1
|
Gao F, Du W, Guo C, Geng P, Liu W, Jin X. α7nACh receptor, a promising target to reduce BBB damage by regulating inflammation and autophagy after ischemic stroke. Biomed Pharmacother 2024; 179:117337. [PMID: 39191022 DOI: 10.1016/j.biopha.2024.117337] [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: 05/04/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Increased blood-brain barrier (BBB) permeability can lead to cerebral vasogenic edema and hemorrhagic transformation (HT) after reperfusion with tissue plasminogen activator (tPA), the only United States Food and Drug Administration (FDA)-approved treatment for acute ischemia stroke (AIS). The therapeutic benefits of tPA after AIS are partially outweighed by a more than a six-fold increase in the risk of symptomatic intracerebral hemorrhage. Therefore, strategies to protect the integrity of BBB are urgently needed to reduce HT and vasogenic edema after tPA thrombolysis or endovascular thrombectomy. Interestingly, an NIH study showed that smokers treated with tPA had a significantly lower prevalence of brain hemorrhage than nonsmokers, suggesting that cigarette smoking may protect patients treated with tPA from the side effects of cerebral hemorrhage. Importantly, we recently showed that treatment with nicotine reduces AIS-induced BBB damage and that modulating α7nAChR by modulation could reduce ischemia/reperfusion-induced BBB damage, suggesting that α7nAChR could be a potential target to reduce BBB after AIS. In this review, we first provide an overview of stroke and the impact of α7nAChR activation on BBB damage. Next, we discuss the features and mechanism of BBB destruction after AIS. We then discuss the effect of nicotine effect on BBB integrity as well as the mechanism underlying those effects. Finally, we discuss the side effects and potential strategies for modulating α7nAChR to reduce AIS-induced BBB damage.
Collapse
Affiliation(s)
- Fengying Gao
- Department of Emergency, Shanxi Provincial People's Hospital, Taiyuan 030001, China
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Wencao Liu
- Department of Emergency, Shanxi Provincial People's Hospital, Taiyuan 030001, China.
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
2
|
Wei L, Chen S, Deng X, Liu Y, Wang H, Gao X, Huang Y. Metabolomic discoveries for early diagnosis and traditional Chinese medicine efficacy in ischemic stroke. Biomark Res 2024; 12:63. [PMID: 38902829 PMCID: PMC11188286 DOI: 10.1186/s40364-024-00608-7] [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: 03/30/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024] Open
Abstract
Ischemic stroke (IS), a devastating cerebrovascular accident, presents with high mortality and morbidity. Following IS onset, a cascade of pathological changes, including excitotoxicity, inflammatory damage, and blood-brain barrier disruption, significantly impacts prognosis. However, current clinical practices struggle with early diagnosis and identifying these alterations. Metabolomics, a powerful tool in systems biology, offers a promising avenue for uncovering early diagnostic biomarkers for IS. By analyzing dynamic metabolic profiles, metabolomics can not only aid in identifying early IS biomarkers but also evaluate Traditional Chinese Medicine (TCM) efficacy and explore its mechanisms of action in IS treatment. Animal studies demonstrate that TCM interventions modulate specific metabolite levels, potentially reflecting their therapeutic effects. Identifying relevant metabolites in cerebral ischemia patients holds immense potential for early diagnosis and improved outcomes. This review focuses on recent metabolomic discoveries of potential early diagnostic biomarkers for IS. We explore variations in metabolites observed across different ages, genders, disease severity, and stages. Additionally, the review examines how specific TCM extracts influence IS development through metabolic changes, potentially revealing their mechanisms of action. Finally, we emphasize the importance of integrating metabolomics with other omics approaches for a comprehensive understanding of IS pathophysiology and TCM efficacy, paving the way for precision medicine in IS management.
Collapse
Affiliation(s)
- Liangzhe Wei
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Siqi Chen
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China
| | - Xinpeng Deng
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Yuchun Liu
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Haifeng Wang
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Xiang Gao
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China.
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China.
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China.
| | - Yi Huang
- Department of Neurosurgery, Ningbo Hospital, Zhejiang University School of Medicine, Ningbo, 315010, China.
- Ningbo Key Laboratory of Neurological Diseases and Brain Function, Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China.
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, 315010, China.
| |
Collapse
|
3
|
Zhi S, Hu X, Ding Y, Chen H, Li X, Tao Y, Li W. An exploration on the machine-learning-based stroke prediction model. Front Neurol 2024; 15:1372431. [PMID: 38742047 PMCID: PMC11089140 DOI: 10.3389/fneur.2024.1372431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction With the rapid development of artificial intelligence technology, machine learning algorithms have been widely applied at various stages of stroke diagnosis, treatment, and prognosis, demonstrating significant potential. A correlation between stroke and cytokine levels in the human body has recently been reported. Our study aimed to establish machine-learning models based on cytokine features to enhance the decision-making capabilities of clinical physicians. Methods This study recruited 2346 stroke patients and 2128 healthy control subjects from Chongqing University Central Hospital. A predictive model was established through clinical experiments and collection of clinical laboratory tests and demographic variables at admission. Three classification algorithms, namely Random Forest, Gradient Boosting, and Support Vector Machine, were employed. The models were evaluated using methods such as ROC curves, AUC values, and calibration curves. Results Through univariate feature selection, we selected 14 features and constructed three machine-learning models: Support Vector Machine (SVM), Random Forest (RF), and Gradient Boosting Machine (GBM). Our results indicated that in the training set, the RF model outperformed the GBM and SVM models in terms of both the AUC value and sensitivity. We ranked the features using the RF algorithm, and the results showed that IL-6, IL-5, IL-10, and IL-2 had high importance scores and ranked at the top. In the test set, the stroke model demonstrated a good generalization ability, as evidenced by the ROC curve, confusion matrix, and calibration curve, confirming its reliability as a predictive model for stroke. Discussion We focused on utilizing cytokines as features to establish stroke prediction models. Analyses of the ROC curve, confusion matrix, and calibration curve of the test set demonstrated that our models exhibited a strong generalization ability, which could be applied in stroke prediction.
Collapse
Affiliation(s)
- Shenshen Zhi
- Department of Blood Transfusion, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Xiefei Hu
- Medicine School of Chongqing University, Chongqing, China
| | - Yan Ding
- Clinical Laboratory, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Huajian Chen
- Clinical Laboratory, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Xun Li
- Clinical Laboratory, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Yang Tao
- Intensive Care Unit, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Wei Li
- Clinical Laboratory, Chongqing Emergency Medical Center, Chongqing University Central Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
4
|
Shi Y, Sheng P, Guo M, Chen K, Zhao Y, Wang X, Wu M, Li B. Banxia Xiexin Decoction Prevents HT22 Cells from High Glucose-induced Neurotoxicity via JNK/SIRT1/Foxo3a Signaling Pathway. Curr Comput Aided Drug Des 2024; 20:911-927. [PMID: 37608672 DOI: 10.2174/1573409920666230822110258] [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: 04/21/2023] [Revised: 06/27/2023] [Accepted: 07/13/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND Type 2 diabetes-associated cognitive dysfunction (DCD) is a chronic complication of diabetes that has gained international attention. The medicinal compound Banxia Xiexin Decoction (BXXXD) from traditional Chinese medicine (TCM) has shown potential in improving insulin resistance, regulating endoplasmic reticulum stress (ERS), and inhibiting cell apoptosis through various pathways. However, the specific mechanism of action and medical value of BXXXD remain unclear. METHODS We utilized TCMSP databases to screen the chemical constituents of BXXXD and identified DCD disease targets through relevant databases. By using Stitch and String databases, we imported the data into Cytoscape 3.8.0 software to construct a protein-protein interaction (PPI) network and subsequently identified core targets through network topology analysis. The core targets were subjected to Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The results were further validated through in vitro experiments. RESULTS Network pharmacology analysis revealed the screening of 1490 DCD-related targets and 190 agents present in BXXXD. The topological analysis and enrichment analysis conducted using Cytoscape software identified 34 core targets. Additionally, GO and KEGG pathway analyses yielded 104 biological targets and 97 pathways, respectively. BXXXD exhibited its potential in treating DCD by controlling synaptic plasticity and conduction, suppressing apoptosis, reducing inflammation, and acting as an antioxidant. In a high glucose (HG) environment, the expression of JNK, Foxo3a, SIRT1, ATG7, Lamp2, and LC3 was downregulated. BXXXD intervention on HT22 cells potentially involved inhibiting excessive oxidative stress, promoting neuronal autophagy, and increasing the expression levels of JNK, SIRT1, Foxo3a, ATG7, Lamp2, and LC3. Furthermore, the neuroprotective effect of BXXXD was partially blocked by SP600125, while quercetin enhanced the favorable role of BXXXD in the HG environment. CONCLUSION BXXXD exerts its effects on DCD through multiple components, targets, levels, and pathways. It modulates the JNK/SIRT1/Foxo3a signaling pathway to mitigate autophagy inhibition and apoptotic damage in HT22 cells induced by HG. These findings provide valuable perspectives and concepts for future clinical trials and fundamental research.
Collapse
Affiliation(s)
- Yinli Shi
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pei Sheng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Guo
- Southeast University, Zhongda Hospital Southeast University, Nanjing, China
| | - Kai Chen
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Zhao
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xu Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mianhua Wu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
5
|
Priori EC, Ratto D, De Luca F, Sandionigi A, Savino E, Giammello F, Romeo M, Brandalise F, Roda E, Rossi P. Hericium erinaceus Extract Exerts Beneficial Effects on Gut-Neuroinflammaging-Cognitive Axis in Elderly Mice. BIOLOGY 2023; 13:18. [PMID: 38248449 PMCID: PMC10813749 DOI: 10.3390/biology13010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/23/2024]
Abstract
Ageing is a biological phenomenon that determines the impairment of cognitive performances, in particular, affecting memory. Inflammation and cellular senescence are known to be involved in the pathogenesis of cognitive decline. The gut microbiota-brain axis could exert a critical role in influencing brain homeostasis during ageing, modulating neuroinflammation, and possibly leading to inflammaging. Due to their anti-ageing properties, medicinal mushrooms can be utilised as a resource for developing pharmaceuticals and functional foods. Specifically, Hericium erinaceus (He), thanks to its bioactive metabolites, exerts numerous healthy beneficial effects, such as reinforcing the immune system, counteracting ageing, and improving cognitive performance. Our previous works demonstrated the capabilities of two months of He1 standardised extract oral supplementation in preventing cognitive decline in elderly frail mice. Herein, we showed that this treatment did not change the overall gut microbiome composition but significantly modified the relative abundance of genera specifically involved in cognition and inflammation. Parallelly, a significant decrease in crucial markers of inflammation and cellular senescence, i.e., CD45, GFAP, IL6, p62, and γH2AX, was demonstrated in the dentate gyrus and Cornus Ammonis hippocampal areas through immunohistochemical experiments. In summary, we suggested beneficial and anti-inflammatory properties of He1 in mouse hippocampus through the gut microbiome-brain axis modulation.
