1
|
Chang Z, Liu Q, Fan P, Xu W, Xie Y, Gong K, Zhang C, Zhao Z, Sun K, Shao G. Hypoxia preconditioning increases Notch1 activity by regulating DNA methylation in vitro and in vivo. Mol Biol Rep 2024; 51:507. [PMID: 38622406 DOI: 10.1007/s11033-024-09308-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/01/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Our previous research has demonstrated that hypoxic preconditioning (HPC) can improve spatial learning and memory abilities in adult mice. Adult hippocampal neurogenesis has been associated with learning and memory. The Neurogenic locus notch homolog protein (Notch) was involved in adult hippocampal neurogenesis, as well as in learning and memory. It is currently unclear whether the Notch pathway regulates hippocampal neuroregeneration by modifying the DNA methylation status of the Notch gene following HPC. METHOD The HPC animal model and cell model were established through repeated hypoxia exposure using mice and the mouse hippocampal neuronal cell line HT22. Step-down test was conducted on HPC mice. Real-time PCR and Western blot analysis were used to assess the mRNA and protein expression levels of Notch1 and hairy and enhancer of split1 (HES1). The presence of BrdU-positive cells and Notch1 expression in the hippocampal dental gyrus (DG) were examined with confocal microscopy. The methylation status of the Notch1 was analyzed using methylation-specific PCR (MS-PCR). HT22 cells were employed to elucidate the impact of HPC on Notch1 in vitro. RESULTS HPC significantly improved the step-down test performance of mice with elevated levels of mRNA and protein expression of Notch1 and HES1 (P < 0.05). The intensities of the Notch1 signal in the control group, the H group and the HPC group were 2.62 ± 0.57 × 107, 2.87 ± 0.84 × 107, and 3.32 ± 0.14 × 107, respectively, and the number of BrdU (+) cells in the hippocampal DG were 1.83 ± 0.54, 3.71 ± 0.64, and 7.29 ± 0.68 respectively. Compared with that in C and H group, the intensity of the Notch1 signal and the number of BrdU (+) cells increased significantly in HPC group (P < 0.05). The methylation levels of the Notch1 promoter 0.82 ± 0.03, 0.65 ± 0.03, and 0.60 ± 0.02 in the C, H, and HPC groups, respectively. The methylation levels of Notch1 decreased significantly (P < 0.05). The effect of HPC on HT22 cells exhibited similarities to that observed in the hippocampus. CONCLUSION HPC may confer neuroprotection by activating the Notch1 signaling pathway and regulating its methylation level, resulting in the regeneration of hippocampal neurons.
Collapse
Affiliation(s)
- Zhehan Chang
- Center for Translational Medicine, The Third People's Hospital of Longgang District, Shenzhen, China
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qi Liu
- Department of Radiology, The Second Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Peijia Fan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wenqiang Xu
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yabin Xie
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kerui Gong
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, USA
| | - Chunyang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Zhijun Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Baotou, China.
| | - Kai Sun
- Center for Translational Medicine, The Third People's Hospital of Longgang District, Shenzhen, China.
| | - Guo Shao
- Center for Translational Medicine, The Third People's Hospital of Longgang District, Shenzhen, China.
- Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China.
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Baotou, China.
- Joint Laboratory of South China Hospital Affiliated to Shenzhen University and Third People's Hospital of Longgang District, Shenzhen University, Shenzhen, China.
| |
Collapse
|
2
|
Costa DP, Favilla AB. Field physiology in the aquatic realm: ecological energetics and diving behavior provide context for elucidating patterns and deviations. J Exp Biol 2023; 226:jeb245832. [PMID: 37843467 DOI: 10.1242/jeb.245832] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Comparative physiology has developed a rich understanding of the physiological adaptations of organisms, from microbes to megafauna. Despite extreme differences in size and a diversity of habitats, general patterns are observed in their physiological adaptations. Yet, many organisms deviate from the general patterns, providing an opportunity to understand the importance of ecology in determining the evolution of unusual adaptations. Aquatic air-breathing vertebrates provide unique study systems in which the interplay between ecology, physiology and behavior is most evident. They must perform breath-hold dives to obtain food underwater, which imposes a physiological constraint on their foraging time as they must resurface to breathe. This separation of two critical resources has led researchers to investigate these organisms' physiological adaptations and trade-offs. Addressing such questions on large marine animals is best done in the field, given the difficulty of replicating the environment of these animals in the lab. This Review examines the long history of research on diving physiology and behavior. We show how innovative technology and the careful selection of research animals have provided a holistic understanding of diving mammals' physiology, behavior and ecology. We explore the role of the aerobic diving limit, body size, oxygen stores, prey distribution and metabolism. We then identify gaps in our knowledge and suggest areas for future research, pointing out how this research will help conserve these unique animals.
Collapse
Affiliation(s)
- Daniel P Costa
- Institute of Marine Sciences, Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, Santa Cruz, CA 95060, USA
| | - Arina B Favilla
- Institute of Marine Sciences, Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, Santa Cruz, CA 95060, USA
| |
Collapse
|
3
|
Liu N, Zhang Y, Zhang P, Gong K, Zhang C, Sun K, Shao G. Vascular Endothelial Growth Factor and Erythropoietin Show Different Expression Patterns in the Early and Late Hypoxia Preconditioning Phases and May Correlate with DNA Methylation Status in the Mouse Hippocampus. High Alt Med Biol 2022; 23:361-368. [PMID: 36449395 DOI: 10.1089/ham.2021.0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Liu, Na, Yanbo Zhang, Pu Zhang, Kerui Gong, Chunyang Zhang, Kai Sun, and Guo Shao. Vascular endothelial growth factor and erythropoietin show different expression patterns in the early and late hypoxia preconditioning phases and may correlate with DNA methylation status in the mouse hippocampus. High Alt Med Biol. 23:361-368, 2022. Background: Vascular endothelial growth factor (VEGF) and erythropoietin (EPO) have been proven to participate in neuroprotection induced by hypoxia preconditioning (HPC), and they can be regulated by hypoxia-inducible factor 1 (HIF-1). It has been reported that DNA methylation can affect VEGF and EPO expression. This study aimed to explore the expression of VEGF and EPO in the early phase and late phase of HPC and whether their expression was affected by DNA methylation. Method: Acute repeated HPC mice were used as the animal model, and detection of molecular changes was performed immediately (early phase) and 1 day (late phase) after HPC treatment. The mRNA and protein expression levels of VEGF, EPO, and DNA methyltransferases (DNMTs) in the hippocampi were measured by real-time polymerase chain reaction and western blotting, respectively. The activity of DNMTs and global methylation levels were analyzed by enzyme-linked immunosorbent assay. DNA methylation levels of VEGF and EPO promoters, which were catalyzed by DNMTs, were determined by bisulfite-modified DNA sequencing. Results: The expression of VEGF was increased in the early phase and late phase of HPC (p < 0.05), whereas the expression of EPO was unchanged in the early phase (p > 0.05) of HPC and was increased in the late phase (p < 0.05). VEGF and EPO expression were negatively correlated with the DNA methylation levels of their promoters. DNMT3A and DNMT3B were decreased in the early phase and late phase (p < 0.05), whereas DNMT1 was unchanged in the early phase and late phase (p > 0.05). Conclusions: Our data demonstrated that DNMTs affect VEGF and EPO expression by regulating the DNA methylation levels of the promoters of VEGF and EPO.
Collapse
Affiliation(s)
- Na Liu
- Department of Laboratory Medicine, Center for Translational Medicine, the Third People's Hospital of Longgang District, Shenzhen, China.,Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College of Neuroscience Institute, Baotou Medical College, Inner Mongolia, China.,Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yanbo Zhang
- Department of Laboratory Medicine, Center for Translational Medicine, the Third People's Hospital of Longgang District, Shenzhen, China.,Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Pu Zhang
- Department of Laboratory Medicine, Center for Translational Medicine, the Third People's Hospital of Longgang District, Shenzhen, China.,Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College of Neuroscience Institute, Baotou Medical College, Inner Mongolia, China.,Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kerui Gong
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, California, USA
| | - Chunyang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia, China
| | - Kai Sun
- Department of Laboratory Medicine, Center for Translational Medicine, the Third People's Hospital of Longgang District, Shenzhen, China.,Joint Laboratory of South China Hospital Affiliated to Shenzhen University and Third People's Hospital of Longgang District, Shenzhen University, Shenzhen, China
| | - Guo Shao
- Department of Laboratory Medicine, Center for Translational Medicine, the Third People's Hospital of Longgang District, Shenzhen, China.,Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College of Neuroscience Institute, Baotou Medical College, Inner Mongolia, China.,Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia, China.,Joint Laboratory of South China Hospital Affiliated to Shenzhen University and Third People's Hospital of Longgang District, Shenzhen University, Shenzhen, China
| |
Collapse
|
4
|
Eaton L, Pamenter ME. What to do with low O 2: Redox adaptations in vertebrates native to hypoxic environments. Comp Biochem Physiol A Mol Integr Physiol 2022; 271:111259. [PMID: 35724954 DOI: 10.1016/j.cbpa.2022.111259] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 01/05/2023]
Abstract
Reactive oxygen species (ROS) are important cellular signalling molecules but sudden changes in redox balance can be deleterious to cells and lethal to the whole organism. ROS production is inherently linked to environmental oxygen availability and many species live in variable oxygen environments that can range in both severity and duration of hypoxic exposure. Given the importance of redox homeostasis to cell and animal viability, it is not surprising that early studies in species adapted to various hypoxic niches have revealed diverse strategies to limit or mitigate deleterious ROS changes. Although research in this area is in its infancy, patterns are beginning to emerge in the suites of adaptations to different hypoxic environments. This review focuses on redox adaptations (i.e., modifications of ROS production and scavenging, and mitigation of oxidative damage) in hypoxia-tolerant vertebrates across a range of hypoxic environments. In general, evidence suggests that animals adapted to chronic lifelong hypoxia are in homeostasis, and do not encounter major oxidative challenges in their homeostatic environment, whereas animals exposed to seasonal chronic anoxia or hypoxia rapidly downregulate redox balance to match a hypometabolic state and employ robust scavenging pathways during seasonal reoxygenation. Conversely, animals adapted to intermittent hypoxia exposure face the greatest degree of ROS imbalance and likely exhibit enhanced ROS-mitigation strategies. Although some progress has been made, research in this field is patchy and further elucidation of mechanisms that are protective against environmental redox challenges is imperative for a more holistic understanding of how animals survive hypoxic environments.
