51
|
Baranova KA, Rybnikova EA, Samoilov MO. The dynamics of HIF-1α expression in the rat brain at different stages of experimental posttraumatic stress disorder and its correction with moderate hypoxia. NEUROCHEM J+ 2017. [DOI: 10.1134/s1819712417020027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
52
|
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: 156] [Impact Index Per Article: 19.5] [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
|
53
|
Yu J, Liu XL, Cheng QG, Lu SS, Xu XQ, Zu QQ, Liu S. G-CSF and hypoxic conditioning improve the proliferation, neural differentiation and migration of canine bone marrow mesenchymal stem cells. Exp Ther Med 2016; 12:1822-1828. [PMID: 27588100 DOI: 10.3892/etm.2016.3535] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/23/2016] [Indexed: 12/15/2022] Open
Abstract
Transplantation using bone marrow mesenchymal stem cells (BMSCs) is emerging as a potential regenerative therapy after ischemic attacks in the brain. However, it has been questioned because very few transplanted BMSCs are detected homing to and survived in the ischemic region. Improving the cell viability and migration ability under the complex ischemic condition seems very important. The aim of our study is to identify whether hypoxic condition and granulocyte colony-stimulating factor (G-CSF) could improve the cell survival and migration ability of transplanted cells or hypoxic condition could promote BMSC's neural differentiation. BMSCs were treated under either normoxic (21% O2) or hypoxic (1% O2) (HP-BMSCs) conditions, no significant apoptosis was observed in hypoxic precondition (HP) group, our study confirmed that HP improves BMSCs proliferation and migration. Meanwhile, neural induction of BMSCs under hypoxic condition exhibited significant superior results than normoxic condition. Additionally, the addition of G-CSF in HP-BMSCs culture media promoted HP efficiency on BMSCs. These findings shed light on novel efficient strategy on the prosperity of BMSCs. Hypoxic preconditioning and cultured with G-CSF may become a promising therapeutics for cell-based therapy in the treatments of ischemia stroke.
Collapse
Affiliation(s)
- Jing Yu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xing-Long Liu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qi-Guang Cheng
- Department of Radiology, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430074, P.R. China
| | - Shan-Shan Lu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiao-Quan Xu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qing-Quan Zu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Sheng Liu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
54
|
Singh A, Singh A, Sen D. Mesenchymal stem cells in cardiac regeneration: a detailed progress report of the last 6 years (2010-2015). Stem Cell Res Ther 2016; 7:82. [PMID: 27259550 PMCID: PMC4893234 DOI: 10.1186/s13287-016-0341-0] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells have been used for cardiovascular regenerative therapy for decades. These cells have been established as one of the potential therapeutic agents, following several tests in animal models and clinical trials. In the process, various sources of mesenchymal stem cells have been identified which help in cardiac regeneration by either revitalizing the cardiac stem cells or revascularizing the arteries and veins of the heart. Although mesenchymal cell therapy has achieved considerable admiration, some challenges still remain that need to be overcome in order to establish it as a successful technique. This in-depth review is an attempt to summarize the major sources of mesenchymal stem cells involved in myocardial regeneration, the significant mechanisms involved in the process with a focus on studies (human and animal) conducted in the last 6 years and the challenges that remain to be addressed.
Collapse
Affiliation(s)
- Aastha Singh
- School of Bio Sciences and Technology, VIT University, Vellore, India
| | - Abhishek Singh
- School of Bio Sciences and Technology, VIT University, Vellore, India
| | - Dwaipayan Sen
- School of Bio Sciences and Technology, VIT University, Vellore, India. .,Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), VIT University, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
55
|
Revisiting cobalt chloride preconditioning to prevent hypobaric hypoxia-induced damage: identification of global proteomic alteration and key networks. Funct Integr Genomics 2016; 16:281-95. [PMID: 26882918 DOI: 10.1007/s10142-016-0483-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/24/2016] [Accepted: 02/02/2016] [Indexed: 10/22/2022]
Abstract
Several studies have supported the hypoxia mimetic roles and cytoprotective properties of cobalt chloride in vitro and in vivo. However, a clear understanding of biological process-based mechanism that integrates the available information remains unknown. This study was aimed to explore the potential mechanism of cobalt chloride deciphering its benefits and well-known physiological challenge caused by hypobaric hypoxia that reportedly affects nearly 24 % of the global population. In order to explore the mechanism of CoCl2, we used global proteomic and systems biology approach in rat model to provide a deeper insight into molecular mechanisms of preconditioning. Furthermore, key conclusions were drawn based on biological network analysis and their enrichment with ontological overlaps. The study was further strengthened by consistent identification of validation of proteins using immunoblotting. CoCl2-pretreated animals exposed to hypoxia showed two significant networks, one lipid metabolism and other cell cycle associated, with a total score of 23 and eight focus molecules. In this study, we delineated two primary routes: one, by direct modulation of reactive oxygen species metabolism and, second, by regulation of lipid metabolism which was not known until now. The previously known benefits of cobalt chloride during physiological challenge by hypobaric hypoxia are convincing and could be explained by some basic set of metabolic and molecular reorganization within the hypoxia model. Interestingly, we also observed some of the completely unknown roles of cobalt chloride such as regulation of lipid that could undulate the translational roles of cobalt chloride supplementation beyond hypoxia preconditioning.
Collapse
|
56
|
Kapilevich LV, Kironenko TA, Zaharova AN, Kotelevtsev YV, Dulin NO, Orlov SN. Skeletal muscle as an endocrine organ: Role of [Na +] i/[K +] i-mediated excitation-transcription coupling. Genes Dis 2015; 2:328-336. [PMID: 27610402 PMCID: PMC5012537 DOI: 10.1016/j.gendis.2015.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/21/2015] [Indexed: 01/20/2023] Open
Abstract
During the last two decades numerous research teams demonstrated that skeletal muscles function as an exercise-dependent endocrine organ secreting dozens of myokines. Variety of physiological and pathophysiological implications of skeletal muscle myokines secretion has been described; however, upstream signals and sensing mechanisms underlying this phenomenon remain poorly understood. It is well documented that in skeletal muscles intensive exercise triggers dissipation of transmembrane gradient of monovalent cations caused by permanent activation of voltage-gated Na+ and K+ channels. Recently, we demonstrated that sustained elevation of the [Na+]i/[K+]i ratio triggers expression of dozens ubiquitous genes including several canonical myokines, such as interleukin 6 and cyclooxygenase 2, in the presence of intra- and extracellular Ca2+ chelators. These data allowed us to suggest a novel [Na+]i/[K+]i-sensitive, Ca2+i-independent mechanism of excitation-transcription coupling which triggers myokine production. This pathway exists in parallel with canonical signaling mediated by Ca2+i, AMP-activated protein kinase and hypoxia-inducible factor 1α (HIF-1α). In our mini-review we briefly summarize data supporting this hypothesis as well as unresolved issues aiming to forthcoming studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Sergei N. Orlov
- National Research Tomsk State University, Tomsk, Russia
- Siberian Medical University, Tomsk, Russia
- M.V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
57
|
Samoilov MO, Sidorova MV, Glushchenko TS. The pattern of the expression of hypoxia-inducible factor (HIF-1α) in the neocortex and hippocampus of rats after the presentation of different modes of hypobaric hypoxia. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415040145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
58
|
Petzinger GM, Holschneider DP, Fisher BE, McEwen S, Kintz N, Halliday M, Toy W, Walsh JW, Beeler J, Jakowec MW. The Effects of Exercise on Dopamine Neurotransmission in Parkinson's Disease: Targeting Neuroplasticity to Modulate Basal Ganglia Circuitry. Brain Plast 2015; 1:29-39. [PMID: 26512345 PMCID: PMC4621077 DOI: 10.3233/bpl-150021] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Animal studies have been instrumental in providing evidence for exercise-induced neuroplasticity of corticostriatal circuits that are profoundly affected in Parkinson’s disease. Exercise has been implicated in modulating dopamine and glutamate neurotransmission, altering synaptogenesis, and increasing cerebral blood flow. In addition, recent evidence supports that the type of exercise may have regional effects on brain circuitry, with skilled exercise differentially affecting frontal-striatal related circuits to a greater degree than pure aerobic exercise. Neuroplasticity in models of dopamine depletion will be reviewed with a focus on the influence of exercise on the dorsal lateral striatum and prefrontal related circuitry underlying motor and cognitive impairment in PD. Although clearly more research is needed to address major gaps in our knowledge, we hypothesize that the potential effects of exercise on inducing neuroplasticity in a circuit specific manner may occur through synergistic mechanisms that include the coupling of an increasing neuronal metabolic demand and increased blood flow. Elucidation of these mechanisms may provide important new targets for facilitating brain repair and modifying the course of disease in PD.
