1
|
Stepien BK, Wielockx B. From Vessels to Neurons-The Role of Hypoxia Pathway Proteins in Embryonic Neurogenesis. Cells 2024; 13:621. [PMID: 38607059 PMCID: PMC11012138 DOI: 10.3390/cells13070621] [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: 02/28/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Embryonic neurogenesis can be defined as a period of prenatal development during which divisions of neural stem and progenitor cells give rise to neurons. In the central nervous system of most mammals, including humans, the majority of neocortical neurogenesis occurs before birth. It is a highly spatiotemporally organized process whose perturbations lead to cortical malformations and dysfunctions underlying neurological and psychiatric pathologies, and in which oxygen availability plays a critical role. In case of deprived oxygen conditions, known as hypoxia, the hypoxia-inducible factor (HIF) signaling pathway is activated, resulting in the selective expression of a group of genes that regulate homeostatic adaptations, including cell differentiation and survival, metabolism and angiogenesis. While a physiological degree of hypoxia is essential for proper brain development, imbalanced oxygen levels can adversely affect this process, as observed in common obstetrical pathologies such as prematurity. This review comprehensively explores and discusses the current body of knowledge regarding the role of hypoxia and the HIF pathway in embryonic neurogenesis of the mammalian cortex. Additionally, it highlights existing gaps in our understanding, presents unanswered questions, and provides avenues for future research.
Collapse
Affiliation(s)
- Barbara K. Stepien
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- Experimental Centre, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
2
|
Fan Y, Li J, Fang B. A Tale of Two: When Neural Stem Cells Encounter Hypoxia. Cell Mol Neurobiol 2023; 43:1799-1816. [PMID: 36308642 PMCID: PMC11412202 DOI: 10.1007/s10571-022-01293-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/02/2022] [Indexed: 11/12/2022]
Abstract
Normoxia is defined as an oxygen concentration of 20.9%, as in room air, whereas hypoxia refers to any oxygen concentration less than this. Any physiological oxygen deficiency or tissue oxygen deficiency relative to demand is called hypoxia. Neural stem cells (NSCs) are multipotent stem cells that can differentiate into multiple cell lines such as neurons, oligodendrocytes, and astrocytes. Under hypoxic conditions, the apoptosis rate of NSCs increases remarkably in vitro or in vivo. However, some hypoxia promotes the proliferation and differentiation of NSCs. The difference is related to the oxygen concentration, the duration of hypoxia, the hypoxia tolerance threshold of the NSCs, and the tissue source of the NSCs. The main mechanism of hypoxia-induced proliferation and differentiation involves an increase in cyclin and erythropoietin concentrations, and hypoxia-inducible factors play a key role. Multiple molecular pathways are activated during hypoxia, including Notch, Wnt/β-catenin, PI3K/Akt, and altered microRNA expression. In addition, we review the protective effect of exogenous NSCs transplantation on ischemic or anoxic organs, the therapeutic potential of hypoxic preconditioning on exogenous NSCs and clinical application of NSCs.
Collapse
Affiliation(s)
- Yiting Fan
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Jinshi Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Li J, Gong SH, He YL, Cao Y, Chen Y, Huang GH, Wang YF, Zhao M, Cheng X, Zhou YZ, Zhao T, Zhao YQ, Fan M, Wu HT, Zhu LL, Wu LY. Autophagy Is Essential for Neural Stem Cell Proliferation Promoted by Hypoxia. Stem Cells 2023; 41:77-92. [PMID: 36208284 DOI: 10.1093/stmcls/sxac076] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/06/2022] [Indexed: 02/02/2023]
Abstract
Hypoxia as a microenvironment or niche stimulates proliferation of neural stem cells (NSCs). However, the underlying mechanisms remain elusive. Autophagy is a protective mechanism by which recycled cellular components and energy are rapidly supplied to the cell under stress. Whether autophagy mediates the proliferation of NSCs under hypoxia and how hypoxia induces autophagy remain unclear. Here, we report that hypoxia facilitates embryonic NSC proliferation through HIF-1/mTORC1 signaling pathway-mediated autophagy. Initially, we found that hypoxia greatly induced autophagy in NSCs, while inhibition of autophagy severely impeded the proliferation of NSCs in hypoxia conditions. Next, we demonstrated that the hypoxia core regulator HIF-1 was necessary and sufficient for autophagy induction in NSCs. Considering that mTORC1 is a key switch that suppresses autophagy, we subsequently analyzed the effect of HIF-1 on mTORC1 activity. Our results showed that the mTORC1 activity was negatively regulated by HIF-1. Finally, we provided evidence that HIF-1 regulated mTORC1 activity via its downstream target gene BNIP3. The increased expression of BNIP3 under hypoxia enhanced autophagy activity and proliferation of NSCs, which was mediated by repressing the activity of mTORC1. We further illustrated that BNIP3 can interact with Rheb, a canonical activator of mTORC1. Thus, we suppose that the interaction of BNIP3 with Rheb reduces the regulation of Rheb toward mTORC1 activity, which relieves the suppression of mTORC1 on autophagy, thereby promoting the rapid proliferation of NSCs. Altogether, this study identified a new HIF-1/BNIP3-Rheb/mTORC1 signaling axis, which regulates the NSC proliferation under hypoxia through induction of autophagy.
Collapse
Affiliation(s)
- Jian Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Sheng-Hui Gong
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yun-Ling He
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yan Cao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Ying Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Guang-Hai Huang
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yu-Fei Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Ming Zhao
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Xiang Cheng
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yan-Zhao Zhou
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Tong Zhao
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yong-Qi Zhao
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Ming Fan
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Hai-Tao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Ling-Ling Zhu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China.,Department of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, People's Republic of China.,Department of Pharmacology, University of Nanhua, Hengyang, China
| | - Li-Ying Wu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
4
|
Brandon A, Cui X, Luan W, Ali AA, Pertile RAN, Alexander SA, Eyles DW. Prenatal hypoxia alters the early ontogeny of dopamine neurons. Transl Psychiatry 2022; 12:238. [PMID: 35672280 PMCID: PMC9174174 DOI: 10.1038/s41398-022-02005-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 11/09/2022] Open
Abstract
Dopaminergic (DA) dysfunction is a significant feature in the pathophysiology of schizophrenia. Established developmental risk factors for schizophrenia such as maternal immune activation (MIA) or developmental vitamin D (DVD) deficiency, when modelled in animals, reveal the differentiation of early DA neurons in foetal brains is delayed suggesting this may be a convergent aetiological pathway. Here we have assessed the effects of prenatal hypoxia, another well-known developmental risk factor for schizophrenia, on developing DA systems. Pregnant mice were exposed to a hypoxic environment of 10% oxygen for 48 h from embryonic day 10 (E10) to E12. Embryonic brains were collected and the positioning of mesencephalic cells, expression of DA specification and maturation factors were examined along with the expression of factors that may govern the migration of these neurons. We show that prenatal hypoxia results in a decrease in dopaminergic progenitors retards early DA neuron lateral migration and reduces expression of the receptors known to govern this process. A second time-point, postnatal day 10 (P10) was also examined in order to assess whether prenatal hypoxia alters early presynaptic architecture in the developing striatum. We show reduced expression of tyrosine hydroxylase (TH) in the postnatal striatum along with increases in the density of high-probability DA release sites within TH varicosities. These findings add to the emerging literature showing that multiple epidemiologically validated environmental risk factors for schizophrenia may induce early alterations to develop DA systems. This may represent a possible convergent mechanism in the onset of presynaptic DA dysfunction in patients.
Collapse
Affiliation(s)
- Anastasia Brandon
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Xiaoying Cui
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia
| | - Wei Luan
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Asad Amanat Ali
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | | | - Suzanne Adele Alexander
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia
| | - Darryl Walter Eyles
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia.
- Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, QLD, Australia.
| |
Collapse
|
5
|
Asschenfeldt B, Evald L, Yun HJ, Heiberg J, Østergaard L, Grant PE, Hjortdal VE, Im K, Eskildsen SF. Abnormal Left-Hemispheric Sulcal Patterns in Adults With Simple Congenital Heart Defects Repaired in Childhood. J Am Heart Assoc 2021; 10:e018580. [PMID: 33745293 PMCID: PMC8174332 DOI: 10.1161/jaha.120.018580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background Children operated on for a simple congenital heart defect (CHD) are at risk of neurodevelopmental abnormalities. Abnormal cortical development and folding have been observed in fetuses with CHD. We examined whether sulcal folding patterns in adults operated on for simple CHD in childhood differ from those of healthy controls, and whether such differences are associated with neuropsychological outcomes. Methods and Results Patients (mean age, 24.5 years) who underwent childhood surgery for isolated atrial septal defect (ASD; n=33) or ventricular septal defect (VSD; n=30) and healthy controls (n=37) were enrolled. Sulcal pattern similarity to healthy controls was determined using magnetic resonance imaging and looking at features of sulcal folds, their intersulcal relationships, and sulcal graph topology. The sulcal pattern similarity values were tested for associations with comprehensive neuropsychological scores. Patients with both ASD and VSD had decreased sulcal pattern similarity in the left hemisphere compared with controls. The differences were found in the left temporal lobe in the ASD group and in the whole left hemisphere in the VSD group (P=0.033 and P=0.039, respectively). The extent of abnormal left hemispheric sulcal pattern similarity was associated with worse neuropsychological scores (intelligence, executive function, and visuospatial abilities) in the VSD group, and special educational support in the ASD group. Conclusions Adults who underwent surgery for simple CHD in childhood display altered left hemisphere sulcal folding patterns, commensurate with neuropsychological scores for patients with VSD and special educational support for ASD. This may indicate that simple CHD affects early brain development. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT03871881.
