1
|
Jiang N, Ying G, Yin Y, Guo J, Lozada J, Valdivia Padilla A, Gómez A, Gomes de Melo BA, Lugo Mestre F, Gansevoort M, Palumbo M, Calá N, Garciamendez-Mijares CE, Kim GA, Takayama S, Gerhard-Herman MD, Zhang YS. A closed-loop modular multiorgan-on-chips platform for self-sustaining and tightly controlled oxygenation. Proc Natl Acad Sci U S A 2024; 121:e2413684121. [PMID: 39541351 PMCID: PMC11588096 DOI: 10.1073/pnas.2413684121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
To mimic physiological microenvironments in organ-on-a-chip systems, physiologically relevant parameters are required to precisely access drug metabolism. Oxygen level is a critical microenvironmental parameter to maintain cellular or tissue functions and modulate their behaviors. Current organ-on-a-chip setups are oftentimes subjected to the ambient incubator oxygen level at 21%, which is higher than most if not all physiological oxygen concentrations. Additionally, the physiological oxygen level in each tissue is different ranging from 0.5 to 13%. Here, a closed-loop modular multiorgan-on-chips platform is developed to enable not only real-time monitoring of the oxygen levels but, more importantly, tight control of them in the range of 4 to 20% across each connected microtissue-on-a-chip in the circulatory culture medium. This platform, which consists of microfluidic oxygen scavenger(s), an oxygen generator, a monitoring/controller system, and bioreactor(s), allows for independent, precise upregulation and downregulation of dissolved oxygen in the perfused culture medium to meet the physiological oxygen level in each modular microtissue compartment, as needed. Furthermore, drug studies using the platform demonstrate that the oxygen level affects drug metabolism in the parallelly connected liver, kidney, and arterial vessel microtissues without organ-organ interactions factored in. Overall, this platform can promote the performances of organ-on-a-chip devices in drug screening by providing more physiologically relevant and independently adjustable oxygen microenvironments for desired organ types on a single- or a multiorgan-on-chip(s) configuration.
Collapse
Affiliation(s)
- Nan Jiang
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
- Department of Physics, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Guoliang Ying
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Yixia Yin
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Jie Guo
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Jorge Lozada
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Alejandra Valdivia Padilla
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Ameyalli Gómez
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Bruna Alice Gomes de Melo
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Francisco Lugo Mestre
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Merel Gansevoort
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Marcello Palumbo
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Noemi Calá
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Carlos Ezio Garciamendez-Mijares
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
| | - Ge-Ah Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA30318
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA30318
| | - Marie Denis Gerhard-Herman
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Yu Shrike Zhang
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02139
- Harvard Stem Cell Institute, Harvard University,Cambridge, MA02138
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA02142
| |
Collapse
|
2
|
Barata P, Camacho O, Lima CG, Pereira AC. The Role of Hyperbaric Oxygen Therapy in Neuroregeneration and Neuroprotection: A Review. Cureus 2024; 16:e62067. [PMID: 38989389 PMCID: PMC11235151 DOI: 10.7759/cureus.62067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Neurogenesis is a high energy-demanding process, which is why blood vessels are an active part of the neurogenic niche since they allow the much-needed oxygenation of progenitor cells. In this regard, although neglected for a long time, the "oxygen niche" should be considered an important intervenient in adult neurogenesis. One possible hypothesis for the failure of numerous neuroprotective trials is that they relied on compounds that target a highly specific neuroprotective pathway. This approach may be too limited, given the complexity of the processes that lead to cell death. Therefore, research should adopt a more multifactorial approach. Among the limited range of agents with multimodal neuromodulatory capabilities, hyperbaric oxygen therapy has demonstrated effectiveness in reducing secondary brain damage in various brain injury models. This therapy functions not only as a neuroprotective mechanism but also as a powerful neuroregenerative mechanism.
Collapse
Affiliation(s)
- Pedro Barata
- Pathology and Laboratory Medicine, Centro Hospitalar Universitário do Porto, Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| | - Oscar Camacho
- Hyperbaric Medicine Unit, Unidade Local de Saúde de Matosinhos, Matosinhos, PRT
| | - Clara G Lima
- Anesthesiology, Hospital Pedro Hispano, Matosinhos, PRT
| | - Ana Claudia Pereira
- Faculty of Health Sciences, Universidade Fernando Pessoa (UFP), Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| |
Collapse
|
3
|
Yasan GT, Gunel-Ozcan A. Hypoxia and Hypoxia Mimetic Agents As Potential Priming Approaches to Empower Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2024; 19:33-54. [PMID: 36642875 DOI: 10.2174/1574888x18666230113143234] [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] [Received: 07/29/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 01/17/2023]
Abstract
Mesenchymal stem cells (MSC) exhibit self-renewal capacity and multilineage differentiation potential, making them attractive for research and clinical application. The properties of MSC can vary depending on specific micro-environmental factors. MSC resides in specific niches with low oxygen concentrations, where oxygen functions as a metabolic substrate and a signaling molecule. Conventional physical incubators or chemically hypoxia mimetic agents are applied in cultures to mimic the original low oxygen tension settings where MSC originated. This review aims to focus on the current knowledge of the effects of various physical hypoxic conditions and widely used hypoxia-mimetic agents-PHD inhibitors on mesenchymal stem cells at a cellular and molecular level, including proliferation, stemness, differentiation, viability, apoptosis, senescence, migration, immunomodulation behaviors, as well as epigenetic changes.
Collapse
Affiliation(s)
| | - Aysen Gunel-Ozcan
- Department of Stem Cell Sciences, Center for Stem Cell Research and Development, Hacettepe University, Ankara, Turkey
| |
Collapse
|
4
|
Zuccoli GS, Nascimento JM, Moraes-Vieira PM, Rehen SK, Martins-de-Souza D. Mitochondrial, cell cycle control and neuritogenesis alterations in an iPSC-based neurodevelopmental model for schizophrenia. Eur Arch Psychiatry Clin Neurosci 2023; 273:1649-1664. [PMID: 37039888 DOI: 10.1007/s00406-023-01605-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/29/2023] [Indexed: 04/12/2023]
Abstract
Schizophrenia is a severe psychiatric disorder of neurodevelopmental origin that affects around 1% of the world's population. Proteomic studies and other approaches have provided evidence of compromised cellular processes in the disorder, including mitochondrial function. Most of the studies so far have been conducted on postmortem brain tissue from patients, and therefore, do not allow the evaluation of the neurodevelopmental aspect of the disorder. To circumvent that, we studied the mitochondrial and nuclear proteomes of neural stem cells (NSCs) and neurons derived from induced pluripotent stem cells (iPSCs) from schizophrenia patients versus healthy controls to assess possible alterations related to energy metabolism and mitochondrial function during neurodevelopment in the disorder. Our results revealed differentially expressed proteins in pathways related to mitochondrial function, cell cycle control, DNA repair and neuritogenesis and their possible implication in key process of neurodevelopment, such as neuronal differentiation and axonal guidance signaling. Moreover, functional analysis of NSCs revealed alterations in mitochondrial oxygen consumption in schizophrenia-derived cells and a tendency of higher levels of intracellular reactive oxygen species (ROS). Hence, this study shows evidence that alterations in important cellular processes are present during neurodevelopment and could be involved with the establishment of schizophrenia, as well as the phenotypic traits observed in adult patients. Neural stem cells (NSCs) and neurons were derived from induced pluripotent stem cells (iPSCs) from schizophrenia patients and controls. Proteomic analyses were performed on the enriched mitochondrial and nuclear fractions of NSCs and neurons. Whole-cell proteomic analysis was also performed in neurons. Our results revealed alteration in proteins related to mitochondrial function, cell cycle control, among others. We also performed energy pathway analysis and reactive oxygen species (ROS) analysis of NSCs, which revealed alterations in mitochondrial oxygen consumption and a tendency of higher levels of intracellular ROS in schizophrenia-derived cells.
Collapse
Affiliation(s)
- Giuliana S Zuccoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Juliana M Nascimento
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil
| | - Pedro M Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, 13083-862, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, São Paulo, Brazil
| | - Stevens K Rehen
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil
- Department of Genetics, Institute of Biology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil.
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil.
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, 13083-862, Brazil.
- Instituto Nacional de Biomarcadores em Neuropsiquiatria (INBION), Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil.
| |
Collapse
|
5
|
Yang S, Yi L, Xia X, Chen X, Hou X, Zhang L, Yang F, Liao J, Han Z, Fu Y. Transcriptome comparative analysis of amygdala-hippocampus in depression: A rat model induced by chronic unpredictable mild stress (CUMS). J Affect Disord 2023; 334:258-270. [PMID: 37105469 DOI: 10.1016/j.jad.2023.04.074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 04/11/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND Depression is a common and complex mental disease, and its pathogenesis involves several brain regions. Abnormalities in the amygdala-hippocampal neural circuits have been shown to be involved in depression. However, the underlying molecular mechanisms remain unclear. METHODS A rat model was used to determine the transcriptome changes in the amygdala-hippocampal neural network under chronic unpredictable mild stress (CUMS). Depression-related modules in this neural network were identified using weighted gene co-expression network analysis (WGCNA). Difference and enrichment analyses were used to determine differential gene expression in the two brain regions. RESULTS The modules in the amygdala and hippocampus associated with depression-like behavior contained 363 and 225 genes, respectively. Forty-two differentially expressed genes were identified in the amygdala candidate module and 37 in the hippocampus. Enrichment analysis showed that candidate genes in the amygdala were associated with neuronal myelination and candidate genes in the hippocampus were associated with synaptic transmission. Finally, based on module hub gene statistics, differential gene expression, and protein-protein interaction networks, 11 central genes were found in the amygdala candidate module, and one central gene was found in the hippocampal module. LIMITATIONS Our study was based on a rat CUMS model. Further evidence is needed to prove that our results are applicable to patients with depression. CONCLUSION This study identified critical modules and central genes involved in the amygdala-hippocampal circuit in the context of depression, and may provide further understanding of the pathogenesis of depression and help identify potential targets for antidepressant therapy.
Collapse
Affiliation(s)
- Shu Yang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Li Yi
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaodi Xia
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaolu Chen
- The First Branch, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiao Hou
- Department of Clinical Medicine, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Longjie Zhang
- Department of Pharmacy, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Fang Yang
- Department of pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Jiaxin Liao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhijie Han
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yixiao Fu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
6
|
Samaja M, Ottolenghi S. The Oxygen Cascade from Atmosphere to Mitochondria as a Tool to Understand the (Mal)adaptation to Hypoxia. Int J Mol Sci 2023; 24:ijms24043670. [PMID: 36835089 PMCID: PMC9960749 DOI: 10.3390/ijms24043670] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/05/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Hypoxia is a life-threatening challenge for about 1% of the world population, as well as a contributor to high morbidity and mortality scores in patients affected by various cardiopulmonary, hematological, and circulatory diseases. However, the adaptation to hypoxia represents a failure for a relevant portion of the cases as the pathways of potential adaptation often conflict with well-being and generate diseases that in certain areas of the world still afflict up to one-third of the populations living at altitude. To help understand the mechanisms of adaptation and maladaptation, this review examines the various steps of the oxygen cascade from the atmosphere to the mitochondria distinguishing the patterns related to physiological (i.e., due to altitude) and pathological (i.e., due to a pre-existing disease) hypoxia. The aim is to assess the ability of humans to adapt to hypoxia in a multidisciplinary approach that correlates the function of genes, molecules, and cells with the physiologic and pathological outcomes. We conclude that, in most cases, it is not hypoxia by itself that generates diseases, but rather the attempts to adapt to the hypoxia condition. This underlies the paradigm shift that when adaptation to hypoxia becomes excessive, it translates into maladaptation.
