101
|
Uddin MS, Hasana S, Ahmad J, Hossain MF, Rahman MM, Behl T, Rauf A, Ahmad A, Hafeez A, Perveen A, Ashraf GM. Anti-Neuroinflammatory Potential of Polyphenols by Inhibiting NF-κB to Halt Alzheimer's Disease. Curr Pharm Des 2021; 27:402-414. [PMID: 33213314 DOI: 10.2174/1381612826666201118092422] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/01/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is an irrevocable chronic brain disorder featured by neuronal loss, microglial accumulation, and progressive cognitive impairment. The proper pathophysiology of this life-threatening disorder is not completely understood and no exact remedies have been found yet. Over the last few decades, research on AD has mainly highlighted pathomechanisms linked to a couple of the major pathological hallmarks, including extracellular senile plaques made of amyloid-β (Aβ) peptides, and intracellular neurofibrillary tangles (NFTs) made of tau proteins. Aβ can induce apoptosis, trigger an inflammatory response, and inhibit the synaptic plasticity of the hippocampus, which ultimately contributes to reducing cognitive functions and memory impairment. Recently, a third disease hallmark, the neuroinflammatory reaction that is mediated by cerebral innate immune cells, has become a spotlight in the current research area, assured by pre-clinical, clinical, and genetic investigations. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), a cytokine producer, is significantly associated with physiological inflammatory proceedings and thus shows a promising candidate for inflammation- based AD therapy. Recent data reveal that phytochemicals, mainly polyphenol compounds, exhibit potential neuroprotective functions and these may be considered as a vital resource for discovering several drug candidates against AD. Interestingly, phytochemicals can easily interfere with the signaling pathway of NF-κB. This review represents the anti-neuroinflammatory potential of polyphenols as inhibitors of NF-κB to combat AD pathogenesis.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Sharifa Hasana
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Jamil Ahmad
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | | | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Khyber Pakhtunkhwa, Pakistan
| | - Ausaf Ahmad
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Uttar Pradesh, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
102
|
Tyrtyshnaia A, Bondar A, Konovalova S, Sultanov R, Manzhulo I. N-Docosahexanoylethanolamine Reduces Microglial Activation and Improves Hippocampal Plasticity in a Murine Model of Neuroinflammation. Int J Mol Sci 2020; 21:ijms21249703. [PMID: 33352646 PMCID: PMC7767308 DOI: 10.3390/ijms21249703] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/16/2022] Open
Abstract
Chronic neuroinflammation is a common pathogenetic link in the development of various neurological and neurodegenerative diseases. Thus, a detailed study of neuroinflammation and the development of drugs that reduce or eliminate the negative effect of neuroinflammation on cognitive processes are among the top priorities of modern neurobiology. N-docosahexanoylethanolamine (DHEA, synaptamide) is an endogenous metabolite and structural analog of anandamide, an essential endocannabinoid produced from arachidonic acid. Our study aims to elucidate the pharmacological activity of synaptamide in lipopolysaccharide (LPS)-induced neuroinflammation. Memory deficits in animals were determined using behavioral tests. To study the effects of LPS (750 µg/kg/day, 7 days) and synaptamide (10 mg/kg/day, 7 days) on synaptic plasticity, long-term potentiation was examined in the CA1 area of acute hippocampal slices. The Golgi-Cox method allowed us to assess neuronal morphology. The production of inflammatory factors and receptors was assessed using ELISA and immunohistochemistry. During the study, functional, structural, and plastic changes within the hippocampus were identified. We found a beneficial effect of synaptamide on hippocampal synaptic plasticity and morphological characteristics of neurons. Synaptamide treatment recovered hippocampal neurogenesis, suppressed microglial activation, and significantly improved hippocampus-dependent memory. The basis of the phenomena described above is probably the powerful anti-inflammatory activity of synaptamide, as shown in our study and several previous works.
Collapse
|
103
|
Hepp Rehfeldt SC, Majolo F, Goettert MI, Laufer S. c-Jun N-Terminal Kinase Inhibitors as Potential Leads for New Therapeutics for Alzheimer's Diseases. Int J Mol Sci 2020; 21:E9677. [PMID: 33352989 PMCID: PMC7765872 DOI: 10.3390/ijms21249677] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's Disease (AD) is becoming more prevalent as the population lives longer. For individuals over 60 years of age, the prevalence of AD is estimated at 40.19% across the world. Regarding the cognitive decline caused by the disease, mitogen-activated protein kinases (MAPK) pathways such as the c-Jun N-terminal kinase (JNK) pathway are involved in the progressive loss of neurons and synapses, brain atrophy, and augmentation of the brain ventricles, being activated by synaptic dysfunction, oxidative stress, and excitotoxicity. Nowadays, AD symptoms are manageable, but the disease itself remains incurable, thus the inhibition of JNK3 has been explored as a possible therapeutic target, considering that JNK is best known for its involvement in propagating pro-apoptotic signals. This review aims to present biological aspects of JNK, focusing on JNK3 and how it relates to AD. It was also explored the recent development of inhibitors that could be used in AD treatment since several drugs/compounds in phase III clinical trials failed. General aspects of the MAPK family, therapeutic targets, and experimental treatment in models are described and discussed throughout this review.
Collapse
Affiliation(s)
- Stephanie Cristine Hepp Rehfeldt
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado CEP 95914-014, Rio Grande do Sul, Brazil; (S.C.H.R.); (F.M.)
| | - Fernanda Majolo
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado CEP 95914-014, Rio Grande do Sul, Brazil; (S.C.H.R.); (F.M.)
