1
|
Guan D, Hou Z, Fan B, Bai Y, Wu H, Yu J, Xie H, Duan Z, Wang F, Wang Q. The Extract of Piper nigrum Improves the Cognitive Impairment and Mood in Sleep-Deprived Mice Through the JAK1/STAT3 Signalling Pathway. Int J Mol Sci 2025; 26:1842. [PMID: 40076470 PMCID: PMC11899911 DOI: 10.3390/ijms26051842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Piper nigrum L. (PN), which contains various bioactive compounds, is a plant with homologous medicine and food. Sleep deprivation (SD) profoundly impacts cognitive function and emotional health. However, the mechanisms by which PN improves cognitive function and depressive mood induced by SD remain unclear. In our study, network pharmacology and molecular docking techniques were used to predict the potential mechanisms by which PN regulates SD. In this study, 220 compounds were identified in PN, and 10 core targets were screened through network pharmacology. Animal experiments showed that PN ameliorated depressive mood and cognitive deficits in sleep-deprived mice, upregulated the serum activities of superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT), and downregulated malondialdehyde (MDA) levels. The ELISA assay showed that PN significantly decreased the tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β) levels. Histopathological staining of brain tissue demonstrated that PN mitigates SD-induced hippocampal damage, enables the hippocampus to produce more neurotransmitters, including 5-hydroxytryptamine (5-HT), gamma-aminobutyric acid (GABA), and dopamine (DA), and reduces glutamate (Glu) levels. RT-qPCR and WB analyses further indicated that PN could exert anti-SD effects by inhibiting the over-activation of the JAK1/STAT3 signalling pathway. In the PC12 cell model, PN could reduce inflammation and prevent apoptosis, exerting neuroprotective effects. In summary, PN has positive effects on alleviating depressive symptoms and cognitive dysfunction induced by SD.
Collapse
Affiliation(s)
- Dongyan Guan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (D.G.); (Z.H.); (B.F.); (Y.B.); (H.W.); (J.Y.)
- National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya 572024, China
| | - Zhiying Hou
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (D.G.); (Z.H.); (B.F.); (Y.B.); (H.W.); (J.Y.)
| | - Bei Fan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (D.G.); (Z.H.); (B.F.); (Y.B.); (H.W.); (J.Y.)
| | - Yajuan Bai
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (D.G.); (Z.H.); (B.F.); (Y.B.); (H.W.); (J.Y.)
| | - Honghong Wu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (D.G.); (Z.H.); (B.F.); (Y.B.); (H.W.); (J.Y.)
| | - Jiawei Yu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (D.G.); (Z.H.); (B.F.); (Y.B.); (H.W.); (J.Y.)
| | - Hui Xie
- Institute of Processing & Design of Agroproducts, Hainan Academy of Agricultural Science, Haikou 571100, China; (H.X.); (Z.D.)
| | - Zhouwei Duan
- Institute of Processing & Design of Agroproducts, Hainan Academy of Agricultural Science, Haikou 571100, China; (H.X.); (Z.D.)
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (D.G.); (Z.H.); (B.F.); (Y.B.); (H.W.); (J.Y.)
| | - Qiong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (D.G.); (Z.H.); (B.F.); (Y.B.); (H.W.); (J.Y.)
- National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya 572024, China
| |
Collapse
|
2
|
Collins B, Lemanski EA, Wright-Jin E. The Importance of Including Maternal Immune Activation in Animal Models of Hypoxic-Ischemic Encephalopathy. Biomedicines 2024; 12:2559. [PMID: 39595123 PMCID: PMC11591850 DOI: 10.3390/biomedicines12112559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a perinatal brain injury that is the leading cause of cerebral palsy, developmental delay, and poor cognitive outcomes in children born at term, occurring in about 1.5 out of 1000 births. The only proven therapy for HIE is therapeutic hypothermia. However, despite this treatment, many children ultimately suffer disability, brain injury, and even death. Barriers to implementation including late diagnosis and lack of resources also lead to poorer outcomes. This demonstrates a critical need for additional treatments for HIE, and to facilitate this, we need translational models that accurately reflect risk factors and interactions present in HIE. Maternal or amniotic infection is a significant risk factor and possible cause of HIE in humans. Maternal immune activation (MIA) is a well-established model of maternal infection and inflammation that has significant developmental consequences largely characterized within the context of neurodevelopmental disorders such as autism spectrum disorder and schizophrenia. MIA can also lead to long-lasting changes within the neuroimmune system, which lead to compounding negative outcomes following a second insult. This supports the importance of understanding the interaction of maternal inflammation and hypoxic-ischemic outcomes. Animal models have been invaluable to understanding the pathophysiology of this injury and to the development of therapeutic hypothermia. However, each model system has its own limitations. Large animal models such as pigs may more accurately represent the brain and organ development and complexity in humans, while rodent models are more cost-effective and offer more possible molecular techniques. Recent studies have utilized MIA or direct inflammation prior to HIE insult. Investigators should thoughtfully consider the risk factors they wish to include in their HIE animal models. In the incorporation of MIA, investigators should consider the type, timing, and dose of the inflammatory stimulus, as well as the timing, severity, and type of hypoxic insult. Using a variety of animal models that incorporate the maternal-placental-fetal system of inflammation will most likely lead to a more robust understanding of the mechanisms of this injury that can guide future clinical decisions and therapies.
Collapse
Affiliation(s)
- Bailey Collins
- Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA; (B.C.); (E.A.L.)
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| | - Elise A. Lemanski
- Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA; (B.C.); (E.A.L.)
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
| | - Elizabeth Wright-Jin
- Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
- Division of Neurology, Nemours Children’s Health, Wilmington, DE 19803, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
3
|
Kilicdag H, Akillioglu K, Kilic Bagır E, Kose S, Erdogan S. Neuroserpin As an Adjuvant Therapy for Hypothermia on Brain Injury in Neonatal Hypoxic-Ischemic Rats. Am J Perinatol 2024; 41:1538-1543. [PMID: 37611639 DOI: 10.1055/a-2159-0488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
OBJECTIVE We aimed to assess the effects of neuroserpin and its combination with hypothermia on hypoxic-ischemic (HI) brain injury in neonatal rats. Neuroserpin is an axon-secreted serine protease inhibitor and is important for brain development, neuronal survival, and synaptic plasticity. STUDY DESIGN Male Wistar-Albino rats on postnatal day 7 (P7) were randomly divided into five groups: sham group (n = 10), (HI; n = 10), hypoxic-ischemic hypothermia (HIH; n = 10), hypoxic-ischemic neuroserpin (HIN; n = 10), and hypoxic-ischemic neuroserpin-hypothermia (HINH; n = 10). The P7 rat brain's maturation is similar to a late preterm human brain at 34 to 36 weeks of gestation. HI was induced in rats on P7 as previously described. A single dose of 0.2 µM neuroserpin (HINH and HIN) or an equivalent volume of phosphate-buffered saline (sham, HIH, and HI) was administered intraventricularly by a Hamilton syringe immediately after hypoxia. In the follow-up, pups were subjected to systemic hypothermia or normothermia for 2 hours. Euthanasia was performed for histopathological evaluation on P10. Apoptosis was detected by caspase-3 activity and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining and was counted in the hippocampus. RESULTS In comparison to the HI group, the TUNEL-positive and caspase-3-positive neurons in the sham, HIN, HIH, and HINH groups were considerably lower (13.4 ± 1.0 vs. 1.9 ± 0.9, 6.0 ± 0.9, 5.3 ± 1.6, and 4.0 ± 1.1; p < 0.001) and (13.5 ± 1.7 vs. 1.2 ± 0.7, 9.1 ± 2.7, 4.8 ± 1.0, and 3.9 ± 1.6; p < 0.001). HIN, HIH, and HINH, compared to the sham group, showed more TUNEL-positive and caspase-3-positive neurons (6.0 ± 0.9, 5.3 ± 1.6, 4.0 ± 1.1 vs. 1.9 ± 0.9 and 9.1 ± 2.7, 4.8 ± 1.0, 3.9 ± 1.6 vs. 1.2 ± 0.7; p < 0.001). The HINH group (synergistic effect) had significantly fewer TUNEL-positive neurons and caspase-3-positive neurons than the HIN group (4.0 ± 1.1 vs. 6.0 ± 0.9 and 3.9 ± 1.6 vs. 9.1 ± 2.7; p < 0.001). CONCLUSION Our study showed that both neuroserpin alone and as an adjuvant treatment for hypothermia may have a neuroprotective effect on brain injury. KEY POINTS · Neuroserpin decreased brain injury.. · Neuroserpin showed a synergistic effect when used as an adjuvant treatment for hypothermia.. · The neuroprotective effect of neuroserpine was related to its antiapoptotic properties..
Collapse
Affiliation(s)
- Hasan Kilicdag
- Division of Neonatology, Department of Pediatrics, Baskent University Faculty of Medicine, Ankara, Turkey
| | - Kubra Akillioglu
- Division of Neurophysiology, Department of Physiology, Medical Faculty, University of Cukurova, Turkey
| | - Emine Kilic Bagır
- Department of Pathology, Cukurova University, Medical Faculty, Adana, Turkey
| | - Seda Kose
- Division of Neurophysiology, Department of Physiology, Medical Faculty, University of Cukurova, Turkey
| | - Seyda Erdogan
- Department of Pathology, Cukurova University, Medical Faculty, Adana, Turkey
| |
Collapse
|
4
|
Guan S, Li Y, Xin Y, Wang D, Lu P, Han F, Xu H. Deciphering the dual role of N-methyl-D-Aspartate receptor in postoperative cognitive dysfunction: A comprehensive review. Eur J Pharmacol 2024; 971:176520. [PMID: 38527701 DOI: 10.1016/j.ejphar.2024.176520] [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: 12/04/2023] [Revised: 03/03/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication following surgery, adversely impacting patients' recovery, increasing the risk of negative outcomes, prolonged hospitalization, and higher mortality rates. The N-methyl-D-aspartate (NMDA) receptor, crucial for learning, memory, and synaptic plasticity, plays a significant role in the development of POCD. Various perioperative factors, including age and anesthetic use, can reduce NMDA receptor function, while surgical stress, inflammation, and pain may lead to its excessive activation. This review consolidates preclinical and clinical research to explore the intricate relationship between perioperative factors affecting NMDA receptor functionality and the onset of POCD. It discusses the influence of aging, anesthetic administration, perioperative injury, pain, and inflammation on the NMDA receptor-related pathophysiology of POCD. The comprehensive analysis presented aims to identify effective treatment targets for POCD, contributing to the improvement of patient outcomes post-surgery.
