1
|
Mukhtar AB, Morgan HJ, Gibbs A, Davies GE, Lovatt C, Patel GK. Targeting CD20-expressing malignant melanoma cells augments BRAF inhibitor killing. Br J Dermatol 2024; 190:729-739. [PMID: 38288865 DOI: 10.1093/bjd/ljad502] [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: 09/17/2022] [Revised: 12/08/2023] [Indexed: 04/19/2024]
Abstract
BACKGROUND Mutant BRAF targeted therapies remain a standard of care for the treatment of metastatic malignant melanoma (MM); however, high initial response rates are tempered by the persistence of residual MM cells that eventually lead to disease recurrence and mortality. As MM recurrence during targeted therapy can present with the simultaneous occurrence of multiple tumour nodules at the original body sites, we hypothesized the presence of an intrinsically resistant MM cell subpopulation. OBJECTIVES To identify an MM cell subpopulation that is intrinsically resistant to targeted therapy and possibly responsible for MM recurrence. METHODS Using melanoma cell lines, we defined culture conditions for the reproducible three-dimensional growth of melanospheres to investigate putative cancer stem cell populations. We undertook RNA sequencing and bioinformatic analysis to characterize cell populations between adherent and nonadherent culture, and cells expressing or not expressing CD20. Furthermore, we defined an in vitro assay to evaluate the killing of melanoma cancer stem cells as a therapeutic test using combination therapies targeting driver mutation and CD20. RESULTS We described the culture conditions that promote MM cells to form melanospheres with a reproducible colony-forming efficiency rate of 0.3-1.3%. RNA sequencing of melanosphere vs. conventional MM cell cultures (n = 6), irrespective of the BRAF mutation status, showed that melanosphere formation was associated with growth and differentiation transcriptional signatures resembling MM tumours. Importantly, melanosphere formation also led to the emergence of a CD20+ MM cell subpopulation, similar to that observed in primary human MM tumours. CD20+ MM cells were resistant to BRAF inhibitor therapy and, consistent with this finding, demonstrated a Forkhead box protein M1 transcriptomic profile (n = 6). Combining BRAF inhibitor and anti-CD20 antibody treatment led to the additional killing of previously resistant CD20+ BRAF mutant MM cells. CONCLUSIONS In patients with MM that harbour a CD20+ subpopulation, combined therapy with BRAF inhibitor and anti-CD20 antibody could potentially kill residual MM cells and prevent disease recurrence.
Collapse
Affiliation(s)
- Abdullahi B Mukhtar
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Huw J Morgan
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Alex Gibbs
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Gemma E Davies
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Charlotte Lovatt
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Girish K Patel
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
2
|
Zhang Y, Li S, Li L, Huang H, Fu Z, Hua Z. Bilirubin impairs neuritogenesis and synaptogenesis in NSPCs by downregulating NMDAR-CREB-BDNF signaling. In Vitro Cell Dev Biol Anim 2024; 60:161-171. [PMID: 38216855 DOI: 10.1007/s11626-023-00844-5] [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: 08/16/2023] [Accepted: 12/01/2023] [Indexed: 01/14/2024]
Abstract
Neonatal jaundice is one of the most common disorders in the first 2 wk after birth. Unconjugated bilirubin (UCB) is neurotoxic and can cause neurological dysfunction; however, the underlying mechanisms remain unclear. Neurogenesis, neuronal growth, and synaptogenesis are exuberant in the early postnatal stage. In this study, the impact of UCB on neuritogenesis and synaptogenesis in the early postnatal stage was evaluated both in vitro and in vivo. Primary culture neuronal stem and progenitor cells (NSPCs) were treated with UCB during differentiation, and then the neurite length and synapse puncta were measured. In the bilirubin encephalopathy (BE) animal model, DCX+-marked developing neurons were used to detect apical length and dendritic arborization. According to the data, UCB significantly reduced neurite length and synapse density, as well as decreased the apical dendrite length and dendritic arborization. Furthermore, the NMDAR subunit NR2B was downregulated in NSPCs, while pCREB expression in the hippocampus progressively decreased during disease progression in the BE model. Next, we tested the expression of NR2B, pCREB, mBDNF, and p-mTOR in NSPCs in vitro, and found that UCB treatment reduced the expression of these proteins. In summary, this suggests that UCB causes chronic neurological impairment and is related to the inhibition of NMDAR-CREB-BDNF signaling in NSPCs, which is associated with reduced neuritogenesis and synaptogenesis. This finding may inspire the development of novel pharmaceuticals and treatments.
Collapse
Affiliation(s)
- Yan Zhang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Siyu Li
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Ling Li
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Hongmei Huang
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Zhou Fu
- Department of Respiratory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Ziyu Hua
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
3
|
Marcellus KA, Bugiel S, Nunnikhoven A, Curran I, Gill SS. Polystyrene Nano- and Microplastic Particles Induce an Inflammatory Gene Expression Profile in Rat Neural Stem Cell-Derived Astrocytes In Vitro. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:429. [PMID: 38470760 DOI: 10.3390/nano14050429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024]
Abstract
Microplastics are considered an emerging environmental pollutant due to their ubiquitous presence in the environment. However, the potential impact of microplastics on human health warrants further research. Recent studies have reported neurobehavioral and neurotoxic effects in marine and rodent models; however, their impact on the underlying cellular physiology in mammals remains unclear. Herein, we exposed neural stem cells and neural stem cell-derived astrocytes, oligodendrocytes, and neurons to various sizes and concentrations of polystyrene nano- and microplastics. We investigated their cellular uptake, impact on cytotoxicity, and alteration of gene expression through transcriptome profiling. The cell type most affected by decreased viability were astrocytes after 7 days of repeated exposure. Transcriptional analysis showed that 1274 genes were differentially expressed in astrocytes exposed to 500 nm microplastics, but only 531 genes were altered in astrocytes exposed to 50 nm nanoplastics. Both canonical pathway and Kyoto Encyclopedia of Genes and Genomes analysis showed that upregulated pathways were involved in neuroinflammation, innate and adaptive immunity, cell migration, proliferation, extracellular matrix remodeling, and cytoskeleton structures. The downregulated pathways were involved in lipid metabolism, specifically fatty acid oxidation and cholesterol metabolism. Our results show that neural stem cell-derived astrocytes repeatedly exposed to nano- and microplastics for 7 days undergo changes that are hallmarks of astrogliosis.
Collapse
Affiliation(s)
- Kristen A Marcellus
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Steven Bugiel
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Andrée Nunnikhoven
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Ivan Curran
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Santokh S Gill
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| |
Collapse
|
4
|
Park HH, Kwon HS, Lee KY, Kim YE, Son JW, Choi NY, Han MH, Park DW, Kim S, Koh SH. GV1001 reduces neurodegeneration and prolongs lifespan in 3xTg-AD mouse model through anti-aging effects. Aging (Albany NY) 2024; 16:1983-2004. [PMID: 38301041 PMCID: PMC10911355 DOI: 10.18632/aging.205489] [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: 12/09/2022] [Accepted: 01/02/2024] [Indexed: 02/03/2024]
Abstract
GV1001, which mimics the activity of human telomerase reverse transcriptase, protects neural cells from amyloid beta (Aβ) toxicity and other stressors through extra-telomeric function, as noted in our prior in vitro studies. As per a recent phase II clinical trial, it improves cognitive function in patients with moderate to severe dementia. However, the underlying protective mechanisms remain unclear. This study aimed to investigate the effects of GV1001 on neurodegeneration, senescence, and survival in triple transgenic Alzheimer's disease (3xTg-AD) mice. GV1001 (1 mg/kg) was subcutaneously injected into old 3xTg-AD mice thrice a week until the endpoint for sacrifice, and survival was analysed. Magnetic resonance imaging (MRI) and Prussian blue staining (PBS) were performed to evaluate entry of GV1001 entrance into the brain. Diverse molecular studies were performed to investigate the effect of GV1001 on neurodegeneration and cellular senescence in AD model mice, with a particular focus on BACE, amyloid beta1-42 (Aβ1-42), phosphorylated tau, volume of dentate gyrus, β-galactosidase positive cells, telomere length, telomerase activity, and ageing-associated proteins. GV1001 crossed the blood-brain barrier, as confirmed by assessing the status of ferrocenecarboxylic acid-conjugated GV1001 using magnetic resonance imaging and PBS. GV1001 increased the survival of 3xTg-AD mice. It decreased BACE and Aβ1-42 levels, neurodegeneration (i.e., reduced CA1, CA3 and dentate gyrus volume, decreased levels of senescence-associated β-galactosidase positive cells, and increased telomere length and telomerase activity), and levels of ageing-associated proteins. We suggest that GV1001 exerts anti-ageing effects in 3xTg-AD mice by reducing neurodegeneration and senescence, which contributes to improved survival.
Collapse
Affiliation(s)
- Hyun-Hee Park
- Department of Neurology, Hanyang University Guri Hospital, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, Korea
| | - Hyuk Sung Kwon
- Department of Neurology, Hanyang University Guri Hospital, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, Korea
| | - Kyu-Yong Lee
- Department of Neurology, Hanyang University Guri Hospital, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, Korea
| | - Ye Eun Kim
- Department of Neurology, Hanyang University Guri Hospital, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, Korea
| | - Jeong-Woo Son
- Department of Neurology, Hanyang University Guri Hospital, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, Korea
| | - Na-Young Choi
- Department of Neurology, Hanyang University Guri Hospital, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, Korea
| | - Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, Korea
| | - Dong Woo Park
- Department of Radiology, Hanyang University Guri Hospital, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, Korea
| | | | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul 04763, Korea
| |
Collapse
|
5
|
Aksoy ZB, Akcali KC. A Three-Step Protocol to Differentiate iPSC into Colon Organoids. Methods Mol Biol 2024; 2835:59-67. [PMID: 39105906 DOI: 10.1007/978-1-0716-3995-5_6] [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: 08/07/2024]
Abstract
Organoids, three-dimensional, stem cell-based structures that mimic the cellular and functional architecture of tissues, have emerged as an innovative in vitro tool. They offer highly efficient models for studying both embryonic development and disease progression processes. Colon organoids can also be generated from biopsies obtained during a colonoscopy. However, the invasive nature of biopsy collection poses practical challenges and introduces biases when studying patients who are already afflicted. Therefore, the use of iPSC-derived colon organoids can be considered a more practical approach for researchers and patients alike. Numerous protocols have been published for generating colon organoids from iPSCs. While most of these protocols share a common developmental process, some are labor-intensive or require additional equipment. Taking these considerations into account, we present a cost-effective and straightforward yet functionally robust colon organoid protocol: (1) definitive endoderm differentiation, (2) hindgut endoderm differentiation, and (3) maturation of colon spheroids into mature organoids.
Collapse
Affiliation(s)
| | - Kamil Can Akcali
- Stem Cell Institute, Ankara University, Ankara, Turkey.
- Faculty of Medicine, Department of Biophysics, Ankara University, Ankara, Turkey.
| |
Collapse
|
6
|
Kameyama T, Miyata M, Shiotani H, Adachi J, Kakuta S, Uchiyama Y, Mizutani K, Takai Y. Heterogeneity of perivascular astrocyte endfeet depending on vascular regions in the mouse brain. iScience 2023; 26:108010. [PMID: 37829206 PMCID: PMC10565786 DOI: 10.1016/j.isci.2023.108010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/14/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Abstract
Astrocytes interact with not only synapses but also brain blood vessels through perivascular astrocyte endfeet (PV-AEF) to form the neurovascular unit (NVU). However, PV-AEF components have not been fully identified. Here, we biochemically isolated blood vessels from mouse brain homogenates and purified PV-AEF. The purified PV-AEF were observed in different sizes, similar to PV-AEF on brain blood vessels. Mass spectrometry analysis identified 9,762 proteins in the purified PV-AEF, including cell adhesion molecules, nectin-2δ, Kirrel2, and podoplanin. Immunofluorescence microscopic analysis revealed that nectin-2δ and podoplanin were concentrated mainly in arteries/arterioles and veins/venules of the mouse brain, whereas Kirrel2 was mainly in arteries/arterioles. Nectin-2α/δ, Kirrel2, and podoplanin were preferentially observed in large sizes of the purified PV-AEF. Furthermore, Kirrel2 potentially has cell adhesion activity of cultured astrocytes. Collectively, these results indicate that PV-AEF have heterogeneity in sizes and molecular components, implying different roles of PV-AEF in NVU function depending on vascular regions.