Collapse
Affiliation(s)
- Liam Eaton
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew E Pamenter
- Department of Biology, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
5
|
Chen Y, Hou G, Jing M, Teng H, Liu Q, Yang X, Wang Y, Qu J, Shi C, Lu L, Zhang J, Zhang Y. Genomic analysis unveils mechanisms of northward invasion and signatures of plateau adaptation in the Asian house rat. Mol Ecol 2021; 30:6596-6610. [PMID: 34564921 DOI: 10.1111/mec.16194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/21/2021] [Accepted: 09/20/2021] [Indexed: 12/20/2022]
Abstract
The Asian house rat (AHR), Rattus tanezumi, has recently invaded the northern half of China. The AHR is a highly adaptive rat species that has also successfully conquered the Qinghai-Tibet Plateau (QTP) and replaced the brown rat (BR), R. norvegicus, at the edge of the QTP. Here, we assembled a draft genome of the AHR and explored the mechanisms of its northward invasion and the genetic basis underlying plateau adaptation in this species. Population genomic analyses revealed that the northwardly invasive AHRs consisted of two independent and genetically distinct populations which might result from multiple independent primary invasion events. One invasive population exhibited reduced genetic diversity and distinct population structure compared with its source population, while the other displayed preserved genetic polymorphisms and little genetic differentiation from its source population. Genes involved in G-protein coupled receptors and carbohydrate metabolism may contribute to the local adaptation of northern AHRs. In particular, RTN4 was identified as a key gene for AHRs in the QTP that favours adaptation to high-altitude hypoxia. Coincidently, the physiological performance and transcriptome profiles of hypoxia-exposed rats both showed better hypoxia adaptation in AHRs than in BRs that failed to colonize the heart of the QTP, which may have facilitated the replacement of the BR population by the invading AHRs at the edge of the QTP. This study provides profound insights into the multiple origins of the northwardly invasive AHR and the great tolerance to hypoxia in this species.
Collapse
Affiliation(s)
- Yi Chen
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Guanmei Hou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Meidong Jing
- School of Life Sciences, Nantong University, Nantong, China
| | - Huajing Teng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Quansheng Liu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Xingen Yang
- Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Institute of Plant Protection, Shanxi Academy of Agricultural Sciences, Taiyuan, China
| | - Yong Wang
- Dongting Lake Station for Wetland Ecosystem Research, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Jiapeng Qu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Qinghai, China
| | - Chengmin Shi
- College of Plant Protection, Hebei Agricultural University, Baoding, China
| | - Liang Lu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jianxu Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Yaohua Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Rocha ACG, Cristina-Silva C, Taxini CL, da Costa Silva KS, Lima VTM, Macari M, Bícego KC, Szawka RE, Gargaglioni LH. Embryonic Thermal Manipulation Affects Ventilation, Metabolism, Thermal Control and Central Dopamine in Newly Hatched and Juvenile Chicks. Front Physiol 2021; 12:699142. [PMID: 34220555 PMCID: PMC8249324 DOI: 10.3389/fphys.2021.699142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/20/2021] [Indexed: 11/29/2022] Open
Abstract
The first third of incubation is critical for embryonic development, and environmental changes during this phase can affect the physiology and survival of the embryos. We evaluated the effects of low (LT), control (CT), and high (HT) temperatures during the first 5 days of incubation on ventilation (V.E), body temperature (Tb), oxygen consumption (V.O2), respiratory equivalent (V.E/V.O2), and brain monoamines on 3-days-old (3d) and 14-days-old (14d) male and female chickens. The body mass of LT animals of both ages and sexes was higher compared to HT and CT animals (except for 3d males). The heart mass of 14d HT animals was higher than that of CT animals. Thermal manipulation did not affect V.E, V.O2 or V.E/V.O2 of 3d animals in normoxia, except for 3d LT males V.E, which was lower than CT. Regarding 14d animals, the HT females showed a decrease in V.E and V.O2 compared to CT and LT groups, while the HT males displayed a lower V.O2 compared to CT males, but no changes in V.E/V.O2. Both sexes of 14d HT chickens presented a greater Tb compared to CT animals. Thermal manipulations increased the dopamine turnover in the brainstem of 3d females. No differences were observed in ventilatory and metabolic parameters in the 3d animals of either sexes, and 14d males under 7% CO2. The hypercapnic hyperventilation was attenuated in the 14d HT females due to changes in V.O2, without alterations in V.E. The 14d LT males showed a lower V.E, during hypercapnia, compared to CT, without changes in V.O2, resulting in an attenuation in V.E/V.O2. During hypoxia, 3d LT females showed an attenuated hyperventilation, modulated by a higher V.O2. In 14d LT and HT females, the increase in V.E was greater and the hypometabolic response was attenuated, compared to CT females, which resulted in no change in the V.E/V.O2. In conclusion, thermal manipulations affect hypercapnia-induced hyperventilation more so than hypoxic challenge, and at both ages, females are more affected by thermal manipulation than males.
Collapse
Affiliation(s)
- Aline C G Rocha
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, São Paulo, Brazil
| | - Caroline Cristina-Silva
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, São Paulo, Brazil
| | | | - Kaoma Stephani da Costa Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Virgínia T M Lima
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Marcos Macari
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, São Paulo, Brazil
| | - Kênia C Bícego
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, São Paulo, Brazil
| | - Raphael E Szawka
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais - UFMG, Belo Horizonte, Brazil
| | - Luciane H Gargaglioni
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinarian Sciences, São Paulo State University, São Paulo, Brazil
| |
Collapse
|
7
|
Xu K, Cai W, Zeng Y, Li J, He J, Cui F, Liu J. Video-assisted thoracoscopic surgery for primary lung cancer resections in patients with moderate to severe chronic obstructive pulmonary diseases. Transl Lung Cancer Res 2021; 10:2603-2613. [PMID: 34295665 PMCID: PMC8264335 DOI: 10.21037/tlcr-21-449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 06/17/2021] [Indexed: 11/06/2022]
Abstract
Background Lung cancer patients with chronic obstructive pulmonary disease (COPD) are considered a high-risk population to receive radical surgical treatment due to the high incidence of cardiopulmonary complications. The aim of this study was to evaluate the clinical factors associated with postoperative complications in primary lung cancer patients with moderate to extremely severe grades of COPD. Methods From December 2015 to June 2020, 138 patients with moderate to extremely severe COPD who underwent video-assisted thoracoscopic surgery (VATS) lung cancer resection (lobectomy or sublobar resection) were retrospectively reviewed. Patients' postoperative complications were collected from clinical records. Clinical factors (such as COPD severity or surgical approaches, etc.) were evaluated to investigate the association with postoperative complications. Results Of the 138 patients included in the study, the mean age was 67 (63-74) years, the mean preoperative forced expiratory volume in one second (FEV1) was 1.33±0.39 L, the mean FEV1% was 51.23% (41.43-60.00%). 33% patients (46/138) had postoperative complications, and no mortality occurred. Univariate analysis revealed that incidence of overall complications (OCs) and respiratory complications (RCs) was markedly higher in extremely severe COPD patients compared to moderate (OCs, P=0.033; RCs, P=0.050) and severe (OCs, P=0.015; RCs, P=0.008) COPD patients, respectively. Multivariate analysis showed that COPD grade was an independent risk factor of RCs (P=0.024). Furthermore, the grades of COPD (moderate, P=0.029; severe, P=0.028; extremely severe, P=0.019) and the surgical procedure (lobectomy or sublobar resection, P=0.043) were independent risk factors for atelectasis, which was the most common postoperative complication. Conclusions The aggravation of COPD was accompanied by an increase in the incidence of respiratory system complications postoperatively, especially atelectasis. For patients with moderate to extremely severe grades of COPD, careful perioperative evaluation should be performed to identify the indicators that influence the surgical choice between lobectomy and sublobar resection.