Collapse
Affiliation(s)
- G M Petzinger
- Department of Neurology, University of Southern California, Los Angeles, CA, 90033 ; Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, 90033
| | - D P Holschneider
- Department of Neurology, University of Southern California, Los Angeles, CA, 90033 ; Department of Psychiatry and the Behavioral Sciences, University of Southern California, Los Angeles, CA, 90033
| | - B E Fisher
- Department of Neurology, University of Southern California, Los Angeles, CA, 90033 ; Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, 90033
| | - S McEwen
- Andrus Gerontology, University of Southern California, Los Angeles, CA, 90033, and Department of Psychiatry & Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, 90095
| | - N Kintz
- Department of Neurology, University of Southern California, Los Angeles, CA, 90033
| | - M Halliday
- Department of Neurology, University of Southern California, Los Angeles, CA, 90033
| | - W Toy
- Department of Neurology, University of Southern California, Los Angeles, CA, 90033
| | - J W Walsh
- Andrus Gerontology, University of Southern California, Los Angeles, CA, 90033, and Department of Psychiatry & Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, 90095
| | - J Beeler
- Department of Psychology, CUNY, New York
| | - M W Jakowec
- Department of Neurology, University of Southern California, Los Angeles, CA, 90033 ; Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, 90033
| |
Collapse
|
59
|
Semenov DG, Belyakov AV, Glushchenko TS, Samoilov MO, Salinska E, Lazarewicz JW. Hypobaric Preconditioning Modifies Group I mGluRs Signaling in Brain Cortex. Neurochem Res 2015; 40:2200-10. [PMID: 26318863 DOI: 10.1007/s11064-015-1708-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 11/24/2022]
Abstract
The study assessed involvement of Ca(2+) signaling mediated by the metabotropic glutamate receptors mGluR1/5 in brain tolerance induced by hypoxic preconditioning. Acute slices of rat piriform cortex were tested 1 day after exposure of adult rats to mild hypobaric hypoxia for 2 h at a pressure of 480 hPa once a day for three consecutive days. We detected 44.1 ± 11.6 % suppression of in vitro anoxia-induced increases of intracellular Ca(2+) levels and a fivefold increase in Ca(2+) transients evoked by selective mGluR1/5 agonist, DHPG. Western blot analysis of cortical homogenates demonstrated a 11 ± 4 % decrease in mGluR1 immunoreactivity (IR), and in the nuclei-enriched fraction a 12 ± 3 % increase in IR of phospholipase Cβ1 (PLCβ1), which is a major mediator of mGluR1/5 signaling. Immunocytochemical analysis of the cortex revealed increase in the mGluR1/5 and PLCβ1 IR in perikarya, and a decrease in IR of the neuronal inositol trisphosphate receptors (IP3Rs). We suggest that enhanced expression of mGluR5 and PLCβ1 and potentiation of Ca(2+) signaling may represent pro-survival upregulation of Ca(2+)-dependent genomic processes, while decrease in mGluR1 and IP3R IR may be attributed to a feedback mechanism preventing excessive intracellular Ca(2+) release.
Collapse
Affiliation(s)
- Dmitry G Semenov
- Pavlov Institute of Physiology, Russian Academy of Sciences, Nab. Makarova, 6, Saint Petersburg, Russia, 199034.
| | - Alexandr V Belyakov
- Pavlov Institute of Physiology, Russian Academy of Sciences, Nab. Makarova, 6, Saint Petersburg, Russia, 199034.
| | - Tatjana S Glushchenko
- Pavlov Institute of Physiology, Russian Academy of Sciences, Nab. Makarova, 6, Saint Petersburg, Russia, 199034.
| | - Mikhail O Samoilov
- Pavlov Institute of Physiology, Russian Academy of Sciences, Nab. Makarova, 6, Saint Petersburg, Russia, 199034.
| | - Elzbieta Salinska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Jerzy W Lazarewicz
- Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| |
Collapse
|
60
|
Scholefield BR, Gao F, Duncan HP, Tasker RC, Parslow RC, Draper ES, McShane P, Davies P, Morris KP. Observational study of children admitted to United Kingdom and Republic of Ireland Paediatric Intensive Care Units after out-of-hospital cardiac arrest. Resuscitation 2015. [PMID: 26206597 DOI: 10.1016/j.resuscitation.2015.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AIMS To estimate the prevalence of children admitted after out-of-hospital cardiac arrest (OHCA) to UK and Republic of Ireland (RoI) Paediatric Intensive Care Units (PICUs) and factors associated with mortality to inform future clinical trial feasibility. METHOD Observational study using a prospectively collected dataset of the Paediatric Intensive Care Audit Network (PICANet) of 33 UK and RoI PICUs (January 2003 to June 2010). Cases (0 to <16 years), with documented OHCA surviving to PICU admission and requiring mechanical ventilation were included. Main outcomes were prevalence for admission and death within PICU. Factors associated with mortality were examined with multiple logistic regression analysis. RESULTS 827 of 111,170 admissions (0.73%; 95% CI [0.48 to 0.98%]) were identified as children admitted following OHCA. PICU mortality for OHCA was 50.5% (418/827). Recruitment into an adequately sized clinical trial would not be feasible with the current prevalence rate. Characteristics at PICU admission associated with increased risk of death included; bilateral unreactive pupils, genetically inherited condition, inter-hospital transfer to PICU, requirement for vasoactive drugs and greater base deficit. Factors associated with reduced risk of death were submersion or a respiratory aetiology and pre-existing respiratory or cardiac conditions. CONCLUSIONS Less than 120 children a year are admitted to PICUs in the UK and RoI after OHCA, limiting options for conducting UK intervention trials. The risk factors associated with mortality identified in this study will allow risk stratification in future studies.
Collapse
Affiliation(s)
- B R Scholefield
- Birmingham Children's Hospital, Paediatric Intensive Care Unit, Steelhouse Lane, Birmingham B4 6NH, United Kingdom.
| | - F Gao
- College of Medical and Dental Sciences, School of Clinical and Experimental Medicine, Medical School Building, University of Birmingham, Room WF28, Birmingham B15 2TT, United Kingdom.
| | - H P Duncan
- Birmingham Children's Hospital, Paediatric Intensive Care Unit, Steelhouse Lane, Birmingham B4 6NH, United Kingdom.
| | - R C Tasker
- Departments of Neurology and Anaesthesia, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - R C Parslow
- Epidemiology, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Room 8.49 Worsley Building, Clarendon Way, Leeds LS2 9JT, United Kingdom.
| | - E S Draper
- Perinatal & Paediatric Epidemiology Department of Health Sciences, College of Medicine, Biological Sciences and Psychology, University of Leicester, 22-28 Princess Road West, Leicester LE1 6TP, United Kingdom.
| | - P McShane
- Leeds Institute of Cardiovascular and Metabolic Medicine School of Medicine, University of Leeds, Room 8.49, Worsley Building, Clarendon Way, Leeds LS2 9JT, United Kingdom.
| | - P Davies
- Birmingham Children's Hospital, Research and Development, Steelhouse Lane, Birmingham B4 6NH, West Midlands, United Kingdom.
| | - K P Morris
- Birmingham Children's Hospital, Paediatric Intensive Care Unit, Steelhouse Lane, Birmingham B4 6NH, United Kingdom.
| |
Collapse
|
61
|
Abstract
SIGNIFICANCE The molecular mechanism of aging is still vigorously debated, although a general consensus exists that mitochondria are significantly involved in this process. However, the previously postulated role of mitochondrial-derived reactive oxygen species (ROS) as the damaging agents inducing functional loss in aging has fallen out of favor in the recent past. In this review, we critically examine the role of ROS in aging in the light of recent advances on the relationship between mitochondrial structure and function. RECENT ADVANCES The functional mitochondrial respiratory chain is now recognized as a reflection of the dynamic association of respiratory complexes in the form of supercomplexes (SCs). Besides providing kinetic advantage (channeling), SCs control ROS generation by the respiratory chain, thus providing a means to regulate ROS levels in the cell. Depending on their concentration, these ROS are either physiological signals essential for the life of the cell or toxic species that damage cell structure and functions. CRITICAL ISSUES We propose that under physiological conditions the dynamic nature of SCs reversibly controls the generation of ROS as signals involved in mitochondrial-nuclear communication. During aging, there is a progressive loss of control of ROS generation so that their production is irreversibly enhanced, inducing a vicious circle in which signaling is altered and structural damage takes place. FUTURE DIRECTIONS A better understanding on the forces affecting SC association would allow the manipulation of ROS generation, directing these species to their physiological signaling role.
Collapse
Affiliation(s)
- Maria Luisa Genova
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| | - Giorgio Lenaz
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| |
Collapse
|
62
|
Min JW, Hu JJ, He M, Sanchez RM, Huang WX, Liu YQ, Bsoul NB, Han S, Yin J, Liu WH, He XH, Peng BW. Vitexin reduces hypoxia-ischemia neonatal brain injury by the inhibition of HIF-1alpha in a rat pup model. Neuropharmacology 2015; 99:38-50. [PMID: 26187393 DOI: 10.1016/j.neuropharm.2015.07.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/16/2015] [Accepted: 07/07/2015] [Indexed: 01/25/2023]
Abstract
Previous studies have demonstrated that the early suppression of HIF-1α after hypoxia-ischemia (HI) injury provides neuroprotection. Vitexin (5, 7, 4-trihydroxyflavone-8-glucoside), an HIF-1α inhibitor, is a c-glycosylated flavone that has been identified in medicinal plants. Therefore, we hypothesized that treatment with vitexin would protect against HI brain injury. Newborn rat pups were subjected to unilateral carotid artery ligation followed by 2.5 h of hypoxia (8% O2 at 37 °C). Vitexin (30, 45 or 60 mg/kg) was administered intraperitoneally at 5 min or 3 h after HI. Vitexin, administered 5 min after HI, was neuroprotective as seen by decreased infarct volume evaluated at 48 h post-HI. This neuroprotection was removed when vitexin was administered 3 h after HI. Neuronal cell death, blood-brain barrier (BBB) integrity, brain edema, HIF-1α and VEGF protein levels were evaluated using a combination of Nissl staining, IgG staining, brain water content, immunohistochemistry and Western blot at 24 and 48 h after HI. The long-term effects of vitexin were evaluated by brain atrophy measurement, Nissl staining and neurobehavioral tests. Vitexin (45 mg/kg) ameliorated brain edema, BBB disruption and neuronal cell death; Upregulation of HIF-1α by dimethyloxalylglycine (DMOG) increased the BBB permeability and brain edema compared to HI alone. Vitexin attenuated the increase in HIF-1α and VEGF. Vitexin also had long-term effects of protecting against the loss of ipsilateral brain and improveing neurobehavioral outcomes. In conclusion, our data indicate early HIF-1α inhibition with vitexin provides both acute and long-term neuroprotection in the developing brain after neonatal HI injury.
