1
|
Deng Q, Parker E, Duan R, Yang L. Preconditioning and Posttreatment Strategies in Neonatal Hypoxic-Ischemic Encephalopathy: Recent Advances and Clinical Challenges. Mol Neurobiol 2025:10.1007/s12035-025-04896-4. [PMID: 40178781 DOI: 10.1007/s12035-025-04896-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a severe neurological disorder caused by impaired cerebral blood flow and brain hypoxia, resulting in high morbidity and mortality rates. While therapeutic hypothermia remains the standard treatment and has been shown to reduce mortality to some extent, its therapeutic efficacy is limited, and it applies only to a select group of neonates who meet stringent inclusion criteria. Advances in our understanding of the pathophysiology of HIE have led to the identification of several promising neuroprotective strategies designed to mitigate or prevent the neurological damage induced by hypoxia-ischemia. Among these, preconditioning has emerged as a potent neuroprotective approach, enhancing cellular resilience to subsequent injury and potentially reducing treatment complexity and healthcare costs. Preconditioning/pretreatment and posttreatment offer significant promise in attenuating the neurological damage associated with HIE. Thus, exploring early intervention strategies for neonatal HIE, focusing on the comparative mechanisms and therapeutic targets of preconditioning and postconditioning, is critical to developing more effective treatment modalities. This review summarizes the current understanding of the pathophysiological mechanisms underlying neonatal HIE and its prevention and treatment strategies, providing new perspectives and a theoretical foundation for future neuroprotective interventions.
Collapse
Affiliation(s)
- Qianting Deng
- Laboratory of Exercise and Neurobiology, College of Physical Education and Sport Science, South China Normal University, Guangzhou, 510006, GD, China
| | - Emily Parker
- Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Rui Duan
- Lab of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, College of Physical Education and Sport Science, South China Normal University, Guangzhou, 510006, GD, China.
| |
Collapse
|
2
|
Guo X, Xu J, Zhao Y, Wang J, Fu T, Richard ML, Sokol H, Wang M, Li Y, Liu Y, Wang H, Wang C, Wang X, He H, Wang Y, Ma B, Peng S. Melatonin alleviates heat stress-induced spermatogenesis dysfunction in male dairy goats by regulating arachidonic acid metabolism mediated by remodeling the gut microbiota. MICROBIOME 2024; 12:233. [PMID: 39533343 PMCID: PMC11559159 DOI: 10.1186/s40168-024-01942-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Heat stress (HS) commonly occurring in summer has gradually become a factor threatening the reproductive performance of male dairy goats by reducing their fecundity. Despite the melatonin is applied to relieve HS, it is still unclear whether melatonin protects against reproductive damage induced by HS in dairy goats and how it works. The purpose of the present study is to evaluate the role of melatonin in alleviating HS-induced spermatogenesis dysfunction in male dairy goats and further explore its mechanism. RESULTS HS impaired spermatogenesis, sperm formation in the testes, and sperm maturation in the epididymis of dairy goats, resulting in decreased sperm quality. Melatonin rescued the decrease of sperm quality induced by HS via decreasing inflammatory and oxidative stress levels in testicular tissue and enhancing intercellular barrier function within the testes. Amplicon-based microbiota analysis revealed that despite gut microbiota differences between melatonin-treated dairy goats and NC dairy goats to some extent, melatonin administration tends to return the gut microbiota of male dairy goats under HS to the levels of natural control dairy goats. To explore whether the protective role of melatonin in sperm quality is mediated by regulating gut microbiota, fecal microbiota of HS dairy goats with or without melatonin treatment were transferred to HS mice, respectively. We found HS mice that had received fecal bacteria of HS dairy goats experienced serious testicular injury and dyszoospermia, while this phenomenon was ameliorated in HS mice that had received fecal bacteria of dairy goats treated with melatonin, indicating melatonin alleviates HS-induced spermatogenic damage in a microbiota dependent manner. We further found that the testicular tissue of both HS dairy goats and mice transplanted with HS dairy goat feces produced large amounts of arachidonic acid (AA)-related metabolites, which were closely associated with semen quality. Consistently, supplementation with AA has been shown to elevate the levels of inflammation and oxidative stress in the testicular tissue of mice, disrupting intercellular connections and ultimately leading to spermatogenic disorders. CONCLUSION This study has revealed that melatonin can effectively alleviate spermatogenic disorders in dairy goats caused by HS. This beneficial effect was primarily achieved through the modulation of gut microbiota, which subsequently inhibited the excessive synthesis of AA in testicular tissue. These discoveries are of great significance for preventing or improving the decline in male livestock reproductive performance caused by HS, enhancing the reproductive efficiency of elite male breeds, and ultimately improving the production efficiency of animal husbandry. Video Abstract.
Collapse
Affiliation(s)
- Xinrui Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jing Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yongkang Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jie Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Tingshu Fu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Mathias L Richard
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, 75012, Paris, France
| | - Harry Sokol
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine, Fédération Hospitalo-Universitaire, 75012, Paris, France
- Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, 75012, Paris, France
| | - Miao Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yu Li
- Animal Engineering Department, Yangling Vocational and Technical College, Yangling, 712100, Shaanxi, China
| | - Yang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Hui Wang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Chenlei Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Xueqin Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Haiyang He
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yazhou Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| | - Baohua Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| | - Sha Peng
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.
| |
Collapse
|
3
|
Endo H, Ogasawara M, Tega Y, Kubo Y, Hosoya KI, Akanuma SI. Upregulation of P-Glycoprotein and Breast Cancer Resistance Protein Activity in Newly Developed in Vitro Rat Blood-Brain Barrier Spheroids Using Advanced Glycation End-Products. Biol Pharm Bull 2024; 47:1893-1903. [PMID: 39551527 DOI: 10.1248/bpb.b24-00481] [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] [Indexed: 11/19/2024]
Abstract
The blood-brain barrier (BBB) is a dynamic interface controlling the compound translocation between the blood and the brain, thereby maintaining neural homeostasis. There is cumulative evidence that BBB impairment during diabetes mellitus (DM) takes part in the progression of cognitive dementia. As tight junction proteins and ATP-binding cassette (ABC) transporters regulate substance exchange between the circulating blood and brain, the expression and function of these molecules under DM should be fully clarified. To understand the alteration of ABC transporter function in the BBB under DM, in vitro multicellular rat BBB spheroids consisting of conditionally immortalized rat brain capillary endothelial cells, astrocytes, and pericytes were newly developed. Immunostaining and permeability analysis of paracellular transport markers suggested the construction of tight junctions on the surface of the BBB spheroids. Transport analyses using fluorescence substrates of P-glycoprotein (P-gp), the breast cancer resistance protein (BCRP), and multidrug resistance-associated protein 4 (MRP4) indicate the functional expression of these transporters in the spheroids. After treatment with advanced glycation end-products (AGEs), involved in various signals during DM, the mRNA expression of tight junction molecules and ABC transporters in the BBB spheroids was upregulated. Furthermore, the functional changes in P-gp and BCRP in the BBB spheroids exposed to AGEs were canceled by the inhibitors of the receptor for AGEs (RAGE). These results suggest that AGE-RAGE interaction upregulates P-gp and BCRP function in the BBB.