Collapse
Affiliation(s)
- Ke Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Weipeng Cai
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Yuan Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Jingpei Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Jianxing He
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Fei Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Jun Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, China
| |
Collapse
|
8
|
Li J, Zhao H, Xing Y, Zhao T, Cai L, Yan Z. A Genome-Wide Analysis of the Gene Expression and Alternative Splicing Events in a Whole-Body Hypoxic Preconditioning Mouse Model. Neurochem Res 2021; 46:1101-1111. [PMID: 33582968 DOI: 10.1007/s11064-021-03241-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/07/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022]
Abstract
Exposure to specific doses of hypoxia can trigger endogenous neuroprotective and neuroplastic mechanisms of the central nervous system. These molecular mechanisms, together referred to as hypoxic preconditioning (HPC), remain poorly understood. In the present study, we applied RNA sequencing and bioinformatics analyses to study HPC in a whole-body HPC mouse model. The preconditioned (H4) and control (H0) groups showed 605 differentially expressed genes (DEGs), of which 263 were upregulated and 342 were downregulated. Gene Ontology enrichment analysis indicated that these DEGs were enriched in several biological processes, including metabolic stress and angiogenesis. The Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed that the FOXO and Notch signaling pathways were involved in hypoxic tolerance and protection during HPC. Furthermore, 117 differential alternative splicing events (DASEs) were identified, with exon skipping being the dominant one (48.51%). Repeated exposure to systemic hypoxia promoted skipping of exon 7 in Edrf1 and exon 9 or 13 in Lrrc45. This study expands the understanding of the endogenous protective mechanisms of HPC and the DASEs that occur during HPC.
Collapse
Affiliation(s)
- Jun Li
- College of Science, Inner Mongolia Agricultural University, Hohhot, 010018, People's Republic of China
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China
| | - Hongyu Zhao
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China
| | - Yongqiang Xing
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China
| | - Tongling Zhao
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China
| | - Lu Cai
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China.
- Inner Mongolia Key Laboratory of Functional Genome Bioinformatics, Inner Mongolia University of Science and Technology, Baotou, 014010, People's Republic of China.
| | - Zuwei Yan
- College of Science, Inner Mongolia Agricultural University, Hohhot, 010018, People's Republic of China.
| |
Collapse
|
9
|
Kim MS, Kim DH, Kang HK, Kook MG, Choi SW, Kang KS. Modeling of Hypoxic Brain Injury through 3D Human Neural Organoids. Cells 2021; 10:cells10020234. [PMID: 33504071 PMCID: PMC7911731 DOI: 10.3390/cells10020234] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/14/2021] [Accepted: 01/22/2021] [Indexed: 01/04/2023] Open
Abstract
Brain organoids have emerged as a novel model system for neural development, neurodegenerative diseases, and human-based drug screening. However, the heterogeneous nature and immature neuronal development of brain organoids generated from pluripotent stem cells pose challenges. Moreover, there are no previous reports of a three-dimensional (3D) hypoxic brain injury model generated from neural stem cells. Here, we generated self-organized 3D human neural organoids from adult dermal fibroblast-derived neural stem cells. Radial glial cells in these human neural organoids exhibited characteristics of the human cerebral cortex trend, including an inner (ventricular zone) and an outer layer (early and late cortical plate zones). These data suggest that neural organoids reflect the distinctive radial organization of the human cerebral cortex and allow for the study of neuronal proliferation and maturation. To utilize this 3D model, we subjected our neural organoids to hypoxic injury. We investigated neuronal damage and regeneration after hypoxic injury and reoxygenation. Interestingly, after hypoxic injury, reoxygenation restored neuronal cell proliferation but not neuronal maturation. This study suggests that human neural organoids generated from neural stem cells provide new opportunities for the development of drug screening platforms and personalized modeling of neurodegenerative diseases, including hypoxic brain injury.
Collapse
Affiliation(s)
| | | | | | | | - Soon Won Choi
- Correspondence: (S.W.C.); (K.-S.K.); Tel.: +82-2-880-1298 (S.W.C.); +82-2-880-1246 (K.-S.K.)
| | - Kyung-Sun Kang
- Correspondence: (S.W.C.); (K.-S.K.); Tel.: +82-2-880-1298 (S.W.C.); +82-2-880-1246 (K.-S.K.)
| |
Collapse
|
10
|
Li S, Ren C, Stone C, Chandra A, Xu J, Li N, Han C, Ding Y, Ji X, Shao G. Hamartin: An Endogenous Neuroprotective Molecule Induced by Hypoxic Preconditioning. Front Genet 2020; 11:582368. [PMID: 33193709 PMCID: PMC7556298 DOI: 10.3389/fgene.2020.582368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/08/2020] [Indexed: 11/23/2022] Open
Abstract
Hypoxic/ischemic preconditioning (HPC/IPC) is an innate neuroprotective mechanism in which a number of endogenous molecules are known to be involved. Tuberous sclerosis complex 1 (TSC1), also known as hamartin, is thought to be one such molecule. It is also known that hamartin is involved as a target in the rapamycin (mTOR) signaling pathway, which functions to integrate a variety of environmental triggers in order to exert control over cellular metabolism and homeostasis. Understanding the role of hamartin in ischemic/hypoxic neuroprotection will provide a novel target for the treatment of hypoxic-ischemic disease. Therefore, the proposed molecular mechanisms of this neuroprotective role and its preconditions are reviewed in this paper, with emphases on the mTOR pathway and the relationship between the expression of hamartin and DNA methylation.
Collapse
Affiliation(s)
- Sijie Li
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China
| | - Christopher Stone
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Ankush Chandra
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Jiali Xu
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ning Li
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Cong Han
- Department of Neurosurgery, The Fifth Medical Centre of PLA General Hospital, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Guo Shao
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Inner Mongolia Key Laboratory of Hypoxic Translational Medicine, Baotou Medical College, Baotou, China.,Public Health Department, Biomedicine Research Center, Basic Medical College, Baotou, China.,Baotou Medical College of Neuroscience Institute, Baotou Medical College, Baotou, China
| |
Collapse
|
11
|
Das T, Soren K, Yerasi M, Kamle A, Kumar A, Chakravarty S. Molecular Basis of Sex Difference in Neuroprotection induced by Hypoxia Preconditioning in Zebrafish. Mol Neurobiol 2020; 57:5177-5192. [PMID: 32862360 DOI: 10.1007/s12035-020-02091-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/24/2020] [Indexed: 02/08/2023]
Abstract
Hypoxia, the major cause of ischemic injury, leads to debilitating disease in infants via birth asphyxia and cerebral palsy, whereas in adults via heart attack and stroke. A widespread, natural protective phenomenon termed 'hypoxic preconditioning' (PH) occurs when prior exposures to hypoxia eventually result in robust hypoxia resistance. Accordingly, we have developed and optimized a novel model of hypoxic preconditioning in adult zebrafish to mimic the tolerance of mini stroke(s) in human, which appears to protect against the severe damage inflicted by a major stroke event. Here, we observed a remarkable difference in the progression pattern of neuroprotection between preconditioning hypoxia followed by acute hypoxia (PH) group, and acute hypoxia (AH) only group, with noticeable sex difference when compared with normoxia behaviour upon recovery. Since gender difference has been reported in stroke risk factors and disease history, it was pertinent to investigate whether any such sex difference also exists in PH's protective mechanism against acute ischemic stroke. In order to elucidate the neural molecular mechanisms behind sex difference in neuroprotection induced by PH, a high throughput proteomics approach utilizing iTRAQ was performed, followed by protein enrichment analysis using ingenuity pathway analysis (IPA) tool. Out of thousands of significantly altered proteins in zebrafish brain, the ones having critical role either in neuroglial proliferation/differentiation or neurotrophic functions were validated by analyzing their expression levels in preconditioned (PH), acute hypoxia (AH), and normoxia groups. The data indicate that female zebrafish brains are more protected against the severity of AH when exposed to the hypoxic preconditioning. The study also sheds light on the involvement of many signalling pathways underlying sex difference in preconditioning-induced neuroprotective mechanism, which can be further validated for the therapeutic approach.
