1
|
Rezaei Adariani S, Agne D, Koska S, Burhop A, Seitz C, Warmers J, Janning P, Metz M, Pahl A, Sievers S, Waldmann H, Ziegler S. Detection of a Mitochondrial Fragmentation and Integrated Stress Response Using the Cell Painting Assay. J Med Chem 2024; 67:13252-13270. [PMID: 39018123 PMCID: PMC11320566 DOI: 10.1021/acs.jmedchem.4c01183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Mitochondria are cellular powerhouses and are crucial for cell function. However, they are vulnerable to internal and external perturbagens that may impair mitochondrial function and eventually lead to cell death. In particular, small molecules may impact mitochondrial function, and therefore, their influence on mitochondrial homeostasis is at best assessed early on in the characterization of biologically active small molecules and drug discovery. We demonstrate that unbiased morphological profiling by means of the cell painting assay (CPA) can detect mitochondrial stress coupled with the induction of an integrated stress response. This activity is common for compounds addressing different targets, is not shared by direct inhibitors of the electron transport chain, and enables prediction of mitochondrial stress induction for small molecules that are profiled using CPA.
Collapse
Affiliation(s)
- Soheila Rezaei Adariani
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
- Faculty
of Chemistry and Chemical Biology, Technical
University Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Daya Agne
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Sandra Koska
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Annina Burhop
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Carina Seitz
- Compound
Management and Screening Center, Max Planck
Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Jens Warmers
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
- Faculty
of Chemistry and Chemical Biology, Technical
University Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Petra Janning
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Malte Metz
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Axel Pahl
- Compound
Management and Screening Center, Max Planck
Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Sonja Sievers
- Compound
Management and Screening Center, Max Planck
Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Herbert Waldmann
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
- Faculty
of Chemistry and Chemical Biology, Technical
University Dortmund, Otto-Hahn-Strasse 6, 44227 Dortmund, Germany
| | - Slava Ziegler
- Department
of Chemical Biology, Max Planck Institute
of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| |
Collapse
|
2
|
Barata P, Camacho O, Lima CG, Pereira AC. The Role of Hyperbaric Oxygen Therapy in Neuroregeneration and Neuroprotection: A Review. Cureus 2024; 16:e62067. [PMID: 38989389 PMCID: PMC11235151 DOI: 10.7759/cureus.62067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Neurogenesis is a high energy-demanding process, which is why blood vessels are an active part of the neurogenic niche since they allow the much-needed oxygenation of progenitor cells. In this regard, although neglected for a long time, the "oxygen niche" should be considered an important intervenient in adult neurogenesis. One possible hypothesis for the failure of numerous neuroprotective trials is that they relied on compounds that target a highly specific neuroprotective pathway. This approach may be too limited, given the complexity of the processes that lead to cell death. Therefore, research should adopt a more multifactorial approach. Among the limited range of agents with multimodal neuromodulatory capabilities, hyperbaric oxygen therapy has demonstrated effectiveness in reducing secondary brain damage in various brain injury models. This therapy functions not only as a neuroprotective mechanism but also as a powerful neuroregenerative mechanism.
Collapse
Affiliation(s)
- Pedro Barata
- Pathology and Laboratory Medicine, Centro Hospitalar Universitário do Porto, Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| | - Oscar Camacho
- Hyperbaric Medicine Unit, Unidade Local de Saúde de Matosinhos, Matosinhos, PRT
| | - Clara G Lima
- Anesthesiology, Hospital Pedro Hispano, Matosinhos, PRT
| | - Ana Claudia Pereira
- Faculty of Health Sciences, Universidade Fernando Pessoa (UFP), Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| |
Collapse
|
3
|
Zhang T, Luo L, He Q, Xiao S, Li Y, Chen J, Qin T, Xiao Z, Ge Q. Research advances on molecular mechanism and natural product therapy of iron metabolism in heart failure. Eur J Med Res 2024; 29:253. [PMID: 38659000 PMCID: PMC11044586 DOI: 10.1186/s40001-024-01809-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
The progression of heart failure (HF) is complex and involves multiple regulatory pathways. Iron ions play a crucial supportive role as a cofactor for important proteins such as hemoglobin, myoglobin, oxidative respiratory chain, and DNA synthetase, in the myocardial energy metabolism process. In recent years, numerous studies have shown that HF is associated with iron dysmetabolism, and deficiencies in iron and overload of iron can both lead to the development of various myocarditis diseases, which ultimately progress to HF. Iron toxicity and iron metabolism may be key targets for the diagnosis, treatment, and prevention of HF. Some iron chelators (such as desferrioxamine), antioxidants (such as ascorbate), Fer-1, and molecules that regulate iron levels (such as lactoferrin) have been shown to be effective in treating HF and protecting the myocardium in multiple studies. Additionally, certain natural compounds can play a significant role by mediating the imbalance of iron-related signaling pathways and expression levels. Therefore, this review not only summarizes the basic processes of iron metabolism in the body and the mechanisms by which they play a role in HF, with the aim of providing new clues and considerations for the treatment of HF, but also summarizes recent studies on natural chemical components that involve ferroptosis and its role in HF pathology, as well as the mechanisms by which naturally occurring products regulate ferroptosis in HF, with the aim of providing reference information for the development of new ferroptosis inhibitors and lead compounds for the treatment of HF in the future.
Collapse
Affiliation(s)
- Tianqing Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Li Luo
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Qi He
- People's Hospital of Ningxiang City, Ningxiang City, China
| | - Sijie Xiao
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Yuwei Li
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Junpeng Chen
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Tao Qin
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Zhenni Xiao
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China
| | - Qingliang Ge
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University, Hunan, China.
| |
Collapse
|
4
|
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
|
5
|
Sarath Kumar K, Kritika S, Karthikeyan NS, Sujatha V, Mahalaxmi S, Ravichandran C. Development of cobalt-incorporated chitosan scaffold for regenerative potential in human dental pulp stem cells: An in vitro study. Int J Biol Macromol 2023; 253:126574. [PMID: 37648130 DOI: 10.1016/j.ijbiomac.2023.126574] [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: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
The aim of the study was to comparatively evaluate chitosan and Cobalt incorporated chitosan (CoCH) scaffold at varying concentrations in terms of their material characteristics, cytotoxicity and cell adhesion potential. In the present study, cobalt incorporated chitosan scaffolds at varying concentrations were prepared and dried. The synthesised scaffolds were characterised using XRD, FTIR, SEM-EDX and BET which revealed amorphous, porous surface of CoCH scaffolds and FTIR analysis showed the complexation confirming the chelation of cobalt with chitosan. The experimental scaffolds proved to be non-cytotoxic when compared to chitosan scaffolds on XTT analysis. Cell-seeding assay revealed enhanced adherence of hDPSCs to CoCH scaffold at 1:1 ratio in the concentration of 100 mL of 100 μmol/L cobalt chloride solution in 100mL of 2% chitosan solution, when compared to other groups. The results highlighted that 100 μmol/L concentration of cobalt chloride when incorporated in 1:1 ratio into 2 % CH solution yields a promising porous, biocompatible scaffold with enhanced cellular adhesion for dentin-pulp regeneration.
Collapse
Affiliation(s)
- K Sarath Kumar
- Department of Conservative Dentistry and Endodontics, SRM Dental College, Ramapuram, SRM Institute of Science & Technology, Ramapuram Campus, Bharathi Salai, Ramapuram, Chennai 600 089, Tamil Nadu, India
| | - Selvakumar Kritika
- Department of Conservative Dentistry and Endodontics, SRM Dental College, Ramapuram, SRM Institute of Science & Technology, Ramapuram Campus, Bharathi Salai, Ramapuram, Chennai 600 089, Tamil Nadu, India
| | | | - Venkatappan Sujatha
- Department of Conservative Dentistry and Endodontics, SRM Dental College, Ramapuram, SRM Institute of Science & Technology, Ramapuram Campus, Bharathi Salai, Ramapuram, Chennai 600 089, Tamil Nadu, India.
| | - Sekar Mahalaxmi
- Department of Conservative Dentistry and Endodontics, SRM Dental College, Ramapuram, SRM Institute of Science & Technology, Ramapuram Campus, Bharathi Salai, Ramapuram, Chennai 600 089, Tamil Nadu, India
| | - Cingaram Ravichandran
- Department of Chemistry, Easwari Engineering College, Bharathi Salai, Ramapuram, Chennai 600 089, Tamil Nadu, India
| |
Collapse
|
6
|
Schlegel C, Liu K, Spring B, Dietz S, Poets CF, Hudalla H, Lajqi T, Köstlin-Gille N, Gille C. Decreased expression of hypoxia-inducible factor 1α (HIF-1α) in cord blood monocytes under anoxia. Pediatr Res 2023; 93:870-877. [PMID: 35906309 PMCID: PMC10033401 DOI: 10.1038/s41390-022-02193-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/24/2022] [Accepted: 06/28/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Infections are a major cause for morbidity and mortality in neonates; however, the underling mechanisms for increased infection susceptibility are incompletely understood. Hypoxia, which is present in inflamed tissues, has been identified as an important activation signal for innate immune cells in adults and is mainly mediated by hypoxia-inducible factor 1α (HIF-1α). Fetal tissue pO2 physiologically is low but rises immediately after birth. METHODS In this study, the effect of low oxygen partial pressure (pO2) on HIF-1α expression and its targets phagocytosis, reactive oxygen species (ROS) production and vascular endothelial growth factor (VEGF) secretion was compared in vitro between immune cells from adult peripheral blood and cord blood using anoxia, HIF-1α stabilizer desferroxamin (DFO) and E. coli as stimuli. RESULTS We show that anoxia-induced HIF-1α protein accumulation, phagocytosis, ROS-production and VEGF-expression were greatly diminished in cord blood compared to adult cells. E. coli led to HIF-1α gene expression in adult and cord blood immune cells; however, cord blood cells failed to accumulate HIF-1α protein and VEGF upon E. coli stimulation. CONCLUSIONS Taken together, our results show a diminished activation of cord blood immune cells by low pO2, which might contribute to impaired reactivity in the context of infection. IMPACT Neonatal immune cells do not accumulate HIF-1α under low oxygen partial pressure leading to decreased phagocytosis and decreased ROS production. We demonstrate a previously unknown mechanism of reduced activation of neonatal immune cells in the context of an inflammatory response. This could contribute to the increased susceptibility of newborns and preterm infants to infection.
Collapse
Affiliation(s)
- Christiane Schlegel
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Kai Liu
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Bärbel Spring
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Stefanie Dietz
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Christian F Poets
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Hannes Hudalla
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany.
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany.
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| |
Collapse
|
7
|
Hadanny A, Rittblat M, Bitterman M, May-Raz I, Suzin G, Boussi-Gross R, Zemel Y, Bechor Y, Catalogna M, Efrati S. Hyperbaric oxygen therapy improves neurocognitive functions of post-stroke patients - a retrospective analysis. Restor Neurol Neurosci 2021; 38:93-107. [PMID: 31985478 PMCID: PMC7081098 DOI: 10.3233/rnn-190959] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Previous studies have shown that hyperbaric oxygen therapy (HBOT) can improve the motor functions and memory of post-stroke patients in the chronic stage. Objective: The aim of this study is to evaluate the effects of HBOT on overall cognitive functions of post-stroke patients in the chronic stage. The nature, type and location of the stroke were investigated as possible modifiers. Methods: A retrospective analysis was conducted on patients who were treated with HBOT for chronic stroke (>3 months) between 2008-2018. Participants were treated in a multi-place hyperbaric chamber with the following protocols: 40 to 60 daily sessions, 5 days per week, each session included 90 min of 100% oxygen at 2 ATA with 5 min air brakes every 20 minutes. Clinically significant improvements (CSI) were defined as > 0.5 standard deviation (SD). Results: The study included 162 patients (75.3% males) with a mean age of 60.75±12.91. Of them, 77(47.53%) had cortical strokes, 87(53.7%) strokes were located in the left hemisphere and 121 suffered ischemic strokes (74.6%). HBOT induced a significant increase in all the cognitive function domains (p < 0.05), with 86% of the stroke victims achieving CSI. There were no significant differences post-HBOT of cortical strokes compared to sub-cortical strokes (p > 0.05). Hemorrhagic strokes had a significantly higher improvement in information processing speed post-HBOT (p < 0.05). Left hemisphere strokes had a higher increase in the motor domain (p < 0.05). In all cognitive domains, the baseline cognitive function was a significant predictor of CSI (p < 0.05), while stroke type, location and side were not significant predictors. Conclusions: HBOT induces significant improvements in all cognitive domains even in the late chronic stage. The selection of post-stroke patients for HBOT should be based on functional analysis and baseline cognitive scores rather than the stroke type, location or side of lesion.