Collapse
Affiliation(s)
- Hiroki Endo
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Miki Ogasawara
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Yuma Tega
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Yoshiyuki Kubo
- Laboratory of Drug Disposition & Pharmacokinetics, Faculty of Pharma-Sciences, Teikyo University
| | - Ken-Ichi Hosoya
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Shin-Ichi Akanuma
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| |
Collapse
|
4
|
Zhang D, Jia X, Lin D, Ma J. Melatonin and ferroptosis: Mechanisms and therapeutic implications. Biochem Pharmacol 2023; 218:115909. [PMID: 37931663 DOI: 10.1016/j.bcp.2023.115909] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Ferroptosis, a regulated form of cell death, is characterized by iron-dependent lipid peroxidation leading to oxidative damage to cell membranes. Cell sensitivity to ferroptosis is influenced by factors such as iron overload, lipid metabolism, and the regulation of the antioxidant system. Melatonin, with its demonstrated capacity to chelate iron, modulate iron metabolism proteins, regulate lipid peroxidation, and regulate antioxidant systems, has promise as a potential therapeutic agent in mediating ferroptosis. The availability of approved drugs targeting ferroptosis is limited; therefore, melatonin is a candidate for broad application due to its safety and efficacy in attenuating ferroptosis in noncancerous diseases. Melatonin has been demonstrated to attenuate ferroptosis in cellular and animal models of noncancerous diseases, showcasing effectiveness in organs such as the heart, brain, lung, liver, kidney, and bone. This review outlines the molecular mechanisms of ferroptosis, investigates melatonin's potential effects on ferroptosis, and discusses melatonin's therapeutic potential as a promising intervention against diseases associated with ferroptosis. Through this discourse, we aim to lay a strong foundation for developing melatonin as a therapeutic strategy to modulate ferroptosis in a variety of disease contexts.
Collapse
Affiliation(s)
- Dongni Zhang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiaotong Jia
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| | - Duomao Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
5
|
Hong QL, Ding YH, Chen JY, Shi SS, Liang RS, Tu XK. Schisandrin B Protects against Ischemic Brain Damage by Regulating PI3K/AKT Signaling in Rats. Chin J Integr Med 2023; 29:885-894. [PMID: 37357242 DOI: 10.1007/s11655-023-3596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2022] [Indexed: 06/27/2023]
Abstract
OBJECTIVE To explore the effect and mechanism of schisandrin B (Sch B) in the treatment of cerebral ischemia in rats. METHODS The cerebral ischemia models were induced by middle cerebral artery occlusion (MCAO) and reperfusion. Sprague-Dawley rats were divided into 6 groups using a random number table, including sham, MCAO, MCAO+Sch B (50 mg/kg), MCAO+Sch B (100 mg/kg), MCAO+Sch B (100 mg/kg)+LY294002, and MCAO+Sch B (100 mg/kg)+wortmannin groups. The effects of Sch B on pathological indicators, including neurological deficit scores, cerebral infarct volume, and brain edema, were subsequently studied. Tissue apoptosis was identified by terminal transferase-mediated dUTP nick end-labeling (TUNEL) staining. The protein expressions involved in apoptosis, inflammation response and oxidative stress were examined by immunofluorescent staining, biochemical analysis and Western blot analysis, respectively. The effect of Sch B on phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling was also explored. RESULTS Sch B treatment decreased neurological deficit scores, cerebral water content, and infarct volume in MCAO rats (P<0.05 or P<0.01). Neuronal nuclei and TUNEL staining indicated that Sch B also reduced apoptosis in brain tissues, as well as the Bax/Bcl-2 ratio and caspase-3 expression (P<0.01). Sch B regulated the production of myeloperoxidase, malondialdehyde, nitric oxide and superoxide dismutase, as well as the release of cytokine interleukin (IL)-1 β and IL-18, in MCAO rats (P<0.05 or P<0.01). Sch B promoted the phosphorylation of PI3K and AKT. Blocking the PI3K/AKT signaling pathway with LY294002 or wortmannin reduced the protective effect of Sch B against cerebral ischemia (P<0.05 or P<0.01). CONCLUSIONS Sch B reduced apoptosis, inflammatory response, and oxidative stress of MCAO rats by modulating the PI3K/AKT pathway. Sch B had a potential for treating cerebral ischemia.
Collapse
Affiliation(s)
- Quan-Long Hong
- Department of Neurology, The First Hospital of Quanzhou Affiliated to Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yi-Hang Ding
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jing-Yi Chen
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Song-Sheng Shi
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Ri-Sheng Liang
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Xian-Kun Tu
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
6
|
Zeng H, Chen YX. MiR-19b-3p Inhibits Hypoxia-Ischemia Encephalopathy by Inhibiting SOX6 Expression via Activating Wnt/β-catenin Pathway. Neurochem Res 2023; 48:874-884. [PMID: 36369428 DOI: 10.1007/s11064-022-03812-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a detrimental factor in infant death and chronic disease. The specific pathogenesis is not entirely clear. Therefore, exploring the pathogenesis of HIE is critical. The expression of miR-19b-3p and SOX6 in umbilical blood of HIE patients was detected by qRT-PCR assay. HT22 cells were triggered with oxygen-glucose deprivation/reoxygenation (OGD/R) to construct the HIE cell model. Cell Counting Kit-8 (CCK-8) assay was used to estimate viability. SOD and MDA levels were detected by enzyme linked immunosorbent assay. Flow cytometry was implemented to ascertain neurocyte apoptosis. Cellular β-catenin immunofluorescence staining was used to detect the expression and distribution of β-catenin protein. Wnt signaling pathway activation was detected by TOPFlash/FOPFlash luciferase reporter assay. The targeting correlation of SOX6 and miR-19b-3p was corroborated by dual-luciferase reporter gene assay and RNA pull-down assay. MiR-19b-3p expression was once down-regulated, whilst SOX6 expression was up-regulated in HIE patients. MiR-19b-3p overexpression promoted cell proliferation, repressed cell apoptosis, oxidative stress response, and Wnt/β-catenin pathway activation in OGD/R-triggered HT22 cells. MiR-19b-3p negatively regulated SOX6 expression. SOX6 knockdown improved OGD/R-triggered HT22 cells injury via Wnt/β-catenin pathway activation. MiR-19b-3p overexpression suppressed OGD/R-triggered HT22 cell injury via inhibiting SOX6 expression via activating Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Hao Zeng
- Department of Neurosurgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, People's Republic of China
| | - Yu-Xia Chen
- Department of Neontal Development, Shenzhen Longhua District Central Hospital, No. 187, Guanlan Avenue, Longhua District, Shenzhen, 518110, Guangdong, People's Republic of China.
| |
Collapse
|
7
|
Niu Y, Du SZ, He R. TNF-α interference ameliorates brain damage in neonatal hypoxic-ischemic encephalopathy rats by regulating the expression of NT-3 and TRKC. IBRAIN 2023; 9:381-389. [PMID: 38680513 PMCID: PMC11045181 DOI: 10.1002/ibra.12089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 01/06/2023] [Accepted: 01/13/2023] [Indexed: 05/01/2024]
Abstract
The aim of this study is to explore the effect of tumor necrosis factor-α (TNF-α) inhibition in rats with neonatal hypoxic-ischemic encephalopathy (HIE) and ascertain the relevant signaling pathways. The Zea-Longa score was used to evaluate the neurological function of the rats. ImageJ was used for quantification of the brain edema volume. Triphenyl tetrazolium chloride (TTC) staining of brain tissue was performed 24 h after hypoxic-ischemic (HI) to detect right brain infarction. The expression of TNF-α was detected by real-time quantitative polymerase chain reaction (RT-qPCR). Immunofluorescence staining was used to identify the localization of TNF-α; Then, the effective shRNA fragment of TNF-α was used to validate the role of TNF-α in HIE rats, and the change of neurotrofin-3 (NT-3) and tyrosine kinase receptor-C (TRKC) was examined after TNF-α-shRNA lentivirus transfection to determine downstream signaling associated with TNF-α. Protein interaction analysis was carried out to predict the links among TNF-α, NT-3, and TRKC. Cerebral edema volume and infarction increased in the right brain after the HI operation. The Zea-Longa score significantly increased within 24 h after the HI operation. The relative expression of TNF-α was upregulated after the HI operation. TNF-α was highly expressed in the right hippocampus post HI through immunofluorescence staining. Bioinformatics analysis found a direct or an indirect link among TNF-α, NT-3, and TRKC. Moreover, the interference of TNF-α increased the expression of NT-3 and TRKC. TNF-α interference might alleviate brain injury in HIE by upregulating NT-3 and TRKC.