Collapse
Affiliation(s)
- Tapatee Das
- Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., 201002, India
| | - Kalyani Soren
- Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., 201002, India
| | - Mounica Yerasi
- Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India
| | - Avijeet Kamle
- CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India
| | - Arvind Kumar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., 201002, India.,CSIR-Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India
| | - Sumana Chakravarty
- Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P., 201002, India.
| |
Collapse
|
12
|
Turovskaya MV, Gaidin SG, Vedunova MV, Babaev AA, Turovsky EA. BDNF Overexpression Enhances the Preconditioning Effect of Brief Episodes of Hypoxia, Promoting Survival of GABAergic Neurons. Neurosci Bull 2020; 36:733-760. [PMID: 32219700 PMCID: PMC7340710 DOI: 10.1007/s12264-020-00480-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/17/2019] [Indexed: 12/18/2022] Open
Abstract
Hypoxia causes depression of synaptic plasticity, hyperexcitation of neuronal networks, and the death of specific populations of neurons. However, brief episodes of hypoxia can promote the adaptation of cells. Hypoxic preconditioning is well manifested in glutamatergic neurons, while this adaptive mechanism is virtually suppressed in GABAergic neurons. Here, we show that brain-derived neurotrophic factor (BDNF) overexpression in neurons enhances the preconditioning effect of brief episodes of hypoxia. The amplitudes of the NMDAR- and AMPAR-mediated Ca2+ responses of glutamatergic and GABAergic neurons gradually decreased after repetitive brief hypoxia/reoxygenation cycles in cell cultures transduced with the (AAV)-Syn-BDNF-EGFP virus construct. In contrast, the amplitudes of the responses of GABAergic neurons increased in non-transduced cultures after preconditioning. The decrease of the amplitudes in GABAergic neurons indicated the activation of mechanisms of hypoxic preconditioning. Preconditioning suppressed apoptotic or necrotic cell death. This effect was most pronounced in cultures with BDNF overexpression. Knockdown of BDNF abolished the effect of preconditioning and promoted the death of GABAergic neurons. Moreover, the expression of the anti-apoptotic genes Stat3, Socs3, and Bcl-xl substantially increased 24 h after hypoxic episodes in the transduced cultures compared to controls. The expression of genes encoding the pro-inflammatory cytokines IL-10 and IL-6 also increased. In turn, the expression of pro-apoptotic (Bax, Casp-3, and Fas) and pro-inflammatory (IL-1β and TNFα) genes decreased after hypoxic episodes in cultures with BDNF overexpression. Inhibition of vesicular BDNF release abolished its protective action targeting inhibition of the oxygen-glucose deprivation (OGD)-induced [Ca2+]i increase in GABAergic and glutamatergic neurons, thus promoting their death. Bafilomycin A1, Brefeldin A, and tetanus toxin suppressed vesicular release (including BDNF) and shifted the gene expression profile towards excitotoxicity, inflammation, and apoptosis. These inhibitors of vesicular release abolished the protective effects of hypoxic preconditioning in glutamatergic neurons 24 h after hypoxia/reoxygenation cycles. This finding indicates a significant contribution of vesicular BDNF release to the development of the mechanisms of hypoxic preconditioning. Thus, our results demonstrate that BDNF plays a pivotal role in the activation and enhancement of the preconditioning effect of brief episodes of hypoxia and promotes tolerance of the most vulnerable populations of GABAergic neurons to hypoxia/ischemia.
Collapse
Affiliation(s)
- M V Turovskaya
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia
| | - S G Gaidin
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia
| | - M V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - A A Babaev
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - E A Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia.
| |
Collapse
|
13
|
Zhang W, Chen M, Li H, Yuan J, Li J, Wu F, Zhang Y. Hypoxia preconditioning attenuates lung injury after thoracoscopic lobectomy in patients with lung cancer: a prospective randomized controlled trial. BMC Anesthesiol 2019; 19:209. [PMID: 31711422 PMCID: PMC6849275 DOI: 10.1186/s12871-019-0854-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/20/2019] [Indexed: 12/31/2022] Open
Abstract
Background Hypoxic preconditioning (HPC) may protect multiple organs from various injuries. We hypothesized that HPC would reduce lung injury in patients undergoing thoracoscopic lobectomy. Methods In a prospective randomized controlled trial, 70 patients undergoing elective thoracoscopic lobectomy were randomly allocated to the HPC group or the control group. Three cycles of 5-min hypoxia and 3-min ventilation applied to the nondependent lung served as the HPC intervention. The primary outcome was the PaO2/FiO2 ratio. Secondary outcomes included postoperative pulmonary complications, pulmonary function, and duration of hospital stay. Results HPC significantly increased the PaO2/FiO2 ratio compared with the control at 30 min after one-lung ventilation and 7 days after operation. Compared with the control, it also significantly improved postoperative pulmonary function and markedly reduced the postoperative hospital stay duration. No significant differences between groups were observed in the incidence of pulmonary complications or overall postoperative morbidity. Conclusions HPC improves postoperative oxygenation, enhances the recovery of pulmonary function, and reduces the duration of hospital stay in patients undergoing thoracoscopic lobectomy. Trial registration This study was registered in the Chinese Clinical Trial Registry (ChiCTR-IPR-17011249) on April 27, 2017.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Anesthesiology, Zhoushan Hospital, No.739 Dingshen Street, Zhoushan, Zhejiang, China
| | - Mo Chen
- Department of Anesthesiology, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, No.242 Guangji Road, Suzhou, Jiangsu, China
| | - Hongbin Li
- Department of Anesthesiology, Zhoushan Hospital, No.739 Dingshen Street, Zhoushan, Zhejiang, China
| | - Jia Yuan
- Department of Anesthesiology, Zhoushan Hospital, No.739 Dingshen Street, Zhoushan, Zhejiang, China
| | - Jingjing Li
- Department of Anesthesiology, Zhoushan Hospital, No.739 Dingshen Street, Zhoushan, Zhejiang, China
| | - Feixiang Wu
- Department of Anesthesiology, Shanghai Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, No.225 Changhai Road, Shanghai, China.
| | - Yan Zhang
- Department of Anesthesiology, Zhoushan Hospital, No.739 Dingshen Street, Zhoushan, Zhejiang, China.
| |
Collapse
|
14
|
|
15
|
Liao WT, Liu J, Zhou SM, Xu G, Gao YQ, Liu WY. UHPLC-QTOFMS-Based Metabolomic Analysis of the Hippocampus in Hypoxia Preconditioned Mouse. Front Physiol 2019; 9:1950. [PMID: 30687133 PMCID: PMC6335317 DOI: 10.3389/fphys.2018.01950] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/22/2018] [Indexed: 01/22/2023] Open
Abstract
Background: Hypoxia appears in a number of extreme environments, including high altitudes, the deep sea, and during aviation, and occurs in cancer, cardiovascular disease, respiratory failures and neurological disorders. Though it is well recognized that hypoxic preconditioning (HPC) exerts endogenous neuroprotective effect against severe hypoxia, the mediators and underlying molecular mechanism for the protective effect are still not fully understood. This study established a hippocampus metabolomics approach to explore the alterations associated with HPC. Methods: In this study, an animal model of HPC was established by exposing the adult BALB/c mice to acute repetitive hypoxia four times. Ultra-high liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-QTOFMS) in combination with univariate and multivariate statistical analyses was employed to deciphering metabolic changes associated with HPC in hippocampus tissue. MetaboAnalyst 3.0 was used to construct HPC related metabolic pathways. Results: The significant metabolic differences in hippocampus between the HPC groups and control were observed, indicating that HPC mouse model was successfully established and HPC could caused significant metabolic changes. Several key metabolic pathways were found to be acutely perturbed, including phenylalanine, tyrosine and tryptophan biosynthesis, taurine and hypotaurine metabolism, phenylalanine metabolism, glutathione metabolism, alanine, aspartate and glutamate metabolism, tyrosine metabolism, tryptophan metabolism, purine metabolism, citrate cycle, and glycerophospholipid metabolism. Conclusion: The results of the present study provided novel insights into the mechanisms involved in the acclimatization of organisms to hypoxia, and demonstrated the neuroprotective mechanism of HPC.
Collapse
Affiliation(s)
- Wen-Ting Liao
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, China
| | - Jie Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Military Medical University, Chongqing, China.,The Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Si-Min Zhou
- The Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China.,Department of High Altitude Military Hygiene, College of High Altitude Military Medicine, Army Military Medical University, Chongqing, China
| | - Gang Xu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Military Medical University, Chongqing, China.,The Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Military Medical University, Chongqing, China.,The Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Wen-Yuan Liu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
16
|
Xu G, Gao YQ, Gao YX, Wu G, Zhang JY, Gao WX. An improved formula for standard hypoxia tolerance time (STT) to evaluate hypoxic tolerance in mice. Mil Med Res 2018; 5:33. [PMID: 30268159 PMCID: PMC6164180 DOI: 10.1186/s40779-018-0180-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 09/12/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypoxia is a primary cause of mountain sickness and a common pathological condition in patients with heart failure, shock, stroke, and chronic obstructive pulmonary disease (COPD). Thus far, little advancement in countering hypoxic damage has been achieved, and one of the main reasons is the absence of an ideal algorithm or calculation method to normalize hypoxia tolerance scores when evaluating an animal model. In this study, we improved a traditional calculation formula for assessment of hypoxia tolerance. METHODS We used a sealed bottle model in which the oxygen is gradually consumed by a mouse inside. To evaluate the hypoxia tolerance of mice, the survival time (ST) of the mouse is recorded and was used to calculate standard hypoxia tolerance time (STT) and adjusted standard hypoxia tolerance time (ASTT). Mice administered with methazolamide and saline were used as positive and negative controls, respectively. RESULTS Since mice were grouped according to either body weight (BW) or bottle volume, we found a strongly negative correlation between STT and BW instead of between STT and bottle volume, suggesting that different BWs could cause false positive or negative errors in the STT results. Furthermore, both false positive and negative errors could be rectified when ASTT was used as the evaluation index. Screening for anti-hypoxic medicines by using mice as the experimental subjects would provide more credible results with the improved ASTT method than with the STT method. CONCLUSION ASTT could be a better index than STT for the evaluation of hypoxia tolerance abilities as it could eliminate the impact of animal BW.