Collapse
Affiliation(s)
- Takeshi Kameyama
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Department of Immunology and Parasitology, Graduate School of Medicine, Tokushima University, Tokushima 770-8503, Japan
| | - Muneaki Miyata
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Division of Pathogenetic Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Hajime Shiotani
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Division of Pathogenetic Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
- Laboratory of Clinical and Analytical Chemistry, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Soichiro Kakuta
- Laboratory of Morphology and Image Analysis, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Department of Cellular Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yasuo Uchiyama
- Department of Cellular Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Kiyohito Mizutani
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
- Division of Pathogenetic Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe 650-0047, Japan
| |
Collapse
|
7
|
Kim JY, Hwang M, Choi NY, Koh SH. Inhibition of the NLRP3 Inflammasome Activation/Assembly through the Activation of the PI3K Pathway by Naloxone Protects Neural Stem Cells from Ischemic Condition. Mol Neurobiol 2023; 60:5330-5342. [PMID: 37300646 DOI: 10.1007/s12035-023-03418-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
Naloxone is a well-known opioid antagonist and has been suggested to have neuroprotective effects in cerebral ischemia. We investigated whether naloxone exhibits anti-inflammatory and neuroprotective effects in neural stem cells (NSCs) injured by oxygen-glucose deprivation (OGD), whether it affects the NOD-like receptor protein 3 (NLRP3) inflammasome activation/assembly, and whether the role of the phosphatidylinositol 3-kinase (PI3K) pathway is important in the control of NLRP3 inflammasome activation/assembly by naloxone. Primary cultured NSCs were subjected to OGD and treated with different concentrations of naloxone. Cell viability, proliferation, and the intracellular signaling proteins associated with the PI3K pathway and NLRP3 inflammasome activation/assembly were evaluated in OGD-injured NSCs. OGD significantly reduced survival, proliferation, and migration and increased apoptosis of NSCs. However, treatment with naloxone significantly restored survival, proliferation, and migration and decreased apoptosis of NSCs. Moreover, OGD markedly increased NLRP3 inflammasome activation/assembly and cleaved caspase-1 and interleukin-1β levels in NSCs, but naloxone significantly attenuated these effects. These neuroprotective and anti-inflammatory effects of naloxone were eliminated when cells were treated with PI3K inhibitors. Our results suggest that NLRP3 inflammasome is a potential therapeutic target and that naloxone reduces ischemic injury in NSCs by inhibiting NLRP3 inflammasome activation/assembly mediated by the activation of the PI3K signaling pathway.
Collapse
Affiliation(s)
- Ji Young Kim
- Department of Nuclear Medicine, Hanyang University College of Medicine, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do, 11923, Republic of Korea
| | - Mina Hwang
- Department of Neurology, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do, 11923, Republic of Korea
| | - Na-Young Choi
- Department of Neurology, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do, 11923, Republic of Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University College of Medicine, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do, 11923, Republic of Korea.
| |
Collapse
|
8
|
Bahia RK, Hao X, Hassam R, Cseh O, Bozek DA, Luchman HA, Weiss S. Epigenetic and molecular coordination between HDAC2 and SMAD3-SKI regulates essential brain tumour stem cell characteristics. Nat Commun 2023; 14:5051. [PMID: 37598220 PMCID: PMC10439933 DOI: 10.1038/s41467-023-40776-y] [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: 09/27/2022] [Accepted: 08/10/2023] [Indexed: 08/21/2023] Open
Abstract
Histone deacetylases are important epigenetic regulators that have been reported to play essential roles in cancer stem cell functions and are promising therapeutic targets in many cancers including glioblastoma. However, the functionally relevant roles of specific histone deacetylases, in the maintenance of key self-renewal and growth characteristics of brain tumour stem cell (BTSC) sub-populations of glioblastoma, remain to be fully resolved. Here, using pharmacological inhibition and genetic loss and gain of function approaches, we identify HDAC2 as the most relevant histone deacetylase for re-organization of chromatin accessibility resulting in maintenance of BTSC growth and self-renewal properties. Furthermore, its specific interaction with the transforming growth factor-β pathway related proteins, SMAD3 and SKI, is crucial for the maintenance of tumorigenic potential in BTSCs in vitro and in orthotopic xenograft models. Inhibition of HDAC2 activity and disruption of the coordinated mechanisms regulated by the HDAC2-SMAD3-SKI axis are thus promising therapeutic approaches for targeting BTSCs.
Collapse
Affiliation(s)
- Ravinder K Bahia
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Xiaoguang Hao
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Rozina Hassam
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Orsolya Cseh
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Danielle A Bozek
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - H Artee Luchman
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada.
| | - Samuel Weiss
- Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
9
|
Radoszkiewicz K, Hribljan V, Isakovic J, Mitrecic D, Sarnowska A. Critical points for optimizing long-term culture and neural differentiation capacity of rodent and human neural stem cells to facilitate translation into clinical settings. Exp Neurol 2023; 363:114353. [PMID: 36841464 DOI: 10.1016/j.expneurol.2023.114353] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/03/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023]
Abstract
Despite several decades of research on the nature and functional properties of neural stem cells, which brought great advances in regenerative medicine, there is still a plethora of ambiguous protocols and interpretations linked to their applications. Here, we present a whole spectrum of protocol elements that should be standardized in order to obtain viable cell cultures and facilitate their translation into clinical settings. Additionally, this review also presents outstanding limitations and possible problems to be encountered when dealing with protocol optimization. Most importantly, we also outline the critical points that should be considered before starting any experiments utilizing neural stem cells or interpreting their results.
Collapse
Affiliation(s)
- Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, 02-106 Warsaw, Poland
| | - Valentina Hribljan
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, Zagreb, Croatia
| | - Jasmina Isakovic
- Omnion Research International Ltd, Heinzelova 4, 10000 Zagreb, Croatia
| | - Dinko Mitrecic
- Laboratory for Stem Cells, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 12, Zagreb, Croatia
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawinskiego 5 Street, 02-106 Warsaw, Poland.
| |
Collapse
|
10
|
Nozawa O, Miyata M, Shiotani H, Kameyama T, Komaki R, Shimizu T, Kuriu T, Kashiwagi Y, Sato Y, Koebisu M, Aiba A, Okabe S, Mizutani K, Takai Y. Necl2/3-mediated mechanism for tripartite synapse formation. Development 2023; 150:285820. [PMID: 36458527 DOI: 10.1242/dev.200931] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022]
Abstract
Ramified, polarized protoplasmic astrocytes interact with synapses via perisynaptic astrocyte processes (PAPs) to form tripartite synapses. These astrocyte-synapse interactions mutually regulate their structures and functions. However, molecular mechanisms for tripartite synapse formation remain elusive. We developed an in vitro co-culture system for mouse astrocytes and neurons that induced astrocyte ramifications and PAP formation. Co-cultured neurons were required for astrocyte ramifications in a neuronal activity-dependent manner, and synaptically-released glutamate and activation of astrocytic mGluR5 metabotropic glutamate receptor were likely involved in astrocyte ramifications. Astrocytic Necl2 trans-interacted with axonal Necl3, inducing astrocyte-synapse interactions and astrocyte functional polarization by recruiting EAAT1/2 glutamate transporters and Kir4.1 K+ channel to the PAPs, without affecting astrocyte ramifications. This Necl2/3 trans-interaction increased functional synapse number. Thus, astrocytic Necl2, synaptically-released glutamate and axonal Necl3 cooperatively formed tripartite glutamatergic synapses in vitro. Studies on hippocampal mossy fiber synapses in Necl3 knockout and Necl2/3 double knockout mice confirmed these previously unreported mechanisms for astrocyte-synapse interactions and astrocyte functional polarization in vivo.
Collapse
Affiliation(s)
- Osamu Nozawa
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0047, Japan
| | - Muneaki Miyata
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0047, Japan
| | - Hajime Shiotani
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0047, Japan
| | - Takeshi Kameyama
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0047, Japan
| | - Ryouhei Komaki
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0047, Japan
| | - Tatsuhiro Shimizu
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0047, Japan
| | - Toshihiko Kuriu
- Osaka Medical and Pharmaceutical University, Research and Development Center, Takatsuki, Osaka 569-8686, Japan
| | - Yutaro Kashiwagi
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuka Sato
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Michinori Koebisu
- Section of Animal Research and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Atsu Aiba
- Section of Animal Research and Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Kiyohito Mizutani
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0047, Japan
| | - Yoshimi Takai
- Division of Pathogenetic Signaling, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
11
|
Redox-dependent Igfbp2 signaling controls Brca1 DNA damage response to govern neural stem cell fate. Nat Commun 2023; 14:444. [PMID: 36707536 PMCID: PMC9883463 DOI: 10.1038/s41467-023-36174-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/17/2023] [Indexed: 01/29/2023] Open
Abstract
Neural stem cell (NSC) maintenance and functions are regulated by reactive oxygen species (ROS). However, the mechanisms by which ROS control NSC behavior remain unclear. Here we report that ROS-dependent Igfbp2 signaling controls DNA repair pathways which balance NSC self-renewal and lineage commitment. Ncf1 or Igfbp2 deficiency constrains NSCs to a self-renewing state and prevents neurosphere formation. Ncf1-dependent oxidation of Igfbp2 promotes neurogenesis by NSCs in vitro and in vivo while repressing Brca1 DNA damage response genes and inducing DNA double-strand breaks (DDSBs). By contrast, Ncf1-/- and Igfbp2-/- NSCs favor the formation of oligodendrocytes in vitro and in vivo. Notably, transient repression of Brca1 DNA repair pathway genes induces DDSBs and is sufficient to rescue the ability of Ncf1-/- and Igfbp2-/- NSCs to lineage-commit to form neurospheres and neurons. NSC lineage commitment is dependent on the oxidizable cysteine-43 residue of Igfbp2. Our study highlights the role of DNA damage/repair in orchestrating NSC fate decisions downstream of redox-regulated Igfbp2.
Collapse
|
12
|
Dittmann NL, Torabi P, Watson AES, Yuzwa SA, Voronova A. Culture Protocol and Transcriptomic Analysis of Murine SVZ NPCs and OPCs. Stem Cell Rev Rep 2023; 19:983-1000. [PMID: 36617597 DOI: 10.1007/s12015-022-10492-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 01/10/2023]
Abstract
The mammalian adult brain contains two neural stem and precursor (NPC) niches: the subventricular zone [SVZ] lining the lateral ventricles and the subgranular zone [SGZ] in the hippocampus. From these, SVZ NPCs represent the largest NPC pool. While SGZ NPCs typically only produce neurons and astrocytes, SVZ NPCs produce neurons, astrocytes and oligodendrocytes throughout life. Of particular importance is the generation and replacement of oligodendrocytes, the only myelinating cells of the central nervous system (CNS). SVZ NPCs contribute to myelination by regenerating the parenchymal oligodendrocyte precursor cell (OPC) pool and by differentiating into oligodendrocytes in the developing and demyelinated brain. The neurosphere assay has been widely adopted by the scientific community to facilitate the study of NPCs in vitro. Here, we present a streamlined protocol for culturing postnatal and adult SVZ NPCs and OPCs from primary neurosphere cells. We characterize the purity and differentiation potential as well as provide RNA-sequencing profiles of postnatal SVZ NPCs, postnatal SVZ OPCs and adult SVZ NPCs. We show that primary neurospheres cells generated from postnatal and adult SVZ differentiate into neurons, astrocytes and oligodendrocytes concurrently and at comparable levels. SVZ OPCs are generated by subjecting primary neurosphere cells to OPC growth factors fibroblast growth factor (FGF) and platelet-derived growth factor-AA (PDGF-AA). We further show SVZ OPCs can differentiate into oligodendrocytes in the absence and presence of thyroid hormone T3. Transcriptomic analysis confirmed the identities of each cell population and revealed novel immune and signalling pathways expressed in an age and cell type specific manner.