Collapse
Affiliation(s)
- Benjamin Asschenfeldt
- Department of Cardiothoracic and Vascular Surgery Aarhus University Hospital Aarhus N Denmark.,Department of Clinical Medicine Aarhus University Aarhus N Denmark
| | - Lars Evald
- Department of Clinical Medicine Aarhus University Aarhus N Denmark.,Hammel Neurorehabilitation Centre and University Research Clinic Hammel Denmark
| | - Hyuk Jin Yun
- Fetal Neonatal Neuroimaging and Developmental Science Center Boston Children's Hospital Boston MA.,Division of Newborn Medicine Boston Children's Hospital Boston MA.,Harvard Medical School Boston MA
| | - Johan Heiberg
- Department of Cardiothoracic and Vascular Surgery Aarhus University Hospital Aarhus N Denmark.,Department of Clinical Medicine Aarhus University Aarhus N Denmark
| | - Leif Østergaard
- Department of Clinical Medicine Aarhus University Aarhus N Denmark.,Center of Functionally Integrative Neuroscience Aarhus University Aarhus C Denmark
| | - P Ellen Grant
- Fetal Neonatal Neuroimaging and Developmental Science Center Boston Children's Hospital Boston MA.,Division of Newborn Medicine Boston Children's Hospital Boston MA.,Department of Radiology Boston Children's Hospital Boston MA.,Harvard Medical School Boston MA
| | - Vibeke Elisabeth Hjortdal
- Department of Clinical Medicine Aarhus University Aarhus N Denmark.,Department of Cardiothoracic Surgery RigshospitaletCopenhagen Denmark.,Institute of Clinical Medicine University of Copenhagen Copenhagen Denmark
| | - Kiho Im
- Fetal Neonatal Neuroimaging and Developmental Science Center Boston Children's Hospital Boston MA.,Division of Newborn Medicine Boston Children's Hospital Boston MA.,Harvard Medical School Boston MA
| | - Simon Fristed Eskildsen
- Department of Clinical Medicine Aarhus University Aarhus N Denmark.,Center of Functionally Integrative Neuroscience Aarhus University Aarhus C Denmark
| |
Collapse
|
6
|
Dalby RB, Eskildsen SF, Videbech P, Frandsen J, Mouridsen K, Sørensen L, Jeppesen P, Bek T, Rosenberg R, Østergaard L. Oxygenation differs among white matter hyperintensities, intersected fiber tracts and unaffected white matter. Brain Commun 2019; 1:fcz033. [PMID: 32954272 PMCID: PMC7425421 DOI: 10.1093/braincomms/fcz033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 08/27/2019] [Accepted: 10/01/2019] [Indexed: 01/15/2023] Open
Abstract
White matter hyperintensities of presumed vascular origin are frequently observed on magnetic resonance imaging in normal aging. They are typically found in cerebral small vessel disease and suspected culprits in the etiology of complex age- and small vessel disease-related conditions, such as late-onset depression. White matter hyperintensities may interfere with surrounding white matter metabolic demands by disrupting fiber tract integrity. Meanwhile, risk factors for small vessel disease are thought to reduce tissue oxygenation, not only by reducing regional blood supply, but also by impairing capillary function. To address white matter oxygen supply–demand balance, we estimated voxel-wise capillary density as an index of resting white matter metabolism, and combined estimates of blood supply and capillary function to calculate white matter oxygen availability. We conducted a cross-sectional study with structural, perfusion- and diffusion-weighted magnetic resonance imaging in 21 patients with late-onset depression and 21 controls. We outlined white matter hyperintensities and used tractography to identify the tracts they intersect. Perfusion data comprised cerebral blood flow, blood volume, mean transit time and relative transit time heterogeneity—the latter a marker of capillary dysfunction. Based on these, white matter oxygenation was calculated as the steady state cerebral metabolic rate of oxygen under the assumption of normal tissue oxygen tension and vice versa. The number, volume and perfusion characteristics of white matter hyperintensities did not differ significantly between groups. Hemodynamic data showed white matter hyperintensities to have lower blood flow and blood volume, but higher relative transit time heterogeneity, than normal-appearing white matter, resulting in either reduced capillary metabolic rate of oxygen or oxygen tension. Intersected tracts showed significantly lower blood flow, blood volume and capillary metabolic rate of oxygen than normal-appearing white matter. Across groups, lower lesion oxygen tension was associated with higher lesion number and volume. Compared with normal-appearing white matter, tissue oxygenation is significantly reduced in white matter hyperintensities as well as the fiber tracts they intersect, independent of parallel late-onset depression. In white matter hyperintensities, reduced microvascular blood volume and concomitant capillary dysfunction indicate a severe oxygen supply–demand imbalance with hypoxic tissue injury. In intersected fiber tracts, parallel reductions in oxygenation and microvascular blood volume are consistent with adaptations to reduced metabolic demands. We speculate, that aging and vascular risk factors impair white matter hyperintensity perfusion and capillary function to create hypoxic tissue injury, which in turn affect the function and metabolic demands of the white matter tracts they disrupt.
Collapse
Affiliation(s)
- Rikke B Dalby
- Center of Functionally Integrative Neuroscience & MINDLab, Aarhus University Hospital, 8200 Aarhus C., Denmark.,Centre for Psychiatric Research, Aarhus University Hospital, 8340 Risskov, Denmark.,Department of Neuroradiology, Aarhus University Hospital, 8200 Aarhus N., Denmark
| | - Simon F Eskildsen
- Center of Functionally Integrative Neuroscience & MINDLab, Aarhus University Hospital, 8200 Aarhus C., Denmark
| | - Poul Videbech
- Center for Neuropsychiatric Depression Research, Mental Health Center Glostrup, 2600 Glostrup, Denmark
| | - Jesper Frandsen
- Center of Functionally Integrative Neuroscience & MINDLab, Aarhus University Hospital, 8200 Aarhus C., Denmark
| | - Kim Mouridsen
- Center of Functionally Integrative Neuroscience & MINDLab, Aarhus University Hospital, 8200 Aarhus C., Denmark
| | - Leif Sørensen
- Department of Neuroradiology, Aarhus University Hospital, 8200 Aarhus N., Denmark
| | - Peter Jeppesen
- Department of Ophthalmology, Aarhus University Hospital, 8200 Aarhus N., Denmark
| | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, 8200 Aarhus N., Denmark
| | - Raben Rosenberg
- Centre of Psychiatry Amager, Mental Health Services in the Capital Region of Denmark, 2300 Copenhagen S., Denmark
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience & MINDLab, Aarhus University Hospital, 8200 Aarhus C., Denmark.,Department of Neuroradiology, Aarhus University Hospital, 8200 Aarhus N., Denmark
| |
Collapse
|
7
|
Wu LY, He YL, Zhu LL. Possible Role of PHD Inhibitors as Hypoxia-Mimicking Agents in the Maintenance of Neural Stem Cells' Self-Renewal Properties. Front Cell Dev Biol 2018; 6:169. [PMID: 30619851 PMCID: PMC6297135 DOI: 10.3389/fcell.2018.00169] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022] Open
Abstract
Hypoxia is the most critical factor for maintaining stemness. During embryonic development, neural stem cells (NSCs) reside in hypoxic niches, and different levels of oxygen pressure and time of hypoxia exposure play important roles in the development of NSCs. Such hypoxic niches exist in adult brain tissue, where the neural precursors originate. Hypoxia-inducible factors (HIFs) are key transcription heterodimers consisting of regulatory α-subunits (HIF-1α, HIF-2α, HIF-3α) and a constitutive β-subunit (HIF-β). Regulation of downstream targets determines the fate of NSCs. In turn, the stability of HIFs-α is regulated by prolyl hydroxylases (PHDs), whose activity is principally modulated by PHD substrates like oxygen (O2), α-ketoglutarate (α-KG), and the co-factors ascorbate (ASC) and ferrous iron (Fe2+). It follows that the transcriptional activity of HIFs is actually determined by the contents of O2, α-KG, ASC, and Fe2+. In normoxia, HIFs-α are rapidly degraded via the ubiquitin-proteasome pathway, in which PHDs, activated by O2, lead to hydroxylation of HIFs-α at residues 402 and 564, followed by recognition by the tumor suppressor protein von Hippel–Lindau (pVHL) as an E3 ligase and ubiquitin labeling. Conversely, in hypoxia, the activity of PHDs is inhibited by low O2 levels and HIFs-α can thus be stabilized. Hence, suppression of PHD activity in normoxic conditions, mimicking the effect of hypoxia, might be beneficial for preserving the stemness of NSCs, and it is clinically relevant as a therapeutic approach for enhancing the number of NSCs in vitro and for cerebral ischemia injury in vivo. This study will review the putative role of PHD inhibitors on the self-renewal of NSCs.
Collapse
Affiliation(s)
- Li-Ying Wu
- Beijing Institute of Cognition and Brain Sciences, Beijing, China.,State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yun-Ling He
- Beijing Institute of Cognition and Brain Sciences, Beijing, China
| | - Ling-Ling Zhu
- Beijing Institute of Cognition and Brain Sciences, Beijing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
8
|
Muñoz‐Sánchez J, Chánez‐Cárdenas ME. The use of cobalt chloride as a chemical hypoxia model. J Appl Toxicol 2018; 39:556-570. [DOI: 10.1002/jat.3749] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/13/2018] [Accepted: 10/07/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Jorge Muñoz‐Sánchez
- Laboratorio de Patología Vascular CerebralInstituto Nacional de Neurología y Neurología (INNN) Insurgentes Sur 3877, la Fama 14269 Tlalpan Ciudad de México Mexico
| | - María E. Chánez‐Cárdenas
- Laboratorio de Patología Vascular CerebralInstituto Nacional de Neurología y Neurología (INNN) Insurgentes Sur 3877, la Fama 14269 Tlalpan Ciudad de México Mexico
| |
Collapse
|
9
|
Østergaard L, Jørgensen MB, Knudsen GM. Low on energy? An energy supply-demand perspective on stress and depression. Neurosci Biobehav Rev 2018; 94:248-270. [DOI: 10.1016/j.neubiorev.2018.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 07/09/2018] [Accepted: 08/13/2018] [Indexed: 12/17/2022]
|
10
|
Abstract
Stroke-induced endothelial cell injury leads to destruction of cerebral microvasculature and significant damage to the brain tissue. A subacute phase of cerebral ischemia is associated with regeneration involving the activation of vascular remodeling, neuroplasticity, neurogenesis, and neuroinflammation processes. Effective restoration and improvement of blood supply to the damaged brain tissue offers a potential therapy for stroke. microRNAs (miRNAs) are recently identified small RNA molecules that regulate gene expression and significantly influence the essential cellular processes associated with brain repair following stroke. A number of specific miRNAs are implicated in regulating the development and propagation of the ischemic tissue damage as well as in mediating post-stroke regeneration. In this review, I discuss the functions of the miRNA miR-155 and the effect of its in vivo inhibition on brain recovery following experimental cerebral ischemia. The article introduces new and unexplored approach to cerebral regeneration: regulation of brain tissue repair through a direct modulation of specific miRNA activity.
Collapse
Affiliation(s)
- Tamara Roitbak
- Department of Neurosurgery, Health Sciences Center, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
11
|
Thomas TM, Yu JS. Metabolic regulation of glioma stem-like cells in the tumor micro-environment. Cancer Lett 2017; 408:174-181. [PMID: 28743531 PMCID: PMC5790120 DOI: 10.1016/j.canlet.2017.07.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/11/2017] [Accepted: 07/15/2017] [Indexed: 12/18/2022]
Abstract
Cancer metabolism has emerged as one of the most interesting old ideas being revisited from a new perspective. In the early 20th century Otto Warburg declared metabolism the prime cause in a disease of many secondary causes, and this statement seems more prescient in view of modern expositions into the true nature of tumor evolution. As the complexity of tumor heterogeneity becomes more clear from a genetic perspective, it is important to consider the inevitably heterogeneous metabolic components of the tumor and the tumor microenvironment. High grade gliomas remain one of the most difficult to treat solid tumors, due in part to the highly vascularized nature of the tumor and the maintenance of more resistant stem-like subpopulations within the tumor. Maintenance of glioma stem cells (GSCs) requires specific alterations within the cells and the greater tumor microenvironment with regards to signaling and metabolism. Specific niches within gliomas help foster the survival of stem-like sub-populations of cells with high tumorigenicity and high metabolic plasticity. Understanding these maintenance pathways and the metabolic dependencies within the niche may highlight potential avenues of addressing tumor resistance and recurrence in glioma patients.