Collapse
Affiliation(s)
- Gang Xu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, 400038, China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, 400038, China
| | - Yi-Xing Gao
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, 400038, China
| | - Gang Wu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, 400038, China
| | - Jian-Yang Zhang
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, 400038, China
| | - Wen-Xiang Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China. .,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, 400038, China.
| |
Collapse
|
17
|
Lu N, Li X, Tan R, An J, Cai Z, Hu X, Wang F, Wang H, Lu C, Lu H. HIF-1α/Beclin1-Mediated Autophagy Is Involved in Neuroprotection Induced by Hypoxic Preconditioning. J Mol Neurosci 2018; 66:238-250. [PMID: 30203298 PMCID: PMC6182618 DOI: 10.1007/s12031-018-1162-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023]
Abstract
Hypoxic preconditioning (HPC) exerts a protective effect against hypoxic/ischemic brain injury, and one mechanism explaining this effect may involve the upregulation of hypoxia-inducible factor-1 (HIF-1). Autophagy, an endogenous protective mechanism against hypoxic/ischemic injury, is correlated with the activation of the HIF-1α/Beclin1 signaling pathway. Based on previous studies, we hypothesize that the protective role of HPC may involve autophagy occurring via activation of the HIF-1α/Beclin1 signaling pathway. To test this hypothesis, we evaluated the effects of HPC on oxygen-glucose deprivation/reperfusion (OGD/R)-induced apoptosis and autophagy in SH-SY5Y cells. HPC significantly attenuated OGD/R-induced apoptosis, and this effect was suppressed by the autophagy inhibitor 3-methyladenine and mimicked by the autophagy agonist rapamycin. In control SH-SY5Y cells, HPC upregulated the expression of HIF-1α and downstream molecules such as BNIP3 and Beclin1. Additionally, HPC increased the LC3-II/LC3-I ratio and decreased p62 levels. The increase in the LC3-II/LC3-I ratio was inhibited by the HIF-1α inhibitor YC-1 or by Beclin1-short hairpin RNA (shRNA). In OGD/R-treated SH-SY5Y cells, HPC also upregulated the expression levels of HIF-1α, BNIP3, and Beclin1, as well as the LC3-II/LC3-I ratio. Furthermore, YC-1 or Beclin1-shRNA attenuated the HPC-mediated cell viability in OGD/R-treated cells. Taken together, our results demonstrate that HPC protects SH-SY5Y cells against OGD/R via HIF-1α/Beclin1-regulated autophagy.
Collapse
Affiliation(s)
- Na Lu
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
- Key Laboratory for the Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Xingxing Li
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Ruolan Tan
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jing An
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Zhenlu Cai
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Xiaoxuan Hu
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Feidi Wang
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Haoruo Wang
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Chengbiao Lu
- Key Laboratory for the Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China
| | - Haixia Lu
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
18
|
Ren C, Li S, Rajah G, Shao G, Lu G, Han R, Huang Q, Li H, Ding Y, Jin K, Ji X. Hypoxia, hibernation and Neuroprotection: An Experimental Study in Mice. Aging Dis 2018; 9:761-768. [PMID: 30090664 PMCID: PMC6065299 DOI: 10.14336/ad.2018.0702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/02/2018] [Indexed: 11/01/2022] Open
Abstract
Hibernation is a unique physiological state that evolved to survive periods of food shortages. It is characterized by profound decreases in metabolic rate, body temperature and physiological functions. Studies have shown that animals in hibernation can resist neurological damage. Here, we aimed to study whether hypoxia can induce a hibernation-like state in a traditionally non-hibernating animal and whether it is neuroprotective. All procedures were conducted according to international guidelines on laboratory animal safety. Mice C57BL/6 (19-21g) were placed into a 125 mL jar with fresh air and the jar was sealed with a rubber plug. For each run, the tolerance limit was judged by the animals' appearance for "air hunger". The animal was removed from the jar as soon as its first gasping breath appeared and was moved to another fresh-air-containing jar of similar volume. This procedure was performed in four runs. The hypoxia exposure significantly decreased oxygen (O2) consumption, carbon dioxide (CO2) production, respiratory rate and heart rate. Meanwhile, rectal temperature reached a minimum of 12.7±2.56°C, which is lower than a wide range of ambient temperatures. The mimicked hibernation decreased the infarct size in a focal cerebral ischemia mouse model. Our findings suggest the possibility of inducing suspended animation-like hibernation states for medical applications post injury.
Collapse
Affiliation(s)
- Changhong Ren
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Gary Rajah
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Guo Shao
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Guowei Lu
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Rongrong Han
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Qingjian Huang
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Haiyan Li
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
| | - Yuchuan Ding
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kunlin Jin
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Translational Medicine, Beijing 100053, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing 100069, China
| |
Collapse
|
19
|
Almohanna AM, Wray S. Hypoxic conditioning in blood vessels and smooth muscle tissues: effects on function, mechanisms, and unknowns. Am J Physiol Heart Circ Physiol 2018; 315:H756-H770. [PMID: 29702009 DOI: 10.1152/ajpheart.00725.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hypoxic preconditioning, the protective effect of brief, intermittent hypoxic or ischemic episodes on subsequent more severe hypoxic episodes, has been known for 30 yr from studies on cardiac muscle. The concept of hypoxic preconditioning has expanded; excitingly, organs beyond the heart, including the brain, liver, and kidney, also benefit. Preconditioning of vascular and visceral smooth muscles has received less attention despite their obvious importance to health. In addition, there has been no attempt to synthesize the literature in this field. Therefore, in addition to overviewing the current understanding of hypoxic conditioning, in the present review, we consider the role of blood vessels in conditioning and explore evidence for conditioning in other smooth muscles. Where possible, we have distinguished effects on myocytes from other cell types in the visceral organs. We found evidence of a pivotal role for blood vessels in conditioning and for conditioning in other smooth muscle, including the bladder, vascular myocytes, and gastrointestinal tract, and a novel response in the uterus of a hypoxic-induced force increase, which helps maintain contractions during labor. To date, however, there are insufficient data to provide a comprehensive or unifying mechanism for smooth muscles or visceral organs and the effects of conditioning on their function. This also means that no firm conclusions can be drawn as to how differences between smooth muscles in metabolic and contractile activity may contribute to conditioning. Therefore, we have suggested what may be general mechanisms of conditioning occurring in all smooth muscles and tabulated tissue-specific mechanistic findings and suggested ideas for further progress.
Collapse
Affiliation(s)
- Asmaa M Almohanna
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine University of Liverpool , Liverpool , United Kingdom.,Princess Nourah bint Abdulrahman University , Riyadh , Saudi Arabia
| | - Susan Wray
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine University of Liverpool , Liverpool , United Kingdom
| |
Collapse
|
20
|
Cheng H, Cui C, Lu S, Xia B, Li X, Xu P, Xue M. Identification and analysis of hub genes and networks related to hypoxia preconditioning in mice (No 035215). Oncotarget 2017; 9:11889-11904. [PMID: 29552280 PMCID: PMC5844716 DOI: 10.18632/oncotarget.23555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/28/2017] [Indexed: 11/25/2022] Open
Abstract
Hypoxia preconditioning is an effective strategy of intrinsic cell protection. An acute repetitive hypoxic mice model was developed. High-throughput microarray analysis was performed to explore the integrative alterations of gene expression in repetitive hypoxic mice. Data obtained was analyzed via multiple bioinformatics approaches to identify the hub genes, pathways and biological processes related to hypoxia preconditioning. The current study, for the first time, provides insights into the gene expression profiles in repetitive hypoxic mice. It was found that a total of 1175 genes expressed differentially between the hypoxic mice and normal mice. Overall, 113 significantly up-regulated and 138 significantly down-regulated functions were identified from the differentially expressed genes in repetitive hypoxic brains. Among them, at least fourteen of these genes were very associated with hypoxia preconditioning. The change trends of these genes were validated by reverse-transcription polymerase chain reaction and were found to be consistent with the microarray data. Combined the results of pathway and gene co-expression networks, we defined Plcb1, Cacna2d1, Atp2b4, Grin2a, Grin2b and Glra1 as the main hub genes tightly related with hypoxia preconditioning. The differential functions mainly included the mitogen-activated protein kinase pathway and ion or neurotransmitter transport. The multiple reactions in cell could be initiated by activating MAPK pathway to prevent hypoxia damage. Plcb1 was an important and hub gene and node in the hypoxia preconditioning signal networks. The findings in the hub genes and integrated gene networks provide very useful information for further exploring the molecular mechanisms of hypoxia preconditioning.