Collapse
Affiliation(s)
- Nicole L Dittmann
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.,Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Pouria Torabi
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Adrianne E S Watson
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada. .,Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada. .,Women and Children's Health Research Institute5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, Alberta, T6G 1C9, Canada. .,Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada. .,Multiple Sclerosis Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
13
|
Claxton M, Pulix M, Seah MKY, Bernardo R, Zhou P, Aljuraysi S, Liloglou T, Arnaud P, Kelsey G, Messerschmidt DM, Plagge A. Variable allelic expression of imprinted genes at the Peg13, Trappc9, Ago2 cluster in single neural cells. Front Cell Dev Biol 2022; 10:1022422. [PMID: 36313557 PMCID: PMC9596773 DOI: 10.3389/fcell.2022.1022422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Genomic imprinting is an epigenetic process through which genes are expressed in a parent-of-origin specific manner resulting in mono-allelic or strongly biased expression of one allele. For some genes, imprinted expression may be tissue-specific and reliant on CTCF-influenced enhancer-promoter interactions. The Peg13 imprinting cluster is associated with neurodevelopmental disorders and comprises canonical imprinted genes, which are conserved between mouse and human, as well as brain-specific imprinted genes in mouse. The latter consist of Trappc9, Chrac1 and Ago2, which have a maternal allelic expression bias of ∼75% in brain. Findings of such allelic expression biases on the tissue level raise the question of how they are reflected in individual cells and whether there is variability and mosaicism in allelic expression between individual cells of the tissue. Here we show that Trappc9 and Ago2 are not imprinted in hippocampus-derived neural stem cells (neurospheres), while Peg13 retains its strong bias of paternal allele expression. Upon analysis of single neural stem cells and in vitro differentiated neurons, we find not uniform, but variable states of allelic expression, especially for Trappc9 and Ago2. These ranged from mono-allelic paternal to equal bi-allelic to mono-allelic maternal, including biased bi-allelic transcriptional states. Even Peg13 expression deviated from its expected paternal allele bias in a small number of cells. Although the cell populations consisted of a mosaic of cells with different allelic expression states, as a whole they reflected bulk tissue data. Furthermore, in an attempt to identify potential brain-specific regulatory elements across the Trappc9 locus, we demonstrate tissue-specific and general silencer activities, which might contribute to the regulation of its imprinted expression bias.
Collapse
Affiliation(s)
- Michael Claxton
- Department of Molecular Physiology and Cell Signaling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Michela Pulix
- Department of Molecular Physiology and Cell Signaling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Michelle K. Y. Seah
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ralph Bernardo
- Department of Molecular Physiology and Cell Signaling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Peng Zhou
- Department of Molecular Physiology and Cell Signaling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Sultan Aljuraysi
- Department of Molecular Physiology and Cell Signaling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Triantafillos Liloglou
- Faculty of Health, Social Care and Medicine, Edge Hill University, Ormskirk, Lancashire, United Kingdom
| | - Philippe Arnaud
- Université Clermont Auvergne, CNRS, Inserm, GReD, Clermont-Ferrand, France
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories, Cambridge, United Kingdom
| | - Daniel M. Messerschmidt
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Antonius Plagge
- Department of Molecular Physiology and Cell Signaling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
14
|
Zhang YF, Li XX, Cao XL, Ji CC, Gao XY, Gao D, Han H, Yu F, Zheng MH. MicroRNA-582-5p Contributes to the Maintenance of Neural Stem Cells Through Inhibiting Secretory Protein FAM19A1. Front Cell Neurosci 2022; 16:866020. [PMID: 35685988 PMCID: PMC9171424 DOI: 10.3389/fncel.2022.866020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/04/2022] [Indexed: 11/15/2022] Open
Abstract
Epigenetic regulations on the maintenance of neural stem cells (NSCs) are complicated and far from been fully understood. Our previous findings have shown that after blocking Notch signaling in NSCs in vivo, the stemness of NSCs decreases, accompanied by the downregulated expression of miR-582-5p. In the current study, we further investigated the function and mechanism of miR-582-5p in the maintenance of NSCs in vitro and in vivo. After transfecting a mimic of miR-582-5p, the formation of neurospheres and proliferation of NSCs and intermediate progenitor cells (NS/PCs) were enhanced, and the expression of stemness markers such as Sox2, Nestin, and Pax6 also increased. The results were reversed after transfection of an inhibitor of miR-582-5p. We further generated miR-582 knock-out (KO) mice to investigate its function in vivo, and we found that the number of NSCs in the subventricular zone (SVZ) region decreased and the number of neuroblasts increased in miR-582 deficient mice, indicating reduced stemness and enhanced neurogenesis of NSCs. Moreover, RNA-sequencing and molecular biological analysis revealed that miR-582-5p regulates the stemness and proliferation of NSCs by inhibiting secretory protein FAM19A1. In summary, our research uncovered a new epigenetic mechanism that regulates the maintenance of NSCs, therefore providing novel targets to amplify NSCs in vitro and to promote neurogenesis in vivo during brain pathology and aging.
Collapse
Affiliation(s)
- Yu-Fei Zhang
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Life Sciences, Northwest A&F University, Yangling, China
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Xin-Xin Li
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Xi’ an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Xiu-Li Cao
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
| | - Chen-Chen Ji
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Xiang-Yu Gao
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Dan Gao
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
| | - Hua Han
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- *Correspondence: Hua Han,
| | - Fei Yu
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Life Sciences, Northwest A&F University, Yangling, China
- Fei Yu,
| | - Min-Hua Zheng
- Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi’an, China
- Min-Hua Zheng,
| |
Collapse
|
15
|
Physiological Electric Field: A Potential Construction Regulator of Human Brain Organoids. Int J Mol Sci 2022; 23:ijms23073877. [PMID: 35409232 PMCID: PMC8999182 DOI: 10.3390/ijms23073877] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Brain organoids can reproduce the regional three-dimensional (3D) tissue structure of human brains, following the in vivo developmental trajectory at the cellular level; therefore, they are considered to present one of the best brain simulation model systems. By briefly summarizing the latest research concerning brain organoid construction methods, the basic principles, and challenges, this review intends to identify the potential role of the physiological electric field (EF) in the construction of brain organoids because of its important regulatory function in neurogenesis. EFs could initiate neural tissue formation, inducing the neuronal differentiation of NSCs, both of which capabilities make it an important element of the in vitro construction of brain organoids. More importantly, by adjusting the stimulation protocol and special/temporal distributions of EFs, neural organoids might be created following a predesigned 3D framework, particularly a specific neural network, because this promotes the orderly growth of neural processes, coordinate neuronal migration and maturation, and stimulate synapse and myelin sheath formation. Thus, the application of EF for constructing brain organoids in a3D matrix could be a promising future direction in neural tissue engineering.
Collapse
|
16
|
Liu H, Dai Q, Yang J, Zhang Y, Zhang B, Zhong L. Zuogui Pill Attenuates Neuroinflammation and Improves Cognitive Function in Cerebral Ischemia Reperfusion-Injured Rats. Neuroimmunomodulation 2022; 29:143-150. [PMID: 34736255 DOI: 10.1159/000519010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/10/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Cerebral ischemia and reperfusion (CI/R) injury is a devasting cerebrovascular disease, accompanied with ischemia stroke, cerebral infarction. Zuogui Pill (ZGP), as a Chinese traditional medicine, is proved to be effective in many diseases and cancers. Our study aimed to detect the roles of ZGP in CI/R injury. METHODS Neural stem cells were isolated from rats and induced by oxygen and glucose deprivation and recovery. CCK-8 and flow cytometry were applied to assess the function of ZGP on cell viability and apoptosis. Rat CI/R injury models were established by the middle cerebral artery occlusion and reperfusion. The function of ZGP on CI/R injury was identified via evaluating modified neurological severity score, infarct area, and cognitive impairment. RESULTS Compared to the control, the cell viability was obviously decreased in the oxygen and glucose deprivation and recovery (OGD/R) group, while the adverse influence on cells was reversed by cultured plus 10% ZGP serum. Consistently, ZGP attenuated the influence of OGD/R on cell apoptosis. More importantly, ZGP could alleviate CI/R injury of rats by reducing neurological damage and infarct area and promoting cognitive function. CONCLUSION This study provided protective roles of ZGP on cell viability and apoptosis induced by OGD/R. In addition, ZGP played protective roles on neuroinflammation and cognitive function in rats.
Collapse
Affiliation(s)
- Hong Liu
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
- Postdoctoral Program, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiaomei Dai
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Yang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuwei Zhang
- Department of Physiology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bo Zhang
- Department of Neurobiology, Research Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lili Zhong
- Department of Pathology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
17
|
Mirzaei R, Gordon A, Zemp FJ, Kumar M, Sarkar S, Luchman HA, Bellail AC, Hao C, Mahoney DJ, Dunn JF, Bose P, Yong VW. PD-1 independent of PD-L1 ligation promotes glioblastoma growth through the NFκB pathway. SCIENCE ADVANCES 2021; 7:eabh2148. [PMID: 34739319 PMCID: PMC8570610 DOI: 10.1126/sciadv.abh2148] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Brain tumor–initiating cells (BTICs) drive glioblastoma growth through not fully understood mechanisms. Here, we found that about 8% of cells within the human glioblastoma microenvironment coexpress programmed cell death 1 (PD-1) and BTIC marker. Gain- or loss-of-function studies revealed that tumor-intrinsic PD-1 promoted proliferation and self-renewal of BTICs. Phosphorylation of tyrosines within the cytoplasmic tail of PD-1 recruited Src homology 2–containing phosphatase 2 and activated the nuclear factor kB in BTICs. Notably, the tumor-intrinsic promoting effects of PD-1 did not require programmed cell death ligand 1(PD-L1) ligation; thus, the therapeutic antibodies inhibiting PD-1/PD-L1 interaction could not overcome the growth advantage of PD-1 in BTICs. Last, BTIC-intrinsic PD-1 accelerated intracranial tumor growth, and this occurred in mice lacking T and B cells. These findings point to a critical role for PD-1 in BTICs and uncover a nonimmune resistance mechanism of patients with glioblastoma to PD-1– or PD-L1–blocking therapies.
Collapse
Affiliation(s)
- Reza Mirzaei
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ashley Gordon
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Franz J. Zemp
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mehul Kumar
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Susobhan Sarkar
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - H. Artee Luchman
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Anita C. Bellail
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chunhai Hao
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Douglas J. Mahoney
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jeff F. Dunn
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
| | - Pinaki Bose
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
- Department of Surgery, University of Calgary, Calgary, Alberta, Canada
| | - V. Wee Yong
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
- Corresponding author.