Collapse
Affiliation(s)
- Tom M Thomas
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - John S Yu
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
12
|
Mu S, Guo S, Wang X, Zhan Y, Li Y, Jiang Y, Zhang R, Zhang B. Effects of deferoxamine on the osteogenic differentiation of human periodontal ligament cells. Mol Med Rep 2017; 16:9579-9586. [PMID: 29039615 DOI: 10.3892/mmr.2017.7810] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 08/17/2017] [Indexed: 11/06/2022] Open
Abstract
Hypoxia regulates a number of cell biological processes, including cell survival, development and differentiation. Deferoxamine (DFO), an oral chelator for blood transfusion patients, has been demonstrated to induce hypoxia and is frequently used as a hypoxia‑mimicking agent. The purpose of the present study was to investigate the influence of DFO on the proliferation, migration and osteogenic differentiation of human periodontal ligament cells (hPDLCs). The effects of DFO on hPDLC viability and migration were measured using an MTT and wound healing assay. To characterize the hypoxia microenvironment, the expression of hypoxia‑inducible factor‑1α (HIF‑1α) in hPDLCs treated with DFO was quantified using the reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). Subsequently, the osteogenic differentiation potential of DFO was determined by RT‑qPCR of the mRNA of osteogenic markers (runt‑related transcription factor 2 [Runx‑2], osteopontin [OPN] and collagen type I [Col‑1]). The alkaline phosphatase activity and mineral deposition were analyzed using alizarin red S staining. The MTT and wound healing assays demonstrated that low‑concentrations of DFO had little impact on hPDLC viability and migration 48 h into the treatment. DFO upregulated the expression of hPDLC genes specific for osteogenic differentiation: HIF‑1α, Runx‑2, OPN and Col‑1. Furthermore, formation of mineralized nodules was enhanced by DFO. The present study suggests that DFO provided favorable culture conditions to promote the osteogenic differentiation and mineralization of hPDLCs. The mechanism underlying these alterations remains to be elucidated.
Collapse
Affiliation(s)
- Sen Mu
- Department of Periodontology and Oral Mucosa, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Shuanlong Guo
- Department of Stomatology, Fenyang Hospital, Fenyang, Shanxi 032200, P.R. China
| | - Xiang Wang
- Department of General Dentistry, Yinzhou Stomatology Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Yuanbo Zhan
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Ying Li
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Ying Jiang
- Pediatric Department of Stomatology, Yinzhou Stomatology Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Ruimin Zhang
- Department of Periodontology and Oral Mucosa, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Bin Zhang
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
13
|
Luan Y, Zhang X, Zhang Y, Dong Y. MicroRNA-210 Protects PC-12 Cells Against Hypoxia-Induced Injury by Targeting BNIP3. Front Cell Neurosci 2017; 11:285. [PMID: 29018330 PMCID: PMC5614931 DOI: 10.3389/fncel.2017.00285] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 09/01/2017] [Indexed: 11/30/2022] Open
Abstract
MicroRNA (miR)-210 is the most consistently and predominantly up-regulated miR in response to hypoxia in multiple cancer cells. The roles of miR-210 in rat adrenal gland pheochromocytoma (PC-12) cells remain unknown. We aimed to explore the possible effect of miR-210 in neonatal brain injury. We explored the potential molecular mechanism by using PC-12 cells under hypoxia. Scramble miRs, miR-210 mimic, miR-210 inhibitor or its negative control were respectively transfected into PC-12 cells. Cell viability, migration, invasion and apoptosis were also assessed to evaluate hypoxia-induced cell injury. The expression level of miR-210 was identified by quantitative real-time polymerase chain reaction (qRT-PCR) analysis. Apoptosis-related protein expression as well as key kinases in the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signal pathway was studied by Western blot analysis. Hypoxia suppressed cell viability, migration and invasion, but promoted apoptosis through activation of mitochondrial- and caspase-dependent pathways. Hypoxia markedly induced up-regulation of miR-210 in PC-12 cells. Overexpression of miR-210 protected PC-12 cells against hypoxia-induced injury. Bcl-2 adenovirus E1B 19 kDa-interacting protein 3 (BNIP3) was proven to be a target gene of miR-210 in PC-12 cells. miR-210 overexpression ameliorated the hypoxia-induced injury in PC-12 cells by down-regulating BNIP3. Hypoxia-induced alterations of key kinases in the PI3K/AKT/mTOR signal pathway were affected by aberrant expression of BNIP3. These findings suggested that miR-210 protected PC-12 cells against hypoxia-induced injury by targeting BNIP3, involving the PI3K/AKT/mTOR signal pathway.
Collapse
Affiliation(s)
- Yonggang Luan
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| | - Xiaoli Zhang
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| | - Yongli Zhang
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| | - Yubin Dong
- Department of Neonatal Intensive Care Unit, Zhoukou Central HospitalZhoukou, China
| |
Collapse
|
14
|
Abstract
One of the differences between normal and cancer cells is lower pH of the extracellular space in tumors. Low pH in the extracellular space activates proteases and stimulates tumor invasion and metastasis. Tumor cells display higher level of the HIF1α transcription factor that promotes cell switch from mitochondrial respiration to glycolysis. The terminal product of glycolysis is lactate. Lactate formation from pyruvate is catalyzed by the specific HIF1α-dependent isoform of lactate dehydrogenase A. Because lactate accumulation is deleterious for the cell, it is actively exported by monocarboxylate transporters. Lactate is cotransported with proton, which acidifies the extracellular space. Another protein that contributes to proton concentration increase in the extracellular space is tumor-specific HIF1α-dependent carbonic anhydrase IX, which generates a proton in the reaction between carbon dioxide and water. The activity of Na+/H+ exchanger (another protein pump) is stimulated by stress factors (e.g. osmotic shock) and proliferation stimuli. This review describes the mechanisms of proton pump activation and reviews results of studies on effects of various proton pump inhibitors on tumor functioning and growth in cell culture and in vivo. The prospects of combined application of proton pump inhibitors and cytostatics in cancer therapy are discussed.
Collapse
Affiliation(s)
- V A Kobliakov
- Blokhin Russian Cancer Research Center, Russian Ministry of Health, Moscow, 115478, Russia.
| |
Collapse
|
15
|
Tang X, Qin H, Gu X, Fu X. China’s landscape in regenerative medicine. Biomaterials 2017; 124:78-94. [DOI: 10.1016/j.biomaterials.2017.01.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 01/24/2017] [Accepted: 01/28/2017] [Indexed: 12/15/2022]
|
16
|
Espinoza I, Sakiyama MJ, Ma T, Fair L, Zhou X, Hassan M, Zabaleta J, Gomez CR. Hypoxia on the Expression of Hepatoma Upregulated Protein in Prostate Cancer Cells. Front Oncol 2016; 6:144. [PMID: 27379206 PMCID: PMC4908134 DOI: 10.3389/fonc.2016.00144] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/26/2016] [Indexed: 01/26/2023] Open
Abstract
Hepatoma upregulated protein (HURP) is a multifunctional protein with clinical promise. This protein has been demonstrated to be a predictive marker for the outcome in high-risk prostate cancer (PCa) patients, besides being a resistance factor in PCa. Although changes in oxygen tension (pO2) are associated with PCa aggressiveness, the role of hypoxia in the regulation of tumor progression genes such as HURP has not yet been described. We hypothesized that pO2 alteration is involved in the regulation of HURP expression in PCa cells. In the present study, PCa cells were incubated at 2% O2 (hypoxia) and 20% O2 (normoxia) conditions. Hypoxia reduced cell growth rate of PCa cells, when compared to the growth rate of cells cultured under normoxia (p < 0.05). The decrease in cell viability was accompanied by fivefold (p < 0.05) elevated rate of vascular endothelial growth factor (VEGF) release. The expression of VEGF and the hypoxia-inducible metabolic enzyme carbonic anhydrase 9 were elevated maximally nearly 61-fold and 200-fold, respectively (p < 0.05). Noted in two cell lines (LNCaP and C4-2B) and independent of the oxygen levels, HURP expression assessed at both mRNA and protein levels was reduced. However, the decrease was more pronounced in cells cultured under hypoxia (p < 0.05). Interestingly, the analysis of patients’ specimens by Western blot revealed a marked increase of HURP protein (fivefold), when compared to control (cystoprostatectomy) tissue (p < 0.05). Immunohistochemistry analysis showed an increase in the immunostaining intensity of HURP and the hypoxia-sensitive molecules, hypoxia-inducible factor 1-alpha (HIF-1α), VEGF, and heat-shock protein 60 (HSP60) in association with tumor grade. The data also suggested a redistribution of subcellular localization for HURP and HIF-1α from the nucleus to the cytoplasmic compartment in relation to increasing tumor grade. Analysis of HURP Promoter for HIF-1-binding sites revealed presence of four putative HIF binding sites on the promoter of DLGAP5/HURP gene in the non-translated region upstream from the start codon, suggesting association between HIF-1α and the regulation of HURP protein. Taken together, our findings suggest a modulatory role of hypoxia on the expression of HURP. Additionally our results provide basis for utilization of tumor-associated molecules as predictors of aggressive PCa.
Collapse
Affiliation(s)
- Ingrid Espinoza
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Preventive Medicine, University of Mississippi Medical Center, Jackson, MS, USA; Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | - Marcelo J Sakiyama
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA; CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Tangeng Ma
- Cancer Institute, University of Mississippi Medical Center , Jackson, MS , USA
| | - Logan Fair
- School of Medicine, University of Mississippi Medical Center , Jackson, MS , USA
| | - Xinchun Zhou
- Department of Pathology, University of Mississippi Medical Center , Jackson, MS , USA
| | - Mohamed Hassan
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jovanny Zabaleta
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA; Department of Pediatrics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Christian R Gomez
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA; Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA; Department of Radiation Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
17
|
Richards R, Jenkinson MD, Haylock BJ, See V. Cell cycle progression in glioblastoma cells is unaffected by pathophysiological levels of hypoxia. PeerJ 2016; 4:e1755. [PMID: 26966676 PMCID: PMC4782743 DOI: 10.7717/peerj.1755] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/12/2016] [Indexed: 12/31/2022] Open
Abstract
Hypoxia is associated with the increased malignancy of a broad range of solid tumours. While very severe hypoxia has been widely shown to induce cell cycle arrest, the impact of pathophysiological hypoxia on tumour cell proliferation is poorly understood. The aim of this study was to investigate the effect of different oxygen levels on glioblastoma (GBM) cell proliferation and survival. GBM is an extremely aggressive brain tumour with a heterogeneous oxygenation pattern. The effects of a range of oxygen tensions on GBM cell lines and primary cells were assessed using flow cytometry. Results indicate that cell cycle distribution and viability are unaffected by long term exposure (24–96 h) to pathophysiological levels of oxygen (1–8% O2). Both transient cell cycle arrest and small amounts of cell death could only be detected when cells were exposed to severe hypoxia (0.1% O2). No significant changes in p21 protein expression levels were detected. These findings reinforce the importance of using physiologically relevant oxygen tensions when investigating tumour hypoxia, and help to explain how solid tumours can be both hypoxic and highly proliferative, as is the case with GBM.