Collapse
Affiliation(s)
- Shaodi Guan
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yali Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yueyang Xin
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Danning Wang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pei Lu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fanglong Han
- Department of Anesthesiology, Xiangyang Maternal and Child Health Hospital, Xiangyang, 441003, China
| | - Hui Xu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
5
|
Chen H, Zhao S, Jian Q, Yan Y, Wang S, Zhang X, Ji Y. The role of ApoE in fatty acid transport from neurons to astrocytes under ischemia/hypoxia conditions. Mol Biol Rep 2024; 51:320. [PMID: 38393618 DOI: 10.1007/s11033-023-08921-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/31/2023] [Indexed: 02/25/2024]
Abstract
BACKGROUND The aim of this study was to investigate whether ischemia/hypoxia conditions induce fatty acid transport from neurons to astrocytes and whether this mechanism is affected by ApoE isoforms. METHODS AND RESULTS A neonatal rat model of hypoxic-ischemic brain damage was established. Excessive accumulation of lipid droplets and upregulation of ApoE expression occurred in the hippocampus and cerebral cortex after hypoxia-ischemia, which implied the occurrence of abnormal fatty acid metabolism. Lipid peroxidation was induced in an oxygen-glucose deprivation and reperfusion (OGDR) model of ApoE-/- primary neurons. The number of BODIPY 558/568 C12-positive particles (fatty acid markers) transferred from neurons to astrocytes was significantly increased with the addition of human recombinant ApoE compared with that in the OGDR group, which significantly increased the efficiency of fatty acid transport from neurons to astrocytes and neuronal viability. However, ApoE4 was found to be associated with lower efficiency in fatty acid transport and less protective effects in OGDR-induced neuronal cell death than both ApoE2 and ApoE3. COG133, an ApoE-mimetic peptide, partially compensated for the adverse effects of ApoE4. FABP5 and SOD1 gene and protein expression levels were upregulated in astrocytes treated with BODIPY 558/568 C12 particles. CONCLUSIONS In conclusion, ApoE plays an important role in mediating the transport of fatty acids from neurons to astrocytes under ischemia/hypoxia conditions, and this transport mechanism is ApoE isoform dependent. ApoE4 has a low transfer efficiency and may be a potential target for the clinical treatment of neonatal hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
- Hongyan Chen
- Department of Central Laboratory, Xi'an No. 1 Hospital, The First Affiliated Hospital of Northwest University, No. 30, South Street, Beilin District, Xi'an, 710002, Shaanxi, China
- Center of Medical Genetics, Xi'an People's Hospital (Xi'an No. 4 Hospital), No. 21, Jiefang Road, Xi'an, 710004, Shaanxi, China
| | - Shaozhi Zhao
- Center of Medical Genetics, Xi'an People's Hospital (Xi'an No. 4 Hospital), No. 21, Jiefang Road, Xi'an, 710004, Shaanxi, China
| | - Qiang Jian
- Center of Medical Genetics, Xi'an People's Hospital (Xi'an No. 4 Hospital), No. 21, Jiefang Road, Xi'an, 710004, Shaanxi, China
| | - Yinfang Yan
- Department of Central Laboratory, Xi'an No. 1 Hospital, The First Affiliated Hospital of Northwest University, No. 30, South Street, Beilin District, Xi'an, 710002, Shaanxi, China
| | - Simin Wang
- Department of Central Laboratory, Xi'an No. 1 Hospital, The First Affiliated Hospital of Northwest University, No. 30, South Street, Beilin District, Xi'an, 710002, Shaanxi, China
| | - Xinwen Zhang
- Center of Medical Genetics, Xi'an People's Hospital (Xi'an No. 4 Hospital), No. 21, Jiefang Road, Xi'an, 710004, Shaanxi, China.
| | - Yuqiang Ji
- Department of Central Laboratory, Xi'an No. 1 Hospital, The First Affiliated Hospital of Northwest University, No. 30, South Street, Beilin District, Xi'an, 710002, Shaanxi, China.
| |
Collapse
|
6
|
Guez-Barber D, Eisch AJ, Cristancho AG. Developmental Brain Injury and Social Determinants of Health: Opportunities to Combine Preclinical Models for Mechanistic Insights into Recovery. Dev Neurosci 2023; 45:255-267. [PMID: 37080174 PMCID: PMC10614252 DOI: 10.1159/000530745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/14/2023] [Indexed: 04/22/2023] Open
Abstract
Epidemiological studies show that social determinants of health are among the strongest factors associated with developmental outcomes after prenatal and perinatal brain injuries, even when controlling for the severity of the initial injury. Elevated socioeconomic status and a higher level of parental education correlate with improved neurologic function after premature birth. Conversely, children experiencing early life adversity have worse outcomes after developmental brain injuries. Animal models have provided vital insight into mechanisms perturbed by developmental brain injuries, which have indicated directions for novel therapeutics or interventions. Animal models have also been used to learn how social environments affect brain maturation through enriched environments and early adverse conditions. We recognize animal models cannot fully recapitulate human social circumstances. However, we posit that mechanistic studies combining models of developmental brain injuries and early life social environments will provide insight into pathways important for recovery. Some studies combining enriched environments with neonatal hypoxic injury models have shown improvements in developmental outcomes, but further studies are needed to understand the mechanisms underlying these improvements. By contrast, there have been more limited studies of the effects of adverse conditions on developmental brain injury extent and recovery. Uncovering the biological underpinnings for early life social experiences has translational relevance, enabling the development of novel strategies to improve outcomes through lifelong treatment. With the emergence of new technologies to analyze subtle molecular and behavioral phenotypes, here we discuss the opportunities for combining animal models of developmental brain injury with social construct models to deconvolute the complex interactions between injury, recovery, and social inequity.
Collapse
Affiliation(s)
- Danielle Guez-Barber
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amelia J. Eisch
- Department of Anesthesiology and Critical Care, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana G. Cristancho
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Baranoglu Kilinc Y, Dilek M, Kilinc E, Torun IE, Saylan A, Erdogan Duzcu S. Capsaicin attenuates excitotoxic-induced neonatal brain injury and brain mast cell-mediated neuroinflammation in newborn rats. Chem Biol Interact 2023; 376:110450. [PMID: 36925032 DOI: 10.1016/j.cbi.2023.110450] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/07/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023]
Abstract
Excitotoxicity and neuroinflammation are key contributors to perinatal brain injuries. Capsaicin, an active ingredient of chili peppers, is a potent exogenous agonist for transient receptor potential vanilloid 1 receptors. Although the neuroprotective and anti-inflammatory effects of capsaicin are well-documented, its effects on excitotoxic-induced neonatal brain injury and neuroinflammation have not previously been investigated. The aim of this study was to investigate the effects of capsaicin on brain damage, brain mast cells, and inflammatory mediators in a model of ibotenate-induced excitotoxic brain injury in neonatal rats. P5 rat-pups were intraperitoneally injected with vehicle, 0.2-, 1-, and 5-mg/kg doses of capsaicin, or the NMDA (N-methyl-d-aspartate) receptor antagonist MK-801 (dizocilpine), 30 min before intracerebral injection of 10 μg ibotenate. The naive-control group received no substance administration. The rat pups were sacrificed one or five days after ibotenate injection. Levels of activin A and interleukin (IL)-1β, IL-6, and IL-10 in brain tissue were measured using the enzyme-linked immunosorbent assay method. Cortex and white matter thicknesses, white matter lesion size, and mast cells were evaluated in brain sections stained with cresyl-violet or toluidine-blue. Capsaicin improved ibotenate-induced white matter lesions and cerebral white and gray matter thicknesses in a dose-dependent manner. In addition, it suppressed the degranulation and increased number of brain mast cells induced by ibotenate. Capsaicin also reduced the excitotoxic-induced production of neuronal survival factor activin A and of the pro-inflammatory cytokines IL-1β, and IL-6 in brain tissue. However, IL-10 levels were not altered by the treatments. MK-801, as a positive control, reversed all these ibotenate-induced changes, further confirming the success of the model. Our findings provide, for the first time, evidence for the therapeutic effects of capsaicin against excitotoxic-induced neonatal brain injury and brain mast cell-mediated neuroinflammation. Capsaicin may therefore be a promising candidate in the prevention and/or reduction of neonatal brain damage.
Collapse
Affiliation(s)
| | - Mustafa Dilek
- Department of Pediatrics, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Erkan Kilinc
- Department of Physiology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey.
| | - Ibrahim Ethem Torun
- Department of Physiology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Aslihan Saylan
- Department of Histology and Embryology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | - Selma Erdogan Duzcu
- Department of Medical Pathology, Faculty of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| |
Collapse
|
8
|
Nascimento ALCS, Fernandes RP, Carvalho ACS, Frigieri I, Alves RC, Chorilli M. Insights for Alzheimer's disease pharmacotherapy and current clinical trials. Neurochem Int 2022; 159:105401. [PMID: 35842055 DOI: 10.1016/j.neuint.2022.105401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/20/2022] [Accepted: 07/09/2022] [Indexed: 12/25/2022]
Abstract
Over the years, the scientific community has sought improvements in the life quality of patients diagnosed with Alzheimer's disease (AD). Synaptic loss and neuronal death observed in the regions responsible for cognitive functions represent an irreversible progressive disease that is clinically characterized by impaired cognitive and functional abilities, along with behavioral symptoms. Currently, image and body fluid biomarkers can provide early dementia diagnostic, being it the best way to slow the disease's progression. The first signs of AD development are still complex, the existence of individual genetic and phenotypic characteristics about the disease makes it difficult to standardize studies on the subject. The answer seems to be related between Aβ and tau proteins. Aβ deposition in the medial parietal cortex appears to be the initial stage of AD, but it does not have a strong correlation with neurodegeneration. The strongest link between symptoms occurs with tau aggregation, which antecede Aβ deposits in the medial temporal lobe, however, the protein can be found in cognitively healthy older people. The answer to the question may lie in some catalytic effect between both proteins. Amid so many doubts, Aducanumab was approved, which raised controversies and results intense debate in the scientific field. Abnormal singling of some blood biomarkers produced by adipocytes under high lipogenesis, such as TNFα, leptin, and interleukin-6, demonstrate to be linked to neuroinflammation worsens, diabetes, and also severe cases of COVID-19, howsoever, under higher lipolysis, seem to have therapeutic anti-inflammatory effects in the brain, which has increasingly contributed to the understanding of AD. In addition, the relationship of severe clinical complications caused by Sars-CoV-2 viral infection and AD, go beyond the term "risk group" and may be related to the development of dementia long-term. Thus, this review summarized the current emerging pharmacotherapies, alternative treatments, and nanotechnology applied in clinical trials, discussing relevant points that may contribute to a more accurate look.
