1
|
Gu CL, Zhang L, Zhu Y, Bao TY, Zhu YT, Chen YT, Pang HQ. Exploring the cellular and molecular basis of nerve growth factor in cerebral ischemia recovery. Neuroscience 2024; 566:190-197. [PMID: 39742942 DOI: 10.1016/j.neuroscience.2024.12.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Vascular obstruction often causes inadequate oxygen and nutrient supply to the brain. This deficiency results in cerebral ischemic injury, which significantly impairs neurological function. This review aimed to explore the neuroprotective and regenerative effects of nerve growth factor (NGF) in cerebral ischemic injury. NGF, a crucial neurotrophic factor, could inhibit neuronal apoptosis, reduce inflammatory responses, and promote axon regeneration and angiogenesis through its interaction with TrkA, a high-affinity receptor. These functions were closely related to the activation of Phosphatidylinositol 3-kinase/Protein kinase B (PI3K/AKT) and Mitogen-Activated Protein Kinase (MAPK) pathways. Moreover, the mechanisms of NGF in the acute and recovery phases, along with the strategies to enhance its therapeutic effects using delivery systems (such as intranasal administration, nanovesicles, and gene therapy) were also summarized. Although NGF shows great potential for clinical application, its delivery efficiency and long-term safety still need more research and improvements. Future research should focus on exploring the specific action mechanism of NGF, optimizing the delivery strategy, and evaluating its long-term efficacy and safety to facilitate its clinical transformation in cerebral ischemic stroke.
Collapse
Affiliation(s)
- Chen-Lin Gu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Lu Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan 030001, China
| | - Yan Zhu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Ting-Yu Bao
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Yu-Ting Zhu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Yu-Tong Chen
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Han-Qing Pang
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
2
|
Huang KC, Chiang YF, Ali M, Hsia SM. Oleocanthal mitigates CoCl 2-induced oxidative damage and apoptosis via regulating MAPK pathway in human retinal pigment epithelial cells. Biomed Pharmacother 2024; 180:117582. [PMID: 39467472 DOI: 10.1016/j.biopha.2024.117582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
Retinal hypoxia causes severe visual impairment and dysfunction in retinal pigment epithelial (RPE) cells, triggering a cascade of events leading to cellular apoptosis. Oxidative stress induced by hypoxia plays a significant role in the development of retinal diseases; however, the precise pathogenesis remains unclear. Oleocanthal, a phenolic compound in extra virgin olive oil, is known for its diverse biological properties. This study aims to investigate the potential anti-oxidative effects of oleocanthal against CoCl2-induced hypoxia in ARPE-19 cells. The cell culture model enabled the evaluation of apoptosis, DNA damage, and ROS levels using MTT assay, Western blot, Annexin V/PI staining, JC-1 staining, MitoSOX, H2DCFDA, immunocytochemistry, and comet assays. Our results showed that oleocanthal effectively protected RPE cells against CoCl2-induced damage by enhancing cell viability, reducing DNA damage, and decreasing ROS levels. Moreover, oleocanthal attenuated CoCl2-induced MMP loss by elevating the JC-1 aggregate/monomer ratio. Furthermore, CoCl2-induced cell apoptosis via up-regulating MAPK signaling, while oleocanthal mitigated this effect. These findings shed light on the molecular mechanisms underlying oleocanthal's protection against oxidative stress induced by hypoxia, offering potential insights for the development of novel therapeutic agents for retinal hypoxia.
Collapse
Affiliation(s)
- Ko-Chieh Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Fen Chiang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Mohamed Ali
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan; School of Food and Safety, Taipei Medical University, Taipei 11031, Taiwan; Nutrition Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan; TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| |
Collapse
|
3
|
Porel P, Bala K, Aran KR. Exploring the role of HIF-1α on pathogenesis in Alzheimer's disease and potential therapeutic approaches. Inflammopharmacology 2024:10.1007/s10787-024-01585-x. [PMID: 39465478 DOI: 10.1007/s10787-024-01585-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
Hypoxia-inducible factor 1α (HIF-1α) is a crucial transcription factor that regulates cellular responses to low oxygen levels (hypoxia). In Alzheimer's disease (AD), emerging evidence suggests a significant involvement of HIF-1α in disease pathogenesis. AD is characterized by the accumulation of amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFTs), leading to neuronal dysfunction and cognitive decline. HIF-1α is implicated in AD through its multifaceted roles in various cellular processes. Firstly, in response to hypoxia, HIF-1α promotes the expression of genes involved in angiogenesis, which is crucial for maintaining cerebral blood flow and oxygen delivery to the brain. However, in the context of AD, dysregulated HIF-1α activation may exacerbate cerebral hypoperfusion, contributing to neuronal damage. Moreover, HIF-1α is implicated in the regulation of Aβ metabolism. It can influence the production and clearance of Aβ peptides, potentially modulating their accumulation and toxicity in the brain. Additionally, HIF-1α activation has been linked to neuroinflammation, a key feature of AD pathology. It can promote the expression of pro-inflammatory cytokines and exacerbate neuronal damage. Furthermore, HIF-1α may play a role in synaptic plasticity and neuronal survival, which are impaired in AD. Dysregulated HIF-1α signaling could disrupt these processes, contributing to cognitive decline and neurodegeneration. Overall, the involvement of HIF-1α in various aspects of AD pathophysiology highlights its potential as a therapeutic target. Modulating HIF-1α activity could offer novel strategies for mitigating neurodegeneration and preserving cognitive function in AD patients. However, further research is needed to elucidate the precise mechanisms underlying HIF-1α dysregulation in AD and to develop targeted interventions.
Collapse
Affiliation(s)
- Pratyush Porel
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Kanchan Bala
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Khadga Raj Aran
- Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
4
|
Gao Y, Zhang J, Tang T, Liu Z. Hypoxia Pathways in Parkinson's Disease: From Pathogenesis to Therapeutic Targets. Int J Mol Sci 2024; 25:10484. [PMID: 39408813 PMCID: PMC11477385 DOI: 10.3390/ijms251910484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
The human brain is highly dependent on oxygen, utilizing approximately 20% of the body's oxygen at rest. Oxygen deprivation to the brain can lead to loss of consciousness within seconds and death within minutes. Recent studies have identified regions of the brain with spontaneous episodic hypoxia, referred to as "hypoxic pockets". Hypoxia can also result from impaired blood flow due to conditions such as heart disease, blood clots, stroke, or hemorrhage, as well as from reduced oxygen intake or excessive oxygen consumption caused by factors like low ambient oxygen, pulmonary diseases, infections, inflammation, and cancer. Severe hypoxia in the brain can manifest symptoms similar to Parkinson's disease (PD), including cerebral edema, mood disturbances, and cognitive impairments. Additionally, the development of PD appears to be closely associated with hypoxia and hypoxic pathways. This review seeks to investigate the molecular interactions between hypoxia and PD, emphasizing the pathological role of hypoxic pathways in PD and exploring their potential as therapeutic targets.
Collapse
Affiliation(s)
- Yuanyuan Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (Y.G.); (J.Z.)
| | - Jiarui Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (Y.G.); (J.Z.)
| | - Tuoxian Tang
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Zhenjiang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; (Y.G.); (J.Z.)
| |
Collapse
|
5
|
Alehashem M, Alcaraz AJ, Hogan N, Weber L, Siciliano SD, Hecker M. Linking pesticide exposure to neurodegenerative diseases: An in vitro investigation with human neuroblastoma cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 933:173041. [PMID: 38723972 DOI: 10.1016/j.scitotenv.2024.173041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/05/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024]
Abstract
Although many organochlorine pesticides (OCPs) have been banned or restricted because of their persistence and linkage to neurodegenerative diseases, there is evidence of continued human exposure. In contrast, registered herbicides are reported to have a moderate to low level of toxicity; however, there is little information regarding their toxicity to humans or their combined effects with OCPs. This study aimed to characterize the mechanism of toxicity of banned OCP insecticides (aldrin, dieldrin, heptachlor, and lindane) and registered herbicides (trifluralin, triallate, and clopyralid) detected at a legacy contaminated pesticide manufacturing and packing site using SH-SY5Y cells. Cell viability, LDH release, production of reactive oxygen species (ROS), and caspase 3/7 activity were evaluated following 24 h of exposure to the biocides. In addition, RNASeq was conducted at sublethal concentrations to investigate potential mechanisms involved in cellular toxicity. Our findings suggested that aldrin and heptachlor were the most toxic, while dieldrin, lindane, trifluralin, and triallate exhibited moderate toxicity, and clopyralid was not toxic to SH-SY5Y cells. While aldrin and heptachlor induced their toxicity through damage to the cell membrane, the toxicity of dieldrin was partially attributed to necrosis and apoptosis. Moreover, toxic effects of lindane, trifluralin, and triallate, at least partially, were associated with ROS generation. Gene expression profiles suggested that decreased cell viability induced by most of the tested biocides was related to inhibited cell proliferation. The dysregulation of genes encoding for proteins with anti-apoptotic properties also supported the absence of caspase activation. Identified enriched terms showed that OCP toxicity in SH-SY5Y cells was mediated through pathways associated with the pathogenesis of neurodegenerative diseases. In conclusion, this study provides a basis for elucidating the molecular mechanisms of pesticide-induced neurotoxicity. Moreover, it introduced SH-SY5Y cells as a relevant in vitro model for investigating the neurotoxicity of pesticides in humans.
Collapse
Affiliation(s)
- M Alehashem
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - A J Alcaraz
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - N Hogan
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada; Department of Animal Science, University of Saskatchewan, Saskatoon, SK, Canada
| | - L Weber
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - S D Siciliano
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada; Department of Soil Science, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - M Hecker
- Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada; School of Environment and Sustainability, University of Saskatchewan, Saskatoon, SK S7N 5C8, Canada.