Collapse
Affiliation(s)
- Michele Samaja
- MAGI GROUP, San Felice del Benaco, 25010 Brescia, Italy
- Correspondence:
| | - Sara Ottolenghi
- School of Medicine and Surgery, University of Milano Bicocca, 20126 Milan, Italy
| |
Collapse
|
7
|
Hydrogel oxygen reservoirs increase functional integration of neural stem cell grafts by meeting metabolic demands. Nat Commun 2023; 14:457. [PMID: 36709345 PMCID: PMC9884236 DOI: 10.1038/s41467-023-36133-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 01/17/2023] [Indexed: 01/29/2023] Open
Abstract
Injectable biomimetic hydrogels have great potential for use in regenerative medicine as cellular delivery vectors. However, they can suffer from issues relating to hypoxia, including poor cell survival, differentiation, and functional integration owing to the lack of an established vascular network. Here we engineer a hybrid myoglobin:peptide hydrogel that can concomitantly deliver stem cells and oxygen to the brain to support engraftment until vascularisation can occur naturally. We show that this hybrid hydrogel can modulate cell fate specification within progenitor cell grafts, resulting in a significant increase in neuronal differentiation. We find that the addition of myoglobin to the hydrogel results in more extensive innervation within the host tissue from the grafted cells, which is essential for neuronal replacement strategies to ensure functional synaptic connectivity. This approach could result in greater functional integration of stem cell-derived grafts for the treatment of neural injuries and diseases affecting the central and peripheral nervous systems.
Collapse
|
8
|
Serial Gene Expression Profiling of Neural Stem Cells Shows Transcriptome Switch by Long-Term Physioxia from Metabolic Adaption to Cell Signaling Profile. Stem Cells Int 2022; 2022:6718640. [PMID: 36411871 PMCID: PMC9675612 DOI: 10.1155/2022/6718640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
Oxygen is an essential factor in the cellular microenvironment with pivotal effects on neural development with a particular sensitivity of midbrain neural stem cells (NSCs) to high atmospheric oxygen tension. However, most experiments are still performed at atmospheric O2 levels (21%, normoxia), whereas mammalian brain tissue is physiologically exposed to substantially lower O2 tensions around 3% (physioxia). We here performed serial Affymetrix gene array analyses to detect expression changes in mouse fetal NSCs from both midbrain and cortical tissues when kept at physioxia compared to normoxia. We identified more than 400 O2-regulated genes involved in cellular metabolism, cell proliferation/differentiation, and various signaling pathways. NSCs from both regions showed a low number but high conformity of regulated genes (9 genes in midbrain vs. 34 in cortical NSCs; 8 concordant expression changes) after short-term physioxia (2 days) with metabolic processes and cellular processes being the most prominent GO categories pointing to cellular adaption to lower oxygen levels. Gene expression profiles changed dramatically after long-term physioxia (13 days) with a higher number of regulated genes and more diverse expression patterns when comparing the two NSC types (338 genes in midbrain vs. 121 in cortical NSCs; 75 concordant changes). Most prominently, we observed a reduction of hits in metabolic processes but an increase in biological regulation and signaling pointing to a switch towards signaling processes and stem cell maintenance. Our data may serve as a basis for identifying potential signaling pathways that maintain stem cell characteristics in cortical versus midbrain physioxic stem cell niches.
Collapse
|
9
|
A Tale of Two: When Neural Stem Cells Encounter Hypoxia. Cell Mol Neurobiol 2022:10.1007/s10571-022-01293-6. [DOI: 10.1007/s10571-022-01293-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/02/2022] [Indexed: 11/12/2022]
|
10
|
Augustyniak J, Lipka G, Kozlowska H, Caloni F, Buzanska L. Oxygen as an important factor modulating in vitro MeHgCl toxicity associated with mitochondrial genes in hiPSCs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 241:113737. [PMID: 35696963 DOI: 10.1016/j.ecoenv.2022.113737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
Mitochondria are energy factories of cells and important targets for methylmercury chloride (MgHgCl). Methylmercury (MeHg) is a well-known environmental toxicant that bioaccumulates in fish and shellfish. It readily crosses the placental barrier, making it a threat to correct fetal development. Despite being comprehensively investigated for years, this compound has not been assessed for its in vitro mitochondrial toxicity under different oxygen conditions. In this study, human induced pluripotent stem cells (hiPSCs) were used to evaluate the dependence of the expression of genes associated with pluripotency and mitochondria on atmospheric (21% O2) and low (5% O2) oxygen concentrations upon MeHgCl treatment. We showed that the toxicity of MeHgCl was strongly related to an increased mtDNA copy number and downregulation of the expression of an mtDNA replication and damage repair-associated gene POLG1 (Mitochondrial Polymerase Gamma Catalytic Subunit) in both tested oxygen conditions. In addition, the viability and mitochondrial membrane potential of hiPSCs were significantly lowered by MeHgCl regardless of the oxygen concentration. However, reactive oxygen species accumulation significantly increased only under atmospheric oxygen conditions; what was associated with increased expression of TFAM (Transcription Factor A, Mitochondrial) and NRF1 (Nuclear Respiratory Factor 1) and downregulation of PARK2 (Parkin RBR E3 Ubiquitin Protein Ligase). Taken together, our results demonstrated that MeHgCl could induce in vitro toxicity in hiPSCs through altering mitochondria-associated genes in an oxygen level-dependent manner. Thus, our work suggests that oxygen should be considered a factor was modulating the in vitro toxicity of environmental pollutants. Typical atmospheric conditions of in vitro culture significantly lower the predictive value of studies of such toxicity.
Collapse
Affiliation(s)
- J Augustyniak
- Department of Neurochemistry, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - G Lipka
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - H Kozlowska
- Laboratory of Advanced Microscopy Technique, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - F Caloni
- Department of Environmental Science and Policy (ESP), Università degli Studi di Milano, Milan, Italy
| | - L Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
11
|
Hokama Y, Nishimura M, Usugi R, Fujiwara K, Katagiri C, Takagi H, Ishiuchi S. Recovery from the damage of cranial radiation modulated by memantine, an NMDA receptor antagonist, combined with hyperbaric oxygen therapy. Neuro Oncol 2022; 25:108-122. [PMID: 35762568 PMCID: PMC9825311 DOI: 10.1093/neuonc/noac162] [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: 02/08/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Radiotherapy is an important treatment option for central nervous system malignancies. However, cranial radiation induces hippocampal dysfunction and white matter injury; this leads to cognitive dysfunction, and results in a reduced quality of life in patients. Excitatory glutamate signaling through N-methyl-d-aspartate receptors (NMDARs) plays a central role both in hippocampal neurogenesis and in the myelination of oligodendrocytes in the cerebrum. METHODS We provide a method for quantifying neurogenesis in human subjects in live brain during cancer therapy. Neuroimaging using originally created behavioral tasks was employed to examine human hippocampal memory pathway in patients with brain disorders. RESULTS Treatment with memantine, a non-competitive NMDAR antagonist, reversed impairment in hippocampal pattern separation networks as detected by functional magnetic resonance imaging. Hyperbaric preconditioning of the patients just before radiotherapy with memantine mostly reversed white matter injury as detected by whole brain analysis with Tract-Based Spatial Statics. Neuromodulation combined with the administration of hyperbaric oxygen therapy and memantine during radiotherapy facilitated the restoration of hippocampal function and white matter integrity, and improved higher cognitive function in patients receiving cranial radiation. CONCLUSIONS The method described herein, for diagnosis of hippocampal dysfunction, and therapeutic intervention can be utilized to restore some of the cognitive decline experienced by patients who have received cranial radiation. The underlying mechanism of restoration is the production of new neurons, which enhances functionality in pattern separation networks in the hippocampi, resulting in an increase in cognitive score, and restoration of microstructural integrity of white matter tracts revealed by Tract-Based Spatial Statics Analysis.
Collapse
Affiliation(s)
- Yohei Hokama
- Department of Neurosurgery, Graduate School of Medicine, University of The Ryukyus, 207 Uehara, Nishihara-machi, Okinawa 903-0215, Japan
| | - Masahiko Nishimura
- Department of Neurosurgery, Graduate School of Medicine, University of The Ryukyus, 207 Uehara, Nishihara-machi, Okinawa 903-0215, Japan
| | - Ryoichi Usugi
- Department of Neurosurgery, Graduate School of Medicine, University of The Ryukyus, 207 Uehara, Nishihara-machi, Okinawa 903-0215, Japan
| | - Kyoko Fujiwara
- Department of Neurosurgery, Graduate School of Medicine, University of The Ryukyus, 207 Uehara, Nishihara-machi, Okinawa 903-0215, Japan
| | - Chiaki Katagiri
- Department of Neurosurgery, Graduate School of Medicine, University of The Ryukyus, 207 Uehara, Nishihara-machi, Okinawa 903-0215, Japan
| | - Hiroshi Takagi
- Department of Neurosurgery, Graduate School of Medicine, University of The Ryukyus, 207 Uehara, Nishihara-machi, Okinawa 903-0215, Japan
| | - Shogo Ishiuchi
- Corresponding Author: Dr. Shogo Ishiuchi, Department of Neurosurgery, Graduate School of Medicine, University of The Ryukyus, 207 Uehara, Nishihara-machi, Okinawa 903-0215, Japan ()
| |
Collapse
|
12
|
Atherton E, Hu Y, Brown S, Papiez E, Ling V, Colvin V, Borton D. A 3D in vitro model of the device-tissue interface: Functional and structural symptoms of innate neuroinflammation are mitigated by antioxidant ceria nanoparticles. J Neural Eng 2022; 19. [PMID: 35447619 DOI: 10.1088/1741-2552/ac6908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/20/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE The recording instability of neural implants due to neuroinflammation at the device-tissue interface is a primary roadblock to broad adoption of brain-machine interfaces. While a multiphasic immune response, marked by glial scaring, oxidative stress (OS), and neurodegeneration, is well-characterized, the independent contributions of systemic and local "innate" immune responses are not well-understood. We aimed to understand and mitigate the isolated the innate neuroinflammatory response to devices. APPROACH Three-dimensional primary neural cultures provide a unique environment for studying the drivers of neuroinflammation by decoupling the innate and systemic immune systems, while conserving an endogenous extracellular matrix and structural and functional network complexity. We created a three-dimensional in vitro model of the DTI by seeding primary cortical cells around microwires. Live imaging of both dye and AAV-mediated functional, structural, and lipid peroxidation fluorescence was employed to characterize the neuroinflammatory response. MAIN RESULTS Live imaging of microtissues over time revealed independent innate neuroinflammation, marked by increased OS, decreased neuronal density, and increased functional connectivity. We demonstrated the use of this model for therapeutic screening by directly applying drugs to neural tissue, bypassing low bioavailability through the in vivo blood brain barrier. As there is growing interest in long-acting antioxidant therapies, we tested efficacy of "perpetual" antioxidant ceria nanoparticles, which reduced OS, increased neuronal density, and protected functional connectivity. SIGNIFICANCE Our 3D in vitro model of the device-tissue interface exhibited symptoms of OS-mediated innate neuroinflammation, indicating a significant local immune response to devices. The dysregulation of functional connectivity of microcircuits surround implants suggests the presence of an observer effect, in which the process of recording neural activity may fundamentally change the neural signal. Finally, the demonstration of antioxidant ceria nanoparticle treatment exhibited substantial promise as a neuroprotective and anti-inflammatory treatment strategy.
