251
|
Vezzani B, Carinci M, Patergnani S, Pasquin MP, Guarino A, Aziz N, Pinton P, Simonato M, Giorgi C. The Dichotomous Role of Inflammation in the CNS: A Mitochondrial Point of View. Biomolecules 2020; 10:E1437. [PMID: 33066071 PMCID: PMC7600410 DOI: 10.3390/biom10101437] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/07/2020] [Accepted: 10/10/2020] [Indexed: 12/14/2022] Open
Abstract
Innate immune response is one of our primary defenses against pathogens infection, although, if dysregulated, it represents the leading cause of chronic tissue inflammation. This dualism is even more present in the central nervous system, where neuroinflammation is both important for the activation of reparatory mechanisms and, at the same time, leads to the release of detrimental factors that induce neurons loss. Key players in modulating the neuroinflammatory response are mitochondria. Indeed, they are responsible for a variety of cell mechanisms that control tissue homeostasis, such as autophagy, apoptosis, energy production, and also inflammation. Accordingly, it is widely recognized that mitochondria exert a pivotal role in the development of neurodegenerative diseases, such as multiple sclerosis, Parkinson's and Alzheimer's diseases, as well as in acute brain damage, such in ischemic stroke and epileptic seizures. In this review, we will describe the role of mitochondria molecular signaling in regulating neuroinflammation in central nervous system (CNS) diseases, by focusing on pattern recognition receptors (PRRs) signaling, reactive oxygen species (ROS) production, and mitophagy, giving a hint on the possible therapeutic approaches targeting mitochondrial pathways involved in inflammation.
Collapse
Affiliation(s)
- Bianca Vezzani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Marianna Carinci
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Simone Patergnani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Matteo P. Pasquin
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Annunziata Guarino
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Department of BioMedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Nimra Aziz
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Department of BioMedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy
| | - Michele Simonato
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Department of BioMedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy
- School of Medicine, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| |
Collapse
|
252
|
Early life overnutrition impairs plasticity of non-neuronal brainstem cells and drives obesity in offspring across development in rats. Int J Obes (Lond) 2020; 44:2405-2418. [PMID: 32999409 DOI: 10.1038/s41366-020-00658-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/17/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prevalence of adolescent obesity has increased dramatically, becoming a serious public health concern. While previous evidence suggests that in utero- and early postnatal overnutrition increases adult-onset obesity risk, the neurobiological mechanisms underlying this outcome are not well understood. Non-neuronal cells play an underestimated role in the physiological responses to metabolic/nutrient signals. Hypothalamic glial-mediated inflammation is now considered a contributing factor in the development and perpetuation of obesity; however, attention on the role of gliosis and microglia activation in other nuclei is still needed. METHODS/RESULTS Here, we demonstrate that early life consumption of high-fat/sucrose diet (HFSD) is sufficient to increase offspring body weight, hyperleptinemia and potentially maladaptive cytoarchitectural changes in the brainstem dorsal-vagal-complex (DVC), an essential energy balance processing hub, across postnatal development. Our data demonstrate that pre- and postnatal consumption of HFSD result in increased body weight, hyperleptinemia and dramatically affects the non-neuronal landscape, and therefore the plasticity of the DVC in the developing offspring. CONCLUSIONS Current findings are very provocative, considering the importance of the DVC in appetite regulation, suggesting that HFSD-consumption during early life may contribute to subsequent obesity risk via DVC cytoarchitectural changes.
Collapse
|
253
|
Poon CH, Wang Y, Fung ML, Zhang C, Lim LW. Rodent Models of Amyloid-Beta Feature of Alzheimer's Disease: Development and Potential Treatment Implications. Aging Dis 2020; 11:1235-1259. [PMID: 33014535 PMCID: PMC7505263 DOI: 10.14336/ad.2019.1026] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/26/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide and causes severe financial and social burdens. Despite much research on the pathogenesis of AD, the neuropathological mechanisms remain obscure and current treatments have proven ineffective. In the past decades, transgenic rodent models have been used to try to unravel this disease, which is crucial for early diagnosis and the assessment of disease-modifying compounds. In this review, we focus on transgenic rodent models used to study amyloid-beta pathology in AD. We also discuss their possible use as promising tools for AD research. There is still no effective treatment for AD and the development of potent therapeutics are urgently needed. Many molecular pathways are susceptible to AD, ranging from neuroinflammation, immune response, and neuroplasticity to neurotrophic factors. Studying these pathways may shed light on AD pathophysiology as well as provide potential targets for the development of more effective treatments. This review discusses the advantages and limitations of these models and their potential therapeutic implications for AD.
Collapse
Affiliation(s)
- Chi Him Poon
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yingyi Wang
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man-Lung Fung
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chengfei Zhang
- 2Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Lee Wei Lim
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
254
|
Kikutani K, Giga H, Hosokawa K, Shime N, Aizawa H. Microglial translocator protein and stressor-related disorder. Neurochem Int 2020; 140:104855. [PMID: 32980493 DOI: 10.1016/j.neuint.2020.104855] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 02/08/2023]
Abstract
Despite the prevalence of neuroinflammation in psychiatric disorders, molecular mechanism underlying it remains elusive. Translocator protein 18 kDa (TSPO), also known as peripheral benzodiazepine receptor, is a mitochondrial protein implicated in the synthesis of steroids in a variety of tissues. Multiple reports have shown increased expression of TSPO in the activated microglia in the CNS. Radioactive probes targeting TSPO have been developed and used for imaging assessment in neurological and psychiatric disorders to examine neuroinflammation. Recent studies revealed that the wide range of stressors ranging from psychological to physical insults induced TSPO in human, suggesting that this protein could be an important tool to explore the contribution of microglia in stressor-related disorders. In this review, we first overview the microglial activation with TSPO in a wide range of stressors in human and animal models to discuss prevalent roles of TSPO in response of CNS to stressors. With recent update of the signaling pathway revealing link connecting TSPO with neuroinflammatory effectors such as reactive oxygen species, we discuss TSPO as a therapeutic targeting tool for suppression of adverse effect of stressors on long-lasting changes in animal behaviors and activities. Targeting TSPO which mediates neuroinflammation under the stress might pave the way to develop therapeutic intervention and prophylaxis of stressor-related disorder.
Collapse
Affiliation(s)
- Kazuya Kikutani
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Japan
| | - Hiroshi Giga
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Japan
| | - Koji Hosokawa
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Japan
| | - Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Science, Hiroshima University, Japan
| | - Hidenori Aizawa
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan.
| |
Collapse
|
255
|
Fan X, Ma W, Zhang Y, Zhang L. P2X7 Receptor (P2X7R) of Microglia Mediates Neuroinflammation by Regulating (NOD)-Like Receptor Protein 3 (NLRP3) Inflammasome-Dependent Inflammation After Spinal Cord Injury. Med Sci Monit 2020; 26:e925491. [PMID: 32952148 PMCID: PMC7518010 DOI: 10.12659/msm.925491] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Microglia participate in mediating neuroinflammation in which P2X7R triggered by adenosine triphosphate has a critical effect after spinal cord injury. However, how the P2X7R of microglia regulate neuroinflammation after spinal cord injury is still unclear. The aim of this study was to explore the mechanism by which the P2X7 receptor of microglia regulates neuroinflammation after spinal cord injury in NLRP3 inflammasome-dependent inflammation. Material/Methods Sixt rats were divided into 5 groups: a sham group, a model group, a BzATP group, an A-438079 group, and a BzATP+CY-09 group. Rats in the sham group were only subjected to laminectomy and rats in the other groups were subjected to spinal cord injury followed by treatment with physiological saline, BzATP, A-438079, and BzATP following CY-09, separately. Real-time polymerase chain reaction, Western blot, immunofluorescence staining, and enzyme-linked immunosorbent assay were used to analyze the scientific hypothesis. Results (i) P2X7R of microglia was upregulated and downregulated by BzATP, and A-438079 was upregulated after spinal cord injury. (ii) Upregulation of P2X7R on microglia is coincident with increase of neuroinflammation after spinal cord injury. (iii) P2X7R of microglia participates in spinal cord-mediated neuroinflammation via regulating NLRP3 inflammasome-dependent inflammation. Conclusions P2X7R of microglia in spinal cord mediates neuroinflammation by regulating NLRP3 inflammasome-dependent inflammation after spinal cord injury.
Collapse
Affiliation(s)
- Xiao Fan
- Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland).,Fujian Universities and Colleges Engineering Research Center of Marine Biopharmaceutical Resources, Xiamen, Fujian, China (mainland)
| | - Wei Ma
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Yingyu Zhang
- Qingdao Municipal Hospital, Qingdao, Shandong, China (mainland)
| | - Li Zhang
- Fujian Universities and Colleges Engineering Research Center of Marine Biopharmaceutical Resources, Xiamen, Fujian, China (mainland).,Xiamen Medical College, Xiamen, Fujian, China (mainland)
| |
Collapse
|
256
|
Lin CW, Fan CH, Chang YC, Hsieh-Li HM. ERK activation precedes Purkinje cell loss in mice with Spinocerebellar ataxia type 17. Neurosci Lett 2020; 738:135337. [PMID: 32877710 DOI: 10.1016/j.neulet.2020.135337] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/27/2020] [Accepted: 08/26/2020] [Indexed: 12/18/2022]
Abstract
Spinocerebellar ataxia type 17 (SCA17) is an autosomal dominant neurodegenerative disease caused by CAG expansion in the gene encoding the TATA-binding protein (TBP). The neurological features of SCA17 are Purkinje cell loss and gliosis. We have generated SCA17 transgenic mice which recapitulate the patients' phenotypes and are suitable for the study of the SCA17 pathomechanism. Our previous study identified the activation of mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) occurred in the SCA17 cerebella, this study aims to study the role of ERK activation in SCA17. The levels of pERK, calbindin, and gliosis markers on the mouse cerebellum at 4-8 weeks old were analyzed to elucidate the correlation among behavioral performance, ERK activation and Purkinje cell degeneration. The motor incoordination was initiated in SCA17 mice at 6 weeks old. We found that the presence of TBP nuclear aggregation and microglia activation were observed at 4 weeks old. Gliosis of astrocytes and Bergmann glia, pERK, Bax/Bcl2 ratio, and caspase-3 were significantly increased in the 6-week-old SCA17 mouse cerebellum. In addition to the polyglutamine-protein aggregation in Purkinje cells caused apoptosis cell-autonomously, a significant body of evidence have shown that ERK pathways involves in neuronal apoptosis. Our study showed that the activation of ERK in the astrocytes and Bergmann glia was identified as preceding motor deficits, which suggest the elevated gliosis by ERK activation may contribute to neuronal apoptosis in SCA17 mice.
Collapse
Affiliation(s)
- Chia-Wei Lin
- Department of Life Science, National Taiwan Normal University, Taiwan
| | - Chia-Hao Fan
- Department of Life Science, National Taiwan Normal University, Taiwan
| | - Ya-Chin Chang
- Department of Pharmacy, Taiwan Adventist Hospital, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taiwan.
| |
Collapse
|
257
|
Fischer I, Barak B. Molecular and Therapeutic Aspects of Hyperbaric Oxygen Therapy in Neurological Conditions. Biomolecules 2020; 10:E1247. [PMID: 32867291 PMCID: PMC7564723 DOI: 10.3390/biom10091247] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
In hyperbaric oxygen therapy (HBOT), the subject is placed in a chamber containing 100% oxygen gas at a pressure of more than one atmosphere absolute. This treatment is used to hasten tissue recovery and improve its physiological aspects, by providing an increased supply of oxygen to the damaged tissue. In this review, we discuss the consequences of hypoxia, as well as the molecular and physiological processes that occur in subjects exposed to HBOT. We discuss the efficacy of HBOT in treating neurological conditions and neurodevelopmental disorders in both humans and animal models. We summarize by discussing the challenges in this field, and explore future directions that will allow the scientific community to better understand the molecular aspects and applications of HBOT for a wide variety of neurological conditions.