Collapse
Affiliation(s)
- Amir Hadanny
- Neurosurgery Department, Galilee Medical Center, Naharyia, Israel.,Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel.,Galilee Faculty of Medicine, Bar Ilan University, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Mor Rittblat
- Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Mor Bitterman
- Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Ido May-Raz
- Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Gil Suzin
- Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Rahav Boussi-Gross
- Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Yonatan Zemel
- Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Yair Bechor
- Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Merav Catalogna
- Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Shai Efrati
- Sagol Center for Hyperbaric Medicine and Research, Assaf Harofeh Medical Center, Zerifin, Israel.,Research and Development Unit, Assaf Harofeh Medical Center, Zerifin, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
8
|
DeFrates KG, Franco D, Heber-Katz E, Messersmith PB. Unlocking mammalian regeneration through hypoxia inducible factor one alpha signaling. Biomaterials 2021; 269:120646. [PMID: 33493769 PMCID: PMC8279430 DOI: 10.1016/j.biomaterials.2020.120646] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/19/2020] [Accepted: 12/29/2020] [Indexed: 02/08/2023]
Abstract
Historically, the field of regenerative medicine has aimed to heal damaged tissue through the use of biomaterials scaffolds or delivery of foreign progenitor cells. Despite 30 years of research, however, translation and commercialization of these techniques has been limited. To enable mammalian regeneration, a more practical approach may instead be to develop therapies that evoke endogenous processes reminiscent of those seen in innate regenerators. Recently, investigations into tadpole tail regrowth, zebrafish limb restoration, and the super-healing Murphy Roths Large (MRL) mouse strain, have identified ancient oxygen-sensing pathways as a possible target to achieve this goal. Specifically, upregulation of the transcription factor, hypoxia-inducible factor one alpha (HIF-1α) has been shown to modulate cell metabolism and plasticity, as well as inflammation and tissue remodeling, possibly priming injuries for regeneration. Since HIF-1α signaling is conserved across species, environmental or pharmacological manipulation of oxygen-dependent pathways may elicit a regenerative response in non-healing mammals. In this review, we will explore the emerging role of HIF-1α in mammalian healing and regeneration, as well as attempts to modulate protein stability through hyperbaric oxygen treatment, intermittent hypoxia therapy, and pharmacological targeting. We believe that these therapies could breathe new life into the field of regenerative medicine.
Collapse
Affiliation(s)
- Kelsey G DeFrates
- Department of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, USA.
| | - Daniela Franco
- Department of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, USA.
| | - Ellen Heber-Katz
- Laboratory of Regenerative Medicine, Lankenau Institute for Medical Research, Wynnewood, PA, USA.
| | - Phillip B Messersmith
- Department of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, USA; Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
9
|
Resveratrol promotes the survival and neuronal differentiation of hypoxia-conditioned neuronal progenitor cells in rats with cerebral ischemia. Front Med 2020; 15:472-485. [PMID: 33263836 DOI: 10.1007/s11684-021-0832-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
Hypoxia conditioning could increase the survival of transplanted neuronal progenitor cells (NPCs) in rats with cerebral ischemia but could also hinder neuronal differentiation partly by suppressing mitochondrial metabolism. In this work, the mitochondrial metabolism of hypoxia-conditioned NPCs (hcNPCs) was upregulated via the additional administration of resveratrol, an herbal compound, to resolve the limitation of hypoxia conditioning on neuronal differentiation. Resveratrol was first applied during the in vitro neuronal differentiation of hcNPCs and concurrently promoted the differentiation, synaptogenesis, and functional development of neurons derived from hcNPCs and restored the mitochondrial metabolism. Furthermore, this herbal compound was used as an adjuvant during hcNPC transplantation in a photothrombotic stroke rat model. Resveratrol promoted neuronal differentiation and increased the long-term survival of transplanted hcNPCs. 18-fluorine fluorodeoxyglucose positron emission tomography and rotarod test showed that resveratrol and hcNPC transplantation synergistically improved the neurological and metabolic recovery of stroke rats. In conclusion, resveratrol promoted the neuronal differentiation and therapeutic efficiency of hcNPCs in stroke rats via restoring mitochondrial metabolism. This work suggested a novel approach to promote the clinical translation of NPC transplantation therapy.
Collapse
|
10
|
Maitra S, Khandelwal N, Kootar S, Sant P, Pathak SS, Reddy S, K. AP, Murty US, Chakravarty S, Kumar A. Histone Lysine Demethylase JMJD2D/KDM4D and Family Members Mediate Effects of Chronic Social Defeat Stress on Mouse Hippocampal Neurogenesis and Mood Disorders. Brain Sci 2020; 10:brainsci10110833. [PMID: 33182385 PMCID: PMC7695311 DOI: 10.3390/brainsci10110833] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022] Open
Abstract
Depression, anxiety and related mood disorders are major psychiatric illnesses worldwide, and chronic stress appears to be one of the primary underlying causes. Therapeutics to treat these debilitating disorders without a relapse are limited due to the incomplete molecular understanding of their etiopathology. In addition to the well-studied genetic component, research in the past two decades has implicated diverse epigenetic mechanisms in mediating the negative effects of chronic stressful events on neural circuits. This includes the cognitive circuitry, where the dynamic hippocampal dentate gyrus (DG) neurogenesis gets affected in depression and related affective disorders. Most of these epigenetic studies have focused on the impact of acetylation/deacetylation and methylation of several histone lysine residues on neural gene expression. However, there is a dearth of investigation into the role of demethylation of these lysine residues in chronic stress-induced changes in neurogenesis that results in altered behaviour. Here, using the chronic social defeat stress (CSDS) paradigm to induce depression and anxiety in C57BL/6 mice and ex vivo DG neural stem/progenitor cell (NSCs/NPCs) culture we show the role of the members of the JMJD2/KDM4 family of histone lysine demethylases (KDMs) in mediating stress-induced changes in DG neurogenesis and mood disorders. The study suggests a critical role of JMJD2D in DG neurogenesis. Altered enrichment of JMJD2D on the promoters of Id2 (inhibitor of differentiation 2) and Sox2 (SRY-Box Transcription Factor 2) was observed during proliferation and differentiation of NSCs/NPCs obtained from the DG. This would affect the demethylation of repressive epigenetic mark H3K9, thus activating or repressing these and possibly other genes involved in regulating proliferation and differentiation of DG NSCs/NPCs. Treatment of the NSCs/NPCs culture with Dimethyloxallyl Glycine (DMOG), an inhibitor of JMJDs, led to attenuation in their proliferation capacity. Additionally, systemic administration of DMOG in mice for 10 days induced depression-like and anxiety-like phenotype without any stress exposure.
Collapse
Affiliation(s)
- Swati Maitra
- Applied Biology, CSIR—Indian Institute of Chemical Technology (IICT), Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India; (S.M.); (U.S.M.)
| | - Nitin Khandelwal
- Epigenetics & Neuropsychiatric Disorders Laboratory, CSIR—Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Habsiguda, Hyderabad 500007, Telangana, India; (N.K.); (S.K.); (P.S.); (S.S.P.); (S.R.); (A.P.K.)
| | - Scherazad Kootar
- Epigenetics & Neuropsychiatric Disorders Laboratory, CSIR—Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Habsiguda, Hyderabad 500007, Telangana, India; (N.K.); (S.K.); (P.S.); (S.S.P.); (S.R.); (A.P.K.)
| | - Pooja Sant
- Epigenetics & Neuropsychiatric Disorders Laboratory, CSIR—Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Habsiguda, Hyderabad 500007, Telangana, India; (N.K.); (S.K.); (P.S.); (S.S.P.); (S.R.); (A.P.K.)
| | - Salil S. Pathak
- Epigenetics & Neuropsychiatric Disorders Laboratory, CSIR—Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Habsiguda, Hyderabad 500007, Telangana, India; (N.K.); (S.K.); (P.S.); (S.S.P.); (S.R.); (A.P.K.)
| | - Sujatha Reddy
- Epigenetics & Neuropsychiatric Disorders Laboratory, CSIR—Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Habsiguda, Hyderabad 500007, Telangana, India; (N.K.); (S.K.); (P.S.); (S.S.P.); (S.R.); (A.P.K.)
| | - Annapoorna P. K.
- Epigenetics & Neuropsychiatric Disorders Laboratory, CSIR—Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Habsiguda, Hyderabad 500007, Telangana, India; (N.K.); (S.K.); (P.S.); (S.S.P.); (S.R.); (A.P.K.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Upadhyayula Suryanarayana Murty
- Applied Biology, CSIR—Indian Institute of Chemical Technology (IICT), Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India; (S.M.); (U.S.M.)
- National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, Assam, India
| | - Sumana Chakravarty
- Applied Biology, CSIR—Indian Institute of Chemical Technology (IICT), Uppal Road, Tarnaka, Hyderabad 500007, Telangana, India; (S.M.); (U.S.M.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
- Correspondence: (S.C.); (A.K.)
| | - Arvind Kumar
- Epigenetics & Neuropsychiatric Disorders Laboratory, CSIR—Centre for Cellular and Molecular Biology (CCMB), Uppal Road, Habsiguda, Hyderabad 500007, Telangana, India; (N.K.); (S.K.); (P.S.); (S.S.P.); (S.R.); (A.P.K.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
- Correspondence: (S.C.); (A.K.)
| |
Collapse
|
11
|
Chang HC, Yang YR, Wang RY. Effects of repetitive hyperbaric oxygen therapy on neuroprotection in middle cerebral artery occlusion rats. Brain Res 2020; 1748:147097. [PMID: 32896522 DOI: 10.1016/j.brainres.2020.147097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023]
Abstract
Hyperbaric oxygen (HBO) has been suggested as a possible therapy for brain injury. However, the effects of HBO after transient brain ischemia are inconsistent and the underlying mechanisms are not fully known. The present study aimed to investigate the effects of repetitive HBO intervention in a transient middle cerebral artery occlusion (MCAO) animal model. Seventy-two Sprague-Dawley rats received MCAO and were randomly assigned to normal air control or HBO intervention groups. Each group was divided into 3 subgroups according to the intervention time period (7, 14, and 21 days). HBO was started 24 h post-MCAO for 1 h/day at 3.0 ATA with no-air breaks. After the final intervention, half of the rats in each subgroup were sacrificed and the right motor cortex was removed to examine levels of Akt phosphorylation and glutathione (GSH), as well as glutathione peroxidase (GPx) and reductase (GR) activity. The other half of the rats were used to examine infarct volume. At 24 h post-MCAO and the end of the final intervention, rats underwent tests to examine motor performance. We noted that 14- and 21-day HBO interventions significantly reduced infarct volume and increased Akt phosphorylation and GSH levels and GPx and GR activity. Motor performance was also significantly improved after 14- and 21-day interventions. No significant differences were observed between the controls and 7-day intervention groups. Repetitive HBO intervention starting 24 h post-MCAO and applied for at least 14 days, provided neuroprotective effects through modulating the cell survival pathway and antioxidative defense system.
