Luke's Hospital, PTA1010 G'Mangia, Malta.
- School of Biomedical Science and Physiology, University of Wolverhampton, Wolverhampton WV1 1LY, UK.
| | - Byron Baron
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta.
| | - Duncan Ayers
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta.
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK.
| |
Collapse
|
36
|
Feuerstein M, Chleilat E, Khakipoor S, Michailidis K, Ophoven C, Roussa E. Expression patterns of key Sonic Hedgehog signaling pathway components in the developing and adult mouse midbrain and in the MN9D cell line. Cell Tissue Res 2017; 370:211-225. [PMID: 28799057 PMCID: PMC5640734 DOI: 10.1007/s00441-017-2664-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/29/2022]
Abstract
The temporal dynamic expression of Sonic Hedgehog (SHH) and signaling during early midbrain dopaminergic (mDA) neuron development is one of the key players in establishing mDA progenitor diversity. However, whether SHH signaling is also required during later developmental stages and in mature mDA neurons is less understood. We study the expression of SHH receptors Ptch1 and Gas1 (growth arrest-specific 1) and of the transcription factors Gli1, Gli2 and Gli3 in mouse midbrain during embryonic development [embryonic day (E) 12.5 onwards)], in newborn and adult mice using in situ hybridization and immunohistochemistry. Moreover, we examine the expression and regulation of dopaminergic neuronal progenitor markers, midbrain dopaminergic neuronal markers and markers of the SHH signaling pathway in undifferentiated and butyric acid-treated (differentiated) MN9D cells in the presence or absence of exogenous SHH in vitro by RT-PCR, immunoblotting and immunocytochemistry. Gli1 was expressed in the lateral mesencephalic domains, whereas Gli2 and Gli3 were expressed dorsolaterally and complemented by ventrolateral expression of Ptch1. Co-localization with tyrosine hydroxylase could not be observed. GAS1 was exclusively expressed in the dorsal mesencephalon at E11.5 and co-localized with Ki67. In contrast, MN9D cells expressed all the genes investigated and treatment of the cells with butyric acid significantly upregulated their expression. The results suggest that SHH is only indirectly involved in the differentiation and survival of mDA neurons and that the MN9D cell line is a valuable model for investigating early development but not the differentiation and survival of mDA neurons.
Collapse
Affiliation(s)
- Melanie Feuerstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Enaam Chleilat
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Shokoufeh Khakipoor
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Konstantinos Michailidis
- Neuroanatomy, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Christian Ophoven
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany
| | - Eleni Roussa
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany. .,Neuroanatomy, Faculty of Medicine, University of Freiburg, Albertstrasse 17, D-79104, Freiburg, Germany.
| |
Collapse
|
37
|
Guerrero-Flores G, Bastidas-Ponce A, Collazo-Navarrete O, Guerra-Crespo M, Covarrubias L. Functional determination of the differentiation potential of ventral mesencephalic neural precursor cells during dopaminergic neurogenesis. Dev Biol 2017; 429:56-70. [PMID: 28733161 DOI: 10.1016/j.ydbio.2017.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/17/2017] [Accepted: 07/17/2017] [Indexed: 11/29/2022]
Abstract
The ventral mesencephalic neural precursor cells (vmNPCs) that give rise to dopaminergic (DA) neurons have been identified by the expression of distinct genes (e.g., Lmx1a, Foxa2, Msx1/2). However, the commitment of these NPCs to the mesencephalic DA neuronal fate has not been functionally determined. Evaluation of the plasticity of vmNPCs suggests that their commitment occurs after E10.5. Here we show that E9.5 vmNPCs implanted in an ectopic area of E10.5 mesencephalic explants, retained their specification marker Lmx1a and efficiently differentiated into neurons but did not express the gene encoding tyrosine hydroxylase (Th), the limiting enzyme for dopamine synthesis. A proportion of E10.5-E11.5 implanted vmNPCs behaved as committed, deriving into Th+ neurons in ectopic sites. Interestingly, implanted cells from E12.5 embryos were unable to give rise to a significant number of Th+ neurons. Concomitantly, differentiation assays in culture and in mesencephalic explants treated with Fgf2+LIF detected vmNPCs with astrogenic potential since E11.5. Despite this, a full suspension of E12.5 vmNPCs give rise to DA neurons in a similar proportion as those of E10.5 when they were transplanted into adult brain, but astrocytes were only detected with the former population. These data suggest that the subventricular postmitotic progenitors present in E12.5 ventral mesencephalon are unable to implant in embryonic explants and are the source of DA neurons in the transplanted adult brain. Based on our findings we propose that during DA differentiation committed vmNPCs emerge at E10.5 and they exhaust their neurogenic capacity with the rise of NPCs with astrogenic potential.