Collapse
Affiliation(s)
- Rosalie Richards
- Institute of Integrative Biology, Department of Biochemistry, University of Liverpool , Liverpool , United Kingdom
| | - Michael D Jenkinson
- Institute of Translational Medicine, Clinical Science Centre, University of Liverpool , Liverpool , United Kingdom
| | - Brian J Haylock
- Department of Clinical Oncology, Clatterbridge Cancer Centre , Bebington , United Kingdom
| | - Violaine See
- Institute of Integrative Biology, Department of Biochemistry, University of Liverpool , Liverpool , United Kingdom
| |
Collapse
|
18
|
Chen Y, Guan R, Zhang C, Huang J, Ji L, Chao H. Two-photon luminescent metal complexes for bioimaging and cancer phototherapy. Coord Chem Rev 2016. [DOI: 10.1016/j.ccr.2015.09.010] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
19
|
Wagenführ L, Meyer AK, Marrone L, Storch A. Oxygen Tension Within the Neurogenic Niche Regulates Dopaminergic Neurogenesis in the Developing Midbrain. Stem Cells Dev 2016; 25:227-38. [PMID: 26577812 DOI: 10.1089/scd.2015.0214] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oxygen tension is an important factor controlling stem cell proliferation and maintenance in various stem cell populations with a particular relevance in midbrain dopaminergic progenitors. Further studies have shown that the oxygen-dependent transcription factor hypoxia-inducible factor 1α (HIF-1α) is involved in these processes. However, all available studies on oxygen effects in dopaminergic neuroprogenitors were performed in vitro and thus it remains unclear whether tissue oxygen tension in the embryonic midbrain is also relevant for the regulation of dopaminergic neurogenesis in vivo. We thus dissect here the effects of oxygen tension in combination with HIF-1α conditional knockout on dopaminergic neurogenesis by using a novel experimental design allowing for the control of oxygen tension within the microenvironment of the neurogenic niche of the murine fetal midbrain in vivo. The microenvironment of the midbrain dopaminergic neurogenic niche was detected as hypoxic with oxygen tensions below 1.1%. Maternal oxygen treatment of 10%, 21%, and 75% atmospheric oxygen tension for 48 h translates into robust changes in fetal midbrain oxygenation. Fetal midbrain hypoxia hampered the generation of dopaminergic neurons and is accompanied with restricted fetal midbrain development. In contrast, induced hyperoxia stimulated proliferation and differentiation of dopaminergic progenitors during early and late embryogenesis. Oxygen effects were not directly mediated through HIF-1α signaling. These data--in agreement with in vitro data-indicate that oxygen is a crucial regulator of developmental dopaminergic neurogenesis. Our study provides the initial framework for future studies on molecular mechanisms mediating oxygen regulation of dopaminergic neurogenesis within the fetal midbrain as its natural environment.
Collapse
Affiliation(s)
- Lisa Wagenführ
- 1 Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden , Dresden, Germany
| | - Anne Karen Meyer
- 1 Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden , Dresden, Germany
| | - Lara Marrone
- 1 Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden , Dresden, Germany .,2 Center for Regenerative Therapies Dresden (CRTD) , Technische Universität Dresden, Dresden, Germany
| | - Alexander Storch
- 1 Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden , Dresden, Germany .,2 Center for Regenerative Therapies Dresden (CRTD) , Technische Universität Dresden, Dresden, Germany .,3 Department of Neurology, University of Rostock , Rostock, Germany .,4 German Centre for Neurodegenerative Diseases (DZNE) , Rostock, Germany
| |
Collapse
|
20
|
Zhang K, Zhao T, Huang X, He Y, Zhou Y, Wu L, Wu K, Fan M, Zhu L. Dissolved oxygen concentration in the medium during cell culture: Defects and improvements. Cell Biol Int 2016; 40:354-60. [PMID: 26648388 DOI: 10.1002/cbin.10570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/01/2015] [Indexed: 12/21/2022]
Abstract
In vitro cell culture has provided a useful model to study the effects of oxygen on cellular behavior. However, it remains unknown whether the in vitro operations themselves affect the medium oxygen levels and the living states of cells. In addition, a prevailing controversy is whether reactive oxygen species (ROS) production is induced by continuous hypoxia or reoxygenation. In this study, we have measured the effects of different types of cell culture containers and the oxygen environment where medium replacement takes place on the actual oxygen tension in the medium. We found that the deviations of oxygen concentrations in the medium are much greater in 25-cm(2) flasks than in 24-well plates and 35-mm dishes. The dissolved oxygen concentrations in the medium were increased after medium replacement in normoxia, but remained unchanged in glove boxes in which the oxygen tension remained at a low level (11.4, 5.7, and 0.5% O2 ). We also found that medium replacement in normoxia increased the number of ROS-positive cells and reduced the cell viability; meanwhile, medium replacement in a glove box did not produce the above effects. Therefore, we conclude that the use of 25-cm(2) flasks should be avoided and demonstrate that continuous hypoxia does not produce ROS, whereas the reoxygenation that occurs during the harvesting of cells leads to ROS and induces cell death.
Collapse
Affiliation(s)
- Kuan Zhang
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China.,Brain Research Center, College of Basic Medical Sciences, Third Military Medical University, Chongqing, 400038, China
| | - Tong Zhao
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xin Huang
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yunlin He
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yanzhao Zhou
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Liying Wu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Kuiwu Wu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ming Fan
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China
| | - Lingling Zhu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, 100850, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, 226001, China
| |
Collapse
|
21
|
Hypoxic Adaptation in the Nervous System: Promise for Novel Therapeutics for Acute and Chronic Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 903:221-43. [PMID: 27343100 DOI: 10.1007/978-1-4899-7678-9_16] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Homeostasis is the process by which cells adapt to stress and prevent or repair injury. Unique programs have evolved to sense and activate these homeostatic mechanisms and as such, homeostatic sensors may be potent therapeutic targets. The hypoxic response mediated by hypoxia inducible factor (HIF) downstream of oxygen sensing by HIF prolyl 4-hydroxylases (PHDs) has been well-studied, revealing cell-type specific regulation of HIF stability, activity, and transcriptional targets. HIF's paradoxical roles in nervous system development, physiology, and pathology arise from its complex roles in hypoxic adaptation and normoxic biology. Understanding how to engage the hypoxic response so as to recapitulate the protective mechanism of ischemic preconditioning is a high priority. Indeed, small molecules that activate the hypoxic response provide broad neuroprotection in several clinically relevant injury models. Screens for PHD inhibitors have identified novel therapeutics for neuroprotection that are ready to proceed to clinical trials for ischemic stroke. Better understanding the mechanisms of how to engage hypoxic adaption without altering development or physiology may identify additional novel therapeutic targets for diverse acute and chronic neuropathologies.
Collapse
|
22
|
Ma T, Schreiber CA, Knutson GJ, Khattouti AE, Sakiyama MJ, Hassan M, Charlesworth MC, Madden BJ, Zhou X, Vuk-Pavlović S, Gomez CR. Effects of oxygen on the antigenic landscape of prostate cancer cells. BMC Res Notes 2015; 8:687. [PMID: 26581192 PMCID: PMC4652345 DOI: 10.1186/s13104-015-1633-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023] Open
Abstract
Background Use of allogeneic cancer cells-based immunotherapy for treatment of established prostate cancer (PCa) has only been marginally effective. One reason for failure could stem from the mismatch of antigenic signatures of vaccine cells and cancer in situ. Hence, it is possible that vaccine cells expressed antigens differently than tumor cells in situ. We hypothesized that cells grown in vitro at low oxygen tension (pO2) provide a better antigen match to tumors in situ and could reveal a more relevant antigenic landscape than cells grown in atmospheric pO2. Methods We tested this hypothesis by comparing PCa cells propagated at pO2 = 2 kPa and 20 kPa. To identify potential tumor-associated antigens (TAAs), we prepared PCa cell lysates, resolved them by two-dimensional electrophoresis and immunoblotting using spontaneous antibodies from plasma derived from PCa patients and control subjects. Antibody-labeled spots were analyzed by MALDI-TOF mass spectrometry and validated by ELISA. We selected hypoxia-regulated HSP70 and hnRNP L and hypoxia-independent HSP60 and determined the frequency of plasma samples reacting with these molecules. Results Frequency of HSP60-reactive plasma was low in healthy controls [1.3 % (1/76)], while it was elevated in PCa patients [13.0 % (7/54); p < 0.05]. These data suggest a humoral immune response to HSP60 in PCa. Levels of autoantibodies to HSP70 did not differ from healthy controls [3.7 % (2/54)] in PCa patients [5.3 % (2/38)]. Similarly, hnRNP L autoantibodies did no differ between healthy controls [6.1 % (3/49)] and PCa patients [5.3 % (2/38)]. Conclusions Overall our results suggest the value of hypoxia as a modifier of the cellular and antigenic landscape of PCa cells. By modifying the immune reactivity of PCa cells in culture, manipulation of pO2 can be proposed as a new avenue for improving diagnosis, prognosis and immunotherapy for PCa.
Collapse
Affiliation(s)
- Tangeng Ma
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | - Claire A Schreiber
- Stem Cell Laboratory, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| | - Gaylord J Knutson
- Stem Cell Laboratory, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| | - Abdelouahid El Khattouti
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | - Marcelo J Sakiyama
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | - Mohamed Hassan
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | | | - Benjamin J Madden
- Proteomics Research Center, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| | - Xinchun Zhou
- Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA.
| | - Stanimir Vuk-Pavlović
- Stem Cell Laboratory, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA. .,Division of Hematology, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA. .,Division of Preventive and Occupational Medicine, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| | - Christian R Gomez
- Cancer Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Department of Pathology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Department of Radiation Oncology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216, USA. .,Stem Cell Laboratory, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA. .,Division of Hematology, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA. .,Division of Preventive and Occupational Medicine, Department of Internal Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
23
|
Zhang K, Zhou Y, Zhao T, Wu L, Huang X, Wu K, Xu L, Li D, Liu S, Zhao Y, Fan M, Zhu L. Reduced Cerebral Oxygen Content in the DG and SVZ In Situ Promotes Neurogenesis in the Adult Rat Brain In Vivo. PLoS One 2015; 10:e0140035. [PMID: 26466323 PMCID: PMC4605722 DOI: 10.1371/journal.pone.0140035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 09/20/2015] [Indexed: 01/09/2023] Open
Abstract
Neurogenesis in the adult brain occurs mainly within two neurogenic structures, the dentate gyrus (DG) of the hippocampus and the sub-ventricular zone (SVZ) of the forebrain. It has been reported that mild hypoxia promoted the proliferation of Neural Stem Cells (NSCs)in vitro. Our previous study further demonstrated that an external hypoxic environment stimulated neurogenesis in the adult rat brain in vivo. However, it remains unknown how external hypoxic environments affect the oxygen content in the brain and result in neurogenesis. Here we use an optical fiber luminescent oxygen sensor to detect the oxygen content in the adult rat brain in situ under normoxia and hypoxia. We found that the distribution of oxygen in cerebral regions is spatiotemporally heterogeneous. The Po2 values in the ventricles (45∼50 Torr) and DG (approximately 10 Torr) were much higher than those of other parts of the brain, such as the cortex and thalamus (approximately 2 Torr). Interestingly, our in vivo studies showed that an external hypoxic environment could change the intrinsic oxygen content in brain tissues, notably reducing oxygen levels in both the DG and SVZ, the major sites of adult neurogenesis. Furthermore, the hypoxic environment also increased the expression of HIF-1α and VEGF, two factors that have been reported to regulate neurogenesis, within the DG and SVZ. Thus, we have demonstrated that reducing the oxygen content of the external environment decreased Po2 levels in the DG and SVZ. This reduced oxygen level in the DG and SVZ might be the main mechanism triggering neurogenesis in the adult brain. More importantly, we speculate that varying oxygen levels may be the physiological basis of the regionally restricted neurogenesis in the adult brain.