| |
Collapse
|
6
|
Hou Y, Fan F, Xie N, Zhang Y, Wang X, Meng X. Rhodiola crenulata alleviates hypobaric hypoxia-induced brain injury by maintaining BBB integrity and balancing energy metabolism dysfunction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155529. [PMID: 38503156 DOI: 10.1016/j.phymed.2024.155529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/25/2024] [Accepted: 03/08/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND/PURPOSE Rhodiola crenulata (Hook. f. et Thoms.) H. Ohba (R. crenulate), a famous and characteristic Tibetan medicine, has been demonstrated to exert an outstanding brain protection role in the treatment of high-altitude hypoxia disease. However, the metabolic effects of R. crenulate on high-altitude hypoxic brain injury (HHBI) are still incompletely understood. Herein, the anti-hypoxic effect and associated mechanisms of R. crenulate were explored through both in vivo and in vitro experiments. STUDY DESIGN/METHODS The mice model of HHBI was established using an animal hypobaric and hypoxic chamber. R. crenulate extract (RCE, 0.5, 1.0 and 2.0 g/kg) and salidroside (Sal, 25, 50 and 100 mg/kg) was given by gavage for 7 days. Pathological changes and neuronal apoptosis of mice hippocampus and cortex were evaluated using H&E and TUNEL staining, respectively. The effects of RCE and Sal on the permeability of blood brain barrier (BBB) were detected by Evans blue staining and NIR-II fluorescence imaging. Meanwhile, the ultrastructural BBB and cerebrovascular damages were observed using a transmission electron microscope (TEM). The levels of tight junction proteins Claudin-1, ZO-1 and occludin were detected by immunofluorescence. Additionally, the metabolites in mice serum and brain were determined using UHPLC-MS and MALDI-MSI analysis. The cell viability of Sal on hypoxic HT22 cells induced by CoCl2 was investigated by cell counting kit-8. The contents of LDH, MDA, SOD, GSH-PX and SDH were detected by using commercial biochemical kits. Meanwhile, intracellular ROS, Ca2+ and mitochondrial membrane potential were determined by corresponding specific labeled probes. The intracellular metabolites of HT22 cells were performed by the targeted metabolomics analysis of the Q300 kit. The cell apoptosis and necrosis were examined by YO-PRO-1/PI, Annexin V/PI and TUNEL staining. In addition, mitochondrial morphology was tested by Mito-tracker red with confocal microscopy and TEM. Real-time ATP production, oxygen consumption rate, and proton efflux rate were measured using a Seahorse analyzer. Subsequently, MCU, OPA1, p-Drp1ser616, p-AMPKα, p-AMPKβ and Sirt1 were determined by immunofluorescent and western blot analyses. RESULTS The results demonstrated that R. crenulate and Sal exert anti-hypoxic brain protection from inhibiting neuronal apoptosis, maintaining BBB integrity, increasing tight junction protein Claudin-1, ZO-1 and occludin and improving mitochondrial morphology and function. Mechanistically, R. crenulate and Sal alleviated HHBI by enhancing the tricarboxylic acid cycle to meet the demand of energy of brain. Additionally, experiments in vitro confirmed that Sal could ameliorate the apoptosis of HT22 cells, improve mitochondrial morphology and energy metabolism by enhancing mitochondrial respiration and glycolysis. Meanwhile, Sal-mediated MCU inhibited the activation of Drp1 and enhanced the expression of OPA1 to maintain mitochondrial homeostasis, as well as activation of AMPK and Sirt1 to enhance ATP production. CONCLUSION Collectively, the findings suggested that RCE and Sal may afford a protective intervention in HHBI through maintaining BBB integrity and improving energy metabolism via balancing MCU-mediated mitochondrial homeostasis by activating the AMPK/Sirt1 signaling pathway.
Collapse
Affiliation(s)
- Ya Hou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Fuhan Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Na Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Xiaobo Wang
- Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, 620010, China.
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China; Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, 620010, China.
| |
Collapse
|
7
|
Sun M, Baker TL, Wilson CT, Brady RD, Yamakawa GR, Wright DK, Mychasiuk R, Vo A, Wilson T, Allen J, McDonald SJ, Shultz SR. Treatment with the vascular endothelial growth factor-A antibody, bevacizumab, has sex-specific effects in a rat model of mild traumatic brain injury. J Cereb Blood Flow Metab 2024; 44:542-555. [PMID: 37933736 PMCID: PMC10981407 DOI: 10.1177/0271678x231212377] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023]
Abstract
Mild traumatic brain injury (mTBI) involves damage to the cerebrovascular system. Vascular endothelial growth factor-A (VEGF-A) is an important modulator of vascular health and VEGF-A promotes the brain's ability to recover after more severe forms of brain injury; however, the role of VEGF-A in mTBI remains poorly understood. Bevacizumab (BEV) is a monoclonal antibody that binds to VEGF-A and neutralises its actions. To better understand the role of VEGF-A in mTBI recovery, this study examined how BEV treatment affected outcomes in rats given a mTBI. Adult Sprague-Dawley rats were assigned to sham-injury + vehicle treatment (VEH), sham-injury + BEV treatment, mTBI + VEH treatment, mTBI + BEV treatment groups. Treatment was administered intracerebroventricularly via a cannula beginning at the time of injury and continuing until the end of the study. Rats underwent behavioral testing after injury and were euthanized on day 11. In both females and males, BEV had a negative impact on cognitive function. mTBI and BEV treatment increased the expression of inflammatory markers in females. In males, BEV treatment altered markers related to hypoxia and vascular health. These novel findings of sex-specific responses to BEV and mTBI provide important insights into the role of VEGF-A in mTBI.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Tamara L Baker
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Campbell T Wilson
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anh Vo
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Trevor Wilson
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Josh Allen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Health Sciences, Vancouver Island University, Nanaimo, BC, Canada
| |
Collapse
|
8
|
Zhu L, Xin YJ, He M, Bian J, Cheng XL, Li R, Li JJ, Wang J, Liu JY, Yang L. Downregulation of miR-337-3p in hypoxia/reoxygenation neuroblastoma cells increases KCTD11 expression. J Biochem Mol Toxicol 2024; 38:e23685. [PMID: 38495002 DOI: 10.1002/jbt.23685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/18/2023] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Neurodegeneration is linked to the progressive loss of neural function and is associated with several diseases. Hypoxia is a hallmark in many of these diseases, and several therapies have been developed to treat this disease, including gene expression therapies that should be tightly controlled to avoid side effects. Cells experiencing hypoxia undergo a series of physiological responses that are induced by the activation of various transcription factors. Modulation of microRNA (miRNA) expression to alter transcriptional regulation has been demonstrated to be beneficial in treating multiple diseases, and in this study, we therefore explored potential miRNA candidates that could influence hypoxia-induced nerve cell death. Our data suggest that in mouse neuroblasts Neuro-2a cells with hypoxia/reoxygenation (H/R), miR-337-3p is downregulated to increase the expression of Potassium channel tetramerization domain containing 11 (KCTD11) and subsequently promote apoptosis. Here, we demonstrate for the first time that KCTD11 plays a role in the cellular response to hypoxia, and we also provide a possible regulatory mechanism by identifying the axis of miR-337-3p/KCTD11 as a promising candidate modulator of nerve cell survival after H/R exposure.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Yi-Juan Xin
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Mu He
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Jun Bian
- Department of General Surgery, Xi'an Jiaotong University Affiliated Children's Hospital, Xi'an, Shaanxi, China
| | - Xiao-Li Cheng
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Rui Li
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Jin-Jie Li
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Juan Wang
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Jia-Yun Liu
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Liu Yang
- Department of Clinical Laboratory, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
9
|
Pena E, San Martin-Salamanca R, El Alam S, Flores K, Arriaza K. Tau Protein Alterations Induced by Hypobaric Hypoxia Exposure. Int J Mol Sci 2024; 25:889. [PMID: 38255962 PMCID: PMC10815386 DOI: 10.3390/ijms25020889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Tauopathies are a group of neurodegenerative diseases whose central feature is dysfunction of the microtubule-associated protein tau (MAPT). Although the exact etiology of tauopathies is still unknown, it has been hypothesized that their onset may occur up to twenty years before the clear emergence of symptoms, which has led to questions about whether the prognosis of these diseases can be improved by, for instance, targeting the factors that influence tauopathy development. One such factor is hypoxia, which is strongly linked to Alzheimer's disease because of its association with obstructive sleep apnea and has been reported to affect molecular pathways related to the dysfunction and aggregation of tau proteins and other biomarkers of neurological damage. In particular, hypobaric hypoxia exposure increases the activation of several kinases related to the hyperphosphorylation of tau in neuronal cells, such as ERK, GSK3β, and CDK5. In addition, hypoxia also increases the levels of inflammatory molecules (IL-β1, IL-6, and TNF-α), which are also associated with neurodegeneration. This review discusses the many remaining questions regarding the influence of hypoxia on tauopathies and the contribution of high-altitude exposure to the development of these diseases.
Collapse
Affiliation(s)
| | | | - Samia El Alam
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1110939, Chile; (E.P.); (R.S.M.-S.); (K.F.); (K.A.)
| | | | | |
Collapse
|
10
|
Sivagurunathan N, Calivarathan L. SARS-CoV-2 Infection to Premature Neuronal Aging and Neurodegenerative Diseases: Is there any Connection with Hypoxia? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:431-448. [PMID: 37073650 DOI: 10.2174/1871527322666230418114446] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 01/28/2023] [Accepted: 02/09/2023] [Indexed: 04/20/2023]
Abstract
The pandemic of coronavirus disease-2019 (COVID-19), caused by SARS-CoV-2, has become a global concern as it leads to a spectrum of mild to severe symptoms and increases death tolls around the world. Severe COVID-19 results in acute respiratory distress syndrome, hypoxia, and multi- organ dysfunction. However, the long-term effects of post-COVID-19 infection are still unknown. Based on the emerging evidence, there is a high possibility that COVID-19 infection accelerates premature neuronal aging and increases the risk of age-related neurodegenerative diseases in mild to severely infected patients during the post-COVID period. Several studies correlate COVID-19 infection with neuronal effects, though the mechanism through which they contribute to the aggravation of neuroinflammation and neurodegeneration is still under investigation. SARS-CoV-2 predominantly targets pulmonary tissues and interferes with gas exchange, leading to systemic hypoxia. The neurons in the brain require a constant supply of oxygen for their proper functioning, suggesting that they are more vulnerable to any alteration in oxygen saturation level that results in neuronal injury with or without neuroinflammation. We hypothesize that hypoxia is one of the major clinical manifestations of severe SARS-CoV-2 infection; it directly or indirectly contributes to premature neuronal aging, neuroinflammation, and neurodegeneration by altering the expression of various genes responsible for the survival of the cells. This review focuses on the interplay between COVID-19 infection, hypoxia, premature neuronal aging, and neurodegenerative diseases and provides a novel insight into the molecular mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Narmadhaa Sivagurunathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| | - Latchoumycandane Calivarathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| |
Collapse
|
11
|
Shushanyan R, Grigoryan A, Abgaryan T, Karapetyan A. Histological and cytochemical analysis of the brain under conditions of hypobaric hypoxia-induced oxygen deficiency in albino rats. Acta Histochem 2023; 125:152114. [PMID: 37980852 DOI: 10.1016/j.acthis.2023.152114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/21/2023]
Abstract
High altitude sickness is a life-threatening disease that occurs among acclimatized individuals working or living at a high altitude accompanied by hypobaric hypoxia exposure. The prolonged influence of hypobaric hypoxia on the brain may trigger neuronal damage and cell death due to an oxygen deficiency. The purpose of the current study was to investigate the histomorphological changes in the hippocampus, cerebral cortex, cerebellar cortex, and striatum of the rat's brain following chronic hypobaric hypoxia. Fourteen albino rats were used for this investigation. The animals were exposed to chronic hypobaric hypoxia in the special decompression chamber at an altitude of 7000 m for 7 days. The histological analysis was conducted via toluidine staining and silver impregnation. DNA damage and cell apoptosis were assessed via Feulgen staining. The histochemical assessment revealed increased dark neurons in the hippocampus with cell swelling. Silver impregnation showed increased argyrophilic neurons in the cerebellar cortex, striatum, CA1 subfield of the hippocampus, and cerebral cortex. The cytochemical analysis determined the increased apoptotic cells with hyperchromatic condensation and pyknosis in the hippocampus subfields and cerebral cortex. In addition, it has been observed that hypoxia has resulted in small hemorrhages and perivascular edema within the cerebellar and cerebral cortex. The results indicate brain injury observed in the various parts of the brain towards hypobaric hypoxia, however, the hippocampus showed greater vulnerability against hypoxic exposure in comparison to the striatum, cerebellum, and cerebral cortex. These changes support our insights regarding brain intolerance under conditions of hypoxia-induced oxygen deficiency and its histomorphological manifestations.