Collapse
Affiliation(s)
- Gilda Guerrero-Flores
- Department of Developmental Genetics and Molecular Physiology, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Aimée Bastidas-Ponce
- Department of Developmental Genetics and Molecular Physiology, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico
| | - Omar Collazo-Navarrete
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
| | - Magdalena Guerra-Crespo
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México 04510, Mexico
| | - Luis Covarrubias
- Department of Developmental Genetics and Molecular Physiology, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos 62210, Mexico.
| |
Collapse
|
38
|
Abstract
Sirtuins are pleiotropic NAD+ dependent histone deacetylases involved in metabolism, DNA damage repair, inflammation and stress resistance. SIRT6, a member of the sirtuin family, regulates the process of normal aging and increases the lifespan of male mice over-expressing Sirt6 by 15%. Neurogenesis, the formation of new neurons within the hippocampus of adult mammals, involves several complex stages including stem cell proliferation, differentiation, migration and network integration. During aging, the number of newly generated neurons continuously declines, and this is correlated with a decline in neuronal plasticity and cognitive behavior. In this study we investigated the involvement of SIRT6 in adult hippocampal neurogenesis. Mice over-expressing Sirt6 exhibit increased numbers of young neurons and decreased numbers of mature neurons, without affecting glial differentiation. This implies of an involvement of SIRT6 in neuronal differentiation and maturation within the hippocampus. This work adds to the expanding body of knowledge on the regulatory mechanisms underlying adult hippocampal neurogenesis, and describes novel roles for SIRT6 as a regulator of cell fate during adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Eitan Okun
- The Mina and Everard Goodman faculty of Life sciences, Bar Ilan University, Ramat Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer's disease research, Bar-Ilan University, Ramat Gan, Israel
- * E-mail:
| | - Daniel Marton
- The Mina and Everard Goodman faculty of Life sciences, Bar Ilan University, Ramat Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Daniel Cohen
- The Mina and Everard Goodman faculty of Life sciences, Bar Ilan University, Ramat Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Kathleen Griffioen
- Department of Biology and Chemistry, Liberty University, Lynchburg, VA, United States of America
| | - Yariv Kanfi
- The Mina and Everard Goodman faculty of Life sciences, Bar Ilan University, Ramat Gan, Israel
| | - Tomer Illouz
- The Mina and Everard Goodman faculty of Life sciences, Bar Ilan University, Ramat Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Ravit Madar
- The Mina and Everard Goodman faculty of Life sciences, Bar Ilan University, Ramat Gan, Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Haim Y. Cohen
- The Mina and Everard Goodman faculty of Life sciences, Bar Ilan University, Ramat Gan, Israel
| |
Collapse
|
39
|
Yurek DM, Fletcher-Turner A. Comparison of Embryonic Stem Cell-Derived Dopamine Neuron Grafts and Fetal Ventral Mesencephalic Tissue Grafts: Morphology and Function. Cell Transplant 2017; 13:295-306. [PMID: 15191167 DOI: 10.3727/000000004783983954] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In this study we compared the function and morphology of two types of neural grafts: allografts of fetal ventral mesencephalic (VM) tissue and xenografts of embryonic stem cell (ESC)-derived dopamine neurons. Mouse embryonic stem cells were cultured and exposed to differentiation factors that induced approximately 10% of the cells to express a dopaminergic phenotype. These cells were then harvested and implanted into the denervated striatum of rats with unilateral lesions of the nigrostriatal pathway. Another group of lesioned rats received allografts of fetal ventral mesencephalic tissue. While both types of grafts yield a similar number of tyrosine hydroxylase (TH)-positive cells, amphetamine-induced rotational behavior was differentially affected by these grafts: rotational behavior was significantly reduced in lesioned rats receiving allografts of fetal VM tissue while ESC grafts had slight but insignificant effects on rotational scores. Densitometry measures of TH+ fiber outgrowth revealed a similar area of reinnervation and a comparable number of TH+ cells for ESC graft when compared with VM grafts. These data suggest there are similarities and also distinct differences in the manner in which ESC and VM grafts interact with the denervated striatum.