Collapse
Affiliation(s)
- Kuan Zhang
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
- Brain Research Center, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Yanzhao Zhou
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Tong Zhao
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Liying Wu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Xin Huang
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Kuiwu Wu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Lun Xu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Dahu Li
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Shuhong Liu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Yongqi Zhao
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
| | - Ming Fan
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Beijing Institute for Brain Disorders, 10 Xitoutiao, You Anmen, Fengtai District, Beijing, P.R. China
- * E-mail: (LZ); (MF)
| | - Lingling Zhu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Beijing, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- * E-mail: (LZ); (MF)
| |
Collapse
|
24
|
Wagenführ L, Meyer AK, Braunschweig L, Marrone L, Storch A. Brain oxygen tension controls the expansion of outer subventricular zone-like basal progenitors in the developing mouse brain. Development 2015; 142:2904-15. [DOI: 10.1242/dev.121939] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mammalian neocortex shows a conserved six-layered structure that differs between species in the total number of cortical neurons produced owing to differences in the relative abundance of distinct progenitor populations. Recent studies have identified a new class of proliferative neurogenic cells in the outer subventricular zone (OSVZ) in gyrencephalic species such as primates and ferrets. Lissencephalic brains of mice possess fewer OSVZ-like progenitor cells and these do not constitute a distinct layer. Most in vitro and in vivo studies have shown that oxygen regulates the maintenance, proliferation and differentiation of neural progenitor cells. Here we dissect the effects of fetal brain oxygen tension on neural progenitor cell activity using a novel mouse model that allows oxygen tension to be controlled within the hypoxic microenvironment in the neurogenic niche of the fetal brain in vivo. Indeed, maternal oxygen treatment of 10%, 21% and 75% atmospheric oxygen tension for 48 h translates into robust changes in fetal brain oxygenation. Increased oxygen tension in fetal mouse forebrain in vivo leads to a marked expansion of a distinct proliferative cell population, basal to the SVZ. These cells constitute a novel neurogenic cell layer, similar to the OSVZ, and contribute to corticogenesis by heading for deeper cortical layers as a part of the cortical plate.
Collapse
Affiliation(s)
- Lisa Wagenführ
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Fetscherstrasse 74, Dresden 01307, Germany
| | - Anne K. Meyer
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Fetscherstrasse 74, Dresden 01307, Germany
- Leibniz Institute for Solid State and Material Research, IFW Dresden, Institute for Integrative Nanosciences, Helmholtzstrasse 20, Dresden 01069, Germany
| | - Lena Braunschweig
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Fetscherstrasse 74, Dresden 01307, Germany
| | - Lara Marrone
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Fetscherstrasse 74, Dresden 01307, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Fetscherstrasse 105, Dresden 01307, Germany
| | - Alexander Storch
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Fetscherstrasse 74, Dresden 01307, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Fetscherstrasse 105, Dresden 01307, Germany
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Arnoldstrasse 18, Dresden 01307, Germany
| |
Collapse
|
25
|
Li R, Wang Y, Yang Z, He Y, Zhao T, Fan M, Wang X, Zhu L, Wang X. Hypoxia-inducible factor-1α regulates the expression of L-type voltage-dependent Ca(2+) channels in PC12 cells under hypoxia. Cell Stress Chaperones 2015; 20:507-16. [PMID: 25648081 PMCID: PMC4406929 DOI: 10.1007/s12192-015-0575-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 01/16/2015] [Accepted: 01/23/2015] [Indexed: 10/24/2022] Open
Abstract
Hypoxia is an important factor in regulation of cell behavior both under physiological and pathological conditions. The mechanisms of hypoxia-induced cell death have not been completely elucidated yet. It is well known that Ca(2+) is critically related to cell survival. Hypoxia-inducible factor-1α (HIF-1α) is a core regulatory factor during hypoxia, and L-type voltage-dependent Ca(2+) channels (L-VDCCs) have been reported to play a critical role in cell survival. This study was conducted to explore the relationship between L-VDCC expression and HIF-1α regulation in PC12 cells under hypoxia. PC12 cells were treated at 20 or 3 % O2 to observe its proliferation and the intracellular calcium concentration. Then, we detected the protein expression of HIF-1α and L-VDCCs subtypes, Cav1.2 and Cav1.3. At last, to verify the relationship between HIF-1α and Cav1.2 and Cav1.3, we got the expression of Cav1.2 and Cav1.3 with Western blot and luciferase report gene assays after PC12 cells were treated by echinomycin, which is an HIF-1α inhibitor. Compared with 20 % O2 (normoxia), 3 % O2 (hypoxia) inhibited cell proliferation, increased the intracellular calcium concentration, and induced protein expression of HIF-1α. The protein expression of two L-VDCCs subtypes expressed in the nervous system, Cav1.2 and Cav1.3, was upregulated by hypoxia and reduced dose dependently by treatment with echinomycin, a HIF-1α inhibitor. Luciferase report gene assays showed that the expression of Cav1.2 and Cav1.3 genes was augmented under 3 % O2. However, echinomycin only slightly and dose dependently decreased expression of the Cav1.2 gene, but not that of the Cav1.3 gene. These data indicated that Cav1.2 might be regulated by HIF-1α as one of its downstream target genes and involved in regulation of PC12 cells death under hypoxia.
Collapse
Affiliation(s)
- Ran Li
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
- />Department of Rehabilitation Medicine, Xuan Xu Hospital, Capital Medical University, 45# Changchun Street, Beijing, 100053 People’s Republic of China
| | - Yong Wang
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
| | - Zhaofei Yang
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
| | - Yunling He
- />Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, 27# Taiping Road, Beijing, 100850 People’s Republic of China
| | - Tong Zhao
- />Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, 27# Taiping Road, Beijing, 100850 People’s Republic of China
| | - Ming Fan
- />Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, 27# Taiping Road, Beijing, 100850 People’s Republic of China
| | - Xuan Wang
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
| | - Lingling Zhu
- />Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, 27# Taiping Road, Beijing, 100850 People’s Republic of China
| | - Xiaomin Wang
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
- />Beijing Institute for Brain Disorder, 10# You An Men, Beijing, 100069 People’s Republic of China
| |
Collapse
|
26
|
Daniel G, Schmidt-Edelkraut U, Spengler D, Hoffmann A. Imprinted Zac1 in neural stem cells. World J Stem Cells 2015; 7:300-314. [PMID: 25815116 PMCID: PMC4369488 DOI: 10.4252/wjsc.v7.i2.300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/24/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Neural stem cells (NSCs) and imprinted genes play an important role in brain development. On historical grounds, these two determinants have been largely studied independently of each other. Recent evidence suggests, however, that NSCs can reset select genomic imprints to prevent precocious depletion of the stem cell reservoir. Moreover, imprinted genes like the transcriptional regulator Zac1 can fine tune neuronal vs astroglial differentiation of NSCs. Zac1 binds in a sequence-specific manner to pro-neuronal and imprinted genes to confer transcriptional regulation and furthermore coregulates members of the p53-family in NSCs. At the genome scale, Zac1 is a central hub of an imprinted gene network comprising genes with an important role for NSC quiescence, proliferation and differentiation. Overall, transcriptional, epigenomic, and genomic mechanisms seem to coordinate the functional relationships of NSCs and imprinted genes from development to maturation, and possibly aging.
Collapse
|
27
|
Radiation therapy for glioma stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 853:85-110. [PMID: 25895709 DOI: 10.1007/978-3-319-16537-0_6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Radiation therapy is the most effective adjuvant treatment modality for virtually all patients with high-grade glioma. Its ability to improve patient survival has been recognized for decades. Cancer stem cells provide new insights into how tumor biology is affected by radiation and the role that this cell population can play in disease recurrence. Glioma stem cells possess a variety of intracellular mechanisms to resist and even flourish in spite of radiation, and their proliferation and maintenance appear tied to supportive stimuli from the tumor microenvironment. This chapter reviews the basis for our current use of radiation to treat high-grade gliomas, and addresses this model in the context of therapeutically resistant stem cells. We discuss the available evidence highlighting current clinical efforts to improve radiosensitivity, and newer targets worthy of further development.
Collapse
|
28
|
Trollmann R, Richter M, Jung S, Walkinshaw G, Brackmann F. Pharmacologic stabilization of hypoxia-inducible transcription factors protects developing mouse brain from hypoxia-induced apoptotic cell death. Neuroscience 2014; 278:327-42. [PMID: 25162122 DOI: 10.1016/j.neuroscience.2014.08.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 07/25/2014] [Accepted: 08/15/2014] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Accumulation of hypoxia-inducible transcription factors (HIFs) by prolyl-4-hydroxylase inhibitors (PHI) has been suggested to induce neuroprotection in the ischemic rodent brain. We aimed to investigate in vivo effects of a novel PHI on HIF-regulated neurotrophic and pro-apoptotic factors in the developing normoxic and hypoxic mouse brain. METHODS Neonatal mice (P7) were treated with PHI FG-4497 (30-100mg/kg, i.p.) followed by exposure to systemic hypoxia (8% O2, 6h) 4h later. Cerebral expression of HIFα-subunits, specific neurotrophic and vasoactive target genes (vascular endothelial growth factor (VEGF), adrenomedullin (ADM), erythropoietin (EPO), inducible nitric oxide synthase (iNOS)) as well as pro-apoptotic (BCL2/adenovirus E1B 19-kDa protein-interacting protein 3 gene (BNIP3), immediate early response 3 (IER3)) and migratory factors (chemokine receptor 4 (CXCR4), stromal cell-derived factor 1 (SDF-1)) was determined (quantitative real-time (RT)., Western blot analysis) in comparison to controls. Apoptotic cell death was analyzed by terminal desoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) and cleaved caspase 3 (CC3) staining. RESULTS Under normoxic conditions, FG-4497 treatment significantly induced the accumulation of both HIF-1α and HIF-2α isoforms in developing mouse brain. In addition, there was a significant up-regulation of HIF target genes (VEGF, ADM, EPO, CXCR4, p<0.01) with FG-4497 treatment compared to controls supporting functional activation of the HIF proteins. Under hypoxia, differential target gene activation was observed in the developing brain including additive effects of FG-4497 and hypoxia on mRNA expression of VEGF and ADM as well as a dose-dependent down-regulation of iNOS. BNIP3 but not IER3 mRNA levels significantly increased in hypoxic brains pre-treated with high-dose FG-4497 compared to controls. Of special interest, FG-4497 treatment significantly diminished apoptotic cell death, quantified by TUNEL and CC3-positive cells, in hypoxic developing brains compared to controls. CONCLUSIONS PHI treatment modulates neurotrophic factors known to be crucially involved in hypoxia-induced cerebral adaptive mechanisms as well as early brain maturation. Pre-treatment with FG-4497 seems to protect the developing brain from hypoxia-induced apoptosis. Present observations provide basic information for further evaluation of neuroprotective properties of PHI treatment in hypoxic injury of the developing brain. However, potential effects on maturational processes need special attention in experimental research targeting HIF-dependent neuroprotective interventions during the very early stage of brain development.