Collapse
Affiliation(s)
- Yong‐Min Niu
- Institute of NeuroscienceKunming Medical UniversityKunmingChina
| | - Steven Z. Du
- Department of Integrative BiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Rong He
- Animal Zoology DepartmentKunming Medical UniversityKunmingChina
| |
Collapse
|
8
|
Pluta R, Furmaga-Jabłońska W, Januszewski S, Tarkowska A. Melatonin: A Potential Candidate for the Treatment of Experimental and Clinical Perinatal Asphyxia. Molecules 2023; 28:1105. [PMID: 36770769 PMCID: PMC9919754 DOI: 10.3390/molecules28031105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Perinatal asphyxia is considered to be one of the major causes of brain neurodegeneration in full-term newborns. The worst consequence of perinatal asphyxia is neurodegenerative brain damage, also known as hypoxic-ischemic encephalopathy. Hypoxic-ischemic encephalopathy is the leading cause of mortality in term newborns. To date, due to the complex mechanisms of brain damage, no effective or causal treatment has been developed that would ensure complete neuroprotection. Although hypothermia is the standard of care for hypoxic-ischemic encephalopathy, it does not affect all changes associated with encephalopathy. Therefore, there is a need to develop effective treatment strategies, namely research into new agents and therapies. In recent years, it has been pointed out that natural compounds with neuroprotective properties, such as melatonin, can be used in the treatment of hypoxic-ischemic encephalopathy. This natural substance with anti-inflammatory, antioxidant, anti-apoptotic and neurofunctional properties has been shown to have pleiotropic prophylactic or therapeutic effects, mainly against experimental brain neurodegeneration in hypoxic-ischemic neonates. Melatonin is a natural neuroprotective hormone, which makes it promising for the treatment of neurodegeneration after asphyxia. It is supposed that melatonin alone or in combination with hypothermia may improve neurological outcomes in infants with hypoxic-ischemic encephalopathy. Melatonin has been shown to be effective in the last 20 years of research, mainly in animals with perinatal asphyxia but, so far, no clinical trials have been performed on a sufficient number of newborns. In this review, we summarize the advantages and limitations of melatonin research in the treatment of experimental and clinical perinatal asphyxia.
Collapse
Affiliation(s)
- Ryszard Pluta
- Ecotech-Complex Analytical and Programme Centre for Advanced Environmentally-Friendly Technologies, Marie Curie-Skłodowska University in Lublin, 20-612 Lublin, Poland
| | - Wanda Furmaga-Jabłońska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Agata Tarkowska
- Department of Neonate and Infant Pathology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
9
|
Zhang D, Xu S, Wu H, Liu J, Wang Y, Zhu G. Melatonin Is Neuroprotective in Escherichia coli Meningitis Depending on Intestinal Microbiota. Int J Mol Sci 2022; 24:ijms24010298. [PMID: 36613745 PMCID: PMC9820133 DOI: 10.3390/ijms24010298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Avian meningitis Escherichia coli (E. coli) can cause acute bacterial meningitis which threatens poultry health, causes great economic losses in the poultry industry, and has recently been speculated as a potential zoonotic pathogen. Melatonin can counteract bacterial meningitis-induced disruption of the blood-brain barrier (BBB), neuroinflammation, and reduce mortality. There are increasing data showing that melatonin's beneficial effects on bacterial meningitis are associated with intestinal microbiota. In this study, our data showed that melatonin alleviated neurological symptoms, enhanced survival rate, protected the integrity of the BBB, reduced the bacterial load in various tissues and blood, and inhibited inflammation and neutrophil infiltration of brain tissue in an APEC TW-XM-meningitis mice model. The results of 16S rRNA showed that melatonin pretreatment significantly maintained the composition of intestinal microbiota in APEC-meningitis mice. The abundance and diversity of intestinal microbiota were disturbed in APEC TW-XM-meningitis mice, with a decreased ratio of Firmicutes to Bacteroides and an increased the abundance of Proteobacteria. Melatonin pretreatment could significantly improve the composition and abundance of harmful bacteria and alleviate the decreased abundance of beneficial bacteria. Importantly, melatonin failed to affect the meningitis neurologic symptoms caused by APEC TW-XM infection in antibiotic-pretreated mice. In conclusion, the results suggest that melatonin can effectively prevent meningitis induced by APEC TW-XM infection in mice, depending on the intestinal microbiota. This finding is helpful to further explore the specific target mechanism of melatonin-mediated intestinal microbiota in the prevention of and protection against Escherichia coli meningitis.
Collapse
Affiliation(s)
- Dong Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Shu Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Hucong Wu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jiaqi Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yiting Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence:
| |
Collapse
|
10
|
Zhang Y, Lang R, Guo S, Luo X, Li H, Liu C, Dong W, Bao C, Yu Y. Intestinal microbiota and melatonin in the treatment of secondary injury and complications after spinal cord injury. Front Neurosci 2022; 16:981772. [PMID: 36440294 PMCID: PMC9682189 DOI: 10.3389/fnins.2022.981772] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/24/2022] [Indexed: 09/12/2023] Open
Abstract
Spinal cord injury (SCI) is a central nervous system (CNS) disease that can cause sensory and motor impairment below the level of injury. Currently, the treatment scheme for SCI mainly focuses on secondary injury and complications. Recent studies have shown that SCI leads to an imbalance of intestinal microbiota and the imbalance is also associated with complications after SCI, possibly through the microbial-brain-gut axis. Melatonin is secreted in many parts of the body including pineal gland and gut, effectively protecting the spinal cord from secondary damage. The secretion of melatonin is affected by circadian rhythms, known as the dark light cycle, and SCI would also cause dysregulation of melatonin secretion. In addition, melatonin is closely related to the intestinal microbiota, which protects the barrier function of the gut through its antioxidant and anti-inflammatory effects, and increases the abundance of intestinal microbiota by influencing the metabolism of the intestinal microbiota. Furthermore, the intestinal microbiota can influence melatonin formation by regulating tryptophan and serotonin metabolism. This paper summarizes and reviews the knowledge on the relationship among intestinal microbiota, melatonin, and SCI in recent years, to provide new theories and ideas for clinical research related to SCI treatment.