Collapse
Affiliation(s)
- A L C S Nascimento
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, 14800-903, Araraquara, São Paulo, Brazil.
| | - R P Fernandes
- Federal University of Mato Grosso (UFMT), Department of Chemistry, 78060-900, Cuiabá, Mato Grosso, Brazil
| | - A C S Carvalho
- São Paulo State University (UNESP), Institute of Chemistry, 14800-060, Araraquara, São Paulo, Brazil
| | - I Frigieri
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, 14800-903, Araraquara, São Paulo, Brazil
| | - R C Alves
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, 14800-903, Araraquara, São Paulo, Brazil
| | - M Chorilli
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, 14800-903, Araraquara, São Paulo, Brazil
| |
Collapse
|
9
|
Activation of TRESK background potassium channels by cloxyquin exerts protective effects against excitotoxic-induced brain injury and neuroinflammation in neonatal rats. J Neuroimmunol 2022; 368:577894. [DOI: 10.1016/j.jneuroim.2022.577894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/25/2022] [Accepted: 05/14/2022] [Indexed: 10/18/2022]
|
10
|
Kratimenos P, Vij A, Vidva R, Koutroulis I, Delivoria-Papadopoulos M, Gallo V, Sathyanesan A. Computational analysis of cortical neuronal excitotoxicity in a large animal model of neonatal brain injury. J Neurodev Disord 2022; 14:26. [PMID: 35351004 PMCID: PMC8966144 DOI: 10.1186/s11689-022-09431-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Neonatal hypoxic brain injury is a major cause of intellectual and developmental disability. Hypoxia causes neuronal dysfunction and death in the developing cerebral cortex due to excitotoxic Ca2+-influx. In the translational piglet model of hypoxic encephalopathy, we have previously shown that hypoxia overactivates Ca2+/Calmodulin (CaM) signaling via Sarcoma (Src) kinase in cortical neurons, resulting in overexpression of proapoptotic genes. However, identifying the exact relationship between alterations in neuronal Ca2+-influx, molecular determinants of cell death, and the degree of hypoxia in a dynamic system represents a significant challenge. METHODS We used experimental and computational methods to identify molecular events critical to the onset of excitotoxicity-induced apoptosis in the cerebral cortex of newborn piglets. We used 2-3-day-old piglets (normoxic [Nx], hypoxic [Hx], and hypoxic + Src-inhibitor-treatment [Hx+PP2] groups) for biochemical analysis of ATP production, Ca2+-influx, and Ca2+/CaM-dependent protein kinase kinase 2 (CaMKK2) expression. We then used SimBiology to build a computational model of the Ca2+/CaM-Src-kinase signaling cascade, simulating Nx, Hx, and Hx+PP2 conditions. To evaluate our model, we used Sobol variance decomposition, multiparametric global sensitivity analysis, and parameter scanning. RESULTS Our model captures important molecular trends caused by hypoxia in the piglet brain. Incorporating the action of Src kinase inhibitor PP2 further validated our model and enabled predictive analysis of the effect of hypoxia on CaMKK2. We determined the impact of a feedback loop related to Src phosphorylation of NMDA receptors and activation kinetics of CaMKII. We also identified distinct modes of signaling wherein Ca2+ level alterations following Src kinase inhibition may not be a linear predictor of changes in Bax expression. Importantly, our model indicates that while pharmacological pre-treatment significantly reduces the onset of abnormal Ca2+-influx, there exists a window of intervention after hypoxia during which targeted modulation of Src-NMDAR interaction kinetics in combination with PP2 administration can reduce Ca2+-influx and Bax expression to similar levels as pre-treatment. CONCLUSIONS Our model identifies new dynamics of critical components in the Ca2+/CaM-Src signaling pathway leading to neuronal injury and provides a feasible framework for drug efficacy studies in translational models of neonatal brain injury for the prevention of intellectual and developmental disabilities.
Collapse
Affiliation(s)
- Panagiotis Kratimenos
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, 111 Michigan Avenue, Washington, DC, 20010, USA. .,Department of Pediatrics, Division of Neonatology, Children's National Hospital, Washington DC, USA. .,George Washington University School of Medicine and Health Sciences, Washington DC, USA.
| | - Abhya Vij
- George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | | | - Ioannis Koutroulis
- George Washington University School of Medicine and Health Sciences, Washington DC, USA.,Department of Pediatrics, Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA.,Center for Genetic Medicine Research, Children's National Research Institute and Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, 111 Michigan Avenue, Washington, DC, 20010, USA.,George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Aaron Sathyanesan
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, 111 Michigan Avenue, Washington, DC, 20010, USA. .,George Washington University School of Medicine and Health Sciences, Washington DC, USA.
| |
Collapse
|
11
|
Heydarifard Z, Zadheidar S, Yavarian J, Shatizadeh Malekshahi S, Kalantari S, Mokhtari-Azad T, Shafiei-Jandaghi NZ. Potential role of viral infections in miscarriage and insights into the underlying molecular mechanisms. Congenit Anom (Kyoto) 2022; 62:54-67. [PMID: 34961973 DOI: 10.1111/cga.12458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 11/30/2022]
Abstract
Intrauterine viruses can infect the decidua and placenta and cause adverse effects on the fetus during gestation. This review discusses the contribution of various viral infections to miscarriage and the molecular mechanisms by which viruses can cause devastating effects on healthy fetuses and induce miscarriage. Severe acute respiratory syndrome coronavirus 2 as newly emerged coronavirus was considered here, due to the concerns about its role during pregnancy and inducing miscarriage, as well. In this narrative review, an extensive literature search was conducted to find all studies investigating viral infections in miscarriage and their molecular mechanisms published over the past 20 years. The results of various studies investigating the roles of 20 viral infections in miscarriage are presented. Then, the mechanisms of pregnancy loss in viral infections were addressed, including alteration of trophoblast invasion and placental dysfunction, inducing excessive maternal immune response, and inducing apoptosis in the placental tissue. Viruses may cause pregnancy loss through different mechanisms and our knowledge about these mechanisms can be helpful for controlling or preventing viral infections and achieving a successful pregnancy.
Collapse
Affiliation(s)
- Zahra Heydarifard
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sevrin Zadheidar
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Jila Yavarian
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shirin Kalantari
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari-Azad
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
12
|
Borozdenko DA, Ezdoglian AA, Shmigol TA, Gonchar DI, Lyakhmun DN, Tarasenko DV, Golubev YV, Cherkashova EA, Namestnikova DD, Gubskiy IL, Lagunin AA, Gubsky LV, Chekhonin VP, Borisevich SS, Gureev MA, Shagina AD, Kiseleva NM, Negrebetsky VV, Baukov YI. A Novel Phenylpyrrolidine Derivative: Synthesis and Effect on Cognitive Functions in Rats with Experimental Ishemic Stroke. Molecules 2021; 26:molecules26206124. [PMID: 34684709 PMCID: PMC8541353 DOI: 10.3390/molecules26206124] [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] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022] Open
Abstract
We performed an in silico, in vitro, and in vivo assessment of a potassium 2-[2-(2-oxo-4-phenylpyrrolidin-1-yl) acetamido]ethanesulfonate (compound 1) as a potential prodrug for cognitive function improvement in ischemic brain injury. Using in silico methods, we predicted the pharmacological efficacy and possible safety in rat models. In addition, in silico data showed neuroprotective features of compound 1, which were further supported by in vitro experiments in a glutamate excitotoxicity-induced model in newborn rat cortical neuron cultures. Next, we checked whether compound 1 is capable of crossing the blood-brain barrier in intact and ischemic animals. Compound 1 improved animal behavior both in intact and ischemic rats and, even though the concentration in intact brains was low, we still observed a significant anxiety reduction and activity escalation. We used molecular docking and molecular dynamics to support our hypothesis that compound 1 could affect the AMPA receptor function. In a rat model of acute focal cerebral ischemia, we studied the effects of compound 1 on the behavior and neurological deficit. An in vivo experiment demonstrated that compound 1 significantly reduced the neurological deficit and improved neurological symptom regression, exploratory behavior, and anxiety. Thus, here, for the first time, we show that compound 1 can be considered as an agent for restoring cognitive functions.
Collapse
Affiliation(s)
- Denis A. Borozdenko
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Aiarpi A. Ezdoglian
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Tatiana A. Shmigol
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Darya I. Gonchar
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Dmitri N. Lyakhmun
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Dmitri V. Tarasenko
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Yaroslav V. Golubev
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Elvira A. Cherkashova
- Department of Neurology, Neurosurgery and Medical Genetics, Faculty of Medicine Federal State Budgetary Institution, Federal Center of Brain Research and Neurotechnologies, Federal Medical Bio-logical Agency, 117997 Moscow, Russia; (E.A.C.); (D.D.N.); (I.L.G.)
| | - Daria D. Namestnikova
- Department of Neurology, Neurosurgery and Medical Genetics, Faculty of Medicine Federal State Budgetary Institution, Federal Center of Brain Research and Neurotechnologies, Federal Medical Bio-logical Agency, 117997 Moscow, Russia; (E.A.C.); (D.D.N.); (I.L.G.)
| | - Ilya L. Gubskiy
- Department of Neurology, Neurosurgery and Medical Genetics, Faculty of Medicine Federal State Budgetary Institution, Federal Center of Brain Research and Neurotechnologies, Federal Medical Bio-logical Agency, 117997 Moscow, Russia; (E.A.C.); (D.D.N.); (I.L.G.)
| | - Alexey A. Lagunin
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
- Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - Leonid V. Gubsky
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
- Department of Neurology, Neurosurgery and Medical Genetics, Faculty of Medicine Federal State Budgetary Institution, Federal Center of Brain Research and Neurotechnologies, Federal Medical Bio-logical Agency, 117997 Moscow, Russia; (E.A.C.); (D.D.N.); (I.L.G.)
| | - Vladimir P. Chekhonin
- Department of Medical Nanobiotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Sophia S. Borisevich
- Laboratory of Physical Chemistry, Ufa Institute of Chemistry UFRS RAS, pr. Oktyabrya 71, 450054 Ufa, Russia;
| | - Maxim A. Gureev
- Laboratory of Bioinformatics, Research Center “Digital Biodesign and Personalized Healthcare”, I.M. Sechenov University, 119991 Moscow, Russia;
- Laboratory of Bioinformatics and Computational Modelling of Biological Systems, Department of Computational Biology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anastasia D. Shagina
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Nina M. Kiseleva
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Vadim V. Negrebetsky
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
| | - Yuri I. Baukov
- Department of Medicinal Chemistry and Toxicology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; (D.A.B.); (A.A.E.); (T.A.S.); (D.I.G.); (D.N.L.); (D.V.T.); (Y.V.G.); (A.A.L.); (L.V.G.); (A.D.S.); (N.M.K.); (V.V.N.)
- Correspondence:
| |
Collapse
|
13
|
Chen JS, Wang HK, Hsu CY, Su YT, Chen JS, Liang CL, Hsieh PCH, Wu CC, Kwan AL. HDAC1 deregulation promotes neuronal loss and deficit of motor function in stroke pathogenesis. Sci Rep 2021; 11:16354. [PMID: 34381129 PMCID: PMC8357973 DOI: 10.1038/s41598-021-95837-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
Stroke is a common cause of death worldwide and leads to disability and cognitive dysfunction. Ischemic stroke and hemorrhagic stroke are major categories of stroke, accounting for 68% and 32% of strokes, respectively. Each year, 15 million people experience stroke worldwide, and the stroke incidence is rising. Epigenetic modifications regulate gene transcription and play a major role in stroke. Accordingly, histone deacetylase 1 (HDAC1) participates in DNA damage repair and cell survival. However, the mechanisms underlying the role of HDAC1 in stroke pathogenesis are still controversial. Therefore, we investigated the role of HDAC1 in stroke by using a rat model of endothelin-1-induced brain ischemia. Our results revealed that HDAC1 was deregulated following stroke, and its expressional level and enzymatic activity were decreased. We also used MS-275 to inhibit HDAC1 function in rats exposed to ischemic insult. We found that HDAC1 inhibition promoted the infarct volume, neuronal loss, DNA damage, neuronal apoptosis after stroke, and levels of reactive oxygen species and inflammation cytokines. Additionally, HDAC1 inhibition deteriorated the behavioral outcomes of rats with ischemic insult. Overall, our findings demonstrate that HDAC1 participates in ischemic pathogenesis in the brain and possesses potential for use as a therapeutic target.