Collapse
Affiliation(s)
- Heng-Chih Chang
- Department of Physical Therapy, Asia University, Taichung, Taiwan, ROC
| | - Yea-Ru Yang
- Department of Physical Therapy and Assistive Technology, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Ray-Yau Wang
- Department of Physical Therapy and Assistive Technology, National Yang-Ming University, Taipei, Taiwan, ROC.
| |
Collapse
|
12
|
Cozene B, Sadanandan N, Gonzales-Portillo B, Saft M, Cho J, Park YJ, Borlongan CV. An Extra Breath of Fresh Air: Hyperbaric Oxygenation as a Stroke Therapeutic. Biomolecules 2020; 10:E1279. [PMID: 32899709 PMCID: PMC7563917 DOI: 10.3390/biom10091279] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 12/15/2022] Open
Abstract
Stroke serves as a life-threatening disease and continues to face many challenges in the development of safe and effective therapeutic options. The use of hyperbaric oxygen therapy (HBOT) demonstrates pre-clinical effectiveness for the treatment of acute ischemic stroke and reports reductions in oxidative stress, inflammation, and neural apoptosis. These pathophysiological benefits contribute to improved functional recovery. Current pre-clinical and clinical studies are testing the applications of HBOT for stroke neuroprotection, including its use as a preconditioning regimen. Mild oxidative stress may be able to prime the brain to tolerate full extensive oxidative stress that occurs during a stroke, and HBOT preconditioning has displayed efficacy in establishing such ischemic tolerance. In this review, evidence on the use of HBOT following an ischemic stroke is examined, and the potential for HBOT preconditioning as a neuroprotective strategy. Additionally, HBOT as a stem cell preconditioning is also discussed as a promising strategy, thus maximizing the use of HBOT for ischemic stroke.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (B.C.); (N.S.); (B.G.-P.); (M.S.); (J.C.); (Y.J.P.)
| |
Collapse
|
13
|
Cheng YJ, Liu CC, Chu FY, Yang CP, Hsiao CW, Chuang CW, Shiau MY, Lee HT, Tsai JN, Chang YH. Oxygenated Water Inhibits Adipogenesis and Attenuates Hepatic Steatosis in High-Fat Diet-Induced Obese Mice. Int J Mol Sci 2020; 21:ijms21155493. [PMID: 32752112 PMCID: PMC7432369 DOI: 10.3390/ijms21155493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 11/16/2022] Open
Abstract
The expansion of adipose tissue mass is the primary characteristic of the process of becoming obesity, which causes chronic adipose inflammation and is closely associated with type 2 diabetes mellitus (T2DM). Adipocyte hypertrophy restricts oxygen availability, leading to microenvironmental hypoxia and adipose dysfunction. This study aimed at investigating the effects of oxygenated water (OW) on adipocyte differentiation (adipogenesis) and the metabolic function of mature adipocytes. The effects of OW on adipogenesis and the metabolic function of mature adipocytes were examined. Meanwhile, the in vivo metabolic effects of long-term OW consumption on diet-induced obesity (DIO) mice were investigated. OW inhibited adipogenesis and lipid accumulation through down-regulating critical adipogenic transcription factors and lipogenic enzymes. While body weight, blood and adipose parameters were not significantly improved by long-term OW consumption, transient circulatory triglyceride-lowering and glucose tolerance-improving effects were identified. Notably, hepatic lipid contents were significantly reduced, indicating that the DIO-induced hepatic steatosis was attenuated, despite no improvements in fibrosis and lipid contents in adipose tissue being observed in the OW-drinking DIO mice. The study provides evidence regarding OW’s effects on adipogenesis and mature adipocytes, and the corresponding molecular mechanisms. OW exhibits transient triglyceride-lowering and glucose tolerance-improving activity as well as hepatic steatosis-attenuating functions.
Collapse
Affiliation(s)
- Yuh-Jen Cheng
- Research Center for Applied Sciences, Academia Sinica, Taipei 115, Taiwan;
| | - Chao-Chi Liu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.L.); (C.-P.Y.); (C.-W.C.)
| | - Fang-Yeh Chu
- Department of Clinical Pathology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu 300, Taiwan
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei 110, Taiwan
| | - Ching-Ping Yang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.L.); (C.-P.Y.); (C.-W.C.)
| | - Chiao-Wan Hsiao
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei 112, Taiwan;
| | - Cheng-Wei Chuang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.L.); (C.-P.Y.); (C.-W.C.)
| | - Ming-Yuh Shiau
- Department of Nursing, College of Nursing, Hungkuang University, Taichung 433, Taiwan;
| | - Hsueh-Te Lee
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan;
| | - Jen-Ning Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan;
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Yih-Hsin Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan; (C.-C.L.); (C.-P.Y.); (C.-W.C.)
- Correspondence: ; Tel.: +886-2-2826-7955; Fax: 886-2-2821-9240
| |
Collapse
|
14
|
Chounchay S, Noctor SC, Chutabhakdikul N. Microglia enhances proliferation of neural progenitor cells in an in vitro model of hypoxic-ischemic injury. EXCLI JOURNAL 2020; 19:950-961. [PMID: 32788909 PMCID: PMC7415932 DOI: 10.17179/excli2020-2249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022]
Abstract
Microglial cells are the primary immune cells in the central nervous system. In the mature brain, microglia perform functions that include eliminating pathogens and clearing dead/dying cells and cellular debris through phagocytosis. In the immature brain, microglia perform functions that include synapse development and the regulation of cell production through extensive contact with and phagocytosis of neural progenitor cells (NPCs). However, the functional role of microglia in the proliferation and differentiation of NPCs under hypoxic-ischemic (HI) injury is not clear. Here, we tested the hypothesis that microglia enhance NPCs proliferation following HI insult. Primary NPCs cultures were divided into four treatment groups: 1) normoxic NPCs (NN); 2) normoxic NPCs cocultured with microglia (NN+M); 3) hypoxic NPCs (HN); and 4) hypoxic NPCs cocultured with microglia (HN+M). Hypoxic-ischemic injury was induced by pretreatment of the cell cultures with 100 µM deferoxamine mesylate (DFO). NPCs treated with 100 µM DFO (HN groups) for 24 hours had significantly increased expression of hypoxia-inducible factor 1 alpha (HIF-1α), a marker of hypoxic cells. Cell number, protein expression, mitosis, and cell cycle phase were examined, and the data were compared between the four groups. We found that the number of cells expressing the NPCs marker Sox2 increased significantly in the HN+M group and that the number of PH3-positive cells increased in the HN+M group; flow cytometry analysis showed a significant increase in the percentage of cells in the G2/M phase in the HN+M group. In summary, these results support the concept that microglia enhance the survival of NPCs under HI injury by increasing NPCs proliferation, survival, and differentiation. These results further suggest that microglia may induce neuroprotective effects after hypoxic injury that can be explored to develop novel therapeutic strategies for the treatment of HI injury in the immature brain.
Collapse
Affiliation(s)
- Supanee Chounchay
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom, 73170, Thailand.,Faculty of Physical Therapy, Huachiew Chalermprakiet University, Samut Prakan, 10540, Thailand
| | - Stephen C Noctor
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, 95817, USA.,MIND Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Nuanchan Chutabhakdikul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom, 73170, Thailand
| |
Collapse
|
15
|
Hadanny A, Efrati S. The Hyperoxic-Hypoxic Paradox. Biomolecules 2020; 10:biom10060958. [PMID: 32630465 PMCID: PMC7355982 DOI: 10.3390/biom10060958] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Effective metabolism is highly dependent on a narrow therapeutic range of oxygen. Accordingly, low levels of oxygen, or hypoxia, are one of the most powerful inducers of gene expression, metabolic changes, and regenerative processes, including angiogenesis and stimulation of stem cell proliferation, migration, and differentiation. The sensing of decreased oxygen levels (hypoxia) or increased oxygen levels (hyperoxia), occurs through specialized chemoreceptor cells and metabolic changes at the cellular level, which regulate the response. Interestingly, fluctuations in the free oxygen concentration rather than the absolute level of oxygen can be interpreted at the cellular level as a lack of oxygen. Thus, repeated intermittent hyperoxia can induce many of the mediators and cellular mechanisms that are usually induced during hypoxia. This is called the hyperoxic-hypoxic paradox (HHP). This article reviews oxygen physiology, the main cellular processes triggered by hypoxia, and the cascade of events triggered by the HHP.
Collapse
Affiliation(s)
- Amir Hadanny
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin 70300, Israel;
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan 5290002, Israel
- Correspondence: ; Tel.: +972-544707381; Fax: +972-8-9779748
| | - Shai Efrati
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin 70300, Israel;
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
- The Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 6997801, Israel
| |
Collapse
|
16
|
Hadanny A, Efrati S. The Hyperoxic-Hypoxic Paradox. Biomolecules 2020; 10:biom10060958. [PMID: 32630465 DOI: 10.3390/biom1006095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 05/21/2023] Open
Abstract
Effective metabolism is highly dependent on a narrow therapeutic range of oxygen. Accordingly, low levels of oxygen, or hypoxia, are one of the most powerful inducers of gene expression, metabolic changes, and regenerative processes, including angiogenesis and stimulation of stem cell proliferation, migration, and differentiation. The sensing of decreased oxygen levels (hypoxia) or increased oxygen levels (hyperoxia), occurs through specialized chemoreceptor cells and metabolic changes at the cellular level, which regulate the response. Interestingly, fluctuations in the free oxygen concentration rather than the absolute level of oxygen can be interpreted at the cellular level as a lack of oxygen. Thus, repeated intermittent hyperoxia can induce many of the mediators and cellular mechanisms that are usually induced during hypoxia. This is called the hyperoxic-hypoxic paradox (HHP). This article reviews oxygen physiology, the main cellular processes triggered by hypoxia, and the cascade of events triggered by the HHP.
Collapse
Affiliation(s)
- Amir Hadanny
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin 70300, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Shai Efrati
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin 70300, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
- The Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 6997801, Israel
| |
Collapse
|
17
|
Sheik Abdul N, Nagiah S, Chuturgoon AA. The neglected foodborne mycotoxin Fusaric acid induces bioenergetic adaptations by switching energy metabolism from mitochondrial processes to glycolysis in a human liver (HepG2) cell line. Toxicol Lett 2020; 318:74-85. [DOI: 10.1016/j.toxlet.2019.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/03/2019] [Accepted: 10/17/2019] [Indexed: 12/21/2022]
|
18
|
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: 17] [Impact Index Per Article: 2.4] [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
|
19
|
Hey SM, Jensen P, Ryding M, Martínez Serrano A, Kristensen BW, Meyer M. Nonhypoxic pharmacological stabilization of Hypoxia Inducible Factor 1α: Effects on dopaminergic differentiation of human neural stem cells. Eur J Neurosci 2018; 49:497-509. [PMID: 30471165 DOI: 10.1111/ejn.14284] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/13/2018] [Accepted: 11/07/2018] [Indexed: 01/01/2023]
Abstract
Parkinson's disease is a neurodegenerative disease resulting in degeneration of midbrain dopaminergic neurons. Exploratory studies using human foetal tissue or predifferentiated stem cells have suggested that intracerebral transplantation of dopaminergic precursor cells may become an effective treatment for patients with Parkinson's disease. However, strategies for dopaminergic stem cell differentiation vary widely in efficiency, and better methods still need to be developed. Hypoxia Inducible Factor 1 (HIF-1) is a transcription factor involved in the regulation of genes important for cellular adaption to hypoxia and low glucose supply. HIF-1 is to a large degree regulated by the availability of oxygen as in its presence, the subunit HIF-1α is degraded by HIF prolyl hydroxylase enzymes (HPHs). Stabilization of HIF-1α through inhibition of HPHs has been shown to increase dopaminergic differentiation of stem cells and to protect dopaminergic neurons against neurotoxins. This study investigated the effects of noncompetitive (FG-0041) and competitive (Compound A and JNJ-42041935) HIF-1α stabilizing compounds on the dopaminergic differentiation of human neural stem cells. Treatment with all HPH inhibitors at high oxygen tension (20%) resulted in HIF-1α stabilization as assessed by immunocytochemistry for HIF-1α and detection of increased levels of vascular endothelial growth factor in the conditioned culture medium. Following 10 days of HIF-1α stabilization, the cultures displayed a slightly reduced proliferative activity and significantly increased relative levels of tyrosine hydroxylase-positive dopaminergic neurons. In conclusion, HIF-1α stabilization may represent a promising strategy for the generation of dopaminergic neurons intended for use in experimental in vitro studies and cell replacement therapies.