Collapse
Affiliation(s)
- Yiwen Zhang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rui Lang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shunyu Guo
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoqin Luo
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Huiting Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Cencen Liu
- Department of Pathology, People’s Hospital of Zhongjiang County, Deyang, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Changshun Bao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yang Yu
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Szczygielski J, Kopańska M, Wysocka A, Oertel J. Cerebral Microcirculation, Perivascular Unit, and Glymphatic System: Role of Aquaporin-4 as the Gatekeeper for Water Homeostasis. Front Neurol 2021; 12:767470. [PMID: 34966347 PMCID: PMC8710539 DOI: 10.3389/fneur.2021.767470] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022] Open
Abstract
In the past, water homeostasis of the brain was understood as a certain quantitative equilibrium of water content between intravascular, interstitial, and intracellular spaces governed mostly by hydrostatic effects i.e., strictly by physical laws. The recent achievements in molecular bioscience have led to substantial changes in this regard. Some new concepts elaborate the idea that all compartments involved in cerebral fluid homeostasis create a functional continuum with an active and precise regulation of fluid exchange between them rather than only serving as separate fluid receptacles with mere passive diffusion mechanisms, based on hydrostatic pressure. According to these concepts, aquaporin-4 (AQP4) plays the central role in cerebral fluid homeostasis, acting as a water channel protein. The AQP4 not only enables water permeability through the blood-brain barrier but also regulates water exchange between perivascular spaces and the rest of the glymphatic system, described as pan-cerebral fluid pathway interlacing macroscopic cerebrospinal fluid (CSF) spaces with the interstitial fluid of brain tissue. With regards to this, AQP4 makes water shift strongly dependent on active processes including changes in cerebral microcirculation and autoregulation of brain vessels capacity. In this paper, the role of the AQP4 as the gatekeeper, regulating the water exchange between intracellular space, glymphatic system (including the so-called neurovascular units), and intravascular compartment is reviewed. In addition, the new concepts of brain edema as a misbalance in water homeostasis are critically appraised based on the newly described role of AQP4 for fluid permeation. Finally, the relevance of these hypotheses for clinical conditions (including brain trauma and stroke) and for both new and old therapy concepts are analyzed.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Institute of Medical Sciences, University of Rzeszów, Rzeszów, Poland.,Department of Neurosurgery, Faculty of Medicine and Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Marta Kopańska
- Department of Pathophysiology, Institute of Medical Sciences, University of Rzeszów, Rzeszów, Poland
| | - Anna Wysocka
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, Lublin, Poland
| | - Joachim Oertel
- Department of Neurosurgery, Faculty of Medicine and Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
12
|
Tu H, Zhang A, Fu X, Xu S, Bai X, Wang H, Gao J. SMPX Deficiency Causes Stereocilia Degeneration and Progressive Hearing Loss in CBA/CaJ Mice. Front Cell Dev Biol 2021; 9:750023. [PMID: 34722533 PMCID: PMC8551870 DOI: 10.3389/fcell.2021.750023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Abstract
The small muscle protein, x-linked (SMPX) encodes a small protein containing 88 amino acids. Malfunction of this protein can cause a sex-linked non-syndromic hearing loss, named X-linked deafness 4 (DFNX4). Herein, we reported a point mutation and a frameshift mutation in two Chinese families who developed gradual hearing loss with age. To explore the impaired sites in the hearing system and the mechanism of DFNX4, we established and validated an Smpx null mouse model using CRISPR-Cas9. By analyzing auditory brainstem response (ABR), male Smpx null mice showed a progressive hearing loss starting from high frequency at the 3rd month. Hearing loss in female mice was milder and occurred later compared to male mice, which was very similar to human beings. Through morphological analyses of mice cochleas, we found the hair cell bundles progressively degenerated from the shortest row. Cellular edema occurred at the end phase of stereocilia degeneration, followed by cell death. By transfecting exogenous fluorescent Smpx into living hair cells, Smpx was observed to be expressed in stereocilia. Through noise exposure, it was shown that Smpx might participate in maintaining hair cell bundles. This Smpx knock-out mouse might be used as a suitable model to explore the pathology of DFNX4.
Collapse
Affiliation(s)
- Hailong Tu
- School of Life Sciences, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Aizhen Zhang
- School of Life Sciences, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Xiaolong Fu
- School of Life Sciences, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Shiqi Xu
- University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Xiaohui Bai
- Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Jinan, China
| | - Haibo Wang
- School of Life Sciences, Shandong Provincial ENT Hospital, Shandong University, Jinan, China.,Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Jinan, China
| | - Jiangang Gao
- School of Life Sciences, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| |
Collapse
|
13
|
Otero-Losada M, Wandosell FG, Capani F. Editorial: Neuroprotection in Synaptic Signalling During Neurological Disorders. Front Synaptic Neurosci 2021; 13:746487. [PMID: 34539374 PMCID: PMC8446195 DOI: 10.3389/fnsyn.2021.746487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/05/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Matilde Otero-Losada
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS.UAI-CONICET, Buenos Aires, Argentina
| | - Francisco G Wandosell
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, CSIC-UAM y Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Capani
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS.UAI-CONICET, Buenos Aires, Argentina.,Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| |
Collapse
|
14
|
Zhang M, Yang JK, Ma J. Regulation of the long noncoding RNA XIST on the inflammatory polarization of microglia in cerebral infarction. Exp Ther Med 2021; 22:924. [PMID: 34306193 PMCID: PMC8281447 DOI: 10.3892/etm.2021.10356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 03/22/2021] [Indexed: 01/09/2023] Open
Abstract
Proinflammatory polarization of microglia aggravates brain injury in cerebral infarction. The present study focused on the role of long non-coding (lnc)RNA X-inactive specific transcript (XIST) in the phenotype modulation of microglia. It was revealed that lncRNA XIST was significantly upregulated in both a mouse cerebral infarction model induced by middle cerebral artery occlusion (MCAO) and an activated microglial model induced by oxygen/glucose deprivation (OGD). The overexpression of XIST enhanced the expression and release of pro-inflammatory mediators [such as tumor necrosis factor (TNF)-α, IL-6, and iNOS] in microglia. Culture supernatant from lncRNA XIST-overexpressed microglial cells induced the apoptosis of primary neurons, while TNF-α antibody counteracted this neurotoxic effect. LncRNA XIST served as a sponge for miR-96-5p, counteracting its inhibitory effect on IKKβ/NF-κB signaling and TNF-α production. Notably, TNF-α was positively regulated by XIST and in turn enhanced XIST expression in microglia. The lncRNA XIST-TNF-α feedback promoted the proinflammatory polarization of microglia, thereby exacerbating cerebral neuron apoptosis.
Collapse
Affiliation(s)
- Min Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Xingtai Medical College, Xingtai, Hebei 054000, P.R. China
| | - Jian-Kai Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jing Ma
- Meteorological Service and Applied Meteorology, Ren County Meteorological Bureau of Hebei Province, Xingtai, Hebei 055150, P.R. China
| |
Collapse
|
15
|
Pang R, Advic-Belltheus A, Meehan C, Fullen DJ, Golay X, Robertson NJ. Melatonin for Neonatal Encephalopathy: From Bench to Bedside. Int J Mol Sci 2021; 22:5481. [PMID: 34067448 PMCID: PMC8196955 DOI: 10.3390/ijms22115481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/21/2022] Open
Abstract
Neonatal encephalopathy is a leading cause of morbidity and mortality worldwide. Although therapeutic hypothermia (HT) is now standard practice in most neonatal intensive care units in high resource settings, some infants still develop long-term adverse neurological sequelae. In low resource settings, HT may not be safe or efficacious. Therefore, additional neuroprotective interventions are urgently needed. Melatonin's diverse neuroprotective properties include antioxidant, anti-inflammatory, and anti-apoptotic effects. Its strong safety profile and compelling preclinical data suggests that melatonin is a promising agent to improve the outcomes of infants with NE. Over the past decade, the safety and efficacy of melatonin to augment HT has been studied in the neonatal piglet model of perinatal asphyxia. From this model, we have observed that the neuroprotective effects of melatonin are time-critical and dose dependent. Therapeutic melatonin levels are likely to be 15-30 mg/L and for optimal effect, these need to be achieved within the first 2-3 h after birth. This review summarises the neuroprotective properties of melatonin, the key findings from the piglet and other animal studies to date, and the challenges we face to translate melatonin from bench to bedside.