Collapse
Affiliation(s)
- Erica Cecilia Priori
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | - Daniela Ratto
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | - Fabrizio De Luca
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | - Anna Sandionigi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy;
- Quantia Consulting S.r.l., Via Petrarca 20, 22066 Mariano Comense, Italy
| | - Elena Savino
- Department of Earth and Environmental Science, University of Pavia, 27100 Pavia, Italy;
| | - Francesca Giammello
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | - Marcello Romeo
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | | | - Elisa Roda
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| |
Collapse
|
6
|
Lee JM, Lee JH, Kim SH, Sim TH, Kim YJ. NXP032 ameliorates cognitive impairment by alleviating the neurovascular aging process in aged mouse brain. Sci Rep 2023; 13:8594. [PMID: 37237085 DOI: 10.1038/s41598-023-35833-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/24/2023] [Indexed: 05/28/2023] Open
Abstract
Vascular aging is well known to be associated with the breakdown of the neurovascular unit (NVU), which is essential for maintaining brain homeostasis and linked to higher cognitive dysfunction. Oxidative stress is believed to be a significant cause of the vascular aging process. Vitamin C is easily oxidized under physiological conditions, so it loses its potent antioxidant activity. We developed a DNA aptamer that enhances the function of vitamin C. NXP032 is the binding form of the aptamer and vitamin C. In this study, we investigated the effect of NXP032 on neurovascular stabilization through the changes of PECAM-1, PDGFR-β, ZO-1, laminin, and glial cells involved in maintaining the integrity of the blood-brain barrier (BBB) in aged mice. NXP032 was orally administered daily for 8 weeks. Compared to young mice and NXP032-treated mice, 20-month-old mice displayed cognitive impairments in Y-maze and passive avoidance tests. NXP032 treatment contributed to reducing the BBB damage by attenuating the fragmentation of microvessels and reducing PDGFR-β, ZO-1, and laminin expression, thereby mitigating astrocytes and microglia activation during normal aging. Based on the results, we suggest that NXP032 reduces vascular aging and may be a novel intervention for aging-induced cognitive impairment.
Collapse
Affiliation(s)
- Jae-Min Lee
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Joo Hee Lee
- Korea Armed Forces Nursing Academy, Daejeon, 34059, Republic of Korea
| | - So Hee Kim
- Department of Nursing, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Tae Hyeok Sim
- Department of Nursing, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Youn-Jung Kim
- College of Nursing Science, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
7
|
Lee HJ, Yoon YS, Lee SJ. Molecular mechanisms of cellular senescence in neurodegenerative diseases. J Mol Biol 2023:168114. [PMID: 37085010 DOI: 10.1016/j.jmb.2023.168114] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/23/2023]
Abstract
Neurodegenerative diseases, such as Alzheimer's and Parkinson's, are characterized by several pathological features, including selective neuronal loss, aggregation of specific proteins, and chronic inflammation. Aging is the most critical risk factor of these disorders. However, the mechanism by which aging contributes to the pathogenesis of neurodegenerative diseases is not clearly understood. Cellular senescence is a cell state or fate in response to stimuli. It is typically associated with a series of changes in cellular phenotypes such as abnormal cellular metabolism and proteostasis, reactive oxygen species (ROS) production, and increased secretion of certain molecules via senescence-associated secretory phenotype (SASP). In this review, we discuss how cellular senescence contributes to brain aging and neurodegenerative diseases, and the relationship between protein aggregation and cellular senescence. Finally, we discuss the potential of senescence modifiers and senolytics in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- He-Jin Lee
- Department of Anatomy, Konkuk University, Seoul 05029, Korea; IBST, Konkuk University, Seoul 05029, Korea.
| | - Ye-Seul Yoon
- Department of Anatomy, Konkuk University, Seoul 05029, Korea; IBST, Konkuk University, Seoul 05029, Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, Korea; Neuramedy, Co., Ltd., Seoul, Korea.
| |
Collapse
|
8
|
Luo BL, Zhang ZZ, Chen J, Liu X, Zhang YM, Yang QG, Chen GH. Effects of gestational inflammation on age-related cognitive decline and hippocampal Gdnf-GFRα1 levels in F1 and F2 generations of CD-1 Mice. BMC Neurosci 2023; 24:26. [PMID: 37055728 PMCID: PMC10103445 DOI: 10.1186/s12868-023-00793-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/23/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND It has been reported that age-associated cognitive decline (AACD) accelerated by maternal lipopolysaccharide (LPS) insult during late pregnancy can be transmitted to the second generation in a sex-specificity manner. In turn, recent studies indicated that glial cell line-derived neurotrophic factor (GDNF) and its cognate receptor (GFRα1) are critical for normal cognitive function. Based on this evidence, we aimed to explore whether Gdnf-GFRα1 expression contributes to cognitive decline in the F1 and F2 generations of mouse dams exposed to lipopolysaccharide (LPS) during late gestation, and to evaluate also the potential interference effect of pro-inflammatory cytokines. METHODS During gestational days 15-17, pregnant CD-1 mice (8-10 weeks old) received a daily intraperitoneal injection of LPS (50 μg/kg) or saline (control). In utero LPS-exposed F1 generation mice were selectively mated to produce F2 generation mice. In F1 and F2 mice aged 3 and 15 months, the Morris water maze (MWM) was used to evaluated the spatial learning and memory ability, the western blotting and RT-PCR were used for analyses of hippocampal Gdnf and GFRα1 expression, and ELISA was used to analyse IL-1β, IL-6 and TNF-α levels in serum. RESULTS Middle-aged F1 offspring from LPS-treated mothers exhibited longer swimming latency and distance during the learning phase, lower percentage swimming time and distance in targe quadrant during memory phase, and lower hippocampal levels of Gdnf and GFRα1 gene products compared to age-matched controls. Similarly, the middle-aged F2 offspring from the Parents-LPS group had longer swimming latency and distance in the learning phase, and lower percentage swimming time and distance in memory phase than the F2-CON group. Moreover, the 3-month-old Parents-LPS and 15-month-old Parents- and Father-LPS groups had lower GDNF and GFRα1 protein and mRNAs levels compared to the age-matched F2-CON group. Furthermore, hippocampal levels of Gdnf and GFRα1 were correlated with impaired cognitive performance in the Morris water maze after controlling for circulating pro-inflammatory cytokine levels. CONCLUSIONS Our findings indicate that accelerated AACD by maternal LPS exposure can be transmitted across at least two generations through declined Gdnf and GFRα1 expression, mainly via paternal linage.
Collapse
Affiliation(s)
- Bao-Ling Luo
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Zhe-Zhe Zhang
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Jing Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Xue Liu
- Department of Geriatrics, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, People's Republic of China
| | - Yue-Ming Zhang
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China
| | - Qi-Gang Yang
- Department of Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, People's Republic of China.
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, 238000, Anhui, People's Republic of China.
| |
Collapse
|
9
|
Kim H, Noh M, Zhang H, Kim Y, Park S, Park J, Kwon YG. Long-term administration of CU06-1004 ameliorates cerebrovascular aging and BBB injury in aging mouse model. Fluids Barriers CNS 2023; 20:9. [PMID: 36726154 PMCID: PMC9893613 DOI: 10.1186/s12987-023-00410-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Age-related changes in the cerebrovasculature, including blood-brain barrier (BBB) disruption and vascular dementia, are emerging as potential risks for many neurodegenerative diseases. Therefore, the endothelial cells that constitute the cerebrovasculature may play key roles in preventing brain injury. Our previous study showed that CU06-1004, an endothelial cell dysfunction blocker, prevented vascular leakage, enhanced vascular integrity in ischemic reperfusion injury, and promoted the normalization of tumor vasculature. Here, we evaluated the effects of CU06-1004 on age-related cerebrovascular functional decline in the aged mouse brain. RESULTS In this study, we investigated the protective effects of CU06-1004 against oxidative stress-induced damage in human brain microvascular endothelial cells (HBMECs). HBMECs were treated with hydrogen peroxide (H2O2) to establish an oxidative stress-induced model of cellular injury. Compared with H2O2 treatment alone, pretreatment of HBMECs with CU06-1004 considerably reduced oxidative stress-induced cytotoxicity, reactive oxygen species generation, senescence-associated β-galactosidase activity, senescence marker expression, and the expression levels of inflammatory proteins. Based on the observed cytoprotective effects of CU06-1004 in HBMECs, we examined whether CU06-1004 displayed protective effects against cerebrovascular aging in mice. Long-term administration of CU06-1004 alleviated age-associated cerebral microvascular rarefaction and cerebrovascular senescence in the aged mouse brain. CU06-1004 supplementation also reduced the extravasation of plasma IgG by improving BBB integrity in the aged mouse brain, associated with reductions in neuronal injury. A series of behavioral tests also revealed improved motor and cognitive functions in aged mice that received long-term CU06-1004 administration. CONCLUSIONS These findings suggest that CU06-1004 may represent a promising therapeutic approach for delaying age-related cerebrovascular impairment and improving cognitive function in old age.
Collapse
Affiliation(s)
- Hyejeong Kim
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Korea
| | - Minyoung Noh
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Korea
| | | | - Yeomyeong Kim
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Korea
| | - Songyi Park
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Korea
| | - Jeongeun Park
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Korea
| | - Young-Guen Kwon
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Korea
| |
Collapse
|
10
|
Gu S, Zhou Z, Zhang S, Cai Y. Advances in Anti-Diabetic Cognitive Dysfunction Effect of Erigeron Breviscapus (Vaniot) Hand-Mazz. Pharmaceuticals (Basel) 2022; 16:ph16010050. [PMID: 36678547 PMCID: PMC9867432 DOI: 10.3390/ph16010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
Diabetic cognitive dysfunction (DCD) is the decline in memory, learning, and executive function caused by diabetes. Although its pathogenesis is unclear, molecular biologists have proposed various hypotheses, including insulin resistance, amyloid β hypothesis, tau protein hyperphosphorylation hypothesis, oxidative stress and neuroinflammation. DCD patients have no particular treatment options and current pharmacological regimens are suboptimal. In recent years, Chinese medicine research has shown that herbs with multi-component, multi-pathway and multi-target synergistic activities can prevent and treat DCD. Yunnan is home to the medicinal herb Erigeron breviscapus (Vant.) Hand-Mazz. (EBHM). Studies have shown that EBHM and its active components have a wide range of pharmacological effects and applications in cognitive disorders. EBHM's anti-DCD properties have been seldom reviewed. Through a literature study, we were able to evaluate the likely pathophysiology of DCD, prescribe anti-DCD medication and better grasp EBHM's therapeutic potential. EBHM's pharmacological mechanism and active components for DCD treatment were also summarized.
Collapse
Affiliation(s)
- Shanye Gu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ziyi Zhou
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Guangzhou 510120, China
| | - Shijie Zhang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Guangzhou 510120, China
| | - Yefeng Cai
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Guangzhou 510120, China
- Correspondence: ; Tel.: +86-136-3133-3842
| |
Collapse
|
11
|
Lymphocyte-to-Monocyte Ratio Is Independently Associated with Progressive Infarction in Patients with Acute Ischemic Stroke. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2290524. [PMID: 36605104 PMCID: PMC9810397 DOI: 10.1155/2022/2290524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/28/2022]
Abstract
Methods From April 2017 to December 2020, we retrospectively recruited 477 patients with acute ischemic stroke (within 48 hours after onset). Progressive infarction was defined as an increase of ≥1 point in motor power or ≥2 points on the total National Institutes of Health Stroke Scale (NIHSS) within 7 days after admission and extension of the original infarction were further confirmed by diffusion-weighted imaging. Demographic characteristics, clinical information, and neuroimaging characteristics were evaluated after admission. All blood draws and initial imaging were completed within 24 hours of admission. Results PI occurred in 147 (30.8%) patients. Univariate analysis comparing the two groups revealed that hypertension, initial NIHSS score, discharge NIHSS score, modified Rankin scale score at 90 days, monocyte level, creatinine level, fasting glucose level, LMR, monocyte-to-high-density lipoprotein ratio (MHR), and lesion location were significantly different (P < 0.05). Multivariate logistic regression analysis showed that the odds ratio of PI increased as the quartile of LMR increased, with the lowest quartile as the reference value. Subgroup analyses showed that a high LMR was an independent predictor of PI only in large artery atherosclerosis (LAA) patients. The receiver operating characteristic (ROC) curve was drawn to estimate the predictive value of LMR for PI. For all cases, the area under the curve was 0.583 (95% CI 0.526-0.641), and the best predictive cutoff value was 3.506, with a sensitivity of 53.1% and a specificity of 63.9%. In patients with LAA, the area under the curve was 0.585 (95% CI 0.505-0.665), and the best predictive cutoff value was 3.944, with a sensitivity of 48.7% and a specificity of 72.8%. Conclusions LMR was an independent predictor for progressive infarction in patients with acute ischemic stroke, especially in LAA cerebral infarction patients.