Collapse
Affiliation(s)
- Ruzanna Shushanyan
- Department of Human and Animal Physiology, Yerevan State University, Yerevan, Armenia.
| | - Anna Grigoryan
- Department of Human and Animal Physiology, Yerevan State University, Yerevan, Armenia.
| | - Tamara Abgaryan
- Department of Human and Animal Physiology, Yerevan State University, Yerevan, Armenia.
| | - Anna Karapetyan
- Department of Human and Animal Physiology, Yerevan State University, Yerevan, Armenia.
| |
Collapse
|
12
|
Zhuang XM, Guo ZY, Zhang M, Chen YH, Qi FN, Wang RQ, Zhang L, Zhao PJ, Lu CJ, Zou CG, Ma YC, Xu J, Zhang KQ, Cao YR, Liang LM. Ethanol mediates the interaction between Caenorhabditis elegans and the nematophagous fungus Purpureocillium lavendulum. Microbiol Spectr 2023; 11:e0127023. [PMID: 37560934 PMCID: PMC10580998 DOI: 10.1128/spectrum.01270-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 08/11/2023] Open
Abstract
Accurately recognizing pathogens by the host is vital for initiating appropriate immune response against infecting microorganisms. Caenorhabditis elegans has no known receptor to recognize pathogen-associated molecular pattern. However, recent studies showed that nematodes have a strong specificity for transcriptomes infected by different pathogens, indicating that they can identify different pathogenic microorganisms. However, the mechanism(s) for such specificity remains largely unknown. In this study, we showed that the nematophagous fungus Purpureocillium lavendulum can infect the intestinal tract of the nematode C. elegans and the infection led to the accumulation of reactive oxygen species (ROS) in the infected intestinal tract, which suppressed fungal growth. Co-transcriptional analysis revealed that fungal genes related to anaerobic respiration and ethanol production were up-regulated during infection. Meanwhile, the ethanol dehydrogenase Sodh-1 in C. elegans was also up-regulated. Together, these results suggested that the infecting fungi encounter hypoxia stress in the nematode gut and that ethanol may play a role in the host-pathogen interaction. Ethanol production in vitro during fungal cultivation in hypoxia conditions was confirmed by gas chromatography-mass spectrometry. Direct treatment of C. elegans with ethanol elevated the sodh-1 expression and ROS accumulation while repressing a series of immunity genes that were also repressed during fungal infection. Mutation of sodh-1 in C. elegans blocked ROS accumulation and increased the nematode's susceptibility to fungal infection. Our study revealed a new recognition and antifungal mechanism in C. elegans. The novel mechanism of ethanol-mediated interaction between the fungus and nematode provides new insights into fungal pathogenesis and for developing alternative biocontrol of pathogenic nematodes by nematophagous fungi. IMPORTANCE Nematodes are among the most abundant animals on our planet. Many of them are parasites in animals and plants and cause human and animal health problems as well as agricultural losses. Studying the interaction of nematodes and their microbial pathogens is of great importance for the biocontrol of animal and plant parasitic nematodes. In this study, we found that the model nematode Caenorhabditis elegans can recognize its fungal pathogen, the nematophagous fungus Purpureocillium lavendulum, through fungal-produced ethanol. Then the nematode elevated the reactive oxygen species production in the gut to inhibit fungal growth in an ethanol dehydrogenase-dependent manner. With this mechanism, novel biocontrol strategies may be developed targeting the ethanol receptor or metabolic pathway of nematodes. Meanwhile, as a volatile organic compound, ethanol should be taken seriously as a vector molecule in the microbial-host interaction in nature.
Collapse
Affiliation(s)
- Xue-Mei Zhuang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Zhi-Yi Guo
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Meng Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Yong-Hong Chen
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Feng-Na Qi
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Ren-Qiao Wang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Ling Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Pei-Ji Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Chao-Jun Lu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Cheng-Gang Zou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Yi-Cheng Ma
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Jianping Xu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Ke-Qin Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| | - Yan-Ru Cao
- College of Agriculture and Life Sciences, Kunming University, Kunming, China
| | - Lian-Ming Liang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and The Key Laboratory for Southwest Microbial Diversity of the Ministry of Education, Yunnan University, Kunming, China
| |
Collapse
|
13
|
Mitroshina EV, Krivonosov MI, Pakhomov AM, Yarullina LE, Gavrish MS, Mishchenko TA, Yarkov RS, Vedunova MV. Unravelling the Collective Calcium Dynamics of Physiologically Aged Astrocytes under a Hypoxic State In Vitro. Int J Mol Sci 2023; 24:12286. [PMID: 37569663 PMCID: PMC10419080 DOI: 10.3390/ijms241512286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/24/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Astrocytes serve many functions in the brain related to maintaining nerve tissue homeostasis and regulating neuronal function, including synaptic transmission. It is assumed that astrocytes are crucial players in determining the physiological or pathological outcome of the brain aging process and the development of neurodegenerative diseases. Therefore, studies on the peculiarities of astrocyte physiology and interastrocytic signaling during aging are of utmost importance. Calcium waves are one of the main mechanisms of signal transmission between astrocytes, and in the present study we investigated the features of calcium dynamics in primary cultures of murine cortical astrocytes in physiological aging and hypoxia modeling in vitro. Specifically, we focused on the assessment of calcium network dynamics and the restructuring of the functional network architecture in primary astrocytic cultures. Calcium imaging was performed on days 21 ("young" astrocyte group) and 150 ("old" astrocyte group) of cultures' development in vitro. While the number of active cells and frequency of calcium events were decreased, we observed a reduced degree of correlation in calcium dynamics between neighboring cells, which was accompanied by a reduced number of functionally connected cells with fewer and slower signaling events. At the same time, an increase in the mRNA expression of anti-apoptotic factor Bcl-2 and connexin 43 was observed in "old" astrocytic cultures, which can be considered as a compensatory response of cells with a decreased level of intercellular communication. A hypoxic episode aggravates the depression of the connectivity of calcium dynamics of "young" astrocytes rather than that of "old" ones.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Mikhail I. Krivonosov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Alexander M. Pakhomov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
- Federal Research Center Institute of Applied Physics of the Russian Academy of Sciences (IAP RAS), 603950 Nizhny Novgorod, Russia
| | - Laysan E. Yarullina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Maria S. Gavrish
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Roman S. Yarkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia; (E.V.M.); (A.M.P.); (L.E.Y.); (M.S.G.); (T.A.M.); (R.S.Y.)
| |
Collapse
|
14
|
Jiang M, Jang SE, Zeng L. The Effects of Extrinsic and Intrinsic Factors on Neurogenesis. Cells 2023; 12:cells12091285. [PMID: 37174685 PMCID: PMC10177620 DOI: 10.3390/cells12091285] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
In the mammalian brain, neurogenesis is maintained throughout adulthood primarily in two typical niches, the subgranular zone (SGZ) of the dentate gyrus and the subventricular zone (SVZ) of the lateral ventricles and in other nonclassic neurogenic areas (e.g., the amygdala and striatum). During prenatal and early postnatal development, neural stem cells (NSCs) differentiate into neurons and migrate to appropriate areas such as the olfactory bulb where they integrate into existing neural networks; these phenomena constitute the multistep process of neurogenesis. Alterations in any of these processes impair neurogenesis and may even lead to brain dysfunction, including cognitive impairment and neurodegeneration. Here, we first summarize the main properties of mammalian neurogenic niches to describe the cellular and molecular mechanisms of neurogenesis. Accumulating evidence indicates that neurogenesis plays an integral role in neuronal plasticity in the brain and cognition in the postnatal period. Given that neurogenesis can be highly modulated by a number of extrinsic and intrinsic factors, we discuss the impact of extrinsic (e.g., alcohol) and intrinsic (e.g., hormones) modulators on neurogenesis. Additionally, we provide an overview of the contribution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection to persistent neurological sequelae such as neurodegeneration, neurogenic defects and accelerated neuronal cell death. Together, our review provides a link between extrinsic/intrinsic factors and neurogenesis and explains the possible mechanisms of abnormal neurogenesis underlying neurological disorders.
Collapse
Affiliation(s)
- Mei Jiang
- Department of Human Anatomy, Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Dongguan Campus, Guangdong Medical University, Dongguan 523808, China
| | - Se Eun Jang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, 11 Mandalay Road, Singapore 308232, Singapore
| |
Collapse
|
15
|
Sun M, Baker TL, Wilson CT, Brady RD, Mychasiuk R, Yamakawa GR, Vo A, Wilson T, McDonald SJ, Shultz SR. Treatment with vascular endothelial growth factor-A worsens cognitive recovery in a rat model of mild traumatic brain injury. Front Mol Neurosci 2022; 15:937350. [PMID: 36385769 PMCID: PMC9643175 DOI: 10.3389/fnmol.2022.937350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/29/2022] [Indexed: 09/08/2023] Open
Abstract
Mild traumatic brain injury (mTBI) is a common and unmet clinical issue, with limited treatments available to improve recovery. The cerebrovascular system is vital to provide oxygen and nutrition to the brain, and a growing body of research indicates that cerebrovascular injury contributes to mTBI symptomatology. Vascular endothelial growth factor-A (VEGF-A) is a potent promoter of angiogenesis and an important modulator of vascular health. While indirect evidence suggests that increased bioavailability of VEGF-A may be beneficial after mTBI, the direct therapeutic effects of VEGF-A in this context remains unknown. This study therefore aimed to determine whether intracerebroventricular administration of recombinant VEGF-A could improve recovery from mTBI in a rat model. Male and female Sprague-Dawley rats were assigned to four groups: sham + vehicle (VEH), sham + VEGF-A, mTBI + VEH, mTBI + VEGF-A. The mTBI was induced using the lateral impact model, and treatment began at the time of the injury and continued until the end of the study. Rats underwent behavioral testing between days 1 and 10 post-injury, and were euthanized on day 11 for post-mortem analysis. In males, the mTBI + VEGF-A group had significantly worse cognitive recovery in the water maze than all other groups. In females, the VEGF treatment worsened cognitive performance in the water maze regardless of mTBI or sham injury. Analysis of hippocampal tissue found that these cognitive deficits occurred in the presence of gene expression changes related to neuroinflammation and hypoxia in both male and female rats. These findings indicate that the VEGF-A treatment paradigm tested in this study failed to improve mTBI outcomes in either male or female rats.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Tamara L. Baker
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Campbell T. Wilson
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rhys D. Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Glenn R. Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anh Vo
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Trevor Wilson
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
- Health and Human Services, Vancouver Island University, Nanaimo, BC, Canada
| |
Collapse
|
16
|
Kosyreva AM, Sentyabreva AV, Tsvetkov IS, Makarova OV. Alzheimer’s Disease and Inflammaging. Brain Sci 2022; 12:brainsci12091237. [PMID: 36138973 PMCID: PMC9496782 DOI: 10.3390/brainsci12091237] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 09/10/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer’s disease is one of the most common age-related neurodegenerative disorders. The main theory of Alzheimer’s disease progress is the amyloid-β cascade hypothesis. However, the initial mechanisms of insoluble forms of amyloid-β formation and hyperphosphorylated tau protein in neurons remain unclear. One of the factors, which might play a key role in senile plaques and tau fibrils generation due to Alzheimer’s disease, is inflammaging, i.e., systemic chronic low-grade age-related inflammation. The activation of the proinflammatory cell phenotype is observed during aging, which might be one of the pivotal mechanisms for the development of chronic inflammatory diseases, e.g., atherosclerosis, metabolic syndrome, type 2 diabetes mellitus, and Alzheimer’s disease. This review discusses the role of the inflammatory processes in developing neurodegeneration, activated during physiological aging and due to various diseases such as atherosclerosis, obesity, type 2 diabetes mellitus, and depressive disorders.