Collapse
Affiliation(s)
- Haiting Cheng
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,Department of Pharmacy, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Can Cui
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shousi Lu
- China Rehabilitation Research Center, Beijing 100068, China
| | - Binbin Xia
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaorong Li
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Pinxiang Xu
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Ming Xue
- Department of Pharmacology, Beijing Laboratory for Biomedical Detection Technology and Instrument, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
21
|
Liu J, Zhan G, Chen D, Chen J, Yuan ZB, Zhang EL, Gao YX, Xu G, Sun BD, Liao W, Gao YQ. UPLC‑QTOFMS‑based metabolomic analysis of the serum of hypoxic preconditioning mice. Mol Med Rep 2017; 16:6828-6836. [PMID: 28901489 PMCID: PMC5865841 DOI: 10.3892/mmr.2017.7493] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/24/2017] [Indexed: 01/06/2023] Open
Abstract
Hypoxic preconditioning (HPC) is well‑known to exert a protective effect against hypoxic injury; however, the underlying molecular mechanism remains unclear. The present study utilized a serum metabolomics approach to detect the alterations associated with HPC. In the present study, an animal model of HPC was established by exposing adult BALB/c mice to acute repetitive hypoxia four times. The serum samples were collected by orbital blood sampling. Metabolite profiling was performed using ultra‑performance liquid chromatography‑quadrupole time‑of‑flight mass spectrometry (UPLC‑QTOFMS), in conjunction with univariate and multivariate statistical analyses. The results of the present study confirmed that the HPC mouse model was established and refined, suggesting significant differences between the control and HPC groups at the molecular levels. HPC caused significant metabolic alterations, as represented by the significant upregulation of valine, methionine, tyrosine, isoleucine, phenylalanine, lysophosphatidylcholine (LysoPC; 16:1), LysoPC (22:6), linoelaidylcarnitine, palmitoylcarnitine, octadecenoylcarnitine, taurine, arachidonic acid, linoleic acid, oleic acid and palmitic acid, and the downregulation of acetylcarnitine, malate, citrate and succinate. Using MetaboAnalyst 3.0, a number of key metabolic pathways were observed to be acutely perturbed, including valine, leucine and isoleucine biosynthesis, in addition to taurine, hypotaurine, phenylalanine, linoleic acid and arachidonic acid metabolism. The results of the present study provided novel insights into the mechanisms involved in the acclimatization of organisms to hypoxia, and demonstrated the protective mechanism of HPC.
Collapse
Affiliation(s)
- Jie Liu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Gang Zhan
- Key Laboratory of High Altitude Medicine, Ministry of Education, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Dewei Chen
- Key Laboratory of High Altitude Medicine, Ministry of Education, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jian Chen
- Key Laboratory of High Altitude Medicine, Ministry of Education, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Zhi-Bin Yuan
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Er-Long Zhang
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yi-Xing Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Gang Xu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Bing-Da Sun
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| | - Wenting Liao
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
22
|
Amaral-Silva LD, Scarpellini CDS, Toro-Velasquez PA, Fernandes MH, Gargaglioni LH, Bícego KC. Hypoxia during embryonic development increases energy metabolism in normoxic juvenile chicks. Comp Biochem Physiol A Mol Integr Physiol 2017; 207:93-99. [DOI: 10.1016/j.cbpa.2017.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 03/01/2017] [Accepted: 03/05/2017] [Indexed: 01/11/2023]
|
23
|
Li S, Hafeez A, Noorulla F, Geng X, Shao G, Ren C, Lu G, Zhao H, Ding Y, Ji X. Preconditioning in neuroprotection: From hypoxia to ischemia. Prog Neurobiol 2017; 157:79-91. [PMID: 28110083 DOI: 10.1016/j.pneurobio.2017.01.001] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/08/2017] [Accepted: 01/13/2017] [Indexed: 01/05/2023]
Abstract
Sublethal hypoxic or ischemic events can improve the tolerance of tissues, organs, and even organisms from subsequent lethal injury caused by hypoxia or ischemia. This phenomenon has been termed hypoxic or ischemic preconditioning (HPC or IPC) and is well established in the heart and the brain. This review aims to discuss HPC and IPC with respect to their historical development and advancements in our understanding of the neurochemical basis for their neuroprotective role. Through decades of collaborative research and studies of HPC and IPC in other organ systems, our understanding of HPC and IPC-induced neuroprotection has expanded to include: early- (phosphorylation targets, transporter regulation, interfering RNA) and late- (regulation of genes like EPO, VEGF, and iNOS) phase changes, regulators of programmed cell death, members of metabolic pathways, receptor modulators, and many other novel targets. The rapid acceleration in our understanding of HPC and IPC will help facilitate transition into the clinical setting.
Collapse
Affiliation(s)
- Sijie Li
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Adam Hafeez
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Fatima Noorulla
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA; Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
| | - Guo Shao
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Guowei Lu
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Heng Zhao
- Department of Neurosurgery, Stanford University, CA, USA
| | - Yuchuan Ding
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China.
| |
Collapse
|
24
|
Zhou T, Wang M, Cheng H, Cui C, Su S, Xu P, Xue M. UPLC-HRMS based metabolomics reveals the sphingolipids with long fatty chains and olefinic bonds up-regulated in metabolic pathway for hypoxia preconditioning. Chem Biol Interact 2015; 242:145-52. [DOI: 10.1016/j.cbi.2015.09.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 09/02/2015] [Accepted: 09/28/2015] [Indexed: 10/23/2022]
|
25
|
Zhang YB, Guo ZD, Li MY, Li SJ, Niu JZ, Yang MF, Ji XM, Lv GW. Cerebrospinal fluid from rats given hypoxic preconditioning protects neurons from oxygen-glucose deprivation-induced injury. Neural Regen Res 2015; 10:1471-6. [PMID: 26604909 PMCID: PMC4625514 DOI: 10.4103/1673-5374.165519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Hypoxic preconditioning activates endogenous mechanisms that protect against cerebral ischemic and hypoxic injury. To better understand these protective mechanisms, adult rats were housed in a hypoxic environment (8% O2/92% N2) for 3 hours, and then in a normal oxygen environment for 12 hours. Their cerebrospinal fluid was obtained to culture cortical neurons from newborn rats for 1 day, and then the neurons were exposed to oxygen-glucose deprivation for 1.5 hours. The cerebrospinal fluid from rats subjected to hypoxic preconditioning reduced oxygen-glucose deprivation-induced injury, increased survival rate, upregulated Bcl-2 expression and downregulated Bax expression in the cultured cortical neurons, compared with control. These results indicate that cerebrospinal fluid from rats given hypoxic preconditioning protects against oxygen-glucose deprivation-induced injury by affecting apoptosis-related protein expression in neurons from newborn rats.
Collapse
Affiliation(s)
- Yan-Bo Zhang
- Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian, Shandong Province, China
| | - Zheng-Dong Guo
- Department of Endocrinology, Affiliated Hospital of Taishan Medical University, Taian, Shandong Province, China
| | - Mei-Yi Li
- Department of Neurology, Shandong Taishan Chronic Disease Hospital, Taian, Shandong Province, China
| | - Si-Jie Li
- Hypoxia Medical Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing-Zhong Niu
- Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian, Shandong Province, China
| | - Ming-Feng Yang
- Department of Neurology, Affiliated Hospital of Taishan Medical University, Taian, Shandong Province, China
| | - Xun-Ming Ji
- Hypoxia Medical Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Guo-Wei Lv
- Hypoxia Medical Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
|
27
|
Zhang S, Zhang Y, Jiang S, Liu Y, Huang L, Zhang T, Lu G, Gong K, Ji X, Shao G. The effect of hypoxia preconditioning on DNA methyltransferase and PP1γ in hippocampus of hypoxia preconditioned mice. High Alt Med Biol 2014; 15:483-90. [PMID: 25531462 DOI: 10.1089/ham.2014.1042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It is well known that hypoxia preconditioning can increase hypoxic tolerance by changing the expressions of some genes in the brain. DNA methylation is important for regulating gene expression and is catalyzed by DNA methyltransferase (DNMT), an enzyme that is abundant in the brain. However, the impact of hypoxia preconditioning on DNA methylation remains unknown. In the current study, mice were randomly divided into three groups: blank control group with no exposure to hypoxia (H0), the hypoxia control group exposed to hypoxia once (H1), and the hypoxia preconditioning group exposed to 4 runs of hypoxia (H4). The mRNA and protein levels of three kinds of DNMTs and the activity of total DMNT were measured. Protein phosphatase 1(PP1)γ, which critically regulates neuroprotective pathways in brain, was measured in mRNA and protein activity and promoter methylation. DNMT1 was unchanged in H1 and H4, while DNMT3A and DNMT3B were decreased in H4. The mRNA and protein levels of PP1γ were decreased in H4. However, there was no detectable change in the level of DNA methylation of the promoter of PP1γ (-321 bp to 95 bp). These findings suggest that DNA methylation may have a role in hypoxia neuroprotection, and the change of PP1γ, which did not depend on the change of its promoter (-321 bp to 95bp) DNA methylation, may be involved in neuroprotection.