Collapse
Affiliation(s)
- Jui-Sheng Chen
- grid.412019.f0000 0000 9476 5696Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ,Department of Neurosurgery, E-Da Dachang Hospital, Kaohsiung, Taiwan ,grid.414686.90000 0004 1797 2180Department of Neurosurgery, E-Da Hospital, Kaohsiung, Taiwan ,grid.28665.3f0000 0001 2287 1366Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan ,grid.411447.30000 0004 0637 1806School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Hao-Kuang Wang
- grid.414686.90000 0004 1797 2180Department of Neurosurgery, E-Da Hospital, Kaohsiung, Taiwan ,grid.411447.30000 0004 0637 1806School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Chien-Yu Hsu
- grid.414686.90000 0004 1797 2180Department of Neurosurgery, E-Da Hospital, Kaohsiung, Taiwan
| | - Yu-Ting Su
- grid.145695.aDepartment of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jia-Shing Chen
- grid.411447.30000 0004 0637 1806School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Cheng-Loong Liang
- grid.414686.90000 0004 1797 2180Department of Neurosurgery, E-Da Hospital, Kaohsiung, Taiwan ,grid.411447.30000 0004 0637 1806School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Patrick Ching-Ho Hsieh
- grid.28665.3f0000 0001 2287 1366Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Cheng-Chun Wu
- grid.411447.30000 0004 0637 1806School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- grid.412019.f0000 0000 9476 5696Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ,grid.412027.20000 0004 0620 9374Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
14
|
Altered neurodevelopmental DNA methylation status after fetal growth restriction with brain-sparing. J Dev Orig Health Dis 2021; 13:378-389. [PMID: 34325767 DOI: 10.1017/s2040174421000374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
It is under debate how preferential perfusion of the brain (brain-sparing) in fetal growth restriction (FGR) relates to long-term neurodevelopmental outcome. Epigenetic modification of neurotrophic genes by altered fetal oxygenation may be involved. To explore this theory, we performed a follow-up study of 21 FGR children, in whom we prospectively measured the prenatal cerebroplacental ratio (CPR) with Doppler sonography. At 4 years of age, we tested their neurodevelopmental outcome using the Wechsler Preschool and Primary Scale of Intelligence, the Child Behavior Checklist, and the Behavior Rating Inventory of Executive Function. In addition, we collected their buccal DNA to determine the methylation status at predefined genetic regions within the genes hypoxia-inducible factor-1 alpha (HIF1A), vascular endothelial growth factor A (VEGFA), erythropoietin (EPO), EPO-receptor (EPOR), brain-derived neurotrophic factor (BDNF), and neurotrophic tyrosine kinase, receptor, type 2 (NTRK2) by pyrosequencing. We found that FGR children with fetal brain-sparing (CPR <1, n = 8) demonstrated a trend (0.05 < p < 0.1) toward hypermethylation of HIF1A and VEGFA at their hypoxia-response element (HRE) compared with FGR children without fetal brain-sparing. Moreover, in cases with fetal brain-sparing, we observed statistically significant hypermethylation at a binding site for cyclic adenosine monophophate response element binding protein (CREB) of BDNF promoter exon 4 and hypomethylation at an HRE located within the NTRK2 promoter (both p <0.05). Hypermethylation of VEGFA was associated with a poorer Performance Intelligence Quotient, while hypermethylation of BDNF was associated with better inhibitory self-control (both p <0.05). These results led us to formulate the hypothesis that early oxygen-dependent epigenetic alterations due to hemodynamic alterations in FGR may be associated with altered neurodevelopmental outcome in later life. We recommend further studies to test this hypothesis.
Collapse
|
15
|
Elsayed NA, Boyer TM, Burd I. Fetal Neuroprotective Strategies: Therapeutic Agents and Their Underlying Synaptic Pathways. Front Synaptic Neurosci 2021; 13:680899. [PMID: 34248595 PMCID: PMC8262796 DOI: 10.3389/fnsyn.2021.680899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/28/2021] [Indexed: 01/31/2023] Open
Abstract
Synaptic signaling is integral for proper brain function. During fetal development, exposure to inflammation or mild hypoxic-ischemic insult may lead to synaptic changes and neurological damage that impairs future brain function. Preterm neonates are most susceptible to these deleterious outcomes. Evaluating clinically used and novel fetal neuroprotective measures is essential for expanding treatment options to mitigate the short and long-term consequences of fetal brain injury. Magnesium sulfate is a clinical fetal neuroprotective agent utilized in cases of imminent preterm birth. By blocking N-methyl-D-aspartate receptors, magnesium sulfate reduces glutamatergic signaling, which alters calcium influx, leading to a decrease in excitotoxicity. Emerging evidence suggests that melatonin and N-acetyl-L-cysteine (NAC) may also serve as novel putative fetal neuroprotective candidates. Melatonin has important anti-inflammatory and antioxidant properties and is a known mediator of synaptic plasticity and neuronal generation. While NAC acts as an antioxidant and a precursor to glutathione, it also modulates the glutamate system. Glutamate excitotoxicity and dysregulation can induce perinatal preterm brain injury through damage to maturing oligodendrocytes and neurons. The improved drug efficacy and delivery of the dendrimer-bound NAC conjugate provides an opportunity for enhanced pharmacological intervention. Here, we review recent literature on the synaptic pathways underlying these therapeutic strategies, discuss the current gaps in knowledge, and propose future directions for the field of fetal neuroprotective agents.
Collapse
Affiliation(s)
- Nada A. Elsayed
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Theresa M. Boyer
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Irina Burd
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
16
|
Zhao Y, Liang L, Liu G, Liu Y, Zheng H, Dai L. The effects of short time hyperoxia on glutamate concentration and glutamate transporters expressions in brain of neonatal rats. Neurosci Lett 2021; 758:136013. [PMID: 34111510 DOI: 10.1016/j.neulet.2021.136013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/27/2021] [Accepted: 06/03/2021] [Indexed: 11/18/2022]
Abstract
Preterm infants often suffer from impaired postnatal brain development, and glutamate excitotoxicity is identified as a pivotal mechanism of hyperoxia-induced neurological abnormality. We aimed to investigate the effect of short time hyperoxia on glutamate homeostasis and glutamate transporters expressions in immature brain. Six-day-old (P6) rat pups were exposed to 80% oxygen for 24 h (the hyperoxia group) or placed in atmospheric air (the control group). The concentrations of glutamate and γ-aminobutyric acid (GABA) in immature cerebrum and cerebellum at P7, P14 and P21 were determined by ELISA. The mRNA levels of glutamate transporters including excitatory amino acid transporter 1 (EAAT1), EAAT2, EAAT3, vesicular glutamate transporter 1 (VGLUT1) and VGLUT2 in brain were determined by qPCR. Glutamate accumulation was induced by hyperoxia both in immature cerebrum and cerebellum at P7 but got gradually attenuated at P14 and P21, as evidenced by the changes of glutamate and GABA concentrations. Hyperoxia also induced sustained glutamatic oxidative stress in both cerebrum and cerebellum, as GSH (reduced glutathione) levels in the hyperoxia group were constantly higher than the control group at three examined time-points. Furthermore, at P7, the expressions of all glutamate transporters decreased in both cerebrum and cerebellum except that of EAAT1. At P21, VGLUT2 in cerebrum and EAAT1, EAAT3 and VGLUT2 in cerebellum still displayed significant decrease in expression levels upon hyperoxia stimulation. Taken together, our results indicate that hyperoxia induces glutamate accumulation in brain of rat pups, which is associated with increased oxidative stress and decreased expressions of glutamate transporters.
Collapse
Affiliation(s)
- Yuwei Zhao
- Neonatology Department, Anhui Provincial Children's Hospital, Hefei, China.
| | - Lei Liang
- Pulmonary Department, Anhui Provincial Children's Hospital, Hefei, China
| | - Guanghui Liu
- Neonatology Department, Anhui Provincial Children's Hospital, Hefei, China
| | - Yongqing Liu
- Neonatology Department, Anhui Provincial Children's Hospital, Hefei, China
| | - Hong Zheng
- Neonatology Department, Anhui Provincial Children's Hospital, Hefei, China
| | - Liying Dai
- Neonatology Department, Anhui Provincial Children's Hospital, Hefei, China
| |
Collapse
|
17
|
Tveden-Nyborg P. Vitamin C Deficiency in the Young Brain-Findings from Experimental Animal Models. Nutrients 2021; 13:1685. [PMID: 34063417 PMCID: PMC8156420 DOI: 10.3390/nu13051685] [Citation(s) in RCA: 9] [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: 04/05/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022] Open
Abstract
Severe and long-term vitamin C deficiency can lead to fatal scurvy, which is fortunately considered rare today. However, a moderate state of vitamin C (vitC) deficiency (hypovitaminosis C)-defined as a plasma concentration below 23 μM-is estimated to affect up to 10% of the population in the Western world, albeit clinical hallmarks in addition to scurvy have not been linked to vitC deficiency. The brain maintains a high vitC content and uniquely high levels during deficiency, supporting vitC's importance in the brain. Actions include both antioxidant and co-factor functions, rendering vitamin C deficiency likely to affect several targets in the brain, and it could be particularly significant during development where a high cellular metabolism and an immature antioxidant system might increase sensitivity. However, investigations of a non-scorbutic state of vitC deficiency and effects on the developing young brain are scarce. This narrative review provides a comprehensive overview of the complex mechanisms that regulate vitC homeostasis in vivo and in the brain in particular. Functions of vitC in the brain and the potential consequences of deficiency during brain development are highlighted, based primarily on findings from experimental animal models. Perspectives for future investigations of vitC are outlined.
Collapse
Affiliation(s)
- Pernille Tveden-Nyborg
- Section of Experimental Animal Models, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Copenhagen, Denmark
| |
Collapse
|
18
|
Memantine ameliorates cognitive impairment induced by exposure to chronic hypoxia environment at high altitude by inhibiting excitotoxicity. Life Sci 2021; 270:119012. [PMID: 33422543 DOI: 10.1016/j.lfs.2020.119012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/19/2020] [Accepted: 12/25/2020] [Indexed: 01/23/2023]
Abstract
AIMS Memantine is a non-competitive antagonist of glutamatergic NMDA receptor that is mainly used in the treatment of Alzheimer's disease. The excitatory toxicity mediated by glutamate via glutamatergic receptor signals is considered to be one of the mechanisms mediating neuronal injury and cognitive impairment after exposure to a hypoxic environment at a high altitude. Therefore, in this study, we hypothesized that inhibiting glutamate signaling using memantine could alleviate neuronal injury and cognitive impairment in rats exposed to chronic hypoxia. MAIN METHODS we made animal models in the natural environment of the Qinghai-Tibet Plateau at an altitude of 4300 m, and used animal behavior, morphology, molecular biology and other methods to evaluate the impact of chronic hypoxia exposure on cognitive function and the neuroprotective effect of Memantine. KEY FINDINGS Our results showed that the expression of NMDA receptors increased, while the expression of AMPA receptors decreased, after 4 weeks of chronic hypoxia exposure. Concomitantly, apoptotic neuronal cell death in the hippocampus and frontal cortex was significantly increased, along with levels of oxidative stress, whereas innate ability to inhibit free radicals decreased. Moreover, after 8 weeks of hypoxia exposure, learning, memory, and space exploration abilities were significantly decreased. Notably, after treatment with memantine, apoptotic neuronal cell death, oxidative stress, and free radical levels decreased, and the cognitive function of the animals improved. SIGNIFICANCE Present study shows that chronic hypoxia can produce the excitatory toxicity leading to neural injury and cognitive impairment that can be suppressed with memantine treatment by inhibiting excitatory toxicity.