Collapse
|
17
|
Zhang T, Guo R, Li Y, Zhao Y, Li Y, Liang ZP. T 2 ' mapping of the brain from water-unsuppressed 1 H-MRSI and turbo spin-echo data. Magn Reson Med 2022; 88:2198-2207. [PMID: 35844075 DOI: 10.1002/mrm.29386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/14/2022] [Accepted: 06/22/2022] [Indexed: 11/08/2022]
Abstract
PURPOSE To obtain high-quality T 2 ' $$ {\mathrm{T}}_2^{\prime } $$ maps of brain tissues from water-unsuppressed magnetic resonance spectroscopic imaging (MRSI) and turbo spin-echo (TSE) data. METHODS T 2 ' $$ {\mathrm{T}}_2^{\prime } $$ mapping can be achieved using T 2 * $$ {\mathrm{T}}_2^{\ast } $$ mapping from water-unsuppressed MRSI data and T 2 $$ {\mathrm{T}}_2 $$ mapping from TSE data. However, T 2 * $$ {\mathrm{T}}_2^{\ast } $$ mapping often suffers from signal dephasing and distortions caused by B 0 $$ {\mathrm{B}}_0 $$ field inhomogeneity; T 2 $$ {\mathrm{T}}_2 $$ measurements may be biased due to system imperfections, especially for T 2 $$ {\mathrm{T}}_2 $$ -weighted image with small number of TEs. In this work, we corrected the B 0 $$ {\mathrm{B}}_0 $$ field inhomogeneity effect on T 2 * $$ {\mathrm{T}}_2^{\ast } $$ mapping using a subspace model-based method, incorporating pre-learned spectral basis functions of the water signals. T 2 $$ {\mathrm{T}}_2 $$ estimation bias was corrected using a TE-adjustment method, which modeled the deviation between measured and reference T 2 $$ {\mathrm{T}}_2 $$ decays as TE shifts. RESULTS In vivo experiments were performed to evaluate the performance of the proposed method. High-quality T 2 * $$ {\mathrm{T}}_2^{\ast } $$ maps were obtained in the presence of large field inhomogeneity in the prefrontal cortex. Bias in T 2 $$ {\mathrm{T}}_2 $$ measurements obtained from TSE data was effectively reduced. Based on the T 2 * $$ {\mathrm{T}}_2^{\ast } $$ and T 2 $$ {\mathrm{T}}_2 $$ measurements produced by the proposed method, high-quality T 2 ' $$ {\mathrm{T}}_2^{\prime } $$ maps were obtained, along with neurometabolite maps, from MRSI and TSE data that were acquired in about 9 min. The results obtained from acute stroke and glioma patients demonstrated the feasibility of the proposed method in the clinical setting. CONCLUSIONS High-quality T 2 ' $$ {\mathrm{T}}_2^{\prime } $$ maps can be obtained from water-unsuppressed 1 H-MRSI and TSE data using the proposed method. With further development, this method may lay a foundation for simultaneously imaging oxygenation and neurometabolic alterations of brain disorders.
Collapse
Affiliation(s)
- Tianxiao Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Rong Guo
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Yudu Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Yibo Zhao
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Yao Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Pei Liang
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
18
|
Gursoy M, Gul Z, Buyukuysal RL. Sigma receptor ligands haloperidol and ifenprodil attenuate hypoxia induced dopamine release in rat striatum. Neurol Res 2022; 44:927-936. [DOI: 10.1080/01616412.2022.2072100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Murat Gursoy
- Bursa Provincial Health Directorate, T.C. Ministry of Health, Bursa, Turkey
| | - Zulfiye Gul
- Department of Medical Pharmacology, Faculty of Medicine, Bahcesehir University, Istanbul, Turkey
| | - R. Levent Buyukuysal
- Department of Medical Pharmacology, Faculty of Medicine, Uludag University, Bursa, Turkey
| |
Collapse
|
19
|
Song D, Zhang J, Li J, Kong X, Jiang Y, Xu J, Zhang X, Zhao Q. Effective Parts of Gentiana straminea Maxim Attenuates Hypoxia-Induced Oxidative Stress and Apoptosis. Dose Response 2022; 20:15593258221100986. [PMID: 35602583 PMCID: PMC9121479 DOI: 10.1177/15593258221100986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxia occurs in physiological situations and several pathological situations, inducing oxidative stress. G straminea Maxim (G.s Maxim) is a traditional Tibetan medicine that exerts several biological effects. This study focused on the protective effects of G.s Maxim in hypoxia-induced oxidative stress and apoptosis. We found that G.s Maxim significantly increased survival and reduced oxidative stress in hypoxic mice. Various extraction parts of G.s Maxim showed antioxidant activity and significantly improved survival in hypoxia-injured PC12 cells. G.s Maxim reduced hypoxia-induced cell apoptosis and leakage of lactate dehydrogenase. Hypoxic cells had increased malondialdehyde levels but reduced superoxide dismutase activity and G.s Maxim reversed these effects. Moreover, G.s Maxim suppressed hypoxia-induced apoptosis by inducing protein expression of B cell leukemia/lymphoma-2 and reducing the expression of hypoxia-inducible factor-1α, Bcl-2-associated X, and nuclear factor-k-gene binding. These findings suggest that G.s Maxim attenuates hypoxia-induced injury associated with oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Dan Song
- Joint Laboratory for Research on Active
Components and Pharmacological Mechanism of Tibetan Materia Medica of Tibetan Medical
Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Jingyu Zhang
- Department of Pathology, Cangzhou Central Hospital, Cangzhou, China
| | - Jie Li
- Joint Laboratory for Research on Active
Components and Pharmacological Mechanism of Tibetan Materia Medica of Tibetan Medical
Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Xiumei Kong
- Joint Laboratory for Research on Active
Components and Pharmacological Mechanism of Tibetan Materia Medica of Tibetan Medical
Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Yi Jiang
- Joint Laboratory for Research on Active
Components and Pharmacological Mechanism of Tibetan Materia Medica of Tibetan Medical
Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Jiaojiao Xu
- Joint Laboratory for Research on Active
Components and Pharmacological Mechanism of Tibetan Materia Medica of Tibetan Medical
Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Xiaoying Zhang
- Joint Laboratory for Research on Active
Components and Pharmacological Mechanism of Tibetan Materia Medica of Tibetan Medical
Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Qin Zhao
- Joint Laboratory for Research on Active
Components and Pharmacological Mechanism of Tibetan Materia Medica of Tibetan Medical
Research Center of Tibet, School of Medicine, Xizang Minzu University, Xianyang, China
| |
Collapse
|
20
|
Tsang HW, Bhatia I, Chan KW, Chan GCF, Ip P, Cheung PT. Transmembrane 29 (Tmem29), a Newly Identified Molecule Showed Downregulation in Hypoxic-Ischemic Brain Damage. NEUROSCI 2022; 3:41-51. [PMID: 39484674 PMCID: PMC11523738 DOI: 10.3390/neurosci3010003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/30/2021] [Indexed: 11/03/2024] Open
Abstract
Transmembrane 29 (Tmem29) gene with unknown function is a gene located on the X chromosome of the mouse genome. The gene showed differential expression in the Vannucci neonatal hypoxic-ischemic mouse brain model. We found the gene expresses with different molecular forms, including a group of long non-coding RNA forming a family of transcripts. It was predominantly expressed in the testes, brain, and kidney of mouse. In vitro identification and functional characterization were carried out in Neuro2a cells. Using fluorescence microscopy, Tmem29 protein was found to be constitutively expressed in mouse cell lines of different origins. Oxygen glucose deprivation (OGD) induced apoptotic cell death in Neuro2a cells and was confirmed by activations of caspase 3. Tmem29 protein was found to be associated with cell death especially at the time points of caspase 3 activations. A similar response was obtained in glucose deprivation (GD) cultures suggesting Tmem29 response to a common mechanism induced by OGD and GD. Downregulation of Tmem29 was induced by OGD and GD, further validating its response to hypoxia-ischemia (HI) insults. Our findings contributed to further understanding of molecular events after hypoxic-ischemic insults and opens new avenues for developing protective and therapeutic strategies for hypoxic-ischemic encephalopathy or even pathological programmed cell death.
Collapse
Affiliation(s)
- Hing-Wai Tsang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.-W.T.); (I.B.); (K.-W.C.); (G.C.-F.C.)
| | - Inderjeet Bhatia
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.-W.T.); (I.B.); (K.-W.C.); (G.C.-F.C.)
| | - Koon-Wing Chan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.-W.T.); (I.B.); (K.-W.C.); (G.C.-F.C.)
| | - Godfrey Chi-Fung Chan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.-W.T.); (I.B.); (K.-W.C.); (G.C.-F.C.)
| | - Patrick Ip
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.-W.T.); (I.B.); (K.-W.C.); (G.C.-F.C.)
- Room 123, 1/F, New Clinical Building, Queen Mary Hospital, Hong Kong, China
| | - Pik-To Cheung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (H.-W.T.); (I.B.); (K.-W.C.); (G.C.-F.C.)