Collapse
Affiliation(s)
- Inbar Fischer
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel;
| | - Boaz Barak
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel;
- The School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
258
|
de Leeuw FA, Schneider JA, Agrawal S, Leurgans SE, Morris MC. Brain tocopherol levels are associated with lower activated microglia density in elderly human cortex. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12021. [PMID: 32864412 PMCID: PMC7444784 DOI: 10.1002/trc2.12021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/17/2020] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Higher brain tocopherol levels have been associated with lower levels of Alzheimer's disease (AD) neuropathology; however, the underlying mechanisms are unclear. METHODS We studied the relations of α- and γ-tocopherol brain levels to microglia density in 113 deceased participants from the Memory and Aging Project. We used linear regression analyses to examine associations between tocopherol levels and microglia densities in a basic model adjusted for age, sex, education, apolipoprotein E (APOE)ε4 genotype (any ε4 allele vs. none) , and post-mortem time interval, and a second model additionally adjusted for total amyloid load and neurofibrillary tangle severity. RESULTS Higher α- and γ-tocopherol levels were associated with lower total and activated microglia density in cortical but not in subcortical brain regions. The association between cortical α-tocopherol and total microglia density remained statistically significant after adjusting for AD neuropathology. DISCUSSION These results suggest that the relation between tocopherols and AD might be partly explained by the alleviating effects of tocopherols on microglia activation.
Collapse
Affiliation(s)
- Francisca A. de Leeuw
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdam UMCAmsterdamThe Netherlands
| | - Julie A. Schneider
- Rush Alzheimer's Disease CenterDepartment of Neurological Sciences and Department of Pathology (Neuropathology)Rush University Medical CenterChicagoIllinois
| | - Sonal Agrawal
- Rush Alzheimer's Disease CenterDepartment of Pathology (Neuropathology)Rush University Medical CenterChicagoIllinois
| | - Sue E. Leurgans
- Rush Alzheimer's Disease Center and Department of Neurological SciencesRush University Medical CenterChicagoIllinois
| | - Martha Clare Morris
- Rush Institute for Healthy AgingRush University Medical CenterChicagoIllinois
| |
Collapse
|
259
|
Simpson DSA, Oliver PL. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants (Basel) 2020; 9:E743. [PMID: 32823544 PMCID: PMC7463655 DOI: 10.3390/antiox9080743] [Citation(s) in RCA: 546] [Impact Index Per Article: 109.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative disorders, such as Alzheimer's disease, are a global public health burden with poorly understood aetiology. Neuroinflammation and oxidative stress (OS) are undoubtedly hallmarks of neurodegeneration, contributing to disease progression. Protein aggregation and neuronal damage result in the activation of disease-associated microglia (DAM) via damage-associated molecular patterns (DAMPs). DAM facilitate persistent inflammation and reactive oxygen species (ROS) generation. However, the molecular mechanisms linking DAM activation and OS have not been well-defined; thus targeting these cells for clinical benefit has not been possible. In microglia, ROS are generated primarily by NADPH oxidase 2 (NOX2) and activation of NOX2 in DAM is associated with DAMP signalling, inflammation and amyloid plaque deposition, especially in the cerebrovasculature. Additionally, ROS originating from both NOX and the mitochondria may act as second messengers to propagate immune activation; thus intracellular ROS signalling may underlie excessive inflammation and OS. Targeting key kinases in the inflammatory response could cease inflammation and promote tissue repair. Expression of antioxidant proteins in microglia, such as NADPH dehydrogenase 1 (NQO1), is promoted by transcription factor Nrf2, which functions to control inflammation and limit OS. Lipid droplet accumulating microglia (LDAM) may also represent a double-edged sword in neurodegenerative disease by sequestering peroxidised lipids in non-pathological ageing but becoming dysregulated and pro-inflammatory in disease. We suggest that future studies should focus on targeted manipulation of NOX in the microglia to understand the molecular mechanisms driving inflammatory-related NOX activation. Finally, we discuss recent evidence that therapeutic target identification should be unbiased and founded on relevant pathophysiological assays to facilitate the discovery of translatable antioxidant and anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Dominic S. A. Simpson
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire OX11 0RD, UK;
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Peter L. Oliver
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, Oxfordshire OX11 0RD, UK;
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| |
Collapse
|
260
|
Woollacott IOC, Toomey CE, Strand C, Courtney R, Benson BC, Rohrer JD, Lashley T. Microglial burden, activation and dystrophy patterns in frontotemporal lobar degeneration. J Neuroinflammation 2020; 17:234. [PMID: 32778130 PMCID: PMC7418403 DOI: 10.1186/s12974-020-01907-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background Microglial dysfunction is implicated in frontotemporal lobar degeneration (FTLD). Although studies have reported excessive microglial activation or senescence (dystrophy) in Alzheimer’s disease (AD), few have explored this in FTLD. We examined regional patterns of microglial burden, activation and dystrophy in sporadic and genetic FTLD, sporadic AD and controls. Methods Immunohistochemistry was performed in frontal and temporal grey and white matter from 50 pathologically confirmed FTLD cases (31 sporadic, 19 genetic: 20 FTLD-tau, 26 FTLD-TDP, four FTLD-FUS), five AD cases and five controls, using markers to detect phagocytic (CD68-positive) and antigen-presenting (CR3/43-positive) microglia, and microglia in general (Iba1-positive). Microglial burden and activation (morphology) were assessed quantitatively for each microglial phenotype. Iba1-positive microglia were assessed semi-quantitatively for dystrophy severity and qualitatively for rod-shaped and hypertrophic morphology. Microglia were compared in each region between FTLD, AD and controls, and between different pathological subtypes of FTLD, including its main subtypes (FTLD-tau, FTLD-TDP, FTLD-FUS), and subtypes of FTLD-tau, FTLD-TDP and genetic FTLD. Microglia were also compared between grey and white matter within each lobe for each group. Results There was a higher burden of phagocytic and antigen-presenting microglia in FTLD and AD cases than controls, but activation was often not increased. Burden was generally higher in white matter than grey matter, but activation was greater in grey matter. However, microglia varied regionally according to FTLD subtype and disease mechanism. Dystrophy was more severe in FTLD and AD than controls, and more severe in white than grey matter, but this also varied regionally and was particularly extensive in FTLD due to progranulin (GRN) mutations. Presence of rod-shaped and hypertrophic microglia also varied by FTLD subtype. Conclusions This study demonstrates regionally variable microglial involvement in FTLD and links this to underlying disease mechanisms. This supports investigation of microglial dysfunction in disease models and consideration of anti-senescence therapies in clinical trials.
Collapse
Affiliation(s)
- Ione O C Woollacott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Christina E Toomey
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Catherine Strand
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Robert Courtney
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Bridget C Benson
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK. .,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
261
|
Xu D, Zhang Y, Xie B, Yao H, Yuan Y, Yuan S, Zhang J. The spleen mediates chronic sleep restriction-mediated enhancement of LPS-induced neuroinflammation, cognitive deficits, and anxiety-like behavior. Aging (Albany NY) 2020; 12:15446-15461. [PMID: 32741775 PMCID: PMC7467362 DOI: 10.18632/aging.103659] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
Abstract
Chronic sleep restriction promotes neuroinflammation and cognitive deficits in neurodegenerative and neurobehavioral diseases. The spleens of mice exposed to chronic and repeated psychological stress serve as a reservoir of inflammatory myeloid cells that are released into the blood and brain following secondary acute stress. Here, we tested whether chronic and repeated short-term sleep restriction (CRSR) would exacerbate lipopolysaccharide (LPS)-induced neuroinflammation, cognitive deficits, and anxiety-like behavior in a spleen-dependent manner. LPS was administered to aged mice 14 days after exposure to CRSR consisting of three cycles of 7 days of sleep restriction with 7-day intervals in between. CRSR increased plasma proinflammatory cytokine levels, blood-brain barrier permeability, hippocampal proinflammatory cytokine levels, and transition of microglia to the M1 phenotype 24 h after LPS treatment. This in turn led to cognitive deficits and anxiety-like behavior. Interestingly, removal of the spleen 14 days prior to CRSR abrogated the enhancement of LPS-induced increases in systemic inflammation, neuroinflammation, cognitive deficits, and anxiety-like behavior. These data indicate that the spleen was essential for CRSR-induced exacerbation of LPS-induced brain damage.
Collapse
Affiliation(s)
- Dan Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yujing Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bing Xie
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hua Yao
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yin Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shiying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiancheng Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
262
|
Yachou Y, El Idrissi A, Belapasov V, Ait Benali S. Neuroinvasion, neurotropic, and neuroinflammatory events of SARS-CoV-2: understanding the neurological manifestations in COVID-19 patients. Neurol Sci 2020; 41:2657-2669. [PMID: 32725449 PMCID: PMC7385206 DOI: 10.1007/s10072-020-04575-3] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/02/2020] [Indexed: 12/19/2022]
Abstract
Respiratory viruses are opportunistic pathogens that infect the upper respiratory tract in humans and cause severe illnesses, especially in vulnerable populations. Some viruses have neuroinvasive properties and activate the immune response in the brain. These immune events may be neuroprotective or they may cause long-term damage similar to what is seen in some neurodegenerative diseases. The new “Severe Acute Respiratory Syndrome Coronavirus 2” (SARS-CoV-2) is one of the Respiratory viruses causing highly acute lethal pneumonia coronavirus disease 2019 (COVID-19) with clinical similarities to those reported in “Severe Acute Respiratory Syndrome Coronavirus”(SARS-CoV) and the “Middle East Respiratory Syndrome Coronavirus”(MERS-CoV) including neurological manifestation. To examine the possible neurological damage induced by SARS-CoV-2, it is necessary to understand the immune reactions to viral infection in the brain, and their short- and long-term consequences. Considering the similarities between SARS-CoV and SARS-CoV-2, which will be discussed, cooperative homological and phylogenetical studies lead us to question if SARS-CoV-2 can have similar neuroinvasive capacities and neuroinflammatiory events that may lead to the same short- and long-term neuropathologies that SARS-CoV had shown in human and animal models. To explain the neurological manifestation caused by SARS-CoV-2, we will present a literature review of 765 COVID-19 patients, in which 18% had neurological symptoms and complications, including encephalopathy, encephalitis and cerebrovascular pathologies, acute myelitis, and Guillain-Barré syndrome. Clinical studies describe anosmia or partial loss of the sense of smell as the most frequent symptom in COVID19 patients, suggesting that olfactory dysfunction and the initial ultrarapid immune responses could be a prognostic factor.