Collapse
Affiliation(s)
- David M Yurek
- Department of Surgery/Neurosurgery, University of Kentucky College of Medicine, Lexington, KY 40536-0305, USA.
| | | |
Collapse
|
40
|
Yurek DM, Fletcher-Turner A, Moore J, Chai L, Mahanthappa N. Co-Grafts of Fetal Ventral Mesencephalon and Fibroblasts Expressing Sonic Hedgehog: Effect on Survival and Function of Dopamine Grafts. Cell Transplant 2017. [DOI: 10.3727/000000001783986305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- David M. Yurek
- Department of Surgery/Neurosurgery, University of Kentucky College of Medicine, Lexington, KY 40536-0305
| | - Anita Fletcher-Turner
- Department of Surgery/Neurosurgery, University of Kentucky College of Medicine, Lexington, KY 40536-0305
| | - Jennifer Moore
- Department of Surgery/Neurosurgery, University of Kentucky College of Medicine, Lexington, KY 40536-0305
| | - Ling Chai
- Curis, Inc., 45 Moulton Street, Cambridge, MA 02138
| | | |
Collapse
|
41
|
Lee JH, Chung YC, Bok E, Lee H, Huh SH, Lee JE, Jin BK, Ko HW. Injury-stimulated Sonic hedgehog expression in microglia contributes to neuroinflammatory response in the MPTP model of Parkinson's disease. Biochem Biophys Res Commun 2017; 482:980-986. [DOI: 10.1016/j.bbrc.2016.11.144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 11/26/2016] [Indexed: 01/20/2023]
|
42
|
Zhou X, Pace J, Filichia E, Lv T, Davis B, Hoffer B, Selman W, Luo Y. Effect of the sonic hedgehog receptor smoothened on the survival and function of dopaminergic neurons. Exp Neurol 2016; 283:235-45. [PMID: 27317298 PMCID: PMC5479305 DOI: 10.1016/j.expneurol.2016.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/24/2016] [Accepted: 06/12/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To determine the influence of the sonic hedgehog (shh) pathway and its receptor smoothened (smo), on the survival and functionality of dopaminergic (DA) neurons. BACKGROUND During early development, shh induces the differentiation of DA neurons. However, it is unknown whether shh signaling is required in the maturation or maintenance of DA neurons during later development and adulthood due to the lethality of traditional shh knockout models. METHODS We utilized the cre-loxP system to achieve late developmental stage and cell type-specific deletion of the shh receptor, smo, in DA neurons by crossing DATcre (dopamine transporter) mice with Smo(loxP/loxP) mice. We assessed for differences between knockout (ko) and wildtype (wt) mice using combined histochemistry, gene expression analysis, and behavioral evaluation. Number and size of DA neurons in ventral midbrain and the DA neural terminal density in striatum were measured using unbiased stereological quantification. The survival of DA neurons under neurotoxin challenge was examined in the unilateral 6-hydroxydopamine (6-OHDA) Parkinson's disease animal model and the more subtle function under challenge of the dopaminergic system was examined by methamphetamine single- and repeated challenge in wt and ko mice. RESULTS Tyrosine hydroxylase (TH) positive neuronal counts and neuronal size in substantia nigra (SN) and ventral tegmental area (VTA) showed no difference between wt and DAT-Smo ko mice in young (5months) or aged (22months) mice. There was also no difference in the striatal DA projections between wt and ko mice in both age groups. In unilateral striatal 6-OHDA lesions modeling Parkinson's disease, using stereotaxic