- 5/F, Virtus Medical Tower, 122 Queen's Road Central, Hong Kong, China
| |
Collapse
|
21
|
Jha NK, Chen WC, Kumar S, Dubey R, Tsai LW, Kar R, Jha SK, Gupta PK, Sharma A, Gundamaraju R, Pant K, Mani S, Singh SK, Maccioni RB, Datta T, Singh SK, Gupta G, Prasher P, Dua K, Dey A, Sharma C, Mughal YH, Ruokolainen J, Kesari KK, Ojha S. Molecular mechanisms of developmental pathways in neurological disorders: a pharmacological and therapeutic review. Open Biol 2022; 12:210289. [PMID: 35291879 PMCID: PMC8924757 DOI: 10.1098/rsob.210289] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 02/01/2022] [Indexed: 01/07/2023] Open
Abstract
Developmental signalling pathways such as Wnt/β-catenin, Notch and Sonic hedgehog play a central role in nearly all the stages of neuronal development. The term 'embryonic' might appear to be a misnomer to several people because these pathways are functional during the early stages of embryonic development and adulthood, albeit to a certain degree. Therefore, any aberration in these pathways or their associated components may contribute towards a detrimental outcome in the form of neurological disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and stroke. In the last decade, researchers have extensively studied these pathways to decipher disease-related interactions, which can be used as therapeutic targets to improve outcomes in patients with neurological abnormalities. However, a lot remains to be understood in this domain. Nevertheless, there is strong evidence supporting the fact that embryonic signalling is indeed a crucial mechanism as is manifested by its role in driving memory loss, motor impairments and many other processes after brain trauma. In this review, we explore the key roles of three embryonic pathways in modulating a range of homeostatic processes such as maintaining blood-brain barrier integrity, mitochondrial dynamics and neuroinflammation. In addition, we extensively investigated the effect of these pathways in driving the pathophysiology of a range of disorders such as Alzheimer's, Parkinson's and diabetic neuropathy. The concluding section of the review is dedicated to neurotherapeutics, wherein we identify and list a range of biological molecules and compounds that have shown enormous potential in improving prognosis in patients with these disorders.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Wei-Chih Chen
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Sanjay Kumar
- Department of Life Science, School of Basic Science and Research, Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Rajni Dubey
- Department of Medicine Research, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Lung-Wen Tsai
- Department of Medicine Research, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Department of Information Technology Office, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei 110, Taiwan
| | - Rohan Kar
- Indian Institute of Management Ahmedabad (IIMA), Gujarat 380015, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Piyush Kumar Gupta
- Department of Life Science, School of Basic Science and Research, Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Ankur Sharma
- Department of Life Science, School of Basic Science and Research, Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Rohit Gundamaraju
- ER Stress and Mucosal Immunology Laboratory, School of Health Sciences, University of Tasmania, Launceston, Tasmania 7248, Australia
| | - Kumud Pant
- Department of Biotechnology, Graphic Era deemed to be University Dehradun Uttarakhand, 248002 Dehradun, India
| | - Shalini Mani
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector 62, Noida, Uttar Pradesh 201301, India
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, Lucknow 226002, India
| | - Ricardo B. Maccioni
- Laboratory of Neurosciences and Functional Medicine, International Center for Biomedicine (ICC) and Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Tirtharaj Datta
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Gaurav Gupta
- Department of Pharmacology, School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, 302017 Jagatpura, Jaipur, India
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum and Energy Studies, Dehradun 248007, Uttarakhand, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, India
- Department of Applied Physics, School of Science, and
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| | - Yasir Hayat Mughal
- Department of Health Administration, College of Public Health and Health Informatics, Qassim University, Buraidah, Saudi Arabia
| | | | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, and
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo 00076, Finland
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| |
Collapse
|
22
|
Lappano R, Todd LA, Stanic M, Cai Q, Maggiolini M, Marincola F, Pietrobon V. Multifaceted Interplay between Hormones, Growth Factors and Hypoxia in the Tumor Microenvironment. Cancers (Basel) 2022; 14:539. [PMID: 35158804 PMCID: PMC8833523 DOI: 10.3390/cancers14030539] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Hormones and growth factors (GFs) are signaling molecules implicated in the regulation of a variety of cellular processes. They play important roles in both healthy and tumor cells, where they function by binding to specific receptors on target cells and activating downstream signaling cascades. The stages of tumor progression are influenced by hormones and GF signaling. Hypoxia, a hallmark of cancer progression, contributes to tumor plasticity and heterogeneity. Most solid tumors contain a hypoxic core due to rapid cellular proliferation that outgrows the blood supply. In these circumstances, hypoxia-inducible factors (HIFs) play a central role in the adaptation of tumor cells to their new environment, dramatically reshaping their transcriptional profile. HIF signaling is modulated by a variety of factors including hormones and GFs, which activate signaling pathways that enhance tumor growth and metastatic potential and impair responses to therapy. In this review, we summarize the role of hormones and GFs during cancer onset and progression with a particular focus on hypoxia and the interplay with HIF proteins. We also discuss how hypoxia influences the efficacy of cancer immunotherapy, considering that a hypoxic environment may act as a determinant of the immune-excluded phenotype and a major hindrance to the success of adoptive cell therapies.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Lauren A. Todd
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Mia Stanic
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Qi Cai
- Kite Pharma Inc., Santa Monica, CA 90404, USA; (Q.C.); (F.M.)
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | | | | |
Collapse
|
23
|
Olson CA, Iñiguez AJ, Yang GE, Fang P, Pronovost GN, Jameson KG, Rendon TK, Paramo J, Barlow JT, Ismagilov RF, Hsiao EY. Alterations in the gut microbiota contribute to cognitive impairment induced by the ketogenic diet and hypoxia. Cell Host Microbe 2021; 29:1378-1392.e6. [PMID: 34358434 PMCID: PMC8429275 DOI: 10.1016/j.chom.2021.07.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/17/2021] [Accepted: 07/12/2021] [Indexed: 01/16/2023]
Abstract
Many genetic and environmental factors increase susceptibility to cognitive impairment (CI), and the gut microbiome is increasingly implicated. However, the identity of gut microbes associated with CI risk, their effects on CI, and their mechanisms remain unclear. Here, we show that a carbohydrate-restricted (ketogenic) diet potentiates CI induced by intermittent hypoxia in mice and alters the gut microbiota. Depleting the microbiome reduces CI, whereas transplantation of the risk-associated microbiome or monocolonization with Bilophila wadsworthia confers CI in mice fed a standard diet. B. wadsworthia and the risk-associated microbiome disrupt hippocampal synaptic plasticity, neurogenesis, and gene expression. The CI is associated with microbiome-dependent increases in intestinal interferon-gamma (IFNg)-producing Th1 cells. Inhibiting Th1 cell development abrogates the adverse effects of both B. wadsworthia and environmental risk factors on CI. Together, these findings identify select gut bacteria that contribute to environmental risk for CI in mice by promoting inflammation and hippocampal dysfunction.
Collapse
Affiliation(s)
- Christine A. Olson
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA,Correspondence to: ,
| | - Alonso J. Iñiguez
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Grace E. Yang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ping Fang
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Geoffrey N. Pronovost
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kelly G. Jameson
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tomiko K. Rendon
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jorge Paramo
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jacob T. Barlow
- Division of Chemistry & Chemical Engineering, California Institute of Technology, Pasadena, CA 91108, USA
| | - Rustem F. Ismagilov
- Division of Chemistry & Chemical Engineering, California Institute of Technology, Pasadena, CA 91108, USA
| | - Elaine Y. Hsiao
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA,Correspondence to: ,
| |
Collapse
|
24
|
Jakubauskienė E, Kanopka A. Alternative Splicing and Hypoxia Puzzle in Alzheimer's and Parkinson's Diseases. Genes (Basel) 2021; 12:genes12081272. [PMID: 34440445 PMCID: PMC8394294 DOI: 10.3390/genes12081272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 01/08/2023] Open
Abstract
Alternative pre-mRNA splicing plays a very important role in expanding protein diversity as it generates numerous transcripts from a single protein-coding gene. Therefore, alterations lead this process to neurological human disorders, including Alzheimer’s and Parkinson’s diseases. Moreover, accumulating evidence indicates that the splicing machinery highly contributes to the cells’ ability to adapt to different altered cellular microenvironments, such as hypoxia. Hypoxia is known to have an effect on the expression of proteins involved in a multiple of biological processes, such as erythropoiesis, angiogenesis, and neurogenesis, and is one of the important risk factors in neuropathogenesis. In this review, we discuss the current knowledge of alternatively spliced genes, which, as it is reported, are associated with Alzheimer’s and Parkinson’s diseases. Additionally, we highlight the possible influence of cellular hypoxic microenvironment for the formation of mRNA isoforms contributing to the development of these neurodegenerative diseases.
Collapse
|
25
|
Bhardwaj S, Kesari KK, Rachamalla M, Mani S, Ashraf GM, Jha SK, Kumar P, Ambasta RK, Dureja H, Devkota HP, Gupta G, Chellappan DK, Singh SK, Dua K, Ruokolainen J, Kamal MA, Ojha S, Jha NK. CRISPR/Cas9 gene editing: New hope for Alzheimer's disease therapeutics. J Adv Res 2021; 40:207-221. [PMID: 36100328 PMCID: PMC9481950 DOI: 10.1016/j.jare.2021.07.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/27/2021] [Accepted: 07/02/2021] [Indexed: 12/16/2022] Open
Abstract
Mutations in APP, PSEN1 and PSEN2 are known factors for AD pathobiology. CRISPR/Cas9 genome editing approach hold promises in AD management. CRISPR/Cas9 is utilized to help correct anomalous genetic functions. Off-target mutations may impair the functionality of edited cells. Non-viral vectors show better efficacy and safety than viral vectors.
Background Alzheimer's disease (AD) is an insidious, irreversible, and progressive neurodegenerative health condition manifesting as cognitive deficits and amyloid beta (Aβ) plaques and neurofibrillary tangles. Approximately 50 million individuals are affected by AD, and the number is rapidly increasing globally. This review explores the role of CRISPR/Cas9 gene editing in the management of AD and its clinical manifestations. Aim of Review This review aims to provide a deep insight into the recent progress in CRISPR/Cas9-mediated genome editing and its use against neurodegenerative disorders, specifically AD. However, we have referred to its use against parkinsons’s disease (PD), Huntington’s disease (HD), and other human diseases, as is one of the most promising and emerging technologies for disease treatment. Key Scientific Concepts of Review The pathophysiology of AD is known to be linked with gene mutations, that is, presenilin (PSEN) and amyloid beta precursor protein (APP). However, clinical trials focused at the genetic level could not meet the desired efficiency. The CRISPR/Cas9 genome editing tool is one of the most powerful technologies for correcting inconsistent genetic signatures and now extensively used for AD management. It has significant potential for the correction of undesired gene mutations associated with AD. This technology has allowed the development of empirical AD models, therapeutic lines, and diagnostic approaches for better understanding the nervous system, from in vitro to in vivo models.
Collapse
Affiliation(s)
- Shanu Bhardwaj
- Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, 00076 Espoo, Finland
| | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada
| | - Shalini Mani
- Department of Biotechnology, Centre for Emerging Disease, Jaypee Institute of Information Technology, Noida, India
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida 201310, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Harish Dureja
- Faculty of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal road, Jagatpura, Jaipur, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India
| | - Janne Ruokolainen
- Department of Applied Physics, School of Science, Aalto University, 00076 Espoo, Finland
| | - Mohammad Amjad Kamal
- West China School of Nursing / Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, NSW 2770; Novel Global Community Educational Foundation, Australia
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, 17666, United Arab Emirates.
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida 201310, India.
| |
Collapse
|
26
|
Roy SS, Taguchi YH. Identification of genes associated with altered gene expression and m6A profiles during hypoxia using tensor decomposition based unsupervised feature extraction. Sci Rep 2021; 11:8909. [PMID: 33903618 PMCID: PMC8076323 DOI: 10.1038/s41598-021-87779-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/05/2021] [Indexed: 11/09/2022] Open
Abstract
Although hypoxia is a critical factor that can drive the progression of various diseases, the mechanism underlying hypoxia itself remains unclear. Recently, m6A has been proposed as an important factor driving hypoxia. Despite successful analyses, potential genes were not selected with statistical significance but were selected based solely on fold changes. Because the number of genes is large while the number of samples is small, it was impossible to select genes using conventional feature selection methods with statistical significance. In this study, we applied the recently proposed principal component analysis (PCA), tensor decomposition (TD), and kernel tensor decomposition (KTD)-based unsupervised feature extraction (FE) to a hypoxia data set. We found that PCA, TD, and KTD-based unsupervised FE could successfully identify a limited number of genes associated with altered gene expression and m6A profiles, as well as the enrichment of hypoxia-related biological terms, with improved statistical significance.