Collapse
Affiliation(s)
- Elaina Atherton
- School of Engineering, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Yue Hu
- Department of Chemistry, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Sophie Brown
- School of Engineering, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Emily Papiez
- School of Engineering, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Vivian Ling
- Department of Chemistry, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Vicki Colvin
- Department of Chemistry, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - David Borton
- School of Engineering, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| |
Collapse
|
13
|
Cao J, Chan WC, Chow MSS. Use of conditional reprogramming cell, patient derived xenograft and organoid for drug screening for individualized prostate cancer therapy: Current and future perspectives (Review). Int J Oncol 2022; 60:52. [PMID: 35322860 DOI: 10.3892/ijo.2022.5342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/14/2022] [Indexed: 11/06/2022] Open
Abstract
Prostate cancer mortality is ranked second among all cancer mortalities in men worldwide. There is a great need for a method of efficient drug screening for precision therapy, especially for patients with existing drug‑resistant prostate cancer. Based on the concept of bacterial cell culture and drug sensitivity testing, the traditional approach of cancer drug screening is inadequate. The current and more innovative use of cancer cell culture and in vivo tumor models in drug screening for potential individualization of anti‑cancer therapy is reviewed and discussed in the present review. An ideal screening model would have the ability to identify drug activity for the targeted cells resembling what would have occurred in the in vivo environment. Based on this principle, three available cell culture/tumor screening models for prostate cancer are reviewed and considered. The culture conditions, advantages and disadvantages for each model together with ideas to best utilize these models are discussed. The first screening model uses conditional reprogramed cells derived from patient cancer cells. Although these cells are convenient to grow and use, they are likely to have different markers and characteristics from original tumor cells and thus not likely to be informative. The second model employs patient derived xenograft (PDX) which resembles an in vivo approach, but its main disadvantages are that it cannot be easily genetically modified and it is not suitable for high‑throughput drug screening. Finally, high‑throughput screening is more feasible with tumor organoids grown from patient cancer cells. The last system still needs a large number of tumor cells. It lacks in situ blood vessels, immune cells and the extracellular matrix. Based on these current models, future establishment of an organoid data bank would allow the selection of a specific organoid resembling that of an individual's prostate cancer and used for screening of suitable anticancer drugs. This can be further confirmed using the PDX model. Thus, this combined organoid‑PDX approach is expected to be able to provide the drug sensitivity testing approach for individualization of prostate cancer therapy in the near future.
Collapse
Affiliation(s)
- Jessica Cao
- College of Osteopathic Medicine of The Pacific, Western University of Health Sciences, Pomona, CA 91766‑1854, USA
| | - Wing C Chan
- City of Hope Comprehensive Cancer Center, City of Hope Medical Center, Duarte, CA 91010‑3012, USA
| | - Moses S S Chow
- College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766‑1854, USA
| |
Collapse
|
14
|
Egger D, Lavrentieva A, Kugelmeier P, Kasper C. Physiologic isolation and expansion of human mesenchymal stem/stromal cells for manufacturing of cell-based therapy products. Eng Life Sci 2022; 22:361-372. [PMID: 35382547 PMCID: PMC8961040 DOI: 10.1002/elsc.202100097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 01/04/2023] Open
Abstract
The utilization of mesenchymal stem/stromal cells raises new hopes in treatment of diseases and pathological conditions, while at the same time bringing immense challenges for researchers, manufacturers and physicians. It is essential to consider all steps along the in vitro fabrication of cell-based products in order to reach efficient and reproducible treatment outcomes. Here, the optimal protocols for isolation, cultivation and differentiation of mesenchymal stem cells are required. In this review we discuss these aspects and their influence on the final cell-based product quality. We demonstrate that physiological in vitro cell cultivation conditions play a crucial role in therapeutic functionalities of cultivated cells. We show that three-dimensional cell culture, dynamic culture conditions and physiologically relevant in vitro oxygen concentrations during isolation and expansion make a decisive contribution towards the improvement of cell-based products in regenerative medicine.
Collapse
Affiliation(s)
- Dominik Egger
- Department of BiotechnologyUniversity of Natural Resources and Life ScienceViennaAustria
| | | | | | - Cornelia Kasper
- Department of BiotechnologyUniversity of Natural Resources and Life ScienceViennaAustria
| |
Collapse
|
15
|
Uribe D, Niechi I, Rackov G, Erices JI, San Martín R, Quezada C. Adapt to Persist: Glioblastoma Microenvironment and Epigenetic Regulation on Cell Plasticity. BIOLOGY 2022; 11:313. [PMID: 35205179 PMCID: PMC8869716 DOI: 10.3390/biology11020313] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is the most frequent and aggressive brain tumor, characterized by great resistance to treatments, as well as inter- and intra-tumoral heterogeneity. GBM exhibits infiltration, vascularization and hypoxia-associated necrosis, characteristics that shape a unique microenvironment in which diverse cell types are integrated. A subpopulation of cells denominated GBM stem-like cells (GSCs) exhibits multipotency and self-renewal capacity. GSCs are considered the conductors of tumor progression due to their high tumorigenic capacity, enhanced proliferation, invasion and therapeutic resistance compared to non-GSCs cells. GSCs have been classified into two molecular subtypes: proneural and mesenchymal, the latter showing a more aggressive phenotype. Tumor microenvironment and therapy can induce a proneural-to-mesenchymal transition, as a mechanism of adaptation and resistance to treatments. In addition, GSCs can transition between quiescent and proliferative substates, allowing them to persist in different niches and adapt to different stages of tumor progression. Three niches have been described for GSCs: hypoxic/necrotic, invasive and perivascular, enhancing metabolic changes and cellular interactions shaping GSCs phenotype through metabolic changes and cellular interactions that favor their stemness. The phenotypic flexibility of GSCs to adapt to each niche is modulated by dynamic epigenetic modifications. Methylases, demethylases and histone deacetylase are deregulated in GSCs, allowing them to unlock transcriptional programs that are necessary for cell survival and plasticity. In this review, we described the effects of GSCs plasticity on GBM progression, discussing the role of GSCs niches on modulating their phenotype. Finally, we described epigenetic alterations in GSCs that are important for stemness, cell fate and therapeutic resistance.
Collapse
Affiliation(s)
- Daniel Uribe
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
| | - Ignacio Niechi
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
| | - Gorjana Rackov
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas (CNB-CSIC), 28049 Madrid, Spain;
| | - José I. Erices
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
| | - Rody San Martín
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
| | - Claudia Quezada
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia 5090000, Chile; (D.U.); (I.N.); (J.I.E.); (R.S.M.)
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| |
Collapse
|
16
|
Mikloska KV, Zrini ZA, Bernier NJ. Severe hypoxia exposure inhibits larval brain development but does not affect the capacity to mount a cortisol stress response in zebrafish. J Exp Biol 2021; 225:274120. [PMID: 34931659 DOI: 10.1242/jeb.243335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022]
Abstract
Fish nursery habitats are increasingly hypoxic and the brain is recognized as highly hypoxia-sensitive, yet there is a lack of information on the effects of hypoxia on the development and function of the larval fish brain. Here, we tested the hypothesis that by inhibiting brain development, larval exposure to severe hypoxia has persistent functional effects on the cortisol stress response in zebrafish (Danio rerio). Exposing 5 days post-fertilization (dpf) larvae to 10% dissolved O2 (DO) for 16 h only marginally reduced survival, but it decreased forebrain neural proliferation by 55%, and reduced the expression of neurod1, gfap, and mbpa, markers of determined neurons, glia, and oligodendrocytes, respectively. The 5 dpf hypoxic exposure also elicited transient increases in whole body cortisol and in crf, uts1, and hsd20b2 expression, key regulators of the endocrine stress response. Hypoxia exposure at 5 dpf also inhibited the cortisol stress response to hypoxia in 10 dpf larvae and increased hypoxia tolerance. However, 10% DO exposure at 5 dpf for 16h did not affect the cortisol stress response to a novel stressor in 10 dpf larvae or the cortisol stress response to hypoxia in adult fish. Therefore, while larval exposure to severe hypoxia can inhibit brain development, it also increases hypoxia tolerance. These effects may transiently reduce the impact of hypoxia on the cortisol stress response but not its functional capacity to respond to novel stressors. We conclude that the larval cortisol stress response in zebrafish has a high capacity to cope with severe hypoxia-induced neurogenic impairment.
Collapse
Affiliation(s)
- Kristina V Mikloska
- Department of Integrative Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Zoe A Zrini
- Department of Integrative Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Nicholas J Bernier
- Department of Integrative Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| |
Collapse
|
17
|
Abstract
Cerebral ischemic injury may lead to a series of serious brain diseases, death or different degrees of disability. Hypoxia-inducible factor-1α (HIF-1α) is an oxygen-sensitive transcription factor, which mediates the adaptive metabolic response to hypoxia and serves a key role in cerebral ischemia. HIF-1α is the main molecule that responds to hypoxia. HIF-1α serves an important role in the development of cerebral ischemia by participating in numerous processes, including metabolism, proliferation and angiogenesis. The present review focuses on the endogenous protective mechanism of cerebral ischemia and elaborates on the role of HIF-1α in cerebral ischemia. In addition, it focuses on cerebral ischemia interventions that act on the HIF-1α target, including biological factors, non-coding RNA, hypoxic-ischemic preconditioning and drugs, and expands upon the measures to strengthen the endogenous compensatory response to support HIF-1α as a therapeutic target, thus providing novel suggestions for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Peiliang Dong
- Institute of Traditional Chinese Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Qingna Li
- College of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Hua Han
- College of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
18
|
Afshari R, Akhavan O, Hamblin MR, Varma RS. Review of Oxygenation with Nanobubbles: Possible Treatment for Hypoxic COVID-19 Patients. ACS APPLIED NANO MATERIALS 2021; 4:11386-11412. [PMID: 37556289 PMCID: PMC8565459 DOI: 10.1021/acsanm.1c01907] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/12/2021] [Indexed: 05/05/2023]
Abstract
The coronavirus disease (COVID-19) pandemic, which has spread around the world, caused the death of many affected patients, partly because of the lack of oxygen arising from impaired respiration or blood circulation. Thus, maintaining an appropriate level of oxygen in the patients' blood by devising alternatives to ventilator systems is a top priority goal for clinicians. The present review highlights the ever-increasing application of nanobubbles (NBs), miniature gaseous vesicles, for the oxygenation of hypoxic patients. Oxygen-containing NBs can exert a range of beneficial physiologic and pharmacologic effects that include tissue oxygenation, as well as tissue repair mechanisms, antiinflammatory properties, and antibacterial activity. In this review, we provide a comprehensive survey of the application of oxygen-containing NBs, with a primary focus on the development of intravenous platforms. The multimodal functions of oxygen-carrying NBs, including antimicrobial, antiinflammatory, drug carrying, and the promotion of wound healing are discussed, including the benefits and challenges of using NBs as a treatment for patients with acute hypoxemic respiratory failure, particularly due to COVID-19.