Collapse
|
12
|
4-Hydroxynonenal Modulates Blood-Brain Barrier Permeability In Vitro through Changes in Lipid Composition and Oxidative Status of Endothelial Cells and Astrocytes. Int J Mol Sci 2022; 23:ijms232214373. [PMID: 36430852 PMCID: PMC9698020 DOI: 10.3390/ijms232214373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Blood brain barrier (BBB) is a dynamic interface responsible for proper functioning of brain, but also a major obstacle for effective treatment of neurological diseases. Increased levels of free radicals, in high ferrous and high lipid content surrounding, induce lipid peroxidation, leading to production of 4-hydroxynonenal (HNE). HNE modifies all key proteins responsible for proper brain functioning thus playing a major role in the onset of neurological diseases. To investigate HNE effects on BBB permeability, we developed two in vitro BBB models-'physiological' and 'pathological'. The latter mimicked HNE modified extracellular matrix under oxidative stress conditions in brain pathologies. We showed that exogenous HNE induce activation of antioxidative defense systems by increasing catalase activity and glutathione content as well as reducing lipid peroxide levels in endothelial cells and astrocytes of 'physiological' model. While in 'pathological' model, exogenous HNE further increased lipid peroxidation levels of endothelial cells and astrocytes, followed by increase in Nrf2 and glutathione levels in endothelial cells. At lipid composition level, HNE caused increase in ω3 polyunsaturated fatty acid (PUFA) level in endothelial cells, followed by decrease in ω3 PUFA level and increase in monounsaturated fatty acid level in astrocytes. Using these models, we showed for the first time that HNE in 'pathological' model can reduce BBB permeability.
Collapse
|
13
|
Adesse D, Gladulich L, Alvarez-Rosa L, Siqueira M, Marcos AC, Heider M, Motta CS, Torices S, Toborek M, Stipursky J. Role of aging in Blood-Brain Barrier dysfunction and susceptibility to SARS-CoV-2 infection: impacts on neurological symptoms of COVID-19. Fluids Barriers CNS 2022; 19:63. [PMID: 35982454 PMCID: PMC9386676 DOI: 10.1186/s12987-022-00357-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 07/18/2022] [Indexed: 12/21/2022] Open
Abstract
COVID-19, which is caused by Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2), has resulted in devastating morbidity and mortality worldwide due to lethal pneumonia and respiratory distress. In addition, the central nervous system (CNS) is well documented to be a target of SARS-CoV-2, and studies detected SARS-CoV-2 in the brain and the cerebrospinal fluid of COVID-19 patients. The blood-brain barrier (BBB) was suggested to be the major route of SARS-CoV-2 infection of the brain. Functionally, the BBB is created by an interactome between endothelial cells, pericytes, astrocytes, microglia, and neurons, which form the neurovascular units (NVU). However, at present, the interactions of SARS-CoV-2 with the NVU and the outcomes of this process are largely unknown. Moreover, age was described as one of the most prominent risk factors for hospitalization and deaths, along with other comorbidities such as diabetes and co-infections. This review will discuss the impact of SARS-CoV-2 on the NVU, the expression profile of SARS-CoV-2 receptors in the different cell types of the CNS and the possible role of aging in the neurological outcomes of COVID-19. A special emphasis will be placed on mitochondrial functions because dysfunctional mitochondria are also a strong inducer of inflammatory reactions and the "cytokine storm" associated with SARS-CoV-2 infection. Finally, we will discuss possible drug therapies to treat neural endothelial function in aged patients, and, thus, alleviate the neurological symptoms associated with COVID-19.
Collapse
Affiliation(s)
- Daniel Adesse
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil.
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| | - Luis Gladulich
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Liandra Alvarez-Rosa
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michele Siqueira
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anne Caroline Marcos
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Marialice Heider
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Caroline Soares Motta
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, Pavilhão Carlos Chagas, sala 307b, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| | - Joice Stipursky
- Laboratório Compartilhado, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Xia L, Xu T, Zhan Z, Wu Y, Xu Y, Cao Y, Han Z. High ratio of monocytes to high-density lipoprotein is associated with hemorrhagic transformation in acute ischemic stroke patients on intravenous thrombolysis. Front Aging Neurosci 2022; 14:977332. [PMID: 36051305 PMCID: PMC9424860 DOI: 10.3389/fnagi.2022.977332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundHemorrhagic transformation (HT) is a frequent, serious complication in acute ischemic stroke patients on intravenous thrombolysis. Here we investigated whether risk of HT is associated with the ratio of monocyte count to high-density lipoprotein level (MHR).Materials and methodsMedical records were retrospectively examined for consecutive patients with acute ischemic stroke who received thrombolytic therapy. HT was diagnosed by computed tomography at 24–36 h after therapy. Potential association between MHR and HT was examined using logistic regression.ResultsA total of 340 patients were analyzed, and their median MHR was 0.44 (0.31–0.59). MHR was higher in the 51 patients (15.0%) with HT than in those who did not suffer HT (0.53 vs. 0.42, P = 0.001). Multivariate logistic regression showed that, after adjusting for potential confounders, MHR was an independent risk factor for HT (OR 7.50, 95% CI 1.64 to 34.35, P = 0.009). Risk of HT was significantly higher among patients whose MHR fell in the third quartile (0.42–0.53) and the fourth quartile (> 0.53) than among those with MHR in the first quartile (< 0.31; OR 3.53, 95% CI 1.11 to 11.20, P = 0.032; OR 4.79, 95% CI 1.49 to 15.42, P = 0.009).ConclusionHigh MHR may be independently associated with higher risk of HT in patients with acute ischemic stroke on intravenous thrombolysis.
Collapse
|
15
|
Sifat AE, Nozohouri S, Archie SR, Chowdhury EA, Abbruscato TJ. Brain Energy Metabolism in Ischemic Stroke: Effects of Smoking and Diabetes. Int J Mol Sci 2022; 23:ijms23158512. [PMID: 35955647 PMCID: PMC9369264 DOI: 10.3390/ijms23158512] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/06/2023] Open
Abstract
Proper regulation of energy metabolism in the brain is crucial for maintaining brain activity in physiological and different pathophysiological conditions. Ischemic stroke has a complex pathophysiology which includes perturbations in the brain energy metabolism processes which can contribute to worsening of brain injury and stroke outcome. Smoking and diabetes are common risk factors and comorbid conditions for ischemic stroke which have also been associated with disruptions in brain energy metabolism. Simultaneous presence of these conditions may further alter energy metabolism in the brain leading to a poor clinical prognosis after an ischemic stroke event. In this review, we discuss the possible effects of smoking and/or diabetes on brain glucose utilization and mitochondrial energy metabolism which, when present concurrently, may exacerbate energy metabolism in the ischemic brain. More research is needed to investigate brain glucose utilization and mitochondrial oxidative metabolism in ischemic stroke in the presence of smoking and/or diabetes, which would provide further insights on the pathophysiology of these comorbid conditions and facilitate the development of therapeutic interventions.
Collapse
|
16
|
Satyam SM, Bairy LK. Neuronutraceuticals Combating Neuroinflammaging: Molecular Insights and Translational Challenges—A Systematic Review. Nutrients 2022; 14:nu14153029. [PMID: 35893883 PMCID: PMC9330442 DOI: 10.3390/nu14153029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
Neuropathologies, such as neuroinflammaging, have arisen as a serious concern for preserving the quality of life due to the global increase in neurodegenerative illnesses. Nowadays, neuronutraceuticals have gained remarkable attention. It is necessary to investigate the bioavailability, off-target effects, and mechanism of action of neuronutraceuticals. To comprehend the comprehensive impact on brain health, well-designed randomized controlled trials testing combinations of neuronutraceuticals are also necessary. Although there is a translational gap between basic and clinical research, the present knowledge of the molecular perspectives of neuroinflammaging and neuronutraceuticals may be able to slow down brain aging and to enhance cognitive performance. The present review also highlights the key emergent issues, such as regulatory and scientific concerns of neuronutraceuticals, including bioavailability, formulation, blood–brain permeability, safety, and efficacy.
Collapse
|
17
|
Groth M, Skrzydlewska E, Dobrzyńska M, Pancewicz S, Moniuszko-Malinowska A. Redox Imbalance and Its Metabolic Consequences in Tick-Borne Diseases. Front Cell Infect Microbiol 2022; 12:870398. [PMID: 35937690 PMCID: PMC9353526 DOI: 10.3389/fcimb.2022.870398] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022] Open
Abstract
One of the growing global health problems are vector-borne diseases, including tick-borne diseases. The most common tick-borne diseases include Lyme disease, tick-borne encephalitis, human granulocytic anaplasmosis, and babesiosis. Taking into account the metabolic effects in the patient's body, tick-borne diseases are a significant problem from an epidemiological and clinical point of view. Inflammation and oxidative stress are key elements in the pathogenesis of infectious diseases, including tick-borne diseases. In consequence, this leads to oxidative modifications of the structure and function of phospholipids and proteins and results in qualitative and quantitative changes at the level of lipid mediators arising in both reactive oxygen species (ROS) and ROS enzyme-dependent reactions. These types of metabolic modifications affect the functioning of the cells and the host organism. Therefore, links between the severity of the disease state and redox imbalance and the level of phospholipid metabolites are being searched, hoping to find unambiguous diagnostic biomarkers. Assessment of molecular effects of oxidative stress may also enable the monitoring of the disease process and treatment efficacy.
Collapse
Affiliation(s)
- Monika Groth
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Inorganic and Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Marta Dobrzyńska
- Department of Inorganic and Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Sławomir Pancewicz
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
18
|
Khilazheva ED, Lychkovskaya EV, Kutyakov VA, Morgun AV, Salmin VV. In vitro Effects of Plasma Acid on Proliferation of Rat Brain Endothelial Cells. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022040196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
The amyloid peptide β disrupts intercellular junctions and increases endothelial permeability in a NADPH oxidase 1-dependent manner. Redox Biol 2022; 52:102287. [PMID: 35358850 PMCID: PMC8966210 DOI: 10.1016/j.redox.2022.102287] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/12/2022] [Accepted: 03/12/2022] [Indexed: 11/20/2022] Open
Abstract
Alzheimer's disease is the most common form of dementia and is associated with the accumulation of amyloid peptide β in the brain parenchyma. Vascular damage and microvascular thrombosis contribute to the neuronal degeneration and the loss of brain function typical of this disease. In this study, we utilised a murine model of Alzheimer's disease to evaluate the neurovascular effects of this disease. Upon detection of an increase in the phosphorylation of the endothelial surface receptor VE-cadherin, we focused our attention on endothelial cells and utilised two types of human endothelial cells cultured in vitro: 1) human umbilical vein endothelial cells (HUVECs) and 2) human brain microvascular endothelial cells (hBMECs). Using an electrical current impedance system (ECIS) and FITC-albumin permeability assays, we discovered that the treatment of human endothelial cells with amyloid peptide β causes a loss in their barrier function, which is oxidative stress-dependent and similarly to our observation in mouse brain associates with VE-cadherin phosphorylation. The activation of the superoxide anion-generating enzyme NADPH oxidase 1 is responsible for the oxidative stress that leads to the disruption of barrier function in human endothelial cells in vitro. In summary, we have identified a novel molecular mechanism explaining how the accumulation of amyloid peptide β in the brain parenchyma may induce the loss of neurovascular barrier function, which has been observed in patients. Neurovascular leakiness plays an important role in brain inflammation and neuronal degeneration driving the progression of the Alzheimer's disease. Therefore, this study provides a novel and promising target for the development of a pharmacological treatment to protect neurovascular function and reduce the progression of the neurodegeneration in Alzheimer's patients. Amyloid peptide β induces oxidative changes in mouse hippocampus. The endothelial barrier function is impaired by amyloid peptide β. Oxidative stress is critical for the increase in endothelial monolayer permeability. NADPH oxidase 1 mediates the endothelial barrier damage caused by amyloid peptide β.