Collapse
Affiliation(s)
- Jia-Wei Min
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jiang-Jian Hu
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Miao He
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Russell M Sanchez
- Department of Surgery, College of Medicine, Texas A&M Health Science Center, Neuroscience Institute, Scott & White Hospital, & Central Texas Veterans Health Care System, Temple, TX, USA
| | - Wen-Xian Huang
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yu-Qiang Liu
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Najeeb Bassam Bsoul
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Song Han
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jun Yin
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wan-Hong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiao-Hua He
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
63
|
Sari Y, Sanada H, Minematsu T, Nakagami G, Nagase T, Huang L, Noguchi H, Mori T, Yoshimura K, Sugama J. Vibration inhibits deterioration in rat deep-tissue injury through HIF1-MMP axis. Wound Repair Regen 2015; 23:386-93. [PMID: 25801385 DOI: 10.1111/wrr.12286] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/16/2015] [Indexed: 11/29/2022]
Abstract
Deep-tissue injury (DTI) is a unique type of pressure ulcer (PU) in which deep-tissue damage expands outwards to the superficial skin. DTI progresses rapidly into a severe PU, despite initially appearing as only a bruise or darkened tissue in the superficial skin. Although some DTI detection methods are available, there is currently no strategy for treating deteriorating DTI. This study investigated the efficacy of vibration therapy for preventing DTI deterioration through down-regulation of the hypoxia-inducible factor-1 matrix metalloproteinase (MMP) axis in rats. We prepared a conventional PU rat model (PU group) and a DTI deterioration rat model (DTI group). The DTI group was further divided into two groups subjected to vibration and control treatments, respectively. Macroscopic and histological features, hypoxia, oxidative stress, apoptosis, and MMP2 and MMP9 activities in compressed skin were analyzed. Hypoxia, oxidative stress, and MMP activity were enhanced in the DTI group compared with the PU group. Vibration remarkably inhibited DTI deterioration, hypoxia, and the expression/activities of MMP2 and MMP9. These results suggest that vibration therapy can effectively attenuate deterioration of DTI. This report provides the first evidence for a therapeutic treatment for deteriorating DTI.
Collapse
Affiliation(s)
- Yunita Sari
- Department of Gerontological Nursing/Wound Care Management, The University of Tokyo, Tokyo, Japan.,Department of Nursing, Jenderal Soedirman University, Purwokerto, Indonesia
| | - Hiromi Sanada
- Department of Gerontological Nursing/Wound Care Management, The University of Tokyo, Tokyo, Japan
| | - Takeo Minematsu
- Department of Gerontological Nursing/Wound Care Management, The University of Tokyo, Tokyo, Japan
| | - Gojiro Nakagami
- Department of Gerontological Nursing/Wound Care Management, The University of Tokyo, Tokyo, Japan
| | - Takashi Nagase
- Department of Gerontological Nursing/Wound Care Management, The University of Tokyo, Tokyo, Japan
| | - Lijuan Huang
- Department of Gerontological Nursing/Wound Care Management, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Noguchi
- Department of Life Support Technology (Molten), The University of Tokyo, Tokyo, Japan
| | - Taketoshi Mori
- Department of Life Support Technology (Molten), The University of Tokyo, Tokyo, Japan
| | - Kotaro Yoshimura
- Department of Plastic Surgery, The University of Tokyo, Tokyo, Japan
| | - Junko Sugama
- Wellness Promotion Science Center, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
64
|
Thushara Vijayakumar N, Sangwan A, Sharma B, Majid A, Rajanikant GK. Cerebral Ischemic Preconditioning: the Road So Far…. Mol Neurobiol 2015; 53:2579-93. [PMID: 26081149 DOI: 10.1007/s12035-015-9278-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 06/02/2015] [Indexed: 12/25/2022]
Abstract
Cerebral preconditioning constitutes the brain's adaptation to lethal ischemia when first exposed to mild doses of a subtoxic stressor. The phenomenon of preconditioning has been largely studied in the heart, and data from in vivo and in vitro models from past 2-3 decades have provided sufficient evidence that similar machinery exists in the brain as well. Since preconditioning results in a transient protective phenotype labeled as ischemic tolerance, it can open many doors in the medical warfare against stroke, a debilitating cerebrovascular disorder that kills or cripples thousands of people worldwide every year. Preconditioning can be induced by a variety of stimuli from hypoxia to pharmacological anesthetics, and each, in turn, induces tolerance by activating a multitude of proteins, enzymes, receptors, transcription factors, and other biomolecules eventually leading to genomic reprogramming. The intracellular signaling pathways and molecular cascades behind preconditioning are extensively being investigated, and several first-rate papers have come out in the last few years centered on the topic of cerebral ischemic tolerance. However, translating the experimental knowledge into the clinical scaffold still evades practicality and faces several challenges. Of the various preconditioning strategies, remote ischemic preconditioning and pharmacological preconditioning appears to be more clinically relevant for the management of ischemic stroke. In this review, we discuss current developments in the field of cerebral preconditioning and then examine the potential of various preconditioning agents to confer neuroprotection in the brain.
Collapse
Affiliation(s)
- N Thushara Vijayakumar
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India
| | - Amit Sangwan
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India
| | - Bhargy Sharma
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India
| | - Arshad Majid
- Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - G K Rajanikant
- School of Biotechnology, DBT-Centre for Bioinformatics, National Institute of Technology Calicut, Calicut, 673601, India.
| |
Collapse
|
65
|
Liu FJ, Kaur P, Karolina DS, Sepramaniam S, Armugam A, Wong PTH, Jeyaseelan K. MiR-335 Regulates Hif-1α to Reduce Cell Death in Both Mouse Cell Line and Rat Ischemic Models. PLoS One 2015; 10:e0128432. [PMID: 26030758 PMCID: PMC4452242 DOI: 10.1371/journal.pone.0128432] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 04/27/2015] [Indexed: 01/25/2023] Open
Abstract
Hypoxia inducible factor-1α facilitates cellular adaptation to hypoxic conditions. Hence its tight regulation is crucial in hypoxia related diseases such as cerebral ischemia. Changes in hypoxia inducible factor-1α expression upon cerebral ischemia influence the expression of its downstream genes which eventually determines the extent of cellular damage. MicroRNAs are endogenous regulators of gene expression that have rapidly emerged as promising therapeutic targets in several diseases. In this study, we have identified miR-335 as a direct regulator of hypoxia inducible factor-1α and as a potential therapeutic target in cerebral ischemia. MiR-335 and hypoxia inducible factor-1α mRNA showed an inverse expression profile, both in vivo and in vitro ischemic conditions. Given the biphasic nature of hypoxia inducible factor-1α expression during cerebral ischemia, miR-335 mimic was found to reduce infarct volume in the early time (immediately after middle cerebral artery occlusion) of embolic stroke animal models while the miR-335 inhibitor appears to be beneficial at the late time of stroke (24 hrs after middle cerebral artery occlusion). Modulation of hypoxia inducible factor-1α expression by miR-335 also influenced the expression of crucial genes implicated in neurovascular permeability, cell death and maintenance of the blood brain barrier. These concerted effects, resulting in a reduction in infarct volume bring about a beneficial outcome in ischemic stroke.
Collapse
Affiliation(s)
- Fu Jia Liu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Prameet Kaur
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Dwi S. Karolina
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Sugunavathi Sepramaniam
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Arunmozhiarasi Armugam
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Peter T. H. Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Kandiah Jeyaseelan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, 3800, Australia
- * E-mail:
| |
Collapse
|
66
|
Li R, Luo X, Wu J, Thangthaeng N, Jung ME, Jing S, Li L, Ellis DZ, Liu L, Ding Z, Forster MJ, Yan LJ. Mitochondrial Dihydrolipoamide Dehydrogenase is Upregulated in Response to Intermittent Hypoxic Preconditioning. Int J Med Sci 2015; 12:432-440. [PMID: 26078703 PMCID: PMC4466405 DOI: 10.7150/ijms.11402] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 05/13/2015] [Indexed: 01/06/2023] Open
Abstract
Intermittent hypoxia preconditioning (IHP) has been shown to protect neurons against ischemic stroke injury. Studying how proteins respond to IHP may identify targets that can help fight stroke. The objective of the present study was to investigate whether mitochondrial dihydrolipoamide dehydrogenase (DLDH) would respond to IHP and if so, whether such a response could be linked to neuroprotection in ischemic stroke injury. To do this, we subjected male rats to IHP for 20 days and measured the content and activity of DLDH as well as the three α-keto acid dehydrogenase complexes that contain DLDH. We also measured mitochondrial electron transport chain enzyme activities. Results show that DLDH content was indeed upregulated by IHP and this upregulation did not alter the activities of the three α-keto acid dehydrogenase complexes. Results also show that the activities of the five mitochondrial complexes (I-V) were not altered either by IHP. To investigate whether IHP-induced DLDH upregulation is linked to neuroprotection against ischemic stroke injury, we subjected both DLDH deficient mouse and DLDH transgenic mouse to stroke surgery followed by measurement of brain infarction volume. Results indicate that while mouse deficient in DLDH had exacerbated brain injury after stroke, mouse overexpressing human DLDH also showed increased brain injury after stroke. Therefore, the physiological significance of IHP-induced DLDH upregulation remains to be further investigated.
Collapse
Affiliation(s)
- Rongrong Li
- 1. Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- 2. Department of Anethesiology, the First Affiliated Hospital of Nanjing University, Nanjing, Jiangsu province, China, 210029
| | - Xiaoting Luo
- 1. Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- 3. Department of Biochemistry and Molecular Biology, Gannan Medical University, Ganzhou, Jiangxi province, China, 341000
| | - Jinzi Wu
- 1. Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Nopporn Thangthaeng
- 4. Department of Pharmacology and Neurosciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Marianna E. Jung
- 4. Department of Pharmacology and Neurosciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Siqun Jing
- 1. Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
- 5. College of Life Sciences and Technology, Xinjiang University, Urumqi, Xinjiang, China, 830046
| | - Linya Li
- 1. Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Dorette Z. Ellis
- 1. Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Li Liu
- 6. Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China 210029
| | - Zhengnian Ding
- 2. Department of Anethesiology, the First Affiliated Hospital of Nanjing University, Nanjing, Jiangsu province, China, 210029
| | - Michael J. Forster
- 4. Department of Pharmacology and Neurosciences, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Liang-Jun Yan
- 1. Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
67
|
Feng Y, Bhatt AJ. Corticosteroid responses following hypoxic preconditioning provide neuroprotection against subsequent hypoxic-ischemic brain injury in the newborn rats. Int J Dev Neurosci 2015; 44:6-13. [PMID: 25937464 DOI: 10.1016/j.ijdevneu.2015.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/03/2015] [Accepted: 04/20/2015] [Indexed: 11/26/2022] Open
Abstract
Limited research has evaluated the corticosteroids (CS) response in hypoxic preconditioning (PC) induced neuroprotection against subsequent hypoxic-ischemic (HI) brain injury in newborns. To measure, CS response to hypoxic PC, at postnatal day 6 (P6), rat pups were randomly divided into sham, NoPC (exposure to 21% O2) and PC (exposure to 8% O2 for 3h) groups. In a separate experiment, at P6, rat pups were randomly divided into three groups (sham, NoPC+HI, PC+HI). Rat pups in NoPC+HI and PC+HI groups, respectively had normoxic or hypoxic exposure for 3h at P6 and then had the right carotid artery permanently ligated followed by 140 min of hypoxia at P7 (HI). Plasma CS levels were measured at 0.5, 1, 3, 6 and 12h after hypoxic PC and hypoxic PC followed by HI. To investigate whether CS response to hypoxic PC provides neuroprotection against HI, at P6, rat pups were randomly divided into five groups. Fifteen minutes prior to PC or normoxic exposure, rat pups in DMSO+PC+HI and DMSO+NoPC+HI groups received DMSO while in RU486+PC+HI and RU486+NoPC+HI groups received RU486 (glucocorticoid receptor blocker, 60 mg/kg) s.c., respectively. Afterwards, rat pups were exposed to normoxia (DMSO+NoPC+HI, RU486+NoPC+HI) or hypoxia (DMSO+PC+HI, RU486+PC+HI) for 3h and then HI 24h later (P7). Rat pups at the corresponding age without any exposure to PC or HI or RU486/DMSO were used as sham. We found that hypoxic PC caused CS surge as well as augmented CS surge and preserved the glucocorticoid feedback regulation after HI. Hypoxic PC reduced HI induced early and delayed brain damage. RU486 partially but significantly inhibited hypoxic PC induced neuroprotection.