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre CEP 90619-900, Rio Grande do Sul, Brazil
| | - Márcia Inês Goettert
- Graduate Program in Biotechnology, University of Vale do Taquari (Univates), Lajeado CEP 95914-014, Rio Grande do Sul, Brazil; (S.C.H.R.); (F.M.)
| | - Stefan Laufer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmaceutical Sciences, Faculty of Sciences, University of Tuebingen, D-72076 Tuebingen, Germany
| |
Collapse
|
104
|
Feng YS, Tan ZX, Wu LY, Dong F, Zhang F. The involvement of NLRP3 inflammasome in the treatment of Alzheimer's disease. Ageing Res Rev 2020; 64:101192. [PMID: 33059089 DOI: 10.1016/j.arr.2020.101192] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/04/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases, and it is characterised by progressive deterioration in cognitive and memory abilities, which can severely influence the elderly population's daily living abilities. Although researchers have made great efforts in the field of AD, there are still no well-established strategies to prevent and treat this disease. Therefore, better clarification of the molecular mechanisms associated with the onset and progression of AD is critical to provide a theoretical basis for the establishment of novel preventive and therapeutic strategies. Currently, it is generally believed that neuroinflammation plays a key role in the pathogenesis of AD. Inflammasome, a multiprotein complex, is involved in the innate immune system, and it can mediate inflammatory responses and pyroptosis, which lead to neurodegeneration. Among the various types of inflammasomes, the NLRP3 inflammasome is the most characterised in neurodegenerative diseases, especially in AD. The activation of the NLRP3 inflammasome causes the generation of caspase-1-mediated interleukin (IL)-1β and IL-18 in microglia cells, where neuroinflammation is involved in the development and progression of AD. Thus, the NLRP3 inflammasome is likely to be a crucial therapeutic molecular target for AD via regulating neuroinflammation. In this review, we summarise the current knowledge on the role and regulatory mechanisms of the NLRP3 inflammasome in the pathogenic mechanisms of AD. We also focus on a series of potential therapeutic treatments targeting NLRP3 inflammasome for AD. Further clarification of the regulatory mechanisms of the NLRP3 inflammasome in AD may provide more useful clues to develop novel AD treatment strategies.
Collapse
|
105
|
Gong P, Chen YQ, Lin AH, Zhang HB, Zhang Y, Ye RD, Yu Y. p47 phox deficiency improves cognitive impairment and attenuates tau hyperphosphorylation in mouse models of AD. ALZHEIMERS RESEARCH & THERAPY 2020; 12:146. [PMID: 33183342 PMCID: PMC7659091 DOI: 10.1186/s13195-020-00714-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/22/2020] [Indexed: 01/02/2023]
Abstract
Background Alzheimer’s disease (AD) is characterized by progressive memory loss and cognitive impairment. The aggregation of amyloid β (Aβ) and hyperphosphorylated tau protein are two major pathological features of AD. Nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase, NOX) has been indicated in Aβ pathology; however, whether and how it affects tau pathology are not yet clear. Methods The role of NOX2 in cognitive function, amyloid plaque formation, and tau hyperphosphorylation were examined in APP/PS1 transgenic mice mated with p47phox-deficient mice (with deletion of the gene of neutrophil cytosolic factor 1, Ncf1) and/or in p47phox-deficient mice receiving intracerebroventricular (ICV) injection of streptozotocin (STZ). The cognitive and non-cognitive functions in these mice were assessed by Morris water maze, Rotarod test, open field, and elevated plus maze. Aβ levels, amyloid plaques, p47phox expression, and astrocyte activation were evaluated using immunofluorescence staining, ELISA, and/or Western blotting. Cultured primary neuronal cells were treated with okadaic acid or conditioned media (CM) from high glucose-stimulated primary astrocytes. The alteration in tau pathology was determined using Western blotting and immunofluorescence staining. Results Deletion of the gene coding for p47phox, the organizer subunit of NOX2, significantly attenuated cognitive impairment and tau pathology in these mice. p47phox deficiency decreased the activation of astrocytes but had no effect on Aβ levels and amyloid plaque formation in the brains of aged APP/PS1 mice, which displayed markedly increased expression of p47phox in neurons and astrocytes. Cell culture studies found that neuronal p47phox deletion attenuated okadaic acid-induced tau hyperphosphorylation at specific sites in primary cultures of neurons. CM from high glucose-treated WT astrocytes increased tau hyperphosphorylation in primary neurons, whereas this effect was absent from p47phox-deficient astrocytes. Conclusions These results suggest that p47phox is associated with cognitive function and tau pathology in AD. p47phox expressed in neurons contributes to tau hyperphosphorylation directly, while p47phox in astrocytes affect tau hyperphosphorylation by activating astrocytes indirectly. Our results provide new insights into the role of NOX2 in AD and indicate that targeted inhibition of p47phox may be a new strategy for the treatment of AD.
Collapse
Affiliation(s)
- Ping Gong
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan-Qing Chen
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ai-Hua Lin
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hai-Bo Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yan Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Richard D Ye
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, 518172, China.
| | - Yang Yu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
106
|
Qu Z, Sun J, Zhang W, Yu J, Zhuang C. Transcription factor NRF2 as a promising therapeutic target for Alzheimer's disease. Free Radic Biol Med 2020; 159:87-102. [PMID: 32730855 DOI: 10.1016/j.freeradbiomed.2020.06.028] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022]
Abstract
Oxidative stress is considered as one of the pathogenesis of Alzheimer's disease (AD) and plays an important role in the occurrence and development of AD. Nuclear factor erythroid 2 related factor 2 (NRF2) is a key regulatory of oxidative stress defence. There is growing evidence indicating the relationship between NRF2 and AD. NRF2 activation mitigates multiple pathogenic processes involved in AD by upregulating antioxidative defense, inhibiting neuroinflammation, improving mitochondrial function, maintaining proteostasis, and inhibiting ferroptosis. In addition, several NRF2 activators are currently being evaluated as AD therapeutic agents in clinical trials. Thus, targeting NRF2 has been the focus of a new strategy for prevention and treatment of AD. In this review, the role of NRF2 in AD and the NRF2 activators advanced into clinical and preclinical studies will be summarized.