Collapse
|
20
|
Yuan W, Xia H, Xu Y, Xu C, Chen N, Shao C, Dai Z, Chen R, Tao A. The role of ferroptosis in endothelial cell dysfunction. Cell Cycle 2022; 21:1897-1914. [PMID: 35579940 DOI: 10.1080/15384101.2022.2079054] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Ferroptosis is a form of iron-dependent cell death caused by an excessive accumulation of reactive oxygen species and lipid peroxidation. The importance of ferroptosis in the occurrence and progression of various diseases is gradually being recognized; however, the exact biological effects and potential mechanisms of endothelial cell ferroptosis remain unclear. The endothelium forms the innermost layer of the blood vessels and lymphatic vessels. It acts as an important functional interface, responds to various pathological stimuli and causes endothelial dysfunction. Here, we review recent findings to elucidate the role of ferroptosis in endothelial cells under different pathophysiologic settings.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hao Xia
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yao Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chong Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Nan Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhiyin Dai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Rui Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Aibin Tao
- Department of Cardiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
21
|
Sung KY, Zhang B, Wang HE, Bai YM, Tsai SJ, Su TP, Chen TJ, Hou MC, Lu CL, Wang YP, Chen MH. Schizophrenia and risk of new-onset inflammatory bowel disease: a nationwide longitudinal study. Aliment Pharmacol Ther 2022; 55:1192-1201. [PMID: 35261051 DOI: 10.1111/apt.16856] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/16/2021] [Accepted: 02/16/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Inflammatory bowel disease (IBD) is a chronic gastrointestinal inflammatory disorder with increasing global prevalence. The risk of IBD in patients with schizophrenia remains unclear. We aim to investigate the risk of new-onset IBD in patients with schizophrenia compared with matched controls. METHODS We conducted a retrospective, population-based cohort study utilising patient data from the Taiwan National Health Insurance Research Database collected between January 1, 2001, and December 31, 2011. Patients diagnosed with schizophrenia by board-certified psychiatrists without prior diagnosis of IBD were enrolled and matched to controls in 1:4 fashion by age, sex, residence, income level and medical comorbidities. Adjusted hazard ratios (HRs) for new-onset IBD and sub-analyses were determined using Cox regression analysis with adjustments. RESULTS Among 116 164 patients with schizophrenia and 464 656 matched controls, overall incidence of IBD among patients was significantly higher (1.14% vs. 0.25%). Average age of IBD diagnosis was 46.82 among patients with schizophrenia, versus 55.30 among controls. The HR of developing IBD among patients was 3.28, with a 95% confidence interval (95% CI) 2.49-4.33. IBD risk was higher among patients with psychiatric admissions more than once per year (HR 7.99, 95% CI 5.25-12.15) compared to those hospitalised less frequently (HR 2.72, 95% CI 2.03-3.66). CONCLUSIONS This population-based cohort study demonstrates a significant association between schizophrenia and subsequent IBD development. Patients with schizophrenia develop IBD at a younger age, and the risk increases with inadequately controlled schizophrenia. Physician vigilance and awareness of this correlation will improve IBD diagnosis and management among this vulnerable patient population.
Collapse
Affiliation(s)
- Kuan-Yi Sung
- Endoscopy Center for Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Bing Zhang
- Department of Medicine, Division of Gastrointestinal and Liver Disease, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hohui E Wang
- Department of Psychiatry, University of California San Francisco, San Francisco, California, USA
| | - Ya-Mei Bai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tung-Ping Su
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Departments of Psychiatry and Medicine, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Tzeng-Ji Chen
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Hospital and Health Care Administration, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Chih Hou
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ching-Liang Lu
- Endoscopy Center for Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Po Wang
- Endoscopy Center for Diagnosis and Treatment, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Brain Science, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
22
|
Archie SR, Sharma S, Burks E, Abbruscato T. Biological determinants impact the neurovascular toxicity of nicotine and tobacco smoke: A pharmacokinetic and pharmacodynamics perspective. Neurotoxicology 2022; 89:140-160. [PMID: 35150755 PMCID: PMC8958572 DOI: 10.1016/j.neuro.2022.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/30/2022] [Accepted: 02/05/2022] [Indexed: 01/01/2023]
Abstract
Accumulating evidence suggests that the detrimental effect of nicotine and tobacco smoke on the central nervous system (CNS) is caused by the neurotoxic role of nicotine on blood-brain barrier (BBB) permeability, nicotinic acetylcholine receptor expression, and the dopaminergic system. The ultimate consequence of these nicotine associated neurotoxicities can lead to cerebrovascular dysfunction, altered behavioral outcomes (hyperactivity and cognitive dysfunction) as well as future drug abuse and addiction. The severity of these detrimental effects can be associated with several biological determinants. Sex and age are two important biological determinants which can affect the pharmacokinetics and pharmacodynamics of several systemically available substances, including nicotine. With regard to sex, the availability of gonadal hormone is impacted by the pregnancy status and menstrual cycle resulting in altered metabolism rate of nicotine. Additionally, the observed lower smoking cessation rate in females compared to males is a consequence of differential effects of sex on pharmacokinetics and pharmacodynamics of nicotine. Similarly, age-dependent alterations in the pharmacokinetics and pharmacodynamics of nicotine have also been observed. One such example is related to severe vulnerability of adolescence towards addiction and long-term behavioral changes which may continue through adulthood. Considering the possible neurotoxic effects of nicotine on the central nervous system and the deterministic role of sex as well as age on these neurotoxic effects of smoking, it has become important to consider sex and age to study nicotine induced neurotoxicity and development of treatment strategies for combating possible harmful effects of nicotine. In the future, understanding the role of sex and age on the neurotoxic actions of nicotine can facilitate the individualization and optimization of treatment(s) to mitigate nicotine induced neurotoxicity as well as smoking cessation therapy. Unfortunately, however, no such comprehensive study is available which has considered both the sex- and age-dependent neurotoxicity of nicotine, as of today. Hence, the overreaching goal of this review article is to analyze and summarize the impact of sex and age on pharmacokinetics and pharmacodynamics of nicotine and possible neurotoxic consequences associated with nicotine in order to emphasize the importance of including these biological factors for such studies.
Collapse
Affiliation(s)
- Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Elizabeth Burks
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA
| | - Thomas Abbruscato
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX, USA.
| |
Collapse
|
23
|
van Vliet EA, Marchi N. Neurovascular unit dysfunction as a mechanism of seizures and epilepsy during aging. Epilepsia 2022; 63:1297-1313. [PMID: 35218208 PMCID: PMC9321014 DOI: 10.1111/epi.17210] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022]
Abstract
The term neurovascular unit (NVU) describes the structural and functional liaison between specialized brain endothelium, glial and mural cells, and neurons. Within the NVU, the blood‐brain barrier (BBB) is the microvascular structure regulating neuronal physiology and immune cross‐talk, and its properties adapt to brain aging. Here, we analyze a research framework where NVU dysfunction, caused by acute insults or disease progression in the aging brain, represents a converging mechanism underlying late‐onset seizures or epilepsy and neurological or neurodegenerative sequelae. Furthermore, seizure activity may accelerate brain aging by sustaining regional NVU dysfunction, and a cerebrovascular pathology may link seizures to comorbidities. Next, we focus on NVU diagnostic approaches that could be tailored to seizure conditions in the elderly. We also examine the impending disease‐modifying strategies based on the restoration of the NVU and, more in general, the homeostatic control of anti‐ and pro‐inflammatory players. We conclude with an outlook on current pre‐clinical knowledge gaps and clinical challenges pertinent to seizure onset and conditions in an aging population.
Collapse
Affiliation(s)
- Erwin A van Vliet
- Amsterdam UMC, University of Amsterdam, dept. of (Neuro)pathology, Amsterdam, the Netherlands.,University of Amsterdam, Swammerdam Institute for Life Sciences, Center for Neuroscience, Amsterdam, the Netherlands
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
24
|
Harati R, Hammad S, Tlili A, Mahfood M, Mabondzo A, Hamoudi R. miR-27a-3p regulates expression of intercellular junctions at the brain endothelium and controls the endothelial barrier permeability. PLoS One 2022; 17:e0262152. [PMID: 35025943 PMCID: PMC8758013 DOI: 10.1371/journal.pone.0262152] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/17/2021] [Indexed: 01/08/2023] Open
Abstract
Background The brain endothelial barrier permeability is governed by tight and adherens junction protein complexes that restrict paracellular permeability at the blood-brain barrier (BBB). Dysfunction of the inter-endothelial junctions has been implicated in neurological disorders such as multiple sclerosis, stroke and Alzheimer’s disease. The molecular mechanisms underlying junctional dysfunction during BBB impairment remain elusive. MicroRNAs (miRNAs) have emerged as versatile regulators of the BBB function under physiological and pathological conditions, and altered levels of BBB-associated microRNAs were demonstrated in a number of brain pathologies including neurodegeneration and neuroinflammatory diseases. Among the altered micro-RNAs, miR-27a-3p was found to be downregulated in a number of neurological diseases characterized by loss of inter-endothelial junctions and disruption of the barrier integrity. However, the relationship between miR-27a-3p and tight and adherens junctions at the brain endothelium remains unexplored. Whether miR-27a-3p is involved in regulation of the junctions at the brain endothelium remains to be determined. Methods Using a gain-and-loss of function approach, we modulated levels of miR-27a-3p in an in-vitro model of the brain endothelium, key component of the BBB, and examined the resultant effect on the barrier paracellular permeability and on the expression of essential tight and adherens junctions. The mechanisms governing the regulation of junctional proteins by miR-27a-3p were also explored. Results Our results showed that miR-27a-3p inhibitor increases the barrier permeability and causes reduction of claudin-5 and occludin, two proteins highly enriched at the tight junction, while miR-27a-3p mimic reduced the paracellular leakage and increased claudin-5 and occludin protein levels. Interestingly, we found that miR-27-3p induces expression of claudin-5 and occludin by downregulating Glycogen Synthase Kinase 3 beta (GSK3ß) and activating Wnt/ß-catenin signaling, a key pathway required for the BBB maintenance. Conclusion For the first time, we showed that miR-27a-3p is a positive regulator of key tight junction proteins, claudin-5 and occludin, at the brain endothelium through targeting GSK3ß gene and activating Wnt/ß-catenin signaling. Thus, miR-27a-3p may constitute a novel therapeutic target that could be exploited to prevent BBB dysfunction and preserves its integrity in neurological disorders characterized by impairment of the barrier’s function.