Collapse
Affiliation(s)
- Sabine Morris Hey
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Pia Jensen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Matias Ryding
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Alberto Martínez Serrano
- Department of Molecular Biology and Center of Molecular Biology Severo Ochoa, Autonomous University of Madrid-C.S.I.C Campus Cantoblanco, Madrid, Spain
| | - Bjarne W Kristensen
- Department of Pathology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
20
|
Krueger K, Catanese L, Sciesielski LK, Kirschner KM, Scholz H. Deletion of an intronic HIF-2α binding site suppresses hypoxia-induced WT1 expression. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:71-83. [PMID: 30468780 DOI: 10.1016/j.bbagrm.2018.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/02/2018] [Accepted: 11/18/2018] [Indexed: 12/20/2022]
Abstract
Hypoxia-inducible factors (HIFs) play a key role in the adaptation to low oxygen by interacting with hypoxia response elements (HREs) in the genome. Cellular levels of the HIF-2α transcription factor subunit influence the histopathology and clinical outcome of neuroblastoma, a malignant childhood tumor of the sympathetic ganglia. Expression of the Wilms tumor gene, WT1, marks a group of high-risk neuroblastoma. Here, we identify WT1 as a downstream target of HIF-2α in Kelly neuroblastoma cells. In chromatin immunoprecipitation assays, HIF-2α bound to a HRE in intron 3 of the WT1 gene, but not to another predicted HIF binding site (HBS) in the first intron. The identified element conferred oxygen sensitivity to otherwise hypoxia-resistant WT1 and SV40 promoter constructs. Deletion of the HBS in the intronic HRE by genome editing abolished WT1 expression in hypoxic neuroblastoma cells. Physical interaction between the HRE and the WT1 promoter in normoxic and hypoxic Kelly cells was shown by chromosome conformation capture assays. These findings demonstrate that binding of HIF-2α to an oxygen-sensitive enhancer in intron 3 stimulates transcription of the WT1 gene in neuroblastoma cells by hypoxia-independent chromatin looping. This novel regulatory mechanism may have implications for the biology and prognosis of neuroblastoma.
Collapse
Affiliation(s)
- Katharina Krueger
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Lorenzo Catanese
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Lina K Sciesielski
- Klinik für Neonatologie, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Karin M Kirschner
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Holger Scholz
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
21
|
Ferreira AC, Sousa N, Bessa JM, Sousa JC, Marques F. Metabolism and adult neurogenesis: Towards an understanding of the role of lipocalin-2 and iron-related oxidative stress. Neurosci Biobehav Rev 2018; 95:73-84. [PMID: 30267731 DOI: 10.1016/j.neubiorev.2018.09.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/20/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
The process of generating new functional neurons in the adult mammalian brain occurs from the local neural stem and progenitor cells and requires tight control of the progenitor cell's activity. Several signaling pathways and intrinsic/extrinsic factors have been well studied over the last years, but recent attention has been given to the critical role of cellular metabolism in determining the functional properties of progenitor cells. Here, we review recent advances in the current understanding of when and how metabolism affects neural stem cell (NSC) behavior and subsequent neuronal differentiation and highlight the role of lipocalin-2 (LCN2), a protein involved in the control of oxidative stress, as a recently emerged regulator of NSC activity and neuronal differentiation.
Collapse
Affiliation(s)
- Ana Catarina Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João M Bessa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João Carlos Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
22
|
DiGuilio KM, Valenzano MC, Rybakovsky E, Mullin JM. Cobalt chloride compromises transepithelial barrier properties of CaCo-2 BBe human gastrointestinal epithelial cell layers. BMC Gastroenterol 2018; 18:2. [PMID: 29304733 PMCID: PMC5756372 DOI: 10.1186/s12876-017-0731-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 12/14/2017] [Indexed: 12/28/2022] Open
Abstract
Background Elevation of the transcription factor HIF-1 is a prominent mediator of not only processes that accompany hypoxia, but also the tumor microenvironment and tissue regeneration. This study uses mediators of “chemical hypoxia” to ask the question whether HIF-1α elevation in a healthy epithelial cell layer leads to leakiness in its tight junctional seals. Methods Transepithelial electrical resistance and transepithelial diffusion of 14C–D-mannitol and other radiolabeled probes are used as indicators of transepithelial barrier function of CaCo-2 BBe human gastrointestinal epithelial cell layers cultured on permeable supports. Western immunoblot analyses of integral tight junctional proteins (occludin and claudins) are used as further indicators of barrier function change. Results Cobalt, an inhibitor of the prolyl hydroxylase enzymes governing HIF-1α breakdown in the cell, induces transepithelial leakiness in CaCo-2 BBe cell layers in a time and concentration-dependent manner. This increased leakiness is accompanied by significant changes in certain specific integral tight junctional (TJ) proteins such as a decreased level of occludin and increased level of claudin-5. Similar results regarding barrier function compromise also occur with other chemical inhibitors of HIF-1α breakdown, namely ciclopiroxolamine (CPX) and dimethyloxalylglycine (DMOG). The increased leak is manifested by both decreased transepithelial electrical resistance (Rt) and increased paracellular diffusion of D-mannitol (Jm). The induced transepithelial leak shows significant size selectivity, consistent with induced effects on TJ permeability. Less-differentiated cell layers were significantly more affected than well-differentiated cell layers regarding induced transepithelial leak. A genetically modified CaCo-2 variant with reduced levels of HIF-1β, showed reduced transepithelial leak in response to cobalt exposure, further indicating that elevation of HIF-1α levels induced by agents of “chemical hypoxia” is responsible for the compromised barrier function of the CaCo-2 BBe cell layers. Conclusions Exposure to inducers of chemical hypoxia elevated HIF-1α levels and increased transepithelial leak. The degree of epithelial differentiation has significant effects on this action, possibly explaining the varying effects of HIF-1 modulation in epithelial and endothelial barrier function in different physiological and pathophysiological conditions. Electronic supplementary material The online version of this article (doi: 10.1186/s12876-017-0731-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- K M DiGuilio
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA.,Present Address: Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA, 19131, USA
| | - M C Valenzano
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA
| | - E Rybakovsky
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA
| | - J M Mullin
- Lankenau Institute for Medical Research, 100 Lancaster Avenue, Wynnewood, PA, 19096, USA. .,Division of Gastroenterology, Lankenau Medical Center, Wynnewood, PA, 19096, USA.
| |
Collapse
|
23
|
Xie Y, Lowry WE. Manipulation of neural progenitor fate through the oxygen sensing pathway. Methods 2017; 133:44-53. [PMID: 28864353 DOI: 10.1016/j.ymeth.2017.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/17/2017] [Accepted: 08/24/2017] [Indexed: 12/15/2022] Open
Abstract
Neural progenitor cells hold significant promise in a variety of clinical settings. While both the brain and spinal cord harbor endogenous neural progenitor or stem cells, they typically are not capable of repopulating neural populations in case of injury or degenerative disease. In vitro systems for the culture of neural progenitors has come a long ways due to advances in the method development. Recently, many groups have shown that manipulation of the oxygen-sensing pathway leading to activation of hypoxia inducible factors (HIFs) that can influence the proliferation, differentiation or maturation of neural progenitors. Moreover, different oxygen concentrations appear to affect lineage specification of neural progenitors upon their differentiation in vitro. Here we summarize some of these studies in an attempt to direct effort towards implementation of best methods to advance the use of neural progenitors from basic development towards clinical application.
Collapse
Affiliation(s)
- Yuan Xie
- Department of Biochemistry and Molecular Biology, University of Chicago, United States
| | - William E Lowry
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, United States; The Molecular Biology Institute, UCLA, United States; The Jonsson Comprehensive Cancer Center, UCLA, United States; Department of Dermatology, David Geffen School of Medicine, UCLA, United States.
| |
Collapse
|
24
|
Laksana K, Sooampon S, Pavasant P, Sriarj W. Cobalt Chloride Enhances the Stemness of Human Dental Pulp Cells. J Endod 2017; 43:760-765. [PMID: 28343926 DOI: 10.1016/j.joen.2017.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 12/16/2016] [Accepted: 01/04/2017] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Hypoxia is a factor in controlling stem cell stemness. We investigated if cobalt chloride (CoCl2), a chemical agent that mimics hypoxia in vitro, affected human dental pulp cell (hDPC) stemness by examining cell proliferation, stem cell marker expression, and osteogenic differentiation. METHODS hDPCs were cultured with or without 25 or 50 μmol/L CoCl2. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was used to determine cell proliferation. The number of STRO-1+ cells was determined by flow cytometry. The messenger RNA expression of the stem cell markers REX1, OCT4, SOX2, and NANOG and the osteogenic-associated genes ALP, COLI, and RUNX2 were evaluated using reverse transcription polymerase chain reaction or real-time polymerase chain reaction. Osteogenic differentiation was assessed by alkaline phosphatase (ALP) activity and mineralization assays. RESULTS Although 25 and 50 μmol/L CoCl2 suppressed hDPC proliferation, 50 μmol/L CoCl2 increased the number of STRO-1+ cells. Moreover, CoCl2 dose dependently induced stem cell marker expression. Additionally, CoCl2 treatment suppressed osteogenic-associated gene expression, ALP activity, and calcium deposition. The addition of apigenin, a hypoxia-inducible factor 1-alpha inhibitor, reversed the inhibitory effect of CoCl2 on ALP activity. CONCLUSIONS This study indicated that CoCl2 may enhance hDPC stemness.