Collapse
Affiliation(s)
- Yangzheng Feng
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Abhay J Bhatt
- Department of Pediatrics, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
68
|
Cui C, Zhou T, Li J, Wang H, Li X, Xiong J, Xu P, Xue M. Proteomic analysis of the mouse brain after repetitive exposure to hypoxia. Chem Biol Interact 2015; 236:57-66. [PMID: 25937538 DOI: 10.1016/j.cbi.2015.04.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 02/04/2015] [Accepted: 04/09/2015] [Indexed: 10/23/2022]
Abstract
Hypoxic preconditioning (HPC) is known to have a protective effect against hypoxic damage; however, the precise mechanisms involved remain unknown. In this study, an acute and repetitive hypoxia mouse model, two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) coupled with matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF/TOF-MS), and Western blot experiments were used to identify the differential expression of key proteins in the mouse brain during HPC. Approximately 2100 2D-DIGE spots were observed following gel imaging and spot detection. Significant differences (p < 0.05) in the expression of 66 proteins were observed between the 3× HPC treatment group and the control group, 45 proteins were observed between the 6× HPC treatment group and the control group, and 70 proteins were observed between the 3× HPC treatment group and the 6× HPC group. Consistent results among Western blot, 2D-DIGE and MS methods were observed for the proteins, ATP synthase subunit alpha, malate dehydrogenase, guanine nucleotide-binding protein subunit beta-1 and proteasome subunit alpha type-2. The proteins associated with ATP synthesis and the citric acid cycle were down-regulated, while those linked to glycolysis and oxygen-binding were up-regulated. This proteomic analysis of the mouse brain after HPC furthers understanding of the molecular pathways involved in the protective effect of HPC and these findings provide new insight into the mechanisms of hypoxia and HPC.
Collapse
Affiliation(s)
- Can Cui
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China
| | - Tao Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China
| | - Jingyi Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China
| | - Hong Wang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China
| | - Xiaorong Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China; Beijing Laboratory for Biomedical Detection Technology and Instrument, Beijing 100069, China
| | - Jie Xiong
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China; Beijing Laboratory for Biomedical Detection Technology and Instrument, Beijing 100069, China
| | - Pingxiang Xu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China; Beijing Laboratory for Biomedical Detection Technology and Instrument, Beijing 100069, China
| | - Ming Xue
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 10069, China; Beijing Laboratory for Biomedical Detection Technology and Instrument, Beijing 100069, China.
| |
Collapse
|
69
|
|
70
|
Bonova P, Gottlieb M. Blood as the carrier of ischemic tolerance in rat brain. J Neurosci Res 2015; 93:1250-7. [PMID: 25787695 DOI: 10.1002/jnr.23580] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/20/2015] [Accepted: 02/09/2015] [Indexed: 11/08/2022]
Abstract
This study provides clear evidence that the factor inducing tolerance to ischemia is transmitted via the circulating blood. By using the remote ischemia and the cross-circulation model, the tolerance to ischemia was transmitted from donor to recipient. For this study, the following experimental groups were designed: I, sham control group; II, group of tolerant hindlimb tourniquet-treated rats; III, positive control group; IV, control for cross-circulation influence; preconditioned animals: V, tolerant animals subjected to middle cerebral artery occlusion (MCAO); VI, tolerant animals cross-circulated with SHC, followed by MCAO; VII, SHC animals cross-circulated with tolerant animals and subsequently subjected to MCAO; VIII, tolerant animals cross-circulated with ischemic rats, followed by MCAO; IX, SHC animals cross-circulated with ischemic animals and subjected to MCAO; postconditioned animals: X, ischemic animals treated with a remote limb tourniquet; XI, ischemic animals cross-circulated with SHC control rats; and XII, ischemic animals cross-circulated with tolerant rats. Results confirmed that remote ischemia induced reduction of infarct volume in the preconditioned (V, 60%) as well as in the postconditioned group (X, 52%). Significant diminution was also observed in group XII (56.6%). In the preconditioned group, decreased infarct volume was detected in groups VI and VII (about 65%) and in group IX (about 50%). The greatest infarct reduction (84%) was induced by the presence of ischemic blood in a tolerant rat before ischemia induction. In summary, the factor inducing tolerance to ischemia is generated by remote ischemia and by ischemia itself; from the site of origin to the rest of the body, it is transported by the systemic blood circulation and can be transferred from animal to animal. The effect of conditioning with two different ischemic events (brain and hindlimb ischemia) led to a cumulative, stronger tolerance response.
Collapse
Affiliation(s)
- Petra Bonova
- Institute of Neurobiology, Slovak Academy of Sciences, Kosice, Slovak Republic
| | - Miroslav Gottlieb
- Institute of Neurobiology, Slovak Academy of Sciences, Kosice, Slovak Republic
| |
Collapse
|
71
|
Koltsova SV, Shilov B, Birulina JG, Akimova OA, Haloui M, Kapilevich LV, Gusakova SV, Tremblay J, Hamet P, Orlov SN. Transcriptomic changes triggered by hypoxia: evidence for HIF-1α-independent, [Na+]i/[K+]i-mediated, excitation-transcription coupling. PLoS One 2014; 9:e110597. [PMID: 25375852 PMCID: PMC4222758 DOI: 10.1371/journal.pone.0110597] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 09/16/2014] [Indexed: 11/19/2022] Open
Abstract
This study examines the relative impact of canonical hypoxia-inducible factor-1alpha- (HIF-1α and Na+i/K+i-mediated signaling on transcriptomic changes evoked by hypoxia and glucose deprivation. Incubation of RASMC in ischemic conditions resulted in ∼3-fold elevation of [Na+]i and 2-fold reduction of [K+]i. Using global gene expression profiling we found that Na+,K+-ATPase inhibition by ouabain or K+-free medium in rat aortic vascular smooth muscle cells (RASMC) led to the differential expression of dozens of genes whose altered expression was previously detected in cells subjected to hypoxia and ischemia/reperfusion. For further investigations, we selected Cyp1a1, Fos, Atf3, Klf10, Ptgs2, Nr4a1, Per2 and Hes1, i.e. genes possessing the highest increments of expression under sustained Na+,K+-ATPase inhibition and whose implication in the pathogenesis of hypoxia was proved in previous studies. In ouabain-treated RASMC, low-Na+, high-K+ medium abolished amplification of the [Na+]i/[K+]i ratio as well as the increased expression of all tested genes. In cells subjected to hypoxia and glucose deprivation, dissipation of the transmembrane gradient of Na+ and K+ completely eliminated increment of Fos, Atf3, Ptgs2 and Per2 mRNAs and sharply diminished augmentation expression of Klf10, Edn1, Nr4a1 and Hes1. In contrast to low-Na+, high-K+ medium, RASMC transfection with Hif-1a siRNA attenuated increments of Vegfa, Edn1, Klf10 and Nr4a1 mRNAs triggered by hypoxia but did not impact Fos, Atf3, Ptgs2 and Per2 expression. Thus, our investigation demonstrates, for the first time, that Na+i/K+i-mediated, Hif-1α- -independent excitation-transcription coupling contributes to transcriptomic changes evoked in RASMC by hypoxia and glucose deprivation.
Collapse
MESH Headings
- Animals
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation
- Hypoxia/genetics
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Ouabain/pharmacology
- Rats
- Signal Transduction/drug effects
- Sodium-Potassium-Exchanging ATPase/metabolism
- Transcriptome
Collapse
Affiliation(s)
- Svetlana V. Koltsova
- Department of Biology, Moscow State University, Moscow, Russia
- Department of Medicine, Centre de recherche, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Boris Shilov
- Department of Physiology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Julia G. Birulina
- Department of Medical Biology, Siberian State Medical University, Tomsk, Russia
| | - Olga A. Akimova
- Department of Biology, Moscow State University, Moscow, Russia
| | - Mounsif Haloui
- Department of Medicine, Centre de recherche, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Leonid V. Kapilevich
- Department of Medical Biology, Siberian State Medical University, Tomsk, Russia
- Department of Physical Education, Tomsk State University, Tomsk, Russia
| | | | - Johanne Tremblay
- Department of Medicine, Centre de recherche, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Pavel Hamet
- Department of Medicine, Centre de recherche, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Sergei N. Orlov
- Department of Biology, Moscow State University, Moscow, Russia
- Department of Medicine, Centre de recherche, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Department of Medical Biology, Siberian State Medical University, Tomsk, Russia
- * E-mail:
| |
Collapse
|
72
|
Cruz Navarro J, Pillai S, Ponce LL, Van M, Goodman JC, Robertson CS. Endothelial nitric oxide synthase mediates the cerebrovascular effects of erythropoietin in traumatic brain injury. Front Immunol 2014; 5:494. [PMID: 25346735 PMCID: PMC4191322 DOI: 10.3389/fimmu.2014.00494] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/23/2014] [Indexed: 11/23/2022] Open
Abstract
Background: Erythropoietin (Epo) improves post-traumatic cerebral blood flow (CBF), pressure autoregulation, and vascular reactivity to l-arginine. This study examines the dependence of these cerebral hemodynamic effects of Epo on nitric oxide generated by endothelial nitric oxide synthase (eNOS). Methods: Using laser Doppler flow imaging, CBF was monitored in wild-type (WT) and eNOS-deficient mice undergoing controlled cortical impact followed by administration of Epo (5000 U/kg) or normal saline. Results: Cerebral blood flow decreased in all groups post-injury with the greatest reductions occurring at the impact site. Epo administration resulted in significantly higher CBF in the peri-contusional sites in the WT mice [70.2 ± 3.35% in Epo-treated compared to 53 ± 3.3% of baseline in saline-treated mice (p < 0.0001)], but no effect was seen in the eNOS-deficient mice. No CBF differences were found at the core impact site where CBF dropped to 20–25% of baseline in all groups. Conclusion: These differences between eNOS-deficient and WT mice indicate that the Epo mediated improvement in CBF in traumatic brain injury is eNOS dependent.