Collapse
Affiliation(s)
- Sanjiban Sekhar Roy
- School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, India
| | - Y-H Taguchi
- Department of Physics, Chuo University, Tokyo, 112-8551, Japan.
| |
Collapse
|
27
|
Ming S, Xie Y, Du X, Huang H, Fan Y, Liang Q, Xie Y. Effect of dexmedetomidine on perioperative hemodynamics and organ protection in children with congenital heart disease: A randomized controlled trial. Medicine (Baltimore) 2021; 100:e23998. [PMID: 33429762 PMCID: PMC7793469 DOI: 10.1097/md.0000000000023998] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/25/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND This study aimed to investigate the effects of dexmedetomidine (Dex) on hemodynamics and organ protection in congenital heart disease (CHD) children who underwent open-heart surgery under cryogenic cardiopulmonary bypass. METHODS Ninety children were randomly allocated to group C (0.9% saline 0.2 μg/kg/hour), group D1 (Dex 0.2 μg/kg/hour), and group D2 (Dex 0.4 μg/kg/hour) (n = 30 per group). All participants received fentanyl, propofol and 1% sevoflurane for anesthesia induction. Hemodynamic data were measured from T0 (before the induction) to T7 (30 minutes after extubation). The difference of arterial internal jugular vein bulbar oxygen difference and cerebral oxygen extraction ratio were calculated according to Fick formula. Enzyme-linked immunosorbent assay was performed to detect the serum myocardial, brain and kidney injury markers. The incidence of acute kidney injury (AKI) was calculated by serum creatinine level. Tracheal extubation time, postoperative pain score and emergence agitation score were also recorded. RESULTS Compared with group C, group D1, and D2 exhibited reduction in hemodynamic parameters, myocardial and brain injury indicators, and tracheal extubation time. There were no significant differences in blood urea nitrogen and neutrophil gelatinase-associated lipocalin or incidence of AKI among the 3 groups. Besides, the incidence of tachycardia, nausea, vomiting and moderate agitation, and the FLACC scale in group D1 and D2 were lower than those in group C. Moreover, Dex 0.4 g/kg/hour could further reduce the dosage of fentanyl and dopamine compared with Dex 0.2 g/kg/hour. CONCLUSIONS Dex anesthesia can effectively maintain hemodynamic stability and diminish organ injuries in CHD children.
Collapse
Affiliation(s)
- Shaopeng Ming
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University
| | - Yongguo Xie
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University
| | - Xueke Du
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University
| | - Haiqing Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Guangxi Medical University
| | - Yue Fan
- Department of Cardiothoracic surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning
| | - Qingxuan Liang
- Department of Anesthesiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine
| | - Yubo Xie
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
28
|
Abstract
Over the last few years, cancer immunotherapy experienced tremendous developments and it is nowadays considered a promising strategy against many types of cancer. However, the exclusion of lymphocytes from the tumor nest is a common phenomenon that limits the efficiency of immunotherapy in solid tumors. Despite several mechanisms proposed during the years to explain the immune excluded phenotype, at present, there is no integrated understanding about the role played by different models of immune exclusion in human cancers. Hypoxia is a hallmark of most solid tumors and, being a multifaceted and complex condition, shapes in a unique way the tumor microenvironment, affecting gene transcription and chromatin remodeling. In this review, we speculate about an upstream role for hypoxia as a common biological determinant of immune exclusion in solid tumors. We also discuss the current state of ex vivo and in vivo imaging of hypoxic determinants in relation to T cell distribution that could mechanisms of immune exclusion and discover functional-morphological tumor features that could support clinical monitoring.
Collapse
|
29
|
Jha NK, Sharma A, Jha SK, Ojha S, Chellappan DK, Gupta G, Kesari KK, Bhardwaj S, Shukla SD, Tambuwala MM, Ruokolainen J, Dua K, Singh SK. Alzheimer's disease-like perturbations in HIV-mediated neuronal dysfunctions: understanding mechanisms and developing therapeutic strategies. Open Biol 2020; 10:200286. [PMID: 33352062 PMCID: PMC7776571 DOI: 10.1098/rsob.200286] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/27/2020] [Indexed: 01/10/2023] Open
Abstract
Excessive exposure to toxic substances or chemicals in the environment and various pathogens, including viruses and bacteria, is associated with the onset of numerous brain abnormalities. Among them, pathogens, specifically viruses, elicit persistent inflammation that plays a major role in Alzheimer's disease (AD) as well as dementia. AD is the most common brain disorder that affects thought, speech, memory and ability to execute daily routines. It is also manifested by progressive synaptic impairment and neurodegeneration, which eventually leads to dementia following the accumulation of Aβ and hyperphosphorylated Tau. Numerous factors contribute to the pathogenesis of AD, including neuroinflammation associated with pathogens, and specifically viruses. The human immunodeficiency virus (HIV) is often linked with HIV-associated neurocognitive disorders (HAND) following permeation through the blood-brain barrier (BBB) and induction of persistent neuroinflammation. Further, HIV infections also exhibited the ability to modulate numerous AD-associated factors such as BBB regulators, members of stress-related pathways as well as the amyloid and Tau pathways that lead to the formation of amyloid plaques or neurofibrillary tangles accumulation. Studies regarding the role of HIV in HAND and AD are still in infancy, and potential link or mechanism between both is not yet established. Thus, in the present article, we attempt to discuss various molecular mechanisms that contribute to the basic understanding of the role of HIV-associated neuroinflammation in AD and HAND. Further, using numerous growth factors and drugs, we also present possible therapeutic strategies to curb the neuroinflammatory changes and its associated sequels.
Collapse
Affiliation(s)
- Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, UP 201310, India
| | - Ankur Sharma
- Department of Life Science, School of Basic Science and Research (SBSR), Sharda University, Greater Noida, UP 201310, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, UP 201310, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, PO Box 17666, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Gaurav Gupta
- School of Phamacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo 00076, Finland
| | - Shanu Bhardwaj
- Department of Biotechnology, HIMT, Greater Noida, CCS University, UP, India
| | - Shakti D. Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, UK
| | - Janne Ruokolainen
- Department of Applied Physics, School of Science, Aalto University, Espoo 00076, Finland
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, New South Wales 2007, Australia
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, PO Box 9, Solan, Himachal Pradesh 173229, India
| | - Sandeep Kumar Singh
- Department of Biomedical Research, Centre of Biomedical Research, SGPGI Campus, Lucknow 226014, UP, India
- Biological Science, Indian Scientific Education and Technology Foundation, Lucknow 226002, UP, India
| |
Collapse
|
30
|
Su Y, Tao X, Xu J. Protective effect of Alpinetin on rats with chronic obstructive pulmonary disease. Food Sci Nutr 2020; 8:6603-6611. [PMID: 33312544 PMCID: PMC7723217 DOI: 10.1002/fsn3.1952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/27/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022] Open
Abstract
To investigate the function and mechanism of Alpinetin on chronic obstructive pulmonary disease (COPD) in rat model. The markers of lung injury (body weight/pulmonary function) were measured, and the protein levels of inflammatory-, apoptosis-, and fibrotic-related were determined by Western blot. HE/TUNEL/Masson staining was performed to investigate the mechanisms involved. The levels of inflammatory factors lung injury were detected by ELISA. The in vivo activities of all molecules were determined using a rat model. Alpinetin suppressed the injury of alveolus pulmonis cells occurred in vivo due to the decrease in inflammatory factors and biochemical markers by reduced the expression of TGF-β1, α-SMA, and TNF-α (p < .05), associated with the declined of Caspase-3 and Caspase-9 (p < .01). Additionally, protective affection of Alpinetin downregulated the IL-6 and upregulated the IL-10 (p < .01). Protective affection of Alpinetin inhibits the apoptosis, inflammation, and fibrosis of alveolus pulmonis cells in rat models.
Collapse
Affiliation(s)
- Yu Su
- Pharmaceutical Administration SectionAnqing Municipal HospitalAnqingChina
| | - Xianqi Tao
- Pharmaceutical Administration SectionAnqing Municipal HospitalAnqingChina
| | - Jianghui Xu
- Pharmaceutical Administration SectionAnqing Municipal HospitalAnqingChina
| |
Collapse
|
31
|
Abstract
Abstract
Purpose
Cu(II)-diacetyl-bis(N4-methylthiosemicarbazone) positron emission tomography (CuATSM PET) is a non-invasive imaging technique that can be used to detect hypoxia and inform prognosis in cancer. Hypoxia and oxidative stress are also hallmarks of various age-related diseases. Whether CuATSM PET has a role in the evaluation of hypoxia and oxidative stress in age-related diseases has yet to be established. The aim of this systematic review is to evaluate the utility of CuATSM PET in the diagnosis and management of age-related diseases.
Methods
EMBASE, Medline, Scopus, Web of Science and Psychinfo were systematically searched for articles published between January 1st 1997 and February 13th 2020. We included articles published in English reporting the use of CuATSM PET in the diagnosis and management of age-related diseases in humans or animals.
Results
Nine articles were included describing CuATSM PET measures in neurological and cardiovascular disease. There was higher CuATSM uptake in diseased compared to control subjects in Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), myocardial ischemia (MI), cardiac dysautonomia and atherosclerosis. Higher CuATSM uptake was seen in diseased compared to control anatomical areas in PD, cerebrovascular disease (CVD), MI and atherosclerosis. CuATSM uptake was associated with disease severity in PD, ALS, CVD and atherosclerosis. An association between CuATSM uptake and disease duration was shown in atherosclerosis.
Conclusion
CuATSM uptake is higher in neurological and cardiovascular diseases and associated with disease severity and duration. Further investigations using CuATSM PET in other age-related diseases are needed.
Collapse
|
32
|
Phenolic Acids-Rich Fractions from Agaricus bitorguis (Quél.) Sacc. Chaidam ZJU-CDMA-12 Mycelia Modulate Hypoxic Stress on Hypoxia-Damaged PC12 Cells. Molecules 2020; 25:molecules25204845. [PMID: 33096616 PMCID: PMC7587959 DOI: 10.3390/molecules25204845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/10/2020] [Accepted: 10/17/2020] [Indexed: 01/06/2023] Open
Abstract
Hypoxia is a common pathological process in various clinical diseases. However, there is still a lack of effective anti-hypoxia active substances. Agaricus bitorguis (Quél.) Sacc Chaidam (ABSC) is a rare wild edible macrofungus that grows underground at high altitudes. Herein, intracellular phenolic acids-rich fractions (IPA) were extracted from ABSC ZJU-CDMA-12, and the structural characterization and anti-hypoxia activity of IPA on PC12 cells were elucidated as well. The results of HPLC-Q-TOF-MS illustrated that five kinds of IPA were isolated from ABSC, including (−)-epicatechin gallate, arabelline, yunnaneic acid D, 2′-O-p-hydroxybenzoyl-6′-O-trans-caffeoylgardoside,4′-O-methylgallocatechin-(4->8)-4′-O-methylepigallocatechin. IPA extracted from ABSC proved to show anti-hypoxia activity on hypoxia-damaged PC12 cells. Hypoxia enhanced reactive oxygen species (ROS) generation and reduced the mitochondrial membrane potential (ΔΨm) in PC12 cells, resulting in the inhibition of survival and induction of apoptosis in PC12 cells. Measurements of 100 μg/mL and 250 μg/mL IPA could significantly reduce hypoxia-induced damage in PC12 cells by decreasing overproduced intracellular ROS, improving ΔΨm, and reducing cell apoptosis rate. Our findings indicated that the IPA from ABSC potentially could be used as novel bioactive components applied to anti-hypoxia functional foods or medicines.