Collapse
Affiliation(s)
- Zhuo Qu
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China
| | - Jiachen Sun
- School of Biotechnology and Food Science, Tianjin University of Commerce, 409 Guangrong Road, Tianjin, 300134, China
| | - Wannian Zhang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China
| | - Jianqiang Yu
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China.
| | - Chunlin Zhuang
- School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, China; School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| |
Collapse
|
107
|
Perea JR, Bolós M, Avila J. Microglia in Alzheimer's Disease in the Context of Tau Pathology. Biomolecules 2020; 10:biom10101439. [PMID: 33066368 PMCID: PMC7602223 DOI: 10.3390/biom10101439] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/09/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022] Open
Abstract
Microglia are the cells that comprise the innate immune system in the brain. First described more than a century ago, these cells were initially assigned a secondary role in the central nervous system (CNS) with respect to the protagonists, neurons. However, the latest advances have revealed the complexity and importance of microglia in neurodegenerative conditions such as Alzheimer’s disease (AD), the most common form of dementia associated with aging. This pathology is characterized by the accumulation of amyloid-β peptide (Aβ), which forms senile plaques in the neocortex, as well as by the aggregation of hyperphosphorylated tau protein, a process that leads to the development of neurofibrillary tangles (NFTs). Over the past few years, efforts have been focused on studying the interaction between Aβ and microglia, together with the ability of the latter to decrease the levels of this peptide. Given that most clinical trials following this strategy have failed, current endeavors focus on deciphering the molecular mechanisms that trigger the tau-induced inflammatory response of microglia. In this review, we summarize the most recent studies on the physiological and pathological functions of tau protein and microglia. In addition, we analyze the impact of microglial AD-risk genes (APOE, TREM2, and CD33) in tau pathology, and we discuss the role of extracellular soluble tau in neuroinflammation.
Collapse
Affiliation(s)
- Juan Ramón Perea
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 1 Nicolás Cabrera, 28049 Madrid, Spain; (J.R.P.); (M.B.)
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), 5 Valderrebollo, 28031 Madrid, Spain
| | - Marta Bolós
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 1 Nicolás Cabrera, 28049 Madrid, Spain; (J.R.P.); (M.B.)
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), 5 Valderrebollo, 28031 Madrid, Spain
| | - Jesús Avila
- Department of Molecular Neuropathology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 1 Nicolás Cabrera, 28049 Madrid, Spain; (J.R.P.); (M.B.)
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), 5 Valderrebollo, 28031 Madrid, Spain
- Correspondence: ; Tel.:+34-196-4564
| |
Collapse
|
108
|
Gritsenko A, Green JP, Brough D, Lopez-Castejon G. Mechanisms of NLRP3 priming in inflammaging and age related diseases. Cytokine Growth Factor Rev 2020; 55:15-25. [PMID: 32883606 PMCID: PMC7571497 DOI: 10.1016/j.cytogfr.2020.08.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023]
Abstract
The NLRP3 inflammasome is a vital part of the innate immune response, whilst its aberrant activation drives the progression of a number of non-communicable diseases. Thus, NLRP3 inflammasome assembly must be tightly controlled at several checkpoints. The priming step of NLRP3 inflammasome activation is associated with increased NLRP3 gene expression, as well as post-translational modifications that control NLRP3 levels and licence the NLRP3 protein for inflammasome assembly. Increasing life expectancy in modern society is accompanied by a growing percentage of elderly individuals. The process of aging is associated with chronic inflammation that drives and/or worsens a range of age related non-communicable conditions. The NLRP3 inflammasome is known to contribute to pathological inflammation in many settings, but the mechanisms that prime NLRP3 for activation throughout aging and related co-morbidities have not been extensively reviewed. Here we dissect the biochemical changes that occur during aging and the pathogenesis of age related diseases and analyse the mechanisms by which they prime the NLRP3 inflammasome, thus exacerbating inflammation.