Collapse
Affiliation(s)
| | - Shibu Pillai
- Department of Neurosurgery, Baylor College of Medicine , Houston, TX , USA
| | - Lucido L Ponce
- Department of Neurosurgery, Baylor College of Medicine , Houston, TX , USA
| | - Mai Van
- Department of Neurosurgery, Baylor College of Medicine , Houston, TX , USA
| | - Jerry Clay Goodman
- Department of Pathology and Immunology, Baylor College of Medicine , Houston, TX , USA ; Department of Neurology, Baylor College of Medicine , Houston, TX , USA
| | | |
Collapse
|
73
|
Lee BJ, Emery-Sinclair EL, Mackenzie RWA, Hussain A, Taylor L, James RS, Thake CD. The impact of submaximal exercise during heat and/or hypoxia on the cardiovascular and monocyte HSP72 responses to subsequent (post 24 h) exercise in hypoxia. EXTREME PHYSIOLOGY & MEDICINE 2014; 3:15. [PMID: 25343025 PMCID: PMC4179935 DOI: 10.1186/2046-7648-3-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 08/21/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND The aims of this study were to describe the cellular stress response to prolonged endurance exercise in acute heat, hypoxia and the combination of heat and hypoxia and to determine whether prior acute exposure to these stressors improved cellular tolerance to a subsequent exercise bout in hypoxia 24 h later. METHODS Twelve males (age 22 ± 4 years, height 1.77 ± 0.05 m, mass 79 ± 12.9 kg, VO2 max 3.57 ± 0.7 L · min(-1)) completed four trials (30-min rest, 90-min cycling at 50% normoxic VO2 max) in normothermic normoxia (NORM; 18°C, FIO2 = 0.21), heat (HEAT; 40°C, 20% RH), hypoxia (HYP; FIO2 = 0.14) or a combination of heat and hypoxia (COM; 40°C, 20% RH, FIO2 = 0.14) separated by at least 7 days. Twenty-four hours after each trial, participants completed a hypoxic stress test (HST; 15-min rest, 60-min cycling at 50% normoxic VO2 max, FIO2 = 0.14). Monocyte heat shock protein 72 (mHSP72) was assessed immediately before and after each exercise bout. RESULTS mHSP72 increased post exercise in NORM (107% ± 5.5%, p > 0.05), HYP (126% ± 16%, p < 0.01), HEAT (153% ± 14%, p < 0.01) and COM (161% ± 32%, p < 0.01). mHSP72 had returned to near-resting values 24 h after NORM (97% ± 8.6%) but was elevated after HEAT (130% ± 19%), HYP (118% ± 17%) and COM (131% ± 19%) (p < 0.05). mHSP72 increased from baseline after HSTNORM (118% ± 12%, p < 0.05), but did not increase further in HSTHEAT, HSTHYP and HSTCOM. CONCLUSIONS The prior induction of mHSP72 as a result of COM, HEAT and HYP attenuated further mHSP72 induction after HST and was indicative of conferred cellular tolerance.
Collapse
Affiliation(s)
- Ben J Lee
- Sport and Exercise Science Applied Research Group, Coventry University, Coventry, UK
| | - Emma L Emery-Sinclair
- Inflammation and Infection Group, School of Science and Technology, University of Westminster, London, UK
| | - Richard WA Mackenzie
- Inflammation and Infection Group, School of Science and Technology, University of Westminster, London, UK
| | - Afthab Hussain
- Sport and Exercise Science Applied Research Group, Coventry University, Coventry, UK
| | - Lee Taylor
- Department of Sport and Exercise Sciences, University of Bedfordshire, Bedford, UK
| | - Rob S James
- Sport and Exercise Science Applied Research Group, Coventry University, Coventry, UK
| | - C Douglas Thake
- Sport and Exercise Science Applied Research Group, Coventry University, Coventry, UK
| |
Collapse
|
74
|
Mitochondrial stress extends lifespan in C. elegans through neuronal hormesis. Exp Gerontol 2014; 56:89-98. [DOI: 10.1016/j.exger.2014.03.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/15/2014] [Accepted: 03/25/2014] [Indexed: 12/19/2022]
|
75
|
Silpanisong J, Pearce WJ. Vasotrophic regulation of age-dependent hypoxic cerebrovascular remodeling. Curr Vasc Pharmacol 2014; 11:544-63. [PMID: 24063376 DOI: 10.2174/1570161111311050002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 06/08/2012] [Accepted: 07/12/2012] [Indexed: 02/07/2023]
Abstract
Hypoxia can induce functional and structural vascular remodeling by changing the expression of trophic factors to promote homeostasis. While most experimental approaches have been focused on functional remodeling, structural remodeling can reflect changes in the abundance and organization of vascular proteins that determine functional remodeling. Better understanding of age-dependent hypoxic macrovascular remodeling processes of the cerebral vasculature and its clinical implications require knowledge of the vasotrophic factors that influence arterial structure and function. Hypoxia can affect the expression of transcription factors, classical receptor tyrosine kinase factors, non-classical G-protein coupled factors, catecholamines, and purines. Hypoxia's remodeling effects can be mediated by Hypoxia Inducible Factor (HIF) upregulation in most vascular beds, but alterations in the expression of growth factors can also be independent of HIF. PPARγ is another transcription factor involved in hypoxic remodeling. Expression of classical receptor tyrosine kinase ligands, including vascular endothelial growth factor, platelet derived growth factor, fibroblast growth factor and angiopoietins, can be altered by hypoxia which can act simultaneously to affect remodeling. Tyrosine kinase-independent factors, such as transforming growth factor, nitric oxide, endothelin, angiotensin II, catecholamines, and purines also participate in the remodeling process. This adaptation to hypoxic stress can fundamentally change with age, resulting in different responses between fetuses and adults. Overall, these mechanisms integrate to assure that blood flow and metabolic demand are closely matched in all vascular beds and emphasize the view that the vascular wall is a highly dynamic and heterogeneous tissue with multiple cell types undergoing regular phenotypic transformation.
Collapse
Affiliation(s)
- Jinjutha Silpanisong
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | | |
Collapse
|
76
|
Varendi K, Airavaara M, Anttila J, Vose S, Planken A, Saarma M, Mitchell JR, Andressoo JO. Short-term preoperative dietary restriction is neuroprotective in a rat focal stroke model. PLoS One 2014; 9:e93911. [PMID: 24705386 PMCID: PMC3976327 DOI: 10.1371/journal.pone.0093911] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/07/2014] [Indexed: 01/13/2023] Open
Abstract
Stroke is a major complication of cardiovascular surgery, resulting in over 100,000 deaths and over a million postoperative encephalopathies annually in the US and Europe. While mitigating damage from stroke after it occurs has proven elusive, opportunities to reduce the incidence and/or severity of stroke prior to surgery in at-risk individuals remain largely unexplored. We tested the potential of short-term preoperative dietary restriction to provide neuroprotection in rat models of focal stroke. Rats were preconditioned with either three days of water-only fasting or six days of a protein free diet prior to induction of transient middle cerebral artery occlusion using two different methods, resulting in either a severe focal stroke to forebrain and midbrain, or a mild focal stroke localized to cortex only. Infarct volume, functional recovery and molecular markers of damage and protection were assessed up to two weeks after reperfusion. Preoperative fasting for 3 days reduced infarct volume after severe focal stroke. Neuroprotection was associated with modulation of innate immunity, including elevation of circulating neutrophil chemoattractant C-X-C motif ligand 1 prior to ischemia and suppression of striatal pro-inflammatory markers including tumor necrosis factor α, its receptor and downstream effector intercellular adhesion molecule-1 after reperfusion. Similarly, preoperative dietary protein restriction for 6 days reduced ischemic injury and improved functional recovery in a milder cortical infarction model. Our results suggest that short-term dietary restriction regimens may provide simple and translatable approaches to reduce perioperative stroke severity in high-risk elective vascular surgery.