injection of 6-OHDA intrastriatally led to loss of dopaminergic neurons in SN and diminished TH positive projections in striatum. However, there was no differences in survival of DA neurons between wt and ko mice. DAT-Smo ko mice demonstrated hyperactivity compared to wt mice at 5months, but showed no difference in activity at 22months. When injected with a one-time bolus of methamphetamine (METH), despite the higher basal locomotion activity, DAT-Smo ko mice showed a diminished response to a single METH challenge. In METH sensitization testing, ko mice showed decreased sensitization compared to wt mice without evidence of a delayed shift in dynamics of sensitization. Gene expression analysis showed decreased gene expression of smo, Gli 1 (known target gene of smo) and BDNF (brain-derived neurotrophic factor) in the SN. Gene expression was not altered in striatum for the genes examined in this study including dopamine receptor genes, neurotropic genes such as Glial cell line-derived neurotrophic factor (GDNF), and bone morphogenetic protein 7 (BMP7). CONCLUSION Our study showed the smo receptor function is not required for the maturation and survival of DA neurons during late development, aging or under stress challenge. However, smo function has an influence on behavior in young adult mice and in responses of mice to a drug that modulates DA neurochemistry through regulation of gene expression in DA neurons. Since young adult DAT-smo ko mice show hyperactivity and altered response to a psychostimulant drug (METH), this may indicate the involvement of the shh pathway in the development of functional changes that manifest as alterations in DA pathway dynamics.
Collapse
Affiliation(s)
- Xiaofei Zhou
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Jonathan Pace
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Emily Filichia
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Tao Lv
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Brandon Davis
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Barry Hoffer
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Warren Selman
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Yu Luo
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA.
| |
Collapse
|
43
|
Pignata A, Ducuing H, Castellani V. Commissural axon navigation: Control of midline crossing in the vertebrate spinal cord by the semaphorin 3B signaling. Cell Adh Migr 2016; 10:604-617. [PMID: 27532244 PMCID: PMC5160037 DOI: 10.1080/19336918.2016.1212804] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The mechanisms governing the navigation of commissural axons during embryonic development have been extensively investigated in the past years, often using the drosophila ventral nerve cord and the spinal cord as model systems. Similarities but also specificities in the general strategies, the molecular signals as well as in the regulatory pathways controlling the response of commissural axons to the guidance cues have been found between species. Whether the semaphorin signaling contributes to midline crossing in the fly nervous system remains unknown, while in contrast, it does play a prominent contribution in vertebrates. In this review we discuss the functions of the semaphorins during commissural axon guidance in the developing spinal cord, focusing on the family member semaphorin 3B (Sema3B) in the context of midline crossing in the spinal cord.