Collapse
|
19
|
Vetrovoy O, Stratilov V, Nimiritsky P, Makarevich P, Tyulkova E. Prenatal Hypoxia Induces Premature Aging Accompanied by Impaired Function of the Glutamatergic System in Rat Hippocampus. Neurochem Res 2021; 46:550-563. [PMID: 33389385 DOI: 10.1007/s11064-020-03191-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 11/17/2020] [Accepted: 11/28/2020] [Indexed: 12/27/2022]
Abstract
Prenatal hypoxia is among leading causes of progressive brain pathologies in postnatal life. This study aimed to analyze the characteristics of the hippocampal glutamatergic system and behavior of rats in early (2 weeks), adult (3 months) and advanced (18 months) postnatal ontogenesis after exposure to prenatal severe hypoxia (PSH, 180 Torr, 5% O2, 3 h) during the critical period in the formation of the hippocampus (days 14-16 of gestation). We have shown an age-dependent progressive decrease in the hippocampal glutamate levels, a decrease of the neuronal cell number in the CA1 hippocampal region, as well as impairment of spatial long-term memory in the Morris water navigation task. The gradual decrease of glutamate was accompanied by decreased expression of the genes that mediate glutamate metabolism and recycling in the hippocampus. That deficiency apparently correlated with an increase of the metabotropic glutamate receptor type 1 (mGluR1) and synaptophysin expression. Generation of the lipid peroxidation products in the hippocampus of adult rats subjected to prenatal severe hypoxia (PSH rats) was not increased compared to the control animals when tested in a model of glutamate excitotoxicity induced by severe hypoxia. This demonstrates that excessive glutamate sensitivity in PSH rats does not compensate for glutamate deficiency. Our results show a significant contribution of the glutamate system dysfunction to age-associated decrease of this mediator, cognitive decline, and early neuronal loss in PSH rats.
Collapse
Affiliation(s)
- Oleg Vetrovoy
- Laboratory of Regulation of Brain Neuron Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Makarova emb. 6, Saint-Petersburg, Russia, 199034. .,Department of Biochemistry, Faculty of Biology, Saint-Petersburg State University, Universitetskaya emb. 7-9, Saint-Petersburg, Russia, 199034.
| | - Viktor Stratilov
- Laboratory of Regulation of Brain Neuron Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Makarova emb. 6, Saint-Petersburg, Russia, 199034
| | - Peter Nimiritsky
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Lomonosov Ave. 27-10, Moscow, Russia, 119192.,Faculty of Medicine, Lomonosov Moscow State University, Lomonosov Ave. 31-5, Moscow, Russia, 119192
| | - Pavel Makarevich
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Lomonosov Ave. 27-10, Moscow, Russia, 119192.,Faculty of Medicine, Lomonosov Moscow State University, Lomonosov Ave. 31-5, Moscow, Russia, 119192
| | - Ekaterina Tyulkova
- Laboratory of Regulation of Brain Neuron Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Makarova emb. 6, Saint-Petersburg, Russia, 199034
| |
Collapse
|
20
|
Ginsberg Y, Gutzeit O, Hadad S, Divon MY, Khatib N, Fainaru O, Weiner Z, Beloosesky R. Maternal Progesterone Treatment Reduces Maternal Inflammation-Induced Fetal Brain Injury in a Mouse Model of Preterm Birth. Reprod Sci 2021; 28:166-176. [PMID: 32833191 DOI: 10.1007/s43032-020-00272-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/22/2020] [Indexed: 01/08/2023]
Abstract
Maternal natural vaginal progesterone (nVP) administration has been shown to reduce the risk of preterm birth (PTB). The largest randomized trial of nVP for PTB (OPPTIMUM) noted a sonographic reduction in neonatal brain injury following nVP treatment. We investigated the neuroinflammatory protective effect of maternal nVP in a mouse model for maternal inflammation. Pregnant mice (n = 24) were randomized to nVP (1 mg/day) or vehicle from days 13-16 of gestation. At days 15 and 16, lipopolysaccharide (30 μg) or saline were administered. Mice were sacrificed 4 h following the last injection. Fetal brains and placentas were collected. Levels of NF-κB, nNOS, IL-6, and TNFα were determined by Western blot. Maternal lipopolysaccharide significantly increased fetal brain levels of IL-6 (0.33 ± 0.02 vs. 0.11 ± 0.01 u), TNFα (0.3 ± 0.02 vs. 0.10 ± 0.01 u), NF-κB (0.32 ± 0.01 vs. 0.17 ± 0.01 u), and nNOS (0.24 ± 0.04 vs. 0.08 ± 0.01 u), and reduced the total glutathione levels (0.014 ± 0.001 vs. 0.026 ± 0.001 pmol/μl; p < 0.01) compared with control. Maternal nVP significantly reduced fetal brain levels of IL-6 (0.14 ± 0.01 vs. 0.33 ± 0.02 u), TNFα (0.2 ± 0.06 vs. 0.3 ± 0.02 u), NF-κB (0.16 ± 0.01 vs 0.32 ± 0.01 u), and nNOS (0.14 ± 0.01 vs 0.24 ± 0.04 u), and prevented the reduction of fetal brain total glutathione levels (0.022 ± 0.001 vs. 0.014 ± 0.001 pmol/μl; p < 0.01) to levels similar to controls. A similar pattern was demonstrated in the placenta. Maternal nVP for PTB may protect the fetal brain from inflammation-induced brain injury by inhibiting specific inflammatory and oxidative pathways in both brain and placenta.
Collapse
Affiliation(s)
- Yuval Ginsberg
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel.
| | - Ola Gutzeit
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Salim Hadad
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Michael Y Divon
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Lenox Hill Hospital, Northwell Health, New York City, NY, USA
| | - Nizar Khatib
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Ofer Fainaru
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Zeev Weiner
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| | - Ron Beloosesky
- Department of Obstetrics and Gynecology,Rambam Health Care Campus, 8 Ha'alya St., 38302, Haifa, Israel
| |
Collapse
|
21
|
Scher MS. "The First Thousand Days" Define a Fetal/Neonatal Neurology Program. Front Pediatr 2021; 9:683138. [PMID: 34408995 PMCID: PMC8365757 DOI: 10.3389/fped.2021.683138] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/27/2021] [Indexed: 01/11/2023] Open
Abstract
Gene-environment interactions begin at conception to influence maternal/placental/fetal triads, neonates, and children with short- and long-term effects on brain development. Life-long developmental neuroplasticity more likely results during critical/sensitive periods of brain maturation over these first 1,000 days. A fetal/neonatal program (FNNP) applying this perspective better identifies trimester-specific mechanisms affecting the maternal/placental/fetal (MPF) triad, expressed as brain malformations and destructive lesions. Maladaptive MPF triad interactions impair progenitor neuronal/glial populations within transient embryonic/fetal brain structures by processes such as maternal immune activation. Destructive fetal brain lesions later in pregnancy result from ischemic placental syndromes associated with the great obstetrical syndromes. Trimester-specific MPF triad diseases may negatively impact labor and delivery outcomes. Neonatal neurocritical care addresses the symptomatic minority who express the great neonatal neurological syndromes: encephalopathy, seizures, stroke, and encephalopathy of prematurity. The asymptomatic majority present with neurologic disorders before 2 years of age without prior detection. The developmental principle of ontogenetic adaptation helps guide the diagnostic process during the first 1,000 days to identify more phenotypes using systems-biology analyses. This strategy will foster innovative interdisciplinary diagnostic/therapeutic pathways, educational curricula, and research agenda among multiple FNNP. Effective early-life diagnostic/therapeutic programs will help reduce neurologic disease burden across the lifespan and successive generations.
Collapse
Affiliation(s)
- Mark S Scher
- Division of Pediatric Neurology, Department of Pediatrics, Fetal/Neonatal Neurology Program, Emeritus Scholar Tenured Full Professor in Pediatrics and Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
22
|
Silymarin Inhibits Glutamate Release and Prevents against Kainic Acid-Induced Excitotoxic Injury in Rats. Biomedicines 2020; 8:biomedicines8110486. [PMID: 33182349 PMCID: PMC7695262 DOI: 10.3390/biomedicines8110486] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Silymarin, a polyphenoic flavonoid derived from the seeds of milk thistle (Silybum marianum), exhibits neuroprotective effects. In this study, we used a model of rat cerebrocortical synaptosomes to investigate whether silymarin affects the release of glutamate, an essential neurotransmitter involved in excitotoxicity. Its possible neuroprotective effect on a rat model of kainic acid (KA)-induced excitotoxicity was also investigated. In rat cortical synaptosomes, silymarin reduced glutamate release and calcium elevation evoked by the K+ channel blocker 4-aminopyridine but did not affect glutamate release caused by the Na+ channel activator veratridine or the synaptosomal membrane potential. Decreased glutamate release by silymarin was prevented by removal of extracellular calcium and blocking of N- and P/Q-type Ca2+ channel or extracellular signal-regulated kinase 1/2 (ERK1/2) but not by blocking of intracellular Ca2+ release. Immunoblotting assay results revealed that silymarin reduced 4-aminopyridine-induced phosphorylation of ERK1/2. Moreover, systemic treatment of rats with silymarin (50 or 100 mg/kg) 30 min before systemic KA (15 mg/kg) administration attenuated KA-induced seizures, glutamate concentration elevation, neuronal damage, glial activation, and heat shock protein 70 expression as well as upregulated KA-induced decrease in Akt phosphorylation in the rat hippocampus. Taken together, the present study demonstrated that silymarin depressed synaptosomal glutamate release by suppressing voltage-dependent Ca2+ entry and ERK1/2 activity and effectively prevented KA-induced in vivo excitotoxicity.
Collapse
|
23
|
Yu H, Shao J, Huang R, Guan Y, Li G, Chen S, Zhou F, Yao Q, Shen J. Targeting PTEN to regulate autophagy and promote the repair of injured neurons. Brain Res Bull 2020; 165:161-168. [PMID: 33049350 DOI: 10.1016/j.brainresbull.2020.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 08/21/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
The effects of autophagy on neuronal damage can be positive or detrimental negative. Through establishing a model of fetal rat cortical neuron hydraulic shock injury, dipotassium bisperoxo (picolinoto) oxovanadate (V) [bpv(pic)] was used to inhibit PTEN at different time points post-injury and autophagy level after neuronal injury was assessed. Neurons were divided into several intervention groups according to the time point at which bpv(pic) was used to inhibit autophagy, normal neurons and injuried neurons were set as two control groups. Growth of neurons in each group was assessed through immunofluorescence staining. Expression of the autophagy-related proteins LC3-II and LC3-I was analyzed by western blot. Expression of PTEN, mTOR and Beclin-1 was detected by RT-PCR. The number of autophagosomes in the normal group, injury control group and 24 h, 36 h intervention groups were assessed by electron microscope. We found that autophagy was enhanced after neuronal injury and that the levels of LC3-II was significantly reduced by bpv (pic) intervention. The growth of the injury control groups was worse than normal groups, while improved through bpv(pic) intervention at 24 h and 30 h after injured. Western blot analysis showed that the LC3-II and LC3-II/LC3-I ratios of cells increased post-injury, and autophagy induction was evident by electron microscopy. These effects were confirmed by RT-PCR analysis. Taken together, these data suggest that autophagy is activated after injury in neurons while can be inhibited by bpv(pic) administration and then promote the repair of injured neurons.