Collapse
Affiliation(s)
- Yassine Yachou
- Neurology Department, Astrakhan State Medical University, Astrakhan, Russia.
| | - Abdeslem El Idrissi
- Center for Developmental Neuroscience, City University of New York, College of Staten Island, New York, USA
| | - Vladimir Belapasov
- Neurology Department, Astrakhan State Medical University, Astrakhan, Russia
| | - Said Ait Benali
- Neurosurgery Department, Mohammed VI University Hospital Center, Cadi Ayyad University, Marrakech, Morocco
| |
Collapse
|
263
|
Ait Wahmane S, Achbani A, Ouhaz Z, Elatiqi M, Belmouden A, Nejmeddine M. The Possible Protective Role of α-Synuclein Against Severe Acute Respiratory Syndrome Coronavirus 2 Infections in Patients With Parkinson's Disease. Mov Disord 2020; 35:1293-1294. [PMID: 32519352 PMCID: PMC7300655 DOI: 10.1002/mds.28185] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/01/2020] [Indexed: 12/22/2022] Open
Affiliation(s)
- Sofiane Ait Wahmane
- Laboratory of Cell Biology and Molecular Genetics, Department of Biology, Faculty of Sciences, University Ibn Zohr, Agadir, Morocco
| | - Abderrahmane Achbani
- Laboratory of Cell Biology and Molecular Genetics, Department of Biology, Faculty of Sciences, University Ibn Zohr, Agadir, Morocco
| | - Zakaria Ouhaz
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Mohamed Elatiqi
- Laboratory of Cell Biology and Molecular Genetics, Department of Biology, Faculty of Sciences, University Ibn Zohr, Agadir, Morocco
| | - Ahmed Belmouden
- Laboratory of Cell Biology and Molecular Genetics, Department of Biology, Faculty of Sciences, University Ibn Zohr, Agadir, Morocco
| | - Mohamed Nejmeddine
- Laboratory of Cell Biology and Molecular Genetics, Department of Biology, Faculty of Sciences, University Ibn Zohr, Agadir, Morocco
| |
Collapse
|
264
|
Alam MB, Kwon YG, Simu SY, Abrar Shahriyar S, Lee SH. Attenuation of Inflammatory Symptoms by Icariside B2 in Carrageenan and LPS-Induced Inflammation Models via Regulation of MAPK/NF-κB Signaling Cascades. Biomolecules 2020; 10:1037. [PMID: 32664577 PMCID: PMC7408066 DOI: 10.3390/biom10071037] [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: 05/19/2020] [Revised: 07/02/2020] [Accepted: 07/08/2020] [Indexed: 12/03/2022] Open
Abstract
: Prolonged inflammatory responses can lead to the development of several chronic diseases, such as autoimmune disorders and the development of natural therapeutic agents is required. A murine model was used to assess the anti-inflammatory effects of the megastigmane glucoside, icariside B2 (ICSB), and the assessment was carried out in vitro, and in vivo. The in vitro anti-inflammatory effects of ICSB were tested using LPS-stimulated BV2 cells, and the protein expression levels of inflammatory genes and cytokines were assessed. Mice were subcutaneously injected with 1% carrageenan (CA) to induce acute phase inflammation in the paw. Inflammation was assessed by measuring paw volumes hourly; subsequently, the mice were euthanized and the right hind paw skin was expunged and processed for reverse transcription-polymerase chain reaction (RT-PCR) and Western blot analyses. ICSB inhibits LPS-stimulated nitric oxide (NO) and prostaglandin E2 (PGE2) generation by reducing the expression of inducible NO synthase (iNOS) and cyclooxygenase 2 (COX-2). ICSB also inhibits the COX-2 enzyme with an IC50 value of 7.80±0.26 µM. Molecular docking analysis revealed that ICSB had a strong binding affinity with both murine and human COX-2 proteins with binding energies of -8 kcal/mol and -7.4 kcal/mol, respectively. ICSB also reduces the manifestation of pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β, at their transcriptional and translational level. ICSB hinders inhibitory protein κBα (IκBα) phosphorylation, thereby terminating the nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB) nuclear translocation. ICSB also represses the mitogen-activated protein kinases (MAPKs) signaling pathways. ICSB (50 mg/kg) showed an anti-edema effect in CA-induced mice and suppressed the CA-induced increases in iNOS and COX-2 protein levels. ICSB attenuated inflammatory responses by downregulating NF-κB expression through interference with extracellular signal-regulated kinase (ERK) and p38 phosphorylation, and by modulating the expression levels of iNOS, COX-2, TNF-α, IL-1β, and IL-6.
Collapse
Affiliation(s)
- Md Badrul Alam
- School of Food Science and Biotechnology, Graduate School, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (Y.-G.K.)
- Food and Bio-Industry Research Institute, Inner Beauty/Antiaging Center, Kyungpook National University, Daegu 41566, Korea
| | - Yoon-Gyung Kwon
- School of Food Science and Biotechnology, Graduate School, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (Y.-G.K.)
| | - Shakina Yesmin Simu
- Department of Brain Science, Ajou University School of Medicine, Suwon 16499, Korea;
| | - Sk Abrar Shahriyar
- Biomedical Science, Graduate School, Ajou University School of Medicine, Suwon 16499, Korea;
| | - Sang Han Lee
- School of Food Science and Biotechnology, Graduate School, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (Y.-G.K.)
- Food and Bio-Industry Research Institute, Inner Beauty/Antiaging Center, Kyungpook National University, Daegu 41566, Korea
- knu BnC, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
265
|
Sa de Almeida J, Vargas M, Fonseca-Gomes J, Tanqueiro SR, Belo RF, Miranda-Lourenço C, Sebastião AM, Diógenes MJ, Pais TF. Microglial Sirtuin 2 Shapes Long-Term Potentiation in Hippocampal Slices. Front Neurosci 2020; 14:614. [PMID: 32625056 PMCID: PMC7315392 DOI: 10.3389/fnins.2020.00614] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 05/18/2020] [Indexed: 11/15/2022] Open
Abstract
Microglial cells have emerged as crucial players in synaptic plasticity during development and adulthood, and also in neurodegenerative and neuroinflammatory conditions. Here we found that decreased levels of Sirtuin 2 (Sirt2) deacetylase in microglia affects hippocampal synaptic plasticity under inflammatory conditions. The results show that long-term potentiation (LTP) magnitude recorded from hippocampal slices of wild type mice does not differ between those exposed to lipopolysaccharide (LPS), a pro-inflammatory stimulus, or BSA. However, LTP recorded from hippocampal slices of microglial-specific Sirt2 deficient (Sirt2–) mice was significantly impaired by LPS. Importantly, LTP values were restored by memantine, an antagonist of N-methyl-D-aspartate (NMDA) receptors. These results indicate that microglial Sirt2 prevents NMDA-mediated excitotoxicity in hippocampal slices in response to an inflammatory signal such as LPS. Overall, our data suggest a key-protective role for microglial Sirt2 in mnesic deficits associated with neuroinflammation.
Collapse
Affiliation(s)
- Joana Sa de Almeida
- Division of Development and Growth, Department of Woman, Child and Adolescent, University Hospitals of Geneva, Geneva, Switzerland.,Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mariana Vargas
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João Fonseca-Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara Ramalho Tanqueiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rita F Belo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria José Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | |
Collapse
|
266
|
Retinal ischemia triggers early microglia activation in the optic nerve followed by neurofilament degeneration. Exp Eye Res 2020; 198:108133. [PMID: 32645332 DOI: 10.1016/j.exer.2020.108133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/14/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022]
Abstract
Retinal ischemia leads to an early severe damage of the retina and thus plays an important role in eye diseases such as angle-closure glaucoma or retinal vascular occlusion. In retinal diseases, there is common sense about the affection of the optic nerve by ischemic injury. However, the exact dynamic processes of this optic nerve degeneration are mainly unclear. In this study, retinal ischemia was induced in one eye of Brown-Norway rats by raising the intraocular pressure 60 min to 140 mmHg followed by natural reperfusion. Optic nerves were analyzed at six different points in time: 2, 6, 12, and 24 h as well as 3 and 7 days after ischemic injury. Cell infiltration and moreover signs of tissue demyelination and dissolution were noticed in optic nerves 7 days after ischemia (hematoxylin & eosin: p < 0.001, luxol fast blue: p = 0.04). Although microglial activation was verified already from 12 h on after ischemia (p = 0.030), the beginning of a structural degeneration of the neurofilament was seen at 3 days (p = 0.02). Interestingly, proliferative microglia were present later on (7 days: p = 0.017). At this point, the number of total microglia was also increased in ischemic nerves (p = 0.003). Concluding, our data indicate that not only retinal tissue is affected by an ischemia, the optic nerve also demonstrates progressive damage. Interestingly, a microglia activation was noted days before structural damage became visible.
Collapse
|
267
|
Iturria-Medina Y, Khan AF, Adewale Q, Shirazi AH. Blood and brain gene expression trajectories mirror neuropathology and clinical deterioration in neurodegeneration. Brain 2020; 143:661-673. [PMID: 31989163 DOI: 10.1093/brain/awz400] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 10/05/2019] [Accepted: 11/04/2019] [Indexed: 12/28/2022] Open
Abstract
Most prevalent neurodegenerative disorders take decades to develop and their early detection is challenged by confounding non-pathological ageing processes. For all neurodegenerative conditions, we continue to lack longitudinal gene expression data covering their large temporal evolution, which hinders the understanding of the underlying dynamic molecular mechanisms. Here, we overcome this key limitation by introducing a novel gene expression contrastive trajectory inference (GE-cTI) method that reveals enriched temporal patterns in a diseased population. Evaluated on 1969 subjects in the spectrum of late-onset Alzheimer's and Huntington's diseases (from ROSMAP, HBTRC and ADNI datasets), this unsupervised machine learning algorithm strongly predicts neuropathological severity (e.g. Braak, amyloid and Vonsattel stages). Furthermore, when applied to in vivo blood samples at baseline (ADNI), it significantly predicts clinical deterioration and conversion to advanced disease stages, supporting the identification of a minimally invasive (blood-based) tool for early clinical screening. This technique also allows the discovery of genes and molecular pathways, in both peripheral and brain tissues, that are highly predictive of disease evolution. Eighty-five to ninety per cent of the most predictive molecular pathways identified in the brain are also top predictors in the blood. These pathways support the importance of studying the peripheral-brain axis, providing further evidence for a key role of vascular structure/functioning and immune system response. The GE-cTI is a promising tool for revealing complex neuropathological mechanisms, with direct implications for implementing personalized dynamic treatments in neurology.
Collapse
Affiliation(s)
- Yasser Iturria-Medina
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada.,Ludmer Centre for NeuroInformatics and Mental Health, Montreal, Canada
| | - Ahmed F Khan
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada.,Ludmer Centre for NeuroInformatics and Mental Health, Montreal, Canada
| | - Quadri Adewale
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada.,Ludmer Centre for NeuroInformatics and Mental Health, Montreal, Canada
| | - Amir H Shirazi
- McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada.,Ludmer Centre for NeuroInformatics and Mental Health, Montreal, Canada
| | | |
Collapse
|
268
|
You M, Miao Z, Sienkiewicz O, Jiang X, Zhao X, Hu F. 10-Hydroxydecanoic acid inhibits LPS-induced inflammation by targeting p53 in microglial cells. Int Immunopharmacol 2020; 84:106501. [DOI: 10.1016/j.intimp.2020.106501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/22/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022]
|
269
|
Qian Z, Chang J, Jiang F, Ge D, Yang L, Li Y, Chen H, Cao X. Excess administration of miR-340-5p ameliorates spinal cord injury-induced neuroinflammation and apoptosis by modulating the P38-MAPK signaling pathway. Brain Behav Immun 2020; 87:531-542. [PMID: 32014577 DOI: 10.1016/j.bbi.2020.01.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 01/30/2020] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) is a destructive polyneuropathy that can result in loss of sensorimotor function and sphincter dysfunction, and even death in critical situations. MicroRNAs (miRs) are a series of non-coding RNA molecules that are involved in transcriptional regulation. Previous studies have demonstrated that modulation of multiple miRs is involved in neurological recovery after SCI. However, the functions of miR-340-5p in SCI remain uncertain. Therefore, we probed the therapeutic effect and mechanism of miR-340-5p in microglia in vitro and in vivo in SCI rats. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were employed to examine the alterations in miR-340-5p and P38 levels in SCI rats. miR-340-5p targets in microglia were ascertained using luciferase reporter assays, immunofluorescence analyses, and western blotting. We also established an SCI model and administered miR-340-5p. The effects of miR-340-5p on the amelioration of inflammation, oxidative stress, and apoptosis following SCI were assessed using immunofluorescence, immunohistochemistry, and histological analyses. Finally, locomotor function recovery was determined using the Basso, Beattie, Bresnahan rating scale. In our study, the expression profiles and luciferase assay results clarified that P38 was a target of miR-340-5p, which was associated with activation of the P38-MAPK signaling pathway. Elevation of miR-340-5p decreased P38 expression, subsequently inhibiting the inflammatory reaction. SCI-induced secondary neuroinflammation was relieved under miR-340-5p treatment. Moreover, by controlling neuroinflammation, the increased levels of miR-340-5p might counter oxidative stress and reduce the degree of apoptosis. We also observed decreasing gliosis and glial scar formation and increasing neurotrophin expression at the chronic stage of SCI. Together, these potential effects of miR-340-5p treatment ultimately improved locomotor function recovery in SCI rats.