Collapse
Affiliation(s)
- Kärt Varendi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jenni Anttila
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Sarah Vose
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Anu Planken
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - James R. Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
- * E-mail: (JRM); (JOA)
| | - Jaan-Olle Andressoo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- * E-mail: (JRM); (JOA)
| |
Collapse
|
77
|
Transcriptome profiling of the newborn mouse brain after hypoxia-reoxygenation: hyperoxic reoxygenation induces inflammatory and energy failure responsive genes. Pediatr Res 2014; 75:517-26. [PMID: 24375083 DOI: 10.1038/pr.2013.249] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 09/13/2013] [Indexed: 01/15/2023]
Abstract
BACKGROUND Supplemental oxygen used during resuscitation can be detrimental to the newborn brain. The aim was to determine how different oxygen therapies affect gene transcription in a hypoxia-reoxygenation model. METHODS C57BL/6 mice (n = 56), postnatal day 7, were randomized either to 120 min of hypoxia 8% O2 followed by 30 min of reoxygenation with 21, 40, 60, or 100% O2, or to normoxia followed by 30 min of 21 or 100% O2. Affymetrix 750k expression array was applied with RT-PCR used for validation. Histopathology and immunohistochemistry 3 d after hypoxia-reoxygenation compared groups reoxygenated with 21 or 100% O2 with normoxic controls (n = 22). RESULTS In total, ~81% of the gene expression changes were altered in response to reoxygenation with 60 or 100% O2 and constituted many inflammatory-responsive genes (i.e., C5ar2, Stat3, and Ccl12). Oxidative phosphorylation was downregulated after 60 or 100% O2. Iba1(+) cells were significantly increased in the striatum and hippocampal CA1 after both 21 and 100% O2. CONCLUSION In the present model, hypoxia-reoxygenation induces microglial accumulation in subregions of the brain. The transcriptional changes dominating after applying hyperoxic reoxygenation regimes include upregulating genes related to inflammatory responses and suppressing the oxidative phosphorylation pathway.
Collapse
|
78
|
The authors reply. Crit Care Med 2014; 42:e242-3. [DOI: 10.1097/ccm.0000000000000183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
79
|
Turovskaya MV, Turovsky EA, Kononov AV, Zinchenko VP. Short-term hypoxia induces a selective death of GABAergic neurons. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2014. [DOI: 10.1134/s199074781305019x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
80
|
Wang L, Duan D, Zhao Z, Teng X, Ge L, Liu B, Lu M. Repair of spinal cord injury by hypoxia-inducible factor-1a-expressing neural stem cells. JOURNAL OF MEDICAL HYPOTHESES AND IDEAS 2014. [DOI: 10.1016/j.jmhi.2013.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
81
|
Chen J, Liao W, Gao W, Huang J, Gao Y. Intermittent hypoxia protects cerebral mitochondrial function from calcium overload. Acta Neurol Belg 2013; 113:507-13. [PMID: 24122478 DOI: 10.1007/s13760-013-0220-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 06/10/2013] [Indexed: 11/29/2022]
Abstract
Hypoxia leads to Ca(2+) overload and results in mitochondrial uncoupling, decreased ATP synthesis, and neuronal death. Inhibition of mitochondrial Ca(2+) overload protects mitochondrial function after hypoxia. The present study was aimed to investigate the effect of intermittent hypoxia on mitochondrial function and mitochondrial tolerance to Ca(2+) overload. Wistar rats were divided into control and intermittent hypoxia (IH) groups. The IH group was subject to hypoxia for 4 h daily in a hypobaric cabin (5,000 m) for 7 days. Brain mitochondria were isolated on day 7 following hypoxia. The baseline mitochondrial functions, such as ST3, ST4, and respiratory control ratio (RCR = ST3/ST4), were measured using a Clark-type oxygen electrode. Mitochondrial adenine nucleotide concentrations were measured by HPLC. Mitochondrial membrane potential was determined by measuring rhodamine 123 (Rh-123) fluorescence in the absence and presence of high Ca(2+) concentration (0.1 M), which simulates Ca(2+) overload. Our results revealed that IH did not affect mitochondrial respiratory functions, but led to a reduction in AMP and an increase in ADP concentrations in mitochondria. Both control and IH groups demonstrated decreased mitochondrial membrane potential in the presence of high Ca(2+) (0.1 M), while the IH group showed a relative higher mitochondrial membrane potential. These results indicated that the neuroprotective effect of intermittent hypoxia was resulted partly from preserving mitochondrial membrane potential, and increasing mitochondrial tolerance to high calcium levels. The increased ADP and decreased AMP in mitochondria following intermittent hypoxia may be a mechanism underlying this protection.
Collapse
|
82
|
Long-lasting changes in DNA methylation following short-term hypoxic exposure in primary hippocampal neuronal cultures. PLoS One 2013; 8:e77859. [PMID: 24205000 PMCID: PMC3808424 DOI: 10.1371/journal.pone.0077859] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 09/04/2013] [Indexed: 12/14/2022] Open
Abstract
While the effects of hypoxia on gene expression have been investigated in the CNS to some extent, we currently do not know what role epigenetics plays in the transcription of many genes during such hypoxic stress. To start understanding the role of epigenetic changes during hypoxia, we investigated the long-term effect of hypoxia on gene expression and DNA methylation in hippocampal neuronal cells. Primary murine hippocampal neuronal cells were cultured for 7 days. Hypoxic stress of 1% O2, 5% CO2 for 24 hours was applied on Day 3, conditions we found to maximize cellular hypoxic stress response without inducing cell death. Cells were returned to normoxia for 4 days following the period of hypoxic stress. On Day 7, Methyl-Sensitive Cut Counting (MSCC) was used to identify a genome-wide methylation profile of the hippocampal cell lines to assess methylation changes resulting from hypoxia. RNA-Seq was also done on Day 7 to analyze changes in gene transcription. Phenotypic analysis showed that neuronal processes were significantly shorter after 1 day of hypoxia, but there was a catch-up growth of these processes after return to normoxia. Transcriptome profiling using RNA-Seq revealed 369 differentially expressed genes with 225 being upregulated, many of which form networks shown to affect CNS development and function. Importantly, the expression level of 59 genes could be correlated to the changes in DNA methylation in their promoter regions. CpG islands, in particular, had a strong tendency to remain hypomethylated long after hypoxic stress was removed. From this study, we conclude that short-term, sub-lethal hypoxia results in long-lasting changes to genome wide DNA methylation status and that some of these changes can be highly correlated with transcriptional modulation in a number of genes involved in functional pathways that have been previously implicated in neural growth and development.
Collapse
|
83
|
Ye Z, Ye W, Deng Y, Wang J, Zhou G, Zhang X. HIF-1-modified BMSCs improve migration and reduce neuronal apoptosis after stroke in rats. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s11434-013-5936-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
84
|
Speer RE, Karuppagounder SS, Basso M, Sleiman SF, Kumar A, Brand D, Smirnova N, Gazaryan I, Khim SJ, Ratan RR. Hypoxia-inducible factor prolyl hydroxylases as targets for neuroprotection by "antioxidant" metal chelators: From ferroptosis to stroke. Free Radic Biol Med 2013; 62:26-36. [PMID: 23376032 PMCID: PMC4327984 DOI: 10.1016/j.freeradbiomed.2013.01.026] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 01/23/2013] [Accepted: 01/23/2013] [Indexed: 01/12/2023]
Abstract
Neurologic conditions including stroke, Alzheimer disease, Parkinson disease, and Huntington disease are leading causes of death and long-term disability in the United States, and efforts to develop novel therapeutics for these conditions have historically had poor success in translating from bench to bedside. Hypoxia-inducible factor (HIF)-1α mediates a broad, evolutionarily conserved, endogenous adaptive program to hypoxia, and manipulation of components of the HIF pathway is neuroprotective in a number of human neurological diseases and experimental models. In this review, we discuss molecular components of one aspect of hypoxic adaptation in detail and provide perspective on which targets within this pathway seem to be ripest for preventing and repairing neurodegeneration. Further, we highlight the role of HIF prolyl hydroxylases as emerging targets for the salutary effects of metal chelators on ferroptosis in vitro as well in animal models of neurological diseases.
Collapse
Affiliation(s)
- Rachel E Speer
- Graduate Program in Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Saravanan S Karuppagounder
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Manuela Basso
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Sama F Sleiman
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Amit Kumar
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA
| | - David Brand
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Natalya Smirnova
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Irina Gazaryan
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Soah J Khim
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA
| | - Rajiv R Ratan
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA; Burke Medical Research Institute, White Plains, NY 10605, USA.
| |
Collapse
|
85
|
Effects of continuous positive airway pressure on cognitition and neuroimaging data in sleep apnea. Int J Psychophysiol 2013; 89:203-12. [DOI: 10.1016/j.ijpsycho.2013.03.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/27/2013] [Accepted: 03/30/2013] [Indexed: 11/22/2022]
|
86
|
Drago D, Cossetti C, Iraci N, Gaude E, Musco G, Bachi A, Pluchino S. The stem cell secretome and its role in brain repair. Biochimie 2013; 95:2271-85. [PMID: 23827856 PMCID: PMC4061727 DOI: 10.1016/j.biochi.2013.06.020] [Citation(s) in RCA: 240] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 06/19/2013] [Indexed: 12/16/2022]
Abstract
Compelling evidence exists that non-haematopoietic stem cells, including mesenchymal (MSCs) and neural/progenitor stem cells (NPCs), exert a substantial beneficial and therapeutic effect after transplantation in experimental central nervous system (CNS) disease models through the secretion of immune modulatory or neurotrophic paracrine factors. This paracrine hypothesis has inspired an alternative outlook on the use of stem cells in regenerative neurology. In this paradigm, significant repair of the injured brain may be achieved by injecting the biologics secreted by stem cells (secretome), rather than implanting stem cells themselves for direct cell replacement. The stem cell secretome (SCS) includes cytokines, chemokines and growth factors, and has gained increasing attention in recent years because of its multiple implications for the repair, restoration or regeneration of injured tissues. Thanks to recent improvements in SCS profiling and manipulation, investigators are now inspired to harness the SCS as a novel alternative therapeutic option that might ensure more efficient outcomes than current stem cell-based therapies for CNS repair. This review discusses the most recent identification of MSC- and NPC-secreted factors, including those that are trafficked within extracellular membrane vesicles (EVs), and reflects on their potential effects on brain repair. It also examines some of the most convincing advances in molecular profiling that have enabled mapping of the SCS.