Collapse
Affiliation(s)
- Ronak Afshari
- Department of Physics, Sharif University
of Technology, P.O. Box 11155-9161, Tehran 14588-89694,
Iran
| | - Omid Akhavan
- Department of Physics, Sharif University
of Technology, P.O. Box 11155-9161, Tehran 14588-89694,
Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science,
University of Johannesburg, Doornfontein 2028, South
Africa
| | - Rajender S. Varma
- Regional Center of Advanced Technologies and Materials,
Czech Advanced Technology and Research Institute, Palacky
University, Šlechtitelů 27, Olomouc 78371, Czech
Republic
| |
Collapse
|
19
|
Mennen RH, de Leeuw VC, Piersma AH. Cell differentiation in the cardiac embryonic stem cell test (ESTc) is influenced by the oxygen tension in its underlying embryonic stem cell culture. Toxicol In Vitro 2021; 77:105247. [PMID: 34537371 DOI: 10.1016/j.tiv.2021.105247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022]
Abstract
Oxygen (O2) levels in the mammalian embryo range between 2.4% and 8%. The cardiac embryonic stem cell test (ESTc) is a model for developmental toxicity predictions, which is usually performed under atmospheric O2 levels of 20%. We investigated the chemical sensitivity of the ESTc carried out under 20% O2, using embryonic stem cells (ESC) cultured under either 20% O2 or 5% O2. ESC viability was more sensitive to valproic acid (VPA) but less sensitive to flusilazole (FLU) when cultured under 5% versus 20% O2. For beating cardiomyocyte differentiation, lower ID50 values were found for FLU and VPA when the ESCs had been cultured under 5% versus 20% O2. At differentiation day 4, gene expression values were primarily driven by the level of O2 during ESC culture instead of exposure to FLU. In addition, using ESCs cultured under 5% O2 tension, VPA enhanced Nes (ectoderm) expression. Bmp4 (mesoderm) was enhanced by VPA when using ESCs cultured under 20% O2. At differentiation day 10, using ESCs cultured under 5% instead of 20% O2, Nkx2.5 and Myh6 (cardiomyocytes) were less affected after exposure to FLU or VPA. These results show that O2 tension in ESC culture influences chemical sensitivity in the ESTc. This enhances awareness of the standard culture conditions, which may impact the application of the ESTc in quantitative hazard assessment of chemicals.
Collapse
Affiliation(s)
- R H Mennen
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands.
| | - V C de Leeuw
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| | - A H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
20
|
Ngo MT, Harley BAC. Progress in mimicking brain microenvironments to understand and treat neurological disorders. APL Bioeng 2021; 5:020902. [PMID: 33869984 PMCID: PMC8034983 DOI: 10.1063/5.0043338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022] Open
Abstract
Neurological disorders including traumatic brain injury, stroke, primary and metastatic brain tumors, and neurodegenerative diseases affect millions of people worldwide. Disease progression is accompanied by changes in the brain microenvironment, but how these shifts in biochemical, biophysical, and cellular properties contribute to repair outcomes or continued degeneration is largely unknown. Tissue engineering approaches can be used to develop in vitro models to understand how the brain microenvironment contributes to pathophysiological processes linked to neurological disorders and may also offer constructs that promote healing and regeneration in vivo. In this Perspective, we summarize features of the brain microenvironment in normal and pathophysiological states and highlight strategies to mimic this environment to model disease, investigate neural stem cell biology, and promote regenerative healing. We discuss current limitations and resulting opportunities to develop tissue engineering tools that more faithfully recapitulate the aspects of the brain microenvironment for both in vitro and in vivo applications.
Collapse
Affiliation(s)
- Mai T. Ngo
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Brendan A. C. Harley
- Author to whom correspondence should be addressed:. Tel.: (217) 244-7112. Fax: (217) 333-5052
| |
Collapse
|
21
|
Hwang I, Tang D, Paik J. Oxidative stress sensing and response in neural stem cell fate. Free Radic Biol Med 2021; 169:74-83. [PMID: 33862161 PMCID: PMC9594080 DOI: 10.1016/j.freeradbiomed.2021.03.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/13/2021] [Accepted: 03/25/2021] [Indexed: 12/22/2022]
Abstract
Neural stem/progenitor cells (NSPCs) contribute to the physiological cellular turnover of the adult brain and make up its regenerative potential. It is thus essential to understand how different factors influence their proliferation and differentiation to gain better insight into potential therapeutic targets in neurodegenerative diseases and traumatic brain injuries. Recent evidences indicate the roles of redox stress sensing and coping mechanisms in mediating the balance between NSPC self-renewal and differentiation. Such mechanisms involve direct cysteine modification, signaling and metabolic reprogramming, epigenetic alterations and transcription changes leading to adaptive responses like autophagy. Here, we discuss emerging findings on the involvement of redox sensors and effectors and their mechanisms in influencing changes in cellular redox potential and NSPC fate.
Collapse
Affiliation(s)
- Inah Hwang
- R&D Center, OneCureGEN Co., Ltd, Daejeon, 34141, Republic of Korea; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Deanna Tang
- University of Chicago, Chicago, IL, 60637, USA
| | - Jihye Paik
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
22
|
Implications of Extended Inhibitory Neuron Development. Int J Mol Sci 2021; 22:ijms22105113. [PMID: 34066025 PMCID: PMC8150951 DOI: 10.3390/ijms22105113] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/23/2022] Open
Abstract
A prolonged developmental timeline for GABA (γ-aminobutyric acid)-expressing inhibitory neurons (GABAergic interneurons) is an amplified trait in larger, gyrencephalic animals. In several species, the generation, migration, and maturation of interneurons take place over several months, in some cases persisting after birth. The late integration of GABAergic interneurons occurs in a region-specific pattern, especially during the early postnatal period. These changes can contribute to the formation of functional connectivity and plasticity, especially in the cortical regions responsible for higher cognitive tasks. In this review, we discuss GABAergic interneuron development in the late gestational and postnatal forebrain. We propose the protracted development of interneurons at each stage (neurogenesis, neuronal migration, and network integration), as a mechanism for increased complexity and cognitive flexibility in larger, gyrencephalic brains. This developmental feature of interneurons also provides an avenue for environmental influences to shape neural circuit formation.
Collapse
|
23
|
Tuy K, Rickenbacker L, Hjelmeland AB. Reactive oxygen species produced by altered tumor metabolism impacts cancer stem cell maintenance. Redox Biol 2021; 44:101953. [PMID: 34052208 PMCID: PMC8212140 DOI: 10.1016/j.redox.2021.101953] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/11/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Controlling reactive oxygen species (ROS) at sustainable levels can drive multiple facets of tumor biology, including within the cancer stem cell (CSC) population. Tight regulation of ROS is one key component in CSCs that drives disease recurrence, cell signaling, and therapeutic resistance. While ROS are well-appreciated to need oxygen and are a product of oxidative phosphorylation, there are also important roles for ROS under hypoxia. As hypoxia promotes and sustains major stemness pathways, further consideration of ROS impacts on CSCs in the tumor microenvironment is important. Furthermore, glycolytic shifts that occur in cancer and may be promoted by hypoxia are associated with multiple mechanisms to mitigate oxidative stress. This altered metabolism provides survival advantages that sustain malignant features, such as proliferation and self-renewal, while producing the necessary antioxidants that reduce damage from oxidative stress. Finally, disease recurrence is believed to be attributed to therapy resistant CSCs which can be quiescent and have changes in redox status. Effective DNA damage response pathways and/or a slow-cycling state can protect CSCs from the genomic catastrophe induced by irradiation and genotoxic agents. This review will explore the delicate, yet complex, relationship between ROS and its pleiotropic role in modulating the CSC.
Collapse
Affiliation(s)
- Kaysaw Tuy
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lucas Rickenbacker
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anita B Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
24
|
Khuu MA, Nallamothu T, Castro-Rivera CI, Arias-Cavieres A, Szujewski CC, Garcia Iii AJ. Stage-dependent effects of intermittent hypoxia influence the outcome of hippocampal adult neurogenesis. Sci Rep 2021; 11:6005. [PMID: 33727588 PMCID: PMC7966401 DOI: 10.1038/s41598-021-85357-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 02/25/2021] [Indexed: 12/14/2022] Open
Abstract
Over one billion adults worldwide are estimated to suffer from sleep apnea, a condition with wide-reaching effects on brain health. Sleep apnea causes cognitive decline and is a risk factor for neurodegenerative conditions such as Alzheimer’s disease. Rodents exposed to intermittent hypoxia (IH), a hallmark of sleep apnea, exhibit spatial memory deficits associated with impaired hippocampal neurophysiology and dysregulated adult neurogenesis. We demonstrate that IH creates a pro-oxidant condition that reduces the Tbr2+ neural progenitor pool early in the process, while also suppressing terminal differentiation of adult born neurons during late adult neurogenesis. We further show that IH-dependent cell-autonomous hypoxia inducible factor 1-alpha (HIF1a) signaling is activated in early neuroprogenitors and enhances the generation of adult born neurons upon termination of IH. Our findings indicate that oscillations in oxygen homeostasis, such as those found in sleep apnea, have complex stage-dependent influence over hippocampal adult neurogenesis.
Collapse
Affiliation(s)
- Maggie A Khuu
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - Thara Nallamothu
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - Carolina I Castro-Rivera
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA.,Committee On Neurobiology, The University of Chicago, Chicago, IL, 60307, USA.,Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, 60637, USA
| | - Alejandra Arias-Cavieres
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA
| | - Caroline C Szujewski
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA.,Committee On Neurobiology, The University of Chicago, Chicago, IL, 60307, USA.,Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, 60637, USA
| | - Alfredo J Garcia Iii
- Institute for Integrative Physiology, Section of Emergency Medicine, The University of Chicago, 5841 S Maryland Ave, Chicago, IL, 60637, USA. .,Committee On Neurobiology, The University of Chicago, Chicago, IL, 60307, USA. .,Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
25
|
Santini MP, Malide D, Hoffman G, Pandey G, D'Escamard V, Nomura-Kitabayashi A, Rovira I, Kataoka H, Ochando J, Harvey RP, Finkel T, Kovacic JC. Tissue-Resident PDGFRα + Progenitor Cells Contribute to Fibrosis versus Healing in a Context- and Spatiotemporally Dependent Manner. Cell Rep 2021; 30:555-570.e7. [PMID: 31940496 DOI: 10.1016/j.celrep.2019.12.045] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/11/2019] [Accepted: 12/12/2019] [Indexed: 11/24/2022] Open
Abstract
PDGFRα+ mesenchymal progenitor cells are associated with pathological fibro-adipogenic processes. Conversely, a beneficial role for these cells during homeostasis or in response to revascularization and regeneration stimuli is suggested, but remains to be defined. We studied the molecular profile and function of PDGFRα+ cells in order to understand the mechanisms underlying their role in fibrosis versus regeneration. We show that PDGFRα+ cells are essential for tissue revascularization and restructuring through injury-stimulated remodeling of stromal and vascular components, context-dependent clonal expansion, and ultimate removal of pro-fibrotic PDGFRα+-derived cells. Tissue ischemia modulates the PDGFRα+ phenotype toward cells capable of remodeling the extracellular matrix and inducing cell-cell and cell-matrix adhesion, likely favoring tissue repair. Conversely, pathological healing occurs if PDGFRα+-derived cells persist as terminally differentiated mesenchymal cells. These studies support a context-dependent "yin-yang" biology of tissue-resident mesenchymal progenitor cells, which possess an innate ability to limit injury expansion while also promoting fibrosis in an unfavorable environment.