Collapse
Affiliation(s)
- Shu Zhang
- 1 Biomedicine Research Center and Basic Medical College, The First Affiliated Hospital of BaoTou Medical College , Inner Mongolia, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zhang YB, Wang X, Meister EA, Gong KR, Yan SC, Lu GW, Ji XM, Shao G. The effects of CoCl2 on HIF-1α protein under experimental conditions of autoprogressive hypoxia using mouse models. Int J Mol Sci 2014; 15:10999-1012. [PMID: 24945310 PMCID: PMC4100194 DOI: 10.3390/ijms150610999] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 12/22/2022] Open
Abstract
It is well known that cobalt chloride (CoCl2) can enhance the stability of hypoxia-inducible factor (HIF)-1α. The aim of this study is to detect the effect of CoCl2 on the hypoxia tolerance of mice which were repeatedly exposed to autoprogressive hypoxia. Balb/c mice were randomly divided into groups of chemical pretreatment and normal saline (NS), respectively injected with CoCl2 and NS 3 h before exposure to hypoxia for 0 run (H0), 1 run (H1), and 4 runs (H4). Western Blot, electrophoretic mobility shift assay (EMSA), extracellular recordings population spikes in area cornus ammonis I (CA 1) of mouse hippocampal slices and real-time were used in this study. Our results demonstrated that the tolerance of mice to hypoxia, the changes of HIF-1α protein level and HIF-1 DNA binding activity in mice hippocampus, the mRNA level of erythropoietin (EPO) and vascular endothelial growth factor (VEGF), and the disappearance time of population spikes of hippocampal slices were substantially different between the control group and the CoCl2 group. Over-induction of HIF-1α by pretreatment with CoCl2 before hypoxia did not increase the hypoxia tolerance.
Collapse
Affiliation(s)
- Yan-Bo Zhang
- Department of Neurology, Affiliated Hospital of Tai Shan Medical University, Taishan 271000, China.
| | - Xiulian Wang
- Department of Intensive Care Unit , 2nd Affiliated Hospital of Baotou Medical College, Baotou 014030, China.
| | - Edward A Meister
- Department of Medicine, University of Arizona, Tucson, AZ 85721, USA.
| | - Ke-Rui Gong
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou 014060, China.
| | - Shao-Chun Yan
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou 014060, China.
| | - Guo-Wei Lu
- Institute for Hypoxia Medicine, Xuanwu Hospital of Capital Medical University, Beijing 10054, China.
| | - Xun-Ming Ji
- Institute for Hypoxia Medicine, Xuanwu Hospital of Capital Medical University, Beijing 10054, China.
| | - Guo Shao
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou 014060, China.
| |
Collapse
|
29
|
|
30
|
Nathaniel TI, Otukonyong EE, Okon M, Chaves J, Cochran T, Nathaniel AI. Metabolic regulatory clues from the naked mole rat: toward brain regulatory functions during stroke. Brain Res Bull 2013; 98:44-52. [PMID: 23886571 DOI: 10.1016/j.brainresbull.2013.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/03/2013] [Accepted: 07/15/2013] [Indexed: 12/30/2022]
Abstract
Resistance to tissue hypoxia is a robust fundamental adaptation to low oxygen supply, and represents a novel neuroscience problem with significance to mammalian physiology as well as human health. With the underlying mechanisms strongly conserved in evolution, the ability to resist tissue hypoxia in natural systems has recently emerged as an interesting model in mammalian physiology research to understand mechanisms that can be manipulated for the clinical management of stroke. The extraordinary ability to resist tissue hypoxia by the naked mole rat (NMR) indicates the presence of a unique mechanism that underlies the remarkable healthy life span and exceptional hypoxia resistance. This opens an interesting line of research into understanding the mechanisms employed by the naked mole rat (Heterocephalus glaber) to protect the brain during hypoxia. In a series of studies, we first examined the presence of neuroprotection in the brain cells of naked mole rats (NMRs) subjected to hypoxic insults, and then characterized the expression of such neuroprotection in a wide range of time intervals. We used oxygen nutrient deprivation (OND), an in vitro model of resistance to tissue hypoxia to determine whether there is evidence of neuronal survival in the hippocampal (CA1) slices of NMRs that are subjected to chronic hypoxia. Hippocampus neurons of NMRs that were kept in hypoxic condition consistently tolerated OND right from the onset time of 5h. This tolerance was maintained for 24h. This finding indicates that there is evidence of resistance to tissue hypoxia by CA1 neurons of NMRs. We further examined the effect of hypoxia on metabolic rate in the NMR. Repeated measurement of metabolic rates during exposure of naked mole rats to hypoxia over a constant ambient temperature indicates that hypoxia significantly decreased metabolic rates in the NMR, suggesting that the observed decline in metabolic rate during hypoxia may contribute to the adaptive mechanism used by the NMR to resist tissue hypoxia. This work is aimed to contribute to the understanding of mechanisms of resistance to tissue hypoxia in the NMR as an important life-sustaining process, which can be translated into therapeutic interventions during stroke.
Collapse
Affiliation(s)
- Thomas I Nathaniel
- University of South Carolina School of Medicine, HSEB, 607 Grove Road, Greenville, SC 29605, United States.
| | | | | | | | | | | |
Collapse
|
31
|
In vivo hypoxic preconditioning protects from warm liver ischemia-reperfusion injury through the adenosine A2B receptor. Transplantation 2013; 94:894-902. [PMID: 23073466 DOI: 10.1097/tp.0b013e31826a9a46] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Liver ischemia-reperfusion injury (IRI) is a known risk factor for the postoperative outcome of patients undergoing liver surgery/transplantation. Attempts to protect from organ damage require multidisciplinary strategies and are of emerging interest in view of patients with higher age and American Society of Anesthesiology status. Ischemic preconditioning has been successfully applied to prevent from IRI during liver resection/transplantation. Because even short periods of ischemia during preconditioning inevitably lead to hypoxia and formation of anti-inflammatory/cytoprotective acting adenosine, we reasoned that short nonischemic hypoxia also protects against hepatic IRI. METHODS Mice underwent hypoxic preconditioning (HPC) by breathing 10% oxygen for 10 min followed by 10 min of 21% oxygen before left liver lobe ischemia (45 min) and reperfusion (4 hr). The interactions of hypoxia→adenosine→adenosine receptors were tested by pharmacologic antagonism at adenosine receptor (AR) sites in wild-type mice and in mice with genetic deletions at the A1, A2A, A2B, and A3 ARs. Hepatocellular damage, inflammation, and metabolic effects were quantified by enzyme activities, cytokines, liver myeloperoxidase, blood adenosine, and tissue AMP, respectively. RESULTS Hepatoprotection by HPC was significant in wild-type and A1, A2A, and A3 AR knockout mice as quantified by lower alanine aminotransferase serum activities, cytokine levels, histologic damage scores, tissue myeloperoxidase concentrations, and preserved AMP concentrations. Protection by HPC was blunted in mice pretreated with the A2B AR antagonist MRS1754 or in A2B AR knockout mice. CONCLUSIONS Because liver protective effects of HPC are negated when the A2B receptor is nonfunctional, the hypoxia→adenosine→A2B receptor pathway plays a critical role in the prevention of warm IRI in vivo. Hypoxic activation of this pathway warrants use of selective A2B AR agonists or even intermittent hypoxia (e.g., in deceased organ donors) to protect from liver IRI.
Collapse
|
32
|
Shao G, Lu GW. Hypoxic preconditioning in an autohypoxic animal model. Neurosci Bull 2012; 28:316-20. [PMID: 22622832 PMCID: PMC5560319 DOI: 10.1007/s12264-012-1222-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 12/30/2011] [Indexed: 11/26/2022] Open
Abstract
Hypoxic preconditioning refers to the exposure of organisms, systems, organs, tissues or cells to moderate hypoxia/ischemia that results in increased resistance to a subsequent episode of severe hypoxia/ischemia. In this article, we review recent research based on a mouse model of repeated exposure to autohypoxia. Pre-exposure markedly increases the tolerance to or protection against hypoxic insult, and preserves the cellular structure of the brain. Furthermore, the hippocampal activity amplitude and frequency of electroencephalogram, latency of cortical somatosensory-evoked potential and spinal somatosensory-evoked potential progressively decrease, while spatial learning and memory improve. In the brain, detrimental neurochemicals such as free radicals are down-regulated, while beneficial ones such as adenosine are up-regulated. Also, antihypoxia factor(s) and gene(s) are activated. We propose that the tolerance and protective effects depend on energy conservation and plasticity triggered by exposure to hypoxia via oxygen-sensing transduction pathways and hypoxia-inducible factor-initiated cascades. A potential path for further research is the development of devices and pharmaceuticals acting on antihypoxia factor(s) and gene(s) for the prevention and treatment of hypoxia and related syndromes.