Collapse
Affiliation(s)
- Anna Gritsenko
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jack P Green
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - David Brough
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Gloria Lopez-Castejon
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
109
|
O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci 2020; 14:187. [PMID: 32792908 PMCID: PMC7390932 DOI: 10.3389/fncel.2020.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Following an injury to the central nervous system (CNS), spontaneous plasticity is observed throughout the neuraxis and affects multiple key circuits. Much of this spontaneous plasticity can elicit beneficial and deleterious functional outcomes, depending on the context of plasticity and circuit affected. Injury-induced activation of the neuroimmune system has been proposed to be a major factor in driving this plasticity, as neuroimmune and inflammatory factors have been shown to influence cellular, synaptic, structural, and anatomical plasticity. Here, we will review the mechanisms through which the neuroimmune system mediates plasticity after CNS injury. Understanding the role of specific neuroimmune factors in driving adaptive and maladaptive plasticity may offer valuable therapeutic insight into how to promote adaptive plasticity and/or diminish maladaptive plasticity, respectively.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
110
|
Michalska P, Mayo P, Fernández-Mendívil C, Tenti G, Duarte P, Buendia I, Ramos MT, López MG, Menéndez JC, León R. Antioxidant, Anti-inflammatory and Neuroprotective Profiles of Novel 1,4-Dihydropyridine Derivatives for the Treatment of Alzheimer's Disease. Antioxidants (Basel) 2020; 9:antiox9080650. [PMID: 32708053 PMCID: PMC7463999 DOI: 10.3390/antiox9080650] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease is a chronic and irreversible pathological process that has become the most prevalent neurodegenerative disease. Currently, it is considered a multifactorial disease where oxidative stress and chronic neuroinflammation play a crucial role in its onset and development. Its characteristic neuronal loss has been related to the formation of neurofibrillary tangles mainly composed by hyperphosphorylated tau protein. Hyperphosphorylation of tau protein is related to the over-activity of GSK-3β, a kinase that participates in several pathological mechanisms including neuroinflammation. Neuronal loss is also related to cytosolic Ca2+ homeostasis dysregulation that triggers apoptosis and free radicals production, contributing to oxidative damage and, finally, neuronal death. Under these premises, we have obtained a new family of 4,7-dihydro-2H-pyrazolo[3–b]pyridines as multitarget directed ligands showing potent antioxidant properties and able to scavenge both oxygen and nitrogen radical species, and also, with anti-inflammatory properties. Further characterization has demonstrated their capacity to inhibit GSK-3β and to block L-type voltage dependent calcium channels. Novel derivatives have also demonstrated an interesting neuroprotective profile on in vitro models of neurodegeneration. Finally, compound 4g revokes cellular death induced by tau hyperphosphorylation in hippocampal slices by blocking reactive oxygen species (ROS) production. In conclusion, the multitarget profile exhibited by these compounds is a novel therapeutic strategy of potential interest in the search of novel treatments for Alzheimer’s disease.
Collapse
Affiliation(s)
- Patrycja Michalska
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.); (P.M.); (C.F.-M.); (P.D.); (I.B.); (M.G.L.)
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Paloma Mayo
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.); (P.M.); (C.F.-M.); (P.D.); (I.B.); (M.G.L.)
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Cristina Fernández-Mendívil
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.); (P.M.); (C.F.-M.); (P.D.); (I.B.); (M.G.L.)
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Giammarco Tenti
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (G.T.); (M.T.R.); (J.C.M.)
| | - Pablo Duarte
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.); (P.M.); (C.F.-M.); (P.D.); (I.B.); (M.G.L.)
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - Izaskun Buendia
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.); (P.M.); (C.F.-M.); (P.D.); (I.B.); (M.G.L.)
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - María Teresa Ramos
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (G.T.); (M.T.R.); (J.C.M.)
| | - Manuela G. López
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.); (P.M.); (C.F.-M.); (P.D.); (I.B.); (M.G.L.)
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - J. Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (G.T.); (M.T.R.); (J.C.M.)
| | - Rafael León
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (P.M.); (P.M.); (C.F.-M.); (P.D.); (I.B.); (M.G.L.)
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
- Correspondence: ; Tel.: +34-914-972-766
| |
Collapse
|
111
|
Cervellati C, Trentini A, Pecorelli A, Valacchi G. Inflammation in Neurological Disorders: The Thin Boundary Between Brain and Periphery. Antioxid Redox Signal 2020; 33:191-210. [PMID: 32143546 DOI: 10.1089/ars.2020.8076] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Accumulating evidence suggests that inflammation is a major contributor in the pathogenesis of several highly prevalent, but also rare, neurological diseases. In particular, the neurodegenerative processes of Alzheimer's disease (AD), vascular dementia (VAD), Parkinson's disease (PD), and multiple sclerosis (MS) are fueled by neuroinflammation, which, in turn, is accompanied by a parallel systemic immune dysregulation. This cross-talk between periphery and the brain becomes substantial when the blood-brain barrier loses its integrity, as often occurs in the course of these diseases. It has been hypothesized that the perpetual bidirectional flux of inflammatory mediators is not a mere "static" collateral effect of the neurodegeneration, but represents a proactive phenomenon sparking and driving the neuropathological processes. However, the upstream/downstream relationship between inflammatory events and neurological pathology is still unclear. Recent Advances: Solid recent evidence clearly suggests that metabolic factors, systemic infections, Microbiota dysbiosis, and oxidative stress are implicated, although to a different extent, in the development in brain diseases. Critical Issues: Here, we reviewed the most solid published evidence supporting the implication of the axis systemic inflammation-neuroinflammation-neurodegeneration in the pathogenesis of AD, VAD, PD, and MS, highlighting the possible cause of the putative downstream component of the axis. Future Directions: Reaching a definitive clinical/epidemiological appreciation of the etiopathogenic significance of the connection between peripheral and brain inflammation in neurologic disorders is pivotal since it could open novel therapeutic avenues for these diseases.