Collapse
Affiliation(s)
- Rania Harati
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Saba Hammad
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mona Mahfood
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Aloïse Mabondzo
- Department of Medicines and Healthcare Technologies, Paris-Saclay University, The French Alternative Energies and Atomic Energy Commission, Gif-sur-Yvette, France
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Division of Surgery and Interventional Science, University College London, London, United Kingdom
| |
Collapse
|
25
|
Scarapicchia V, MacDonald S, Gawryluk JR. The relationship between cardiovascular risk and lifestyle activities on hippocampal volumes in normative aging. AGING BRAIN 2022; 2:100033. [PMID: 36908897 PMCID: PMC9999441 DOI: 10.1016/j.nbas.2022.100033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 01/17/2022] [Accepted: 01/31/2022] [Indexed: 10/19/2022] Open
Abstract
Background Despite the life-course perspective of popular aging models, few studies on healthy aging to date have examined both younger and older adulthood. The current study examined how cumulative vascular risk factors and self-reported levels of physical, social, and cognitive activity are associated with differences in hippocampal volumes in healthy younger and older adults. Methods 34 neurologically healthy participants were separated into two age cohorts: a younger adult group (age 25-35, n = 17) and an older adult group (age 65-82, n = 17). Participants underwent a 3 T T1 MRI and completed a series of questionnaires. Voxel-based morphometry examined whole-brain grey matter density differences between groups. Hippocampal volumes were computed. Analyses examined the association between hippocampal volumes, cumulative vascular risk, and self-reported levels of physical, social, and cognitive activity, both within and across groups. Results Between-group comparisons revealed greater cortical atrophy in older relative to young adults in regions including the left and right hippocampus and temporal fusiform cortex. Across-group analyses revealed a significant negative association between cardiovascular risk scores and bilateral hippocampal volumes across age groups. A significant negative association was identified between frequency of social activities and bilateral hippocampal volumes in older adults only. No significant associations were found between left or right hippocampal volumes and total, cognitive, or physical activities in both within- and across-group analyses. Conclusion Greater cumulative vascular risk is associated with smaller hippocampal volumes across age cohorts. Findings suggest that social activities with low cognitive load may not be beneficial to structural brain outcomes in older age.
Collapse
Affiliation(s)
- Vanessa Scarapicchia
- Department of Psychology, University of Victoria, Victoria, British Columbia, Canada.,Institute on Aging and Lifelong Health, University of Victoria, British Columbia, Canada
| | - Stuart MacDonald
- Department of Psychology, University of Victoria, Victoria, British Columbia, Canada.,Institute on Aging and Lifelong Health, University of Victoria, British Columbia, Canada
| | - Jodie R Gawryluk
- Department of Psychology, University of Victoria, Victoria, British Columbia, Canada.,Institute on Aging and Lifelong Health, University of Victoria, British Columbia, Canada.,Division of Medical Sciences, University of Victoria, British Columbia, Canada
| |
Collapse
|
26
|
Mhatre SD, Iyer J, Puukila S, Paul AM, Tahimic CGT, Rubinstein L, Lowe M, Alwood JS, Sowa MB, Bhattacharya S, Globus RK, Ronca AE. Neuro-consequences of the spaceflight environment. Neurosci Biobehav Rev 2021; 132:908-935. [PMID: 34767877 DOI: 10.1016/j.neubiorev.2021.09.055] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 08/03/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022]
Abstract
As human space exploration advances to establish a permanent presence beyond the Low Earth Orbit (LEO) with NASA's Artemis mission, researchers are striving to understand and address the health challenges of living and working in the spaceflight environment. Exposure to ionizing radiation, microgravity, isolation and other spaceflight hazards pose significant risks to astronauts. Determining neurobiological and neurobehavioral responses, understanding physiological responses under Central Nervous System (CNS) control, and identifying putative mechanisms to inform countermeasure development are critically important to ensuring brain and behavioral health of crew on long duration missions. Here we provide a detailed and comprehensive review of the effects of spaceflight and of ground-based spaceflight analogs, including simulated weightlessness, social isolation, and ionizing radiation on humans and animals. Further, we discuss dietary and non-dietary countermeasures including artificial gravity and antioxidants, among others. Significant future work is needed to ensure that neural, sensorimotor, cognitive and other physiological functions are maintained during extended deep space missions to avoid potentially catastrophic health and safety outcomes.
Collapse
Affiliation(s)
- Siddhita D Mhatre
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; KBR, Houston, TX, 77002, USA; COSMIAC Research Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Janani Iyer
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA
| | - Stephanie Puukila
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA; Flinders University, Adelaide, Australia
| | - Amber M Paul
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; KBR, Houston, TX, 77002, USA; Department of Biology, University of North Florida, Jacksonville, FL, 32224, USA
| | - Linda Rubinstein
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Universities Space Research Association, Columbia, MD, 21046, USA
| | - Moniece Lowe
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Blue Marble Space Institute of Science, Seattle, WA, 98154, USA
| | - Joshua S Alwood
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Marianne B Sowa
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Sharmila Bhattacharya
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - April E Ronca
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA; Wake Forest Medical School, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
27
|
Li W, Yuan W, Zhang D, Cai S, Luo J, Zeng K. LCZ696 Possesses a Protective Effect Against Homocysteine (Hcy)-Induced Impairment of Blood-Brain Barrier (BBB) Integrity by Increasing Occludin, Mediated by the Inhibition of Egr-1. Neurotox Res 2021; 39:1981-1990. [PMID: 34542838 DOI: 10.1007/s12640-021-00414-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/24/2021] [Accepted: 09/03/2021] [Indexed: 12/14/2022]
Abstract
Homocysteine (Hcy) is a non-essential amino acid produced from methionine. It has been reported that high concentrations of Hcy are related to the pathogenesis of neurodegenerative diseases and induce the disruption of the blood-brain barrier (BBB) by triggering oxidative stress and inflammation. LCZ696 is a novel antihypertensive agent that has been recently reported to possess promising anti-inflammatory properties. However, whether it has a protective effect on the BBB disruption is still unknown. For the first time, in this study, we aim to investigate whether LCZ696 exerts anti-inflammatory effects on Hcy-induced injury in brain endothelial cells and explore its neuroprotective properties. In in vivo experiments, we found that treatment with LCZ696 ameliorated oxidative stress by reducing malondialdehyde (MDA) and increasing glutathione (GSH). Furthermore, LCZ696 downregulated the excessive release of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) at mRNA and protein levels. Importantly, it reversed the disruption of the BBB induced by Hcy stimulation. In the in vitro human brain microvascular endothelial cell (HBMVEC) experiments, compared to the control, the permeability of the endothelial monolayer was significantly enlarged, the expression level of occludin declined, and Egr-1 upregulated by the introduction of Hcy, and these were all reversed by the treatment with LCZ696. Lastly, we found that the protective effects of LCZ696 against Hcy-induced reduction of occludin and hyper-permeability of the endothelial monolayer were greatly abolished by the overexpression of Egr-1. Taken together, we found that LCZ696 protected against Hcy-induced impairment of BBB integrity by increasing the expression of occludin, all mediated by the inhibition of Egr-1.
Collapse
Affiliation(s)
- Wenfeng Li
- Department of Cardiology, The First Affiliated Hospital of Ji'nan University, Guangzhou, 510630, Guangdong, China.,Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Wenjin Yuan
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Dandan Zhang
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Shuchun Cai
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Jun Luo
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Kanghua Zeng
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
28
|
Elmaleh DR, Downey MA, Kundakovic L, Wilkinson JE, Neeman Z, Segal E. New Approaches to Profile the Microbiome for Treatment of Neurodegenerative Disease. J Alzheimers Dis 2021; 82:1373-1401. [PMID: 34219718 DOI: 10.3233/jad-210198] [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] [Indexed: 02/06/2023]
Abstract
Progressive neurodegenerative diseases represent some of the largest growing treatment challenges for public health in modern society. These diseases mainly progress due to aging and are driven by microglial surveillance and activation in response to changes occurring in the aging brain. The lack of efficacious treatment options for Alzheimer's disease (AD), as the focus of this review, and other neurodegenerative disorders has encouraged new approaches to address neuroinflammation for potential treatments. Here we will focus on the increasing evidence that dysbiosis of the gut microbiome is characterized by inflammation that may carry over to the central nervous system and into the brain. Neuroinflammation is the common thread associated with neurodegenerative diseases, but it is yet unknown at what point and how innate immune function turns pathogenic for an individual. This review will address extensive efforts to identify constituents of the gut microbiome and their neuroactive metabolites as a peripheral path to treatment. This approach is still in its infancy in substantive clinical trials and requires thorough human studies to elucidate the metabolic microbiome profile to design appropriate treatment strategies for early stages of neurodegenerative disease. We view that in order to address neurodegenerative mechanisms of the gut, microbiome and metabolite profiles must be determined to pre-screen AD subjects prior to the design of specific, chronic titrations of gut microbiota with low-dose antibiotics. This represents an exciting treatment strategy designed to balance inflammatory microglial involvement in disease progression with an individual's manifestation of AD as influenced by a coercive inflammatory gut.
Collapse
Affiliation(s)
- David R Elmaleh
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,AZTherapies, Inc., Boston, MA, USA
| | | | | | - Jeremy E Wilkinson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ziv Neeman
- Department of Radiology, Emek Medical Center, Afula, Israel.,Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel.,Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
29
|
Li J, Zheng M, Shimoni O, Banks WA, Bush AI, Gamble JR, Shi B. Development of Novel Therapeutics Targeting the Blood-Brain Barrier: From Barrier to Carrier. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101090. [PMID: 34085418 PMCID: PMC8373165 DOI: 10.1002/advs.202101090] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/11/2021] [Indexed: 05/05/2023]
Abstract
The blood-brain barrier (BBB) is a highly specialized neurovascular unit, initially described as an intact barrier to prevent toxins, pathogens, and potentially harmful substances from entering the brain. An intact BBB is also critical for the maintenance of normal neuronal function. In cerebral vascular diseases and neurological disorders, the BBB can be disrupted, contributing to disease progression. While restoration of BBB integrity serves as a robust biomarker of better clinical outcomes, the restrictive nature of the intact BBB presents a major hurdle for delivery of therapeutics into the brain. Recent studies show that the BBB is actively engaged in crosstalk between neuronal and the circulatory systems, which defines another important role of the BBB: as an interfacing conduit that mediates communication between two sides of the BBB. This role has been subject to extensive investigation for brain-targeted drug delivery and shows promising results. The dual roles of the BBB make it a unique target for drug development. Here, recent developments and novel strategies to target the BBB for therapeutic purposes are reviewed, from both barrier and carrier perspectives.