Collapse
Affiliation(s)
- R Trollmann
- Department of Pediatrics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany.
| | - M Richter
- Department of Pediatrics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - S Jung
- Department of Pediatrics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | | | - F Brackmann
- Department of Pediatrics, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
29
|
Hawkins KE, Sharp TV, McKay TR. The role of hypoxia in stem cell potency and differentiation. Regen Med 2014; 8:771-82. [PMID: 24147532 DOI: 10.2217/rme.13.71] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Regenerative medicine relies on harnessing the capacity of stem cells to grow, divide and differentiate safely and predictably. This may be in the context of expanding stem cells in vitro or encouraging their expansion, mobilization and capacity to regenerate tissues either locally or remotely in vivo. In either case, understanding the stem cell niche is fundamental to recapitulating or manipulating conditions to enable therapy. It has become obvious that hypoxia plays a fundamental role in the maintenance of the stem cell niche. Low O2 benefits the self-renewal of human embryonic, hematopoietic, mesenchymal and neural stem cells, as well as improving the efficiency of genetic reprogramming to induced pluripotency. There is emerging evidence that harnessing or manipulating the hypoxic response can result in safer, more efficacious methodologies for regenerative medicine.
Collapse
Affiliation(s)
- Kate E Hawkins
- Division of Biomedical Sciences, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | | | | |
Collapse
|
30
|
Wang W, Wang P, Li S, Yang J, Liang X, Tang Y, Li Y, Yang R, Wu Y, Shen H. Methylprednisolone inhibits the proliferation and affects the differentiation of rat spinal cord-derived neural progenitor cells cultured in low oxygen conditions by inhibiting HIF-1α and Hes1 in vitro. Int J Mol Med 2014; 34:788-95. [PMID: 24992925 DOI: 10.3892/ijmm.2014.1835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 06/05/2014] [Indexed: 11/05/2022] Open
Abstract
Although there is much controversy over the use of methylprednisolone (MP), it is one of the main drugs used in the treatment of acute spinal cord injury (SCI). The induction of the proliferation and differentiation of endogenous neural progenitor cells (NPCs) is considered a promising mode of treatment for SCI. However, the effects of MP on spinal cord-derived endogenous NPCs in a low oxygen enviroment remain to be delineated. Thus, the aim of this study was to investigate the potential effects of MP on NPCs cultured under low oxygen conditions in vitro and to elucidate the molecular mechanisms involved. Fetal rat spinal cord-derived NPCs were harvested and divided into 4 groups: 2 groups of cells cultured under normal oxygen conditions and treated with or without MP, and 2 groups incubated in 3% O2 (low oxygen) treated in a similar manner. We found that MP induced suppressive effects on NPC proliferation even under low oxygen conditions (3% O2). The proportion of nestin-positive NPCs decreased from 51.8±2.46% to 36.17±3.55% following the addition of MP and decreased more significantly to 27.20±2.68% in the cells cultured in 3% O2. In addition, a smaller number of glial fibrillary acidic protein (GFAP)-positive cells and a greater number of microtubule-associated protein 2 (MAP2)-positive cells was observed following the addition of MP under both normal (normoxic) and low oxygen (hypoxic) conditions. In response to MP treatment, hypoxia-inducible factor-1α (HIF-1α) and the Notch signaling pathway downstream protein, Hes1, but not the upstream Notch-1 intracelluar domain (NICD), were inhibited. After blocking NICD with a γ-secretase inhibitor (DAPT) MP still inhibited the expression of Hes1. Our results provide insight into the molecular mechanisms responsible for the MP-induced inhibition of proliferation and its effects on differentiation and suggest that HIF-1α and Hes1 play an important role in this effect.
Collapse
Affiliation(s)
- Wenhao Wang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Peng Wang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Shiyuan Li
- Foshan Hospital of Sun Yat-sen University, Foshan, Guangdong 528000, P.R. China
| | - Jiewen Yang
- Biotherapy Center, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong 510120, P.R. China
| | - Xinjun Liang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yong Tang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yuxi Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Rui Yang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Yanfeng Wu
- Biotherapy Center, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong 510120, P.R. China
| | - Huiyong Shen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
31
|
No changes in cerebellar microvessel length density in sudden infant death syndrome: implications for pathogenetic mechanisms. J Neuropathol Exp Neurol 2014; 73:312-23. [PMID: 24607967 DOI: 10.1097/nen.0000000000000055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sudden infant death syndrome (SIDS) is the leading cause of mortality in infants younger than 1 year in developed countries, but its primary cause remains unknown. Some studies suggest that there may be hypoxia in the cerebellum in SIDS subjects, but mean total Purkinje cell numbers in SIDS versus controls was recently found not to be different. Probably the best marker for chronic hypoxia in a brain region is the microvessel length per unit volume of tissue, that is, the microvessel length density (MLD). Here, we investigated MLDs using a rigorous design-based stereologic approach in all cell layers and white matter in postmortem cerebella from 9 SIDS cases who died between ages 2 and 10 months and from 14 control children, 9 of which were age- and sex- matched to the SIDS cases. We found no differences either in mean MLDs in the cerebellar layers between the SIDS cases and the controls or between controls with a low likelihood of hypoxia and those with a higher likelihood of hypoxia. Immunohistochemical detection of the astrocytosis marker glial fibrillary acidic protein showed no differences between the SIDS and the matched control cases. These data indicate that there is no association of chronic hypoxia in the cerebellum with SIDS.
Collapse
|
32
|
Zhang K, Zhao T, Huang X, Wu LY, Wu K, Zhu LL, Fan M. Notch1 mediates postnatal neurogenesis in hippocampus enhanced by intermittent hypoxia. Neurobiol Dis 2014; 64:66-78. [DOI: 10.1016/j.nbd.2013.12.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 11/23/2013] [Accepted: 12/12/2013] [Indexed: 12/17/2022] Open
|
33
|
Varela-Nallar L, Rojas-Abalos M, Abbott AC, Moya EA, Iturriaga R, Inestrosa NC. Chronic hypoxia induces the activation of the Wnt/β-catenin signaling pathway and stimulates hippocampal neurogenesis in wild-type and APPswe-PS1ΔE9 transgenic mice in vivo. Front Cell Neurosci 2014; 8:17. [PMID: 24574965 PMCID: PMC3918655 DOI: 10.3389/fncel.2014.00017] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 01/10/2014] [Indexed: 01/01/2023] Open
Abstract
Hypoxia modulates proliferation and differentiation of cultured embryonic and adult stem cells, an effect that includes β-catenin, a key component of the canonical Wnt signaling pathway. Here we studied the effect of mild hypoxia on the activity of the Wnt/β-catenin signaling pathway in the hippocampus of adult mice in vivo. The hypoxia-inducible transcription factor-1α (HIF-1α) was analyzed as a molecular control of the physiological hypoxic response. Exposure to chronic hypoxia (10% oxygen for 6–72 h) stimulated the activation of the Wnt/β-catenin signaling pathway. Because the Wnt/β-catenin pathway is a positive modulator of adult neurogenesis, we evaluated whether chronic hypoxia was able to stimulate neurogenesis in the subgranular zone (SGZ) of the hippocampal dentate gyrus. Results indicate that hypoxia increased cell proliferation and neurogenesis in adult wild-type mice as determined by Ki67 staining, Bromodeoxyuridine (BrdU) incorporation and double labeling with doublecortin (DCX). Chronic hypoxia also induced neurogenesis in a double transgenic APPswe-PS1ΔE9 mouse model of Alzheimer’s disease (AD), which shows decreased levels of neurogenesis in the SGZ. Our results show for the first time that exposure to hypoxia in vivo can induce the activation of the Wnt/β-catenin signaling cascade in the hippocampus, suggesting that mild hypoxia may have a therapeutic value in neurodegenerative disorders associated with altered Wnt signaling in the brain and also in pathological conditions in which hippocampal neurogenesis is impaired.
Collapse
Affiliation(s)
- Lorena Varela-Nallar
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile Santiago, Chile ; Centro de Investigaciones Biomédicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello Santiago, Chile
| | - Macarena Rojas-Abalos
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Ana C Abbott
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Esteban A Moya
- Laboratorio de Neurobiología, Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratorio de Neurobiología, Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
34
|
Baranov PY, Tucker BA, Young MJ. Low-oxygen culture conditions extend the multipotent properties of human retinal progenitor cells. Tissue Eng Part A 2014; 20:1465-75. [PMID: 24320879 DOI: 10.1089/ten.tea.2013.0361] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Development of an effective cell-based therapy is highly dependent upon having a reproducible cell source suitable for transplantation. One potential source, isolated from the developing fetal neural retina, is the human retinal progenitor cell (hRPC). One limiting factor for the use of hRPCs is their in vitro expansion limit. As such, the aim of this study was to determine whether culturing hRPCs under 3% O2 would support their proliferative capacity while maintaining multipotency. METHODS To determine the effect of low oxygen on the ability of hRPCs to self-renew, rates of proliferation and apoptosis, telomerase activity, and expression of proliferative, stemness, and differentiation markers were assessed for hRPCs cultured in 3% and 20% oxygen conditions. RESULTS Culture under 3% oxygen increases the proliferation rate and shifts the proliferation limit of hRPCs to greater 40 divisions. This increased capacity for proliferation is correlated with an upregulation of Ki67, CyclinD1, and telomerase activity and a decrease in p53 expression and apoptosis. Increased expression of cMyc, Klf4, Oct4, and Sox2 in 3% O₂ is correlated with stabilization of both HIF1α and HIF2α. The eye field development markers Pax6, Sox2, and Otx2 are present in hRPCs up to passage 16 in 3% O₂ . Following in vitro differentiation hRPCs expanded in the 3% O₂ were able to generate specialized retinal cells, including rods and cones. CONCLUSIONS Low-oxygen culture conditions act to maintain both multipotency and self-renewal properties of hRPCs in vitro. The extended expansion limits permit the development of a clinical-grade reagent for transplantation.
Collapse
Affiliation(s)
- Petr Y Baranov
- 1 The Schepens Eye Research Institute , Boston, Massachusetts
| | | | | |
Collapse
|
35
|
Dmitriev RI, Zhdanov AV, Nolan YM, Papkovsky DB. Imaging of neurosphere oxygenation with phosphorescent probes. Biomaterials 2013; 34:9307-17. [DOI: 10.1016/j.biomaterials.2013.08.065] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 08/21/2013] [Indexed: 02/04/2023]
|
36
|
Ye Z, Ye W, Deng Y, Wang J, Zhou G, Zhang X. HIF-1-modified BMSCs improve migration and reduce neuronal apoptosis after stroke in rats. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s11434-013-5936-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
37
|
Necdin controls proliferation and apoptosis of embryonic neural stem cells in an oxygen tension-dependent manner. J Neurosci 2013; 33:10362-73. [PMID: 23785149 DOI: 10.1523/jneurosci.5682-12.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neural stem cells (NSCs) reside in vivo in hypoxic environments, and NSC proliferation is enhanced in vitro under hypoxic conditions. Various adaptive responses to hypoxia are mediated by hypoxia-inducible factors (HIFs), a family of basic helix-loop-helix Per-Arnt-Sim (PAS) transcription factors. Necdin, a MAGE (melanoma antigen) family protein, is expressed abundantly in postmitotic neurons and possesses potent antimitotic and antiapoptotic activities. We here report that hypoxia induces degradation of the necdin protein in primary NSCs by HIF-mediated ubiquitin-proteasome system. Necdin was expressed in primary NSCs prepared from the ganglionic eminences of mouse embryos. Hypoxia enhanced neurosphere formation of NSCs, in which the necdin protein level was significantly reduced. Primary NSCs prepared from necdin-deficient mice exhibited higher rates of proliferation and apoptosis than those from wild-type mice in normoxia, whereas there were no significant differences in the proliferation and apoptosis rates between necdin-deficient and wild-type NSCs in hypoxia. HIF-2α was predominantly expressed in hypoxic NSCs, where expression of HIF-responsive genes was upregulated. HIF-2α interacted with necdin via its PAS domain, which enhanced necdin ubiquitination. Lentivirus-mediated expression of the PAS domain in primary NSCs promoted necdin degradation and enhanced NSC proliferation in normoxia, whereas a small-molecule inhibitor of HIF-2α translation stabilized the necdin protein and reduced NSC proliferation in hypoxia. These results suggest that oxygen tension regulates the necdin protein level in NSCs through HIF-2α-mediated proteasomal degradation to modulate their proliferation and apoptosis.