Collapse
Affiliation(s)
- Haoyuan Yu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Junjie Shao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Runxin Huang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Yixiang Guan
- Department of Neurosurgery, Affiliated HaianHospital of Nantong University, Nantong, 226001, China
| | - Guicai Li
- Key Laboratory of Neuroregenerationof Jiangsu and Ministry of Education, Co-innovation Center ofNeuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Shiyu Chen
- Key Laboratory of Neuroregenerationof Jiangsu and Ministry of Education, Co-innovation Center ofNeuroregeneration, Nantong University, Nantong, 226001, Jiangsu, China
| | - Fei Zhou
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Qi Yao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Jianhong Shen
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, 226001, China.
| |
Collapse
|
24
|
Bratek E, Ziembowicz A, Salinska E. N-Acetylaspartylglutamate (NAAG) Pretreatment Reduces Hypoxic-Ischemic Brain Damage and Oxidative Stress in Neonatal Rats. Antioxidants (Basel) 2020; 9:antiox9090877. [PMID: 32957477 PMCID: PMC7555246 DOI: 10.3390/antiox9090877] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
N-acetylaspartylglutamate (NAAG), the most abundant peptide transmitter in the mammalian nervous system, activates mGluR3 at presynaptic sites, inhibiting the release of glutamate, and acts on mGluR3 on astrocytes, stimulating the release of neuroprotective growth factors (TGF-β). NAAG can also affect N-methyl-d-aspartate (NMDA) receptors in both synaptic and extrasynaptic regions. NAAG reduces neurodegeneration in a neonatal rat model of hypoxia-ischemia (HI), although the exact mechanism is not fully recognized. In the present study, the effect of NAAG application 24 or 1 h before experimental birth asphyxia on oxidative stress markers and the potential mechanisms of neuroprotection on 7-day old rats was investigated. The intraperitoneal application of NAAG at either time point before HI significantly reduced the weight deficit of the ischemic brain hemisphere, radical oxygen species (ROS) content and activity of antioxidant enzymes, and increased the concentration of reduced glutathione (GSH). No additional increase in the TGF-β concentration was observed after NAAG application. The fast metabolism of NAAG and the decrease in TGF-β concentration that resulted from NAAG pretreatment, performed up to 24 h before HI, excluded the involvement mGluR3 in neuroprotection. The observed effect may be explained by the activation of NMDA receptors induced by NAAG pretreatment 24 h before HI. Inhibition of the NAAG effect by memantine supports this conclusion. NAAG preconditioning 1 h before HI results in a mixture of mGluR3 and NMDA receptor activation. Preconditioning with NAAG induces the antioxidative defense system triggered by mild excitotoxicity in neurons. Moreover, this response to NAAG pretreatment is consistent with the commonly accepted mechanism of preconditioning. However, this theory requires further investigation.
Collapse
|
25
|
Abstract
Perinatal hypoxia is still one of the greatest threats to the newborn child, even in developed countries. However, there is a lack of works which summarize up-to-date information about that huge topic. Our review covers a broader spectrum of recent results from studies on mechanisms leading to hypoxia-induced injury. It also resumes possible primary causes and observed behavioral outcomes of perinatal hypoxia. In this review, we recognize two types of hypoxia, according to the localization of its primary cause: environmental and placental. Later we analyze possible pathways of prenatal hypoxia-induced injury including gene expression changes, glutaminergic excitatory damage (and a role of NMDA receptors in it), oxidative stress with ROS and RNS production, inflammation and apoptosis. Moreover, we focus on the impact of these pathophysiological changes on the structure and development of the brain, especially on its regions: corpus striatum and hippocampus. These brain changes of the offspring lead to impairments in their postnatal growth and sensorimotor development, and in their motor functions, activity, emotionality and learning ability in adulthood. Later we compare various animal models used to investigate the impact of prenatal and postnatal injury (hypoxic, ischemic or combinatory) on living organisms, and show their advantages and limitations.
Collapse
Affiliation(s)
- M Piešová
- Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | |
Collapse
|
26
|
Wang XG, Zhu DD, Li N, Huang YL, Wang YZ, Zhang T, Wang CM, Wang B, Peng Y, Ge BY, Li S, Zhao J. Scorpion Venom Heat-Resistant Peptide is Neuroprotective against Cerebral Ischemia-Reperfusion Injury in Association with the NMDA-MAPK Pathway. Neurosci Bull 2020; 36:243-253. [PMID: 31502213 PMCID: PMC7056763 DOI: 10.1007/s12264-019-00425-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/03/2019] [Indexed: 12/19/2022] Open
Abstract
Scorpion venom heat-resistant peptide (SVHRP) is a component purified from Buthus martensii Karsch scorpion venom. Our previous studies have shown that SVHRP is neuroprotective in models of Alzheimer's disease and Parkinson's disease. The present study aimed to explore the potential neuroprotective effects of SVHRP on cerebral ischemia/reperfusion (I/R) injury, using a mouse model of middle cerebral artery occlusion/reperfusion (MCAO/R) and a cellular model of oxygen-glucose deprivation/reoxygenation (OGD/R). Our results showed that SVHRP treatment decreased the neurological deficit scores, edema formation, infarct volume and neuronal loss in the MCAO/R mice, and protected primary neurons against OGD/R insult. SVHRP pretreatment suppressed the alterations in protein levels of N-methyl-D-aspartate receptors (NMDARs) and phosphorylated p38 MAPK as well as some proinflammatory factors in both the animal and cellular models. These results suggest that SVHRP has neuroprotective effects against cerebral I/R injury, which might be associated with inhibition of the NMDA-MAPK-mediated excitotoxicity.
Collapse
Affiliation(s)
- Xu-Gang Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
- National-Local Joint Engineering Research Center for Drug-Research and Development of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116000, China
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Dan-Dan Zhu
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Na Li
- National-Local Joint Engineering Research Center for Drug-Research and Development of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116000, China
| | - Yue-Lin Huang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ying-Zi Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
- The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Ting Zhang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Chen-Mei Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bin Wang
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yan Peng
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bi-Ying Ge
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
- National-Local Joint Engineering Research Center for Drug-Research and Development of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116000, China.
| | - Jie Zhao
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
- National-Local Joint Engineering Research Center for Drug-Research and Development of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116000, China.
| |
Collapse
|
27
|
Abbasi H, Unsworth CP. Applications of advanced signal processing and machine learning in the neonatal hypoxic-ischemic electroencephalogram. Neural Regen Res 2020; 15:222-231. [PMID: 31552887 PMCID: PMC6905345 DOI: 10.4103/1673-5374.265542] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/24/2019] [Indexed: 01/15/2023] Open
Abstract
Perinatal hypoxic-ischemic-encephalopathy significantly contributes to neonatal death and life-long disability such as cerebral palsy. Advances in signal processing and machine learning have provided the research community with an opportunity to develop automated real-time identification techniques to detect the signs of hypoxic-ischemic-encephalopathy in larger electroencephalography/amplitude-integrated electroencephalography data sets more easily. This review details the recent achievements, performed by a number of prominent research groups across the world, in the automatic identification and classification of hypoxic-ischemic epileptiform neonatal seizures using advanced signal processing and machine learning techniques. This review also addresses the clinical challenges that current automated techniques face in order to be fully utilized by clinicians, and highlights the importance of upgrading the current clinical bedside sampling frequencies to higher sampling rates in order to provide better hypoxic-ischemic biomarker detection frameworks. Additionally, the article highlights that current clinical automated epileptiform detection strategies for human neonates have been only concerned with seizure detection after the therapeutic latent phase of injury. Whereas recent animal studies have demonstrated that the latent phase of opportunity is critically important for early diagnosis of hypoxic-ischemic-encephalopathy electroencephalography biomarkers and although difficult, detection strategies could utilize biomarkers in the latent phase to also predict the onset of future seizures.
Collapse
Affiliation(s)
- Hamid Abbasi
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - Charles P. Unsworth
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
28
|
Liu Y, Wang S, Kan J, Zhang J, Zhou L, Huang Y, Zhang Y. Chinese Herbal Medicine Interventions in Neurological Disorder Therapeutics by Regulating Glutamate Signaling. Curr Neuropharmacol 2020; 18:260-276. [PMID: 31686629 PMCID: PMC7327939 DOI: 10.2174/1570159x17666191101125530] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 10/23/2019] [Accepted: 10/31/2019] [Indexed: 12/31/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system, and its signaling is critical for excitatory synaptic transmission. The well-established glutamate system involves glutamate synthesis, presynaptic glutamate release, glutamate actions on the ionotropic glutamate receptors (NMDA, AMPA, and kainate receptors) and metabotropic glutamate receptors, and glutamate uptake by glutamate transporters. When the glutamate system becomes dysfunctional, it contributes to the pathogenesis of neurodegenerative and neuropsychiatric diseases such as Alzheimer's disease, Parkinson's disease, depression, epilepsy, and ischemic stroke. In this review, based on regulating glutamate signaling, we summarize the effects and underlying mechanisms of natural constituents from Chinese herbal medicines on neurological disorders. Natural constituents from Chinese herbal medicine can prevent the glutamate-mediated excitotoxicity via suppressing presynaptic glutamate release, decreasing ionotropic and metabotropic glutamate receptors expression in the excitatory synapse, and promoting astroglial glutamate transporter expression to increase glutamate clearance from the synaptic cleft. However, some natural constituents from Chinese herbal medicine have the ability to restore the collapse of excitatory synapses by promoting presynaptic glutamate release and increasing ionotropic and metabotropic glutamate receptors expression. These regulatory processes involve various signaling pathways, which lead to different mechanistic routes of protection against neurological disorders. Hence, our review addresses the underlying mechanisms of natural constituents from Chinese herbal medicines that regulate glutamate systems and serve as promising agents for the treatment of the above-mentioned neurological disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yunlong Zhang
- Address correspondence to this author at the Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China; Tel: +86-20-37105182;, E-mail: and Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan 528300, China; E-mail:
| |
Collapse
|
29
|
Abbasi H, Unsworth CP. Electroencephalogram studies of hypoxic ischemia in fetal and neonatal animal models. Neural Regen Res 2020; 15:828-837. [PMID: 31719243 PMCID: PMC6990791 DOI: 10.4103/1673-5374.268892] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Alongside clinical achievements, experiments conducted on animal models (including primate or non-primate) have been effective in the understanding of various pathophysiological aspects of perinatal hypoxic/ischemic encephalopathy (HIE). Due to the reasonably fair degree of flexibility with experiments, most of the research around HIE in the literature has been largely concerned with the neurodevelopmental outcome or how the frequency and duration of HI seizures could relate to the severity of perinatal brain injury, following HI insult. This survey concentrates on how EEG experimental studies using asphyxiated animal models (in rodents, piglets, sheep and non-human primate monkeys) provide a unique opportunity to examine from the exact time of HI event to help gain insights into HIE where human studies become difficult.