Collapse
Affiliation(s)
- Kantaporn Laksana
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Sireerat Sooampon
- Department of Pharmacology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Wannakorn Sriarj
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
25
|
A. Ahern M, 1 Department of Physics, University of Colorado Denver, Denver, CO 80212, USA, P. Black C, J. Seedorf G, D. Baker C, P. Shepherd D, 2 Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora CO 80045, USA, † Current address: Department of Physiology, School of Medicine, University of Arizona, Tucson, USA. Hyperoxia impairs pro-angiogenic RNA production in preterm endothelial colony-forming cells. AIMS BIOPHYSICS 2017. [DOI: 10.3934/biophy.2017.2.284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
26
|
Moon J, Schwarz SC, Lee H, Kang JM, Lee Y, Kim B, Sung M, Höglinger G, Wegner F, Kim JS, Chung H, Chang SW, Cha KY, Kim K, Schwarz J. Preclinical Analysis of Fetal Human Mesencephalic Neural Progenitor Cell Lines: Characterization and Safety In Vitro and In Vivo. Stem Cells Transl Med 2016; 6:576-588. [PMID: 28191758 PMCID: PMC5442800 DOI: 10.5966/sctm.2015-0228] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 05/16/2016] [Indexed: 12/21/2022] Open
Abstract
We have developed a good manufacturing practice for long‐term cultivation of fetal human midbrain‐derived neural progenitor cells. The generation of human dopaminergic neurons may serve as a tool of either restorative cell therapies or cellular models, particularly as a reference for phenotyping region‐specific human neural stem cell lines such as human embryonic stem cells and human inducible pluripotent stem cells. We cultivated 3 different midbrain neural progenitor lines at 10, 12, and 14 weeks of gestation for more than a year and characterized them in great detail, as well as in comparison with Lund mesencephalic cells. The whole cultivation process of tissue preparation, cultivation, and cryopreservation was developed using strict serum‐free conditions and standardized operating protocols under clean‐room conditions. Long‐term‐cultivated midbrain‐derived neural progenitor cells retained stemness, midbrain fate specificity, and floorplate markers. The potential to differentiate into authentic A9‐specific dopaminergic neurons was markedly elevated after prolonged expansion, resulting in large quantities of functional dopaminergic neurons without genetic modification. In restorative cell therapeutic approaches, midbrain‐derived neural progenitor cells reversed impaired motor function in rodents, survived well, and did not exhibit tumor formation in immunodeficient nude mice in the short or long term (8 and 30 weeks, respectively). We conclude that midbrain‐derived neural progenitor cells are a promising source for human dopaminergic neurons and suitable for long‐term expansion under good manufacturing practice, thus opening the avenue for restorative clinical applications or robust cellular models such as high‐content or high‐throughput screening. Stem Cells Translational Medicine2017;6:576–588
Collapse
Affiliation(s)
- Jisook Moon
- Department of Biotechnology, College of Life Science, CHA University, Seongnam‐si, Gyeonggi‐do, Korea
- General Research Division, Korea Research‐Driven Hospital, Bundang CHA Medical Center, CHA University, Seongnam‐si, Gyeonggi‐do, Korea
| | - Sigrid C. Schwarz
- German Center for Neurodegenerative Diseases, Technical University Munich, Munich, Germany
| | - Hyun‐Seob Lee
- General Research Division, Korea Research‐Driven Hospital, Bundang CHA Medical Center, CHA University, Seongnam‐si, Gyeonggi‐do, Korea
| | - Jun Mo Kang
- General Research Division, Korea Research‐Driven Hospital, Bundang CHA Medical Center, CHA University, Seongnam‐si, Gyeonggi‐do, Korea
| | - Young‐Eun Lee
- General Research Division, Korea Research‐Driven Hospital, Bundang CHA Medical Center, CHA University, Seongnam‐si, Gyeonggi‐do, Korea
| | - Bona Kim
- Development Division, CHA Biotech, Seongnam‐si, Gyeonggi‐do, Korea
| | - Mi‐Young Sung
- Development Division, CHA Biotech, Seongnam‐si, Gyeonggi‐do, Korea
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases, Technical University Munich, Munich, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Jin Su Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hyung‐Min Chung
- Department of Stem Cell Biology, Graduate School of Medicine, Konkuk University, Gwangjin‐gu, Seoul, Korea
| | - Sung Woon Chang
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam‐si, Gyeonggi‐do, Korea
| | - Kwang Yul Cha
- General Research Division, Korea Research‐Driven Hospital, Bundang CHA Medical Center, CHA University, Seongnam‐si, Gyeonggi‐do, Korea
| | - Kwang‐Soo Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, Massachusetts, USA
| | - Johannes Schwarz
- German Center for Neurodegenerative Diseases, Technical University Munich, Munich, Germany
| |
Collapse
|
27
|
Hartman-Petrycka M, Knefel G, Lebiedowska A, Kosmala J, Klimacka-Nawrot E, Kawecki M, Nowak M, Błońska-Fajfrowska B. Alterations in taste perception as a result of hyperbaric oxygen therapy. Appetite 2016; 107:159-165. [PMID: 27497834 DOI: 10.1016/j.appet.2016.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 06/29/2016] [Accepted: 08/02/2016] [Indexed: 12/22/2022]
Abstract
The present study evaluates the effect of hyperbaric oxygen therapy on taste sensitivity, hedonic perception of taste, and food preferences. The studied groups included 197 people in total (79 in the study group; 118 in the control group). All patients from the study group were treated with hyperbaric oxygen therapy due to chronic non-healing wounds. The control group consisted of healthy people, who did not receive hyperbaric oxygen therapy. The taste intensity, recognition thresholds, and hedonic perception were examined using gustatory tests. The aqueous solutions of sucrose for sweet, sodium chloride for salty, citric acid for sour, quinine hydrochloride for bitter, and monosodium glutamate for umami taste were used. The participants fulfilled the questionnaire to examine pleasure derived from eating certain types of dishes. Gustatory tests and analyses of the pleasure derived from eating in the study group were carried out before the first exposure to hyperbaric oxygen and then at the end of therapy, after at least 25 sessions of treatment. In the control group, examination of perception of taste sensations was conducted only once. The results of comparing patients with non-healing wounds with healthy people are characterized by reduced taste sensitivity. After participation in hyperbaric oxygen therapy, the improvement in perception of taste sensations and changes in hedonic evaluation have occurred among patients with non-healing wounds. In terms of food preference, a decreased desire for eating sweet desserts, chocolate, and crisps was observed in those patients who received hyperbaric oxygen therapy.
Collapse
Affiliation(s)
- Magdalena Hartman-Petrycka
- Department of Basic Biomedical Sciences, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, 3, Kasztanowa Street, 41-205 Sosnowiec, Poland.
| | - Grzegorz Knefel
- Dr. Stanisław Sakiel Centre for Burn Treatment, Siemianowice Śląskie, 2, Jana Pawła II Street, 41-100 Siemianowice Śląskie, Poland.
| | - Agata Lebiedowska
- Department of Basic Biomedical Sciences, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, 3, Kasztanowa Street, 41-205 Sosnowiec, Poland.
| | - Joanna Kosmala
- Dr. Stanisław Sakiel Centre for Burn Treatment, Siemianowice Śląskie, 2, Jana Pawła II Street, 41-100 Siemianowice Śląskie, Poland.
| | - Ewa Klimacka-Nawrot
- Department of Basic Biomedical Sciences, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, 3, Kasztanowa Street, 41-205 Sosnowiec, Poland.
| | - Marek Kawecki
- Dr. Stanisław Sakiel Centre for Burn Treatment, Siemianowice Śląskie, 2, Jana Pawła II Street, 41-100 Siemianowice Śląskie, Poland.
| | - Mariusz Nowak
- Dr. Stanisław Sakiel Centre for Burn Treatment, Siemianowice Śląskie, 2, Jana Pawła II Street, 41-100 Siemianowice Śląskie, Poland.
| | - Barbara Błońska-Fajfrowska
- Department of Basic Biomedical Sciences, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, 3, Kasztanowa Street, 41-205 Sosnowiec, Poland.
| |
Collapse
|
28
|
The stabilization of hypoxia inducible factor modulates differentiation status and inhibits the proliferation of mouse embryonic stem cells. Chem Biol Interact 2016; 244:204-14. [DOI: 10.1016/j.cbi.2015.12.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/26/2015] [Accepted: 12/17/2015] [Indexed: 01/16/2023]
|
29
|
Hypoxia-inducible factor-1α upregulates tyrosine hydroxylase and dopamine transporter by nuclear receptor ERRγ in SH-SY5Y cells. Neuroreport 2015; 26:380-6. [PMID: 25807177 DOI: 10.1097/wnr.0000000000000356] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor relevant to the development of many mammalian organs including the brain. However, the molecular mechanisms by which signaling events mediate neuronal differentiation have not been fully elucidated. In the present study, we show for the first time that the orphan nuclear receptor estrogen-related receptor γ (ERRγ) is upregulated by HIF-1α and plays essential roles in HIF-1α-induced upregulation of dopaminergic marker molecules such as tyrosine hydroxylase and dopamine transporter. We found that deferoxamine upregulated HIF-1α and enhanced the dopaminergic phenotype and neurite outgrowth of SH-SY5Y cells. Deferoxamine activated transcription and protein expression of ERRγ, and deferoxamine-induced upregulation of tyrosine hydroxylase and dopamine transporter was attenuated by using the ERRγ inverse agonist or silencing ERRγ. Altogether, these results suggest that HIF-1α can positively regulate the dopaminergic phenotype through ERRγ. This study could provide new perspectives for understanding the mechanisms underlying the promotion of dopaminergic neuronal differentiation by hypoxia.
Collapse
|
30
|
Current Neurogenic and Neuroprotective Strategies to Prevent and Treat Neurodegenerative and Neuropsychiatric Disorders. Neuromolecular Med 2015; 17:404-22. [PMID: 26374113 DOI: 10.1007/s12017-015-8369-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/22/2015] [Indexed: 12/31/2022]
Abstract
The adult central nervous system is commonly known to have a very limited regenerative capacity. The presence of functional stem cells in the brain can therefore be seen as a paradox, since in other organs these are known to counterbalance cell loss derived from pathological conditions. This fact has therefore raised the possibility to stimulate neural stem cell differentiation and proliferation or survival by either stem cell replacement therapy or direct administration of neurotrophic factors or other proneurogenic molecules, which in turn has also originated regenerative medicine for the treatment of otherwise incurable neurodegenerative and neuropsychiatric disorders that take a huge toll on society. This may be facilitated by the fact that many of these disorders converge on similar pathophysiological pathways: excitotoxicity, oxidative stress, neuroinflammation, mitochondrial failure, excessive intracellular calcium and apoptosis. This review will therefore focus on the most promising achievements in promoting neuroprotection and neuroregeneration reported to date.
Collapse
|
31
|
Esfahani M, Karimi F, Afshar S, Niknazar S, Sohrabi S, Najafi R. Prolyl hydroxylase inhibitors act as agents to enhance the efficiency of cell therapy. Expert Opin Biol Ther 2015; 15:1739-55. [PMID: 26325448 DOI: 10.1517/14712598.2015.1084281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION In stem cell-based therapy as a subtype of regenerative medicine, stem cells can be used to replace or repair injured tissue and cells in order to treat disease. Stem cells have the ability to integrate into injured areas and produce new cells via processes of proliferation and differentiation. Several studies have demonstrated that hypoxia increases self-renewal, proliferation and post-homing differentiation of stem cells through the regulation of hypoxia-inducible factor-1 (HIF-1)-mediated gene expression. Thus, pharmacological interventions including prolyl hydroxylase (PHD) inhibitors are considered as promising solutions for stem cell-based therapy. PHD inhibitors stabilize the HIF-1 and activate its pathway through preventing proteasomal degradation of HIF-1. AREAS COVERED This review focuses on the role of hypoxia, HIF-1 and especially PHD inhibitors on cell therapy. PHD structure and function are discussed as well as their inhibitors. In addition, we have investigated several preclinical studies in which PHD inhibitors improved the efficiency of cell-based therapies. EXPERT OPINION The data reviewed here suggest that PHD inhibitors are effective operators in improving stem cell therapy. However, because of some limitations, these compounds should be properly examined before clinical application.