Collapse
Affiliation(s)
- Carlo Cervellati
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandro Trentini
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Alessandra Pecorelli
- Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina, USA
| | - Giuseppe Valacchi
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy.,Animal Science Department, Plants for Human Health Institute, NC State University, Kannapolis, North Carolina, USA.,Department of Food and Nutrition, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
112
|
Zhang H, Jia L, Jia J. Oxiracetam Offers Neuroprotection by Reducing Amyloid β-Induced Microglial Activation and Inflammation in Alzheimer's Disease. Front Neurol 2020; 11:623. [PMID: 32765394 PMCID: PMC7380077 DOI: 10.3389/fneur.2020.00623] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/28/2020] [Indexed: 01/06/2023] Open
Abstract
Background: Alzheimer's disease (AD) is characterized by amyloid beta (Aβ) accumulation in the brain, which triggers the activation of microglia; in turn, microglia release neuroinflammatory factors capable of damaging neurons. Thus, a therapeutic approach targeting this sustained microglia-induced inflammatory response deserves investigation. Here, we examined whether oxiracetam (ORC), a nootropic of the racetam family, can indirectly prevent Aβ-induced neurotoxicity by attenuating microglial activation. Methods: Aβ42 oligomers were used to stimulate BV2 microglial cells, and the morphological changes and phagocytic capacity of BV2 cells were evaluated using fluorescence microscopy. We used quantitative reverse transcription polymerase chain reaction to assess the inhibitory effects of ORC on Aβ-induced mRNA levels of interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α); enzyme-linked immunosorbent assay was used to examine the productions of these cytokines. We also assessed the mRNA level of inducible nitric oxide synthase and the production of nitric oxide (NO). The conditioned medium from BV2 cells was used to culture hippocampal HT22 cells to assess indirect toxicity using the MTT assay. Results: ORC prevented the Aβ-induced activation of BV2 cells, as reflected by reduced morphological changes and phagocytic ability. In addition, ORC downregulated the expression of Aβ-induced cytokines (IL-1β, IL-6, and TNF-α) and the production of NO in BV2 cells. Furthermore, ORC protected HT22 cells from indirect damage evoked by Aβ-treated BV2 cell-conditioned medium, but not from direct Aβ-induced toxicity. Conclusions: ORC suppressed the activation of BV2 cells, decreased the production of Aβ-induced inflammatory molecules and NO in BV2 cells, and protected HT22 cells against indirect toxicity mediated by Aβ-treated BV2 cell-conditioned medium. Thus, ORC may exert a protective role in AD through attenuating the damage caused by inflammation and oxidative stress.
Collapse
Affiliation(s)
- Heng Zhang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
113
|
Cherry JD, Babcock KJ, Goldstein LE. Repetitive Head Trauma Induces Chronic Traumatic Encephalopathy by Multiple Mechanisms. Semin Neurol 2020; 40:430-438. [PMID: 32674181 DOI: 10.1055/s-0040-1713620] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Exposure to repetitive neurotrauma increases lifetime risk for developing progressive cognitive deficits, neurobehavioral abnormalities, and chronic traumatic encephalopathy (CTE). CTE is a tau protein neurodegenerative disease first identified in boxers and recently described in athletes participating in other contact sports (notably American football, ice hockey, rugby, and wrestling) and in military veterans with blast exposure. Currently, CTE can only be diagnosed by neuropathological examination of the brain after death. The defining diagnostic lesion of CTE consists of patchy perivascular accumulations of hyperphosphorylated tau protein that localize in the sulcal depths of the cerebral cortex. Neuronal abnormalities, axonopathy, neurovascular dysfunction, and neuroinflammation are triggered by repetitive head impacts (RHIs) and likely act as catalysts for CTE pathogenesis and progression. However, the specific mechanisms that link RHI to CTE are unknown. This review will explore two important areas of CTE pathobiology. First, we will review what is known about the biomechanical properties of RHI that initiate CTE-related pathologies. Second, we will provide an overview of key features of CTE neuropathology and how these contribute to abnormal tau hyperphosphorylation, accumulation, and spread.
Collapse
Affiliation(s)
- Jonathan D Cherry
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts.,Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts.,VA Boston Healthcare System, Boston, Massachusetts
| | - Katharine J Babcock
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts.,VA Boston Healthcare System, Boston, Massachusetts.,Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts.,Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, Massachusetts
| | - Lee E Goldstein
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts.,Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts.,Boston University College of Engineering, Boston University, Boston, Massachusetts
| |
Collapse
|
114
|
Fagiani F, Lanni C, Racchi M, Govoni S. Targeting dementias through cancer kinases inhibition. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12044. [PMID: 32671184 PMCID: PMC7341824 DOI: 10.1002/trc2.12044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
The failures in Alzheimer's disease (AD) therapy strongly suggest the importance of reconsidering the research strategies analyzing other mechanisms that may take place in AD as well as, in general, in other neurodegenerative dementias. Taking into account that in AD a variety of defects result in neurotransmitter activity and signaling efficiency imbalance, neuronal cell degeneration and defects in damage/repair systems, aberrant and abortive cell cycle, glial dysfunction, and neuroinflammation, a target may be represented by the intracellular signaling machinery provided by the kinome. In particular, based on the observations of a relationship between cancer and AD, we focused on cancer kinases for targeting neurodegeneration, highlighting the importance of targeting the intracellular pathways at the intersection between cell metabolism control/duplication, the inhibition of which may stop a progression in neurodegeneration.
Collapse
Affiliation(s)
- Francesca Fagiani
- Department of Drug Sciences (Pharmacology Section)University of PaviaPaviaItaly
- Scuola Universitaria Superiore IUSS PaviaPaviaItaly
| | - Cristina Lanni
- Department of Drug Sciences (Pharmacology Section)University of PaviaPaviaItaly
| | - Marco Racchi
- Department of Drug Sciences (Pharmacology Section)University of PaviaPaviaItaly
| | - Stefano Govoni
- Department of Drug Sciences (Pharmacology Section)University of PaviaPaviaItaly
| |
Collapse
|
115
|
Stuve O, Weideman RA, McMahan DM, Jacob DA, Little BB. Diclofenac reduces the risk of Alzheimer's disease: a pilot analysis of NSAIDs in two US veteran populations. Ther Adv Neurol Disord 2020; 13:1756286420935676. [PMID: 32647537 PMCID: PMC7325551 DOI: 10.1177/1756286420935676] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/04/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Our aim was to determine whether specific nonsteroidal anti-inflammatory (NSAID) agents are associated with a decreased frequency of Alzheimer’s disease (AD). Materials and methods: Days of drug exposure were determined for diclofenac, etodolac, and naproxen using US Department of Veterans Affairs (VA) pharmacy transaction records, combined from two separate VA sites. AD diagnosis was established by the International Classification of Diseases, ninth revision (ICD-9)/ICD-10 diagnostic codes and the use of AD medications. Cox regression survival analysis was used to evaluate the association between AD frequency and NSAID exposure over time. Age at the end of the study and the medication-based disease burden index (a comorbidity index) were used as covariates. Results: Frequency of AD was significantly lower in the diclofenac group (4/1431, 0.28%) compared with etodolac (328/14,646, 2.24%), and naproxen (202/12,203, 1.66%). For regression analyses, naproxen was chosen as the comparator drug, since it has been shown to have no effect on the development of AD. Compared with naproxen, etodolac had no effect on the development of AD, hazard ratio (HR) 1.00 [95% confidence interval (CI): 0.84–1.20, p = 0.95]. In contrast, diclofenac had a significantly lower HR of AD compared with naproxen, HR 0.25 (95% CI: 0.09–0.68, p <0.01). After site effects were controlled for, age at end of the study (HR = 1.08, 95% CI: 1.07–1.09, p <0.001) was also found to influence the development of AD, and the medication-based disease burden index was a strong predictor for AD, HR 5.17 (95% CI: 4.60–5.81) indicating that as comorbidities increase, the risk for AD increases very significantly. Conclusion: Diclofenac, which has been shown to have active transport into the central nervous system, and which has been shown to lower amyloid beta and interleukin 1 beta, is associated with a significantly lower frequency of AD compared with etodolac and naproxen. These results are compelling, and parallel animal studies of the closely related fenamate NSAID drug class.