Collapse
Affiliation(s)
- Maria Paola Santini
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA.
| | - Daniela Malide
- Light Microscopy Core Facility, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Gabriel Hoffman
- Icahn Institute for Data Science and Genomic Technology, ISMMS, New York, NY 10029, USA
| | - Gaurav Pandey
- Icahn Institute for Data Science and Genomic Technology, ISMMS, New York, NY 10029, USA
| | - Valentina D'Escamard
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - Aya Nomura-Kitabayashi
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA
| | - Ilsa Rovira
- Center for Molecular Medicine, NHLBI, NIH, Bethesda, MD 20892, USA
| | | | - Jordi Ochando
- Department of Medicine and Oncological Sciences, ISMMS, New York, NY 10029, USA
| | - Richard P Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, UNSW Sydney, Kensington, NSW 2052, Australia; Stem Cells Australia, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Toren Finkel
- Aging Institute, University of Pittsburgh/UPMC, 100 Technology Drive, Pittsburgh, PA 15219, USA
| | - Jason C Kovacic
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY 10029, USA.
| |
Collapse
|
26
|
Hypoxia-inducible factor-2α is crucial for proper brain development. Sci Rep 2020; 10:19146. [PMID: 33154420 PMCID: PMC7644612 DOI: 10.1038/s41598-020-75838-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 10/21/2020] [Indexed: 11/09/2022] Open
Abstract
Sufficient tissue oxygenation is required for regular brain function; thus oxygen supply must be tightly regulated to avoid hypoxia and irreversible cell damage. If hypoxia occurs the transcription factor complex hypoxia-inducible factor (HIF) will accumulate and coordinate adaptation of cells to hypoxia. However, even under atmospheric O2 conditions stabilized HIF-2α protein was found in brains of adult mice. Mice with a neuro-specific knockout of Hif-2α showed a reduction of pyramidal neurons in the retrosplenial cortex (RSC), a brain region responsible for a range of cognitive functions, including memory and navigation. Accordingly, behavioral studies showed disturbed cognitive abilities in these mice. In search of the underlying mechanisms for the specific loss of pyramidal cells in the RSC, we found deficits in migration in neural stem cells from Hif-2α knockout mice due to altered expression patterns of genes highly associated with neuronal migration and positioning.
Collapse
|
27
|
Wang X, Shen K, Wang J, Liu K, Wu G, Li Y, Luo L, Zheng Z, Hu D. Hypoxic preconditioning combined with curcumin promotes cell survival and mitochondrial quality of bone marrow mesenchymal stem cells, and accelerates cutaneous wound healing via PGC-1α/SIRT3/HIF-1α signaling. Free Radic Biol Med 2020; 159:164-176. [PMID: 32745765 DOI: 10.1016/j.freeradbiomed.2020.07.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/10/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022]
Abstract
Restrained survival and function of relocated bone marrow mesenchymal stem cells (BMSCs) is a major impediment to BMSCs-mediated tissue repair. Accumulating evidences have indicated that hypoxic preconditioning of BMSCs could enhance BMSCs' adaptability after transplantation and thus improve their therapeutic properties. Curcumin, a natural dietary product, is known to exert profound protective effects on various cellular processes. Here we showed that mild hypoxic preconditioning combined with curcumin significantly increased cell survival, enriched more cells in G2/M and S phase, and improved mitochondrial function in BMSCs. Meanwhile, hypoxic preconditioning combined with curcumin altered mitochondrial cristae shape and strongly inhibited mitochondrial cytochrome c release, which consequently suppressed an apoptosis signal as revealed by reduced caspase-3 cleavage in BMSCs. Moreover, hypoxic preconditioning remarkably promoted mitochondrial quality via increasing mitochondrial fusion and elevating the activity of oxidative phosphorylation (OXPHOS) and mitochondrial complex Ⅰ enzyme in BMSCs, which were in accordance with the up-regulated expression of OPA1, PINK1 and Parkin. At the mechanistic level, the destabilization of HIF-1α and the up-regulated expression of PGC-1α and SIRT3 synergistically contributed to the protective effects of hypoxic preconditioning combined with curcumin in BMSCs. The proteasome inhibitor MG132 stabilized HIF-1a expression, but not PGC-1α or SIRT3, and dramatically restrained BMSCs survival under hypoxia combined with curcumin condition. MG132 also increased mitochondrial superoxide and intracellular hydrogen peroxide (H2O2) production and caspase-3 activation in hypoxia combined with curcumin-treated BMSCs. Furthermore, knockdown of SIRT3 and PGC-1α by RNAi both led to caspase-3 activation in BMSCs after hypoxia and curcumin treatment. Notably, SIRT3 RNAi suppressed OXPHOS activity, while PGC-1α RNAi triggered mitochondrial superoxide and intracellular H2O2 production in hypoxia combined with curcumin-treated BMSCs. Finally, we showed that hypoxia combined with curcumin-treated BMSCs accelerated the cutaneous wound healing process in a mice wound model. Overall, this study suggests that hypoxic preconditioning combined with curcumin could serve as an attractive strategy for facilitating BMSCs-mediated tissue repair, and further sheds new light on the rich repertoire of PGC-1α/SIRT3/HIF-1α signaling involved in the regulation of mitochondrial quality and function for cellular adaption to hypoxia.
Collapse
Affiliation(s)
- Xujie Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, China
| | - Kuo Shen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, China
| | - Jing Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, China
| | - Kaituo Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, China
| | - Gaofeng Wu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, China
| | - Yan Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, China
| | - Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, China
| | - Zhao Zheng
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, China.
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shannxi 710032, China.
| |
Collapse
|
28
|
Voss DM, Sloan A, Spina R, Ames HM, Bar EE. The Alternative Splicing Factor, MBNL1, Inhibits Glioblastoma Tumor Initiation and Progression by Reducing Hypoxia-Induced Stemness. Cancer Res 2020; 80:4681-4692. [PMID: 32928918 DOI: 10.1158/0008-5472.can-20-1233] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/23/2020] [Accepted: 09/09/2020] [Indexed: 12/31/2022]
Abstract
Muscleblind-like proteins (MBNL) belong to a family of tissue-specific regulators of RNA metabolism that control premessenger RNA splicing. Inactivation of MBNL causes an adult-to-fetal alternative splicing transition, resulting in the development of myotonic dystrophy. We have previously shown that the aggressive brain cancer, glioblastoma (GBM), maintains stem-like features (glioma stem cell, GSC) through hypoxia-induced responses. Accordingly, we hypothesize here that hypoxia-induced responses in GBM might also include MBNL-based alternative splicing to promote tumor progression. When cultured in hypoxia condition, GSCs rapidly exported muscleblind-like-1 (MBNL1) out of the nucleus, resulting in significant inhibition of MBNL1 activity. Notably, hypoxia-regulated inhibition of MBNL1 also resulted in evidence of adult-to-fetal alternative splicing transitions. Forced expression of a constitutively active isoform of MBNL1 inhibited GSC self-renewal and tumor initiation in orthotopic transplantation models. Induced expression of MBNL1 in established orthotopic tumors dramatically inhibited tumor progression, resulting in significantly prolonged survival. This study reveals that MBNL1 plays an essential role in GBM stemness and tumor progression, where hypoxic responses within the tumor inhibit MBNL1 activity, promoting stem-like phenotypes and tumor growth. Reversing these effects on MBNL1 may therefore, yield potent tumor suppressor activities, uncovering new therapeutic opportunities to counter this disease. SIGNIFICANCE: This study describes an unexpected mechanism by which RNA-binding protein, MBNL1, activity is inhibited in hypoxia by a simple isoform switch to regulate glioma stem cell self-renewal, tumorigenicity, and progression.
Collapse
Affiliation(s)
- Dillon M Voss
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Anthony Sloan
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Raffaella Spina
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Heather M Ames
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Eli E Bar
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland. .,Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
29
|
An efficient neuron-astrocyte differentiation protocol from human embryonic stem cell-derived neural progenitors to assess chemical-induced developmental neurotoxicity. Reprod Toxicol 2020; 98:107-116. [PMID: 32931842 DOI: 10.1016/j.reprotox.2020.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/13/2020] [Accepted: 09/07/2020] [Indexed: 01/10/2023]
Abstract
Human embryonic stem cell neuronal differentiation models provide promising in vitro tools for the prediction of developmental neurotoxicity of chemicals. Such models mimic essential elements of human relevant neuronal development, including the differentiation of a variety of brain cell types and their neuronal network formation as evidenced by specific gene and protein biomarkers. However, the reproducibility and lengthy culture duration of cell models present drawbacks and delay regulatory implementation. Here we present a relatively short and robust protocol to differentiate H9-derived neural progenitor cells (NPCs) into a neuron-astrocyte co-culture. When frozen-stored NPCs were re-cultured and induced into neuron-astrocyte differentiation, they showed gene- and protein expression typical for these cells, and most notably they exhibited spontaneous electrical activity within three days of culture as measured by a multi-well micro-electrode array. Modulating the ratio of astrocytes and neurons through different growth factors including glial cell line-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), and ciliary neurotrophic factor (CNTF) did not compromise the ability to develop spontaneous electrical activity. This robust neuronal differentiation model may serve as a functional component of a testing strategy for unravelling mechanisms of developmental neurotoxicity.
Collapse
|
30
|
Baccino-Calace M, Prieto D, Cantera R, Egger B. Compartment and cell-type specific hypoxia responses in the developing Drosophila brain. Biol Open 2020; 9:9/8/bio053629. [PMID: 32816692 PMCID: PMC7449796 DOI: 10.1242/bio.053629] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Environmental factors such as the availability of oxygen are instructive cues that regulate stem cell maintenance and differentiation. We used a genetically encoded biosensor to monitor the hypoxic state of neural cells in the larval brain of Drosophila. The biosensor reveals brain compartment and cell-type specific levels of hypoxia. The values correlate with differential tracheolation that is observed throughout development between the central brain and the optic lobe. Neural stem cells in both compartments show the strongest hypoxia response while intermediate progenitors, neurons and glial cells reveal weaker responses. We demonstrate that the distance between a cell and the next closest tracheole is a good predictor of the hypoxic state of that cell. Our study indicates that oxygen availability appears to be the major factor controlling the hypoxia response in the developing Drosophila brain and that cell intrinsic and cell-type specific factors contribute to modulate the response in an unexpected manner. This article has an associated First Person interview with the first author of the paper. Summary: A fluorescent biosensor reveals cell type specific hypoxia levels in the Drosophila brain in unprecedented detail. It paves the way for further functional studies addressing the role of oxygen in neural stem cell maintenance and differentiation.