Collapse
|
38
|
Hermann A, Loewenbrück KF, Boldt Ä, Fiedler C, Maisel M, Kalucka J, Schwarz J, Breier G, Storch A. Lack of vascular endothelial growth factor receptor-2/Flk1 signaling does not affect substantia nigra development. Neurosci Lett 2013; 553:142-7. [PMID: 23994060 DOI: 10.1016/j.neulet.2013.08.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 07/29/2013] [Accepted: 08/11/2013] [Indexed: 01/09/2023]
Abstract
Oxygen tension is critical for proliferation of human and murine midbrain-derived neural precursor cells (mNPCs). Lack of hypoxia-inducible factor-1α (HIF1α) impairs midbrain dopaminergic neurogenesis which could be rescued by vascular endothelial growth factor (VEGF) via VEGFR-2 signaling. Here, we conditionally inactivated the VEGFR-2, encoded by the fetal liver kinase 1 (Flk1) gene, in murine NPCs to determine its role in proliferation and survival in vitro as well as survival of dopaminergic neurons in vivo. Flk1 conditional knock-out (Flk1 CKO) mice showed no general brain phenotype. There was no midbrain-specific impairment of NPC proliferation as seen in HIF1α CKO mice. In the substantia nigra (SN) of adult Flk1 CKO mice, nonbiased stereological cell counts revealed no reduction of TH-positive neurons of Flk1 CKO mice compared with control Cre/wt mice (in which the wild-type Flk1 allele is expressed in parallel with the Cre recombinase allele). In conclusion, VEGF receptor signaling seems not to be relevant to the development and survival of substantia nigra dopaminergic neurons within the hypoxia-HIF1α signaling pathway.
Collapse
Affiliation(s)
- Andreas Hermann
- Division of Neurodegenerative Diseases, Department of Neurology, Dresden University of Technology, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany; German Centre for Neurodegenerative Diseases (DZNE) Dresden, Dresden University of Technology, Dresden, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wang F, Xiong L, Huang X, Zhao T, Wu LY, Liu ZH, Ding X, Liu S, Wu Y, Zhao Y, Wu K, Zhu LL, Fan M. miR-210 suppresses BNIP3 to protect against the apoptosis of neural progenitor cells. Stem Cell Res 2013; 11:657-67. [PMID: 23688833 DOI: 10.1016/j.scr.2013.04.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 04/08/2013] [Accepted: 04/08/2013] [Indexed: 12/16/2022] Open
Abstract
MiR-210 is a hypoxia-inducible factor (HIF)-1 target gene and is the most consistently and predominantly upregulated miRNA in response to hypoxia in various cancer cell lines. Our recent study shows that hypoxia increased miR-210 expression in neural progenitor cells (NPCs) in a time-dependent manner. However, the role of miR-210 in NPCs remains unknown. Following the identification of the miR-210 putative target genes, we demonstrated that the Bcl-2 adenovirus E1B 19kDa-interacting protein 3 (BNIP3), which is regulated by HIF-1 and activates cell death, is regulated by miR-210 in NPCs under hypoxia. Moreover, the over-expression of miR-210 decreased apoptosis in NPCs, and the inhibition of miR-210 expression remarkably increased the number of TUNEL-positive NPCs by 30% in response to hypoxia. Importantly, miR-210 mimics reduced both BNIP3 protein expression and the translocation of AIF into the nucleus, which reduced cell death, whereas miR-210 inhibitors reversed this process, leading to cell death during hypoxia. Taken together, we report a novel feedback loop of BNIP3 regulation in NPCs under hypoxia. HIF-1 is activated under hypoxia and then induces the expression of both BNIP3 and miR-210. The upregulation of miR-210 then directly suppresses BNIP3 expression to maintain the survival of NPCs under hypoxia. This negative feedback regulation might partially contribute to protection against hypoxia-induced cell death via the inhibition of AIF nuclear translocation.
Collapse
Affiliation(s)
- Fei Wang
- Department of Cognitive Sciences, Institute of Basic Medical Sciences, Beijing 100850, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Xiong L, Wang F, Huang X, Liu ZH, Zhao T, Wu LY, Wu K, Ding X, Liu S, Wu Y, Zhao Y, Zhu LL, Fan M. DNA demethylation regulates the expression of miR-210 in neural progenitor cells subjected to hypoxia. FEBS J 2012; 279:4318-26. [DOI: 10.1111/febs.12021] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 08/15/2012] [Accepted: 10/03/2012] [Indexed: 12/18/2022]
Affiliation(s)
| | - Fei Wang
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Xin Huang
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Zhao-hui Liu
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Tong Zhao
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Li-ying Wu
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Kuiwu Wu
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Xuefeng Ding
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Shuhong Liu
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Yan Wu
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Yongqi Zhao
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Ling-ling Zhu
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| | - Ming Fan
- Department of Cognitive Sciences; Institute of Basic Medical Sciences; Beijing; China
| |
Collapse
|
41
|
Zhang S, Luo X, Wan F, Lei T. The roles of hypoxia-inducible factors in regulating neural stem cells migration to glioma stem cells and determinating their fates. Neurochem Res 2012; 37:2659-66. [PMID: 22991140 DOI: 10.1007/s11064-012-0879-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/07/2012] [Accepted: 08/29/2012] [Indexed: 02/07/2023]
Abstract
The mortality of patients with malignant gliomas remains high despite the advancement in multi-modal therapy including surgery, radio- and chemotherapy. Glioma stem cells (GSCs), sharing some characteristics with normal neural stem cells (NSCs), contribute to the cellular origin for primary gliomas and the recurrence of malignant gliomas after current conventional therapy. Accordingly, targeting GSCs proves to be a promising avenue of therapeutic intervention. The specific tropism of NSCs to GSCs provides a novel platform for targeted delivery of therapeutic agents. Tropism and mobilization of NSCs are enhanced by hypoxia through upregulating chemotactic cytokines and activating several signaling pathways. Moreover, hypoxia-inducible factors (HIFs) produced under hypoxic microenvironment of the stem cell niche play critical roles in the growth and stemness phenotypes regulation of both NSCs and GSCs. However, the definite cellular and molecular mechanisms of HIFs involvement in the process remain obscure. In this review, we focus on the pivotal roles of HIFs in migration of NSCs to GSCs and potential roles of HIFs in dictating the fates of migrated NSCs and targeted GSCs.
Collapse
Affiliation(s)
- Suojun Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095#, Wuhan 430030, People's Republic of China
| | | | | | | |
Collapse
|
42
|
Crossin KL. Oxygen levels and the regulation of cell adhesion in the nervous system: a control point for morphogenesis in development, disease and evolution? Cell Adh Migr 2012; 6:49-58. [PMID: 22647940 DOI: 10.4161/cam.19582] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In this article, I discuss the hallmarks of hypoxia in vitro and in vivo and review work showing that many types of stem cell proliferate more robustly in lowered oxygen. I then discuss recent studies showing that alterations in the levels and the types of cell and substrate adhesion molecules are a notable response to reduced O(2) levels in both cultured primary neural stem cells and brain tissues in response to hypoxia in vivo. The ability of O(2) levels to regulate adhesion molecule expression is linked to the Wnt signaling pathway, which can control and be controlled by adhesion events. The ability of O(2) levels to influence cell adhesion also has far-reaching implications for development, ischemic trauma and neural regeneration, as well as for cancer and other diseases. Finally I discuss the possibility that the fluctuations in O(2) levels known to have occurred over evolutionary time could, by influencing adhesion systems, have contributed to early symbiotic events in unicellular organisms and to the emergence of multicellularity. It is not my intention to be exhaustive in these domains, which are far from my own field of study. Rather this article is meant to provoke and stimulate thinking about molecular evolution involving O(2) sensing and signaling during eras of geologic and atmospheric change that might inform modern studies on development and disease.
Collapse
Affiliation(s)
- Kathryn L Crossin
- Department of Neurobiology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
43
|
Liang D, Ma Y, Liu J, Trope CG, Holm R, Nesland JM, Suo Z. The hypoxic microenvironment upgrades stem-like properties of ovarian cancer cells. BMC Cancer 2012; 12:201. [PMID: 22642602 PMCID: PMC3407800 DOI: 10.1186/1471-2407-12-201] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 05/29/2012] [Indexed: 12/26/2022] Open
Abstract
Background To study whether hypoxia influences the stem-like properties of ovarian cancer cells and their biological behavior under hypoxia. Method Ovarian cancer cell lines ES-2 and OVCAR-3 were cultivated in different oxygen tensions for proliferation, cell cycling and invasion analyses. The clonogenic potential of cells was examined by colony formation and sphere formation assays. Stem cell surface markers, SP and CD44bright and CD44dim cells were analyzed by flow cytometry. Protein expression of HIF-1α, HIF-2α, Ot3/4 and Sox2 were investigated by Western blotting. Results Both cell lines cultivated at hypoxic condition grew relatively slowly with extended G0/G1 phase. However, if the cells were pre-treated under 1% O2 for 48 hrs before brought back to normoxia, the cells showed significantly higher proliferation rate with higher infiltration capability, and significant more colonies and spheres, in comparison to the cells always cultivated under normoxia. CD44bright cells expressed significantly higher levels of Oct3/4 and Sox2 than the CD44dim cells and formed significantly more clones and spheres examined in vitro. Hypoxic treatment of the cells resulted in stronger CD44 expression in both cell lines, and stronger CD133 expression in the OVCAR-3 cell line. In parallel with these findings, significantly increased number of side population (SP) cells and up-regulated expression of Oct3/4 and Sox2 in both ES-2 and OVCAR-3 cell lines were observed. Conclusion We conclude that ovarian cancer cells survive hypoxia by upgrading their stem-like properties through up-regulation of stemness-related factors and behave more aggressively when brought back to higher oxygen environment.