Collapse
Affiliation(s)
- Olaf Stuve
- Department of Neurology and Neurotherapeutics, University of Texas, Southwestern Medical School, Neurology Section (111H), Dallas VA Medical Center, 4500 Lancaster Road, Dallas, TX 75216, USA
| | | | | | - David A Jacob
- Pharmacy Service, Veterans Integrated Service Network 17, Arlington, TX, USA
| | - Bertis B Little
- School of Public Health and Information Sciences, University of Louisville, KY, USA
| |
Collapse
|
116
|
Schnöder L, Gasparoni G, Nordström K, Schottek A, Tomic I, Christmann A, Schäfer KH, Menger MD, Walter J, Fassbender K, Liu Y. Neuronal deficiency of p38α-MAPK ameliorates symptoms and pathology of APP or Tau-transgenic Alzheimer's mouse models. FASEB J 2020; 34:9628-9649. [PMID: 32475008 DOI: 10.1096/fj.201902731rr] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/30/2020] [Accepted: 05/11/2020] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia with very limited therapeutic options. Amyloid β (Aβ) and phosphorylated Tau (p-Tau) are key pathogenic molecules in AD. P38α-MAPK is specifically activated in AD lesion sites. However, its effects on AD pathogenesis, especially on p-Tau-associated brain pathology, and the underlying molecular mechanisms remain unclear. We mated human APP-transgenic mice and human P301S Tau-transgenic mice with mapk14-floxed and neuron-specific Cre-knock-in mice. We observed that deletion of p38α-MAPK specifically in neurons improves the cognitive function of both 9-month-old APP and Tau-transgenic AD mice, which is associated with decreased Aβ and p-Tau load in the brain. We further used next-generation sequencing to analyze the gene transcription in brains of p38α-MAPK deficient and wild-type APP-transgenic mice, which indicated that deletion of p38α-MAPK regulates the transcription of calcium homeostasis-related genes, especially downregulates the expression of grin2a, a gene encoding NMDAR subunit NR2A. Cell culture experiments further verified that deletion of p38α-MAPK inhibits NMDA-triggered calcium influx and neuronal apoptosis. Our systemic studies of AD pathogenic mechanisms using both APP- and Tau-transgenic mice suggested that deletion of neuronal p38α-MAPK attenuates AD-associated brain pathology and protects neurons in AD pathogenesis. This study supports p38α-MAPK as a novel target for AD therapy.
Collapse
Affiliation(s)
- Laura Schnöder
- Department of Neurology, Saarland University, Homburg, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg, Germany
| | - Gilles Gasparoni
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Karl Nordström
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Andrea Schottek
- Department of Neurology, Saarland University, Homburg, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg, Germany
| | - Inge Tomic
- Department of Neurology, Saarland University, Homburg, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg, Germany
| | - Anne Christmann
- Working Group Enteric Nervous System, University of Applied Sciences, Zweibrücken, Germany
| | - Karl H Schäfer
- Working Group Enteric Nervous System, University of Applied Sciences, Zweibrücken, Germany
| | - Michael D Menger
- Department of Experimental Surgery, Saarland University, Homburg, Germany
| | - Jörn Walter
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Klaus Fassbender
- Department of Neurology, Saarland University, Homburg, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg, Germany
| | - Yang Liu
- Department of Neurology, Saarland University, Homburg, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg, Germany
| |
Collapse
|
117
|
Dionisio-Santos DA, Behrouzi A, Olschowka JA, O'Banion MK. Evaluating the Effect of Interleukin-4 in the 3xTg Mouse Model of Alzheimer's Disease. Front Neurosci 2020; 14:441. [PMID: 32528242 PMCID: PMC7247853 DOI: 10.3389/fnins.2020.00441] [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: 02/05/2020] [Accepted: 04/09/2020] [Indexed: 11/13/2022] Open
Abstract
Chronic neuroinflammation has long been hypothesized to be involved in Alzheimer's Disease (AD) progression. Previous research suggests that both anti-inflammatory and inflammatory microglia ameliorate amyloid pathology, but the latter worsen tau pathology. In this study, we sought to determine whether induction of arginase-1 positive microglia with the anti-inflammatory cytokine IL-4 modulates pathology in the 3xTg mouse model of AD. Our findings indicate that a single intracranial IL-4 injection positively modulated performance of 3xTg AD mice in a Novel Object Recognition task, and locally increased the levels of arginase-1 positive myeloid cells when assessed one-week post injection. Furthermore, immunohistochemical analysis revealed decreased tau phosphorylation in IL-4 injected animals; however, we were not able to detect significant changes in tau phosphorylation utilizing Western blot. Lastly, IL-4 injection did not appear to cause significant changes in amyloid β load. In conclusion, acute intracranial IL-4 led to some positive benefits in the 3xTg mouse model of AD. Although more work remains, these results support therapeutic strategies aimed at modifying microglial activation states in neurodegenerative diseases.