| |
Collapse
|
18
|
Liu Y, Wu X, Du D, Liu J, Zhang W, Gao Y, Zhang H. p53 Inhibition Provides a Pivotal Protective Effect against Cerebral Ischemia-Reperfusion Injury via the Wnt Signaling Pathway. Cerebrovasc Dis 2021; 50:682-690. [PMID: 34340236 DOI: 10.1159/000516889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/26/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Cerebral ischemia-reperfusion injury enhances brain injury and increases its morbidity and mortality. The purpose of our study was to further explore the specific pathogenesis of cerebral ischemia disease by studying the role of p53 in cerebral ischemia-reperfusion injury and its mechanism to provide a new target for the treatment of cerebral ischemia. METHODS Middle cerebral artery occlusion (MCAo) was established in rats. The changes in p53 and apoptotic proteins in the rat model were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. The effects of p53 inhibitors on cerebral is-chemia-reperfusion injury in rats were evaluated by modified neurological severity score (mNSS) and infarct area. Subsequently, neural stem cells (NSCs) were isolated and cultured in vitro, and oxygen and glucose deprivation (OGD) was induced to establish an in vitro ischemia-reperfusion injury model. Cell viability and migration were detected by CCK-8 and transwell assays. Apoptosis of NSCs was detected by flow cytometry. Finally, protein expression in the Wnt pathway activated by p53 was detected by Western blotting. RESULTS Compared with the sham group, p53 levels, mNSS, cerebral infarction area, and apoptosis were significantly increased in the MCAo group (p < 0.05). When the p53 inhibitor PFT-α was injected, the increase in these levels was reversed. Also, the viability and migration of cells decreased and apo-ptosis increased in the in vitro OGD model, whereas the viability, migration, and apoptosis were significantly reversed after the addition of p53 inhibitors (p < 0.05). Finally, p53 induced Wnt signaling pathway proteins β-catenin and cyclin D1 decrease in the MCAo group, while p53 inhibitors reversed their inhibitory effect on the Wnt signaling pathway. CONCLUSION We confirmed in vivo and in vitro that inhibition of p53 has a protective effect on the cerebral ischemia-reperfusion injury, which may be related to the activation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Yanwei Liu
- Department of Emergency Internal Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinning Wu
- Department of Cardiovascular Medicine, People's Hospital of Rizhao, Rizhao, China
| | - Deyong Du
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, China
| | - Jing Liu
- Bincheng Municipal Hospital, Binzhou, China
| | - Wensheng Zhang
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, China
| | - Yang Gao
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, China
| | - Haitao Zhang
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
19
|
Wu X, Shen Q, Zhang Z, Zhang D, Gu Y, Xing D. Photoactivation of TGFβ/SMAD signaling pathway ameliorates adult hippocampal neurogenesis in Alzheimer's disease model. Stem Cell Res Ther 2021; 12:345. [PMID: 34116709 PMCID: PMC8196501 DOI: 10.1186/s13287-021-02399-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Background Adult hippocampal neurogenesis (AHN) is restricted under the pathological conditions of neurodegenerative diseases, especially in Alzheimer’s disease (AD). The drop of AHN reduces neural circuit plasticity, resulting in the decrease of the generation of newborn neurons in dentate gyrus (DG), which makes it difficult to recover from learning/memory dysfunction in AD, therefore, it is imperative to find a therapeutic strategy to promote neurogenesis and clarify its underlying mechanism involved. Methods Amyloid precursor protein/presenilin 1 (APP/PS1) mice were treated with photobiomodulation therapy (PBMT) for 0.1 mW/mm2 per day in the dark for 1 month (10 min for each day). The neural stem cells (NSCs) were isolated from hippocampus of APP/PS1 transgenic mice at E14, and the cells were treated with PBMT for 0.667 mW/mm2 in the dark (5 min for each time). Results In this study, photobiomodulation therapy (PBMT) is found to promote AHN in APP/PS1 mice. The latent transforming growth factor-β1 (LTGFβ1) was activated in vitro and in vivo during PBMT-induced AHN, which promoted the differentiation of hippocampal APP/PS1 NSCs into newborn neurons. In particular, behavioral experiments showed that PBMT enhanced the spatial learning/memory ability of APP/PS1 mice. Mechanistically, PBMT-stimulated reactive oxygen species (ROS) activates TGFβ/Smad signaling pathway to increase the interaction of the transcription factors Smad2/3 with Smad4 and competitively reduce the association of Smad1/5/9 with Smad4, thereby significantly upregulating the expression of doublecortin (Dcx)/neuronal class-III β-tubulin (Tuj1) and downregulating the expression of glial fibrillary acidic protein (GFAP). These in vitro effects were abrogated when eliminating ROS. Furthermore, specific inhibition of TGFβ receptor I (TGFβR I) attenuates the DNA-binding efficiency of Smad2/3 to the Dcx promotor triggered by PBMT. Conclusion Our study demonstrates that PBMT, as a viable therapeutic strategy, directs the adult hippocampal APP/PS1 NSCs differentiate towards neurons, which has great potential value for ameliorating the drop of AHN in Alzheimer’s disease mice. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02399-2.
Collapse
Affiliation(s)
- Xiaolei Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Qi Shen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Zhan Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Di Zhang
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Ying Gu
- Department of Laser Medicine, First Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China. .,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
20
|
Carroll JA, Foliaki ST, Haigh CL. A 3D cell culture approach for studying neuroinflammation. J Neurosci Methods 2021; 358:109201. [PMID: 33932455 DOI: 10.1016/j.jneumeth.2021.109201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/13/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neurodegenerative diseases are highly complex making them challenging to model in cell culture. All cell types of the brain have been implicated as exerting an effect on pathogenesis, and disease progression is likely influenced by the cross-talk between the different cell types. Sophisticated investigation of the cellular level consequences of cross-talk between different cells types requires three-dimensional (3D) co-culture systems. NEW METHOD Murine neural stem cells were differentiated into mixed-neuronal lineage populations in 3D culture. By seeding these differentiated cultures with microglia from adult brain, we have generated a 3D ex-vivo model of murine brain tissue populated with microglia. RESULTS Monitoring the infiltration of GFP-expressing microglia into the 3D neuronal lineage cultures showed population throughout the tissue and assumption of ramified homeostatic morphology by the microglia. The co-cultures showed good longevity and were functionally responsive to external stimuli. COMPARISON WITH EXISTING METHODS We have previously used 2-dimensional adhered cultures to model cell-cell interactions between microglia and neuronal lineage cells. While the microglia integrate well into these cultures and demonstrate inter-cellular cross-talk, it is known that adhered culture can change their activation state and therefore a 3D system better represents communication throughout a network of neuronal and support cells. CONCLUSIONS Our system offers a straight-forward and time effective way to model 3D mouse brain tissue that is responsive to external neuroinflammatory stimulus. It not only allows inter-cellular interactions to be studied in live tissue but additionally permits study of changes within any available mouse genotype.
Collapse
Affiliation(s)
- James A Carroll
- TSE/Prion and Retroviral Pathogenesis Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - Simote T Foliaki
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - Cathryn L Haigh
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA.
| |
Collapse
|
21
|
Kwon HS, Ha J, Kim JY, Park HH, Lee EH, Choi H, Lee KY, Lee YJ, Koh SH. Telmisartan Inhibits the NLRP3 Inflammasome by Activating the PI3K Pathway in Neural Stem Cells Injured by Oxygen-Glucose Deprivation. Mol Neurobiol 2021; 58:1806-1818. [PMID: 33404978 DOI: 10.1007/s12035-020-02253-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 12/08/2020] [Indexed: 01/01/2023]
Abstract
Angiotensin II receptor blockers (ARBs) have been shown to exert neuroprotective effects by suppressing inflammatory and apoptotic responses. In the present study, the effects of the ARB telmisartan on the NLRP3 inflammasome induced by oxygen-glucose deprivation (OGD) in neural stem cells (NSCs) were investigated, as well as their possible association with the activation of the PI3K pathway. Cultured NSCs were treated with different concentrations of telmisartan and subjected to various durations of OGD. Cell counting, lactate dehydrogenase, bromodeoxyuridine, and colony-forming unit assays were performed to measure cell viability and proliferation. In addition, the activity of intracellular signaling pathways associated with the PI3K pathway and NLRP3 inflammasome was evaluated. Telmisartan alone did not affect NSCs up to a concentration of 10 μM under normal conditions but showed toxicity at a concentration of 100 μM. Moreover, OGD reduced the viability of NSCs in a time-dependent manner. Nevertheless, treatment with telmisartan increased the viability and proliferation of OGD-injured NSCs. Furthermore, telmisartan promoted the expression of survival-related proteins and mRNA while inhibiting the expression of death-related proteins induced by OGD. In particular, telmisartan attenuated OGD-dependent expression of the NLRP3 inflammasome and its related signaling proteins. These beneficial effects of telmisartan were blocked by a PI3K inhibitor. Together, these results indicate that telmisartan attenuated the activation of the NLRP3 inflammasome by triggering the PI3K pathway, thereby contributing to neuroprotection.
Collapse
Affiliation(s)
- Hyuk Sung Kwon
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Jungsoon Ha
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
- GemVax & Kael Co., Ltd, Seongnam-si, Republic of Korea
| | - Ji Young Kim
- Department of Nuclear Medicine, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hyun-Hee Park
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Eun-Hye Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Republic of Korea
| | - Hojin Choi
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Kyu-Yong Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Young Joo Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri, 11923, South Korea.
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Mastrototaro G, Zaghi M, Massimino L, Moneta M, Mohammadi N, Banfi F, Bellini E, Indrigo M, Fagnocchi G, Bagliani A, Taverna S, Rohm M, Herzig S, Sessa A. TBL1XR1 Ensures Balanced Neural Development Through NCOR Complex-Mediated Regulation of the MAPK Pathway. Front Cell Dev Biol 2021; 9:641410. [PMID: 33708771 PMCID: PMC7940385 DOI: 10.3389/fcell.2021.641410] [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: 12/14/2020] [Accepted: 02/04/2021] [Indexed: 12/19/2022] Open
Abstract
TBL1XR1 gene is associated with multiple developmental disorders presenting several neurological aspects. The relative protein is involved in the modulation of important cellular pathways and master regulators of transcriptional output, including nuclear receptor repressors, Wnt signaling, and MECP2 protein. However, TBL1XR1 mutations (including complete loss of its functions) have not been experimentally studied in a neurological context, leaving a knowledge gap in the mechanisms at the basis of the diseases. Here, we show that Tbl1xr1 knock-out mice exhibit behavioral and neuronal abnormalities. Either the absence of TBL1XR1 or its point mutations interfering with stability/regulation of NCOR complex induced decreased proliferation and increased differentiation in neural progenitors. We suggest that this developmental unbalance is due to a failure in the regulation of the MAPK cascade. Taken together, our results broaden the molecular and functional aftermath of TBL1XR1 deficiency associated with human disorders.
Collapse
Affiliation(s)
- Giuseppina Mastrototaro
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mattia Zaghi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Moneta
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Neda Mohammadi
- Neurimmunology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Banfi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Edoardo Bellini
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marzia Indrigo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Fagnocchi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Bagliani
- Medical Oncology Unit, ASST Ovest Milanese, Legnano Hospital, Legnano, Italy
| | - Stefano Taverna
- Neurimmunology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Rohm
- Institute for Diabetes and Cancer IDC, Helmholtz Center, Munich, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.,Medical Faculty, Technical University Munich, Munich, Germany.,German Center for Diabetes Research, Oberschleissheim, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer IDC, Helmholtz Center, Munich, Germany.,Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany.,Medical Faculty, Technical University Munich, Munich, Germany.,German Center for Diabetes Research, Oberschleissheim, Germany
| | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
23
|
Song D, Chen Y, Chen C, Chen L, Cheng O. GABA B receptor antagonist promotes hippocampal neurogenesis and facilitates cognitive function recovery following acute cerebral ischemia in mice. Stem Cell Res Ther 2021; 12:22. [PMID: 33413637 PMCID: PMC7792056 DOI: 10.1186/s13287-020-02059-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/27/2020] [Indexed: 02/02/2023] Open
Abstract
PURPOSE AND BACKGROUND Previous studies have suggested that promoting endogenous neurogenesis has great significance for the recovery of cognitive dysfunction caused by cerebral ischemia (CI). Pharmacological inhibition of GABAB receptor can enhance neurogenesis in adult healthy and depressed mice. In the study, we intended to investigate the effects of GABAB receptor antagonists on cognitive function and hippocampal neurogenesis in mice following CI. METHODS Adult mice were subjected to bilateral common carotid artery occlusion (BCCAO) for 20 min to induce CI and treated with CGP52432 (antagonist of GABAB receptor, CGP, 10 mg/kg intraperitoneal injection) starting 24 h after CI. The Morris water maze test was performed to test spatial learning and memory at day 28. Immunofluorescence was applied to detect neurogenesis in the DG region at day 14 and 28. In in vitro experiments, cell proliferation was detected by CCK8 and immunofluorescence, and the expression of cAMP/CREB signaling pathway-related proteins was detected by ELISA assay and Western blot. RESULTS CGP significantly improved spatial learning and memory disorders caused by CI, and it enhanced the proliferation of neural stem cells (NSCs), the number of immature neurons, and the differentiation from newborn cells to neurons. In vitro experiments further confirmed that CGP dose-dependently enhanced the cell viability of NSCs, and immunofluorescence staining showed that CGP promoted the proliferation of NSCs. In addition, treatment with CGP increased the expression of cAMP, PKA, and pCREB in cultured NSCs. CONCLUSION Inhibition of GABAB receptor can effectively promote hippocampal neurogenesis and improve spatial learning and memory in adult mice following CI.
Collapse
Affiliation(s)
- Dan Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yaohua Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Cheng Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lili Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
24
|
Glia-Like Cells from Human Mesenchymal Stem Cells Protect Neural Stem Cells in an In Vitro Model of Alzheimer's Disease by Reducing NLRP-3 Inflammasome. Dement Neurocogn Disord 2020; 20:1-8. [PMID: 33552214 PMCID: PMC7847801 DOI: 10.12779/dnd.2021.20.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 01/22/2023] Open
Abstract
Background and Purpose Neural stem cells (NSCs) have the ability to regenerate, proliferate, and differentiate, enabling them to play important roles in the recovery of the damaged nervous system. However, in neurodegenerative diseases such as Alzheimer's disease (AD), the NSCs are damaged as well. Glia-like cells from human mesenchymal stem cells (ghMSCs) are functionally enhanced adult stem cells. In the present study, we investigated whether ghMSCs could protect NSCs from amyloid beta (Aβ)-mediated toxicity. Methods Rat NSCs were obtained from E13–14 fetal rat cortices. NSCs were seeded in pre-coated plates, and the next day, cells were simultaneously treated with 20 μM Aβ and 0.4 μm pore insert well-seeded ghMSCs. After 48 hours of co-treatment, cell viability and proliferation were evaluated. After 2 hours of co-treatment, western blotting was performed to measure inflammasome-related factors, such as NOD-like receptor family pyrin domain containing 3, caspase-1, and interleukin-1β. Results The results showed that ghMSCs increased viability and proliferation and reduced the toxicity of NSCs injured by Aβ by reducing the NRLP3 inflammasome activation of NSCs induced by Aβ. Conclusions In this study, we confirmed that ghMSCs could protect NSCs in an in vitro model of AD through the regulation of inflammatory response.