Collapse
Affiliation(s)
- Hamid Abbasi
- Department of Engineering Science, the University of Auckland, Auckland, New Zealand
| | - Charles P Unsworth
- Department of Engineering Science, the University of Auckland, Auckland, New Zealand
| |
Collapse
|
30
|
Richter AE, Scherjon SA, Dikkers R, Bos AF, Kooi EMW. Antenatal Magnesium Sulfate and Preeclampsia Differentially Affect Neonatal Cerebral Oxygenation. Neonatology 2020; 117:331-340. [PMID: 32516784 DOI: 10.1159/000507705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/25/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Magnesium sulfate (MgSO4) is frequently administered for maternal and fetal neuroprotection in preeclampsia (PE) and imminent preterm birth, respectively. OBJECTIVE To assess whether MgSO4 affects neonatal cerebral oxygenation, blood flow, and cerebral autoregulation (CAR) during the first postnatal days independently from PE. METHODS 148 neonates <32 weeks gestational age were included. Cerebral fractional tissue oxygen extraction (cFTOE) was extracted from a daily 2-h period, during which peak systolic blood flow velocity (PSV) and resistance index (RI) of the pericallosal artery were obtained. The percent time of impaired CAR (correlation coefficient between mean arterial blood pressure and cerebral oxygen saturation >0.5) was determined. Linear mixed models were applied. RESULTS MgSO4 exposure was recorded in 77 neonates. Twenty-nine neonates were born following PE. MgSO4 independently lowered cFTOE (B: -0.026, 95% CI: -0.050 to 0.002, p < 0.05) but did not affect PSV and RI. PE was associated with a lower cFTOE (B: -0.041, 95% CI: -0.067 to -0.015, p < 0.05) and a tendency towards both lower PSV (B: -4.285, 95% CI: -9.067 to 0.497, p < 0.1) and more impaired CAR (B: 4.042, 95% CI: -0.028 to 8.112, p < 0.1), which seemed to be strongly mediated by fetal brain sparing. MgSO4 did not alter CAR. CONCLUSIONS In contrast to fetal brain sparing in PE, MgSO4 seems to lower cFTOE by lowering cerebral oxygen demands in preterm neonates without affecting the cerebrovasculature.
Collapse
Affiliation(s)
- Anne E Richter
- Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands,
| | - Sicco A Scherjon
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Riksta Dikkers
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Arend F Bos
- Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elisabeth M W Kooi
- Division of Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Scher MS. Fetal neurology: Principles and practice with a life-course perspective. HANDBOOK OF CLINICAL NEUROLOGY 2019; 162:1-29. [PMID: 31324306 DOI: 10.1016/b978-0-444-64029-1.00001-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Clinical service, educational, and research components of a fetal/neonatal neurology program are anchored by the disciplines of developmental origins of health and disease and life-course science as programmatic principles. Prenatal participation provides perspectives on maternal, fetal, and placental contributions to health or disease for fetal and subsequent neonatal neurology consultations. This program also provides an early-life diagnostic perspective for neurologic specialties concerned with brain health and disease throughout childhood and adulthood. Animal models and birth cohort studies have demonstrated how the science of epigenetics helps to understand gene-environment interactions to better predict brain health or disease. Fetal neurology consultations provide important diagnostic contributions during critical or sensitive periods of brain development when future neurotherapeutic interventions will maximize adaptive neuroplasticity. Age-specific normative neuroinformatics databases that employ computer-based strategies to integrate clinical/demographic, neuroimaging, neurophysiologic, and genetic datasets will more accurately identify either symptomatic patients or those at risk for brain disorders who would benefit from preventive, rescue, or reparative treatment choices throughout the life span.
Collapse
Affiliation(s)
- Mark S Scher
- Division of Pediatric Neurology, Case Western Reserve University, Cleveland, OH, United States.
| |
Collapse
|
32
|
Confortim HD, Deniz BF, de Almeida W, Miguel PM, Bronauth L, Vieira MC, de Oliveira BC, Pereira LO. Neonatal hypoxia-ischemia caused mild motor dysfunction, recovered by acrobatic training, without affecting morphological structures involved in motor control in rats. Brain Res 2018; 1707:27-44. [PMID: 30448443 DOI: 10.1016/j.brainres.2018.11.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/29/2018] [Accepted: 11/14/2018] [Indexed: 10/27/2022]
Abstract
The aim of this study was to evaluated motor function and morphological aspects of the components involved in motor control (sensorimotor cortex, spinal cord, sciatic nerve, neuromuscular junctions and skeletal muscle) in male Wistar rats exposed to a model of neonatal hypoxic-ischemic encephalopathy (HIE) and the possible influence of different physical exercise protocols - treadmill and acrobatic. Male Wistar rats at the 7th post-natal day (PND) were submitted to the HIE model and from the 22nd until 60th PND the exercise protocols (treadmill or acrobatic training) were running. After the training, the animals were evaluated in Open Field, Ladder Rung Walking and Rotarod tasks and after samples of the motor control components were collected. Our results evidenced that the acrobatic training reversed the hyperactivity and anxiety, caused locomotion improvement and decreased brain atrophy in HIE animals. We did not find morphological differences on sensorimotor cortex, spinal cord, sciatic nerve, neuromuscular junctions and skeletal muscle in the animals submitted to HIE model. These intriguing data support the statement of the Rice-Vannucci model does not seem to reproduce, in structures involved in control function, the damage found in humans that suffer HIE. Regarding the protocols of exercise, we proposed that the acrobatic exercise could be a good therapeutic option especially in children affected by neonatal HIE and can be responsible for good results in cognitive and motor aspects.
Collapse
Affiliation(s)
- Heloísa Deola Confortim
- Programa de Pós-Graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, sala 107, 90050-170 Porto Alegre, RS, Brazil; Departamento de Ciências Morfológicas, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - Bruna Ferrary Deniz
- Programa de Pós-Graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, sala 107, 90050-170 Porto Alegre, RS, Brazil; Departamento de Ciências Morfológicas, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - Wellington de Almeida
- Programa de Pós-Graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, sala 107, 90050-170 Porto Alegre, RS, Brazil; Departamento de Ciências Morfológicas, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - Patrícia Maidana Miguel
- Programa de Pós-Graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, sala 107, 90050-170 Porto Alegre, RS, Brazil; Departamento de Ciências Morfológicas, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - Loise Bronauth
- Departamento de Ciências Morfológicas, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - Milene Cardoso Vieira
- Departamento de Ciências Morfológicas, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - Bruna Chaves de Oliveira
- Departamento de Ciências Morfológicas, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil
| | - Lenir Orlandi Pereira
- Programa de Pós-Graduação em Neurociências, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, sala 107, 90050-170 Porto Alegre, RS, Brazil; Departamento de Ciências Morfológicas, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite, 500, 90050-170 Porto Alegre, RS, Brazil.
| |
Collapse
|
33
|
Fitzgerald E, Boardman JP, Drake AJ. Preterm Birth and the Risk of Neurodevelopmental Disorders - Is There a Role for Epigenetic Dysregulation? Curr Genomics 2018; 19:507-521. [PMID: 30386170 PMCID: PMC6158617 DOI: 10.2174/1389202919666171229144807] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 04/06/2017] [Accepted: 12/17/2017] [Indexed: 12/29/2022] Open
Abstract
Preterm Birth (PTB) accounts for approximately 11% of all births worldwide each year and is a profound physiological stressor in early life. The burden of neuropsychiatric and developmental impairment is high, with severity and prevalence correlated with gestational age at delivery. PTB is a major risk factor for the development of cerebral palsy, lower educational attainment and deficits in cognitive functioning, and individuals born preterm have higher rates of schizophrenia, autistic spectrum disorder and attention deficit/hyperactivity disorder. Factors such as gestational age at birth, systemic inflammation, respiratory morbidity, sub-optimal nutrition, and genetic vulnerability are associated with poor outcome after preterm birth, but the mechanisms linking these factors to adverse long term outcome are poorly understood. One potential mechanism linking PTB with neurodevelopmental effects is changes in the epigenome. Epigenetic processes can be defined as those leading to altered gene expression in the absence of a change in the underlying DNA sequence and include DNA methylation/hydroxymethylation and histone modifications. Such epigenetic modifications may be susceptible to environmental stimuli, and changes may persist long after the stimulus has ceased, providing a mechanism to explain the long-term consequences of acute exposures in early life. Many factors such as inflammation, fluctuating oxygenation and excitotoxicity which are known factors in PTB related brain injury, have also been implicated in epigenetic dysfunction. In this review, we will discuss the potential role of epigenetic dysregulation in mediating the effects of PTB on neurodevelopmental outcome, with specific emphasis on DNA methylation and the α-ketoglutarate dependent dioxygenase family of enzymes.
Collapse
Affiliation(s)
| | | | - Amanda J. Drake
- Address correspondence to this author at the University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh EH16 4TJ, UK; Tel: 44 131 2426748; Fax: 44 131 2426779; E-mail:
| |
Collapse
|
34
|
Lee JS, Kim WY, Jeon YJ, Lee SK, Son CG. Aquilariae Lignum extract attenuates glutamate-induced neuroexcitotoxicity in HT22 hippocampal cells. Biomed Pharmacother 2018; 106:1031-1038. [PMID: 30119168 DOI: 10.1016/j.biopha.2018.07.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 01/08/2023] Open
Abstract
An imbalance between excitatory and inhibitory neurotransmitters is known to induce neuronal excitotoxicity which is a major cause of neurodegenerative disorders. Excessive glutamate concentration leads to the neuronal death by increasing oxidative stress and affecting the apoptotic signaling pathway. We investigated the anti-excitotoxic effects and associated working mechanisms of 30% ethanol extract of Aquilariae Lignum (ALE) against hippocampal neuronal death by glutamate. HT22 cells were treated with glutamate (20 mM) for 24 h following pretreatment with ALE (5, 10, 25 μg/mL). Cell viability, biochemical analysis, flow chemistry, and Western blotting assays were performed. Glutamate treatment substantially increased the intracellular level of reactive oxygen species (ROS) and Ca2+ influx into the cell, which were followed by apoptosis. ALE pretreatment, however, significantly attenuated these excitotoxicity-related features according to the results of Annexin V analysis and the lactate dehydrogenase assay, in which the calpain pathway (in a caspase 3-independent manner) may be involved. ALE pretreatment also significantly attenuated the glutamate-induced activation of both inflammation-associated molecules (extracellular signal-regulated kinase, c-Jun N-terminal kinases and p38) and death-related molecules (p53, apoptosis-inducing factor). The inactivation of brain-derived neurotrophic factor (BDNF) was restored by ALE pretreatment. Our results verified that A. Lignum has potential neuroprotective effects on glutamate-induced excitotoxicity in hippocampal neuron cells, and its underlying mechanism may involve the regulation of ROS-mediated cell death pathways.
Collapse
Affiliation(s)
- Jin-Seok Lee
- Liver and Immunology Research Center, Oriental Medical Collage of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon, 301-724, Republic of Korea
| | - Won-Yong Kim
- Liver and Immunology Research Center, Oriental Medical Collage of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon, 301-724, Republic of Korea
| | - Yoo-Jin Jeon
- Liver and Immunology Research Center, Oriental Medical Collage of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon, 301-724, Republic of Korea
| | - Sam-Keun Lee
- Department of Applied Chemistry, Oriental Medicine Collage of Daejeon University, 62, Daehak-ro, Dong-gu, Daejeon, 300-716, Republic of Korea
| | - Chang-Gue Son
- Liver and Immunology Research Center, Oriental Medical Collage of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon, 301-724, Republic of Korea.
| |
Collapse
|
35
|
Abstract
Perinatal brain injury may lead to long-term morbidity and neurodevelopmental impairment. Improvements in perinatal care have resulted in the survival of more infants with perinatal brain injury. The effects of hypoxia-ischemia, inflammation, and infection during critical periods of development can lead to a common pathway of perinatal brain injury marked by neuronal excitotoxicity, cellular apoptosis, and microglial activation. Various interventions can prevent or improve the outcomes of different types of perinatal brain injury. The objective of this article is to review the mechanisms of perinatal brain injury, approaches to prevention, and outcomes among children with perinatal brain injury.