Collapse
|
33
|
Jakubauskienė E, Vilys L, Pečiulienė I, Kanopka A. The role of hypoxia on Alzheimer's disease-related APP and Tau mRNA formation. Gene 2020; 766:145146. [PMID: 32941952 DOI: 10.1016/j.gene.2020.145146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 12/26/2022]
Abstract
The removal of introns from mRNA precursors (pre-mRNAs) is an essential step in eukaryotic gene expression. The splicing machinery heavily contributes to biological complexity and especially to the ability of cells to adapt to altered cellular conditions. Hypoxia also plays a key role in the pathophysiology of many diseases, including Alzheimer's disease (AD). In the presented study, we have examined the influence of cellular hypoxia on mRNA splice variant formation from Alzheimer's disease-related Tau and APP genes in brain cells. We have shown that the hypoxic microenvironment influenced the formation of Tau mRNA splice variants, but had no effect on APP mRNA splice variant formation. Additionally, our presented results indicate that splicing factor SRSF1 but not SRSF5 alters the formation of Tau cellular mRNA splice variants in hypoxic cells. Obtained results have also shown that hypoxic brain cells possess enhanced CLK1-4 kinase mRNA levels. This study underlines that cellular hypoxia can influence disease development through changing pre-mRNA splicing.
Collapse
Affiliation(s)
| | - Laurynas Vilys
- Institute of Biotechnology, Vilnius University, Vilnius, Lithuania
| | - Inga Pečiulienė
- Institute of Biotechnology, Vilnius University, Vilnius, Lithuania
| | - Arvydas Kanopka
- Institute of Biotechnology, Vilnius University, Vilnius, Lithuania.
| |
Collapse
|
34
|
Nicaise AM, Willis CM, Crocker SJ, Pluchino S. Stem Cells of the Aging Brain. Front Aging Neurosci 2020; 12:247. [PMID: 32848716 PMCID: PMC7426063 DOI: 10.3389/fnagi.2020.00247] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
The adult central nervous system (CNS) contains resident stem cells within specific niches that maintain a self-renewal and proliferative capacity to generate new neurons, astrocytes, and oligodendrocytes throughout adulthood. Physiological aging is associated with a progressive loss of function and a decline in the self-renewal and regenerative capacities of CNS stem cells. Also, the biggest risk factor for neurodegenerative diseases is age, and current in vivo and in vitro models of neurodegenerative diseases rarely consider this. Therefore, combining both aging research and appropriate interrogation of animal disease models towards the understanding of the disease and age-related stem cell failure is imperative to the discovery of new therapies. This review article will highlight the main intrinsic and extrinsic regulators of neural stem cell (NSC) aging and discuss how these factors impact normal homeostatic functions within the adult brain. We will consider established in vivo animal and in vitro human disease model systems, and then discuss the current and future trajectories of novel senotherapeutics that target aging NSCs to ameliorate brain disease.
Collapse
Affiliation(s)
- Alexandra M Nicaise
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Cory M Willis
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
35
|
Savyuk M, Krivonosov M, Mishchenko T, Gazaryan I, Ivanchenko M, Khristichenko A, Poloznikov A, Hushpulian D, Nikulin S, Tonevitsky E, Abuzarova G, Mitroshina E, Vedunova M. Neuroprotective Effect of HIF Prolyl Hydroxylase Inhibition in an In Vitro Hypoxia Model. Antioxidants (Basel) 2020; 9:E662. [PMID: 32722310 PMCID: PMC7463909 DOI: 10.3390/antiox9080662] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 01/19/2023] Open
Abstract
A novel potent analog of the branched tail oxyquinoline group of hypoxia-inducible factor (HIF) prolyl hydroxylase inhibitors, neuradapt, has been studied in two treatment regimes in an in vitro hypoxia model on murine primary hippocampal cultures. Neuradapt activates the expression of HIF1 and HIF2 target genes and shows no toxicity up to 20 μM, which is more than an order of magnitude higher than its biologically active concentration. Cell viability, functional activity, and network connectivity between the elements of neuronal networks have been studied using a pairwise correlation analysis of the intracellular calcium fluctuations in the individual cells. An immediate treatment with 1 μМ and 15 μМ neuradapt right at the onset of hypoxia not only protects from the death, but also maintains the spontaneous calcium activity in nervous cells at the level of the intact cultures. A similar neuroprotective effect in the post-treatment scenario is observed for 15 μМ, but not for 1 μМ neuradapt. Network connectivity is better preserved with immediate treatment using 1 μМ neuradapt than with 15 μМ, which is still beneficial. Post-treatment with neuradapt did not restore the network connectivity despite the observation that neuradapt significantly increased cell viability at 1 μМ and functional activity at 15 μМ. The preservation of cell viability and functional activity makes neuradapt promising for further studies in a post-treatment scenario, since it can be combined with other drugs and treatments restoring the network connectivity of functionally competent cells.
Collapse
Affiliation(s)
- Maria Savyuk
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia; (M.S.); (T.M.); (E.M.)
| | - Mikhail Krivonosov
- Department of Applied Mathematics, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia; (M.K.); (M.I.)
| | - Tatiana Mishchenko
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia; (M.S.); (T.M.); (E.M.)
| | - Irina Gazaryan
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow 125284, Russia; (I.G.); (A.K.); or (A.P.); (D.H.); (G.A.)
- Chemical Enzymology Department, Chemistry Faculty, M. V. Lomonosov Moscow State University, Moscow 119992, Russia
| | - Mikhail Ivanchenko
- Department of Applied Mathematics, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia; (M.K.); (M.I.)
| | - Anna Khristichenko
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow 125284, Russia; (I.G.); (A.K.); or (A.P.); (D.H.); (G.A.)
| | - Andrey Poloznikov
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow 125284, Russia; (I.G.); (A.K.); or (A.P.); (D.H.); (G.A.)
- Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow 101000, Russia;
| | - Dmitry Hushpulian
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow 125284, Russia; (I.G.); (A.K.); or (A.P.); (D.H.); (G.A.)
- School of Biomedicine, Far Eastern Federal University, Vladivostok 690091, Russia
| | - Sergey Nikulin
- Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow 101000, Russia;
| | - Evgeny Tonevitsky
- Development Fund of the Innovation Science and Technology Center “Mendeleev Valley”, Moscow 125480, Russia;
| | - Guzal Abuzarova
- P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow 125284, Russia; (I.G.); (A.K.); or (A.P.); (D.H.); (G.A.)
| | - Elena Mitroshina
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia; (M.S.); (T.M.); (E.M.)
| | - Maria Vedunova
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia; (M.S.); (T.M.); (E.M.)
| |
Collapse
|
36
|
Intracellular Neuroprotective Mechanisms in Neuron-Glial Networks Mediated by Glial Cell Line-Derived Neurotrophic Factor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1036907. [PMID: 31827666 PMCID: PMC6885812 DOI: 10.1155/2019/1036907] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 10/19/2019] [Indexed: 12/28/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) has a pronounced neuroprotective effect in various nervous system pathologies, including ischaemic brain damage and neurodegenerative diseases. In this work, we studied the effect of GDNF on the ultrastructure and functional activity of neuron-glial networks during acute hypoxic exposure, a key damaging factor in numerous brain pathologies. We analysed the molecular mechanisms most likely involved in the positive effects of GDNF. Hypoxia modelling was performed on day 14 of culturing primary hippocampal cells obtained from mouse embryos (E18). GDNF (1 ng/ml) was added to the culture medium 20 min before oxygen deprivation. Acute hypoxia-induced irreversible changes in the ultrastructure of neurons and astrocytes led to the loss of functional Сa2+ activity and neural network disruption. Destructive changes in the mitochondrial apparatus and its functional activity characterized by an increase in the basal oxygen consumption rate and respiratory chain complex II activity during decreased stimulated respiration intensity were observed 24 hours after hypoxic injury. At a concentration of 1 ng/ml, GDNF maintained the functional metabolic network activity in primary hippocampal cultures and preserved the structure of the synaptic apparatus and number of mature chemical synapses, confirming its neuroprotective effect. GDNF maintained the normal structure of mitochondria in neuronal outgrowth but not in the soma. Analysis of the possible GDNF mechanism revealed that RET kinase, a component of the receptor complex, and the PI3K/Akt pathway are crucial for the neuroprotective effect of GDNF. The current study also revealed the role of GDNF in the regulation of HIF-1α transcription factor expression under hypoxic conditions.
Collapse
|
37
|
Borjini N, Sivilia S, Giuliani A, Fernandez M, Giardino L, Facchinetti F, Calzà L. Potential biomarkers for neuroinflammation and neurodegeneration at short and long term after neonatal hypoxic-ischemic insult in rat. J Neuroinflammation 2019; 16:194. [PMID: 31660990 PMCID: PMC6819609 DOI: 10.1186/s12974-019-1595-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Hypoxic-ischemic (HI) encephalopathy causes life-long morbidity and premature mortality in term neonates. Therapies in addition to whole-body cooling are under development to treat the neonate at risk for HI encephalopathy, but are not a quickly measured serum inflammatory or neuronal biomarkers to rapidly and accurately identify brain injury in order to follow the efficacy of therapies. METHODS In order to identify potential biomarkers for early inflammatory and neurodegenerative events after neonatal hypoxia-ischemia, both male and female Wistar rat pups at postnatal day 7 (P7) were used and had their right carotid artery permanently doubly occluded and exposed to 8% oxygen for 90 min. Sensory and cognitive parameters were assessed by open field, rotarod, CatWalk, and Morris water maze (MWM) test. Plasma and CSF biomarkers were investigated on the acute (24 h and 72 h) and chronic phase (4 weeks). Brains were assessed for gene expression analysis by quantitative RT-PCR Array. RESULTS We found a delay of neurological reflex maturation in HI rats. We observed anxiolytic-like baseline behavior in males more than females following HI injury. HI rats held on the rotarod for a shorter time comparing to sham. HI injury impaired spatial learning ability on MWM test. The CatWalk assessment demonstrated a long-term deficit in gait parameters related to the hind paw. Proinflammatory biomarkers such as IL-6 in plasma and CCL2 and TNF-α in CSF showed an upregulation at 24 h after HI while other cytokines, such as IL-17A and CCL5, were upregulated after 72 h in CSF. At 24 h post-injury, we observed an increase of Edn1, Hif1-α, and Mmp9 mRNA levels in the ipsilateral vs the contralateral hemisphere of HI rats. An upregulation of genes involved with clotting and hematopoietic processes was observed 72 h post-injury. CONCLUSIONS Our work showed that, in the immature brain, the HI injury induced an early increased production of several proinflammatory mediators detectable in plasma and CSF, followed by tissue damage in the hypoxic hemisphere and short-term as well as long-lasting neurobehavioral deficits.