Collapse
|
25
|
Zhou H, Yang H, Lu L, Li X, Pan B, Fu Z, Chu T, Liu J, Kang Y, Liu L, Ning G, Ding W, Wu P, Kong X, Feng S. A modified protocol for the isolation, culture, and cryopreservation of rat embryonic neural stem cells. Exp Ther Med 2020; 20:156. [PMID: 33093894 DOI: 10.3892/etm.2020.9285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 05/16/2019] [Indexed: 11/05/2022] Open
Abstract
Neural stem cells (NSCs) are characterized by their potential for self-renewal and ability to differentiate into neurons, astrocytes, and oligodendrocytes. They are of great value to scientific studies and clinical applications. Culturing NSCs in vitro is important for characterizing their properties under controlled environmental conditions that may be modified and monitored accurately. The present study explored a modified, detailed and efficient protocol for the isolation, culture and cryopreservation of rat embryonic NSCs. In particular, the viability, nestin expression, and self-renewal and multi-differentiation capabilities of NSCs cryopreserved for various periods of time (7 days, or 1, 6 or 12 months) were characterized and compared. Rat embryonic NSCs were successfully obtained and maintained their self-renewal and multipotent differentiation capabilities even following long-term cryopreservation (for up to 12 months).
Collapse
Affiliation(s)
- Hengxing Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Huan Yang
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Lu Lu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Xueying Li
- Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Department of Immunology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Bin Pan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Zheng Fu
- Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Department of Immunology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Tianci Chu
- Department of Pediatrics, Kosair Children's Hospital Research Institute, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Jun Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Yi Kang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Lu Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Wenyuan Ding
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Ping Wu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1043, USA
| | - Xiaohong Kong
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| |
Collapse
|
26
|
MicroRNAs are indispensable for the proliferation and differentiation of adult neural progenitor cells in mice. Biochem Biophys Res Commun 2020; 530:209-214. [PMID: 32828287 DOI: 10.1016/j.bbrc.2020.06.143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 06/26/2020] [Indexed: 01/18/2023]
Abstract
More than two decades after the discovery of adult neurogenesis in humans, researchers still struggle to elucidate the underlying transcriptional and post-transcriptional mechanisms. RNA interference is a crucially important process in the central nervous system, and its role in adult neurogenesis is poorly understood. In this work, we address the role of Dicer-dependent microRNA biogenesis in neuronal differentiation of adult neural stem cells within the subventricular zone of the mouse brain. Loss of the Dicer1 gene in the tailless (Tlx)-positive cells did not cause the decline in their numbers, but severely affected differentiation. Thus, our findings identify yet another phenomenon associated with microRNA pathway deregulation in adult neural stem cells which might be of relevance both for neuroscience and clinical practice.
Collapse
|
27
|
Zhang L, Yu H, Yuan Y, Yu JS, Lou Z, Xue Y, Liu Y. The necessity for standardization of glioma stem cell culture: a systematic review. Stem Cell Res Ther 2020; 11:84. [PMID: 32102678 PMCID: PMC7045630 DOI: 10.1186/s13287-020-01589-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/15/2019] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The cancer stem cell hypothesis is an old idea which has been revived in recent years for many cancers, including gliomas. However, this concept has become controversial due to a series of studies with conflicting results. METHODS A systematic literature search was conducted in PubMed and the Web of Science database to analyze studies using serum-free medium and its components in glioma stem cells, glioma stem-like cells, glioma-initiating cells, or glioma neurosphere cultures. All the studies reviewed were published between 1970 and 2019. We found that no standardized culture method was used, and the data were incomparable due to differing culture conditions and the use of media with different components. CONCLUSIONS Here, we review the most commonly used serum-free media and added components for glioma stem cell culture while highlighting the function of each component used in the media. We emphasize the necessity for standardization of glioma stem cell culture and propose a standard culture medium to prevent bias in glioma stem cell research.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China.,Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, USA.,Department of Oncology, Mayo Clinic, Rochester, USA
| | - Hongwei Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China
| | - Yuhui Yuan
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China
| | - John S Yu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, Rochester, USA
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China.
| |
Collapse
|
28
|
Zhang S, Sun P, Lin K, Chan FHL, Gao Q, Lau WF, Roy VAL, Zhang H, Lai KWC, Huang Z, Yung KKL. Extracellular Nanomatrix-Induced Self-Organization of Neural Stem Cells into Miniature Substantia Nigra-Like Structures with Therapeutic Effects on Parkinsonian Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901822. [PMID: 31871862 PMCID: PMC6918115 DOI: 10.1002/advs.201901822] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/29/2019] [Indexed: 05/14/2023]
Abstract
Substantia nigra (SN) is a complex and critical region of the brain wherein Parkinson's disease (PD) arises from the degeneration of dopaminergic neurons. Miniature SN-like structures (mini-SNLSs) constructed from novel combination of nanomaterials and cell technologies exhibit promise as potentially curative cell therapies for PD. In this work, a rapid self-organization of mini-SNLS, with an organizational structure and neuronal identities similar to those of the SN in vivo, is achieved by differentiating neural stem cells in vitro on biocompatible silica nanozigzags (NZs) sculptured by glancing angle deposition, without traditional chemical growth factors. The differentiated neurons exhibit electrophysiological activity in vitro. Diverse physical cues and signaling pathways that are determined by the nanomatrices and lead to the self-organization of the mini-SNLSs are clarified and elucidated. In vivo, transplantation of the neurons from a mini-SNLS results in an early and progressive amelioration of PD in rats. The sculptured medical device reported here enables the rapid and specific self-organization of region-specific and functional brain-like structures without an undesirable prognosis. This development provides promising and significant insights into the screening of potentially curative drugs and cell therapies for PD.
Collapse
Affiliation(s)
- Shiqing Zhang
- Department of BiologyHong Kong Baptist University (HKBU)Kowloon TongKowloonHong Kong SAR China
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
- HKBU Institute of Research and Continuing Education, 9FThe Industrialization Complex of Shenzhen Virtual University ParkNo. 2 Yuexing 3rd Road, South Zone, Hi‐tech Industrial Park, Nanshan DistrictShenzhen518057Guangdong ProvinceChina
| | - Peng Sun
- Department of PhysicsHKBUKowloon TongKowloonHong Kong SAR China
- Department of Materials Science and EngineeringSouthern University of Science and TechnologyShenzhen518000Guangdong ProvinceChina
| | - Kaili Lin
- Department of BiologyHong Kong Baptist University (HKBU)Kowloon TongKowloonHong Kong SAR China
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
| | - Florence Hiu Ling Chan
- Department of Biomedical EngineeringCity University of Hong Kong (CityU)Tat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Qi Gao
- Department of Biomedical EngineeringCity University of Hong Kong (CityU)Tat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Wai Fung Lau
- Department of PhysicsHKBUKowloon TongKowloonHong Kong SAR China
| | - Vellaisamy A. L. Roy
- Department of Materials Science and EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Hongqi Zhang
- School of Chinese MedicineHKBUKowloon TongKowloonHong Kong SAR China
| | - King Wai Chiu Lai
- Department of Biomedical EngineeringCity University of Hong Kong (CityU)Tat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Zhifeng Huang
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
- HKBU Institute of Research and Continuing Education, 9FThe Industrialization Complex of Shenzhen Virtual University ParkNo. 2 Yuexing 3rd Road, South Zone, Hi‐tech Industrial Park, Nanshan DistrictShenzhen518057Guangdong ProvinceChina
- Department of PhysicsHKBUKowloon TongKowloonHong Kong SAR China
- Institute of Advanced MaterialsState Key Laboratory of Environmental and Biological AnalysisHKBUKowloon TongKowloonHong Kong SAR China
| | - Ken Kin Lam Yung
- Department of BiologyHong Kong Baptist University (HKBU)Kowloon TongKowloonHong Kong SAR China
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
- HKBU Institute of Research and Continuing Education, 9FThe Industrialization Complex of Shenzhen Virtual University ParkNo. 2 Yuexing 3rd Road, South Zone, Hi‐tech Industrial Park, Nanshan DistrictShenzhen518057Guangdong ProvinceChina
- Institute of Advanced MaterialsState Key Laboratory of Environmental and Biological AnalysisHKBUKowloon TongKowloonHong Kong SAR China
| |
Collapse
|
29
|
Molecular characterization of bovine amniotic fluid derived stem cells with an underlying focus on their comparative neuronal potential at different passages. Ann Anat 2019; 228:151452. [PMID: 31778790 DOI: 10.1016/j.aanat.2019.151452] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/17/2019] [Accepted: 11/13/2019] [Indexed: 11/21/2022]
Abstract
BACKGROUND The excellence in the field of stem cell therapy demands alternative and more convenient stem cells for potential applications. Researchers have opted for least invasive and broadly multipotent cells with minimum ethical concerns. Bovine amniotic fluid derived mesenchymal stem cells (BAF-MSCs) due to their ease of collection and owing similar gestational length to that of human could be presumed as an attractive large animal model for biomedical and biotechnology research. METHODS Bovine amniotic fluid derived stem cells were isolated from abattoir based samples and characterized for epithelial, neuronal, mesenchymal and pluripotent markers by qPCR and immunofluorescence studies at P1, P3, P5 and P7 alongside population doubling time, growth curve and multilineage differentiation studies. RESULTS The cells were explored for unique expression of Sox2, which was observed to be up regulated with increase in passage number and Nestin was found to be downregulated during further passaging of mesenchymal cells in this study. The cells also co-expressed Oct ¾ at initial passages which diminished within further passages. Evidence regarding diversity and heterogeneity in different cell population in amniotic fluid was recorded by positive expression of epithelial cell markers like pan Cytokeratin and p63 during early passages. The study suggested that cells with higher expression of Sox2 generated comparatively larger neurospheres with comparative strong expression of Sox2 and Nestin by immunofluorescence staining and qPCR analysis. Besides BAF-MSCs derived neurospheres were also shown to express pro-neuronal markers like ß-III Tubulin, GAP43 and ASCL-1. CONCLUSIONS This study explores and characterizes BAF-MSCs for their multipotent and neurogenic potentials and their use for clinical applications, though more detailed studies are needed to determine the exact pathways linked with neurogenic capacities of these cells and their morphological assessments at different gestational ages in bovines. The knowledge from the bovine model after detailed studies, proven safety and efficacy could also be used to understand substitutive strategies to investigate MSCs physiology at different trimesters and potential application of these cells for human and veterinary regenerative medicine provided the animal ethics are carefully monitored.
Collapse
|
30
|
Park HH, Han MH, Choi H, Lee YJ, Kim JM, Cheong JH, Ryu JI, Lee KY, Koh SH. Mitochondria damaged by Oxygen Glucose Deprivation can be Restored through Activation of the PI3K/Akt Pathway and Inhibition of Calcium Influx by Amlodipine Camsylate. Sci Rep 2019; 9:15717. [PMID: 31673096 PMCID: PMC6823474 DOI: 10.1038/s41598-019-52083-y] [Citation(s) in RCA: 16] [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: 06/18/2019] [Accepted: 10/13/2019] [Indexed: 12/25/2022] Open
Abstract
Amlodipine, a L-type calcium channel blocker, has been reported to have a neuroprotective effect in brain ischemia. Mitochondrial calcium overload leads to apoptosis of cells in neurologic diseases. We evaluated the neuroprotective effects of amlodipine camsylate (AC) on neural stem cells (NSCs) injured by oxygen glucose deprivation (OGD) with a focus on mitochondrial structure and function. NSCs were isolated from rodent embryonic brains. Effects of AC on cell viability, proliferation, level of free radicals, and expression of intracellular signaling proteins were assessed in OGD-injured NSCs. We also investigated the effect of AC on mitochondrial structure in NSCs under OGD by transmission electron microscopy. AC increased the viability and proliferation of NSCs. This beneficial effect of AC was achieved by strong protection of mitochondria. AC markedly enhanced the expression of mitochondrial biogenesis-related proteins and mitochondrial anti-apoptosis proteins. Together, our results indicate that AC protects OGD-injured NSCs by protecting mitochondrial structure and function. The results of the present study provide insight into the mechanisms underlying the protective effects of AC on NSCs.