Collapse
Affiliation(s)
- Denise Drago
- CNS Repair Unit, Institute of Experimental Neurology, Division of Neurosciences, San Raffaele Scientific Institute, 20132 Milan, Italy; Biomolecular Mass Spectrometry Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | | | | | | | | | | | | |
Collapse
|
87
|
WANG HAIYING, CHEN XIAOLONG. Effects of a Rho kinase inhibitor on the sequential expression of ICAM-1, HIF-1α, Bcl-2 and caspase-3 in the retina of rats with oxygen-induced retinopathy. Int J Mol Med 2013; 32:457-63. [DOI: 10.3892/ijmm.2013.1410] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/03/2013] [Indexed: 11/06/2022] Open
|
88
|
Cui M, Bai X, Li T, Chen F, Dong Q, Zhao Y, Liu X. Decreased extracellular adenosine levels lead to loss of hypoxia-induced neuroprotection after repeated episodes of exposure to hypoxia. PLoS One 2013; 8:e57065. [PMID: 23437309 PMCID: PMC3578825 DOI: 10.1371/journal.pone.0057065] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Accepted: 01/17/2013] [Indexed: 12/20/2022] Open
Abstract
Achieving a prolonged neuroprotective state following transient ischemic attacks (TIAs) is likely to effectively reduce the brain damage and neurological dysfunction associated with recurrent stroke. HPC is a phenomenon in which advanced exposure to mild hypoxia reduces the stroke volume produced by a subsequent TIA. However, this neuroprotection is not long-lasting, with the effects reaching a peak after 3 days. Therefore, in this study, we investigated the use of multiple episodes of hypoxic exposure at different time intervals to induce longer-term protection in a mouse stroke model. C57BL/6 mice were subjected to different hypoxic preconditioning protocols: a single episode of HPC or five identical episodes at intervals of 3 days (E3d HPC) or 6 days (E6d HPC). Three days after the last hypoxic exposure, temporary middle cerebral artery occlusion (MCAO) was induced. The effects of these HPC protocols on hypoxia-inducible factor (HIF) regulated gene mRNA expression were measured by quantitative PCR. Changes in extracellular adenosine concentrations, known to exert neuroprotective effects, were also measured using in vivo microdialysis and high pressure liquid chromatography (HPLC). Neuroprotection was provided by E6d HPC but not E3d HPC. HIF-regulated target gene expression increased significantly following all HPC protocols. However, E3d HPC significantly decreased extracellular adenosine and reduced cerebral blood flow in the ischemic region with upregulated expression of the adenosine transporter, equilibrative nucleoside transporter 1 (ENT1). An ENT1 inhibitor, propentofylline increased the cerebral blood flow and re-established neuroprotection in E3d HPC. Adenosine receptor specific antagonists showed that adenosine mainly through A1 receptor mediates HPC induced neuroprotection. Our data indicate that cooperation of HIF-regulated genes and extracellular adenosine is necessary for HPC-induced neuroprotection.
Collapse
Affiliation(s)
- Mei Cui
- Department of Neurology, Huashan hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
89
|
Epigenetics and the environment: in search of the "toleroasome" vital to execution of ischemic preconditioning. Transl Stroke Res 2013; 4:56-62. [PMID: 24323190 DOI: 10.1007/s12975-012-0235-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 11/29/2012] [Accepted: 11/29/2012] [Indexed: 01/02/2023]
Abstract
Activation and repression of gene expression are key features of ischemic tolerance. Converging lines of inquiry from several groups suggests that epigenetic proteins may transduce sublethal stresses, including bioenergetic or oxidative stress into durable (2-3 days) changes in gene expression that mediate ischemic tolerance. Here we discuss the potential mechanisms by which changes in cell state (e.g., ATP, NAD+, and oxygen) can modify specific targets including polycomb complexes, jumonji domain histone demethylases, and zinc and NAD-dependent histone decetylases and thus trigger an adaptive program. A major unanswered question is whether these proteins work in parallel or convergently as part of a "tolerosome" (tolero is the Latin word for tolerance), a multiprotein complex recruited to promoters or enhancers of specific genes, to mediate preconditioning. Whatever the case may be, epigenetic proteins are fertile targets for the treatment of stroke.
Collapse
|
90
|
Intermittent hypobaric hypoxia induces neuroprotection in kainate-induced oxidative stress in rats. J Mol Neurosci 2013; 50:402-10. [PMID: 23288703 DOI: 10.1007/s12031-012-9945-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 12/20/2012] [Indexed: 01/04/2023]
Abstract
Severe hypoxia induces oxidative stress, which can lead to brain injury. In this study, we wanted to determine whether intermittent hypobaric hypoxia induces oxidative stress in the brain. In adult rats exposed to 380 mmHg in a hypobaric chamber for 3 h/day for 6 days, we determined the levels of malondialdehyde and nitric oxide derivatives in the brain, which indicated that there was no oxidative stress. The levels of N-acetylaspartate indicated that there was no neuronal loss or mitochondrial dysfunction and finally because apoptotic proteins such as caspase-3 and nuclear factor-kappa B (NF-κB) were not activated, apoptosis was probably not induced. The increase in the expression of erythropoietin (EPO) in the brain of rats exposed to hypoxia confirms the efficacy of the method used to induce hypoxia in the brain. Because EPO have antioxidant effects on the brain, the results suggest that intermittent hypoxia can increase the antioxidant capacity of the brain. This effect of intermittent hypoxia was studied using the systemic administration of kainate, as a model of brain oxidative stress. Kainate treatment induces oxidative stress in the brain, which is measured by an increase in lipid peroxidation and nitric oxide. Furthermore, in rats treated with kainate, both caspase-3 and NF-κB activity increased. However, in rats previously exposed to intermittent hypobaric hypoxia, 3 h per day for 6 days, the effect of kainate treatment resulted in the reduction of both oxidative stress and apoptotic activity. This study demonstrates that intermittent hypobaric hypoxia can increase brain antioxidant capacity in rats and induces neuroprotection in kainate-induced oxidative injury.
Collapse
|
91
|
Gonchar O, Mankovska I. Moderate hypoxia/hyperoxia attenuates acute hypoxia-induced oxidative damage and improves antioxidant defense in lung mitochondria. ACTA ACUST UNITED AC 2012; 99:436-46. [DOI: 10.1556/aphysiol.99.2012.4.8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
92
|
Abstract
The preconditioning phenomena have been well established in heart as well as brain. In this review, we detail some of the original studies on preconditioning as well as studies from our lab using rodents and a genetic model system (fruit fly). We have used Drosophila in our lab to solve some of the questions related to tolerance or susceptibility to hypoxia. We believe that these pro-survival strategies and genetic pathways help us understand some of the preconditioning mechanisms that protect the brain from ischemia or ischemia.
Collapse
Affiliation(s)
- Priti Azad
- Department of Pediatrics (Section of Respiratory Medicine), University of California-San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
93
|
Wacker BK, Perfater JL, Gidday JM. Hypoxic preconditioning induces stroke tolerance in mice via a cascading HIF, sphingosine kinase, and CCL2 signaling pathway. J Neurochem 2012; 123:954-62. [PMID: 23043544 DOI: 10.1111/jnc.12047] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 07/26/2012] [Accepted: 10/04/2012] [Indexed: 12/30/2022]
Abstract
The induction of ischemic tolerance by preconditioning provides a platform to elucidate endogenous mechanisms of stroke protection. In these studies, we characterize the relationship between hypoxia-inducible factor (HIF), sphingosine kinase 2 (SphK2), and chemokine (C-C motif) ligand 2 (CCL2) in models of hypoxic or pharmacological preconditioning-induced ischemic tolerance. A genetics-based approach using SphK2- and CCL2-null mice showed both SphK2 and CCL2 to be necessary for the induction of ischemic tolerance following preconditioning with hypoxia, the hypoxia-mimetic cobalt chloride, or the sphingosine-1-phosphate (S1P) agonist FTY720. A pharmacological approach confirmed the necessity of HIF signaling for all three preconditioning stimuli, and showed that the SphK/S1P pathway transduces tolerance via the S1P(1) receptor. In addition, our data suggest significant cross-talk between HIF and SphK2-produced S1P signaling, which together act to up-regulate CCL2 expression. Overall, HIF, SphK, S1P, and CCL2 participate in a signaling cascade to induce the gene expression responsible for the stroke-tolerant phenotype established by hypoxic and FTY720 preconditioning. The identification of these common molecular mediators involved in signaling the genomic response to multiple preconditioning stimuli provides several targets for therapeutic manipulation.
Collapse
Affiliation(s)
- Bradley K Wacker
- Department of Neurosurgery, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | |
Collapse
|
94
|
Obstructive sleep apnea: impact of hypoxemia on memory. Sleep Breath 2012; 17:811-7. [PMID: 23065547 DOI: 10.1007/s11325-012-0769-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 08/27/2012] [Accepted: 09/13/2012] [Indexed: 10/27/2022]
Abstract
PURPOSE Attempts to understand the causes of cognitive impairment in obstructive sleep apnea (OSA) are complicated by the overlap among clinical and demographic factors that may impact cognition. The goal of the current study was to isolate the contribution of hypoxemia to cognitive impairment in OSA. METHODS Two groups of 20 patients with newly diagnosed OSA were compared. The groups differed on severity of hypoxemia but not other demographic (e.g., age, gender, education, estimated premorbid IQ) or clinical (e.g., sleep related respiratory disturbances, daytime sleepiness, depressive symptoms) variables. Participants completed polysonmography and cognitive assessment. RESULTS We compared patients with high and low hypoxemia on measures of memory, attention, executive functioning, and motor coordination using independent sample t-tests. The high hypoxemia group performed significantly better on immediate recall (Hopkins Verbal Learning Test - Revised; t = -2.50, p < 0.02) than the low hypoxemia group. No group differences were observed on other neuropsychological measures. CONCLUSIONS This study is one of the first to compare the cognitive performance of patients with high and low hypoxemia after controlling for demographic factors and aspects of OSA severity that could confound the relationship. In our carefully matched sample, we observed an unexpected advantage of higher hypoxemia on memory. These preliminary findings are discussed in the context of basic science literature on the protective effects of adaptation to intermittent hypoxemia. Our data suggest that the association between hypoxemia and cognition may not straightforward. Future research targeting the effects of hypoxemia on cognition controlling for other clinical factors in large groups of patients with OSA will be important.