Collapse
|
36
|
Association between maternal exposure to phthalates and lower language ability in offspring derived from hair metabolome analysis. Sci Rep 2018; 8:6745. [PMID: 29712949 PMCID: PMC5928220 DOI: 10.1038/s41598-018-24936-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 04/10/2018] [Indexed: 12/03/2022] Open
Abstract
The fetus undergoes a crucial period of neurodevelopment in utero. The maternal hair metabolome provides an integrated record of the metabolic state of the mother prior to, and during pregnancy. We investigated whether variation in the maternal hair metabolome was associated with neurodevelopmental differences across infants. Maternal hair samples and infant neurocognitive assessments (using the Bayley III Scales of Infant Development at 24 months) were obtained for 373 infant-mother dyads between 26–28 weeks’ gestation from the Growing Up in Singapore Towards Healthy Outcomes cohort. The hair metabolome was analysed using gas chromatography-mass spectrometry. Intensity measurements were obtained for 276 compounds. After controlling for maternal education, ethnicity, and infant sex, associations between metabolites and expressive language skills were detected, but not for receptive language, cognitive or motor skills. The results confirm previous research associating higher levels of phthalates with lower language ability. In addition, scores were positively associated with a cluster of compounds, including adipic acid and medium-chain fatty acids. The data support associations between the maternal hair metabolome and neurodevelopmental processes of the fetus. The association between phthalates and lower language ability highlights a modifiable risk factor that warrants further investigation.
Collapse
|
37
|
Johnston MV. Cognitive Development One Year After Infantile Critical Pertussis. Pediatr Crit Care Med 2018; 19:161-162. [PMID: 29394223 PMCID: PMC5798490 DOI: 10.1097/pcc.0000000000001394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Michael V Johnston
- Kennedy Krieger Institute and Johns Hopkins School of Medicine, Baltimore, MD
| |
Collapse
|
38
|
Li T, Luo Z, Liu Y, Wang M, Yu X, Cao C, Liao Z, Ding Y, Yue S. Excessive Activation of NMDA Receptors Induced Neurodevelopmental Brain Damage and Cognitive Deficits in Rats Exposed to Intrauterine Hypoxia. Neurochem Res 2017; 43:566-580. [DOI: 10.1007/s11064-017-2451-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 11/14/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022]
|
39
|
Jiang S, Fang DF, Chen Y. Involvement of N-Methyl-D-Aspartic Acid Receptor and DL-α-Amino-3-Hydroxy-5- Methyl-4-Isoxazole Propionic Acid Receptor in Ginsenosides Rb1 and Rb3 against Oxygen-Glucose Deprivation-Induced Injury in Hippocampal Slices from Rat. Pharmacology 2017; 101:133-139. [PMID: 29207398 DOI: 10.1159/000481710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/22/2017] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Ginsenosides, Rb1 and Rb3, are the major protopanaxadiol components of ginseng saponin. In the present study, the influences of ginsenosides Rb1 and Rb3 on N-methyl-D-aspartic acid (NMDA) receptor or DL-α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor-mediated synaptic transmission after oxygen-glucose deprivation (OGD) were investigated. METHODS NMDA receptor population spike (NMDA-PS) or AMPA receptor-mediated population spike (AMPA-PS) was recorded in the CA1 pyramidal cell layer of rat hippocampal slices by electrophysiological techniques. RESULTS Under normal conditions, ginsenosides Rb3 and Rb1 depressed glutamate receptors-mediated synaptic transmission. Fourteen min of OGD resulted in a poor recovery amplitude of NMDA-PS or AMPA-PS after reoxygenation. Ginsenoside Rb3 significantly delayed the appearance of transient recovery of PS during OGD, and improved the recovery amplitudes of NMDA-PS and AMPA-PS after reoxygenation. However, the similar protective effects of ginsenoside Rb1 were observed only on NMDA-PS but not AMPA-PS. CONCLUSION These results suggest that ginsenosides Rb1 and Rb3 have the different inhibitions on NMDA and AMPA receptors-mediated response, which may partially explain the different protective effects of these agents on ischemic neuronal death.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Anatomy and Physiology, Lianyungang Branch of Traditional Chinese Medicine, Jiangsu Union Technical Institute, Lianyungang, China
| | - De-Fang Fang
- Department of Anatomy and Physiology, Lianyungang Branch of Traditional Chinese Medicine, Jiangsu Union Technical Institute, Lianyungang, China
| | - Ying Chen
- Department of Anesthesiology, The First People's Hospital of Lianyungang, Lianyungang, China
| |
Collapse
|
40
|
Kannan G, Kambhampati SP, Kudchadkar SR. Effect of anesthetics on microglial activation and nanoparticle uptake: Implications for drug delivery in traumatic brain injury. J Control Release 2017; 263:192-199. [DOI: 10.1016/j.jconrel.2017.03.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/01/2017] [Accepted: 03/19/2017] [Indexed: 02/01/2023]
|
41
|
Fineschi V, Viola RV, La Russa R, Santurro A, Frati P. A Controversial Medicolegal Issue: Timing the Onset of Perinatal Hypoxic-Ischemic Brain Injury. Mediators Inflamm 2017; 2017:6024959. [PMID: 28883688 PMCID: PMC5572618 DOI: 10.1155/2017/6024959] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/18/2017] [Indexed: 12/11/2022] Open
Abstract
Perinatal hypoxic-ischemic brain injury, as a result of chronic, subacute, and acute insults, represents the pathological consequence of fetal distress and birth or perinatal asphyxia, that is, "nonreassuring fetal status." Hypoxic-ischemic injury (HII) is typically characterized by an early phase of damage, followed by a delayed inflammatory local response, in an apoptosis-necrosis continuum. In the early phase, the cytotoxic edema and eventual acute lysis take place; with reperfusion, additional damage should be assigned to excitotoxicity and oxidative stress. Finally, a later phase involves all the inflammatory activity and long-term neural tissue repairing and remodeling. In this model mechanism, loss of mitochondrial function is supposed to be the hallmark of secondary injury progression, and autophagy which is lysosome-mediated play a role in enhancing brain injury. Early-induced molecules driven by hypoxia, as chaperonins HSPs and ORP150, besides common markers for inflammatory responses, have predictive value in timing the onset of neonatal HII; on the other hand, clinical biomarkers for HII diagnosis, as CK-BB, LDH, S-100beta, and NSE, could be useful to predict outcomes.
Collapse
Affiliation(s)
- Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Rocco Valerio Viola
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
| | - Raffaele La Russa
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Alessandro Santurro
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
| | - Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00185 Rome, Italy
- IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| |
Collapse
|
42
|
Lee YA. White Matter Injury of Prematurity: Its Mechanisms and Clinical Features. J Pathol Transl Med 2017; 51:449-455. [PMID: 28797157 PMCID: PMC5611534 DOI: 10.4132/jptm.2017.07.25] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/19/2017] [Accepted: 07/25/2017] [Indexed: 11/17/2022] Open
Abstract
A developing central nervous system is vulnerable to various insults such as infection and ischemia. While increased understanding of the dynamic nature of brain development allows a deeper insight into the pathophysiology of perinatal brain injury, the precise nature of specific fetal and neonatal brain injuries and their short- and long-term clinical consequences need special attention and further elucidation. The current review will describe the pathophysiological aspects and clinical significance of white matter injury of prematurity, a main form of perinatal brain injury in premature newborns, with a particular emphasis on its potential antenatal components.
Collapse
Affiliation(s)
- Young Ah Lee
- Division of Pediatric Neurology, Department of Pediatrics, Beaumont Hospital, Oakland University School of Medicine, Royal Oak, MI, USA
| |
Collapse
|
43
|
The zebrafish miR-125c is induced under hypoxic stress via hypoxia-inducible factor 1α and functions in cellular adaptations and embryogenesis. Oncotarget 2017; 8:73846-73859. [PMID: 29088751 PMCID: PMC5650306 DOI: 10.18632/oncotarget.17994] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/06/2017] [Indexed: 12/15/2022] Open
Abstract
Hypoxia is a unique environmental stress. Hypoxia inducible factor-lα (HIF-lα) is a major transcriptional regulator of cellular adaptations to hypoxic stress. MicroRNAs (miRNAs) as posttranscriptional gene expression regulators occupy a crucial role in cell survival under low-oxygen environment. Previous evidences suggested that miR-125c is involved in hypoxia adaptation, but its precise biological roles and the regulatory mechanism underlying hypoxic responses remain unknown. The present study showed that zebrafish miR-125c is upregulated by hypoxia in a Hif-lα-mediated manner in vitro and in vivo. Dual-luciferase assay revealed that cdc25a is a novel target of miR-125c. An inverse correlation between miR-125c and cdc25a was further confirmed in vivo, suggesting miR-125c as a crucial physiological inhibitor of cdc25a which responds to cellular hypoxia. Overexpression of miR-125c suppressed cell proliferation, led to cell cycle arrest at the G1 phase in ZF4 cells and induced apoptotic responses during embryo development. More importantly, miR-125c overexpression resulted in severe malformation and reduction of motility during zebrafish embryonic development. Taken together, we conclude that miR-125c plays a pivotal role in cellular adaptations to hypoxic stress at least in part through the Hif-1α/miR-125c/cdc25a signaling and has great impact on zebrafish early embryonic development.
Collapse
|
44
|
Millar LJ, Shi L, Hoerder-Suabedissen A, Molnár Z. Neonatal Hypoxia Ischaemia: Mechanisms, Models, and Therapeutic Challenges. Front Cell Neurosci 2017; 11:78. [PMID: 28533743 PMCID: PMC5420571 DOI: 10.3389/fncel.2017.00078] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/07/2017] [Indexed: 12/11/2022] Open
Abstract
Neonatal hypoxia-ischaemia (HI) is the most common cause of death and disability in human neonates, and is often associated with persistent motor, sensory, and cognitive impairment. Improved intensive care technology has increased survival without preventing neurological disorder, increasing morbidity throughout the adult population. Early preventative or neuroprotective interventions have the potential to rescue brain development in neonates, yet only one therapeutic intervention is currently licensed for use in developed countries. Recent investigations of the transient cortical layer known as subplate, especially regarding subplate's secretory role, opens up a novel set of potential molecular modulators of neonatal HI injury. This review examines the biological mechanisms of human neonatal HI, discusses evidence for the relevance of subplate-secreted molecules to this condition, and evaluates available animal models. Neuroserpin, a neuronally released neuroprotective factor, is discussed as a case study for developing new potential pharmacological interventions for use post-ischaemic injury.
Collapse
Affiliation(s)
- Lancelot J. Millar
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Lei Shi
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan UniversityGuangzhou, China
| | | | - Zoltán Molnár
- Molnár Group, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| |
Collapse
|
45
|
Abstract
Despite the prevalence of viral infections in the American population, we still have a limited understanding of how they affect pregnancy and fetal development. Viruses can gain access to the decidua and placenta by ascending from the lower reproductive tract or via hematogenous transmission. Viral tropism for the decidua and placenta is then dependent on viral entry receptor expression in these tissues as well as on the maternal immune response to the virus. These factors vary by cell type and gestational age and can be affected by changes to the in utero environment and maternal immunity. Some viruses can directly infect the fetus at specific times during gestation, while some only infect the placenta. Both scenarios can result in severe birth defects or pregnancy loss. Systemic maternal viral infections can also affect the pregnancy, and these can be especially dangerous, because pregnant women suffer higher virus-associated morbidity and mortality than do nonpregnant counterparts. In this Review, we discuss the potential contributions of maternal, placental, and fetal viral infection to pregnancy outcome, fetal development, and maternal well-being.
Collapse
|