Collapse
Affiliation(s)
- Hyun-Hee Park
- Departments of Neurology, Hanyang University Guri Hospital, 11923, Guri, Korea
| | - Myung-Hoon Han
- Departments of Neurosurgery, Hanyang University Guri Hospital, 11923, Guri, Korea
| | - Hojin Choi
- Departments of Neurology, Hanyang University Guri Hospital, 11923, Guri, Korea
| | - Young Joo Lee
- Departments of Neurology, Hanyang University Guri Hospital, 11923, Guri, Korea
| | - Jae Min Kim
- Departments of Neurosurgery, Hanyang University Guri Hospital, 11923, Guri, Korea
| | - Jin Hwan Cheong
- Departments of Neurosurgery, Hanyang University Guri Hospital, 11923, Guri, Korea
| | - Je Il Ryu
- Departments of Neurosurgery, Hanyang University Guri Hospital, 11923, Guri, Korea
| | - Kyu-Yong Lee
- Departments of Neurology, Hanyang University Guri Hospital, 11923, Guri, Korea.
| | - Seong-Ho Koh
- Departments of Neurology, Hanyang University Guri Hospital, 11923, Guri, Korea.
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, 04763, Seoul, Korea.
| |
Collapse
|
31
|
Fawal MA, Jungas T, Kischel A, Audouard C, Iacovoni JS, Davy A. Cross Talk between One-Carbon Metabolism, Eph Signaling, and Histone Methylation Promotes Neural Stem Cell Differentiation. Cell Rep 2019; 23:2864-2873.e7. [PMID: 29874574 DOI: 10.1016/j.celrep.2018.05.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/25/2018] [Accepted: 05/02/2018] [Indexed: 12/29/2022] Open
Abstract
Metabolic pathways, once seen as a mere consequence of cell states, have emerged as active players in dictating different cellular events such as proliferation, self-renewal, and differentiation. Several studies have reported a role for folate-dependent one-carbon (1C) metabolism in stem cells; however, its exact mode of action and how it interacts with other cues are largely unknown. Here, we report a link between the Eph:ephrin cell-cell communication pathway and 1C metabolism in controlling neural stem cell differentiation. Transcriptional and functional analyses following ephrin stimulation revealed alterations in folate metabolism-related genes and enzymatic activity. In vitro and in vivo data indicate that Eph-B forward signaling alters the methylation state of H3K4 by regulating 1C metabolism and locks neural stem cell in a differentiation-ready state. Our study highlights a functional link between cell-cell communication, metabolism, and epigenomic remodeling in the control of stem cell self-renewal.
Collapse
Affiliation(s)
- Mohamad-Ali Fawal
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Thomas Jungas
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Anthony Kischel
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Christophe Audouard
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Jason S Iacovoni
- Bioinformatic Plateau I2MC, INSERM and University of Toulouse, 31432 Toulouse, France
| | - Alice Davy
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France.
| |
Collapse
|
32
|
Choi H, Choi NY, Park HH, Lee KY, Lee YJ, Koh SH. Sublethal Doses of Zinc Protect Rat Neural Stem Cells Against Hypoxia Through Activation of the PI3K Pathway. Stem Cells Dev 2019; 28:769-780. [PMID: 30896367 DOI: 10.1089/scd.2018.0138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cerebral infarction is one of the major causes of severe morbidity and mortality, and thus, research has focused on developing treatment options for this condition. Zinc (Zn) is an essential element in the central nervous system and has several neuroprotective effects in the brain. In this study, we examined the neuroprotective effects of Zn on neural stem cells (NSCs) exposed to hypoxia. After treatment with several concentrations of Zn, the viability of NSCs under hypoxic conditions was measured by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, Trypan blue staining, and a lactate dehydrogenase assay. To evaluate the effect of Zn on the proliferation of NSCs, bromodeoxyuridine/5-bromo-2'-deoxyuridine (BrdU) labeling and colony formation assays were performed. Apoptosis was also examined in NSCs exposed to hypoxia with and without Zn treatment. In addition, a western blot analysis was performed to evaluate the effect of Zn on intracellular signaling proteins. NSC viability and proliferation were decreased under hypoxic conditions, but treatment with sublethal doses of Zn restored viability and proliferation. Sublethal doses of Zn reduced apoptosis caused by hypoxia, increased the expression levels of proteins related to the phosphatidylinositol-3 kinase (PI3K) pathway, and decreased the expression levels of proteins associated with neuronal cell death. These findings confirm that in vivo, sublethal doses of Zn protect NSCs against hypoxia through the activation of the PI3K pathway. Thus, Zn could be employed as a therapeutic option to protect NSCs in ischemic stroke.
Collapse
Affiliation(s)
- Hojin Choi
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Na-Young Choi
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Hyun-Hee Park
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Kyu-Yong Lee
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Young Joo Lee
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
| | - Seong-Ho Koh
- 1 Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
- 2 Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, Korea
| |
Collapse
|
33
|
Abstract
Cancer stem cells (CSCs) have been identified in glioblastoma (GBM) and are proposed to be the main actors of post-treatment recurrence contributing to the very dismal prognosis of this devastating disease. Consequently, this important population of cells needs to be further studied to uncover potential vulnerabilities, identify novel therapeutic targets, and develop drugs that can be translated to the clinic. One obstacle preventing progress in understanding the biology of GBM and the development of novel therapies has arguably been the absence of biologically relevant in vitro models representative of the CSC population in GBM. Adherent and non-adherent serum-free culture methods, initially developed for culturing neural stem cells, have been adapted to identify, isolate, maintain, and expand brain tumor stem cells (BTSCs) from GBM. In this chapter, we describe a method to isolate and culture these BTSCs from fresh GBM patient samples.
Collapse
|
34
|
Modrek AS, Prado J, Bready D, Dhaliwal J, Golub D, Placantonakis DG. Modeling Glioma with Human Embryonic Stem Cell-Derived Neural Lineages. Methods Mol Biol 2018; 1741:227-237. [PMID: 29392705 DOI: 10.1007/978-1-4939-7659-1_19] [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] [Indexed: 02/19/2023]
Abstract
Gliomas are malignant primary tumors of the central nervous system. Their cell-of-origin is thought to be a neural progenitor or stem cell that acquires mutations leading to oncogenic transformation. Thanks to advances in human stem cell biology, it has become possible to derive human cell types that represent putative cells-of-origin in vitro and model the gliomagenesis process by systematically introducing genetic alterations in these human cells. Here, we present methods to derive human neural stem cells (NSCs) from human embryonic stem cells (hESCs). Because these NSCs are genetically unmodified at baseline, they can be used as a cellular platform to study the effects of individual oncogenic hits in a well-controlled manner in the backdrop of a human genetic background. We also present some key applications of these NSCs, which include their transduction with lentiviral vectors, their neuroglial differentiation and xenografting methods into immunocompromised mice to assess in vivo behavior.
Collapse
Affiliation(s)
- Aram S Modrek
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| | - Jod Prado
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| | - Devin Bready
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| | - Joravar Dhaliwal
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| | - Danielle Golub
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA
| | - Dimitris G Placantonakis
- Department of Neurosurgery, NYU School of Medicine, New York, NY, USA. .,Kimmel Center for Stem Cell Biology, NYU School of Medicine, New York, NY, USA. .,Laura and Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA. .,Brain Tumor Center, NYU School of Medicine, New York, NY, USA. .,Neuroscience Institute, NYU School of Medicine, New York, NY, USA.
| |
Collapse
|
35
|
Kawauchi S, Horibe S, Sasaki N, Hirata KI, Rikitake Y. A novel in vitro co-culture model to examine contact formation between astrocytic processes and cerebral vessels. Exp Cell Res 2018; 374:333-341. [PMID: 30553966 DOI: 10.1016/j.yexcr.2018.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/06/2018] [Accepted: 12/10/2018] [Indexed: 10/27/2022]
Abstract
Here, we developed a novel in vitro co-culture model, in which process-bearing astrocytes and isolated cerebral microvessels from mice were co-cultured. Astrocytes formed contacts with microvessels from both adult and neonatal mice. However, concentrated localization of the immunofluorescence signal for aquaporin-4 (AQP4) at contact sites between perivascular endfoot processes and blood vessels was only detected with neonatal mouse microvessels. Contact between astrocytic processes and microvessels was retained, whereas concentrated localization of AQP4 signal at contact sites was lost, by knockdown of dystroglycan or α-syntrophin, reflecting polarized localization of AQP4 at perivascular regions in the brain. Further, using our in vitro co-culture model, we found that astrocytes predominantly extend processes to pericytes located at the abluminal surface of microvessels, providing additional evidence that this model is representative of the in vivo situation. Altogether, we have developed a novel in vitro co-culture model that can reproduce aspects of the in vivo situation and is useful for assessing contact formation between astrocytes and blood vessels.
Collapse
Affiliation(s)
- Shoji Kawauchi
- Educational and Research Center for Clinical Pharmacy, Kobe Pharmaceutical University, Kobe 658-8558, Japan
| | - Sayo Horibe
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Kobe 658-8558, Japan
| | - Naoto Sasaki
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Kobe 658-8558, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yoshiyuki Rikitake
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Kobe 658-8558, Japan.
| |
Collapse
|
36
|
Sato J, Horibe S, Kawauchi S, Sasaki N, Hirata KI, Rikitake Y. Involvement of aquaporin-4 in laminin-enhanced process formation of mouse astrocytes in 2D culture: Roles of dystroglycan and α-syntrophin in aquaporin-4 expression. J Neurochem 2018; 147:495-513. [PMID: 29981530 DOI: 10.1111/jnc.14548] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/27/2018] [Accepted: 06/27/2018] [Indexed: 01/13/2023]
Abstract
In the central nervous system, astrocytes extend endfoot processes to ensheath synapses and microvessels. However, the mechanisms underlying this astrocytic process extension remain unclear. A limitation of the use of 2D cultured astrocytes for such studies is that they display a flat, epithelioid morphology, with no or very few processes, which is markedly different from the stellate morphology observed in vivo. In this study, we obtained 2D cultured astrocytes with a rich complexity of processes using differentiation of neurospheres in vitro. Using these process-bearing astrocytes, we showed that laminin, an extracellular matrix molecule abundant in perivascular sites, efficiently induced process formation and branching. Specifically, the numbers of the first- and second-order branch processes and the maximal process length of astrocytes were increased when cultured on laminin, compared with when they were cultured on poly-L-ornithine or type IV collagen. Knockdown of dystroglycan or α-syntrophin, constituent proteins of the dystrophin-glycoprotein complex that provides a link between laminin and the cytoskeleton, using small interference RNAs inhibited astrocyte process formation and branching, and down-regulated expression of the water channel aquaporin-4 (AQP4). Direct knockdown and a specific inhibitor of AQP4 also inhibited, whereas over-expression of AQP4 enhanced astrocyte process formation and branching. Knockdown of AQP4 decreased phosphorylation of focal adhesion kinase (FAK) that is critically implicated in actin remodeling. Collectively, these results indicate that the laminin-dystroglycan-α-syntrophin-AQP4 axis is important for process formation and branching of 2D cultured astrocytes. OPEN PRACTICES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Read the Editorial Highlight for this article on page 436.