Collapse
Affiliation(s)
- Maryam Esfahani
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Fatemeh Karimi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Saeid Afshar
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Somayeh Niknazar
- b 2 Shahid Beheshti University of Medical Science, Hearing Disorders Research Center , Tehran, the Islamic Republic of Iran
| | - Sareh Sohrabi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| | - Rezvan Najafi
- a 1 Research center for molecular medicine, Hamadan University of Medical Sciences , Hamadan, the Islamic Republic of Iran
| |
Collapse
|
32
|
Piersanti S, Burla R, Licursi V, Brito C, La Torre M, Alves PM, Simao D, Mottini C, Salinas S, Negri R, Tagliafico E, Kremer EJ, Saggio I. Transcriptional Response of Human Neurospheres to Helper-Dependent CAV-2 Vectors Involves the Modulation of DNA Damage Response, Microtubule and Centromere Gene Groups. PLoS One 2015. [PMID: 26207738 PMCID: PMC4514711 DOI: 10.1371/journal.pone.0133607] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Brain gene transfer using viral vectors will likely become a therapeutic option for several disorders. Helper-dependent (HD) canine adenovirus type 2 vectors (CAV-2) are well suited for this goal. These vectors are poorly immunogenic, efficiently transduce neurons, are retrogradely transported to afferent structures in the brain and lead to long-term transgene expression. CAV-2 vectors are being exploited to unravel behavior, cognition, neural networks, axonal transport and therapy for orphan diseases. With the goal of better understanding and characterizing HD-CAV-2 for brain therapy, we analyzed the transcriptomic modulation induced by HD-CAV-2 in human differentiated neurospheres derived from midbrain progenitors. This 3D model system mimics several aspects of the dynamic nature of human brain. We found that differentiated neurospheres are readily transduced by HD-CAV-2 and that transduction generates two main transcriptional responses: a DNA damage response and alteration of centromeric and microtubule probes. Future investigations on the biochemistry of processes highlighted by probe modulations will help defining the implication of HD-CAV-2 and CAR receptor binding in enchaining these functional pathways. We suggest here that the modulation of DNA damage genes is related to viral DNA, while the alteration of centromeric and microtubule probes is possibly enchained by the interaction of the HD-CAV-2 fibre with CAR.
Collapse
Affiliation(s)
- Stefania Piersanti
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Romina Burla
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Valerio Licursi
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Rome, Italy
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780–901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780–157, Oeiras, Portugal
| | - Mattia La Torre
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780–901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780–157, Oeiras, Portugal
| | - Daniel Simao
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780–901, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780–157, Oeiras, Portugal
| | - Carla Mottini
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Sara Salinas
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Rodolfo Negri
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Rome, Italy
| | - Enrico Tagliafico
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Isabella Saggio
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci Bolognetti Foundation, Rome, Italy
- Institute of Molecular Biology and Pathology, CNR, Rome, Italy
- * E-mail:
| |
Collapse
|
33
|
Sakthiswary R, Raymond AA. Stem cell therapy in neurodegenerative diseases: From principles to practice. Neural Regen Res 2015; 7:1822-31. [PMID: 25624807 PMCID: PMC4302533 DOI: 10.3969/j.issn.1673-5374.2012.23.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 06/13/2012] [Indexed: 12/11/2022] Open
Abstract
The lack of curative therapies for neurodegenerative diseases has high economic impact and places huge burden on the society. The contribution of stem cells to cure neurodegenerative diseases has been unraveled and explored extensively over the past few years. Beyond substitution of the lost neurons, stem cells act as immunomodulators and neuroprotectors. A large number of preclinical and a small number of clinical studies have shown beneficial outcomes in this context. In this review, we have summarized the current concepts of stem cell therapy in neurodegenerative diseases and the recent advances in this field, particularly between 2010 and 2012. Further studies should be encouraged to resolve the clinical issues and vague translational findings for maximum optimization of the efficacy of stem cell therapy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Rajalingham Sakthiswary
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak 56000, Kuala Lumpur, Malaysia
| | - Azman Ali Raymond
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Bandar Tun Razak 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
34
|
Cobalt chloride supplementation induces stem-cell marker expression and inhibits osteoblastic differentiation in human periodontal ligament cells. Arch Oral Biol 2015; 60:29-36. [DOI: 10.1016/j.archoralbio.2014.08.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 07/31/2014] [Accepted: 08/30/2014] [Indexed: 12/15/2022]
|
35
|
Liu Y, Ma T. Metabolic regulation of mesenchymal stem cell in expansion and therapeutic application. Biotechnol Prog 2014; 31:468-81. [PMID: 25504836 DOI: 10.1002/btpr.2034] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/28/2014] [Indexed: 12/13/2022]
Abstract
Human mesenchymal or stromal cells (hMSCs) isolated from various adult tissues are primary candidates in cell therapy and tissue regeneration. Despite promising results in preclinical studies, robust therapeutic responses to MSC treatment have not been reproducibly demonstrated in clinical trials. In the translation of MSC-based therapy to clinical application, studies of MSC metabolism have significant implication in optimizing bioprocessing conditions to obtain therapeutically competent hMSC population for clinical application. In addition, understanding the contribution of metabolic cues in directing hMSC fate also provides avenues to potentiate their therapeutic effects by modulating their metabolic properties. This review focuses on MSC metabolism and discusses their unique metabolic features in the context of common metabolic properties shared by stem cells. Recent advances in the fundamental understanding of MSC metabolic characteristics in relation to their in vivo origin and metabolic regulation during proliferation, lineage-specific differentiation, and exposure to in vivo ischemic conditions are summarized. Metabolic strategies in directing MSC fate to enhance their therapeutic potential in tissue engineering and regenerative medicine are discussed.
Collapse
Affiliation(s)
- Yijun Liu
- Dept. of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL, 32310
| | | |
Collapse
|
36
|
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: 26] [Impact Index Per Article: 2.4] [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
|
37
|
Kim DY, Rhee I, Paik J. Metabolic circuits in neural stem cells. Cell Mol Life Sci 2014; 71:4221-41. [PMID: 25037158 DOI: 10.1007/s00018-014-1686-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/25/2014] [Accepted: 07/14/2014] [Indexed: 12/15/2022]
Abstract
Metabolic activity indicative of cellular demand is emerging as a key player in cell fate decision. Numerous studies have demonstrated that diverse metabolic pathways have a critical role in the control of the proliferation, differentiation and quiescence of stem cells. The identification of neural stem/progenitor cells (NSPCs) and the characterization of their development and fate decision process have provided insight into the regenerative potential of the adult brain. As a result, the potential of NSPCs in cell replacement therapies for neurological diseases is rapidly growing. The aim of this review is to discuss the recent findings on the crosstalk among key regulators of NSPC development and the metabolic regulation crucial for the function and cell fate decisions of NSPCs. Fundamental understanding of the metabolic circuits in NSPCs may help to provide novel approaches for reactivating neurogenesis to treat degenerative brain conditions and cognitive decline.
Collapse
Affiliation(s)
- Do-Yeon Kim
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, NY, 10065, USA
| | | | | |
Collapse
|
38
|
Skardelly M, Hempel E, Hirrlinger J, Wegner F, Meixensberger J, Milosevic J. Fluorescent protein-expressing neural progenitor cells as a tool for transplantation studies. PLoS One 2014; 9:e99819. [PMID: 24932758 PMCID: PMC4059690 DOI: 10.1371/journal.pone.0099819] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 05/14/2014] [Indexed: 01/17/2023] Open
Abstract
The purpose of this study was to generate quadruple fluorescent protein (QFP) transgenic mice as a source for QFP-expressing neural stem and progenitor cells (NSCs/NPCs) that could be utilized as a tool for transplantation research. When undifferentiated, these NSCs only express cyan fluorescent protein (CFP); however, upon neuronal differentiation, the cells express yellow fluorescent protein (YFP). During astrocytic differentiation, the cells express green fluorescent protein (GFP), and during oligodendrocytic differentiation, the cells express red fluorescent protein (DsRed). Using immunocytochemistry, immunoblotting, flow cytometry and electrophysiology, quadruple transgenic NPCs (Q-NPCs) and GFP-sorted NPCs were comprehensively characterized in vitro. Overall, the various transgenes did not significantly affect proliferation and differentiation of transgenic NPCs in comparison to wild-type NPCs. In contrast to a strong CFP and GFP expression in vitro, NPCs did not express YFP and dsRed either during proliferation or after differentiation in vitro. GFP-positive sorted NPCs, expressing GFP under the control of the human GFAP promoter, demonstrated a significant improvement in astroglial differentiation in comparison to GFP-negative sorted NPCs. In contrast to non-sorted and GFP-positive sorted NPCs, GFP-negative sorted NPCs demonstrated a high proportion of neuronal differentiation and proved to be functional in vitro. At 6 weeks after the intracerebroventricular transplantation of Q-NPCs into neonatal wild-type mice, CFP/DCX (doublecortin) double-positive transplanted cells were observed. The Q-NPCs did not express any other fluorescent proteins and did not mature into neuronal or glial cells. Although this model failed to visualize NPC differentiation in vivo, we determined that activation of the NPC glial fibrillary acid protein (GFAP) promoter, as indicated by GFP expression, can be used to separate neuronal and glial progenitors as a valuable tool for transplantation studies.
Collapse
Affiliation(s)
- Marco Skardelly
- Translational Centre for Regenerative Medicine, University of Leipzig, Leipzig, Germany
- Department of Neurosurgery, University Hospital, Leipzig, Germany
- * E-mail:
| | - Eileen Hempel
- Department of Neurosurgery, University Hospital, Leipzig, Germany
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | - Javorina Milosevic
- Translational Centre for Regenerative Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
39
|
Hu Q, Liang X, Chen D, Chen Y, Doycheva D, Tang J, Tang J, Zhang JH. Delayed hyperbaric oxygen therapy promotes neurogenesis through reactive oxygen species/hypoxia-inducible factor-1α/β-catenin pathway in middle cerebral artery occlusion rats. Stroke 2014; 45:1807-14. [PMID: 24757104 DOI: 10.1161/strokeaha.114.005116] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Hyperbaric oxygen (HBO) has been reported to be neuroprotective and to improve neurofunctional outcomes in acute stroke. However, it is not clear whether delayed HBO enhances endogenous neurogenesis and promotes neurofunctional recovery. The aim of this study is to evaluate the effects of delayed HBO therapy on neurogenesis and its potential mechanisms. METHODS One hundred eleven male Sprague-Dawley rats that survived for 7 days from 2 hours of middle cerebral artery occlusion and reperfusion were used. Delayed and multiple HBO were administrated beginning at 7 days after middle cerebral artery occlusion and lasting for 42 days with 3 HBO-free intervals (5 days each). Motor sensory deficits were measured by foot-fault test, and learning and memory abilities were evaluated by Morris water maze. Neurogenesis was examined by double immunostaining of bromodeoxyuridine and doublecortin, bromodeoxyuridine and neuronal nuclei at day 42. For mechanism studies, inhibitors for reactive oxygen species (ROS), hypoxia-inducible factor (HIF)-1α, and β-catenin were administrated, and the levels of ROS, HIF-1α, β-catenin, lymphoid enhancer-binding factor-1, T-cell factor-1, neurogenin-1, doublecortin, and synapsin-1 were assessed by ELISA or Western blot at day 14. RESULTS Delayed HBO treatment promoted neurogenesis and improved neurofunctional recovery at day 42, and the improvements were reversed by inhibition of ROS and HIF-1α. Delayed HBO significantly increased ROS and HIF-1α, and upregulated the expression of neurogenin-1, doublecortin, and synapsin-1. Inhibition of ROS and HIF-1α removed the effects of delayed HBO. CONCLUSIONS Delayed HBO enhanced endogenous neurogenesis and improved neurofunctional recovery in the late-chronic phase of stroke possibly mediated by ROS/HIF-1α/β-catenin pathway. Delayed HBO may serve as an alternative treatment to improve long-term recovery of stroke survivors.