Collapse
|
95
|
Lipopolysaccharide-preconditioning protects against endotoxin-induced white matter injury in the neonatal rat brain. Brain Res 2012; 1489:81-9. [PMID: 23063716 DOI: 10.1016/j.brainres.2012.10.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/12/2012] [Accepted: 10/06/2012] [Indexed: 11/22/2022]
Abstract
BACKGROUND Exposing the brain to a sub-damaging stimulus can protect against a subsequent lethal insult, a phenomenon termed preconditioning. The aim of this study was to investigate the neuroprotective effect of low dose LPS (lipopolysaccharide) pretreatment in endotoxin induced periventricular leukomalacia (PVL) in a rat model. METHODS Wistar rats with dated pregnancies were allocated to 5 groups: (i) no LPS administered, intraperitoneally (i.p.) pyrogen-free saline injected (Control group), (ii) 500μg/kg LPS administrated i.p. on days 18 and 19 (PVL group), (iii) 50μg/kg LPS administrated i.p. on day 17 followed by 500μg/kg LPS i.p. on days 18 and 19 (PC-PVL group), (iv) 50μg/kg LPS administrated on day 17 (PC only), and (v) i.p. pyrogen-free saline injected control group on day 17. RESULTS LPS-preconditioning given 24h before potent LPS exposure significantly reduced the number of apoptotic cell deaths and prevented hypomyelination. Antioxidant enzyme gene expression levels (Superoxide Dismutase-SOD1, SOD2, and SOD3) were increased and Tumor Necrosis Factor (TNF)α expression levels were decreased in the PC+PVL group when compared with the PVL group. CONCLUSION Low-dose LPS given one day before potent doses of LPS reduces antepartum LPS-induced brain damage. The mechanisms of protection might involve oxidation and inflammation.
Collapse
|
96
|
Lee CS, Burnsed OA, Raghuram V, Kalisvaart J, Boyan BD, Schwartz Z. Adipose stem cells can secrete angiogenic factors that inhibit hyaline cartilage regeneration. Stem Cell Res Ther 2012; 3:35. [PMID: 22920724 PMCID: PMC3580473 DOI: 10.1186/scrt126] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 08/06/2012] [Indexed: 12/13/2022] Open
Abstract
Introduction Adipose stem cells (ASCs) secrete many trophic factors that can stimulate tissue repair, including angiogenic factors, but little is known about how ASCs and their secreted factors influence cartilage regeneration. Therefore, the aim of this study was to determine the effects ASC-secreted factors have in repairing chondral defects. Methods ASCs isolated from male Sprague Dawley rats were cultured in monolayer or alginate microbeads supplemented with growth (GM) or chondrogenic medium (CM). Subsequent co-culture, conditioned media, and in vivo cartilage defect studies were performed. Results ASC monolayers and microbeads cultured in CM had decreased FGF-2 gene expression and VEGF-A secretion compared to ASCs cultured in GM. Chondrocytes co-cultured with GM-cultured ASCs for 7 days had decreased mRNAs for col2, comp, and runx2. Chondrocytes treated for 12 or 24 hours with conditioned medium from GM-cultured ASCs had reduced sox9, acan, and col2 mRNAs; reduced proliferation and proteoglycan synthesis; and increased apoptosis. ASC-conditioned medium also increased endothelial cell tube lengthening whereas conditioned medium from CM-cultured ASCs had no effect. Treating ASCs with CM reduced or abolished these deleterious effects while adding a neutralizing antibody for VEGF-A eliminated ASC-conditioned medium induced chondrocyte apoptosis and restored proteoglycan synthesis. FGF-2 also mitigated the deleterious effects VEGF-A had on chondrocyte apoptosis and phenotype. When GM-grown ASC pellets were implanted in 1 mm non-critical hyaline cartilage defects in vivo, cartilage regeneration was inhibited as evaluated by radiographic and equilibrium partitioning of an ionic contrast agent via microCT imaging. Histology revealed that defects with GM-cultured ASCs had no tissue ingrowth from the edges of the defect whereas empty defects and defects with CM-grown ASCs had similar amounts of neocartilage formation. Conclusions ASCs must be treated to reduce the secretion of VEGF-A and other factors that inhibit cartilage regeneration, which can significantly influence how ASCs are used for repairing hyaline cartilage.
Collapse
|
97
|
Raida Z, Hundahl CA, Kelsen J, Nyengaard JR, Hay-Schmidt A. Reduced infarct size in neuroglobin-null mice after experimental stroke in vivo. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2012; 4:15. [PMID: 22901501 PMCID: PMC3487987 DOI: 10.1186/2040-7378-4-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/11/2012] [Indexed: 01/09/2023]
Abstract
BACKGROUND Neuroglobin is considered to be a novel important pharmacological target in combating stroke and neurodegenerative disorders, although the mechanism by which this protection is accomplished remains an enigma. We hypothesized that if neuroglobin is directly involved in neuroprotection, then permanent cerebral ischemia would lead to larger infarct volumes in neuroglobin-null mice than in wild-type mice. METHODS Using neuroglobin-null mice, we estimated the infarct volume 24 hours after permanent middle cerebral artery occlusion using Cavalieri's Principle, and compared the infarct volume in neuroglobin-null and wild-type mice. Neuroglobin antibody staining was used to examine neuroglobin expression in the infarct area of wild-type mice. RESULTS Infarct volumes 24 hours after permanent middle cerebral artery occlusion were significantly smaller in neuroglobin-null mice than in wild-types (p < 0.01). Neuroglobin immunostaining of the penumbra area revealed no visible up-regulation of neuroglobin protein in ischemic wild-type mice when compared to uninjured wild-type mice. In uninjured wild-type mice, neuroglobin protein was seen throughout cortical layer II and sparsely in layer V. In contrast, no neuroglobin-immunoreactive neurons were observed in the aforementioned layers of the ischemia injured cortical area, or in the surrounding penumbra of ischemic wild-type mice. This suggests no selective sparing of neuroglobin expressing neurons in ischemia. CONCLUSIONS Neuroglobin-deficiency resulted in reduced tissue infarction, suggesting that, at least at endogenous expression levels, neuroglobin in itself is non-protective against ischemic injury.
Collapse
Affiliation(s)
- Zindy Raida
- Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Ansgar Hundahl
- Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Physiology, University of Tartu, Tartu, Estonia
- Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia
- Department of Clinical Biochemistry, University Hospital Bispebjerg, Copenhagen, Denmark
| | - Jesper Kelsen
- Department of Neurosurgery, University Hospital Copenhagen (Rigshospitalet), Copenhagen, Denmark
| | - Jens Randel Nyengaard
- Stereology and Electron Microscopy Research Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Anders Hay-Schmidt
- Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- The Panum Institute; Department of Neuroscience and Pharmacology, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark
| |
Collapse
|
98
|
López-Aguilera F, Plateo-Pignatari M, Biaggio V, Ayala C, Seltzer A. Hypoxic preconditioning induces an AT2-R/VEGFR-2(Flk-1) interaction in the neonatal brain microvasculature for neuroprotection. Neuroscience 2012; 216:1-9. [DOI: 10.1016/j.neuroscience.2012.04.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 04/13/2012] [Accepted: 04/27/2012] [Indexed: 10/28/2022]
|
99
|
Hypoxia-inducible factor prolyl hydroxylase inhibition: robust new target or another big bust for stroke therapeutics? J Cereb Blood Flow Metab 2012; 32:1347-61. [PMID: 22415525 PMCID: PMC3390817 DOI: 10.1038/jcbfm.2012.28] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A major challenge in developing stroke therapeutics that augment adaptive pathways to stress has been to identify targets that can activate compensatory programs without inducing or adding to the stress of injury. In this regard, hypoxia-inducible factor prolyl hydroxylases (HIF PHDs) are central gatekeepers of posttranscriptional and transcriptional adaptation to hypoxia, oxidative stress, and excitotoxicity. Indeed, some of the known salutary effects of putative 'antioxidant' iron chelators in ischemic and hemorrhagic stroke may derive from their abilities to inhibit this family of iron, 2-oxoglutarate, and oxygen-dependent enzymes. Evidence from a number of laboratories supports the notion that HIF PHD inhibition can improve histological and functional outcomes in ischemic and hemorrhagic stroke models. In this review, we discuss this evidence and highlight important gaps in our understanding that render HIF PHD inhibition a promising but not yet preclinically validated target for protection and repair after stroke.
Collapse
|
100
|
Singh N, Sharma G, Mishra V, Raghubir R. Hypoxia inducible factor-1: its potential role in cerebral ischemia. Cell Mol Neurobiol 2012; 32:491-507. [PMID: 22297543 PMCID: PMC11498632 DOI: 10.1007/s10571-012-9803-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 01/13/2012] [Indexed: 12/16/2022]
Abstract
A divergence in the supply and consumption of oxygen in brain tissue initiates complex cycle of biochemical and molecular events resulting in neuronal death. To overcome such adverse situation, the tissue has to adopt some cellular mechanisms such as induction of various transcription factors, such as hypoxia inducible factor (HIF). It is a transcriptional regulator of oxygen homeostasis and key factor to generate the adaptive responses through upregulation of various target genes involved in the erythropoiesis, angiogenesis as well as glucose metabolism and transport. On the other hand, some studies do suggest that HIF also plays a detrimental role in ischemic reperfusion injury by inducing the pro apoptotic molecules, cytokines such as Nix, BNip3, and IL-20 which cause mitochondrial dysfunction leading to cell death. Hence, modulation of HIF-1 activity seems to provide an innovative therapeutic target to reduce the cellular damage, which arises from ischemic injury. Apart from traditional oxygen dependent HIF regulation, the focus has now shifted toward oxygen independent regulation in cell specific manner through reactive oxygen species involving hypoxia-associated factor, and heat shock protein 90, etc. Therefore, future development of such small molecule regulators for HIF-1 stability and signaling may prove useful to therapeutically target for enhancing recovery and repair in I/R injury.
Collapse
Affiliation(s)
- Neetu Singh
- Division of Pharmacology, CSIR-Central Drug Research Institute, P.O. Box 173, Lucknow, 226001 UP India
| | - Gaurav Sharma
- Division of Pharmacology, CSIR-Central Drug Research Institute, P.O. Box 173, Lucknow, 226001 UP India
| | - Vikas Mishra
- Division of Pharmacology, CSIR-Central Drug Research Institute, P.O. Box 173, Lucknow, 226001 UP India
| | - Ram Raghubir
- Division of Pharmacology, CSIR-Central Drug Research Institute, P.O. Box 173, Lucknow, 226001 UP India
| |
Collapse
|