Collapse
Affiliation(s)
- Junya Sato
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan.,Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan
| | - Sayo Horibe
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan
| | - Shoji Kawauchi
- Educational Center for Clinical Pharmacy, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan
| | - Naoto Sasaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan.,Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Yoshiyuki Rikitake
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan.,Division of Signal Transduction, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| |
Collapse
|
37
|
Song P, Xia X, Han T, Fang H, Wang Y, Dong F, Zhang R, Ge P, Shen C. BMSCs promote the differentiation of NSCs into oligodendrocytes via mediating Id2 and Olig expression through BMP/Smad signaling pathway. Biosci Rep 2018; 38:BSR20180303. [PMID: 30143582 PMCID: PMC6147919 DOI: 10.1042/bsr20180303] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/02/2018] [Accepted: 08/08/2018] [Indexed: 01/01/2023] Open
Abstract
Neural stem cells (NSCs) have emerged as a promising treatment for spinal cord injuries. However, the increasing expression of bone morphogenetic proteins (BMPs) in spinal cord injury lesion sites seems to have contributed to the limited oligodendroglial differentiation and the majority of the astroglial differentiation of NSCs. In the present study, we demonstrate that BMPs promote NSCs differentiation toward astrocytes and prevent them from differentiating into oligodendrocytes. This effect is accompanied by the increasing expression of Id2 and the reduction in Oilg1/2 expression. Treatment with bone marrow stromal cells (BMSCs) can enhance the development of oligodendrocytes in the presence of BMPs. The analysis of Id2, as well as Olig1 and Olig2 gene expression, reveals that the effect of BMPs on these gene expressions is reversed with the addition of BMSCs. In sum, these data strongly suggest that BMSCs can promote the differentiation of NSCs into oligodendrocytes through mediating Id2 and Olig1/2 expression by blocking the BMP/Smad signaling pathway.
Collapse
Affiliation(s)
- Peiwen Song
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Xiang Xia
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Tianyu Han
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Huang Fang
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Ying Wang
- Department of Medical Imaging, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Fulong Dong
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Renjie Zhang
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Peng Ge
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| | - Cailiang Shen
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Shushan District, Hefei City, Anhui Province, China
| |
Collapse
|
38
|
Sahin I, Zhang Y, McAllister F. Tumor Spheres Quantification with Smoothed Euclidean Distance Transform. ACTA ACUST UNITED AC 2018; 8. [PMID: 30319887 PMCID: PMC6179360 DOI: 10.4172/2155-9937.1000143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Tumor sphere quantification plays an important role in cancer research and drugs screening. Even though the number and size of tumor spheres can be found manually, this process is time-consuming, prone to making errors, and may not be viable when the number of images is very large. This manuscript presents a method for automated quantification of spheres with a novel segmentation technique. The segmentation method relies on initial watershed algorithm which detects the minima of the distance transform and finds a tumor sphere for each minimum. Due to the irregular edges of tumor spheres, the distance transform matrix has often more number of minima than the true number of spheres. This leads to the over segmentation problem. The proposed approach uses the smoothed form of the distance transform to effectively eliminate superfluous minima and then seeds the watershed algorithm with the remaining minima. The proposed method was validated over pancreatic tumor spheres images achieving high efficiency for tumor spheres quantification.
Collapse
Affiliation(s)
- Ismet Sahin
- Department of Engineering, Texas Southern University, Houston, TX, USA
| | - Yu Zhang
- Department of Clinical Cancer Prevention, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
39
|
Atorvastatin Rejuvenates Neural Stem Cells Injured by Oxygen–Glucose Deprivation and Induces Neuronal Differentiation Through Activating the PI3K/Akt and ERK Pathways. Mol Neurobiol 2018; 56:2964-2977. [DOI: 10.1007/s12035-018-1267-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 07/18/2018] [Indexed: 01/01/2023]
|
40
|
Kishimoto TE, Uchida K, Thongtharb A, Shibato T, Chambers JK, Nibe K, Kagawa Y, Nakayama H. Expression of Oligodendrocyte Precursor Cell Markers in Canine Oligodendrogliomas. Vet Pathol 2018; 55:634-644. [DOI: 10.1177/0300985818777794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Oligodendroglioma is a common brain tumor in dogs, particularly brachycephalic breeds. Oligodendrocyte precursor cells (OPCs) are suspected to be a possible origin of oligodendroglioma, although it has not been well elucidated. In the present study, 27 cases of canine brain oligodendrogliomas were histologically and immunohistochemically examined. The most commonly affected breed was the French Bulldog ( n = 19 of 27, 70%). Seizure was the most predominant clinical sign ( n = 17 of 25, 68%). The tumors were located mainly in the cerebrum, particularly in the frontal lobe ( n = 10 of 27, 37%). All cases were diagnosed as anaplastic oligodendroglioma (AO) and had common histologic features characterized by the proliferation of round to polygonal cells with pronounced atypia and conspicuous mitotic activity (average, 10.7 mitoses per 10 high-power fields). Honeycomb pattern ( n = 5 of 27, 19%), myxoid matrix ( n = 10, 37%), cyst formation ( n = 6, 22%), necrosis ( n = 19, 70%), pseudopalisading ( n = 5, 18.5%), glomeruloid vessels ( n = 16, 59%), and microcalcification ( n = 5, 19%) were other histopathologic features of the present tumors. Immunohistochemically, the tumor cells were positive for Olig2 in all cases and for other markers of OPCs in most cases, including SOX10 ( n = 24 of 27, 89%), platelet-derived growth factor receptor α ( n = 24, 89%), and NG2 ( n = 23, 85%). The present AO also consisted of heterogeneous cell populations that were positive for nestin ( n = 13 of 27, 48%), glial fibrillary acidic protein ( n = 5, 19%), doublecortin ( n = 22, 82%), and βIII-tubulin ( n = 15, 56%). Moreover, cultured AO cells obtained from 1 case retained expression of OPC markers and exhibited multipotent characteristics in a serum culture condition. Overall, the findings suggest that transformed multipotent OPCs may be a potential origin of canine AO.
Collapse
Affiliation(s)
- Takuya E. Kishimoto
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Kazuyuki Uchida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Atigan Thongtharb
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | | | - James K. Chambers
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Kazumi Nibe
- Japan Animal Referral Medical Center Kawasaki, Kanagawa, Japan
| | | | - Hiroyuki Nakayama
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| |
Collapse
|
41
|
Tekin H, Simmons S, Cummings B, Gao L, Adiconis X, Hession CC, Ghoshal A, Dionne D, Choudhury SR, Yesilyurt V, Sanjana NE, Shi X, Lu C, Heidenreich M, Pan JQ, Levin JZ, Zhang F. Effects of 3D culturing conditions on the transcriptomic profile of stem-cell-derived neurons. Nat Biomed Eng 2018; 2:540-554. [PMID: 30271673 PMCID: PMC6157920 DOI: 10.1038/s41551-018-0219-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Understanding neurological diseases requires tractable genetic systems. Engineered 3D neural tissues are an attractive choice, but how the cellular transcriptomic profiles in these tissues are affected by the encapsulating materials and are related to the human-brain transcriptome is not well understood. Here, we report the characterization of the effects of culturing conditions on the transcriptomic profiles of induced neuronal cells, as well as a method for the rapid generation of 3D co-cultures of neuronal and astrocytic cells from the same pool of human embryonic stem cells. By comparing the gene-expression profiles of neuronal cells in culture conditions relevant to the developing human brain, we found that modifying the degree of crosslinking of composite hydrogels can tune expression patterns so they correlate with those of specific brain regions and developmental stages. Moreover, by using single-cell sequencing, we show that our engineered tissues recapitulate transcriptional patterns of cell types in the human brain. The analysis of culturing conditions will inform the development of 3D neural tissues for use as tractable models of brain diseases.
Collapse
Affiliation(s)
- Halil Tekin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Sean Simmons
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beryl Cummings
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Linyi Gao
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xian Adiconis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Ayan Ghoshal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Sourav R Choudhury
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Volkan Yesilyurt
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Neville E Sanjana
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,New York Genome Center and Department of Biology, New York University, New York, NY, USA
| | - Xi Shi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Congyi Lu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,New York Genome Center and Department of Biology, New York University, New York, NY, USA
| | - Matthias Heidenreich
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jen Q Pan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Feng Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
42
|
B-1a lymphocytes promote oligodendrogenesis during brain development. Nat Neurosci 2018; 21:506-516. [PMID: 29507409 DOI: 10.1038/s41593-018-0106-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 01/08/2018] [Indexed: 01/22/2023]
Abstract
During brain development, the immune system mediates neurogenesis, gliogenesis and synapse formation. However, it remains unclear whether peripheral lymphocytes contribute to brain development. Here we identified the subtypes of lymphocytes that are present in neonatal mouse brains and investigated their functions. We found that B-1a cells, a subtype of B cells, were abundant in the neonatal mouse brain and infiltrated into the brain in a CXCL13-CXCR5-dependent manner. B-1a cells promoted the proliferation of oligodendrocyte-precursor cells (OPCs) in vitro, and depletion of B-1a cells from developing brains resulted in a reduction of numbers of OPCs and mature oligodendrocytes. Furthermore, neutralizing Fcα/μR, the receptor for the Fc region of IgM secreted by B-1a cells, inhibited OPC proliferation and reduced the proportion of myelinated axons in neonatal mouse brains. Our results demonstrate that B-1a cells infiltrate into the brain and contribute to oligodendrogenesis and myelination by promoting OPC proliferation via IgM-Fcα/μR signaling.
Collapse
|
43
|
Lee RHC, Lee MHH, Wu CYC, Couto e Silva A, Possoit HE, Hsieh TH, Minagar A, Lin HW. Cerebral ischemia and neuroregeneration. Neural Regen Res 2018; 13:373-385. [PMID: 29623912 PMCID: PMC5900490 DOI: 10.4103/1673-5374.228711] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Cerebral ischemia is one of the leading causes of morbidity and mortality worldwide. Although stroke (a form of cerebral ischemia)-related costs are expected to reach 240.67 billion dollars by 2030, options for treatment against cerebral ischemia/stroke are limited. All therapies except anti-thrombolytics (i.e., tissue plasminogen activator) and hypothermia have failed to reduce neuronal injury, neurological deficits, and mortality rates following cerebral ischemia, which suggests that development of novel therapies against stroke/cerebral ischemia are urgently needed. Here, we discuss the possible mechanism(s) underlying cerebral ischemia-induced brain injury, as well as current and future novel therapies (i.e., growth factors, nicotinamide adenine dinucleotide, melatonin, resveratrol, protein kinase C isozymes, pifithrin, hypothermia, fatty acids, sympathoplegic drugs, and stem cells) as it relates to cerebral ischemia.
Collapse
Affiliation(s)
- Reggie H. C. Lee
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Michelle H. H. Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China
| | - Celeste Y. C. Wu
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Alexandre Couto e Silva
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Harlee E. Possoit
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Tsung-Han Hsieh
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
- Cardiovascular and Metabolomics Research Center, Hualien Tzu Chi Hospital, Hualien, Taiwan, China
| |
Collapse
|
44
|
Morton PD, Korotcova L, Lewis BK, Bhuvanendran S, Ramachandra SD, Zurakowski D, Zhang J, Mori S, Frank JA, Jonas RA, Gallo V, Ishibashi N. Abnormal neurogenesis and cortical growth in congenital heart disease. Sci Transl Med 2018; 9:9/374/eaah7029. [PMID: 28123074 DOI: 10.1126/scitranslmed.aah7029] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 08/04/2016] [Accepted: 10/05/2016] [Indexed: 12/21/2022]
Abstract
Long-term neurological deficits due to immature cortical development are emerging as a major challenge in congenital heart disease (CHD). However, cellular mechanisms underlying dysregulation of perinatal corticogenesis in CHD remain elusive. The subventricular zone (SVZ) represents the largest postnatal niche of neural stem/progenitor cells (NSPCs). We show that the piglet SVZ resembles its human counterpart and displays robust postnatal neurogenesis. We present evidence that SVZ NSPCs migrate to the frontal cortex and differentiate into interneurons in a region-specific manner. Hypoxic exposure of the gyrencephalic piglet brain recapitulates CHD-induced impaired cortical development. Hypoxia reduces proliferation and neurogenesis in the SVZ, which is accompanied by reduced cortical growth. We demonstrate a similar reduction in neuroblasts within the SVZ of human infants born with CHD. Our findings demonstrate that SVZ NSPCs contribute to perinatal corticogenesis and suggest that restoration of SVZ NSPCs' neurogenic potential is a candidate therapeutic target for improving cortical growth in CHD.