Collapse
Affiliation(s)
- Aurora Pignata
- a University of Lyon, Université Claude Bernard Lyon 1, NeuroMyogene Institute (INMG), UMR CNRS 5310, INSERM U1217 Lyon , France
| | - Hugo Ducuing
- a University of Lyon, Université Claude Bernard Lyon 1, NeuroMyogene Institute (INMG), UMR CNRS 5310, INSERM U1217 Lyon , France
| | - Valérie Castellani
- a University of Lyon, Université Claude Bernard Lyon 1, NeuroMyogene Institute (INMG), UMR CNRS 5310, INSERM U1217 Lyon , France
| |
Collapse
|
44
|
Mesocortical Dopamine Phenotypes in Mice Lacking the Sonic Hedgehog Receptor Cdon. eNeuro 2016; 3:eN-NWR-0009-16. [PMID: 27419218 PMCID: PMC4942720 DOI: 10.1523/eneuro.0009-16.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/21/2022] Open
Abstract
Motivated behaviors and many psychopathologies typically involve changes in dopamine release from the projections of the ventral tegmental area (VTA) and/or the substantia nigra pars compacta (SNc). The morphogen Sonic Hedgehog (Shh) specifies fates of midbrain dopamine neurons, but VTA-specific effects of Shh signaling are also being uncovered. In this study, we assessed the role of the Shh receptor Cdon in the development of VTA and SNc dopamine neurons. We find that Cdon is expressed in the proliferating progenitor zone of the embryonic ventral midbrain and that the number of proliferating cells in this region is increased in mouse Cdon(-/-) embryos. Consistent with a role of Shh in the regulation of neuronal proliferation in this region, we find that the number of tyrosine hydroxylase (TH)-positive neurons is increased in the VTA of Cdon(-/-) mice at birth and that this effect endures into adulthood. In contrast, the number of TH-positive neurons in the SNc is not altered in Cdon(-/-) mice at either age. Moreover, adult Cdon(-/-) mice have a greater number of medial prefrontal cortex (mPFC) dopamine presynaptic sites, and increased baseline concentrations of dopamine and dopamine metabolites selectively in this region. Finally, consistent with increased dopamine function in the mPFC, we find that adult Cdon(-/-) mice fail to exhibit behavioral plasticity upon repeated amphetamine treatment. Based on these data, we suggest that Cdon plays an important role encoding the diversity of dopamine neurons in the midbrain, influencing both the development of the mesocortical dopamine pathway and behavioral outputs that involve this neural circuitry.
Collapse
|
45
|
Heng BC, Lim LW, Wu W, Zhang C. An Overview of Protocols for the Neural Induction of Dental and Oral Stem Cells In Vitro. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:220-50. [PMID: 26757369 DOI: 10.1089/ten.teb.2015.0488] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To date, various adult stem cells have been identified within the oral cavity, including dental pulp stem cells, dental follicle stem cells, stem cells from apical papilla, stem cells from human exfoliated deciduous teeth, periodontal ligament stem cells, and mesenchymal stem cells from the gingiva. All of these possess neurogenic potential due to their common developmental origin from the embryonic neural crest. Besides the relative ease of isolation of these adult stem cells from readily available biological waste routinely produced during dental treatment, these cells also possess the advantage of immune compatibility in autologous transplantation. In recent years, much interest has been focused on the derivation of neural lineages from these adult stem cells for therapeutic applications in the brain, spinal cord, and peripheral nerve regeneration. In addition, there are also promising nontherapeutic applications of stem cell-derived neurons in pharmacological and toxicological screening of neuroactive drugs, and for in vitro modeling of neurodevelopmental and neurodegenerative diseases. Hence, this review will critically examine the diverse array of in vitro neural induction protocols that have been devised for dental and oral-derived stem cells. These protocols are defined not only by the culture milieu comprising the basal medium plus growth factors, small molecules, and other culture supplements but also by the substrata/surface coatings utilized, the presence of multiple culture stages, the total culture duration, the initial seeding density, and whether the spheroid/neurosphere formation is being utilized to recapitulate the three-dimensional neural differentiation microenvironment that is naturally present physiologically in vivo.