Collapse
Affiliation(s)
- Zhanyang Qian
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Chang
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fan Jiang
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dawei Ge
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lei Yang
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - You Li
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongtao Chen
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Xiaojian Cao
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
270
|
Porte Alcon S, Gorojod RM, Kotler ML. Kinetic and protective role of autophagy in manganese-exposed BV-2 cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118787. [PMID: 32592735 DOI: 10.1016/j.bbamcr.2020.118787] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/01/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
Manganese (Mn) plays an important role in many physiological processes. Nevertheless, Mn accumulation in the brain can cause a parkinsonian-like syndrome known as manganism. Unfortunately, the therapeutic options for this disease are scarce and of limited efficacy. For this reason, a great effort is being made to understand the cellular and molecular mechanisms involved in Mn toxicity in neuronal and glial cells. Even though evidence indicates that Mn activates autophagy in microglia, the consequences of this activation in cell death remain unknown. In this study, we demonstrated a key role of reactive oxygen species in Mn-induced damage in microglial cells. These species generated by Mn2+ induce lysosomal alterations, LMP, cathepsins release and cell death. Besides, we described for the first time the kinetic of Mn2+-induced autophagy in BV-2 microglial cells and its relevance to cell fate. We found that Mn promotes a time-dependent increase in LC3-II and p62 expression levels, suggesting autophagy activation. Possibly, cells trigger autophagy to neutralize the risks associated with lysosomal rupture. In addition, pre-treatment with both Rapamycin and Melatonin enhanced autophagy and retarded Mn2+ cytotoxicity. In summary, our results demonstrated that, despite the damage inflicted on a subset of lysosomes, the autophagic pathway plays a protective role in Mn-induced microglial cell death. We propose that 2 h Mn2+ exposure will not induce disturbances in the autophagic flux. However, as time passes, the accumulated damage inside the cell could trigger a dysfunction of this mechanism. These findings may represent a valuable contribution to future research concerning manganism therapies.
Collapse
Affiliation(s)
- Soledad Porte Alcon
- CONICET- Universidad de Buenos Aires. Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN). Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina. Ciudad Autónoma de Buenos Aires, Argentina.
| | - Roxana Mayra Gorojod
- CONICET- Universidad de Buenos Aires. Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN). Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina. Ciudad Autónoma de Buenos Aires, Argentina.
| | - Mónica Lidia Kotler
- CONICET- Universidad de Buenos Aires. Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN). Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Laboratorio de Disfunción Celular en Enfermedades Neurodegenerativas y Nanomedicina. Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
271
|
Yahfoufi N, Matar C, Ismail N. Adolescence and Aging: Impact of Adolescence Inflammatory Stress and Microbiota Alterations on Brain Development, Aging, and Neurodegeneration. J Gerontol A Biol Sci Med Sci 2020; 75:1251-1257. [PMID: 31917834 PMCID: PMC7302172 DOI: 10.1093/gerona/glaa006] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Indexed: 12/16/2022] Open
Abstract
Puberty/adolescence is a critical phase during neurodevelopment with numerous structural, neurochemical, and molecular changes occurring in response to genetic and environmental signals. A consequence of this major neuronal reorganizing and remodeling is a heightened level of vulnerability to stressors and immune challenges. The gut microbiota is a fundamental modulator of stress and immune responses and has been found to play a role in mental health conditions and neurodegenerative disorders. Environmental insults (stress, infection, neuroinflammation, and use of antibiotics) during adolescence can result in dysbiosis subsidizing the development of brain disorders later in life. Also, pubertal neuroinflammatory insults can alter neurodevelopment, impact brain functioning in an enduring manner, and contribute to neurological disorders related to brain aging, such as Alzheimer's disease, Parkinson's disease, and depression. Exposure to probiotics during puberty can mitigate inflammation, reverse dysbiosis, and decrease vulnerabilities to brain disorders later in life. The goal of this review is to reveal the consequences of pubertal exposure to stress and immune challenges on the gut microbiota, immune reactivity within the brain, and the risk or resilience to stress-induced mental illnesses and neurodegenerative disorders. We propose that the consumption of probiotics during adolescence contribute to the prevention of brain pathologies in adulthood.
Collapse
Affiliation(s)
- Nour Yahfoufi
- Cellular and Molecular Medicine Department, Faculty of Medicine, University of Ottawa, Ontario, Canada
| | - Chantal Matar
- Cellular and Molecular Medicine Department, Faculty of Medicine, University of Ottawa, Ontario, Canada
- School of Nutrition, Faculty of Health Sciences, University of Ottawa, Ontario, Canada
| | - Nafissa Ismail
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Ontario, Canada
| |
Collapse
|
272
|
Duffy KB, Ray B, Lahiri DK, Tilmont EM, Tinkler GP, Herbert RL, Greig NH, Ingram DK, Ottinger MA, Mattison JA. Effects of Reducing Norepinephrine Levels via DSP4 Treatment on Amyloid-β Pathology in Female Rhesus Macaques (Macaca Mulatta). J Alzheimers Dis 2020; 68:115-126. [PMID: 30689563 DOI: 10.3233/jad-180487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The degeneration in the locus coeruleus associated with Alzheimer's disease suggests an involvement of the noradrenergic system in the disease pathogenesis. The role of depleted norepinephrine was tested in adult and aged rhesus macaques to develop a potential model for testing Alzheimer's disease interventions. Monkeys were injected with the noradrenergic neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4) or vehicle at 0, 3, and 6 months; brains were harvested at 9 months. Reduced norepinephrine in the locus coeruleus was accompanied by decreased dopamine β-hydroxylase staining and increased amyloid-β load in the aged group, and the proportion of potentially toxic amyloid-β42 peptide was increased. Immunohistochemistry revealed no effects on microglia or astrocytes. DSP4 treatment altered amyloid processing, but these changes were not associated with the induction of chronic neuroinflammation. These findings suggest norepinephrine deregulation is an essential component of a nonhuman primate model of Alzheimer's disease, but further refinement is necessary.
Collapse
Affiliation(s)
- Kara B Duffy
- Animal and Avian Sciences Department, University of Maryland, College Park, MD, USA
| | - Balmiki Ray
- Myriad Neuroscience (Assurex Health), Mason, OH, USA (present address).,Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Debomoy K Lahiri
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Edward M Tilmont
- Translational Gerontology Branch, National Institute on Aging, NIH Animal Center, Dickerson, MD, USA
| | - Gregory P Tinkler
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Richard L Herbert
- Clinical Medicine Branch, National Institute of Allergy and Infectious Disease, NIH, Dickerson, MD, USA
| | - Nigel H Greig
- Translational Gerontology Branch, NIA/NIH, Baltimore, MD, USA
| | - Donald K Ingram
- Nutritional Neuroscience and Aging Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Mary Ann Ottinger
- Animal and Avian Sciences Department, University of Maryland, College Park, MD, USA.,Department of Biology and Biochemistry, University of Houston, Houston, TX, USA (present address)
| | - Julie A Mattison
- Translational Gerontology Branch, National Institute on Aging, NIH Animal Center, Dickerson, MD, USA
| |
Collapse
|
273
|
Nzou G, Wicks RT, VanOstrand NR, Mekky GA, Seale SA, El-Taibany A, Wicks EE, Nechtman CM, Marrotte EJ, Makani VS, Murphy SV, Seeds MC, Jackson JD, Atala AJ. Multicellular 3D Neurovascular Unit Model for Assessing Hypoxia and Neuroinflammation Induced Blood-Brain Barrier Dysfunction. Sci Rep 2020; 10:9766. [PMID: 32555384 PMCID: PMC7299970 DOI: 10.1038/s41598-020-66487-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
The blood-brain barrier (BBB) is a dynamic component of the brain-vascular interface that maintains brain homeostasis and regulates solute permeability into brain tissue. The expression of tight junction proteins between adjacent endothelial cells and the presence of efflux proteins prevents entry of foreign substances into the brain parenchyma. BBB dysfunction, however, is evident in many neurological disorders including ischemic stroke, trauma, and chronic neurodegenerative diseases. Currently, major contributors to BBB dysfunction are not well understood. Here, we employed a multicellular 3D neurovascular unit organoid containing human brain microvascular endothelial cells, pericytes, astrocytes, microglia, oligodendrocytes and neurons to model the effects of hypoxia and neuroinflammation on BBB function. Organoids were cultured in hypoxic chamber with 0.1% O2 for 24 hours. Organoids cultured under this hypoxic condition showed increased permeability, pro-inflammatory cytokine production, and increased oxidative stress. The anti-inflammatory agents, secoisolariciresinol diglucoside and 2-arachidonoyl glycerol, demonstrated protection by reducing inflammatory cytokine levels in the organoids under hypoxic conditions. Through the assessment of a free radical scavenger and an anti-inflammatory endocannabinoid, we hereby report the utility of the model in drug development for drug candidates that may reduce the effects of ROS and inflammation under disease conditions. This 3D organoid model recapitulates characteristics of BBB dysfunction under hypoxic physiological conditions and when exposed to exogenous neuroinflammatory mediators and hence may have potential in disease modeling and therapeutic development.
Collapse
Affiliation(s)
- Goodwell Nzou
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA.
| | - Robert T Wicks
- Department of Neurology and Neurological Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Nicole R VanOstrand
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Gehad A Mekky
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
- Zoology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Stephanie A Seale
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Aya El-Taibany
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Elizabeth E Wicks
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Carl M Nechtman
- Department of Neurology and Neurological Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Eric J Marrotte
- Department of Neurology and Neurological Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Vishruti S Makani
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - M C Seeds
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - John D Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Anthony J Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| |
Collapse
|
274
|
Nzou G, Wicks RT, VanOstrand NR, Mekky GA, Seale SA, El-Taibany A, Wicks EE, Nechtman CM, Marrotte EJ, Makani VS, Murphy SV, Seeds MC, Jackson JD, Atala AJ. Multicellular 3D Neurovascular Unit Model for Assessing Hypoxia and Neuroinflammation Induced Blood-Brain Barrier Dysfunction. Sci Rep 2020; 10:9766. [PMID: 32555384 PMCID: PMC7299970 DOI: 10.1038/s41598-020-66487-8 10.1038/s41598-020-77348-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/20/2020] [Indexed: 05/10/2025] Open
Abstract
The blood-brain barrier (BBB) is a dynamic component of the brain-vascular interface that maintains brain homeostasis and regulates solute permeability into brain tissue. The expression of tight junction proteins between adjacent endothelial cells and the presence of efflux proteins prevents entry of foreign substances into the brain parenchyma. BBB dysfunction, however, is evident in many neurological disorders including ischemic stroke, trauma, and chronic neurodegenerative diseases. Currently, major contributors to BBB dysfunction are not well understood. Here, we employed a multicellular 3D neurovascular unit organoid containing human brain microvascular endothelial cells, pericytes, astrocytes, microglia, oligodendrocytes and neurons to model the effects of hypoxia and neuroinflammation on BBB function. Organoids were cultured in hypoxic chamber with 0.1% O2 for 24 hours. Organoids cultured under this hypoxic condition showed increased permeability, pro-inflammatory cytokine production, and increased oxidative stress. The anti-inflammatory agents, secoisolariciresinol diglucoside and 2-arachidonoyl glycerol, demonstrated protection by reducing inflammatory cytokine levels in the organoids under hypoxic conditions. Through the assessment of a free radical scavenger and an anti-inflammatory endocannabinoid, we hereby report the utility of the model in drug development for drug candidates that may reduce the effects of ROS and inflammation under disease conditions. This 3D organoid model recapitulates characteristics of BBB dysfunction under hypoxic physiological conditions and when exposed to exogenous neuroinflammatory mediators and hence may have potential in disease modeling and therapeutic development.