Collapse
Affiliation(s)
- Dongming Liang
- Departments of Pathology, the Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Montebello, Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
44
|
Heddleston JM, Hitomi M, Venere M, Flavahan WA, Yang K, Kim Y, Minhas S, Rich JN, Hjelmeland AB. Glioma stem cell maintenance: the role of the microenvironment. Curr Pharm Des 2012; 17:2386-401. [PMID: 21827414 DOI: 10.2174/138161211797249260] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 07/25/2011] [Indexed: 12/23/2022]
Abstract
Glioblastomas are highly lethal cancers for which conventional therapies provide only palliation. The cellular heterogeneity of glioblastomas is manifest in genetic and epigenetic variation with both stochastic and hierarchical models informing cellular phenotypes. At the apex of the hierarchy is a self-renewing, tumorigenic, cancer stem cell (CSC). The significance of CSCs is underscored by their resistance to cytotoxic therapies, invasive potential, and promotion of angiogenesis. Thus, targeting CSCs may offer therapeutic benefit and sensitize tumors to conventional treatment, demanding elucidation of CSC regulation. Attention has been paid to intrinsic cellular systems in CSCs, but recognition of extrinsic factors is evolving. Glioma stem cells (GSCs) are enriched in functional niches--prominently the perivascular space and hypoxic regions. These niches provide instructive cues to maintain GSCs and induce cellular plasticity towards a stem-like phenotype. GSC-maintaining niches may therefore offer novel therapeutic targets but also signal additional complexity with perhaps different pools of GSCs governed by different molecular mechanisms that must be targeted for tumor control.
Collapse
Affiliation(s)
- John M Heddleston
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Serebrovskaya TV, Nikolsky IS, Nikolska VV, Mallet RT, Ishchuk VA. Intermittent hypoxia mobilizes hematopoietic progenitors and augments cellular and humoral elements of innate immunity in adult men. High Alt Med Biol 2012; 12:243-52. [PMID: 21962068 DOI: 10.1089/ham.2010.1086] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
This study tested the hypothesis that intermittent hypoxia treatment (IHT) modulates circulating hematopoietic stem and progenitor cells (HSPC) and augments humoral and cellular components of innate immunity in young, healthy men. Ten subjects (group 1: age 31±4 yr) were studied before and at 1 and 7 days after a 14-day IHT program consisting of four 5-min bouts/day of breathing 10% O2, lowering arterial O2 saturation to 84% to 85%, with intervening 5-min room-air exposures. Five more subjects (group 2: age 29±5 yr) were studied during 1 IHT session. Immunofluorescence detected HSPCs as CD45+CD34+ cells in peripheral blood. Phagocytic and bactericidal activities of neutrophils, circulating immunoglobulins (IgM, IgG, IgA), immune complexes, complement, and cytokines (erythropoietin, TNF-α, IL-4, IFN-γ) were measured. In group 1, the HSPC count fell 27% below pre-IHT baseline 1 week after completing IHT, without altering erythrocyte and reticulocyte counts. The IHT program also activated complement, increased circulating platelets, augmented phagocytic and bactericidal activities of neutrophils, sharply lowered circulating TNF-α and IL-4 by >90% and ∼75%, respectively, and increased IFN-γ, particularly 1 week after IHT. During acute IHT (group 2), HSPC increased by 51% after the second hypoxia bout and by 19% after the fourth bout, and total leukocyte, neutrophil, monocyte, and lymphocyte counts also increased; but these effects subsided by 30 min post-IHT. Collectively, these results demonstrate that IHT enhances innate immunity by mobilizing HSPC, activating neutrophils, and increasing circulating complement and immunoglobulins. These findings support the potential for eventual application of IHT for immunotherapy.
Collapse
|
46
|
Cunningham LA, Candelario K, Li L. Roles for HIF-1α in neural stem cell function and the regenerative response to stroke. Behav Brain Res 2011; 227:410-7. [PMID: 21871501 DOI: 10.1016/j.bbr.2011.08.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 07/29/2011] [Accepted: 08/01/2011] [Indexed: 12/20/2022]
Abstract
Stroke represents a leading cause of long-term disability worldwide, with few therapeutic options available for improving behavioral recovery. Identification of endogenous neural stem and progenitor cells (NSPCs) that are capable of promoting reparative responses following brain injury and stroke make these cells attractive therapeutic targets for stimulating cell replacement and neuronal plasticity. Interest in the mechanisms that support NSPC survival and replenishment of damaged cells within the ischemic brain has led to elucidation of new roles for hypoxia-inducible factor-1α (HIF-1α) in NSPC function. HIF-1α is a well-studied mediator of adaptive cellular responses to hypoxia through direct transcriptional regulation of cellular metabolism and angiogenesis. Recent evidence also indicates novel roles for HIF-1α in stem cell differentiation through modulation of Notch and Wnt/β-catenin signaling pathways. In this review, we will explore the hypothesis that HIF-1α represents an important mediator of NSPC function under both non-pathological conditions and stroke; and plays a central role in the regulation of NSPC response to hypoxia, metabolism and maintenance of the vascular environment of the neural stem cell niche.
Collapse
Affiliation(s)
- Lee Anna Cunningham
- Department of Neurosciences, MSC08 4740, 1 University of New Mexico Health Sciences Center, Albuquerque, NM 87131-0001, USA.
| | | | | |
Collapse
|
47
|
Zhu LL, Zhao T, huang X, Liu ZH, Wu LY, Wu KW, Fan M. Gene expression profiles and metabolic changes in embryonic neural progenitor cells under low oxygen. Cell Reprogram 2011; 13:113-20. [PMID: 21473688 DOI: 10.1089/cell.2010.0043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Hypoxia promotes the proliferation of neural progenitor cells (NPCs), and low oxygen is a useful tool for expansion of NPCs in vitro. To further understand the regulation of the mechanisms involved, we first identified the gene expression profile of NPCs and characterized their metabolic changes in vitro under 3% oxygen. NPCs derived from E13.5 rat mesencephalon were cultured under either normoxia or hypoxia for 24 h and 72 h. Total RNA was subjected to cDNA microarray analysis of 5705 genes. The results showed that approximately 1.24% of gene expression changed under low oxygen at the two time points. Among the 142 differentially expressed genes, the greatest number was involved in glycolysis and metabolism. The metabolic changes of NPCs under low oxygen conditions were also assayed. The glucose content in the conditioned medium incubated in low oxygen decreased significantly; however, the levels of pyruvate and lactic acid increased compared to conditioned medium cultured in normoxia. The NPCs under low oxygen consumed more glucose and produced energy by glycolysis. The information gained from gene expression and metabolic analyses of NPCs under low oxygen conditions will provide new approaches for the evaluation of NPCs as potential in vivo cellular therapeutics.
Collapse
Affiliation(s)
- Ling-Ling Zhu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, No. 27 Taiping Road, Beijing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
48
|
Efficient derivation of NPCs, spinal motor neurons and midbrain dopaminergic neurons from hESCs at 3% oxygen. Nat Protoc 2011; 6:1229-40. [PMID: 21799491 DOI: 10.1038/nprot.2011.380] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This protocol has been designed to generate neural precursor cells (NPCs) from human embryonic stem cells (hESCs) using a physiological oxygen (O(2)) level of 3% (previously termed hypoxia) and chemically defined conditions. The first stage involves suspension culture of hESC colonies at 3% O(2), where they acquire a neuroepithelial identity over a period of 2 weeks. This timescale is comparable to that observed at 20% O(2), but survival is enhanced. Sequential application of retinoic acid and purmorphamine (PM), from day 14 to day 28, directs differentiation toward spinal motor neurons. Alternatively, addition of fibroblast growth factor-8 and PM generates midbrain dopaminergic neurons. OLIG2 (encoding oligodendrocyte lineage transcription factor 2) induction in motor neuron precursors is twofold greater than that at 20% O(2), whereas EN1 (encoding engrailed homeobox 1) expression is enhanced fivefold. NPCs (at 3% O(2)) can be differentiated into all three neural lineages, and such cultures can be maintained long term in the absence of neurotrophins. The ability to generate defined cell types at 3% O(2) should represent a significant advancement for in vitro disease modeling and potentially for cell-based therapies.
Collapse
|
49
|
Proteomic analysis of mitochondrial proteins in cardiomyocytes from rats subjected to intermittent hypoxia. Eur J Appl Physiol 2011; 112:1037-46. [PMID: 21735218 DOI: 10.1007/s00421-011-2050-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 06/15/2011] [Indexed: 10/18/2022]
Abstract
Intermittent hypoxia (IH) markedly enhances cardiac tolerance against ischemia/reperfusion injury, but its mechanism and molecular basis remain unclear. For exploring the expression of mitochondrial proteins induced by IH, two-dimensional electrophoresis and Thermo Finnigan LTQ mass spectrometer (MS) were applied. After comparing the protein profiles of myocardial mitochondria between IH and normoxic hearts, 14 protein spots were found to be altered more than threefold between the two groups, 11 of which were identified by Finnigan LTQ MS. Among these 11 proteins, 9 were involved in energy metabolism, including 7 that were increased after IH. The latter were identified as aldehyde dehydrogenase, methylmalonate-semialdehyde dehydrogenase, ATP synthase β chain, mitochondrial aconitase, malate dehydrogenase, electron transfer flavoprotein α subunit and sirtuin 5. Two other proteins, ubiquinol-cytochrome C reductase iron-sulfur subunit and aspartate aminotransferase, were decreased after IH. Biochemical tests for energy metabolism in mitochondria supported the proteomic results. IH exposure also increased the expression of a molecular chaperone-heat shock protein 60 and an antioxidant protein, peroxiredoxin 5. These findings will provide clues for understanding the mechanism of IH-induced cardiac protection and may lead to the development of interventional strategies designed to utilize the advantages of IH clinically.
Collapse
|
50
|
Stacpoole SRL, Bilican B, Webber DJ, Luzhynskaya A, He XL, Compston A, Karadottir R, Franklin RJM, Chandran S. Derivation of neural precursor cells from human ES cells at 3% O(2) is efficient, enhances survival and presents no barrier to regional specification and functional differentiation. Cell Death Differ 2011; 18:1016-23. [PMID: 21274009 PMCID: PMC3091847 DOI: 10.1038/cdd.2010.171] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 11/05/2010] [Accepted: 11/25/2010] [Indexed: 12/21/2022] Open
Abstract
In vitro stem cell systems traditionally employ oxygen levels that are far removed from the in vivo situation. This study investigates whether an ambient environment containing a physiological oxygen level of 3% (normoxia) enables the generation of neural precursor cells (NPCs) from human embryonic stem cells (hESCs) and whether the resultant NPCs can undergo regional specification and functional maturation. We report robust and efficient neural conversion at 3% O(2), demonstration of tri-lineage potential of resultant NPCs and the subsequent electrophysiological maturation of neurons. We also show that NPCs derived under 3% O(2) can be differentiated long term in the absence of neurotrophins and can be readily specified into both spinal motor neurons and midbrain dopaminergic neurons. Finally, modelling the oxygen stress that occurs during transplantation, we demonstrate that in vitro transfer of NPCs from a 20 to 3% O(2) environment results in significant cell death, while maintenance in 3% O(2) is protective. Together these findings support 3% O(2) as a physiologically relevant system to study stem cell-derived neuronal differentiation and function as well as to model neuronal injury.
Collapse
Affiliation(s)
- S R L Stacpoole
- University of Cambridge, Department of Clinical Neurosciences, Cambridge, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|