Collapse
Affiliation(s)
- Paul D Morton
- Center for Neuroscience Research, Children's National Health System, Washington, DC 20010, USA.,Children's National Heart Institute, Children's National Health System, Washington, DC 20010, USA
| | - Ludmila Korotcova
- Center for Neuroscience Research, Children's National Health System, Washington, DC 20010, USA.,Children's National Heart Institute, Children's National Health System, Washington, DC 20010, USA
| | - Bobbi K Lewis
- Frank Laboratory and Laboratory of Diagnostic Radiology Research, Department of Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shivaprasad Bhuvanendran
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Shruti D Ramachandra
- Center for Neuroscience Research, Children's National Health System, Washington, DC 20010, USA.,Children's National Heart Institute, Children's National Health System, Washington, DC 20010, USA
| | - David Zurakowski
- Departments of Anesthesia and Surgery, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | - Jiangyang Zhang
- Department of Biomedical Engineering and The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Susumu Mori
- Department of Biomedical Engineering and The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Joseph A Frank
- Frank Laboratory and Laboratory of Diagnostic Radiology Research, Department of Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD 20892, USA.,Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard A Jonas
- Center for Neuroscience Research, Children's National Health System, Washington, DC 20010, USA. .,Children's National Heart Institute, Children's National Health System, Washington, DC 20010, USA
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Health System, Washington, DC 20010, USA.
| | - Nobuyuki Ishibashi
- Center for Neuroscience Research, Children's National Health System, Washington, DC 20010, USA. .,Children's National Heart Institute, Children's National Health System, Washington, DC 20010, USA
| |
Collapse
|
45
|
Park HH, Lee KY, Park DW, Choi NY, Lee YJ, Son JW, Kim S, Moon C, Kim HW, Rhyu IJ, Koh SH. Tracking and protection of transplanted stem cells using a ferrocenecarboxylic acid-conjugated peptide that mimics hTERT. Biomaterials 2017; 155:80-91. [PMID: 29169040 DOI: 10.1016/j.biomaterials.2017.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/08/2017] [Accepted: 11/12/2017] [Indexed: 02/07/2023]
Abstract
In vivo tracking of transplanted stem cells has been a central aim of stem cell therapy. Although many tracking systems have been introduced, no method has yet been validated for clinical applications. We developed a novel sophisticated peptide (GV1001) that mimics hTERT (human telomerase reverse transcriptase) and analysed its ability to track and protect stem cells after transplantation. Ferrocenecarboxylic acid-conjugated GV1001 (Fe-GV1001) efficiently penetrated stem cells with no adverse effects. Moreover, Fe-GV1001 improved the viability, proliferation, and migration of stem cells under hypoxia. After Fe-GV1001-labelled stem cells were transplanted into the brains of rats after stroke, the labelled cells were easily tracked by MRI. Our findings indicate that Fe-GV1001 can be used for the in vivo tracking of stem cells after transplantation into the brain and can improve the efficacy of stem cell therapy by sustaining and enhancing stem cell characteristics under disease conditions.
Collapse
Affiliation(s)
- Hyun-Hee Park
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Kyu-Yong Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Dong Woo Park
- Department of Radiology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Na-Young Choi
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Young Joo Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Jeong-Woo Son
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, 04763, South Korea
| | - Sangjae Kim
- Teloid Inc., 920 Westholme Ave, Los Angeles (City), CA 90024, USA
| | - Chanil Moon
- Department of Neuroscience, GemVax & KAEL Co., Ltd., Seoul, South Korea
| | - Hyun-Wook Kim
- Brain Korea 21 PLUS, KU Medical Science Center for Convergent Translational Research, 73 Inchonro, Seongbuk-gu, Seoul, 136-705, South Korea; Department of Anatomy, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul, 136-705, South Korea
| | - Im Joo Rhyu
- Brain Korea 21 PLUS, KU Medical Science Center for Convergent Translational Research, 73 Inchonro, Seongbuk-gu, Seoul, 136-705, South Korea; Department of Anatomy, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul, 136-705, South Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea; Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, 04763, South Korea.
| |
Collapse
|
46
|
Abdraboh ME, Abdeen SH, Salama M, El-Husseiny M, El-Sherbini YM, Eldeen NM. Developmental neurotoxic effects of a low dose of TCE on a 3-D neurosphere system. Biochem Cell Biol 2017; 96:50-56. [PMID: 29040813 DOI: 10.1139/bcb-2017-0089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Trichloroethylene (TCE) is one of the industrial toxic byproducts that now persist in the air, soil, and water. Several studies have already illustrated the toxic effect of high doses of TCE on the biological functions of several organs. This study aims to highlight the toxic impact of a low dose of TCE (1 μmol/L) on the development of rat neural stem cells (NSCs). The subventricular zones (SVZ) of rat pup's brains were collected and minced, and the harvested cells were cultured in the presence of neural growth factors B27/N2 to develop neurospheres. The cells were then exposed to a dose of 1 μmol/L TCE for 1 or 2 weeks. The outcomes indicated a remarkable inhibitory effect of TCE on the differentiation capacity of NSCs, which was confirmed by down-regulation of the astrocyte marker GFAP The inhibitory effect of TCE on the proliferation of NSCs was identified by the reductions in neurosphere diameter, Ki67 expression, and cell cycle arrest at the G1/S phase. Immunolabelling with annexin V indicated the proapoptotic effect of TCE exposure. PCR results revealed a TCE-mediated suppression of the expression of the antioxidant enzyme SOD1. This paper illustrates, for the first time, a detailed examination of the toxic effects of an environmentally low dose of TCE on NCSs at the transcriptional, translational, and functional levels.
Collapse
Affiliation(s)
- M E Abdraboh
- a Department of Zoology, Faculty of Science, Mansoura University, Mansoura 35116, Egypt
| | - S H Abdeen
- a Department of Zoology, Faculty of Science, Mansoura University, Mansoura 35116, Egypt
| | - M Salama
- b Department of Toxicology, Faculty of Medicine, Mansoura University, Mansoura 35116, Egypt
| | - M El-Husseiny
- b Department of Toxicology, Faculty of Medicine, Mansoura University, Mansoura 35116, Egypt
| | - Y M El-Sherbini
- c Faculty of Health and Life Science, Oxford Brookes University, Headington Campus, Oxford, OX3 0BP, UK
| | - N M Eldeen
- a Department of Zoology, Faculty of Science, Mansoura University, Mansoura 35116, Egypt
| |
Collapse
|
47
|
Dolci S, Pino A, Berton V, Gonzalez P, Braga A, Fumagalli M, Bonfanti E, Malpeli G, Pari F, Zorzin S, Amoroso C, Moscon D, Rodriguez FJ, Fumagalli G, Bifari F, Decimo I. High Yield of Adult Oligodendrocyte Lineage Cells Obtained from Meningeal Biopsy. Front Pharmacol 2017; 8:703. [PMID: 29075188 PMCID: PMC5643910 DOI: 10.3389/fphar.2017.00703] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/21/2017] [Indexed: 12/25/2022] Open
Abstract
Oligodendrocyte loss can lead to cognitive and motor deficits. Current remyelinating therapeutic strategies imply either modulation of endogenous oligodendrocyte precursors or transplantation of in vitro expanded oligodendrocytes. Cell therapy, however, still lacks identification of an adequate source of oligodendrocyte present in adulthood and able to efficiently produce transplantable cells. Recently, a neural stem cell-like population has been identified in meninges. We developed a protocol to obtain high yield of oligodendrocyte lineage cells from one single biopsy of adult rat meningeal tissue. From 1 cm2 of adult rat spinal cord meninges, we efficiently expanded a homogenous culture of 10 millions of meningeal-derived oligodendrocyte lineage cells in a short period of time (approximately 4 weeks). Meningeal-derived oligodendrocyte lineage cells show typical mature oligodendrocyte morphology and express specific oligodendrocyte markers, such as galactosylceramidase and myelin basic protein. Moreover, when transplanted in a chemically demyelinated spinal cord model, meningeal-derived oligodendrocyte lineage cells display in vivo-remyelinating potential. This oligodendrocyte lineage cell population derives from an accessible and adult source, being therefore a promising candidate for autologous cell therapy of demyelinating diseases. In addition, the described method to differentiate meningeal-derived neural stem cells into oligodendrocyte lineage cells may represent a valid in vitro model to dissect oligodendrocyte differentiation and to screen for drugs capable to promote oligodendrocyte regeneration.
Collapse
Affiliation(s)
- Sissi Dolci
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Annachiara Pino
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Valeria Berton
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Pau Gonzalez
- Group of Molecular Neurology, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - Alice Braga
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Elisabetta Bonfanti
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Giorgio Malpeli
- Section of General and Pancreatic Surgery, Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, Verona, Italy
| | - Francesca Pari
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Stefania Zorzin
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Clelia Amoroso
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Denny Moscon
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Guido Fumagalli
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Ilaria Decimo
- Section of Pharmacology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| |
Collapse
|
48
|
Effects of aspirin and clopidogrel on neural stem cells. Cell Biol Toxicol 2017; 34:219-232. [DOI: 10.1007/s10565-017-9412-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022]
|
49
|
Bryant DT, Landles C, Papadopoulou AS, Benjamin AC, Duckworth JK, Rosahl T, Benn CL, Bates GP. Disruption to schizophrenia-associated gene Fez1 in the hippocampus of HDAC11 knockout mice. Sci Rep 2017; 7:11900. [PMID: 28928414 PMCID: PMC5605701 DOI: 10.1038/s41598-017-11630-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 08/25/2017] [Indexed: 12/18/2022] Open
Abstract
Histone Deacetylase 11 (HDAC11) is highly expressed in the central nervous system where it has been reported to have roles in neural differentiation. In contrast with previous studies showing nuclear and cytoplasmic localisation, we observed synaptic enrichment of HDAC11. Knockout mouse models for HDACs 1-9 have been important for guiding the development of isoform specific HDAC inhibitors as effective therapeutics. Given the close relationship between HDAC11 and neural cells in vitro, we examined neural tissue in a previously uncharacterised Hdac11 knockout mouse (Hdac11 KO/KO). Loss of HDAC11 had no obvious impact on brain morphology and neural stem/precursor cells isolated from Hdac11 KO/KO mice had comparable proliferation and differentiation characteristics. However, in differentiating neural cells we observed decreased expression of schizophrenia-associated gene Fez1 (fasciculation and elongation protein zeta 1), a gene previously reported to be regulated by HDAC11 activity. FEZ1 has been associated with the dendritic growth of neurons and risk of schizophrenia via its interaction with DISC1 (disrupted in schizophrenia 1). Examination of cortical, cerebellar and hippocampal tissue reveal decreased Fez1 expression specifically in the hippocampus of adult mice. The results of this study demonstrate that loss of HDAC11 has age dependent and brain-region specific consequences.
Collapse
Affiliation(s)
- Dale T Bryant
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom.,Neusentis, Pfizer Ltd, The Portway, Granta Park, Abington, Cambridge, United Kingdom
| | - Christian Landles
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Aikaterini S Papadopoulou
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Agnesska C Benjamin
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom
| | - Joshua K Duckworth
- Neusentis, Pfizer Ltd, The Portway, Granta Park, Abington, Cambridge, United Kingdom
| | | | - Caroline L Benn
- Neusentis, Pfizer Ltd, The Portway, Granta Park, Abington, Cambridge, United Kingdom
| | - Gillian P Bates
- UCL Huntington's Disease Centre, Sobell Department of Motor Neuroscience, UCL Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
50
|
Abstract
Traditional primary and secondary cell cultures have been used for the investigation of prion biology and disease for many years. While both types of cultures produce highly valid and immensely valuable results, they also have their limitations; traditional cell lines are often derived from cancers, therefore subject to numerous DNA changes, and primary cultures are labor-intensive and expensive to produce requiring sacrifice of many animals. Neural stem cell (NSC) cultures are a relatively new technology to be used for the study of prion biology and disease. While NSCs are subject to their own limitations-they are generally cultured ex vivo in environments that artificially force their growth-they also have their own unique advantages. NSCs retain the ability for self-renewal and can therefore be propagated in culture similarly to secondary cultures without genetic manipulation. In addition, NSCs are multipotent; they can be induced to differentiate into mature cells of central nervous system (CNS) linage. The combination of self-renewal and multipotency allows NSCs to be used as a primary cell line over multiple generations saving time, costs, and animal harvests, thus providing a valuable addition to the existing cell culture repertoire used for investigation of prion biology and disease. Furthermore, NSC cultures can be generated from mice of any genotype, either by embryonic harvest or harvest from adult brain, allowing gene expression to be studied without further genetic manipulation. This chapter describes a standard method of culturing adult NSCs and assays for monitoring NSC growth, migration, and differentiation and revisits basic reactive oxygen species detection in the context of NSC cultures.
Collapse
Affiliation(s)
- Cathryn L Haigh
- Department of Medicine, Melbourne Brain Centre, Royal Melbourne Hospital, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3010, Australia. .,Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA.
| |
Collapse
|