Collapse
Affiliation(s)
- Jia Li
- School of PharmacyHenan UniversityKaifeng475001China
- Centre for Motor Neuron DiseaseDepartment of Biomedical SciencesFaculty of Medicine & Health SciencesMacquarie UniversitySydneyNew South Wales2109Australia
| | - Meng Zheng
- Henan‐Macquarie University Joint Center for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifengHenan475004China
| | - Olga Shimoni
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesFaculty of ScienceUniversity of Technology SydneySydneyNew South Wales2007Australia
| | - William A. Banks
- Geriatric Research Education and Clinical CenterVeterans Affairs Puget Sound Health Care System and Division of Gerontology and Geriatric MedicineDepartment of MedicineUniversity of Washington School of MedicineSeattleWA98108USA
| | - Ashley I. Bush
- Melbourne Dementia Research CenterThe Florey Institute for Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoria3052Australia
| | - Jennifer R. Gamble
- Center for the EndotheliumVascular Biology ProgramCentenary InstituteThe University of SydneySydneyNew South Wales2042Australia
| | - Bingyang Shi
- School of PharmacyHenan UniversityKaifeng475001China
- Centre for Motor Neuron DiseaseDepartment of Biomedical SciencesFaculty of Medicine & Health SciencesMacquarie UniversitySydneyNew South Wales2109Australia
- Henan‐Macquarie University Joint Center for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifengHenan475004China
| |
Collapse
|
30
|
Rinaldi C, Donato L, Alibrandi S, Scimone C, D’Angelo R, Sidoti A. Oxidative Stress and the Neurovascular Unit. Life (Basel) 2021; 11:767. [PMID: 34440511 PMCID: PMC8398978 DOI: 10.3390/life11080767] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022] Open
Abstract
The neurovascular unit (NVU) is a relatively recent concept that clearly describes the relationship between brain cells and their blood vessels. The components of the NVU, comprising different types of cells, are so interrelated and associated with each other that they are considered as a single functioning unit. For this reason, even slight disturbances in the NVU could severely affect brain homeostasis and health. In this review, we aim to describe the current state of knowledge concerning the role of oxidative stress on the neurovascular unit and the role of a single cell type in the NVU crosstalk.
Collapse
Affiliation(s)
- Carmela Rinaldi
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (C.R.); (L.D.); (S.A.); (R.D.); (A.S.)
| | - Luigi Donato
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (C.R.); (L.D.); (S.A.); (R.D.); (A.S.)
- Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Via Michele Miraglia, 90139 Palermo, Italy
| | - Simona Alibrandi
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (C.R.); (L.D.); (S.A.); (R.D.); (A.S.)
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Concetta Scimone
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (C.R.); (L.D.); (S.A.); (R.D.); (A.S.)
- Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Via Michele Miraglia, 90139 Palermo, Italy
| | - Rosalia D’Angelo
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (C.R.); (L.D.); (S.A.); (R.D.); (A.S.)
| | - Antonina Sidoti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy; (C.R.); (L.D.); (S.A.); (R.D.); (A.S.)
| |
Collapse
|
31
|
Sivandzade F, Alqahtani F, Cucullo L. Impact of chronic smoking on traumatic brain microvascular injury: An in vitro study. J Cell Mol Med 2021; 25:7122-7134. [PMID: 34160882 PMCID: PMC8335687 DOI: 10.1111/jcmm.16741] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/26/2022] Open
Abstract
Traumatic brain injury (TBI) is a major reason of cerebrovascular and neurological damage. Premorbid conditions such as tobacco smoking (TS) can worsen post‐TBI injuries by promoting vascular endothelial impairments. Indeed, TS‐induced oxidative stress (OS) and inflammation can hamper the blood‐brain barrier (BBB) endothelium. This study evaluated the subsequence of chronic TS exposure on BBB endothelial cells in an established in vitro model of traumatic cell injury. Experiments were conducted on confluent TS‐exposed mouse brain microvascular endothelial cells (mBMEC‐P5) following scratch injury. The expression of BBB integrity–associated tight junction (TJ) proteins was assessed by immunofluorescence imaging (IF), Western blotting (WB) and quantitative RT‐PCR. We evaluated reactive oxygen species (ROS) generation, the nuclear factor 2–related (Nrf2) with its downstream effectors and several inflammatory markers. Thrombomodulin expression was used to assess the endothelial haemostatic response to injury and TS exposure. Our results show that TS significantly decreased Nrf2, thrombomodulin and TJ expression in the BBB endothelium injury models while increased OS and inflammation compared to parallel TS‐free cultures. These data suggest that chronic TS exposure exacerbates traumatic endothelial injury and abrogates the protective antioxidative cell responses. The downstream effect was a more significant decline of BBB endothelial viability, which could aggravate subsequent neurological impairments.
Collapse
Affiliation(s)
- Farzane Sivandzade
- Department of Biological Sciences, Oakland University, Rochester, MI, USA.,Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Luca Cucullo
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| |
Collapse
|
32
|
Aryal R, Patabendige A. Blood-brain barrier disruption in atrial fibrillation: a potential contributor to the increased risk of dementia and worsening of stroke outcomes? Open Biol 2021; 11:200396. [PMID: 33878948 PMCID: PMC8059575 DOI: 10.1098/rsob.200396] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Atrial fibrillation (AF) has become one of the most significant health problems worldwide, warranting urgent answers to currently pending questions on the effects of AF on brain function. Recent evidence has emerged to show an association between AF and an increased risk of developing dementia and worsening of stroke outcomes. A healthy brain is protected by the blood–brain barrier (BBB), which is formed by the endothelial cells that line cerebral capillaries. These endothelial cells are continuously exposed to shear stress (the frictional force generated by blood flow), which affects endothelial cell structure and function. Flow disturbances as experienced during AF can disrupt the BBB and leave the brain vulnerable to damage. Investigating the plausible mechanisms in detail, linking AF to cerebrovascular damage is difficult in humans, leading to paucity of available clinical data. Here, we discuss the available evidence for BBB disruption during AF due to altered cerebral blood flow, and how this may contribute to an increased risk of dementia and worsening of stroke outcomes.
Collapse
Affiliation(s)
- Ritambhara Aryal
- Brain Barriers Group, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia.,Brain and Mental Health Research Programme, Hunter Medical Research Institute, Newcastle, Australia
| | - Adjanie Patabendige
- Brain Barriers Group, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia.,Brain and Mental Health Research Programme, Hunter Medical Research Institute, Newcastle, Australia.,Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
33
|
Hou XH, Xu W, Bi YL, Shen XN, Ma YH, Dong Q, Tan L, Yu JT. Associations of healthy lifestyles with cerebrospinal fluid biomarkers of Alzheimer's disease pathology in cognitively intact older adults: the CABLE study. ALZHEIMERS RESEARCH & THERAPY 2021; 13:81. [PMID: 33875016 PMCID: PMC8056495 DOI: 10.1186/s13195-021-00822-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/06/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVE We aimed to investigate the associations between healthy lifestyles and Alzheimer's disease (AD) biomarkers in cerebrospinal fluid (CSF). METHODS A total of 1108 cognitively intact individuals from Chinese Alzheimer's Biomarker and LifestylE (CABLE) study were examined to evaluate the associations of AD biomarkers with healthy lifestyle factors, including no current smoking, no harmful drinking, absence of social isolation, and regular physical activity. The participants were categorized into groups of favorable, intermediate, and unfavorable lifestyles according to the lifestyle factors. The associations between overall lifestyle and CSF biomarkers were also analyzed. RESULTS Among cognitively intact older adults, those having more social engagement had lower CSF tau (p = 0.009) and p-tau (p < 0.001) than those who had social isolation. Regular physical activity was associated with higher CSF Aβ42 (p = 0.013) and lower levels of CSF tau (p = 0.036) and p-tau (p = 0.007). However, no significant associations were found of smoking status or alcohol intake with CSF biomarkers. When the overall lifestyle of the participants was evaluated by all the four lifestyle factors, favorable lifestyle profiles were related to lower levels of CSF tau (p < 0.001) and p-tau (p < 0.001). CONCLUSIONS These findings suggest that healthy lifestyles had a beneficial effect on AD pathology among cognitively intact elders.
Collapse
Affiliation(s)
- Xiao-He Hou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan-Lin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
34
|
Delgado A, Cholevas C, Theoharides TC. Neuroinflammation in Alzheimer's disease and beneficial action of luteolin. Biofactors 2021; 47:207-217. [PMID: 33615581 DOI: 10.1002/biof.1714] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), already the world's most common form of dementia, is projected to continue increasing in prevalence over the next several decades. The current lack of understanding of the pathogenesis of AD has hampered the development of effective treatments. Historically, AD research has been predicated on the amyloid cascade hypothesis (ACH), which attributes disease progression to the build-up of amyloid protein. However, multiple clinical studies of drugs interfering with ACH have failed to show any benefit demonstrating that AD etiology is more complex than previously thought. Here we review the current literature on the emerging key role of neuroinflammation, especially activation of microglia, in AD pathogenesis. Moreover, we provide compelling evidence that certain flavonoids, especially luteolin formulated in olive pomace oil together with hydroxytyrosol, offers a reasonable prophylactic treatment approach due to its many beneficial actions.
Collapse
Affiliation(s)
- Alejandro Delgado
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Biomedical Sciences Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Christos Cholevas
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
- BrainGate, Thessaloniki, Greece
| | - Theoharis C Theoharides
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Biomedical Sciences Program, Tufts University School of Medicine, Boston, Massachusetts, USA
- BrainGate, Thessaloniki, Greece
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Department of Internal Medicine, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts, USA
- Department of Psychiatry, Tufts University School of Medicine and Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Gul G, Ileri-Ercan N. Fullerene translocation through peroxidized lipid membranes. RSC Adv 2021; 11:7575-7586. [PMID: 35423238 PMCID: PMC8694942 DOI: 10.1039/d1ra00272d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/08/2021] [Indexed: 12/20/2022] Open
Abstract
Recent cytotoxicity research suggests that fullerenes can enter the cell and cross the blood-brain barrier. However, the underlying toxicity mechanism behind the penetration of fullerenes through biological membranes is still not well understood. Here we perform coarse-grained molecular dynamics simulations to investigate the interactions of fullerenes and their polar derivatives (Janus) with model regular and peroxidized bilayers. We show that the translocation of fullerenes and their residence time in bulk water vary depending on the bilayer's peroxidation degree and fullerene polarity. The distribution of fullerenes inside the bilayer is mainly determined by the peroxidation degree and the saturation level of lipid acyl chains. The transport of pristine fullerenes through bilayers occurs at nano timescale while the complete diffusion may not be achieved for Janus fullerenes in micro timescale. As for the toxic response of fullerenes in terms of membrane damage, no mechanical disruption of model bilayers is observed throughout the studied simulation times.
Collapse
Affiliation(s)
- Gulsah Gul
- Department of Chemical Engineering, Bogazici University Istanbul Turkey
| | - Nazar Ileri-Ercan
- Department of Chemical Engineering, Bogazici University Istanbul Turkey
| |
Collapse
|
36
|
Dal Magro R, Vitali A, Fagioli S, Casu A, Falqui A, Formicola B, Taiarol L, Cassina V, Marrano CA, Mantegazza F, Anselmi-Tamburini U, Sommi P, Re F. Oxidative Stress Boosts the Uptake of Cerium Oxide Nanoparticles by Changing Brain Endothelium Microvilli Pattern. Antioxidants (Basel) 2021; 10:antiox10020266. [PMID: 33572224 PMCID: PMC7916071 DOI: 10.3390/antiox10020266] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/21/2022] Open
Abstract
Vascular oxidative stress is considered a worsening factor in the progression of Alzheimer's disease (AD). Increased reactive oxygen species (ROS) levels promote the accumulation of amyloid-β peptide (Aβ), one of the main hallmarks of AD. In turn, Aβ is a potent inducer of oxidative stress. In early stages of AD, the concomitant action of oxidative stress and Aβ on brain capillary endothelial cells was observed to compromise the blood-brain barrier functionality. In this context, antioxidant compounds might provide therapeutic benefits. To this aim, we investigated the antioxidant activity of cerium oxide nanoparticles (CNP) in human cerebral microvascular endothelial cells (hCMEC/D3) exposed to Aβ oligomers. Treatment with CNP (13.9 ± 0.7 nm in diameter) restored basal ROS levels in hCMEC/D3 cells, both after acute or prolonged exposure to Aβ. Moreover, we found that the extent of CNP uptake by hCMEC/D3 was +43% higher in the presence of Aβ. Scanning electron microscopy and western blot analysis suggested that changes in microvilli structures on the cell surface, under pro-oxidant stimuli (Aβ or H2O2), might be involved in the enhancement of CNP uptake. This finding opens the possibility to exploit the modulation of endothelial microvilli pattern to improve the uptake of anti-oxidant particles designed to counteract ROS-mediated cerebrovascular dysfunctions.