Collapse
Affiliation(s)
- Guo Shao
- Medical School, Lishui University, Lishui, 323000 China
- Institute for Hypoxia Medicine, Capital Medical University, Beijing, 100069 China
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou, 014030 China
| | - Guo-Wei Lu
- Institute for Hypoxia Medicine, Capital Medical University, Beijing, 100069 China
- School of Basic Medical Sciences, Department of Neurobiology, Capital Medical University, Beijing, 100069 China
| |
Collapse
|
33
|
Aviles-Reyes RX, Angelo MF, Villarreal A, Rios H, Lazarowski A, Ramos AJ. Intermittent hypoxia during sleep induces reactive gliosis and limited neuronal death in rats: implications for sleep apnea. J Neurochem 2010; 112:854-69. [DOI: 10.1111/j.1471-4159.2009.06535.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
34
|
Mild hypobaric hypoxia preconditioning up-regulates expression of transcription factors c-Fos and NGFI-A in rat neocortex and hippocampus. Neurosci Res 2009; 65:360-6. [PMID: 19723547 DOI: 10.1016/j.neures.2009.08.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 08/10/2009] [Accepted: 08/24/2009] [Indexed: 01/19/2023]
Abstract
Transcription factors c-Fos and NGFI-A encoded by immediate early genes largely participate in the biochemical cascade leading to genomically driven lasting adaptation by neurons to injurious exposures including hypoxia/ischemia. Present study was designed to examine the involvement of c-Fos and NGFI-A in the development of brain hypoxic tolerance induced by mild hypoxic preconditioning. Earlier we have reported that preconditioning by repetitive mild hypobaric hypoxia (MHH) considerably increases neuronal resistance to subsequent severe injurious exposures. Herein, changes of c-Fos and NGFI-A expression in vulnerable rat brain areas (hippocampus, neocortex) in response to preconditioning MHH itself were studied using quantitative immunocytochemistry. Exposure to MHH differentially enhanced c-Fos and NGFI-A expression in neocortex and hippocampal fields 3-24h following the last MHH trial. The c-Fos up-regulation was the most pronounced in neocortex, CA1, and dentate gyrus, but it was twice lower in CA3/CA4. The up-regulation of NGFI-A in CA1, dentate gyrus and neocortex was 1.5-2-fold lower than that of c-Fos; but in CA3 and CA4 the rates of the c-Fos and NGFI-A induction were comparable. The present findings indicate that cooperative but differential activation of c-Fos and NGFI-A expression in vulnerable brain areas contribute to the development of tolerance achieved by MHH preconditioning.
Collapse
|
35
|
Abstract
Oxygen is essential for the maintenance of life, and when oxygen levels decline to critical levels, a program of complex mechanisms exists to i) sense hypoxia, ii) respond to minimize acute tissue injury, and iii) result in adaptations that offer protection against further hypoxia challenges. Alternative adaptation-related protection may also be inducible through the increased activity of hypoxia-inducible factors activated by hypoxia mimics such as iron chelation with deferoxamine (DFA). We have characterized a set of hypoxia-related responses at the microvasculature and postulated that microvascular injury in response to hypoxia could be reproduced by the reduction of bioavailable iron through chelation by DFA. We were able to induce a similar degree of leukocyte adherence and emigration and vascular leak with DFA infusion as compared with hypoxia exposure in an intact physiological rodent model. However, in contrast to hypoxia-exposed groups, we were unable to detect reactive oxygen species or alter the injury pattern with reactive oxygen species scavenger in the groups treated with DFA. Thus, we demonstrate that DFA mimics the pattern and intensity of hypoxia-related injury on the microvasculature; however, differences in the time course and mechanism of injury were identified. In addition, DFA saturated with iron did not completely reverse the effects of DFA, suggesting a mechanism(s) beyond a reduction in the bioavailability of iron. These findings may have importance in the targeting of iron for the development of hypoxia mimics that may offer protection against subsequent hypoxia exposure in clinical setting such as myocardial infarction and stroke.
Collapse
|
36
|
Yao SY, Soutto M, Sriram S. Preconditioning with cobalt chloride or desferrioxamine protects oligodendrocyte cell line (MO3.13) from tumor necrosis factor-alpha-mediated cell death. J Neurosci Res 2008; 86:2403-13. [PMID: 18438939 DOI: 10.1002/jnr.21697] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hypoxia-inducible factor-1alpha (HIF-1alpha) is a transcription factor induced under hypoxic conditions. HIF-1alpha promotes the expression of genes encoding proteins that increase the cellular supply of oxygen and promote survival in periods of cellular stress and availability of cellular energy. We examined the effect of desferrioxamine (DFO) and cobalt chloride (CoCl(2)), two agents known to increase the stability of HIF-1alpha, and its effect on the survivability of an oligodendroglial cell line, MO3.13, when cultured with tumor necrosis factor-alpha (TNFalpha). Our studies showed that, unlike a murine microglial cell line (BV-2), MO3.13 cells do not induce HIF-1alpha in the presence of TNFalpha. MO3.13 cells do stabilize HIF-1alpha in the presence of DFO or CoCl(2). When MO3.13 cell were preconditioned with either DFO or CoCl(2), addition of TNFalpha further increased protein levels of HIF-1alpha. The mechanisms that underlie the increase in protein levels of HIF -1alpha seen, following addition of TNFalpha in preconditioned cells is due to an increase in transcription of the HIF-1alpha gene. Increased cellular levels of HIF-1alpha is associated with improved survival of MO3.13 cells, when cultured with TNFalpha after a period of preconditioning by DFO or CoCl(2). These studies suggest that compoundsthat increase HIF-1alpha can function as neuroprotective agents in inflammatory disorders of the CNS.
Collapse
Affiliation(s)
- Song-yi Yao
- Department of Neurology, Multiple Sclerosis Research Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, USA
| | | | | |
Collapse
|
37
|
Weller ML, Stone IM, Goss A, Rau T, Rova C, Poulsen DJ. Selective overexpression of excitatory amino acid transporter 2 (EAAT2) in astrocytes enhances neuroprotection from moderate but not severe hypoxia-ischemia. Neuroscience 2008; 155:1204-11. [PMID: 18620031 DOI: 10.1016/j.neuroscience.2008.05.059] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Accepted: 05/26/2008] [Indexed: 02/08/2023]
Abstract
Attempts have been made to elevate excitatory amino acid transporter 2 (EAAT2) expression in an effort to compensate for loss of function and expression associated with disease or pathology. Increased EAAT2 expression has been noted following treatment with beta-lactam antibiotics, and during ischemic preconditioning (IPC). However, both of these conditions induce multiple changes in addition to alterations in EAAT2 expression that could potentially contribute to neuroprotection. Therefore, the aim of this study was to selectively overexpress EAAT2 in astrocytes and characterize the cell type specific contribution of this transporter to neuroprotection. To accomplish this we used a recombinant adeno-associated virus vector, AAV1-glial fibrillary acidic protein (GFAP)-EAAT2, designed to selectively drive the overexpression of EAAT2 within astrocytes. Both viral-mediated gene delivery and beta-lactam antibiotic (penicillin-G) treatment of rat hippocampal slice cultures resulted in a significant increase in both the expression of EAAT2, and dihydrokainate (DHK) sensitive glutamate uptake. Penicillin-G provided significant neuroprotection in rat hippocampal slice cultures under conditions of both moderate and severe oxygen glucose deprivation (OGD). In contrast, viral-mediated overexpression of EAAT2 in astrocytes provided enhanced neuroprotection only following a moderate OGD insult. These results indicate that functional EAAT2 can be selectively overexpressed in astrocytes, leading to enhanced neuroprotection. However, this cell type specific increase in EAAT2 expression offers only limited protection compared to treatment with penicillin-G.
Collapse
Affiliation(s)
- M L Weller
- NIH COBRE Center for Structural and Functional Neuroscience, Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Rybnikova E, Gluschenko T, Tulkova E, Churilova A, Jaroshevich O, Baranova K, Samoilov M. Preconditioning induces prolonged expression of transcription factors pCREB and NF-kappa B in the neocortex of rats before and following severe hypobaric hypoxia. J Neurochem 2008; 106:1450-8. [PMID: 18547368 DOI: 10.1111/j.1471-4159.2008.05516.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Preconditioning using mild repetitive hypobaric hypoxia is known to increase a tolerance of brain neurons to severe hypoxia and other injurious exposures. In the present study, the effects of mild hypoxic preconditioning on the expression of transcription factors NF-kappaB and phosphorylated CREB (pCREB) has been studied in the neocortex of rats exposed to severe hypobaric hypoxia. As revealed by quantitative immunocytochemistry, the injurious severe hypobaric hypoxia (180 Torr, 3 h) remarkably reduced the neocortical levels of pCREB and NF-kappaB. The three-trial hypoxic preconditioning (360 Torr, 2 h, 3 days) induced persistent up-regulation of pCREB and NF-kappaB expression in the neocortex of rats 3-24 h following the severe hypoxia. In addition, the preconditioning alone which was not followed by the severe hypoxia, considerably increased neocortical pCREB and NF-kappaB levels. The findings suggest a role for transcription factors cAMP response element-binding protein and NF-kappaB in the neuroprotective mechanisms activated by the hypoxic preconditioning.
Collapse
Affiliation(s)
- Elena Rybnikova
- Laboratory of Neuroendocrinology, Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russian Federation.
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Adaptation is one of physiology's fundamental tenets, operating not only at the level of species, as Darwin proposed, but also at the level of tissues, cells, molecules and, perhaps, genes. During recent years, stroke neurobiologists have advanced a considerable body of evidence supporting the hypothesis that, with experimental coaxing, the mammalian brain can adapt to injurious insults such as cerebral ischaemia to promote cell survival in the face of subsequent injury. Establishing this protective phenotype in response to stress depends on a coordinated response at the genomic, molecular, cellular and tissue levels. Here, I summarize our current understanding of how 'preconditioning' stimuli trigger a cerebroprotective state known as cerebral 'ischaemic tolerance'.
Collapse
Affiliation(s)
- Jeffrey M Gidday
- Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri 63110, USA.
| |
Collapse
|
40
|
|