Collapse
Affiliation(s)
- Raymand Pang
- Institute for Women’s Health, University College London, London WC1E 6HU, UK; (R.P.); (A.A.-B.); (C.M.)
| | - Adnan Advic-Belltheus
- Institute for Women’s Health, University College London, London WC1E 6HU, UK; (R.P.); (A.A.-B.); (C.M.)
| | - Christopher Meehan
- Institute for Women’s Health, University College London, London WC1E 6HU, UK; (R.P.); (A.A.-B.); (C.M.)
| | - Daniel J. Fullen
- Translational Research Office, University College London, London W1T 7NF, UK;
| | - Xavier Golay
- Department of Brain Repair and Rehabilitation, Institute of Neurology, University College London, London WC1N 3BG, UK;
| | - Nicola J. Robertson
- Institute for Women’s Health, University College London, London WC1E 6HU, UK; (R.P.); (A.A.-B.); (C.M.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| |
Collapse
|
16
|
Reyes-Corral M, Sola-Idígora N, de la Puerta R, Montaner J, Ybot-González P. Nutraceuticals in the Prevention of Neonatal Hypoxia-Ischemia: A Comprehensive Review of their Neuroprotective Properties, Mechanisms of Action and Future Directions. Int J Mol Sci 2021; 22:2524. [PMID: 33802413 PMCID: PMC7959318 DOI: 10.3390/ijms22052524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
Neonatal hypoxia-ischemia (HI) is a brain injury caused by oxygen deprivation to the brain due to birth asphyxia or reduced cerebral blood perfusion, and it often leads to lifelong limiting sequelae such as cerebral palsy, seizures, or mental retardation. HI remains one of the leading causes of neonatal mortality and morbidity worldwide, and current therapies are limited. Hypothermia has been successful in reducing mortality and some disabilities, but it is only applied to a subset of newborns that meet strict inclusion criteria. Given the unpredictable nature of the obstetric complications that contribute to neonatal HI, prophylactic treatments that prevent, rather than rescue, HI brain injury are emerging as a therapeutic alternative. Nutraceuticals are natural compounds present in the diet or used as dietary supplements that have antioxidant, anti-inflammatory, or antiapoptotic properties. This review summarizes the preclinical in vivo studies, mostly conducted on rodent models, that have investigated the neuroprotective properties of nutraceuticals in preventing and reducing HI-induced brain damage and cognitive impairments. The natural products reviewed include polyphenols, omega-3 fatty acids, vitamins, plant-derived compounds (tanshinones, sulforaphane, and capsaicin), and endogenous compounds (melatonin, carnitine, creatine, and lactate). These nutraceuticals were administered before the damage occurred, either to the mothers as a dietary supplement during pregnancy and/or lactation or to the pups prior to HI induction. To date, very few of these nutritional interventions have been investigated in humans, but we refer to those that have been successful in reducing ischemic stroke in adults. Overall, there is a robust body of preclinical evidence that supports the neuroprotective properties of nutraceuticals, and these may represent a safe and inexpensive nutritional strategy for the prevention of neonatal HI encephalopathy.
Collapse
Affiliation(s)
- Marta Reyes-Corral
- Neurodevelopment Research Group, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain; (M.R.-C.); (N.S.-I.); (P.Y.-G.)
| | - Noelia Sola-Idígora
- Neurodevelopment Research Group, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain; (M.R.-C.); (N.S.-I.); (P.Y.-G.)
| | - Rocío de la Puerta
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain;
| | - Joan Montaner
- Neurovascular Research Lab, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain
- Department of Neurology and Neurophysiology, Hospital Universitario Virgen Macarena, 41009 Seville, Spain
| | - Patricia Ybot-González
- Neurodevelopment Research Group, Institute of Biomedicine of Seville, IBIS/HUVR/CSIC/US, 41013 Seville, Spain; (M.R.-C.); (N.S.-I.); (P.Y.-G.)
- Department of Neurology and Neurophysiology, Hospital Universitario Virgen Macarena, 41009 Seville, Spain
| |
Collapse
|
17
|
Gou Z, Su X, Hu X, Zhou Y, Huang L, Fan Y, Li J, Lu L. Melatonin improves hypoxic-ischemic brain damage through the Akt/Nrf2/Gpx4 signaling pathway. Brain Res Bull 2020; 163:40-48. [DOI: 10.1016/j.brainresbull.2020.07.011] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 06/08/2020] [Accepted: 07/11/2020] [Indexed: 12/13/2022]
|
18
|
Chumboatong W, Khamchai S, Tocharus C, Govitrapong P, Tocharus J. Agomelatine protects against permanent cerebral ischaemia via the Nrf2-HO-1 pathway. Eur J Pharmacol 2020; 874:173028. [PMID: 32084418 DOI: 10.1016/j.ejphar.2020.173028] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/06/2020] [Accepted: 02/17/2020] [Indexed: 12/12/2022]
Abstract
Stroke is a major cause of death and permanent disability worldwide. It has been reported that 85% of stroke patients undergo an ischaemic stroke. The standard treatment is currently recanalization. However, only 5% of patients have access to this treatment. Therefore, new strategies for permanent ischaemic stroke treatment need to be investigated. Agomelatine is a melatonergic agonist that acts on MT1/2 receptors and is an antagonist of 5-HT2c receptors, and melatonergic has pleiotropic effects, such as antioxidation or anti-inflammation effects. In this study, we focused on the effect of agomelatine on permanent cerebral ischaemia in a rat model. Male Wistar rats were randomly divided into the following four groups (n = 6/group): sham operating group, permanent ischaemic model group, permanent ischaemic model plus agomelatine (40 mg/kg, i.p) group and permanent ischaemic model plus melatonin (10 mg/kg, i.p) group. Twenty-four h after ischaemic onset, we investigated the neurological deficits and infarct volume using neurological deficit scores, 2,3,5-triphenyltetrazolium chloride (TTC) and transmission electron microscopy (Kochanski et al.). Moreover, we analysed Nrf2-HO-1 protein expression by Western blot. The results showed that agomelatine and melatonin decreased neuronal injury and promoted the Nrf2-HO-1 signalling pathway. These findings suggest that agomelatine and melatonin exert beneficial effects on permanent cerebral ischaemia.
Collapse
Affiliation(s)
- Wijitra Chumboatong
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Graduate School, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Satchakorn Khamchai
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Kamphaeng Phet 6 Road, Lak Si, Bangkok, 10210, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
19
|
Tan HY, Ng KY, Koh RY, Chye SM. Pharmacological Effects of Melatonin as Neuroprotectant in Rodent Model: A Review on the Current Biological Evidence. Cell Mol Neurobiol 2020; 40:25-51. [PMID: 31435851 PMCID: PMC11448813 DOI: 10.1007/s10571-019-00724-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/07/2019] [Indexed: 12/21/2022]
Abstract
The progressive loss of structure and functions of neurons, including neuronal death, is one of the main factors leading to poor quality of life. Promotion of functional recovery of neuron after injury is a great challenge in neuroregenerative studies. Melatonin, a hormone is secreted by pineal gland and has antioxidative, anti-inflammatory, and anti-apoptotic properties. Besides that, melatonin has high cell permeability and is able to cross the blood-brain barrier. Apart from that, there are no reported side effects associated with long-term usage of melatonin at both physiological and pharmacological doses. Thus, in this review article, we summarize the pharmacological effects of melatonin as neuroprotectant in central nervous system injury, ischemic-reperfusion injury, optic nerve injury, peripheral nerve injury, neurotmesis, axonotmesis, scar formation, cell degeneration, and apoptosis in rodent models.