Collapse
Affiliation(s)
- Qin Hu
- From the Departments of Physiology and Pharmacology (Q.H., X.L., D.C., Y.C., D.D., Junjia Tang, Jiping Tang, J.H.Z.) and Department of Neurosurgery, Loma Linda University School of Medicine, CA (J.H.Z.)
| | - Xiping Liang
- From the Departments of Physiology and Pharmacology (Q.H., X.L., D.C., Y.C., D.D., Junjia Tang, Jiping Tang, J.H.Z.) and Department of Neurosurgery, Loma Linda University School of Medicine, CA (J.H.Z.)
| | - Di Chen
- From the Departments of Physiology and Pharmacology (Q.H., X.L., D.C., Y.C., D.D., Junjia Tang, Jiping Tang, J.H.Z.) and Department of Neurosurgery, Loma Linda University School of Medicine, CA (J.H.Z.)
| | - Yujie Chen
- From the Departments of Physiology and Pharmacology (Q.H., X.L., D.C., Y.C., D.D., Junjia Tang, Jiping Tang, J.H.Z.) and Department of Neurosurgery, Loma Linda University School of Medicine, CA (J.H.Z.)
| | - Desislava Doycheva
- From the Departments of Physiology and Pharmacology (Q.H., X.L., D.C., Y.C., D.D., Junjia Tang, Jiping Tang, J.H.Z.) and Department of Neurosurgery, Loma Linda University School of Medicine, CA (J.H.Z.)
| | - Junjia Tang
- From the Departments of Physiology and Pharmacology (Q.H., X.L., D.C., Y.C., D.D., Junjia Tang, Jiping Tang, J.H.Z.) and Department of Neurosurgery, Loma Linda University School of Medicine, CA (J.H.Z.)
| | - Jiping Tang
- From the Departments of Physiology and Pharmacology (Q.H., X.L., D.C., Y.C., D.D., Junjia Tang, Jiping Tang, J.H.Z.) and Department of Neurosurgery, Loma Linda University School of Medicine, CA (J.H.Z.)
| | - John H Zhang
- From the Departments of Physiology and Pharmacology (Q.H., X.L., D.C., Y.C., D.D., Junjia Tang, Jiping Tang, J.H.Z.) and Department of Neurosurgery, Loma Linda University School of Medicine, CA (J.H.Z.).
| |
Collapse
|
40
|
Reischl S, Li L, Walkinshaw G, Flippin LA, Marti HH, Kunze R. Inhibition of HIF prolyl-4-hydroxylases by FG-4497 reduces brain tissue injury and edema formation during ischemic stroke. PLoS One 2014; 9:e84767. [PMID: 24409307 PMCID: PMC3883663 DOI: 10.1371/journal.pone.0084767] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 11/18/2013] [Indexed: 01/06/2023] Open
Abstract
Ischemic stroke results in disruption of the blood-brain barrier (BBB), edema formation and neuronal cell loss. Some neuroprotective factors such as vascular endothelial growth factor (VEGF) favor edema formation, while others such as erythropoietin (Epo) can mitigate it. Both factors are controlled by hypoxia inducible transcription factors (HIF) and the activity of prolyl hydroxylase domain proteins (PHD). We hypothesize that activation of the adaptive hypoxic response by inhibition of PHD results in neuroprotection and prevention of vascular leakage. Mice, subjected to cerebral ischemia, were pre- or post-treated with the novel PHD inhibitor FG-4497. Inhibition of PHD activity resulted in HIF-1α stabilization, increased expression of VEGF and Epo, improved outcome from ischemic stroke and reduced edema formation by maintaining BBB integrity. Additional in vitro studies using brain endothelial cells and primary astrocytes confirmed that FG-4497 induces the HIF signaling pathway, leading to increased VEGF and Epo expression. In an in vitro ischemia model, using combined oxygen and glucose deprivation, FG-4497 promoted the survival of neurons. Furthermore, FG-4497 prevented the ischemia-induced rearrangement and gap formation of the tight junction proteins zonula occludens 1 and occludin, both in cultured endothelial cells and in infarcted brain tissue in vivo. These results indicate that FG-4497 has the potential to prevent cerebral ischemic damage by neuroprotection and prevention of vascular leakage.
Collapse
Affiliation(s)
- Stefan Reischl
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Lexiao Li
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Gail Walkinshaw
- FibroGen, Inc., San Francisco, California, United States of America
| | - Lee A. Flippin
- FibroGen, Inc., San Francisco, California, United States of America
| | - Hugo H. Marti
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Reiner Kunze
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
41
|
Kim MK, Park HJ, Kim YD, Ryu MH, Takata T, Bae SK, Bae MK. Hinokitiol increases the angiogenic potential of dental pulp cells through ERK and p38MAPK activation and hypoxia-inducible factor-1α (HIF-1α) upregulation. Arch Oral Biol 2013; 59:102-10. [PMID: 24370180 DOI: 10.1016/j.archoralbio.2013.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/14/2013] [Accepted: 10/23/2013] [Indexed: 12/30/2022]
Abstract
Hinokitiol, a natural iron-chelating agent, is known to have diverse biological and pharmacological activities in various cell types. However, the effect of hinokitiol on dental pulp cells has not yet been reported. In this study, hinokitiol increases hypoxia-inducible factor-1α (HIF-1α) protein levels and vascular endothelial growth factor (VEGF) secretion in human dental pulp cells. The extracellular-signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) pathways are involved in hinokitiol-induced HIF-1α protein expression in dental pulp cells. Conditioned media from hinokitiol-treated pulp cells enhances angiogenesis in vitro and in vivo. Overall, these results show that hinokitiol promotes ERK and p38MAPK activation and HIF-1α-induced VEGF production, thus increasing the angiogenic potential of dental pulp cells.
Collapse
Affiliation(s)
- Mi-Kyoung Kim
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 626-870, South Korea
| | - Hyun-Joo Park
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 626-870, South Korea; Department of Dental Pharmacology, School of Dentistry, Pusan National University, Yangsan 626-870, South Korea
| | - Yong-Deok Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, Yangsan 626-870, South Korea
| | - Mi Heon Ryu
- Department of Oral Pathology, School of Dentistry, Pusan National University, Yangsan 626-870, South Korea
| | - Takashi Takata
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Soo-Kyung Bae
- Department of Dental Pharmacology, School of Dentistry, Pusan National University, Yangsan 626-870, South Korea
| | - Moon-Kyoung Bae
- Department of Oral Physiology, School of Dentistry, Pusan National University, Yangsan 626-870, South Korea.
| |
Collapse
|
42
|
Piersanti S, Tagliafico E, Saggio I. DNA microarray to analyze adenovirus-host interactions. Methods Mol Biol 2013; 1089:89-104. [PMID: 24132480 DOI: 10.1007/978-1-62703-679-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Defining the molecular toxicity of viral vectors that are or will be in use for clinical trials is a prerequisite for their safe application in humans. DNA chips allow high-throughput evaluation of the profile of transduced cells and have contributed to underlining specific aspects of vector toxicity both in in vitro and in vivo assets. With gene chips we have been able to identify vector-specific properties, such as the cell cycle alteration induced by vector genomic DNA, along with the activation of specific innate immune pathways that can be ascribed to viral particles. We herein describe a detailed protocol for the use of gene chips to dissect the toxicogenomic signature of human and canine helper-dependent adenoviral vectors. We suggest specific procedures suited for the study of these viral vectors, but we also give indications that can be applied to different experimental contexts. In addition, we discuss the in silico elaboration of gene chip raw data which is a crucial step to extrapolate biological information from gene chip studies.
Collapse
Affiliation(s)
- Stefania Piersanti
- Dipartimento di Biologia e Biotecnologie, Sapienza, Università di Roma, Roma, Italy
| | | | | |
Collapse
|
43
|
Porzionato A, Macchi V, Zaramella P, Sarasin G, Grisafi D, Dedja A, Chiandetti L, De Caro R. Effects of postnatal hyperoxia exposure on the rat dentate gyrus and subventricular zone. Brain Struct Funct 2013; 220:229-47. [PMID: 24135771 DOI: 10.1007/s00429-013-0650-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 10/04/2013] [Indexed: 12/27/2022]
Abstract
Premature newborns may be exposed to hyperoxia in the first postnatal period, but clinical and experimental works have raised the question of oxygen toxicity for the developing brain. However, specific analysis of hyperoxia exposure on neurogenesis is still lacking. Thus, the aim of the present study was to evaluate possible changes in the morphometric parameters of the main neurogenic sites in newborn rats exposed to 60 or 95 % oxygen for the first 14 postnatal days. The optical disector, a morphometric method based upon unbiased sampling principles of stereology, was applied to analyse cell densities, total volumes, and total cell numbers of the dentate gyrus (DG) and subventricular zone (SVZ). Apoptosis and proliferation were also studied by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling method and anti-ki67 immunohistochemistry, respectively. Severe hyperoxia increased the percentage of apoptotic cells in the DG. Moderate and severe hyperoxia induced a proliferative response both in the DG and SVZ, but the two neurogenic sites showed different changes in their morphometric parameters. The DG of both the hyperoxic groups showed lower volume and total cell number than that of the normoxic one. Conversely, the SVZ of newborn rats exposed to 95 % hyperoxia showed statistically significant higher volume and total cell number than SVZ of rats raised in normoxia. Our findings indicate that hyperoxia exposure in the first postnatal period affects both the neurogenic areas, although in different ways, i.e. reduction of DG and expansion of SVZ.
Collapse
Affiliation(s)
- Andrea Porzionato
- Section of Anatomy, Department of Molecular Medicine, University of Padova, Via A Gabelli 65, 35127, Padua, Italy
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Piersanti S, Astrologo L, Licursi V, Costa R, Roncaglia E, Gennetier A, Ibanes S, Chillon M, Negri R, Tagliafico E, Kremer EJ, Saggio I. Differentiated neuroprogenitor cells incubated with human or canine adenovirus, or lentiviral vectors have distinct transcriptome profiles. PLoS One 2013; 8:e69808. [PMID: 23922808 PMCID: PMC3724896 DOI: 10.1371/journal.pone.0069808] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 06/13/2013] [Indexed: 12/13/2022] Open
Abstract
Several studies have demonstrated the potential for vector-mediated gene transfer to the brain. Helper-dependent (HD) human (HAd) and canine (CAV-2) adenovirus, and VSV-G-pseudotyped self-inactivating HIV-1 vectors (LV) effectively transduce human brain cells and their toxicity has been partly analysed. However, their effect on the brain homeostasis is far from fully defined, especially because of the complexity of the central nervous system (CNS). With the goal of dissecting the toxicogenomic signatures of the three vectors for human neurons, we transduced a bona fide human neuronal system with HD-HAd, HD-CAV-2 and LV. We analysed the transcriptional response of more than 47,000 transcripts using gene chips. Chip data showed that HD-CAV-2 and LV vectors activated the innate arm of the immune response, including Toll-like receptors and hyaluronan circuits. LV vector also induced an IFN response. Moreover, HD-CAV-2 and LV vectors affected DNA damage pathways--but in opposite directions--suggesting a differential response of the p53 and ATM pathways to the vector genomes. As a general response to the vectors, human neurons activated pro-survival genes and neuron morphogenesis, presumably with the goal of re-establishing homeostasis. These data are complementary to in vivo studies on brain vector toxicity and allow a better understanding of the impact of viral vectors on human neurons, and mechanistic approaches to improve the therapeutic impact of brain-directed gene transfer.