Collapse
Affiliation(s)
- Goodwell Nzou
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA.
| | - Robert T Wicks
- Department of Neurology and Neurological Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Nicole R VanOstrand
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Gehad A Mekky
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
- Zoology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Stephanie A Seale
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Aya El-Taibany
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Elizabeth E Wicks
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Carl M Nechtman
- Department of Neurology and Neurological Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Eric J Marrotte
- Department of Neurology and Neurological Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Vishruti S Makani
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - M C Seeds
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - John D Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Anthony J Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| |
Collapse
|
275
|
Postmortem Studies of Neuroinflammation in Autism Spectrum Disorder: a Systematic Review. Mol Neurobiol 2020; 57:3424-3438. [DOI: 10.1007/s12035-020-01976-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
|
276
|
He Z, Yang Y, Xing Z, Zuo Z, Wang R, Gu H, Qi F, Yao Z. Intraperitoneal injection of IFN-γ restores microglial autophagy, promotes amyloid-β clearance and improves cognition in APP/PS1 mice. Cell Death Dis 2020; 11:440. [PMID: 32514180 PMCID: PMC7280212 DOI: 10.1038/s41419-020-2644-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 01/07/2023]
Abstract
Autophagy is a major self-degradative process that maintains cellular homeostasis and function in mammalian cells. Autophagic dysfunction occurs in the early pathogenesis of Alzheimer’s disease (AD) and directly regulates amyloid-β (Aβ) metabolism. Although it has been proven that the cytokine IFN-γ enhances autophagy in macrophage cell lines, whether the signaling cascade is implicated in Aβ degradation in AD mouse models remains to be elucidated. Here, we found that 9 days of the intraperitoneal administration of IFN-γ significantly increased the LC3II/I ratio and decreased the level of p62 in APP/PS1 mice, an AD mouse model. In vitro, IFN-γ protected BV2 cells from Aβ toxicity by upregulating the expressions of Atg7 and Atg5 and the LC3II/I ratio, whereas these protective effects were ablated by interference with Atg5 expression. Moreover, IFN-γ enhanced autophagic flux, probably through suppressing the AKT/mTOR pathway both in vivo and in vitro. Importantly, using intravital two-photon microscopy and fluorescence staining, we found that microglia interacted with exogenous IFN-γ and Aβ, and surrounded Aβ in APP/PS1;CX3CR1-GFP+/− mice. In addition, IFN-γ treatment decreased the Aβ plaque load in the cortex and hippocampus and rescued cognitive deficits in APP/PS1 mice. Our data suggest a possible mechanism by which the peripheral injection of IFN-γ restores microglial autophagy to induce the phagocytosis of cerebral Aβ, which represents a potential therapeutic approach for the use of exogenous IFN-γ in AD.
Collapse
Affiliation(s)
- Zitian He
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China
| | - Yunjie Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China
| | - Zhiwei Xing
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China
| | - Rui Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China
| | - Huaiyu Gu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China.
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China. .,Teaching and Research Bureau of Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, Guangdong, China.
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China. .,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, #74, Zhongshan No. 2 Road, 510080, Guangzhou, China.
| |
Collapse
|
277
|
TNF-alpha-induced microglia activation requires miR-342: impact on NF-kB signaling and neurotoxicity. Cell Death Dis 2020; 11:415. [PMID: 32488063 PMCID: PMC7265562 DOI: 10.1038/s41419-020-2626-6] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/07/2020] [Accepted: 05/05/2020] [Indexed: 12/22/2022]
Abstract
Growing evidences suggest that sustained neuroinflammation, caused by microglia overactivation, is implicated in the development and aggravation of several neurological and psychiatric disorders. In some pathological conditions, microglia produce increased levels of cytotoxic and inflammatory mediators, such as tumor necrosis factor alpha (TNF-α), which can reactivate microglia in a positive feedback mechanism. However, specific molecular mediators that can be effectively targeted to control TNF-α-mediated microglia overactivation, are yet to be uncovered. In this context, we aim to identify novel TNF-α-mediated micro(mi)RNAs and to dissect their roles in microglia activation, as well as to explore their impact on the cellular communication with neurons. A miRNA microarray, followed by RT-qPCR validation, was performed on TNF-α-stimulated primary rat microglia. Gain- and loss-of-function in vitro assays and proteomic analysis were used to dissect the role of miR-342 in microglia activation. Co-cultures of microglia with hippocampal neurons, using a microfluidic system, were performed to understand the impact on neurotoxicity. Stimulation of primary rat microglia with TNF-α led to an upregulation of Nos2, Tnf, and Il1b mRNAs. In addition, ph-NF-kB p65 levels were also increased. miRNA microarray analysis followed by RT-qPCR validation revealed that TNF-α stimulation induced the upregulation of miR-342. Interestingly, miR-342 overexpression in N9 microglia was sufficient to activate the NF-kB pathway by inhibiting BAG-1, leading to increased secretion of TNF-α and IL-1β. Conversely, miR-342 inhibition led to a strong decrease in the levels of these cytokines after TNF-α activation. In fact, both TNF-α-stimulated and miR-342-overexpressing microglia drastically affected neuron viability. Remarkably, increased levels of nitrites were detected in the supernatants of these co-cultures. Globally, our findings show that miR-342 is a crucial mediator of TNF-α-mediated microglia activation and a potential target to tackle microglia-driven neuroinflammation.
Collapse
|
278
|
Yang Q, Wu B, Eles JR, Vazquez AL, Kozai TDY, Cui XT. Zwitterionic Polymer Coating Suppresses Microglial Encapsulation to Neural Implants In Vitro and In Vivo. ADVANCED BIOSYSTEMS 2020; 4:e1900287. [PMID: 32363792 PMCID: PMC7686959 DOI: 10.1002/adbi.201900287] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 01/08/2023]
Abstract
For brain computer interfaces (BCI), the immune response to implanted electrodes is a major biological cause of device failure. Bioactive coatings such as neural adhesion molecule L1 have been shown to improve the biocompatibility, but are difficult to handle or produce in batches. Here, a synthetic zwitterionic polymer coating, poly(sulfobetaine methacrylate) (PSBMA) is developed for neural implants with the goal of reducing the inflammatory host response. In tests in vitro, the zwitterionic coating inhibits protein adsorption and the attachment of fibroblasts and microglia, and remains stable for at least 4 weeks. In vivo two-photon microscopy on CX3CR1-GFP mice shows that the zwitterionic coating significantly suppresses the microglial encapsulation of neural microelectrodes over a 6 h observation period. Furthermore, the lower microglial encapsulation on zwitterionic polymer-coated microelectrodes is revealed to originate from a reduction in the size but not the number of microglial end feet. This work provides a facile method for coating neural implants with zwitterionic polymers and illustrates the initial interaction between microglia and coated surface at high temporal and spatial resolution.
Collapse
Affiliation(s)
- Qianru Yang
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| | - Bingchen Wu
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| | - James R Eles
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| | - Alberto L Vazquez
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 3025 East Carson Street, Pittsburgh, PA, 15219, USA
| | - Takashi D Y Kozai
- Center for Biotechnology and Bioengineering, University of Pittsburgh, 300 Technology Dr, Pittsburgh, PA, 15213, USA
| | - X Tracy Cui
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| |
Collapse
|
279
|
Hilliard A, Mendonca P, Soliman KFA. Involvement of NFƙB and MAPK signaling pathways in the preventive effects of Ganoderma lucidum on the inflammation of BV-2 microglial cells induced by LPS. J Neuroimmunol 2020; 345:577269. [PMID: 32480240 PMCID: PMC7382303 DOI: 10.1016/j.jneuroim.2020.577269] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
Abstract
Ganoderma lucidum extract (GLE) is a potent ancient Asian remedy for the treatment of various diseases. This study investigated GLE preventive effects on LPS-stimulated inflammation of BV-2 microglial cells. The results show that pre-treatment with GLE decreased expression of pro-inflammatory cytokines: G-CSF, IL1-α, MCP-5, MIP3α, and, with a higher effect in MIP3α. In RT-PCR assays, pre-treatment with GLE decreased mRNA expression of CHUK, NFκB1/p150, and IKBKE (NFƙB signaling), which may be associated with the neuropathology of Alzheimer's disease. The data show GLE inhibiting ability on pro-inflammatory mediators' release and suggest a potential role of GLE in neurodegenerative disease prevention.
Collapse
Affiliation(s)
- Aaron Hilliard
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, United States of America
| | - Patricia Mendonca
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, United States of America
| | - Karam F A Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, United States of America.
| |
Collapse
|
280
|
He T, Itano MS, Earley LF, Hall NE, Riddick N, Samulski RJ, Li C. The Influence of Murine Genetic Background in Adeno-Associated Virus Transduction of the Mouse Brain. HUM GENE THER CL DEV 2020; 30:169-181. [PMID: 31749390 DOI: 10.1089/humc.2019.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Adeno-associated virus (AAV) vectors have become an important tool for delivering therapeutic genes for a wide range of neurological diseases. AAV serotypes possess differential cellular tropism in the central nervous system. Although several AAV serotypes or mutants have been reported to transduce the brain efficiently, conflicting data occur across studies with the use of various rodent strains from different genetic backgrounds. Herein, we performed a systematic comparison of the brain transduction properties among five AAV serotypes (AAV2, 5, 7, 8, and 9) in two common rodent strains (C57BL/6J and FVB/N), following local intrastriatal injection of AAV vectors encoding enhanced green fluorescent protein (EGFP) driven by the CBh promoter. Important differences were found regarding overall cellular tropism and transduction efficiency, including contralateral transduction among the AAV serotypes and between the mouse strains. We have further found loss of NeuN-immunoreactivity and microglial activation from AAV transduction in the different mouse strains. The important strain-specific differences from our study suggest that the genetic background of the mouse may affect AAV serotype transduction properties in the brain. These data can provide valuable information about how to choose an effective AAV vector for clinical application and interpret the data obtained from preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Ting He
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michelle S Itano
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina.,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lauriel F Earley
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nikita E Hall
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Natallia Riddick
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
281
|
Synergistic depletion of gut microbial consortia, but not individual antibiotics, reduces amyloidosis in APPPS1-21 Alzheimer's transgenic mice. Sci Rep 2020; 10:8183. [PMID: 32424118 PMCID: PMC7235236 DOI: 10.1038/s41598-020-64797-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/16/2020] [Indexed: 01/11/2023] Open
Abstract
In preceding efforts, we demonstrated that antibiotic (ABX) cocktail-mediated perturbations of the gut microbiome in two independent transgenic lines, termed APPSWE/PS1ΔE9 and APPPS1-21, leads to a reduction in Aβ deposition in male mice. To determine whether these observed reductions of cerebral Aβ amyloidosis are specific to any individual antibiotic or require the synergistic effects of several antibiotics, we treated male APPPS1-21 transgenic mice with either individual ABX or an ABX cocktail and assessed amyloid deposition. Specifically, mice were subject to oral gavage with high dose kanamycin, gentamicin, colistin, metronidazole, vancomycin, individually or in a combination (ABX cocktail) from postnatal days (PND) 14 to 21, followed by ad libitum, low-dose individual ABX or ABX cocktail in the drinking water until the time of sacrifice. A control group was subject to gavage with water from PND 14 to 21 and received drinking water till the time of sacrifice. At the time of sacrifice, all groups showed distinct cecal microbiota profiles with the highest differences between control and ABX cocktail-treated animals. Surprisingly, only the ABX cocktail significantly reduced brain Aβ amyloidosis compared to vehicle-treated animals. In parallel studies, and to assess the potential exposure of ABX to the brain, we quantified the levels of each ABX in the brain by liquid chromatography-mass spectrometry (LC-MS) at PND 22 or at 7 weeks of age. With the exception of metronidazole (which was observed at less than 3% relative to the spiked control brains), we were unable to detect the other individual ABX in brain homogenates. Our findings suggest that synergistic alterations of gut microbial consortia, rather than individual antimicrobial agents, underlie the observed reductions in brain amyloidosis.