Collapse
Affiliation(s)
- Boon Chin Heng
- 1 Comprehensive Dental Care, Endodonthics, Faculty of Dentistry, The University of Hong Kong , Pokfulam, Hong Kong
| | - Lee Wei Lim
- 2 School of Biomedical Sciences, The University of Hong Kong , Pokfulam, Hong Kong
| | - Wutian Wu
- 2 School of Biomedical Sciences, The University of Hong Kong , Pokfulam, Hong Kong
| | - Chengfei Zhang
- 1 Comprehensive Dental Care, Endodonthics, Faculty of Dentistry, The University of Hong Kong , Pokfulam, Hong Kong
| |
Collapse
|
46
|
Majumdar D, Kanafi M, Bhonde R, Gupta P, Datta I. Differential Neuronal Plasticity of Dental Pulp Stem Cells From Exfoliated Deciduous and Permanent Teeth Towards Dopaminergic Neurons. J Cell Physiol 2016; 231:2048-63. [PMID: 26773559 DOI: 10.1002/jcp.25314] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/14/2016] [Indexed: 12/17/2022]
Abstract
Based on early occurrence in chronological age, stem-cells from human exfoliated deciduous teeth (SHED) has been reported to possess better differentiation-potential toward certain cell-lineage in comparison to stem-cells from adult teeth (DPSCs). Whether this same property between them extends for the yield of functional central nervous system neurons is still not evaluated. Hence, we aim to assess the neuronal plasticity of SHED in comparison to DPSCs toward dopaminergic-neurons and further, if the difference is reflected in a differential expression of sonic-hedgehog (SHH)-receptors and basal-expressions of tyrosine-hydroxylase [TH; through cAMP levels]. Human SHED and DPSCs were exposed to midbrain-cues [SHH, fibroblast growth-factor8, and basic fibroblast growth-factor], and their molecular, immunophenotypical, and functional characterization was performed at different time-points of induction. Though SHED and DPSCs spontaneously expressed early-neuronal and neural-crest marker in their naïve state, only SHED expressed a high basal-expression of TH. The upregulation of dopaminergic transcription-factors Nurr1, Engrailed1, and Pitx3 was more pronounced in DPSCs. The yield of TH-expressing cells decreased from 49.8% to 32.16% in SHED while it increased from 8.09% to 77.47% in DPSCs. Dopamine release and intracellular-Ca(2+) influx upon stimulation (KCl and ATP) was higher in induced DPSCs. Significantly lower-expression of SHH-receptors was noted in naïve SHED than DPSCs, which may explain the differential neuronal plasticity. In addition, unlike DPSCs, SHED showed a down-regulation of cyclic adenosine-monophosphate (cAMP) upon exposure to SHH; possibly another contributor to the lesser differentiation-potential. Our data clearly demonstrates for the first time that DPSCs possess superior neuronal plasticity toward dopaminergic-neurons than SHED; influenced by higher SHH-receptor and lower basal TH expression. J. Cell. Physiol. 231: 2048-2063, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Debanjana Majumdar
- Manipal Institute of Regenerative Medicine, University of Manipal, Bengaluru, Karnataka, India
| | - Mohammad Kanafi
- Manipal Institute of Regenerative Medicine, University of Manipal, Bengaluru, Karnataka, India
| | - Ramesh Bhonde
- Manipal Institute of Regenerative Medicine, University of Manipal, Bengaluru, Karnataka, India
| | - Pawan Gupta
- Stempeutics Research Pvt Limited, Bengaluru, Karnataka, India
| | - Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, Karnataka, India
| |
Collapse
|
47
|
Wagenführ L, Meyer AK, Marrone L, Storch A. Oxygen Tension Within the Neurogenic Niche Regulates Dopaminergic Neurogenesis in the Developing Midbrain. Stem Cells Dev 2016; 25:227-38. [PMID: 26577812 DOI: 10.1089/scd.2015.0214] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oxygen tension is an important factor controlling stem cell proliferation and maintenance in various stem cell populations with a particular relevance in midbrain dopaminergic progenitors. Further studies have shown that the oxygen-dependent transcription factor hypoxia-inducible factor 1α (HIF-1α) is involved in these processes. However, all available studies on oxygen effects in dopaminergic neuroprogenitors were performed in vitro and thus it remains unclear whether tissue oxygen tension in the embryonic midbrain is also relevant for the regulation of dopaminergic neurogenesis in vivo. We thus dissect here the effects of oxygen tension in combination with HIF-1α conditional knockout on dopaminergic neurogenesis by using a novel experimental design allowing for the control of oxygen tension within the microenvironment of the neurogenic niche of the murine fetal midbrain in vivo. The microenvironment of the midbrain dopaminergic neurogenic niche was detected as hypoxic with oxygen tensions below 1.1%. Maternal oxygen treatment of 10%, 21%, and 75% atmospheric oxygen tension for 48 h translates into robust changes in fetal midbrain oxygenation. Fetal midbrain hypoxia hampered the generation of dopaminergic neurons and is accompanied with restricted fetal midbrain development. In contrast, induced hyperoxia stimulated proliferation and differentiation of dopaminergic progenitors during early and late embryogenesis. Oxygen effects were not directly mediated through HIF-1α signaling. These data--in agreement with in vitro data-indicate that oxygen is a crucial regulator of developmental dopaminergic neurogenesis. Our study provides the initial framework for future studies on molecular mechanisms mediating oxygen regulation of dopaminergic neurogenesis within the fetal midbrain as its natural environment.