Collapse
Affiliation(s)
- Martin Baccino-Calace
- Developmental Neurobiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | - Daniel Prieto
- Developmental Neurobiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | - Rafael Cantera
- Developmental Neurobiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay.,Zoology Department, Stockholm University, Stockholm 106 91, Sweden
| | - Boris Egger
- Department of Biology, University of Fribourg, Fribourg CH-1700, Switzerland
| |
Collapse
|
31
|
Wu X, Wang S, Li M, Li J, Shen J, Zhao Y, Pang J, Wen Q, Chen M, Wei B, Kaboli PJ, Du F, Zhao Q, Cho CH, Wang Y, Xiao Z, Wu X. Conditional reprogramming: next generation cell culture. Acta Pharm Sin B 2020; 10:1360-1381. [PMID: 32963937 PMCID: PMC7488362 DOI: 10.1016/j.apsb.2020.01.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Long-term primary culture of mammalian cells has been always difficult due to unavoidable senescence. Conventional methods for generating immortalized cell lines usually require manipulation of genome which leads to change of important biological and genetic characteristics. Recently, conditional reprogramming (CR) emerges as a novel next generation tool for long-term culture of primary epithelium cells derived from almost all origins without alteration of genetic background of primary cells. CR co-cultures primary cells with inactivated mouse 3T3-J2 fibroblasts in the presence of RHO-related protein kinase (ROCK) inhibitor Y-27632, enabling primary cells to acquire stem-like characteristics while retain their ability to fully differentiate. With only a few years' development, CR shows broad prospects in applications in varied areas including disease modeling, regenerative medicine, drug evaluation, drug discovery as well as precision medicine. This review is thus to comprehensively summarize and assess current progress in understanding mechanism of CR and its wide applications, highlighting the value of CR in both basic and translational researches and discussing the challenges faced with CR.
Collapse
Key Words
- 3T3-J2 fibroblast
- AACR, American Association for Cancer Research
- ACC, adenoid cystic carcinoma
- AR, androgen receptor
- CFTR, cystic fibrosis transmembrane conductance regulators
- CR, conditional reprogramming
- CYPs, cytochrome P450 enzymes
- Conditional reprogramming
- DCIS, ductal carcinoma in situ
- ECM, extracellular matrix
- ESC, embryonic stem cell
- HCMI, human cancer model initiatives
- HGF, hepatocyte growth factor
- HNE, human nasal epithelial
- HPV, human papillomaviruses
- ICD, intracellular domain
- LECs, limbal epithelial cells
- NCI, National Cancer Institute
- NGFR, nerve growth factor receptor
- NSCLC, non-small cell lung cancer
- NSG, NOD/SCID/gamma
- PDAC, pancreatic ductal adenocarcinoma
- PDX, patient derived xenograft
- PP2A, protein phosphatase 2A
- RB, retinoblastoma-associated protein
- ROCK
- ROCK, Rho kinase
- SV40, simian virus 40 large tumor antigen
- Senescence
- UVB, ultraviolet radiation b
- Y-27632
- dECM, decellularized extracellular matrix
- hASC, human adipose stem cells
- hTERT, human telomerase reverse transcriptase
- iPSCs, induction of pluripotent stem cells
- ΔNP63α, N-terminal truncated form of P63α
Collapse
Affiliation(s)
- Xiaoxiao Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jing Li
- Department of Oncology and Hematology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jun Pang
- Center of Radiation Oncology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou 646000, China
| | - Qinglian Wen
- Department of Oncology, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou 646000, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Parham Jabbarzadeh Kaboli
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Qijie Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| |
Collapse
|
32
|
Mennen RH, de Leeuw VC, Piersma AH. Oxygen tension influences embryonic stem cell maintenance and has lineage specific effects on neural and cardiac differentiation. Differentiation 2020; 115:1-10. [PMID: 32738735 DOI: 10.1016/j.diff.2020.07.001] [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: 05/19/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023]
Abstract
The importance of oxygen tension in in vitro cultures and its effect on embryonic stem cell (ESC) differentiation has been widely acknowledged. Research has mainly focussed on ESC maintenance or on one line of differentiation and only few studies have examined the potential relation between oxygen tension during ESC maintenance and differentiation. In this study we investigated the influence of atmospheric (20%) versus physiologic (5%) oxygen tension in ESC cultures and their differentiation within the cardiac and neural embryonic stem cell tests (ESTc, ESTn). Oxygen tension was set at 5% or 20% and cells were kept in these conditions from starting up cell culture until use for differentiation. Under these oxygen tensions, ESC culture showed no differences in proliferation and gene and protein expression levels. Differentiation was either performed in the same or in the alternative oxygen tension compared to ESC culture creating four different experimental conditions. Cardiac differentiation in 5% instead of 20% oxygen resulted in reduced development of spontaneously beating cardiomyocytes and lower expression of cardiac markers Nkx2.5, Myh6 and MF20 (myosin), regardless whether ESC had been cultured in 5% or 20% oxygen tension. As compared to the control (20% oxygen during stem cell maintenance and differentiation), neural differentiation in 5% oxygen with ESC cultured in 20% oxygen led to more cardiac and neural crest cell differentiation. The opposite experimental condition of neural differentiation in 20% oxygen with ESC cultured in 5% oxygen resulted in more glial differentiation. ESC that were maintained and differentiated in 5% oxygen showed an increase in neural crest and oligodendrocytes as compared to 20% oxygen during stem cell maintenance and differentiation. This study showed major effects on ESC differentiation in ESTc and ESTn of oxygen tension, which is an important variable to consider when designing and developing a stem cell-based in vitro system.
Collapse
Affiliation(s)
- Regina H Mennen
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands.
| | - Victoria C de Leeuw
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| | - Aldert H Piersma
- Center for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
33
|
Lee HY, Hong IS. Metabolic Regulation and Related Molecular Mechanisms in Various Stem Cell Functions. Curr Stem Cell Res Ther 2020; 15:531-546. [PMID: 32394844 DOI: 10.2174/1574888x15666200512105347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/11/2020] [Accepted: 03/02/2020] [Indexed: 02/07/2023]
Abstract
Recent studies on the mechanisms that link metabolic changes with stem cell fate have deepened our understanding of how specific metabolic pathways can regulate various stem cell functions during the development of an organism. Although it was originally thought to be merely a consequence of the specific cell state, metabolism is currently known to play a critical role in regulating the self-renewal capacity, differentiation potential, and quiescence of stem cells. Many studies in recent years have revealed that metabolic pathways regulate various stem cell behaviors (e.g., selfrenewal, migration, and differentiation) by modulating energy production through glycolysis or oxidative phosphorylation and by regulating the generation of metabolites, which can modulate multiple signaling pathways. Therefore, a more comprehensive understanding of stem cell metabolism could allow us to establish optimal culture conditions and differentiation methods that would increase stem cell expansion and function for cell-based therapies. However, little is known about how metabolic pathways regulate various stem cell functions. In this context, we review the current advances in metabolic research that have revealed functional roles for mitochondrial oxidative phosphorylation, anaerobic glycolysis, and oxidative stress during the self-renewal, differentiation and aging of various adult stem cell types. These approaches could provide novel strategies for the development of metabolic or pharmacological therapies to promote the regenerative potential of stem cells and subsequently promote their therapeutic utility.
Collapse
Affiliation(s)
- Hwa-Yong Lee
- Department of Biomedical Science, Jungwon University, 85 Goesan-eup, Munmu-ro, Goesan-gun, Chungcheongbuk-do 367-700, Korea
| | - In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Korea
| |
Collapse
|
34
|
Gabrielyan A, Quade M, Gelinsky M, Rösen-Wolff A. IL-11 and soluble VCAM-1 are important components of Hypoxia Conditioned Media and crucial for Mesenchymal Stromal Cells attraction. Stem Cell Res 2020; 45:101814. [PMID: 32334367 DOI: 10.1016/j.scr.2020.101814] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/03/2020] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Bone marrow stromal cells (BMSC) are highly attractive for tissue engineering due to their ability to differentiate into different cell types, to expand extensively in vitro and to release paracrine soluble factors with a high regenerative potential. They were observed to migrate towards the sites of injury in response to chemotactic signals in vivo. During the last years hypoxia has become a proven method to control proliferation, differentiation and multipotency of BMSC. Conditioned medium from hypoxia-treated BMSC (Hypoxia-conditioned Medium; HCM) has been shown to have various favorable properties on tissue regeneration - such as on cell recruitment, wound healing, angiogenesis and revascularization. Due to this regenerative potential many studies attempt to further characterize HCM and its main functional components. In this study we used HCM generated from umbilical cord mesenchymal stem cells (UC-MSC) instead of BMSC, because GMP-verified methods were used to isolate and cultivate the cells and ensure their constant quality. UC-MSC have a high regenerative potential and are still immunologically naive and therefore highly unlikely to cause an immune reaction. In our article we took the first steps to closer investigate the role of umbilical cord MSC-derived HCM components, namely stromal cell-derived factor 1 (SDF-1α), interleukin 11 (IL-11) and soluble vascular cell adhesion molecule 1 (sVCAM-1). RESULTS Our results show previously unknown roles of IL-11 and sVCAM-1 in the attraction of BMSC. The synergistic effect of the investigated protein mixture consisting of IL-11, sVCAM-1 and SDF-1α as well as those recombinant proteins alone revealed a significantly higher chemoattractive capacity towards human BMSC compared to normoxic control medium. Both, the protein mixtures and proteins alone as well as UC-HCM showed an angiogenic effect by promoting the formation of significantly longer tubule structures and higher amounts of junctions and tubules compared to normoxic control medium. CONCLUSIONS By showing the prominent upregulation of IL-11, sVCAM-1 and SDF-1α under hypoxic conditions compared to normoxic control and revealing their crucial role in migration of human BMSC we took a further step forward in characterization of the chemoattractive components of HCM.
Collapse
Affiliation(s)
- Anastasia Gabrielyan
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany.
| | - Mandy Quade
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Michael Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Angela Rösen-Wolff
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| |
Collapse
|
35
|
Kullmann JA, Trivedi N, Howell D, Laumonnerie C, Nguyen V, Banerjee SS, Stabley DR, Shirinifard A, Rowitch DH, Solecki DJ. Oxygen Tension and the VHL-Hif1α Pathway Determine Onset of Neuronal Polarization and Cerebellar Germinal Zone Exit. Neuron 2020; 106:607-623.e5. [PMID: 32183943 DOI: 10.1016/j.neuron.2020.02.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/04/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023]
Abstract
Postnatal brain circuit assembly is driven by temporally regulated intrinsic and cell-extrinsic cues that organize neurogenesis, migration, and axo-dendritic specification in post-mitotic neurons. While cell polarity is an intrinsic organizer of morphogenic events, environmental cues in the germinal zone (GZ) instructing neuron polarization and their coupling during postnatal development are unclear. We report that oxygen tension, which rises at birth, and the von Hippel-Lindau (VHL)-hypoxia-inducible factor 1α (Hif1α) pathway regulate polarization and maturation of post-mitotic cerebellar granule neurons (CGNs). At early postnatal stages with low GZ vascularization, Hif1α restrains CGN-progenitor cell-cycle exit. Unexpectedly, cell-intrinsic VHL-Hif1α pathway activation also delays the timing of CGN differentiation, germinal zone exit, and migration initiation through transcriptional repression of the partitioning-defective (Pard) complex. As vascularization proceeds, these inhibitory mechanisms are downregulated, implicating increasing oxygen tension as a critical switch for neuronal polarization and cerebellar GZ exit.