Collapse
Affiliation(s)
- Nozha Borjini
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A, Largo Belloli 11/A, 43122, Parma, Italy. .,Health Science and Technologies Interdepartmental Center for Industrial Research, University of Bologna, Via Tolara di Sopra 41/E, I-40064, Ozzano Emilia, BO, Italy. .,IRET Foundation, Via Tolara di Sopra 41/E, 40064, Ozzano Emilia, BO, Italy.
| | - Sandra Sivilia
- IRET Foundation, Via Tolara di Sopra 41/E, 40064, Ozzano Emilia, BO, Italy
| | - Alessandro Giuliani
- IRET Foundation, Via Tolara di Sopra 41/E, 40064, Ozzano Emilia, BO, Italy.,Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra 50, 40064, Ozzano Emilia, BO, Italy
| | - Mercedes Fernandez
- IRET Foundation, Via Tolara di Sopra 41/E, 40064, Ozzano Emilia, BO, Italy.,Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra 50, 40064, Ozzano Emilia, BO, Italy
| | - Luciana Giardino
- Health Science and Technologies Interdepartmental Center for Industrial Research, University of Bologna, Via Tolara di Sopra 41/E, I-40064, Ozzano Emilia, BO, Italy.,IRET Foundation, Via Tolara di Sopra 41/E, 40064, Ozzano Emilia, BO, Italy.,Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra 50, 40064, Ozzano Emilia, BO, Italy
| | - Fabrizio Facchinetti
- Corporate Pre-clinical R&D, Chiesi Farmaceutici S.p.A, Largo Belloli 11/A, 43122, Parma, Italy
| | - Laura Calzà
- Health Science and Technologies Interdepartmental Center for Industrial Research, University of Bologna, Via Tolara di Sopra 41/E, I-40064, Ozzano Emilia, BO, Italy.,IRET Foundation, Via Tolara di Sopra 41/E, 40064, Ozzano Emilia, BO, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Via Tolara di Sopra 41, 40064, Ozzano Emilia, BO, Italy
| |
Collapse
|
38
|
Sun X, Yin Y, Kong L, Chen W, Miao C, Chen J. The effect of propofol on hypoxia-modulated expression of heat shock proteins: potential mechanism in modulating blood–brain barrier permeability. Mol Cell Biochem 2019; 462:85-96. [DOI: 10.1007/s11010-019-03612-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/10/2019] [Indexed: 12/11/2022]
|
39
|
Hypoxia and Inflammation as a Consequence of β-Fibril Accumulation: A Perspective View for New Potential Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7935310. [PMID: 31346362 PMCID: PMC6618348 DOI: 10.1155/2019/7935310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/22/2019] [Indexed: 11/30/2022]
Abstract
Amyloidoses are heterogeneous diseases that result from the deposition of toxic insoluble β-sheet fibrillar protein aggregates in different tissues. The cascade of molecular events leading to amyloidoses and to the related clinical manifestations is not completely understood. Nevertheless, it is known that tissue damage associated to this disease involves alteration of tissue architecture, interaction with cell surface receptors, inflammation elicited by the amyloid protein deposition, oxidative stress, and apoptosis. However, another important aspect to consider is that systemic protein massive deposition not only subverts tissue architecture but also determines a progressive cellular hypertrophy and dilation of the extracellular space enlarging the volume of the organ. Such an alteration increases the distance between cells and vessels with a drop in pO2 that, in turn, causes both necrotic cell death and activation of the hypoxia transcription factor HIF-1α. Herewith, we propose the hypothesis that both cell death and hypoxia represent two important events for the pathogenesis of damage and progression of amyloidoses. In fact, molecules released by necrotic cells activate inflammatory cells from one side while binding to HIF-1α-dependent membrane receptors expressed on hypoxic parenchymal cells on the other side. This latter event generates a signaling cascade triggering NFκB activation and chronic inflammation. Finally, we also suggest that this scenario, once proved and detailed, might suggest important targets for new therapeutic interventions.
Collapse
|
40
|
Zimmerman MA, Biggers CD, Li PA. Rapamycin treatment increases hippocampal cell viability in an mTOR-independent manner during exposure to hypoxia mimetic, cobalt chloride. BMC Neurosci 2018; 19:82. [PMID: 30594149 PMCID: PMC6310999 DOI: 10.1186/s12868-018-0482-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cobalt chloride (CoCl2) induces chemical hypoxia through activation of hypoxia-inducible factor-1 alpha (HIF-1α). Mammalian target of rapamycin (mTOR) is a multifaceted protein capable of regulating cell growth, angiogenesis, metabolism, proliferation, and survival. In this study, we tested the efficacy of a well-known mTOR inhibitor, rapamycin, in reducing oxidative damage and increasing cell viability in the mouse hippocampal cell line, HT22, during a CoCl2-simulated hypoxic insult. RESULTS CoCl2 caused cell death in a dose-dependent manner and increased protein levels of cleaved caspase-9 and caspase-3. Rapamycin increased viability of HT22 cells exposed to CoCl2 and reduced activation of caspases-9 and -3. Cells exposed to CoCl2 displayed increased reactive oxygen species (ROS) production and hyperpolarization of the mitochondrial membrane, both of which rapamycin successfully blocked. mTOR protein itself, along with its downstream signaling target, phospho-S6 ribosomal protein (pS6), were significantly inhibited with CoCl2 and rapamycin addition did not significantly lower expression further. Rapamycin promoted protein expression of Beclin-1 and increased conversion of microtubule-associated protein light chain 3 (LC3)-I into LC3-II, suggesting an increase in autophagy. Pro-apoptotic protein, Bcl-2 associated × (Bax), exhibited a slight, but significant decrease with rapamycin treatment, while its anti-apoptotic counterpart, B cell lymphoma-2 (Bcl-2), was to a similar degree upregulated. Finally, the protein expression ratio of phosphorylated mitogen-activated protein kinase (pMAPK) to its unphosphorylated form (MAPK) was dramatically increased in rapamycin and CoCl2 co-treated cells. CONCLUSIONS Our results indicate that rapamycin confers protection against CoCl2-simulated hypoxic insults to neuronal cells. This occurs, as suggested by our results, independent of mTOR modification, and rather through stabilization of the mitochondrial membrane with concomitant decreases in ROS production. Additionally, inhibition of caspase-9 and -3 activation and stimulation of protective autophagy reduces cell death, while a decrease in the Bax/Bcl-2 ratio and an increase in pMAPK promotes cell survival during CoCl2 exposure. Together these results demonstrate the therapeutic potential of rapamycin against hypoxic injury and highlight potential pathways mediating the protective effects of rapamycin treatment.
Collapse
Affiliation(s)
- Mary A. Zimmerman
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC USA
| | - Christan D. Biggers
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC USA
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, Durham, NC USA
| |
Collapse
|
41
|
Hahr JY. Physiology of aging. Med Hypotheses 2018; 123:83-85. [PMID: 30696601 DOI: 10.1016/j.mehy.2018.12.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/05/2018] [Accepted: 12/22/2018] [Indexed: 10/27/2022]
Abstract
In a natural state, death of the neuronal cells of the central nervous system is a hallmark of the aging process and occurs very insidiously. As we age, a natural loss of the body water occurs simultaneously with a rise in plasma osmolality. When a progressive rise in plasma osmolality occurs with age, it increases the effective viscosity of the circulating blood, and the thickened blood fails to perfuse the body systems and organs. If red blood cells fail to perfuse the central nervous system, the neuronal cells die from hypoxia and a lack of essential nutrients. This process is insidious but unavoidable. Hyperosmolality occurs in all mammals and it is a natural phenomenon of mammalian aging.
Collapse
Affiliation(s)
- Jung Y Hahr
- Children's Hospital of Wisconsin, 9200 W. Wisconsin Avenue, Milwaukee, WI 53226, USA.
| |
Collapse
|
42
|
Macheda T, Roberts K, Lyons DN, Higgins E, Ritter KJ, Lin AL, Alilain WJ, Bachstetter AD. Chronic Intermittent Hypoxia Induces Robust Astrogliosis in an Alzheimer's Disease-Relevant Mouse Model. Neuroscience 2018; 398:55-63. [PMID: 30529693 DOI: 10.1016/j.neuroscience.2018.11.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/19/2018] [Accepted: 11/27/2018] [Indexed: 12/17/2022]
Abstract
Sleep disturbances are a common early symptom of neurodegenerative diseases, including Alzheimer's disease (AD) and other age-related dementias, and emerging evidence suggests that poor sleep may be an important contributor to development of amyloid pathology. Of the causes of sleep disturbances, it is estimated that 10-20% of adults in the United States have sleep-disordered breathing (SDB) disorder, with obstructive sleep apnea accounting for the majority of the SBD cases. The clinical and epidemiological data clearly support a link between sleep apnea and AD; yet, almost no experimental research is available exploring the mechanisms associated with this correlative link. Therefore, we exposed an AD-relevant mouse model (APP/PS1 KI) to chronic intermittent hypoxia (IH) (an experimental model of sleep apnea) to begin to describe one of the potential mechanisms by which SDB could increase the risk of dementia. Previous studies have found that astrogliosis is a contributor to neuropathology in models of chronic IH and AD; therefore, we hypothesized that a reactive astrocyte response might be a contributing mechanism in the neuroinflammation associated with sleep apnea. To test this hypothesis, 10-11-month-old wild-type (WT) and APP/PS1 KI mice were exposed to 10 hours of IH, daily for four weeks. At the end of four weeks brains were analyzed from amyloid burden and astrogliosis. No effect was found for chronic IH exposure on amyloid-beta levels or plaque load in the APP/PS1 KI mice. A significant increase in GFAP staining was found in the APP/PS1 KI mice following chronic IH exposure, but not in the WT mice. Profiling of genes associated with different phenotypes of astrocyte activation identified GFAP, CXCL10, and Ggta1 as significant responses activated in the APP/PS1 KI mice exposed to chronic IH.
Collapse
Affiliation(s)
- Teresa Macheda
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Kelly Roberts
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Danielle N Lyons
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Emma Higgins
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Kyle J Ritter
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Ai-Ling Lin
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, United States; Department of Nutrition and Pharmacology, University of Kentucky, Lexington, KY, United States
| | - Warren J Alilain
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Adam D Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States; Department of Neuroscience, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
43
|
Dexmedetomidine Protects Against Chemical Hypoxia-Induced Neurotoxicity in Differentiated PC12 Cells Via Inhibition of NADPH Oxidase 2-Mediated Oxidative Stress. Neurotox Res 2018; 35:139-149. [PMID: 30112693 DOI: 10.1007/s12640-018-9938-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/16/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022]
Abstract
Dexmedetomidine (Dex) is a widely used sedative in anesthesia and critical care units, and it exhibits neuroprotective activity. However, the precise mechanism of Dex-exerted neuroprotection is not clear. Increased neuronal NADPH oxidase 2 (NOX2) contributes to oxidative stress and neuronal damage in various hypoxia-related neurodegenerative disorders. The present study investigated whether Dex regulated neuronal NOX2 to exert its protective effects under hypoxic conditions. Well-differentiated PC12 cells were exposed to cobalt chloride (CoCl2) to mimic a neuronal model of chemical hypoxia-mediated neurotoxicity. The data showed that Dex pretreatment of PC12 cells significantly suppressed CoCl2-induced neurotoxicity, as evidenced by the enhanced cell viability, restoration of cellular morphology, and reduction in apoptotic cells. Dex improved mitochondrial function and inhibited CoCl2-induced mitochondrial apoptotic pathways. We further demonstrated that Dex attenuated oxidative stress, downregulated NOX2 protein expression and activity, and inhibited intracellular calcium ([Ca2+]i) overload in CoCl2-treated PC12 cells. Moreover, knockdown of the NOX2 gene markedly improved mitochondrial function and attenuated apoptosis under hypoxic conditions. These results demonstrated that the protective effects of Dex against hypoxia-induced neurotoxicity in neural cells were mediated, at least partially, via inhibition of NOX2-mediated oxidative stress.
Collapse
|