Collapse
Affiliation(s)
- Lisa Wagenführ
- 1 Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden , Dresden, Germany
| | - Anne Karen Meyer
- 1 Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden , Dresden, Germany
| | - Lara Marrone
- 1 Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden , Dresden, Germany .,2 Center for Regenerative Therapies Dresden (CRTD) , Technische Universität Dresden, Dresden, Germany
| | - Alexander Storch
- 1 Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden , Dresden, Germany .,2 Center for Regenerative Therapies Dresden (CRTD) , Technische Universität Dresden, Dresden, Germany .,3 Department of Neurology, University of Rostock , Rostock, Germany .,4 German Centre for Neurodegenerative Diseases (DZNE) , Rostock, Germany
| |
Collapse
|
48
|
Rodríguez-Traver E, Solís O, Díaz-Guerra E, Ortiz Ó, Vergaño-Vera E, Méndez-Gómez HR, García-Sanz P, Moratalla R, Vicario-Abejón C. Role of Nurr1 in the Generation and Differentiation of Dopaminergic Neurons from Stem Cells. Neurotox Res 2015; 30:14-31. [PMID: 26678495 DOI: 10.1007/s12640-015-9586-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/27/2015] [Accepted: 12/06/2015] [Indexed: 12/23/2022]
Abstract
NURR1 is an essential transcription factor for the differentiation, maturation, and maintenance of midbrain dopaminergic neurons (DA neurons) as it has been demonstrated using knock-out mice. DA neurons of the substantia nigra pars compacta degenerate in Parkinson's disease (PD) and mutations in the Nurr1 gene have been associated with this human disease. Thus, the study of NURR1 actions in vivo is fundamental to understand the mechanisms of neuron generation and degeneration in the dopaminergic system. Here, we present and discuss findings indicating that NURR1 is a valuable molecular tool for the in vitro generation of DA neurons which could be used for modeling and studying PD in cell culture and in transplantation approaches. Transduction of Nurr1 alone or in combination with other transcription factors such as Foxa2, Ngn2, Ascl1, and Pitx3, induces the generation of DA neurons, which upon transplantation have the capacity to survive and restore motor behavior in animal models of PD. We show that the survival of transplanted neurons is increased when the Nurr1-transduced olfactory bulb stem cells are treated with GDNF. The use of these and other factors with the induced pluripotent stem cell (iPSC)-based technology or the direct reprogramming of astrocytes or fibroblasts into human DA neurons has produced encouraging results for the study of the cellular and molecular mechanisms of neurodegeneration in PD and for the search of new treatments for this disease.