Collapse
Affiliation(s)
- Dawling A Dionisio-Santos
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Adib Behrouzi
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - John A Olschowka
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - M Kerry O'Banion
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| |
Collapse
|
118
|
Yuan T, Manohar K, Latorre R, Orock A, Greenwood-Van Meerveld B. Inhibition of Microglial Activation in the Amygdala Reverses Stress-Induced Abdominal Pain in the Male Rat. Cell Mol Gastroenterol Hepatol 2020; 10:527-543. [PMID: 32408032 PMCID: PMC7394753 DOI: 10.1016/j.jcmgh.2020.04.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Psychological stress is a trigger for the development of irritable bowel syndrome and associated symptoms including abdominal pain. Although irritable bowel syndrome patients show increased activation in the limbic brain, including the amygdala, the underlying molecular and cellular mechanisms regulating visceral nociception in the central nervous system are incompletely understood. In a rodent model of chronic stress, we explored the role of microglia in the central nucleus of the amygdala (CeA) in controlling visceral sensitivity. Microglia are activated by environmental challenges such as stress, and are able to modify neuronal activity via synaptic remodeling and inflammatory cytokine release. Inflammatory gene expression and microglial activity are regulated negatively by nuclear glucocorticoid receptors (GR), which are suppressed by the stress-activated pain mediator p38 mitogen-activated protein kinases (MAPK). METHODS Fisher-344 male rats were exposed to water avoidance stress (WAS) for 1 hour per day for 7 days. Microglia morphology and the expression of phospho-p38 MAPK and GR were analyzed via immunofluorescence. Microglia-mediated synaptic remodeling was investigated by quantifying the number of postsynaptic density protein 95-positive puncta. Cytokine expression levels in the CeA were assessed via quantitative polymerase chain reaction and a Luminex assay (Bio-Rad, Hercules, CA). Stereotaxic infusion into the CeA of minocycline to inhibit, or fractalkine to activate, microglia was followed by colonic sensitivity measurement via a visceromotor behavioral response to isobaric graded pressures of tonic colorectal distension. RESULTS WAS induced microglial deramification in the CeA. Moreover, WAS induced a 3-fold increase in the expression of phospho-p38 and decreased the ratio of nuclear GR in the microglia. The number of microglia-engulfed postsynaptic density protein 95-positive puncta in the CeA was increased 3-fold by WAS, while cytokine levels were unchanged. WAS-induced changes in microglial morphology, microglia-mediated synaptic engulfment in the CeA, and visceral hypersensitivity were reversed by minocycline whereas in stress-naïve rats, fractalkine induced microglial deramification and visceral hypersensitivity. CONCLUSIONS Our data show that chronic stress induces visceral hypersensitivity in male rats and is associated with microglial p38 MAPK activation, GR dysfunction, and neuronal remodeling in the CeA.
Collapse
Affiliation(s)
- Tian Yuan
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Krishna Manohar
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Rocco Latorre
- Department of Basic Science and Craniofacial Biology, New York University, New York City, New York
| | - Albert Orock
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Beverley Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma,Oklahoma City VA Health Care System, Oklahoma City, Oklahoma,Correspondence Address correspondence to: Beverley Greenwood-Van Meerveld, PhD, O’Donoghue Building, Room 332, 1122 NE 13th Street, Oklahoma City, Oklahoma 73117.
| |
Collapse
|
119
|
LeBlang CJ, Medalla M, Nicoletti NW, Hays EC, Zhao J, Shattuck J, Cruz AL, Wolozin B, Luebke JI. Reduction of the RNA Binding Protein TIA1 Exacerbates Neuroinflammation in Tauopathy. Front Neurosci 2020; 14:285. [PMID: 32327969 PMCID: PMC7161592 DOI: 10.3389/fnins.2020.00285] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammatory processes play an integral role in the exacerbation and progression of pathology in tauopathies, a class of neurodegenerative disease characterized by aggregation of hyperphosphorylated tau protein. The RNA binding protein (RBP) T-cell Intracellular Antigen 1 (TIA1) is an important regulator of the innate immune response in the periphery, dampening cytotoxic inflammation and apoptosis during cellular stress, however, its role in neuroinflammation is unknown. We have recently shown that TIA1 regulates tau pathophysiology and toxicity in part through the binding of phospho-tau oligomers into pathological stress granules, and that haploinsufficiency of TIA1 in the P301S mouse model of tauopathy results in reduced accumulation of toxic tau oligomers, pathologic stress granules, and the development of downstream pathological features of tauopathy. The putative role of TIA1 as a regulator of the peripheral immune response led us to investigate the effects of TIA1 on neuroinflammation in the context of tauopathy, a chronic stressor in the neural environment. Here, we evaluated indicators of neuroinflammation including; reactive microgliosis and phagocytosis, pro-inflammatory cytokine release, and oxidative stress in hippocampal neurons and glia of wildtype and P301S transgenic mice expressing TIA1+/+, TIA1+/-, and TIA1-/- in both early (5 month) and advanced (9 month) disease states through biochemical, ultrastructural, and histological analyses. Our data show that both TIA1 haploinsufficiency and TIA1 knockout exacerbate neuroinflammatory processes in advanced stages of tauopathy, suggesting that TIA1 dampens the immune response in the central nervous system during chronic stress.