Collapse
|
282
|
Findley CA, Bartke A, Hascup KN, Hascup ER. Amyloid Beta-Related Alterations to Glutamate Signaling Dynamics During Alzheimer's Disease Progression. ASN Neuro 2020; 11:1759091419855541. [PMID: 31213067 PMCID: PMC6582288 DOI: 10.1177/1759091419855541] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) ranks sixth on the Centers for Disease Control and Prevention Top 10 Leading Causes of Death list for 2016, and the Alzheimer’s Association attributes 60% to 80% of dementia cases as AD related. AD pathology hallmarks include accumulation of senile plaques and neurofibrillary tangles; however, evidence supports that soluble amyloid beta (Aβ), rather than insoluble plaques, may instigate synaptic failure. Soluble Aβ accumulation results in depression of long-term potentiation leading to cognitive deficits commonly characterized in AD. The mechanisms through which Aβ incites cognitive decline have been extensively explored, with a growing body of evidence pointing to modulation of the glutamatergic system. The period of glutamatergic hypoactivation observed alongside long-term potentiation depression and cognitive deficits in later disease stages may be the consequence of a preceding period of increased glutamatergic activity. This review will explore the Aβ-related changes to the tripartite glutamate synapse resulting in altered cell signaling throughout disease progression, ultimately culminating in oxidative stress, synaptic dysfunction, and neuronal loss.
Collapse
Affiliation(s)
- Caleigh A Findley
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- 3 Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.,4 Department of Molecular Biology, Microbiology & Biochemistry, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
283
|
Tramadol: a Potential Neurotoxic Agent Affecting Prefrontal Cortices in Adult Male Rats and PC-12 Cell Line. Neurotox Res 2020; 38:385-397. [PMID: 32378056 DOI: 10.1007/s12640-020-00214-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 12/25/2022]
Abstract
Tramadol is a synthetic analogue of codeine that is often prescribed for the treatment of mild to moderate pains. It has a number of side effects including emotional instability and anxiety. In this study, we focus on the structural and functional changes of prefrontal cortex under chronic exposure to tramadol. At the cellular level, the amounts of ROS and annexin V in PC12 cells were evidently increased upon exposure to tramadol (at a concentration of 600 μM for 48 h). To this end, the rats were daily treated with tramadol at doses of 50 mg/kg for 3 weeks. Our findings reveal that tramadol provokes atrophy and apoptosis by the induction of apoptotic markers such as Caspase 3 and 8, pro-inflammatory markers, and downregulation of GDNF. Moreover, it triggers microgliosis and astrogliosis along with neuronal death in the prefrontal cortex. Behavioral disturbance and cognitive impairment are other side effects of tramadol. Overall, our results indicate tramadol-induced neurodegeneration in the prefrontal cortex mainly through activation of neuroinflammatory response.
Collapse
|
284
|
The gut microbiome regulates the increases in depressive-type behaviors and in inflammatory processes in the ventral hippocampus of stress vulnerable rats. Mol Psychiatry 2020; 25:1068-1079. [PMID: 30833676 DOI: 10.1038/s41380-019-0380-x] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 01/24/2019] [Accepted: 02/11/2019] [Indexed: 12/11/2022]
Abstract
Chronic exposure to stress is associated with increased incidence of depression, generalized anxiety, and PTSD. However, stress induces vulnerability to such disorders only in a sub-population of individuals, as others remain resilient. Inflammation has emerged as a putative mechanism for promoting stress vulnerability. Using a rodent model of social defeat, we have previously shown that rats with short-defeat latencies (SL/vulnerable rats) show increased anxiety- and depression-like behaviors, and these behaviors are mediated by inflammation in the ventral hippocampus. The other half of socially defeated rats show long-latencies to defeat (LL/resilient) and are similar to controls. Because gut microbiota are important activators of inflammatory substances, we assessed the role of the gut microbiome in mediating vulnerability to repeated social defeat stress. We analyzed the fecal microbiome of control, SL/vulnerable, and LL/resilient rats using shotgun metagenome sequencing and observed increased expression of immune-modulating microbiota, such as Clostridia, in SL/vulnerable rats. We then tested the importance of gut microbiota to the SL/vulnerable phenotype. In otherwise naive rats treated with microbiota from SL/vulnerable rats, there was higher microglial density and IL-1β expression in the vHPC, and higher depression-like behaviors relative to rats that received microbiota from LL/resilient rats, non-stressed control rats, or vehicle-treated rats. However, anxiety-like behavior during social interaction was not altered by transplant of the microbiome of SL/vulnerable rats into non-stressed rats. Taken together, the results suggest the gut microbiome contributes to the depression-like behavior and inflammatory processes in the vHPC of stress vulnerable individuals.
Collapse
|
285
|
Bjørklund G, Meguid NA, El-Bana MA, Tinkov AA, Saad K, Dadar M, Hemimi M, Skalny AV, Hosnedlová B, Kizek R, Osredkar J, Urbina MA, Fabjan T, El-Houfey AA, Kałużna-Czaplińska J, Gątarek P, Chirumbolo S. Oxidative Stress in Autism Spectrum Disorder. Mol Neurobiol 2020; 57:2314-2332. [PMID: 32026227 DOI: 10.1007/s12035-019-01742-2] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
According to the United States Centers for Disease Control and Prevention (CDC), as of July 11, 2016, the reported average incidence of children diagnosed with an autism spectrum disorder (ASD) was 1 in 68 (1.46%) among 8-year-old children born in 2004 and living within the 11 monitoring sites' surveillance areas in the United States of America (USA) in 2012. ASD is a multifaceted neurodevelopmental disorder that is also considered a hidden disability, as, for the most part; there are no apparent morphological differences between children with ASD and typically developing children. ASD is diagnosed based upon a triad of features including impairment in socialization, impairment in language, and repetitive and stereotypic behaviors. The increasing incidence of ASD in the pediatric population and the lack of successful curative therapies make ASD one of the most challenging disorders for medicine. ASD neurobiology is thought to be associated with oxidative stress, as shown by increased levels of reactive oxygen species and increased lipid peroxidation, as well as an increase in other indicators of oxidative stress. Children with ASD diagnosis are considered more vulnerable to oxidative stress because of their imbalance in intracellular and extracellular glutathione levels and decreased glutathione reserve capacity. Several studies have suggested that the redox imbalance and oxidative stress are integral parts of ASD pathophysiology. As such, early assessment and treatment of antioxidant status may result in a better prognosis as it could decrease the oxidative stress in the brain before it can induce more irreversible brain damage. In this review, many aspects of the role of oxidative stress in ASD are discussed, taking into account that the process of oxidative stress may be a target for therapeutic interventions.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, 8610, Mo i Rana, Norway.
| | - Nagwa A Meguid
- Research on Children with Special Needs Department, National Research Centre, Giza, Egypt
- CONEM Egypt Child Brain Research Group, National Research Center, Giza, Egypt
| | - Mona A El-Bana
- CONEM Egypt Child Brain Research Group, National Research Center, Giza, Egypt
- Medical Biochemistry Department, National Research Centre, Giza, Egypt
| | - Alexey A Tinkov
- Yaroslavl State University, Yaroslavl, Russia
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
- IM Sechenov First Moscow State Medical University, Moscow, Russia
| | - Khaled Saad
- Department of Pediatrics, Faculty of Medicine, Assiut University, Assiut, Egypt
- CONEM Upper Egypt Pediatric Research Group, Assiut University, Assiut, Egypt
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Maha Hemimi
- Research on Children with Special Needs Department, National Research Centre, Giza, Egypt
- CONEM Egypt Child Brain Research Group, National Research Center, Giza, Egypt
| | - Anatoly V Skalny
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
- IM Sechenov First Moscow State Medical University, Moscow, Russia
- Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia
- Taipei Medical University, Taipei, Taiwan
| | - Božena Hosnedlová
- CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic
- Faculty of Pharmacy, Department of Human Pharmacology and Toxicology, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic
| | - Rene Kizek
- CONEM Metallomics Nanomedicine Research Group (CMNRG), Brno, Czech Republic
- Faculty of Pharmacy, Department of Human Pharmacology and Toxicology, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic
| | - Joško Osredkar
- Institute of Clinical Chemistry and Biochemistry (KIKKB), Ljubljana University Medical Centre, Ljubljana, Slovenia
| | - Mauricio A Urbina
- Departamento de Zoología, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, Concepción, Chile
| | - Teja Fabjan
- Institute of Clinical Chemistry and Biochemistry (KIKKB), Ljubljana University Medical Centre, Ljubljana, Slovenia
| | - Amira A El-Houfey
- CONEM Upper Egypt Pediatric Research Group, Assiut University, Assiut, Egypt
- Department of Community Health Nursing, Faculty of Nursing, Assiut University, Assiut, Egypt
- Department of Community Health Nursing, Sabia University College, Jazan University, Jizan, Saudi Arabia
| | - Joanna Kałużna-Czaplińska
- Institute of General and Ecological Chemistry, Department of Chemistry, Technical University of Lodz, Lodz, Poland
- CONEM Poland Chemistry and Nutrition Research Group, Lodz University of Technology, Lodz, Poland
| | - Paulina Gątarek
- Institute of General and Ecological Chemistry, Department of Chemistry, Technical University of Lodz, Lodz, Poland
- CONEM Poland Chemistry and Nutrition Research Group, Lodz University of Technology, Lodz, Poland
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
- CONEM Scientific Secretary, Verona, Italy
| |
Collapse
|
286
|
Isaiah S, Loots DT, Solomons R, van der Kuip M, Tutu Van Furth AM, Mason S. Overview of Brain-to-Gut Axis Exposed to Chronic CNS Bacterial Infection(s) and a Predictive Urinary Metabolic Profile of a Brain Infected by Mycobacterium tuberculosis. Front Neurosci 2020; 14:296. [PMID: 32372900 PMCID: PMC7186443 DOI: 10.3389/fnins.2020.00296] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/16/2020] [Indexed: 12/12/2022] Open
Abstract
A new paradigm in neuroscience has recently emerged - the brain-gut axis (BGA). The contemporary focus in this paradigm has been gut → brain ("bottom-up"), in which the gut-microbiome, and its perturbations, affects one's psychological state-of-mind and behavior, and is pivotal in neurodegenerative disorders. The emerging brain → gut ("top-down") concept, the subject of this review, proposes that dysfunctional brain health can alter the gut-microbiome. Feedback of this alternative bidirectional highway subsequently aggravates the neurological pathology. This paradigm shift, however, focuses upon non-communicable neurological diseases (progressive neuroinflammation). What of infectious diseases, in which pathogenic bacteria penetrate the blood-brain barrier and interact with the brain, and what is this effect on the BGA in bacterial infection(s) that cause chronic neuroinflammation? Persistent immune activity in the CNS due to chronic neuroinflammation can lead to irreversible neurodegeneration and neuronal death. The properties of cerebrospinal fluid (CSF), such as immunological markers, are used to diagnose brain disorders. But what of metabolic markers for such purposes? If a BGA exists, then chronic CNS bacterial infection(s) should theoretically be reflected in the urine. The premise here is that chronic CNS bacterial infection(s) will affect the gut-microbiome and that perturbed metabolism in both the CNS and gut will release metabolites into the blood that are filtered (kidneys) and excreted in the urine. Here we assess the literature on the effects of chronic neuroinflammatory diseases on the gut-microbiome caused by bacterial infection(s) of the CNS, in the context of information attained via metabolomics-based studies of urine. Furthermore, we take a severe chronic neuroinflammatory infectious disease - tuberculous meningitis (TBM), caused by Mycobacterium tuberculosis, and examine three previously validated CSF immunological biomarkers - vascular endothelial growth factor, interferon-gamma and myeloperoxidase - in terms of the expected changes in normal brain metabolism. We then model the downstream metabolic effects expected, predicting pivotal altered metabolic pathways that would be reflected in the urinary profiles of TBM subjects. Our cascading metabolic model should be adjustable to account for other types of CNS bacterial infection(s) associated with chronic neuroinflammation, typically prevalent, and difficult to distinguish from TBM, in the resource-constrained settings of poor communities.