Collapse
Affiliation(s)
- Roberta Dal Magro
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (S.F.); (B.F.); (L.T.); (V.C.); (C.A.M.); (F.M.); (F.R.)
- Correspondence:
| | - Agostina Vitali
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy; (A.V.); (U.A.-T.)
| | - Stefano Fagioli
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (S.F.); (B.F.); (L.T.); (V.C.); (C.A.M.); (F.M.); (F.R.)
| | - Alberto Casu
- NABLA Lab, Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (A.C.); (A.F.)
| | - Andrea Falqui
- NABLA Lab, Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia; (A.C.); (A.F.)
| | - Beatrice Formicola
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (S.F.); (B.F.); (L.T.); (V.C.); (C.A.M.); (F.M.); (F.R.)
| | - Lorenzo Taiarol
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (S.F.); (B.F.); (L.T.); (V.C.); (C.A.M.); (F.M.); (F.R.)
| | - Valeria Cassina
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (S.F.); (B.F.); (L.T.); (V.C.); (C.A.M.); (F.M.); (F.R.)
| | - Claudia Adriana Marrano
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (S.F.); (B.F.); (L.T.); (V.C.); (C.A.M.); (F.M.); (F.R.)
| | - Francesco Mantegazza
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (S.F.); (B.F.); (L.T.); (V.C.); (C.A.M.); (F.M.); (F.R.)
| | | | - Patrizia Sommi
- Human Physiology Unit, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Francesca Re
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (S.F.); (B.F.); (L.T.); (V.C.); (C.A.M.); (F.M.); (F.R.)
| |
Collapse
|
37
|
Jurcau A. The Role of Natural Antioxidants in the Prevention of Dementia-Where Do We Stand and Future Perspectives. Nutrients 2021; 13:282. [PMID: 33498262 PMCID: PMC7909256 DOI: 10.3390/nu13020282] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Dementia, and especially Alzheimer's disease (AD), puts significant burden on global healthcare expenditure through its increasing prevalence. Research has convincingly demonstrated the implication of oxidative stress in the pathogenesis of dementia as well as of the conditions which increase the risk of developing dementia. However, drugs which target single pathways have so far failed in providing significant neuroprotection. Natural antioxidants, due to their effects in multiple pathways through which oxidative stress leads to neurodegeneration and triggers neuroinflammation, could prove valuable weapons in our fight against dementia. Although efficient in vitro and in animal models of AD, natural antioxidants in human trials have many drawbacks related to the limited bioavailability, unknown optimal dose, or proper timing of the treatment. Nonetheless, trials evaluating several of these natural compounds are ongoing, as are attempts to modify these compounds to achieve improved bioavailability.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, nr 1 Universitatii Street, 410087 Oradea, Romania;
- Neurology Ward, Clinical Municipal Hospital “Dr. G. Curteanu”, nr 12 Corneliu Coposu Street, 410469 Oradea, Romania
| |
Collapse
|
38
|
From "Leaky Gut" to Impaired Glia-Neuron Communication in Depression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1305:129-155. [PMID: 33834399 DOI: 10.1007/978-981-33-6044-0_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In the last three decades, the robust scientific data emerged, demonstrating that the immune-inflammatory response is a fundamental component of the pathophysiology of major depressive disorder (MDD). Psychological stress and various inflammatory comorbidities contribute to such immune activation. Still, this is not uncommon that patients with depression do not have defined inflammatory comorbidities, and alternative mechanisms of immune activation need to take place. The gastrointestinal (GI) tract, along with gut-associated lymphoid tissue (GALT), constitutes the largest lymphatic organ in the human body and forms the biggest surface of contact with the external environment. It is also the most significant source of bacterial and food-derived antigenic material. There is a broad range of reciprocal interactions between the GI tract, intestinal microbiota, increased intestinal permeability, activation of immune-inflammatory response, and the CNS that has crucial implications in brain function and mental health. This intercommunication takes place within the microbiota-gut-immune-glia (MGIG) axis, and glial cells are the main orchestrator of this communication. A broad range of factors, including psychological stress, inflammation, dysbiosis, may compromise the permeability of this barrier. This leads to excessive bacterial translocation and the excessive influx of food-derived antigenic material that contributes to activation of the immune-inflammatory response and depressive psychopathology. This chapter summarizes the role of increased intestinal permeability in MDD and mechanisms of how the "leaky gut" may contribute to immune-inflammatory response in this disorder.
Collapse
|
39
|
Kim D, Kim KA, Kim JH, Kim EH, Bae ON. Methylglyoxal-Induced Dysfunction in Brain Endothelial Cells via the Suppression of Akt/HIF-1α Pathway and Activation of Mitophagy Associated with Increased Reactive Oxygen Species. Antioxidants (Basel) 2020; 9:antiox9090820. [PMID: 32899154 PMCID: PMC7554889 DOI: 10.3390/antiox9090820] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/20/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022] Open
Abstract
Methylglyoxal (MG) is a dicarbonyl compound, the level of which is increased in the blood of diabetes patients. MG is reported to be involved in the development of cerebrovascular complications in diabetes, but the exact mechanisms need to be elucidated. Here, we investigated the possible roles of oxidative stress and mitophagy in MG-induced functional damage in brain endothelial cells (ECs). Treatment of MG significantly altered metabolic stress as observed by the oxygen-consumption rate and barrier-integrity as found in impaired trans-endothelial electrical resistance in brain ECs. The accumulation of MG adducts and the disturbance of the glyoxalase system, which are major detoxification enzymes of MG, occurred concurrently. Reactive oxygen species (ROS)-triggered oxidative damage was observed with increased mitochondrial ROS production and the suppressed Akt/hypoxia-inducible factor 1 alpha (HIF-1α) pathway. Along with the disturbance of mitochondrial bioenergetic function, parkin-1-mediated mitophagy was increased by MG. Treatment of N-acetyl cysteine significantly reversed mitochondrial damage and mitophagy. Notably, MG induced dysregulation of tight junction proteins including occludin, claudin-5, and zonula occluden-1 in brain ECs. Here, we propose that diabetic metabolite MG-associated oxidative stress may contribute to mitochondrial damage and autophagy in brain ECs, resulting in the dysregulation of tight junction proteins and the impairment of permeability.
Collapse
|
40
|
Rudzki L, Maes M. The Microbiota-Gut-Immune-Glia (MGIG) Axis in Major Depression. Mol Neurobiol 2020; 57:4269-4295. [DOI: 10.1007/s12035-020-01961-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023]
|
41
|
Blood-brain barrier dysfunction: the undervalued frontier of hypertension. J Hum Hypertens 2020; 34:682-691. [PMID: 32424144 DOI: 10.1038/s41371-020-0352-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) constitutes the complex anatomic and physiologic interface between the intravascular compartment and the central nervous system, and its integrity is paramount for the maintenance of the very sensitive homeostasis of the central nervous system. Arterial hypertension is a leading cause of morbidity and mortality. The BBB has been shown to be disrupted in essential hypertension. BBB integrity is important for central autonomic control and this may be implicated in the pathophysiology of hypertension. On the other hand, evidence from experimental studies indicates that BBB disruption can be present in both hypertensive disease and dementia syndromes, suggesting a possibly key position of loss of BBB integrity in the pathophysiological pathways linking arterial hypertension with cognitive decline. Although much still remains to be elucidated with respect to the exact underlying mechanisms, the discovery of novel pathological pathways has changed our understanding of adult dementia and central nervous system disease overall, pointing out-in parallel-new potential therapeutic targets. The aim of this review is to summarize current scientific knowledge relevant to the pathophysiologic pathways that are involved in the disruption of the BBB function and potentially mediate hypertension-induced cognitive impairment. In parallel, we underline the differential cognition-preserving effect of several antihypertensive agents of similar blood pressure-lowering capacity, highlighting the presence of previously under-recognized BBB-protective actions of these drugs.
Collapse
|
42
|
Sivandzade F, Alqahtani F, Sifat A, Cucullo L. The cerebrovascular and neurological impact of chronic smoking on post-traumatic brain injury outcome and recovery: an in vivo study. J Neuroinflammation 2020; 17:133. [PMID: 32340626 PMCID: PMC7184717 DOI: 10.1186/s12974-020-01818-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is among the most prevalent causes of cerebrovascular and neurological damage worldwide. To this end, tobacco smoke (TS) has been shown to promote vascular inflammation, neurovascular impairments, and risk of cerebrovascular and neurological disorders through oxidative stress (OS) stimuli targeting the blood-brain barrier (BBB) endothelium among others. It has been recently suggested that premorbid conditions such as TS may exacerbate post-TBI brain damage and impact recovery. METHODS Our study investigated the mechanisms underlying the exacerbation of TBI injury by TS using a weight drop model. For this purpose, male C57BL/6J mice, age range 6-8 weeks, were chronically exposed to premorbid TS for 3 weeks. Test animals were then subjected to TBI by guided vertical head weight drop using a 30 g metal weight free felling from an 80 cm distance before reaching the target. We analyzed the physical activity and body weight of the mice before TBI and 1 h, 24 h, and 72 h post-injury. Finally, mice were sacrificed to collect blood and brain samples for subsequent biochemical and molecular analysis. Western blotting was applied to assess the expression of Nrf2 (a critical antioxidant transcription factor) as well as tight junction proteins associated with BBB integrity including ZO-1, Occludin, and Claudin-5 from brain tissues homogenates. Levels of NF-kB (a pro-inflammatory transcript factor which antagonizes Nrf2 activity) and pro-inflammatory cytokines IL-6, IL-10, and TNF-α were assessed in blood samples. RESULTS Our data revealed that premorbid TS promoted significantly increased inflammation and loss of BBB integrity in TBI when compared to TS-Free test mice. Additionally, mice chronically exposed to TS before TBI experienced a more significant weight loss, behavioral and motor activity deficiency, and slower post-TBI recovery when compared to TS-free TBI mice. CONCLUSION The effects of premorbid TS appear consequential to the abrogation of physiological antioxidative and anti-inflammatory response to TBI leading to worsening impairments of the BBB, OS damage, and inflammation. These factors are also likely responsible for the retardation of post-traumatic recovery observed in these animals.