Collapse
Affiliation(s)
- Jan A Kullmann
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Molecular Neurobiology Group, Institute of Physiological Chemistry, Philipps University of Marburg, 35032 Marburg, Germany
| | - Niraj Trivedi
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Danielle Howell
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christophe Laumonnerie
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Vien Nguyen
- Department of Pediatrics and Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shalini S Banerjee
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Daniel R Stabley
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David H Rowitch
- Department of Pediatrics and Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pediatrics and Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Hills Road, Cambridge CB2 0AN, UK
| | - David J Solecki
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
36
|
Modulation of oxygen tension, acidosis, and cell density is crucial for neural differentiation of human induced pluripotent stem cells. Neurosci Res 2020; 163:34-42. [PMID: 32014574 DOI: 10.1016/j.neures.2020.01.015] [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: 01/20/2020] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/30/2022]
Abstract
Human induced pluripotent stem cell (hiPSC)-derived neural cells provide valuable disease models for pathophysiological analysis and drug discovery for intractable neurodegenerative diseases. However, neural differentiation of hiPSCs requires a complex and long culture procedure, which has been a bottleneck for analysis. We previously demonstrated rapid, efficient, and simple motor neuron differentiation from human pluripotent stem cells (hPSCs). Although optimization of the microenvironment for the differentiation of hPSCs has been considered to achieve more efficient differentiation, it has never been investigated in detail. Here, we demonstrated that three microenvironmental modifiers, oxygen (O2) tension, pH, and cell density, critically affect neural differentiation of hiPSCs. Hypoxia is known to be involved in neural development in vivo and to promote neural differentiation of PSCs. However, in this study, it caused significant cell death in aggregation culture of human embryoid bodies (hEBs) and negatively affected neural differentiation. Modulation of pH by optimized carbon dioxide (CO2) tension improved neural differentiation of hiPSCs, but mild acidosis caused by increased CO2 tension suppressed neural differentiation without cell death. Moreover, high-cell density culture resulted in prominent acidosis and cell death under hypoxic conditions, which synergistically suppressed neural differentiation of hiPSCs. These results suggest that optimization of the microenvironment via O2 tension, pH, and cell density enables more efficient neural differentiation of hiPSCs for the analysis of neurological diseases.
Collapse
|
37
|
Yan J, Goerne T, Zelmer A, Guzman R, Kapfhammer JP, Wellmann S, Zhu X. The RNA-Binding Protein RBM3 Promotes Neural Stem Cell (NSC) Proliferation Under Hypoxia. Front Cell Dev Biol 2019; 7:288. [PMID: 31824945 PMCID: PMC6881237 DOI: 10.3389/fcell.2019.00288] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/04/2019] [Indexed: 01/06/2023] Open
Abstract
Neural stem cells (NSCs) reside physiologically in a hypoxic niche to maintain self-renewal and multipotency. Whereas mild hypoxia is known to promote NSC proliferation, severe hypoxia in pathological conditions exerts the reverse effect. The multi-functional RNA-binding protein RBM3 is abundant in NSCs and can be regulated by hypoxic exposure. Although RBM3 has been shown to accelerate cell growth in many cell types, whether and how it affects NSC proliferation in hypoxic environment remains largely unknown. In this study, we tested how RBM3 regulates cell proliferation under hypoxia in C17.2 mouse NSC cell line and in primary mouse NSCs from both the forebrain of postnatal day 0 (P0) mice and the subgranular zone (SGZ) of adult mice. Our results demonstrated that RBM3 expression was highly sensitive to hypoxia, and NSCs were arrested in G0/G1 phase by 5, 2.5, and 1% O2 treatment. When we overexpressed RBM3, hypoxia-induced cell cycle arrest in G0/G1 phase was relieved and more cell transit into S phase was observed. Furthermore, cell viability under hypoxia was also increased by RBM3. In contrast, in RBM3-depleted primary NSCs, less BrdU-incorporated cells were detected, indicating exacerbated cell cycle arrest in G1 to S phase transition. Instead, overexpressed RBM3 significantly increased proliferation ratio in primary NSCs. Our findings indicate RBM3 as a potential target to maintain the proliferation capacity of NSCs under hypoxia, which can be important in NSC-based therapies of acute brain injury and chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Jingyi Yan
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Tessa Goerne
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Andrea Zelmer
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Raphael Guzman
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Sven Wellmann
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland.,Department of Neonatology, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | - Xinzhou Zhu
- Department of Neonatology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| |
Collapse
|
38
|
Lukmanto D, Khanh VC, Shirota S, Kato T, Takasaki MM, Ohneda O. Dynamic Changes of Mouse Embryonic Stem Cell-Derived Neural Stem Cells Under In Vitro Prolonged Culture and Hypoxic Conditions. Stem Cells Dev 2019; 28:1434-1450. [DOI: 10.1089/scd.2019.0101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Donny Lukmanto
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Vuong Cat Khanh
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Saori Shirota
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Toshiki Kato
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Mami Matsuo Takasaki
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Osamu Ohneda
- Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
39
|
Komabayashi-Suzuki M, Yamanishi E, Watanabe C, Okamura M, Tabata H, Iwai R, Ajioka I, Matsushita J, Kidoya H, Takakura N, Okamoto T, Kinoshita K, Ichihashi M, Nagata KI, Ema M, Mizutani KI. Spatiotemporally Dependent Vascularization Is Differently Utilized among Neural Progenitor Subtypes during Neocortical Development. Cell Rep 2019; 29:1113-1129.e5. [DOI: 10.1016/j.celrep.2019.09.048] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 06/19/2019] [Accepted: 09/18/2019] [Indexed: 01/07/2023] Open
|
40
|
Takashima S, Watanabe C, Ema M, Mizutani KI. Interaction of the nervous system and vascular system is required for the proper assembly of the neocortex. Neurochem Int 2019; 129:104481. [DOI: 10.1016/j.neuint.2019.104481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/07/2019] [Accepted: 05/27/2019] [Indexed: 12/15/2022]
|
41
|
Seo Y, Shin TH, Kim HS. Current Strategies to Enhance Adipose Stem Cell Function: An Update. Int J Mol Sci 2019; 20:E3827. [PMID: 31387282 PMCID: PMC6696067 DOI: 10.3390/ijms20153827] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) emerged as a promising therapeutic tool targeting a variety of inflammatory disorders due to their multiple remarkable properties, such as superior immunomodulatory function and tissue-regenerative capacity. Although bone marrow (BM) is a dominant source for adult MSCs, increasing evidence suggests that adipose tissue-derived stem cells (ASCs), which can be easily obtained at a relatively high yield, have potent therapeutic advantages comparable with BM-MSCs. Despite its outstanding benefits in pre-clinical settings, the practical efficacy of ASCs remains controversial since clinical trials with ASC application often resulted in unsatisfactory outcomes. To overcome this challenge, scientists established several strategies to generate highly functional ASCs beyond the naïve cells, including (1) pre-conditioning of ASCs with various stimulants such as inflammatory agents, (2) genetic manipulation of ASCs and (3) modification of culture conditions with three-dimensional (3D) aggregate formation and hypoxic culture. Also, exosomes and other extracellular vesicles secreted from ASCs can be applied directly to recapitulate the beneficial performance of ASCs. This review summarizes the current strategies to improve the therapeutic features of ASCs for successful clinical implementation.
Collapse
Affiliation(s)
- Yoojin Seo
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea
| | - Tae-Hoon Shin
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hyung-Sik Kim
- Dental and Life Science Institute, Pusan National University, Yangsan 50612, Korea.
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| |
Collapse
|
42
|
Boso D, Rampazzo E, Zanon C, Bresolin S, Maule F, Porcù E, Cani A, Della Puppa A, Trentin L, Basso G, Persano L. HIF-1α/Wnt signaling-dependent control of gene transcription regulates neuronal differentiation of glioblastoma stem cells. Am J Cancer Res 2019; 9:4860-4877. [PMID: 31410187 PMCID: PMC6691379 DOI: 10.7150/thno.35882] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/05/2019] [Indexed: 12/16/2022] Open
Abstract
HIF-1α has been suggested to interplay with Wnt signaling components in order to activate a neuronal differentiation process in both normal brain and glioblastoma (GBM). Based on these data, we explored the molecular mechanisms underlying the observed capability of GBM cells to acquire a neuronal phenotype upon Wnt signaling stimulation and how the microenvironment, particularly hypoxia, modulates this process. Methods: here, the employment of ChIP-seq techniques together with co-immunoprecipitation approaches allowed to reconstruct the molecular interactions responsible for activating specific pro-differentiating transcriptional programs in GBM cells. Moreover, gene silencing/over-expression approaches coupled with the functional analysis of cell phenotype were applied to confirm ChIP-driven hypotheses. Finally, we combined the use of publicly available gene expression datasets with protein expression data by immunohistochemistry to test the clinical relevance of obtained results. Results: our data clearly suggest that HIF-1α is recruited by the β-catenin/TCF1 complex to foster neuronal differentiation gene transcription in hypoxic GBM cells. Conversely, at higher oxygen levels, the increased expression of TCF4 exerts a transcriptional inhibitory function on the same genomic regions, thus counteracting differentiation. Moreover, we demonstrate the existence of a positive correlation between the expression levels of HIF-1α, TCF1 and neuronal phenotype in GBM tumors, accompanied by the over-expression of several Wnt signaling components, finally affecting patient prognosis. Conclusion: we unveiled a peculiar mechanism by which TCF1 and HIF-1α can induce a reminiscent neuronal differentiation of hypoxic GBM cells, which is hampered, in normoxia, by high levels of TCF4, thus not only de facto controlling the balance between differentiation and stemness, but also impacting on intra-tumoral heterogeneity and eventually patient outcome.
Collapse
|
43
|
Nordström T, Andersson LC, Åkerman KE. Regulation of intracellular pH by electrogenic Na+/HCO3– co-transporters in embryonic neural stem cell-derived radial glia-like cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1037-1048. [DOI: 10.1016/j.bbamem.2019.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/11/2019] [Accepted: 03/14/2019] [Indexed: 01/19/2023]
|
44
|
Zhao RZ, Jiang S, Ru NY, Jiao B, Yu ZB. Comparison of hypoxic effects induced by chemical and physical hypoxia on cardiomyocytes. Can J Physiol Pharmacol 2019; 97:980-988. [PMID: 31136722 DOI: 10.1139/cjpp-2019-0092] [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] [Indexed: 01/01/2023]
Abstract
The degree and duration of chemical hypoxia induced by sodium dithionite (Na2S2O4) have not been reported. It is not yet clear how much reduction in the O2 concentration (physical hypoxia) can lead to hypoxia in cultured cardiomyocytes. In this study, oxygen microelectrodes were used to measure changes in the O2 concentration in media containing different concentrations of Na2S2O4. Then, hypoxic effects of 0.8, 1.0, and 2.0 mM Na2S2O4 or 1%, 3%, and 5% O2 in cultured cardiomyocytes from neonatal rats were observed and compared. The results showed that the O2 concentration failed to remain constant by Na2S2O4 treatment during the 180-minute observation period. Only the 2.0 mM Na2S2O4 group significantly increased the expression of hypoxia-inducible factor 1α (HIF-1α) and hypoxic responses. Notably, 3% O2 only significantly increased the expression of HIF-1α in cardiomyocytes, while 1% O2 not only increased the expression of HIF-1α but also increased the apoptotic rate in cardiomyocytes. These results suggest that Na2S2O4 is not suitable for establishing a hypoxic model in cultured neonatal rat cardiomyocytes, and neonatal rat cardiomyocytes cultured at or below 1% O2 induced significant hypoxic effects, which can be used as a starting O2 concentration for establishing a hypoxic cell model.