Collapse
Affiliation(s)
- Stefania Piersanti
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
| | - Letizia Astrologo
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
| | - Valerio Licursi
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
| | - Rossella Costa
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
| | - Enrica Roncaglia
- Dipartimento di Scienze Biomediche, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Aurelie Gennetier
- Institut de Génétique Moléculaire de Montpellier, University Montpellier I & II, Montpellier, France
| | - Sandy Ibanes
- Institut de Génétique Moléculaire de Montpellier, University Montpellier I & II, Montpellier, France
| | - Miguel Chillon
- Istituto Pasteur Fondazione Cenci Bolognetti, Roma, Italy
| | - Rodolfo Negri
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Roma, Italy
| | - Enrico Tagliafico
- Dipartimento di Scienze Biomediche, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Eric J. Kremer
- Institut de Génétique Moléculaire de Montpellier, University Montpellier I & II, Montpellier, France
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie “Chrales Darwin”, Sapienza, Università di Roma, Roma, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Roma, Italy
- Istituto di Biologia e Patologia Molecolari del CNR, Roma, Italy
- * E-mail:
| |
Collapse
|
45
|
Corcoran A, Kunze R, Harney SC, Breier G, Marti HH, O'Connor JJ. A role for prolyl hydroxylase domain proteins in hippocampal synaptic plasticity. Hippocampus 2013; 23:861-72. [PMID: 23674383 DOI: 10.1002/hipo.22142] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 03/26/2013] [Accepted: 04/26/2013] [Indexed: 01/17/2023]
Abstract
Hypoxia-inducible factors (HIFs) are key transcriptional regulators that play a major role in oxygen homeostasis. HIF activity is tightly regulated by oxygen-dependent hydroxylases, which additionally require iron and 2-oxoglutarate as cofactors. Inhibition of these enzymes has become a novel target to modulate the hypoxic response for therapeutic benefit. Inhibition of prolyl-4-hydroxylase domains (PHDs) have been shown to delay neuronal cell death and protect against ischemic injury in the hippocampus. In this study we have examined the effects of prolyl hydroxylase inhibition on synaptic transmission and plasticity in the hippocampus. Field excitatory postsynaptic potentials (fEPSPs) and excitatory postsynaptic currents (EPSCs) were elicited by stimulation of the Schaffer collateral pathway in the CA1 region of the hippocampus. Treatment of rat hippocampal slices with low concentrations (10 µM) of the iron chelator deferosoxamine (DFO) or the 2-oxoglutarate analogue dimethyloxalyl glycine (DMOG) had no effect on fEPSP. In contrast, application of 1 mM DMOG resulted in a significant decrease in fEPSP slope. Antagonism of the NMDA receptor attenuated the effects of DMOG on baseline synaptic signalling. In rat hippocampal slices pretreated with DMOG and DFO the induction of long-term potentiation (LTP) by tetanic stimulation was strongly impaired. Similarly, neuronal knockout of the single PHD family member PHD2 prevented murine hippocampal LTP. Preconditioning of PHD2 deficient hippocampi with either DMOG, DFO, or the PHD specific inhibitor JNJ-42041935, did not further decrease LTP suggesting that DMOG and DFO influences synaptic plasticity primarily by inhibiting PHDs rather than unspecific effects. These findings provide striking evidence for a modulatory role of PHD proteins on synaptic plasticity in the hippocampus.
Collapse
Affiliation(s)
- Alan Corcoran
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, 4, Ireland
| | | | | | | | | | | |
Collapse
|
46
|
Najafi R, Sharifi AM. Deferoxamine preconditioning potentiates mesenchymal stem cell homing in vitro and in streptozotocin-diabetic rats. Expert Opin Biol Ther 2013; 13:959-72. [PMID: 23536977 DOI: 10.1517/14712598.2013.782390] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Today, cell therapy is considered a promising alternative in treatment of several diseases such as type 1 diabetes. Loss of transplanted stem cell and more importantly scarcity in the number of cells reaching to target tissue is a major obstacle in cell therapy. There is evidences showing that deferoxamine (DFO), an iron chelator, increases the mobilization and homing of progenitor cells through increasing the stability of hypoxia-inducible factor 1α (HIF-1α) protein. In this study, the effect of DFO on some factors involved in homing of bone marrow-derived mesenchymal stem cell was investigated, and the other objectives of this research were to determine whether DFO is able to increase migration and subsequent homing of mesenchymal stem cell (MSCs) both in vitro and in vivo in streptozotocin-diabetic rats. RESEARCH DESIGN AND METHODS MSCs were treated by DFO in minimal essential medium α (αMEM) for 24 h. The expression and localization of HIF-1α were evaluated by western blotting and immunocytochemistry. The expression of C-X-C chemokine receptor type 4 (CXCR-4) and chemokine receptor 2 (CCR2) were assessed by western blotting and RT-PCR. The activity of matrix metalloproteinases (MMP) -2 and -9 were measured by gelatin zymography. Finally, in vitro migration of MSCs toward different concentrations of stromal cell-derived factor and monocyte chemotactic protein-1 were also evaluated. To demonstrate the homing of MSCs in vivo, DFO-treated chloromethyl-benzamidodialkylcarbocyanine-labeled MSCs were injected into the tail vein of rats, and the number of stained MSCs reaching to the pancreas were determined after 24 h. RESULTS In DFO-treated MSCs, expression of HIF-1α (p < 0.001), CXCR4 (p < 0.001), CCR2 (p < 0.001), and the activity of MMP-2 (p < 0.01) and MMP-9 (p < 0.05) were significantly increased compared to control groups. Elevation of HIF-1α, upregulation of CXCR4/CCR2 and higher activity of MMP-2/MMP-9 in DFO-treated MSCs were reversed by 2-methoxyestradiol (2-ME; 5 μmol), a HIF-1α inhibitor. The in vitro migrations as well as in vivo homing of DFO-treated MSCs were also significantly higher than control groups (p < 0.05). CONCLUSIONS Preconditioning of MSCs by DFO prior to transplantation could increase homing of MSCs through affecting some chemokine receptors as well as proteases involved and eventually improving the efficacy of cell therapy.
Collapse
Affiliation(s)
- R Najafi
- Tehran University of Medical Sciences, School of Medicine, Razi Drug Research Center, Department of Pharmacology, Tehran, Iran
| | | |
Collapse
|
47
|
Wu YM, Jin R, Yang L, Zhang J, Yang Q, Guo YY, Li XB, Liu SB, Luo XX, Zhao MG. Phosphatidylinositol 3 kinase/protein kinase B is responsible for the protection of paeoniflorin upon H₂O₂-induced neural progenitor cell injury. Neuroscience 2013; 240:54-62. [PMID: 23485815 DOI: 10.1016/j.neuroscience.2013.02.037] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 01/23/2013] [Accepted: 02/15/2013] [Indexed: 12/26/2022]
Abstract
Promoting neural stem/progenitor cell (NSC/NPC) survival in the pro-apoptotic environment is critical to stem cell replacement for neurodegenerative disease therapy. Paeoniflorin (PF), one of the principal bioactive components in Paeoniae Radix, has been used widely in central nervous system (CNS) diseases treatment and serves as an antioxidant to protect neurons against oxidative stress. The present study investigated the protective effects of PF on NPC injury induced by hydrogen peroxide (H₂O₂). After challenge with 200 μM H₂O₂ for 2h, loss of cell viability and excessive apoptotic cell death were observed in cultured NPC, PF treatment conferred protective effects against the loss of cellular viability in a concentration-dependent manner. PF pretreatment also inhibited NPC apoptosis induced by H₂O₂ by reversing the decreased level of Procaspase-3 and balancing Bcl-2 and Bax expression. Furthermore, PF-mediated NPC protection was associated with an increase in phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt-1) phosphorylation in a time- and concentration-dependent manner. Selective inhibition of PI3K using LY294002 abolished PF-mediated phosphorylation of Akt-1 and NPC protection upon oxidative stress. These data suggest that PF-mediated NPC protection on H₂O₂ injury is reliant on the activation of the PI3K/Akt-1 pathway, giving insight to an essential role of PF in NPC protection.
Collapse
Affiliation(s)
- Y-M Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Transmembrane prolyl 4-hydroxylase is a fourth prolyl 4-hydroxylase regulating EPO production and erythropoiesis. Blood 2012; 120:3336-44. [DOI: 10.1182/blood-2012-07-441824] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AbstractAn endoplasmic reticulum transmembrane prolyl 4-hydroxylase (P4H-TM) is able to hydroxylate the α subunit of the hypoxia-inducible factor (HIF) in vitro and in cultured cells, but nothing is known about its roles in mammalian erythropoiesis. We studied such roles here by administering a HIF-P4H inhibitor, FG-4497, to P4h-tm−/− mice. This caused larger increases in serum Epo concentration and kidney but not liver Hif-1α and Hif-2α protein and Epo mRNA levels than in wild-type mice, while the liver Hepcidin mRNA level was lower in the P4h-tm−/− mice than in the wild-type. Similar, but not identical, differences were also seen between FG-4497–treated Hif-p4h-2 hypomorphic (Hif-p4h-2gt/gt) and Hif-p4h-3−/− mice versus wild-type mice. FG-4497 administration increased hemoglobin and hematocrit values similarly in the P4h-tm−/− and wild-type mice, but caused higher increases in both values in the Hif-p4h-2gt/gt mice and in hematocrit value in the Hif-p4h-3−/− mice than in the wild-type. Hif-p4h-2gt/gt/P4h-tm−/− double gene-modified mice nevertheless had increased hemoglobin and hematocrit values without any FG-4497 administration, although no such abnormalities were seen in the Hif-p4h-2gt/gt or P4h-tm−/− mice. Our data thus indicate that P4H-TM plays a role in the regulation of EPO production, hepcidin expression, and erythropoiesis.
Collapse
|
49
|
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
|
50
|
Kunze R, Zhou W, Veltkamp R, Wielockx B, Breier G, Marti HH. Neuron-specific prolyl-4-hydroxylase domain 2 knockout reduces brain injury after transient cerebral ischemia. Stroke 2012; 43:2748-56. [PMID: 22933585 DOI: 10.1161/strokeaha.112.669598] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Numerous factors involved in the adaptive response to hypoxia, including erythropoietin and vascular endothelial growth factor are transcriptionally regulated by hypoxia-inducible factors (HIFs). During normoxia, prolyl-4-hydroxylase domain (PHD) proteins hydroxylate HIF-α subunits, resulting in their degradation. We investigated the effect of neuronal deletion of PHD2, the most abundant isoform in brain, for stroke outcome. METHODS We generated neuron-specific Phd2 knockout mice and subjected animals to systemic hypoxia or transient middle cerebral artery occlusion. Infarct volume and cell death were determined by histology. HIF-1α, HIF-2α, and HIF target genes were analyzed by immunoblotting and real-time polymerase chain reaction, respectively. RESULTS Neuron-specific ablation of Phd2 significantly increased protein stability of HIF-1α and HIF-2α in the forebrain and enhanced expression of the neuroprotective HIF target genes erythropoietin and vascular endothelial growth factor as well as glucose transporter and glycolysis-related enzymes under hypoxic and ischemic conditions. Mice with Phd2-deficient neurons subjected to transient cerebral ischemia exhibited a strong reduction in infarct size, and cell death of hippocampal CA1 neurons located in the peri-infarct region was dramatically reduced in these mice. Vessel density in forebrain subregions, except for caudate-putamen, was not altered in Phd2-deficient animals. CONCLUSIONS Our findings denote that the endogenous adaptive response on hypoxic-ischemic insults in the brain is at least partly dependent on the activity of HIFs and identify PHD2 as the key regulator for the protective hypoxia response. The results suggest that specific inhibition of PHD2 may provide a useful therapeutic strategy to protect brain tissue from ischemic injury.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|