Collapse
Affiliation(s)
- Hui Ying Tan
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Rhun Yian Koh
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia.
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
20
|
Berger HR, Nyman AKG, Morken TS, Widerøe M. Transient effect of melatonin treatment after neonatal hypoxic-ischemic brain injury in rats. PLoS One 2019; 14:e0225788. [PMID: 31860692 PMCID: PMC6924669 DOI: 10.1371/journal.pone.0225788] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Melatonin has potential neuroprotective capabilities after neonatal hypoxia-ischemia (HI), but long-term effects have not been investigated. We hypothesized that melatonin treatment directly after HI could protect against early and delayed brain injury. Unilateral HI brain injury was induced in postnatal day 7 rats. An intraperitoneal injection of either melatonin or vehicle was given at 0, 6 and 25 hours after hypoxia. In-vivo MRI was performed 1, 7, 20 and 43 days after HI, followed by histological analysis. Forelimb asymmetry and memory were assessed at 12–15 and at 36–43 days after HI. More melatonin treated than vehicle treated animals (54.5% vs 15.8%) developed a mild injury characterized by diffusion tensor values, brain volumes, histological scores and behavioral parameters closer to sham. However, on average, melatonin treatment resulted only in a tendency towards milder injury on T2-weighted MRI and apparent diffusion coefficient maps day 1 after HI, and not improved long-term outcome. These results indicate that the melatonin treatment regimen of 3 injections of 10 mg/kg within the first 25 hours only gave a transient and subtle neuroprotective effect, and may not have been sufficient to mitigate long-term brain injury development following HI.
Collapse
Affiliation(s)
- Hester Rijkje Berger
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Pediatrics, St. Olav University Hospital, Trondheim, Norway
| | - Axel K. G. Nyman
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neurology, St. Olav University Hospital, Trondheim, Norway
| | - Tora Sund Morken
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Ophthalmology, St. Olav University Hospital, Trondheim, Norway
| | - Marius Widerøe
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- * E-mail:
| |
Collapse
|
21
|
Cardinali DP. An Assessment of Melatonin's Therapeutic Value in the Hypoxic-Ischemic Encephalopathy of the Newborn. Front Synaptic Neurosci 2019; 11:34. [PMID: 31920617 PMCID: PMC6914689 DOI: 10.3389/fnsyn.2019.00034] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/26/2019] [Indexed: 12/17/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is one of the most frequent causes of brain injury in the newborn. From a pathophysiological standpoint, a complex process takes place at the cellular and tissue level during the development of newborn brain damage in the absence of oxygen. Initially, the lesion is triggered by a deficit in the supply of oxygen to cells and tissues, causing a primary energy insufficiency. Subsequently, high energy phosphate levels recover transiently (the latent phase) that is followed by a secondary phase, in which many of the pathophysiological mechanisms involved in the development of neonatal brain damage ensue (i.e., excitotoxicity, massive influx of Ca2+, oxidative and nitrosative stress, inflammation). This leads to cell death by necrosis or apoptosis. Eventually, a tertiary phase occurs, characterized by the persistence of brain damage for months and even years after the HI insult. Hypothermia is the only therapeutic strategy against HIE that has been incorporated into neonatal intensive care units with limited success. Thus, there is an urgent need for agents with the capacity to curtail acute and chronic damage in HIE. Melatonin, a molecule of unusual phylogenetic conservation present in all known aerobic organisms, has a potential role as a neuroprotective agent both acutely and chronically in HIE. Melatonin displays a remarkable antioxidant and anti-inflammatory activity and is capable to cross the blood-brain barrier readily. Moreover, in many animal models of brain degeneration, melatonin was effective to impair chronic mechanisms of neuronal death. In animal models, and in a limited number of clinical studies, melatonin increased the level of protection developed by hypothermia in newborn asphyxia. This review article summarizes briefly the available therapeutic strategies in HIE and assesses the role of melatonin as a potentially relevant therapeutic tool to cover the hypoxia-ischemia phase and the secondary and tertiary phases following a hypoxic-ischemic insult.
Collapse
Affiliation(s)
- Daniel P. Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| |
Collapse
|
22
|
Zhou R, Sun X, Li Y, Huang Q, Qu Y, Mu D, Li X. Low-dose Dexamethasone Increases Autophagy in Cerebral Cortical Neurons of Juvenile Rats with Sepsis Associated Encephalopathy. Neuroscience 2019; 419:83-99. [PMID: 31682824 DOI: 10.1016/j.neuroscience.2019.09.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 12/21/2022]
Abstract
Studies have shown that a certain dose of dexamethasone can improve the survival rate of patients with sepsis, and in sepsis associated encephalopathy (SAE), autophagy plays a regulatory role in brain function. Here, we proved for the first time that small-dose dexamethasone (SdDex) can regulate the autophagy of cerebral cortex neurons in SAE rats and plays a protective role. Cortical neurons were cultured in vitro in a septic microenvironment and a sepsis rat model was established. The small-dose dexamethasone (SdDex) or high-dose dexamethasone (HdDex) was used to intervene in neurons or SAE rats. Through fluorescence microscopy and western blot analysis, the expressions of microtubule-associated protein 1 light chain 3 (LC3), p62/sequestosome1 (p62/SQSTM1), mammalian target of rapamycin (mTOR) signaling pathway related proteins, and apoptosis-related proteins were detected. Theresultsshowthat compared with those in SAE rats, the cortical pathological changes in SAE rats treated with SdDex were improved, and damaged substances were encapsulated and degraded by autophagosomes in neurons. Additionally, similar to neurons in vitro, cortical autophagy was further activated and the mTOR signaling pathway was inhibited. After HdDex treatment, the mTOR signaling pathway in cortex is inhibited, but further activation of autophagy is not obvious, the cortical pathological changes were further worsened and the ultrastructure of neurons was disturbed. Furthermore, the HdDex group exhibited the most obvious apoptosis. SdDex can regulate autophagy of cortical neurons by inhibiting the mTOR signaling pathway and plays a protective role. Brain damage induced by HdDex may be related to the activation of apoptosis.