Collapse
Affiliation(s)
- Ru-Zhou Zhao
- Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an 710032, China
| | - Shuai Jiang
- Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an 710032, China
| | - Ning-Yu Ru
- Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an 710032, China.,Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an 710032, China
| | - Bo Jiao
- Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an 710032, China.,Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an 710032, China
| | - Zhi-Bin Yu
- Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an 710032, China.,Department of Aerospace Physiology, Fourth Military Medical University, Key Laboratory of Aerospace Medicine, Ministry of China, Xi'an 710032, China
| |
Collapse
|
45
|
Baakdhah T, van der Kooy D. Expansion of retinal stem cells and their progeny using cell microcarriers in a bioreactor. Biotechnol Prog 2019; 35:e2800. [DOI: 10.1002/btpr.2800] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/10/2019] [Accepted: 02/24/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Tahani Baakdhah
- Institute of Medical ScienceUniversity of Toronto Toronto Ontario Canada
| | - Derek van der Kooy
- Institute of Medical ScienceUniversity of Toronto Toronto Ontario Canada
- Department of Molecular GeneticsUniversity of Toronto Toronto Ontario Canada
| |
Collapse
|
46
|
Culture in 10% O 2 enhances the production of active hormones in neuro-endocrine cells by up-regulating the expression of processing enzymes. Biochem J 2019; 476:827-842. [PMID: 30787050 DOI: 10.1042/bcj20180832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/17/2019] [Accepted: 02/19/2019] [Indexed: 12/22/2022]
Abstract
To closely mimic physiological conditions, low oxygen cultures have been employed in stem cell and cancer research. Although in vivo oxygen concentrations in tissues are often much lower than ambient 21% O2 (ranging from 3.6 to 12.8% O2), most cell cultures are maintained at 21% O2 To clarify the effects of the O2 culture concentration on the regulated secretion of peptide hormones in neuro-endocrine cells, we examined the changes in the storage and release of peptide hormones in neuro-endocrine cell lines and endocrine tissues cultured in a relatively lower O2 concentration. In both AtT-20 cells derived from the mouse anterior pituitary and freshly prepared mouse pituitaries cultured in 10% O2 for 24 h, the storage and regulated secretion of the mature peptide hormone adrenocorticotropic hormone were significantly increased compared with those in cells and pituitaries cultured in ambient 21% O2, whereas its precursor proopiomelanocortin was not increased in the cells and tissues after being cultured in 10% O2 Simultaneously, the prohormone-processing enzymes PC1/3 and carboxypeptidase E were up-regulated in cells cultured in 10% O2, thus facilitating the conversion of prohormones to their active form. Similarly, culturing the mouse β-cell line MIN6 and islet tissue in 10% O2 also significantly increased the conversion of proinsulin into mature insulin, which was secreted in a regulated manner. These results suggest that culture under 10% O2 is more optimal for endocrine tissues/cells to efficiently generate and secrete active peptide hormones than ambient 21% O2.
Collapse
|
47
|
Sobrino V, Annese V, Navarro-Guerrero E, Platero-Luengo A, Pardal R. The carotid body: a physiologically relevant germinal niche in the adult peripheral nervous system. Cell Mol Life Sci 2019; 76:1027-1039. [PMID: 30498994 PMCID: PMC11105339 DOI: 10.1007/s00018-018-2975-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/05/2018] [Accepted: 11/22/2018] [Indexed: 12/26/2022]
Abstract
Oxygen constitutes a vital element for the survival of every single cell in multicellular aerobic organisms like mammals. A complex homeostatic oxygen-sensing system has evolved in these organisms, including detectors and effectors, to guarantee a proper supply of the element to every cell. The carotid body represents the most important peripheral arterial chemoreceptor organ in mammals and informs about hypoxemic situations to the effectors at the brainstem cardiorespiratory centers. To optimize organismal adaptation to maintained hypoxemic situations, the carotid body has evolved containing a niche of adult tissue-specific stem cells with the capacity to differentiate into both neuronal and vascular cell types in response to hypoxia. These neurogenic and angiogenic processes are finely regulated by the niche and by hypoxia itself. Our recent data on the cellular and molecular mechanisms underlying the functioning of this niche might help to comprehend a variety of different diseases coursing with carotid body failure, and might also improve our capacity to use these stem cells for the treatment of neurological disease. Herein, we review those data about the recent characterization of the carotid body niche, focusing on the study of the phenotype and behavior of multipotent stem cells within the organ, comparing them with other well-documented neural stem cells within the adult nervous system.
Collapse
Affiliation(s)
- Verónica Sobrino
- Instituto de Biomedicina de Sevilla (IBiS), Laboratory 103, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Dpto. de Fisiología Médica y Biofísica, Avda, Manuel Siurot, s/n., 41013, Sevilla, Spain
| | - Valentina Annese
- Instituto de Biomedicina de Sevilla (IBiS), Laboratory 103, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Dpto. de Fisiología Médica y Biofísica, Avda, Manuel Siurot, s/n., 41013, Sevilla, Spain
| | - Elena Navarro-Guerrero
- Instituto de Biomedicina de Sevilla (IBiS), Laboratory 103, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Dpto. de Fisiología Médica y Biofísica, Avda, Manuel Siurot, s/n., 41013, Sevilla, Spain
| | - Aida Platero-Luengo
- Instituto de Biomedicina de Sevilla (IBiS), Laboratory 103, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Dpto. de Fisiología Médica y Biofísica, Avda, Manuel Siurot, s/n., 41013, Sevilla, Spain
| | - Ricardo Pardal
- Instituto de Biomedicina de Sevilla (IBiS), Laboratory 103, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Dpto. de Fisiología Médica y Biofísica, Avda, Manuel Siurot, s/n., 41013, Sevilla, Spain.
| |
Collapse
|
48
|
Intermittent Hypoxia Disrupts Adult Neurogenesis and Synaptic Plasticity in the Dentate Gyrus. J Neurosci 2018; 39:1320-1331. [PMID: 30587544 DOI: 10.1523/jneurosci.1359-18.2018] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 09/03/2018] [Accepted: 09/27/2018] [Indexed: 01/06/2023] Open
Abstract
Individuals with sleep apnea often exhibit changes in cognitive behaviors consistent with alterations in the hippocampus. It is hypothesized that adult neurogenesis in the dentate gyrus is an ongoing process that maintains normal hippocampal function in many mammalian species, including humans. However, the impact of chronic intermittent hypoxia (IH), a principal consequence of sleep apnea, on hippocampal adult neurogenesis remains unclear. Using a murine model, we examined the impact of 30 d of IH (IH30) on adult neurogenesis and synaptic plasticity in the dentate gyrus. Although IH30 did not affect paired-pulse facilitation, IH30 suppressed long-term potentiation (LTP). Immunohistochemical experiments also indicate that IH perturbs multiple aspects of adult neurogenesis. IH30 increased the number of proliferating Sox2+ neural progenitor cells in the subgranular zone yet reduced the number of doublecortin-positive neurons. Consistent with these findings, cell lineage tracing revealed that IH30 increased the proportion of radial glial cells in the subgranular zone, yet decreased the proportion of adult-born neurons in the dentate gyrus. While administration of a superoxide anion scavenger during IH did not prevent neural progenitor cell proliferation, it mitigated the IH-dependent suppression of LTP and prevented adult-born neuron loss. These data demonstrate that IH causes both reactive oxygen species-dependent and reactive oxygen species-independent effects on adult neurogenesis and synaptic plasticity in the dentate gyrus. Our findings identify cellular and neurophysiological changes in the hippocampus that may contribute to cognitive and behavioral deficits occurring in sleep apnea.SIGNIFICANCE STATEMENT Individuals with sleep apnea experience periods of intermittent hypoxia (IH) that can negatively impact many aspects of brain function. Neurons are continually generated throughout adulthood to support hippocampal physiology and behavior. This study demonstrates that IH exposure attenuates hippocampal long-term potentiation and reduces adult neurogenesis. Antioxidant treatment mitigates these effects indicating that oxidative signaling caused by IH is a significant factor that impairs synaptic plasticity and reduces adult neurogenesis in the hippocampus.
Collapse
|
49
|
Carrica L, Li L, Newville J, Kenton J, Gustus K, Brigman J, Cunningham LA. Genetic inactivation of hypoxia inducible factor 1-alpha (HIF-1α) in adult hippocampal progenitors impairs neurogenesis and pattern discrimination learning. Neurobiol Learn Mem 2018; 157:79-85. [PMID: 30521851 DOI: 10.1016/j.nlm.2018.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/16/2018] [Accepted: 12/01/2018] [Indexed: 01/10/2023]
Abstract
HIF-1α is a hypoxia-inducible protein that regulates many cellular processes, including neural stem cell maintenance. Previous work demonstrated constitutive stabilization of HIF-1α in neural stem cells (NSCs) of the adult mouse subventricular zone (SVZ) and hippocampal subgranular zone (SGZ). Genetic inactivation of NSC-encoded HIF-1α in the adult SVZ results in gradual loss of NSCs, but whether HIF-1α is required for the maintenance of SGZ hippocampal progenitors and adult hippocampal neurogenesis has not been determined. Here we tested the hypothesis that HIF-1α plays an essential role in the maintenance of adult hippocampal neurogenesis using Nestin-CreERT2/R26R-YFP/Hif1afl/fl triple transgenic mice, in which HIF-1α was genetically inactivated in nestin+ hippocampal progenitors and their downstream progeny following tamoxifen exposure. We found that disruption of HIF-1α gene expression resulted in a marked 50% reduction of adult-generated dentate granule cells (DGCs) that was highly correlated with impaired hippocampal function, as assessed using two behavioral assays of pattern discrimination. These behavioral tests included the A-B contextual fear-conditioning task and the trial-unique, delayed nonmatching-to-location (TUNL) touch-screen operant chamber task. Our findings identify HIF-1α as a novel regulator of adult hippocampal neurogenesis under non-pathological conditions, and underscore the importance of neurogenesis for pattern discrimination learning.
Collapse
Affiliation(s)
- Lauren Carrica
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Lu Li
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Jessie Newville
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Johnny Kenton
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Kymberly Gustus
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Jonathan Brigman
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Lee Anna Cunningham
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.
| |
Collapse
|
50
|
Andreopoulou E, Arampatzis A, Patsoni M, Kazanis I. Being a Neural Stem Cell: A Matter of Character But Defined by the Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:81-118. [PMID: 29204830 DOI: 10.1007/978-3-319-69194-7_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cells that build the nervous system, either this is a small network of ganglia or a complicated primate brain, are called neural stem and progenitor cells. Even though the very primitive and the very recent neural stem cells (NSCs) share common basic characteristics that are hard-wired within their character, such as the expression of transcription factors of the SoxB family, their capacity to give rise to extremely different neural tissues depends significantly on instructions from the microenvironment. In this chapter we explore the nature of the NSC microenvironment, looking through evolution, embryonic development, maturity and even disease. Experimental work undertaken over the last 20 years has revealed exciting insight into the NSC microcosmos. NSCs are very capable in producing their own extracellular matrix and in regulating their behaviour in an autocrine and paracrine manner. Nevertheless, accumulating evidence indicates an important role for the vasculature, especially within the NSC niches of the postnatal brain; while novel results reveal direct links between the metabolic state of the organism and the function of NSCs.
Collapse
Affiliation(s)
- Evangelia Andreopoulou
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Asterios Arampatzis
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK
- School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Melina Patsoni
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Ilias Kazanis
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece.
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|