Collapse
Affiliation(s)
- Eva Rodríguez-Traver
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Oscar Solís
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Eva Díaz-Guerra
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Óscar Ortiz
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain
| | - Eva Vergaño-Vera
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Héctor R Méndez-Gómez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Patricia García-Sanz
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Carlos Vicario-Abejón
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
49
|
Bissonette GB, Roesch MR. Development and function of the midbrain dopamine system: what we know and what we need to. GENES BRAIN AND BEHAVIOR 2015; 15:62-73. [PMID: 26548362 DOI: 10.1111/gbb.12257] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/11/2015] [Accepted: 10/01/2015] [Indexed: 01/29/2023]
Abstract
The past two decades have seen an explosion in our understanding of the origin and development of the midbrain dopamine system. Much of this work has been focused on the aspects of dopamine neuron development related to the onset of movement disorders such as Parkinson's disease, with the intent of hopefully delaying, preventing or fixing symptoms. While midbrain dopamine degeneration is a major focus for treatment and research, many other human disorders are impacted by abnormal dopamine, including drug addiction, autism and schizophrenia. Understanding dopamine neuron ontogeny and how dopamine connections and circuitry develops may provide us with key insights into potentially important avenues of research for other dopamine-related disorders. This review will provide a brief overview of the major molecular and genetic players throughout the development of midbrain dopamine neurons and what we know about the behavioral- and disease-related implications associated with perturbations to midbrain dopamine neuron development. We intend to combine the knowledge of two broad fields of neuroscience, both developmental and behavioral, with the intent on fostering greater discussion between branches of neuroscience in the service of addressing complex cognitive questions from a developmental perspective and identifying important gaps in our knowledge for future study.
Collapse
Affiliation(s)
- G B Bissonette
- Department of Psychology, University of Maryland, College Park, MD, USA.,Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA
| | - M R Roesch
- Department of Psychology, University of Maryland, College Park, MD, USA.,Program in Neuroscience and Cognitive Science, University of Maryland, College Park, MD, USA
| |
Collapse
|
50
|
Hung HC, Hsiao YH, Gean PW. Sonic hedgehog signaling regulates amygdalar neurogenesis and extinction of fear memory. Eur Neuropsychopharmacol 2015; 25:1723-32. [PMID: 26271722 DOI: 10.1016/j.euroneuro.2015.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 06/09/2015] [Accepted: 07/14/2015] [Indexed: 10/23/2022]
Abstract
It is now recognized that neurogenesis occurs throughout life predominantly in the subgranular zone (SGZ) of the hippocampus and the subventricular zone (SVZ) of the lateral ventricle. In the present study, we investigated the relationship between neurogenesis in the amygdala and extinction of fear memory. Mice received 15 tone-footshock pairings. Twenty-four hours after training, the mice were given 15 tone-alone trials (extinction training) once per day for 7 days. Two hours before extinction training, the mice were injected intraperitoneally with 5-bromo-3-deoxyuridine (BrdU). BrdU-positive and NeuN-positive cells were analyzed 52 days after the training. A group of mice that received tone-footshock pairings but no extinction training served as controls (FC+No-Ext). The number of BrdU(+)/NeuN(+) cells was significantly higher in the extinction (FC+Ext) than in the FC+No-Ext mice. Proliferation inhibitor methylazoxymethanol acetate (MAM) or DNA synthesis inhibitor cytosine arabinoside (Ara-C) reduced neurogenesis and retarded extinction. Silencing Sonic hedgehog (Shh) gene with short hairpin interfering RNA (shRNA) by means of a retrovirus expression system to knockdown Shh specifically in the mitotic neurons reduced neurogenesis and retarded extinction. By contrast, over-expression of Shh increased neurogenesis and facilitated extinction. These results suggest that amygdala neurogenesis and Shh signaling are involved in the extinction of fear memory.
Collapse
Affiliation(s)
- Hui-Chi Hung
- Institute of Basic Medical Sciences and Department of Pharmacology, National Cheng-Kung University, Tainan 701, Taiwan
| | - Ya-Hsin Hsiao
- Institute of Basic Medical Sciences and Department of Pharmacology, National Cheng-Kung University, Tainan 701, Taiwan
| | - Po-Wu Gean
- Institute of Basic Medical Sciences and Department of Pharmacology, National Cheng-Kung University, Tainan 701, Taiwan.
| |
Collapse
|