Collapse
Affiliation(s)
- Ruixi Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Xuemei Sun
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Yuyao Li
- Medical College, Xiamen University, Xiamen 361102, China
| | - Qun Huang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Xihong Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China; Department of Emergency Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
23
|
Chen W, Chen LF, Zhang MB, Xia YP, Zhao YH, Li GZ, Wang XL. [Effects of different melatonin treatment regimens on the proliferation of endogenous neural stem cells in neonatal rats with hypoxic-ischemic brain damage]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:830-835. [PMID: 31416511 PMCID: PMC7389905 DOI: 10.7499/j.issn.1008-8830.2019.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/05/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To study the effects of different melatonin treatment regimens on the proliferation of neural stem cells (NSCs) and long-term histopathology in neonatal rats with hypoxic-ischemic brain damage (HIBD), and to identify better melatonin treatment regimens. METHODS A total of 96 Sprague-Dawley rats aged 7 days were randomly divided into normal control, HIBD, single-dose immediate melatonin treatment (SDIT), and 7-day continuous melatonin treatment (7DCT) groups, with 24 rats in each group. The rat model of HIBD was prepared by isolation and electrocoagulation of the right common carotid artery as well as hypoxic treatment in a hypoxic chamber (oxygen concentration 8.00% ± 0.01%) for 2 hours. On day 7 after modeling, proliferating cell nuclear antigen/Nestin double-labeling immunofluorescence was used to measure the proliferation of endogenous NSCs in the subventricular zone (SVZ) and the hippocampal dentate gyrus (DG) region in 8 rats in each group, and Western blot was used to measure the protein expression of Nestin in brain. On day 28 after modeling, hematoxylin-eosin (HE) staining and Nissl staining were used to observe the changes in the histopathology and the number of pyramidal cells in the hippocampal CA1 region in 8 rats in each group. RESULTS Immunofluorescent staining showed that compared with the HIBD group, the SDIT and 7DCT groups had a significant increase in the number of PCNA+Nestin+DAPI+ cells, and the 7DCT group had a significantly higher number than the SDIT group (P<0.01). Western blot showed that the SDIT and 7DCT groups had significantly higher protein expression of Nestin than the HIBD group, and the 7DCT group had significantly higher expression than the SDIT group (P<0.05). HE staining showed that the SDIT and 7DCT groups had alleviated cell injury, and Nissl staining showed that compared with the HIBD group, the SDIT and 7DCT groups had a significant increase in the number of pyramidal cells, and the 7DCT group had a significantly higher number than the SDIT group (P<0.01). CONCLUSIONS Both single-dose immediate melatonin treatment and 7-day continuous melatonin treatment can promote the proliferation of endogenous NSCs and alleviate long-term histological injury in the brain of neonatal rats with HIBD. A 7-day continuous melatonin treatment has a better effect than single-dose immediate melatonin treatment.
Collapse
Affiliation(s)
- Wei Chen
- Department of Medical Imaging, Weifang Medical University, Weifang, Shandong 261053, China.
| | | | | | | | | | | | | |
Collapse
|
24
|
Effect of Acetyl-L-carnitine Used for Protection of Neonatal Hypoxic-Ischemic Brain Injury on Acute Kidney Changes in Male and Female Rats. Neurochem Res 2019; 44:2405-2412. [PMID: 31041669 DOI: 10.1007/s11064-019-02807-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 10/26/2022]
Abstract
Neonatal hypoxia-ischemia (HI) is a common cause of brain injury in infants. Acute kidney injury frequently occurs after birth asphyxia and is associated with adverse outcome. Treatment with acetyl-L-carnitine (ALCAR) after HI protects brain and improves outcome. Rat pups underwent carotid ligation and 75 min hypoxia on postnatal day 7 to determine effects of HI on kidney which is understudied in this model. HI + ALCAR pups were treated at 0, 4 and 24 h after HI. The organic cation/carnitine transporter 2 (OCTN2), transports ALCAR and functions to reabsorb carnitine and acylcarnitines from urine. At 24 h after injury OCTN2 levels were significantly decreased in kidney from HI pups, 0.80 ± 0.04 (mean ± SEM, p < 0.01), compared to sham controls 1.03 ± 0.04, and HI + ALCAR pups 1.11 ± 0.06. The effect of HI on the level of pyruvate dehydrogenase (PDH) was determined since kidney has high energy requirements. At 24 h after HI, kidney PDH/β-actin ratios were significantly lower in HI pups, 0.98 ± 0.05 (mean ± SEM, p < 0.05), compared to sham controls 1.16 ± 0.06, and HI + ALCAR pups 1.24 ± 0.03, p < 0.01. Treatment of pups with ALCAR after HI prevented the decrease in renal OCTN2 and PDH levels at 24 h after injury. Protection of PDH and OCTN2 after HI would improve energy metabolism in kidney, maintain tissue carnitine levels and overall carnitine homeostasis which is essential for neonatal health.
Collapse
|
25
|
Neuroprotective Effects of Mitochondria-Targeted Plastoquinone in a Rat Model of Neonatal Hypoxic⁻Ischemic Brain Injury. Molecules 2018; 23:molecules23081871. [PMID: 30060443 PMCID: PMC6222533 DOI: 10.3390/molecules23081871] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/13/2018] [Accepted: 07/25/2018] [Indexed: 12/29/2022] Open
Abstract
Neonatal hypoxia⁻ischemia is one of the main causes of mortality and disability of newborns. To study the mechanisms of neonatal brain cell damage, we used a model of neonatal hypoxia⁻ischemia in seven-day-old rats, by annealing of the common carotid artery with subsequent hypoxia of 8% oxygen. We demonstrate that neonatal hypoxia⁻ischemia causes mitochondrial dysfunction associated with high production of reactive oxygen species, which leads to oxidative stress. Targeted delivery of antioxidants to the mitochondria can be an effective therapeutic approach to treat the deleterious effects of brain hypoxia⁻ischemia. We explored the neuroprotective properties of the mitochondria-targeted antioxidant SkQR1, which is the conjugate of a plant plastoquinone and a penetrating cation, rhodamine 19. Being introduced before or immediately after hypoxia⁻ischemia, SkQR1 affords neuroprotection as judged by the diminished brain damage and recovery of long-term neurological functions. Using vital sections of the brain, SkQR1 has been shown to reduce the development of oxidative stress. Thus, the mitochondrial-targeted antioxidant derived from plant plastoquinone can effectively protect the brain of newborns both in pre-ischemic and post-stroke conditions, making it a promising candidate for further clinical studies.
Collapse
|
26
|
Tani N, Ikeda T, Watanabe M, Toyomura J, Ohyama A, Ishikawa T. Prolactin selectively transported to cerebrospinal fluid from blood under hypoxic/ischemic conditions. PLoS One 2018; 13:e0198673. [PMID: 29949606 PMCID: PMC6021042 DOI: 10.1371/journal.pone.0198673] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/23/2018] [Indexed: 12/04/2022] Open
Abstract
Aim The aim of this study was to determine and to verify the correlation between the amount of prolactin (PRL) levels in the blood and in the cerebrospinal fluid (CSF) by various causes of death as an indicator for acute hypoxia in autopsy cases. It is to confirm the cause of the change in prolactin level in CSF by in vitro system. Materials and methods In autopsy materials, the PRL levels in blood from the right heart ventricle and in the CSF were measured by chemiluminescent enzyme immunoassay, and changes in the percentage of PRL-positive cells in the pituitary gland were examined using an immunohistochemical method. Furthermore, an inverted culture method was used as an in vitro model of the blood-CSF barrier using epithelial cells of the human choroid plexus (HIBCPP cell line) and SDR-P-1D5 or MSH-P3 (PRL-secreting cell line derived from miniature swine hypophysis) under normoxic or hypoxic (5% oxygen) conditions, and as an index of cell activity, we used Vascular Endothelial Growth Factor (VEGF). Results and discussion Serum PRL levels were not significantly different between hypoxia/ischemia cases and other causes of death. However, PRL levels in CSF were three times higher in cases of hypoxia/ischemia than in those of the other causes of death. In the cultured cell under the hypoxia condition, PRL and VEGF showed a high concentration at 10 min. We established a brain-CSF barrier model to clarify the mechanism of PRL transport to CSF from blood, the PRL concentrations from blood to CSF increased under hypoxic conditions from 5 min. These results suggested that PRL moves in CSF through choroidal epithelium from blood within a short time. PRL is hypothesized to protect the hypoxic/ischemic brain, and this may be because of the increased transportation of the choroid plexus epithelial cells.