Collapse
Affiliation(s)
- Simon Isaiah
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Regan Solomons
- Department of Pediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Martijn van der Kuip
- Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Academic Medical Center, Emma Children’s Hospital, Amsterdam, Netherlands
| | - A. Marceline Tutu Van Furth
- Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Academic Medical Center, Emma Children’s Hospital, Amsterdam, Netherlands
| | - Shayne Mason
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
287
|
Phạm TL, Yin Y, Kwon HH, Shin N, Kim SI, Park H, Shin J, Shin HJ, Hwang JA, Song HJ, Kim SR, Lee JH, Hwang PTJ, Jun HW, Kim DW. miRNA 146a-5p-loaded poly(d,l-lactic-co-glycolic acid) nanoparticles impair pain behaviors by inhibiting multiple inflammatory pathways in microglia. Nanomedicine (Lond) 2020; 15:1113-1126. [PMID: 32292108 DOI: 10.2217/nnm-2019-0462] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aims: We investigated whether miRNA (miR) 146a-5p-loaded nanoparticles (NPs) can attenuate neuropathic pain behaviors in the rat spinal nerve ligation-induced neuropathic pain model by inhibiting activation of the NF-κB and p38 MAPK pathways in spinal microglia. Materials & methods: After NP preparation, miR NPs were assessed for their physical characteristics and then injected intrathecally into the spinal cords of rat spinal nerve ligation rats to test their analgesic effects. Results: miR NPs reduced pain behaviors for 11 days by negatively regulating the inflammatory response in spinal microglia. Conclusion: The anti-inflammatory effects of miR 146a-5p along with nanoparticle-based materials make miR NPs promising tools for treating neuropathic pain.
Collapse
Affiliation(s)
- Thuỳ Linh Phạm
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Yuhua Yin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anesthesia, Guangzhou Women and Children's Medical Center, Guangzhou, 510623, Guangdong Province, PR China
| | - Hyeok Hee Kwon
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Nara Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Song I Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyewon Park
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Jeong-Ah Hwang
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Hee-Jung Song
- Department of Neurology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Sang Ryong Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Institute of Life Science & Biotechnology, Brain Science & Engineering Institute, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Joo Hyoung Lee
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Patrick T J Hwang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ho-Wook Jun
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.,Department of Anatomy and Cell Biology, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| |
Collapse
|
288
|
Characterization of the SIM-A9 cell line as a model of activated microglia in the context of neuropathic pain. PLoS One 2020; 15:e0231597. [PMID: 32287325 PMCID: PMC7156095 DOI: 10.1371/journal.pone.0231597] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/26/2020] [Indexed: 12/21/2022] Open
Abstract
Resident microglia of the central nervous system are being increasingly recognized as key players in diseases such as neuropathic pain. Biochemical and behavioral studies in neuropathic pain rodent models have documented compelling evidence of the critical role of ATP mediated-P2X4R-brain-derived neurotrophic factor (BDNF) signaling pathway in the initiation and maintenance of pain hypersensitivity, a feature driving neuropathic pain-related behavior. The goal of this study was to develop and characterize an in vitro cell line model of activated microglia that can be subsequently utilized for screening neuropathic pain therapeutics. In the present study, we characterized the SIM-A9 microglia cell line for key molecules in the P2X4R-BDNF signaling axis using a combination of biochemical techniques and developed an ATP-activated SIM-A9 microglia model. We present three novel findings: first, SIM-A9 cells expressed P2X4R and BDNF proteins, second, ATP, but not LPS, was cytocompatible with SIM-A9 cells and third, exposure of cells to optimized ATP concentrations for defined periods increased intracellular expression of Iba1 and BDNF proteins. Increased Iba1 levels confirmed microglia activation and increased BDNF expression confirmed ATP-mediated stimulation of the P2X4R signaling pathway. We propose that this ATP-activated SIM-A9 cell line model system can be utilized for screening both small- as well as macro-molecular neuropathic pain therapeutics targeting BDNF and/or P2X4R knockdown.
Collapse
|
289
|
Investigation on the mechanism of mafenide in inhibiting pyroptosis and the release of inflammatory factors. Eur J Pharm Sci 2020; 147:105303. [DOI: 10.1016/j.ejps.2020.105303] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/19/2020] [Accepted: 03/08/2020] [Indexed: 11/19/2022]
|
290
|
Bhute S, Sarmah D, Datta A, Rane P, Shard A, Goswami A, Borah A, Kalia K, Dave KR, Bhattacharya P. Molecular Pathogenesis and Interventional Strategies for Alzheimer's Disease: Promises and Pitfalls. ACS Pharmacol Transl Sci 2020; 3:472-488. [PMID: 32566913 DOI: 10.1021/acsptsci.9b00104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a debilitating disorder characterized by age-related dementia, which has no effective treatment to date. β-Amyloid depositions and hyperphosphorylated tau proteins are the main pathological hallmarks, along with oxidative stress, N-methyl-d-aspartate (NMDA) receptor-mediated excitotoxicity, and low levels of acetylcholine. Current pharmacotherapy for AD only provides symptomatic relief and limited improvement in cognitive functions. Many molecules have been explored that show promising outcomes in AD therapy and can regulate cellular survival through different pathways. To have a vivid approach to strategize the treatment regimen, AD physiopathology should be better explained considering diverse etiological factors in conjunction with biochemical disturbances. This Review attempts to discuss different disease modification approaches and address the novel therapeutic targets of AD that might pave the way for new drug discovery using the well-defined targets for therapy of the disease.
Collapse
Affiliation(s)
- Shashikala Bhute
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Aishika Datta
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Pallavi Rane
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Avirag Goswami
- Department of Neurology, Albert Einstein Medical Center, Philadelphia, Pennsylvania 19141, United States
| | - Anupom Borah
- Department of Life Science and Bioinformatics, Assam University, Silchar, Assam-788011, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Kunjan R Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| |
Collapse
|
291
|
Ganbold T, Bao Q, Zandan J, Hasi A, Baigude H. Modulation of Microglia Polarization through Silencing of NF-κB p65 by Functionalized Curdlan Nanoparticle-Mediated RNAi. ACS APPLIED MATERIALS & INTERFACES 2020; 12:11363-11374. [PMID: 32073249 DOI: 10.1021/acsami.9b23004] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Microglia polarization plays an important role in poststroke recovery. Inhibition of proinflammatory (M1) polarization and promotion of anti-inflammatory (M2) polarization of microglia are potential therapeutic strategies for inflammation reduction and neuronal recovery after stroke. Here, we evaluated the central nervous system (CNS)-targeted short interfering RNA (siRNA) delivery ability of functionalized curdlan nanoparticles (CMI) and investigated the nuclear factor-κB (NF-κB) p65 silencing efficiency of CMI-mediated siRNA in microglia, as well as the resulting neuroprotective effect of microglia polarization and neuroprotection in vitro and in vivo. The systemic delivery of NF-κB p65 siRNA (sip65) complexed to CMI nanoparticles in the mouse model of transient middle cerebral artery occlusion (tMCAO) resulted in the distribution of siRNA in microglia and significant silencing in NF-κB p65 in the peri-infarct region. Knockdown of NF-κB p65 resulted in M1 to M2 phenotypic transition of microglia, evidenced by the change in the expression pattern of signature cytokines as well as inducible nitric oxide synthase and CD206. Moreover, the CMI-mediated silencing of p65 increased the density of neurons and decreased pyknosis and edema in the peri-infarct region. Assessment of the neurological deficit score on the Bederson scale revealed a significantly reduced score in the mouse model of tMCAO treated with the sip65/CMI complex. Collectively, our data suggest that CMI nanoparticles are a promising CNS-targeting siRNA delivery system, and NF-κB p65 may be a potential therapeutic target for inflammation reduction and poststroke recovery.
Collapse
Affiliation(s)
- Tsogzolmaa Ganbold
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P. R. China
- School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Hohhot, Inner Mongolia Autonomous Region 010020, PR China
| | - Qingming Bao
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P. R. China
| | - Jargalmaa Zandan
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P. R. China
| | - Agula Hasi
- School of Life Sciences, Inner Mongolia University, 24 Zhaojun Road, Hohhot, Inner Mongolia Autonomous Region 010020, PR China
| | - Huricha Baigude
- Institute of Mongolian Medicinal Chemistry, School of Chemistry & Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia 010020, P. R. China
| |
Collapse
|
292
|
de Araújo Boleti AP, de Oliveira Flores TM, Moreno SE, Anjos LD, Mortari MR, Migliolo L. Neuroinflammation: An overview of neurodegenerative and metabolic diseases and of biotechnological studies. Neurochem Int 2020; 136:104714. [PMID: 32165170 DOI: 10.1016/j.neuint.2020.104714] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/19/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
Neuroinflammation is an important factor contributing to cognitive impairment and neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), ischemic injury, and multiple sclerosis (MS). These diseases are characterized by inexorable progressive injury of neuron cells, and loss of motor or cognitive functions. Microglia, which are the resident macrophages in the brain, play an important role in both physiological and pathological conditions. In this review, we provide an updated discussion on the role of ROS and metabolic disease in the pathological mechanisms of activation of the microglial cells and release of cytotoxins, leading to the neurodegenerative process. In addition, we also discuss in vivo models, such as zebrafish and Caenorhabditis elegans, and provide new insights into therapeutics bioinspired by neuropeptides from venomous animals, supporting high throughput drug screening in the near future, searching for a complementary approach to elucidating crucial mechanisms associated with neurodegenerative disorders.