Collapse
Affiliation(s)
- Farzane Sivandzade
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center | Jerry H. Hodge School of pharmacy, 1300 S. Coulter Street, Amarillo, TX 79106 USA
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451 Saudi Arabia
| | - Ali Sifat
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center | Jerry H. Hodge School of pharmacy, 1300 S. Coulter Street, Amarillo, TX 79106 USA
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center | Jerry H. Hodge School of pharmacy, 1300 S. Coulter Street, Amarillo, TX 79106 USA
- Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX 79106 USA
| |
Collapse
|
43
|
Lymphocyte-to-monocyte ratio and risk of hemorrhagic transformation in patients with acute ischemic stroke. Neurol Sci 2020; 41:2511-2520. [DOI: 10.1007/s10072-020-04355-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/16/2020] [Indexed: 02/08/2023]
|
44
|
Blood total antioxidant status is associated with cortical glucose uptake and factors related to accelerated aging. Brain Struct Funct 2020; 225:841-851. [PMID: 32048020 DOI: 10.1007/s00429-020-02039-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022]
Abstract
Identifying cerebral vulnerability in late life is of paramount importance to prevent pathological trajectories of aging before the onset of symptoms. Considerable evidence suggests that impaired antioxidant mechanisms are a fingerprint of aging-related conditions, but there is a lack of human research linking total antioxidant capacity (TAC) measured in peripheral blood to in vivo brain changes and other factors featuring accelerated aging. To address this issue, we have assessed in cognitively normal elderly subjects (N = 100) correlations between serum TAC, using the oxygen radical absorbance capacity assay, surface-based cortical thickness, surface-based 18F-fluorodeoxyglucose positron emission tomography cortical uptake, and different factors associated with accelerated aging [i.e., serum homocysteine (HCY), self-reported memory problems, and self-reported patterns of physical activity]. While no relationship was observed between serum TAC and variations in cortical thickness, decreased TAC level was significantly associated with lower FDG uptake in temporal lobes bilaterally. Remarkably, decreased TAC level was linked to increased HCY concentrations, more subjective memory complaints, and lower frequency of physical activity. Overall, our results suggest that decreased serum TAC level may be helpful to detect vulnerable trajectories of aging.
Collapse
|
45
|
Alamu O, Rado M, Ekpo O, Fisher D. Differential Sensitivity of Two Endothelial Cell Lines to Hydrogen Peroxide Toxicity: Relevance for In Vitro Studies of the Blood-Brain Barrier. Cells 2020; 9:cells9020403. [PMID: 32050666 PMCID: PMC7072657 DOI: 10.3390/cells9020403] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/20/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress (OS) has been linked to blood–brain barrier (BBB) dysfunction which in turn has been implicated in the initiation and propagation of some neurological diseases. In this study, we profiled, for the first time, two endothelioma cell lines of mouse brain origin, commonly used as in vitro models of the blood–brain barrier, for their resistance against oxidative stress using viability measures and glutathione contents as markers. OS was induced by exposing cultured cells to varying concentrations of hydrogen peroxide and fluorescence microscopy/spectrometry was used to detect and estimate cellular glutathione contents. A colorimetric viability assay was used to determine changes in the viability of OS-exposed cells. Both the b.End5 and bEnd.3 cell lines investigated showed demonstrable content of glutathione with a statistically insignificant difference in glutathione quantity per unit cell, but with a statistically significant higher capacity for the b.End5 cell line for de novo glutathione synthesis. Furthermore, the b.End5 cells demonstrated greater oxidant buffering capacity to higher concentrations of hydrogen peroxide than the bEnd.3 cells. We concluded that mouse brain endothelial cells, derived from different types of cell lines, differ enormously in their antioxidant characteristics. We hereby recommend caution in making comparisons across BBB models utilizing distinctly different cell lines and require further prerequisites to ensure that in vitro BBB models involving these cell lines are reliable and reproducible.
Collapse
Affiliation(s)
- Olufemi Alamu
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town 7530, South Africa; (O.A.); (M.R.); (O.E.)
- Anatomy Department, Ladoke Akintola University of Technology, Ogbomoso 210241, Nigeria
| | - Mariam Rado
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town 7530, South Africa; (O.A.); (M.R.); (O.E.)
| | - Okobi Ekpo
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town 7530, South Africa; (O.A.); (M.R.); (O.E.)
| | - David Fisher
- Department of Medical Bioscience, University of the Western Cape, Bellville, Cape Town 7530, South Africa; (O.A.); (M.R.); (O.E.)
- Correspondence: ; Tel.: +27-21-959-2185
| |
Collapse
|
46
|
Dicarbonyl Stress and S-Glutathionylation in Cerebrovascular Diseases: A Focus on Cerebral Cavernous Malformations. Antioxidants (Basel) 2020; 9:antiox9020124. [PMID: 32024152 PMCID: PMC7071005 DOI: 10.3390/antiox9020124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/25/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
Dicarbonyl stress is a dysfunctional state consisting in the abnormal accumulation of reactive α-oxaldehydes leading to increased protein modification. In cells, post-translational changes can also occur through S-glutathionylation, a highly conserved oxidative post-translational modification consisting of the formation of a mixed disulfide between glutathione and a protein cysteine residue. This review recapitulates the main findings supporting a role for dicarbonyl stress and S-glutathionylation in the pathogenesis of cerebrovascular diseases, with specific emphasis on cerebral cavernous malformations (CCM), a vascular disease of proven genetic origin that may give rise to various clinical signs and symptoms at any age, including recurrent headaches, seizures, focal neurological deficits, and intracerebral hemorrhage. A possible interplay between dicarbonyl stress and S-glutathionylation in CCM is also discussed.
Collapse
|
47
|
Padureanu R, Albu CV, Mititelu RR, Bacanoiu MV, Docea AO, Calina D, Padureanu V, Olaru G, Sandu RE, Malin RD, Buga AM. Oxidative Stress and Inflammation Interdependence in Multiple Sclerosis. J Clin Med 2019; 8:jcm8111815. [PMID: 31683787 PMCID: PMC6912446 DOI: 10.3390/jcm8111815] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/27/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
The study aims to explore the oxidative status related to inflammation in peripheral blood of stable relapsing-remitting multiple sclerosis (MS) patients with low disability. In this study, 31 people were included and divided into two groups: an MS group in which 16 relapsing-remitting MS patients with a low disability level (age 38.9 ± 7.08, EDSS median 2.5) were included and a control group that contains 15 healthy volunteers of similar age to the MS group. Thiobarbituric acid reactive substances (TBARS), protein carbonyl level (PCO), total antioxidant capacity (TAC) as oxidative stress markers, neutrophil/lymphocyte ratio (NLR), and erythrocyte sedimentation rate (ESR) were analyzed in the peripheral blood sample of the healthy and the MS patients to establish the oxidative stress/inflammatory level using conventional plasma markers. In this study, we showed that the pro-inflammatory status of the relapse-remitting stage of diseases can be easily and accurately appreciated by NLR. An increased NLR is associated with a decreased antioxidant capacity, even in the early stage of neuronal damage. Oxidative stress associated with inflammation aggravates the functional outcome, potentiates neuronal damage, and can accelerate the progression of the disease.
Collapse
Affiliation(s)
- Rodica Padureanu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Carmen Valeria Albu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Radu Razvan Mititelu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Manuela Violeta Bacanoiu
- Department of Physical Therapy and Sports Medicine, University of Craiova, 200207 Craiova, Romania.
- Department of Laboratory Medicine, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania.
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Vlad Padureanu
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Gabriela Olaru
- Department of Physical Therapy and Sports Medicine, University of Craiova, 200207 Craiova, Romania.
| | - Raluca Elena Sandu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
- Department of Neurology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Ramona Denise Malin
- Department of Neurology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| | - Ana-Maria Buga
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania.
| |
Collapse
|
48
|
Cruz S, Narayanaswami V. Cellular Uptake and Clearance of Oxidatively-modified Apolipoprotein E3 by Cerebral Cortex Endothelial Cells. Int J Mol Sci 2019; 20:ijms20184582. [PMID: 31533203 PMCID: PMC6769588 DOI: 10.3390/ijms20184582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 12/12/2022] Open
Abstract
Apolipoprotein E3 (apoE3) plays a critical role in the metabolism of lipoproteins and lowers plasma lipid levels by serving as a ligand for the low-density lipoprotein receptor (LDLr) family of proteins and by promoting macrophage cholesterol efflux. The current study examines the effect of acrolein (an endogenously generated metabolite and an environmental pollutant) modification on the structure and function of apoE3. Acrolein modification was confirmed in Western blots by reactivity with acrolein–lysine-specific antibody and by the presence of oligomeric species due to cross-linking. LC-MS/MS analysis revealed modification of 10 out of 12 lysines in apoE3, with Nε-(3-methylpyridinium)-lysine being the predominant form of modification, and Lys75 being a ‘hot spot’ in terms of susceptibility to oxidation. Circular dichroism spectroscopy showed no major change in overall secondary structure compared to unmodified apoE3. Reconstituted high density lipoprotein (HDL) bearing acrolein modified apoE3 showed loss of binding to soluble LDLr; however, incubation with mouse endothelioma bEnd.3 cells showed that it was internalized. Incubation with excess LDL did not abolish cellular uptake of acrolein modified apoE3, suggesting alternative mechanism(s) not involving LDLr. Incubation with anti-CD36 antibody did not show a decrease in internalization while incubation with anti- lectin-like oxidized LDL receptor 1 (LOX1) showed partial internalization. However, incubation with anti-scavenger receptor class B type I (SRB1) antibody abolished internalization of acrolein modified apoE3. Taken together, our studies suggest that acrolein modification of apoE3 at lysine residues leads to increase in net negative charge, and as a consequence, results in clearance by LOX1 and SRB1 on endothelial cells. Overall, oxidative modification of apoE3 likely impairs its role in regulating plasma cholesterol homeostasis, eventually leading to lipid disorders.
Collapse
Affiliation(s)
- Siobanth Cruz
- Department of Chemistry and Biochemistry 1250 Bellflower Blvd., California State University Long Beach, Long Beach, CA 90840, USA.
| | - Vasanthy Narayanaswami
- Department of Chemistry and Biochemistry 1250 Bellflower Blvd., California State University Long Beach, Long Beach, CA 90840, USA.
| |
Collapse
|
49
|
Anti-Diabetic Countermeasures Against Tobacco Smoke-Dependent Cerebrovascular Toxicity: Use and Effect of Rosiglitazone. Int J Mol Sci 2019; 20:ijms20174225. [PMID: 31470514 PMCID: PMC6747143 DOI: 10.3390/ijms20174225] [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: 07/13/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/19/2022] Open
Abstract
Tobacco smoking (TS) is one of the most addictive habit sand a main public health hazards, impacting the vascular endothelium through oxidative stress (OS) stimuli, exposure to nicotine, and smoking-induced inflammation in a dose-dependent manner. Increasing evidence also suggested that TS increases glucose intolerance and the risk factor of developing type-2 diabetes mellitus (2DM), which, along with TS, is connected to blood–brain barrier (BBB) injuries, and heightens the risk of cerebrovascular disorders. Although the exact mechanism of rosiglitazone (RSG) is unknown, our previous in vitro work showed how RSG, an oral anti-diabetic drug belonging to the family of thiazolidinedione class, can protect BBB integrity through enhancement of nuclear factor erythroid 2-related factor (Nrf2) activity. Herein, we have validated the protective role of rosiglitazone against TS-induced BBB impairment in vivo. Our results revealed that RSG as a peroxisome proliferator-activated receptor gamma (PPARγ), activates counteractive mechanisms primarily associated with the upregulation of Nrf2 and PPARγ pathways which reduce TS-dependent toxicity at the cerebrovascular level. In line with these findings, our results show that RSG reduces inflammation and protects BBB integrity. In conclusion, RSG offers a novel and promising therapeutic application to reduce TS-induced cerebrovascular dysfunction through activation of the PPARγ-dependent and/or PPARγ-independent Nrf2 pathway.
Collapse
|
50
|
Peña-Bautista C, Baquero M, Vento M, Cháfer-Pericás C. Free radicals in Alzheimer's disease: Lipid peroxidation biomarkers. Clin Chim Acta 2019; 491:85-90. [DOI: 10.1016/j.cca.2019.01.021] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 01/09/2023]
|