Collapse
Affiliation(s)
- Naoto Tani
- Department of Legal Medicine, Osaka City University Medical School, Osaka, Japan
- Forensic Autopsy Section, Medico-legal Consultation and Postmortem Investigation Support Center, Osaka, Japan
- * E-mail:
| | - Tomoya Ikeda
- Department of Legal Medicine, Osaka City University Medical School, Osaka, Japan
- Forensic Autopsy Section, Medico-legal Consultation and Postmortem Investigation Support Center, Osaka, Japan
| | - Miho Watanabe
- Department of NDU Life Sciences, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan
- Field of Oral and Maxillofacial Surgery and System Medicine, Course of Clinical Science, Nippon Dental University, Graduate School of Life Dentistry at Niigata, Niigata, Japan
| | - Junko Toyomura
- Department of NDU Life Sciences, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan
| | - Akihiro Ohyama
- Department of NDU Life Sciences, School of Life Dentistry at Tokyo, The Nippon Dental University, Tokyo, Japan
| | - Takaki Ishikawa
- Department of Legal Medicine, Osaka City University Medical School, Osaka, Japan
- Forensic Autopsy Section, Medico-legal Consultation and Postmortem Investigation Support Center, Osaka, Japan
| |
Collapse
|
27
|
Li X, Liu H, Yang Y. Magnesium sulfate attenuates brain edema by lowering AQP4 expression and inhibits glia-mediated neuroinflammation in a rodent model of eclampsia. Behav Brain Res 2017; 364:403-412. [PMID: 29288747 DOI: 10.1016/j.bbr.2017.12.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/23/2017] [Accepted: 12/26/2017] [Indexed: 12/26/2022]
Abstract
Eclampsia is characterized by high morbidity and mortality wordwide. Magnesium sulfate (MgSO4) is used frequently as a prophylaxis for eclamptic seizure in clinical settings. However, the underlying mechanism is less studied, we have previously demonstrated that MgSO4 pretreatment decreases eclampsia-like seizure threshold. Here, we further evaluated the hypothesis that MgSO4 exert neuroprotective actions in eclampsia-like rats model by ameliorating neuroinflammation and brain edema. In this study, the eclampsia-like model was established by administering lipopolysaccharide plus pentylenetetrazol in pregnant Sprague-Dawley rats. Rats were given MgSO4 from gestation day14-19. Then, Iba-1 (a marker for microglia) and S100-B (a marker for astrocytes) expression levels in the hippocampus CA3 region were detected by Enzyme-linked immunosorbent assay. Cerebrospinal fluid (CSF) levels of inflammatory cytokines were measured by Luminex assays. Aquaporin-4 (a transmembrane water channel protein) expression levels in cortex were analyzed using immunohistochemistry. Astrocyte and microglia expressions were detected by immunofluorescence, neuronal damage were evaluated by Nissl staining, and changes in neuronal number in the hippocampal CA3 region (CA3) among different groups were detected by neuronal nuclei staining. Our results demonstrated that MgSO4 effectively attenuated astrocyte and microglia activation and promoted the neuronal survival in the CA3. Additionally, MgSO4 significantly reduced inflammatory cytokines response in the CSF, and decreased the expression of AQP-4 protein in the cortex. Collectively, the findings of this study indicated that MgSO4 has a neuroprotective role in eclampsia-like seizure rats through its anti-neuroninflammatory and brain edema-attenuating properties.
Collapse
Affiliation(s)
- Xiaolan Li
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Obstetrics, Guangzhou Women and Children's Medical Center, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China
| | - Huishu Liu
- Department of Obstetrics, Guangzhou Women and Children's Medical Center, China
| | - Yuanyuan Yang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.
| |
Collapse
|
28
|
Liu X, Ma Y, Wei X, Fan T. Neuroprotective effect of licochalcone A against oxygen-glucose deprivation/reperfusion in rat primary cortical neurons by attenuating oxidative stress injury and inflammatory response via the SIRT1/Nrf2 pathway. J Cell Biochem 2017; 119:3210-3219. [PMID: 29105819 DOI: 10.1002/jcb.26477] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 10/31/2017] [Indexed: 12/31/2022]
Abstract
Perinatal hypoxic-ischemic encephalopathy (HIE) is a leading cause of neonatal death and neurological disability. Oxidative stress and neuroinflammation are typical pathogenic factors of HIE. Licochalcone A (LCA) exerts various biological properties, including anti-inflammatory and antioxidant activities. However, no data have been reported to elucidate the role of LCA in the development of HIE. In the present study, primary cultured rat cortical neurons were exposed to oxygen-glucose deprivation/reoxygenation (OGD/R) in vitro to simulate the in vivo situation of neonatal HIE. Interestingly, LCA significantly antagonized cell injury under OGD/R by increasing cell survival, inhibiting lactate dehydrogenase (LDH) release and cell apoptosis. Furthermore, treatment with LCA suppressed oxidative stress by decreasing reactive oxygen species (ROS) production and malondialdehyde (MDA) content, and increasing superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities in primary rat cortical neurons after OGD/R. LCA stimulation also restrained OGD/R-triggered increase in pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) production. Importantly, LCA treatment effectively counteracts OGD/R-mediated downregulation of silent information regulator 1 (SIRT1), nuclear factor erythroid2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1), and upregulation of nuclear factor kappa B p65 (NF-κB p65). Moreover, administration with SIRT1 inhibitor EX527 partly abolished LCA-induced neuroprotective effects on rat cortical neurons exposed to OGD/R. In conclusion, our study indicates that LCA exerts a neuroprotective effect against OGD/R-induced neuronal injury in rat primary cortical neurons, suggesting that LCA might act as a candidate therapeutic target drug used for HIE and related diseases.
Collapse
Affiliation(s)
- Xiaohong Liu
- Department of Neonatology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Ying Ma
- Department of Neonatology, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Xiaodi Wei
- Department of Neonatology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Ting Fan
- Department of Neonatology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
29
|
Sun ZZ, Lv ZY, Tian WJ, Yang Y. MicroRNA-132 protects hippocampal neurons against oxygen-glucose deprivation-induced apoptosis. Int J Immunopathol Pharmacol 2017. [PMID: 28627974 PMCID: PMC5815264 DOI: 10.1177/0394632017715837] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Hypoxic-ischemic brain injury (HIBI) results in death or long-term neurologic impairment in both adults and children. In this study, we investigated the effects of microRNA-132 (miR-132) dysregulation on oxygen-glucose deprivation (OGD)-induced apoptosis in fetal rat hippocampal neurons, in order to reveal the therapeutic potential of miR-132 on HIBI. MiR-132 dysregulation was induced prior to OGD exposure by transfection of primary fetal rat hippocampal neurons with miR-132 mimic or miR-132 inhibitor. The effects of miR-132 overexpression and suppression on OGD-stimulated hippocampal neurons were evaluated by detection of cell viability, apoptotic cells rate, and the expression of apoptosis-related proteins. Besides, TargetScan database and dual luciferase activity assay were used to seek a target gene of miR-132. As a result, miR-132 was highly expressed in hippocampal neurons following 2 h of OGD exposure. MiR-132 overexpression significantly increased OGD-diminished cell viability and reduced OGD-induced apoptosis at 12, 24, and 48 h post-OGD. MiR-132 overexpression significantly down-regulated the expressions of Bax, cytochrome c, and caspase-9, but up-regulated BCl-2. Caspase-3 activity was also significantly decreased by miR-132 overexpression. Furthermore, FOXO3 was a direct target of miR-132, and it was negatively regulated by miR-132. To conclude, our results provide evidence that miR-132 protects hippocampal neurons against OGD injury by inhibiting apoptosis.
Collapse
Affiliation(s)
- Zu-Zhen Sun
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Zhan-Yun Lv
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Wen-Jing Tian
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Yan Yang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| |
Collapse
|