Collapse
Affiliation(s)
- Ana Paula de Araújo Boleti
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil
| | - Taylla Michelle de Oliveira Flores
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil; Programa de Pós-graduação em Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Susana Elisa Moreno
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil
| | - Lilian Dos Anjos
- Laboratório de Neurofarmacologia, Departmento Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brazil
| | - Márcia Renata Mortari
- Laboratório de Neurofarmacologia, Departmento Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brazil
| | - Ludovico Migliolo
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil; Programa de Pós-graduação em Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Brazil; Programa de Pós-graduação em Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, Brazil.
| |
Collapse
|
293
|
Cordaro M, Cuzzocrea S, Crupi R. An Update of Palmitoylethanolamide and Luteolin Effects in Preclinical and Clinical Studies of Neuroinflammatory Events. Antioxidants (Basel) 2020; 9:antiox9030216. [PMID: 32150935 PMCID: PMC7139331 DOI: 10.3390/antiox9030216] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 02/06/2023] Open
Abstract
The inflammation process represents of a dynamic series of phenomena that manifest themselves with an intense vascular reaction. Neuroinflammation is a reply from the central nervous system (CNS) and the peripheral nervous system (PNS) to a changed homeostasis. There are two cell systems that mediate this process: the glia of the CNS and the lymphocites, monocytes, and macrophages of the hematopoietic system. In both the peripheral and central nervous systems, neuroinflammation plays an important role in the pathogenesis of neurodegenerative diseases, such as Parkinson’s and Alzheimer’s diseases, and in neuropsychiatric illnesses, such as depression and autism spectrum disorders. The resolution of neuroinflammation is a process that allows for inflamed tissues to return to homeostasis. In this process the important players are represented by lipid mediators. Among the naturally occurring lipid signaling molecules, a prominent role is played by the N-acylethanolamines, namely N-arachidonoylethanolamine and its congener N-palmitoylethanolamine, which is also named palmitoylethanolamide or PEA. PEA possesses a powerful neuroprotective and anti-inflammatory power but has no antioxidant effects per se. For this reason, its co-ultramicronization with the flavonoid luteolin is more efficacious than either molecule alone. Inhibiting or modulating the enzymatic breakdown of PEA represents a complementary therapeutic approach to treating neuroinflammation. The aim of this review is to discuss the role of ultramicronized PEA and co-ultramicronized PEA with luteolin in several neurological diseases using preclinical and clinical approaches.
Collapse
Affiliation(s)
- Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98100 Messina, Italy;
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Via F. Stagno D’Alcontres 31, 98166 Messina, Italy;
- Department of Pharmacology and Physiology, Saint Louis University, St. Louis, MO 63103, USA
- Correspondence: ; Tel.: +390-906-765-208
| | - Rosalia Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Via F. Stagno D’Alcontres 31, 98166 Messina, Italy;
| |
Collapse
|
294
|
Hypothalamic NAD +-Sirtuin Axis: Function and Regulation. Biomolecules 2020; 10:biom10030396. [PMID: 32143417 PMCID: PMC7175325 DOI: 10.3390/biom10030396] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
The rapidly expanding elderly population and obesity endemic have become part of continuing global health care problems. The hypothalamus is a critical center for the homeostatic regulation of energy and glucose metabolism, circadian rhythm, and aging-related physiology. Nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase sirtuins are referred to as master metabolic regulators that link the cellular energy status to adaptive transcriptional responses. Mounting evidence now indicates that hypothalamic sirtuins are essential for adequate hypothalamic neuronal functions. Owing to the NAD+-dependence of sirtuin activity, adequate hypothalamic NAD+ contents are pivotal for maintaining energy homeostasis and circadian physiology. Here, we comprehensively review the regulatory roles of the hypothalamic neuronal NAD+-sirtuin axis in a normal physiological context and their changes in obesity and the aging process. We also discuss the therapeutic potential of NAD+ biology-targeting drugs in aging/obesity-related metabolic and circadian disorders.
Collapse
|
295
|
Arndtsen C, Ballon J, Blackshear K, Corbett CB, Lee K, Peyer J, Holloway KS, Duncan KA. Atypical gene expression of neuroinflammatory and steroid related genes following injury in the photoperiodic Japanese quail. Gen Comp Endocrinol 2020; 288:113361. [PMID: 31830471 DOI: 10.1016/j.ygcen.2019.113361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Clara Arndtsen
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY 12604, USA
| | - Jason Ballon
- Department of Biology, Vassar College, Poughkeepsie, NY 12604, USA
| | - Katie Blackshear
- Department of Biology, Vassar College, Poughkeepsie, NY 12604, USA
| | - Cali B Corbett
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY 12604, USA
| | - Kenneth Lee
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY 12604, USA
| | - Jordan Peyer
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY 12604, USA
| | - Kevin S Holloway
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY 12604, USA; Psychological Science, Vassar College, Poughkeepsie, NY 12604, USA
| | - Kelli A Duncan
- Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY 12604, USA; Department of Biology, Vassar College, Poughkeepsie, NY 12604, USA.
| |
Collapse
|
296
|
Wang H, Liu X, Tan C, Zhou W, Jiang J, Peng W, Zhou X, Mo L, Chen L. Bacterial, viral, and fungal infection-related risk of Parkinson's disease: Meta-analysis of cohort and case-control studies. Brain Behav 2020; 10:e01549. [PMID: 32017453 PMCID: PMC7066372 DOI: 10.1002/brb3.1549] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/24/2022] Open
Abstract
AIMS Recent studies showed that patients with various bacterial, viral, and fungal infections might be at increased risk of Parkinson's disease (PD). However, the risk of PD in patients with each specific infection varied. This meta-analysis estimated the association between various infections and PD risk. METHODS Literature published from January 1965 to October 2019 in PubMed and EMBASE databases was searched. Data were extracted and pooled using random/fixed effects model. Sensitivity analysis and meta-regression were also performed to analyze the source of heterogeneity. Publication bias was estimated by the trim and fill. RESULTS Twenty-three out of 6,609 studies were included. Helicobacter pylori (HP; pooled OR = 1.653, 1.426-1.915, p < .001), hepatitis C virus (HCV; pooled OR = 1.195, 1.012-1.410, p = .035), Malassezia (pooled OR = 1.694, 1.367-2.100, p < .001), and pneumoniae (pooled OR = 1.595, 1.020-2.493, p = .041) infection were associated with increased PD risk. Influenza virus, herpes virus, hepatitis B virus, scarlet fever, mumps virus, chicken pox, pertussis, German measles, and measles were not associated with PD risk. After antiviral treatment against HCV reduced the risk of PD in patients with HCV infection (OR = 0.672, 0.571-0.791, p < .001). Significant heterogeneity exists among the included studies. CONCLUSION Patients with infection of HP, HCV, Malassezia, pneumoniae might be an increased risk of PD. Antiviral treatment of HCV could reduce the risk of PD.
Collapse
Affiliation(s)
- Hui Wang
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xi Liu
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Changhong Tan
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Wen Zhou
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jin Jiang
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Wuxue Peng
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xuan Zhou
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Lijuan Mo
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Lifen Chen
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
297
|
Neuroinflammation in CNS diseases: Molecular mechanisms and the therapeutic potential of plant derived bioactive molecules. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100176] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
298
|
Joffre C, Dinel AL, Chataigner M, Pallet V, Layé S. n-3 Polyunsaturated Fatty Acids and Their Derivates Reduce Neuroinflammation during Aging. Nutrients 2020; 12:nu12030647. [PMID: 32121189 PMCID: PMC7146513 DOI: 10.3390/nu12030647] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/15/2022] Open
Abstract
: Aging is associated to cognitive decline, which can lead to loss of life quality, personal suffering, and ultimately neurodegenerative diseases. Neuroinflammation is one of the mechanisms explaining the loss of cognitive functions. Indeed, aging is associated to the activation of inflammatory signaling pathways, which can be targeted by specific nutrients with anti-inflammatory effects. Dietary n-3 polyunsaturated fatty acids (PUFAs) are particularly attractive as they are present in the brain, possess immunomodulatory properties, and are precursors of lipid derivates named specialized pro-resolving mediators (SPM). SPMs are crucially involved in the resolution of inflammation that is modified during aging, resulting in chronic inflammation. In this review, we first examine the effect of aging on neuroinflammation and then evaluate the potential beneficial effect of n-3 PUFA as precursors of bioactive derivates, particularly during aging, on the resolution of inflammation. Lastly, we highlight evidence supporting a role of n-3 PUFA during aging.
Collapse
Affiliation(s)
- Corinne Joffre
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.C.); (V.P.); (S.L.)
- Correspondence:
| | - Anne-Laure Dinel
- NutriBrain Research and Technology Transfer, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Mathilde Chataigner
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.C.); (V.P.); (S.L.)
- Abyss Ingredients, 56850 Caudan, France
| | - Véronique Pallet
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.C.); (V.P.); (S.L.)
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France; (M.C.); (V.P.); (S.L.)
| |
Collapse
|
299
|
An Y, Zhang H, Huang S, Pei G. PL201, a Reported Rhamnoside Against Alzheimer's Disease Pathology, Alleviates Neuroinflammation and Stimulates Nrf2 Signaling. Front Immunol 2020; 11:162. [PMID: 32174909 PMCID: PMC7056876 DOI: 10.3389/fimmu.2020.00162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/21/2020] [Indexed: 01/14/2023] Open
Abstract
Neuroinflammation induced by overactivated glia cells is believed to be a major hallmark of Alzheimer's disease (AD) and a hopeful target against AD. A rhamnoside PL201 was previously reported to promote neurogenesis and ameliorate AD, and in this study, we revealed that PL201 also significantly reduced accumulation of the activated microglia and proinflammatory cytokines in APP/PS1 mice. In vitro, PL201 consistently suppressed the microglia induction of proinflammatory cytokines after stimulation with lipopolysaccharides and Aβ42. Further mechanistic studies demonstrated that PL201 considerably enhanced the expression level and the nuclear translocation of Nrf2, a key regulator of neuroinflammation. Moreover, PL201 effectively stimulated Nrf2 signaling cascade, including upregulation of HO-1 and downregulation of NF-κB pathway. Thus, our findings indicated the anti-neuroinflammatory effect by PL201 in vivo and suggested that PL201 or the like, with multiple functions such as neurogenesis, mitochondria maintenance, and anti-neuroinflammation, could be a promising candidate in AD treatment.
Collapse
Affiliation(s)
- Yuqian An
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hong Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Shichao Huang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Gang Pei
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,Shanghai Key Laboratory of Signaling and Disease Research, Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
300
|
Paudel YN, Angelopoulou E, Semple B, Piperi C, Othman I, Shaikh MF. Potential Neuroprotective Effect of the HMGB1 Inhibitor Glycyrrhizin in Neurological Disorders. ACS Chem Neurosci 2020; 11:485-500. [PMID: 31972087 DOI: 10.1021/acschemneuro.9b00640] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glycyrrhizin (glycyrrhizic acid), a bioactive triterpenoid saponin constituent of Glycyrrhiza glabra, is a traditional medicine possessing a plethora of pharmacological anti-inflammatory, antioxidant, antimicrobial, and antiaging properties. It is a known pharmacological inhibitor of high mobility group box 1 (HMGB1), a ubiquitous protein with proinflammatory cytokine-like activity. HMGB1 has been implicated in an array of inflammatory diseases when released extracellularly, mainly by activating intracellular signaling upon binding to the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4). HMGB1 neutralization strategies have demonstrated disease-modifying outcomes in several preclinical models of neurological disorders. Herein, we reveal the potential neuroprotective effects of glycyrrhizin against several neurological disorders. Emerging findings demonstrate the therapeutic potential of glycyrrhizin against several HMGB1-mediated pathological conditions including traumatic brain injury, neuroinflammation and associated conditions, epileptic seizures, Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Glycyrrhizin's effects in neurological disorders are mainly attributed to the attenuation of neuronal damage by inhibiting HMGB1 expression and translocation as well as by downregulating the expression of inflammatory cytokines. A large number of preclinical findings supports the notion that glycyrrhizin might be a promising therapeutic alternative to overcome the shortcomings of the mainstream therapeutic strategies against neurological disorders, mainly by halting disease progression. However, future research is warranted for a deeper exploration of the precise underlying molecular mechanism as well as for clinical translation.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Bridgette Semple
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| |
Collapse
|