Collapse
Affiliation(s)
- Chelsey Jenna LeBlang
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Maria Medalla
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Nicholas William Nicoletti
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Emma Catherine Hays
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - James Zhao
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Jenifer Shattuck
- Laboratory of Neurodegeneration, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Anna Lourdes Cruz
- Laboratory of Neurodegeneration, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Benjamin Wolozin
- Laboratory of Neurodegeneration, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- Department of Neuroscience, Boston University, Boston, MA, United States
| | - Jennifer Irene Luebke
- Laboratory of Cellular Neuroscience, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
120
|
Zhang H, Wang D, Gong P, Lin A, Zhang Y, Ye RD, Yu Y. Formyl Peptide Receptor 2 Deficiency Improves Cognition and Attenuates Tau Hyperphosphorylation and Astrogliosis in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2020; 67:169-179. [PMID: 30475772 DOI: 10.3233/jad-180823] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is characterized by progressive loss of memory and other cognitive functions. Accumulation of amyloid-β (Aβ) and hyperphosphorylated tau are two major neuropathological features of AD. Formyl peptide receptor 2 (FPR2), contributing to innate immunity and inflammation, has been implicated in the uptake and clearance of Aβ. It remains unclear whether FPR2 affects cognition and tau phosphorylation. The effects of FPR2 in cognition and tau phosphorylation were examined using FPR2 knock-out (Fpr2-/-) mice receiving intracerebroventricular (ICV) injection of streptozotocin (STZ). The general behaviors and cognitive functions were evaluated using rotarod, open field test, and Morris water maze test. The alteration in tau hyperphosphorylation and activation of astrocytes were determined by using western blotting and/or immunofluorescence staining. ICV injection of STZ impaired spatial learning and memory of mice in Morris water maze. FPR2 deficiency improved spatial learning and memory of ICV-STZ mice. In the hippocampus and cortex of ICV-STZ mice, a marked increase was observed in tau phosphorylation at Ser199, Thr205, and Ser396 compared with ICV-saline control mice. However, FPR2 deficiency attenuated the hyperphosphorylation of tau at Ser199 and Ser396. In addition, the expression of GFAP was significantly increased in hippocampus and cortex of ICV-STZ mice. FPR2 deletion reduced the increase of GFAP expression induced by ICV injection of STZ. These results indicate that FPR2 deficiency is associate with improved cognition, reduced tau hyperphosphorylation, and activation of astrocytes in the mouse AD model tested. FPR2 may be a potential target in AD prevention and therapy.
Collapse
Affiliation(s)
- Haibo Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Ding Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Ping Gong
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Aihua Lin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Yan Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Richard D Ye
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, P. R. China.,Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Yang Yu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
121
|
Neuroinflammation and Neurogenesis in Alzheimer's Disease and Potential Therapeutic Approaches. Int J Mol Sci 2020; 21:ijms21030701. [PMID: 31973106 PMCID: PMC7037892 DOI: 10.3390/ijms21030701] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/17/2020] [Accepted: 01/19/2020] [Indexed: 12/17/2022] Open
Abstract
In adult brain, new neurons are generated throughout adulthood in the subventricular zone and the dentate gyrus; this process is commonly known as adult neurogenesis. The regulation or modulation of adult neurogenesis includes various intrinsic pathways (signal transduction pathway and epigenetic or genetic modulation pathways) or extrinsic pathways (metabolic growth factor modulation, vascular, and immune system pathways). Altered neurogenesis has been identified in Alzheimer's disease (AD), in both human AD brains and AD rodent models. The exact mechanism of the dysregulation of adult neurogenesis in AD has not been completely elucidated. However, neuroinflammation has been demonstrated to alter adult neurogenesis. The presence of various inflammatory components, such as immune cells, cytokines, or chemokines, plays a role in regulating the survival, proliferation, and maturation of neural stem cells. Neuroinflammation has also been considered as a hallmark neuropathological feature of AD. In this review, we summarize current, state-of-the art perspectives on adult neurogenesis, neuroinflammation, and the relationship between these two phenomena in AD. Furthermore, we discuss the potential therapeutic approaches, focusing on the anti-inflammatory and proneurogenic interventions that have been reported in this field.
Collapse
|
122
|
Lahiani-Cohen I, Touloumi O, Lagoudaki R, Grigoriadis N, Rosenmann H. Exposure to 3-Nitropropionic Acid Mitochondrial Toxin Induces Tau Pathology in Tangle-Mouse Model and in Wild Type-Mice. Front Cell Dev Biol 2020; 7:321. [PMID: 32010684 PMCID: PMC6971403 DOI: 10.3389/fcell.2019.00321] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/21/2019] [Indexed: 01/24/2023] Open
Abstract
Oxidative stress, particularly of mitochondrial origin, plays an important role in the pathogenesis of neurodegenerative disorders, including Alzheimer’s disease (AD) and other tauopathies. Controversies regarding the responses of tau phosphorylation state to various stimuli causing oxidative stress have been reported. Here we investigated the effect of 3-nitropropionic acid (3NP), a mitochondrial toxin which induces oxidative stress, on the tangle-pathology in our previously generated double mutant (E257T/P301S, DM) -Tau-tg mice and in WT-mice. We detected an increase in tangle pathology in the hippocampus and cortex of the DM-Tau-tg mice following exposure of the mice to the toxin, as well as generation of tangles in WT-mice. This increase was accompanied with alterations in the level of the glycogen synthase kinase 3β (GSK3β), the kinase which phosphorylates the tau protein, and in the phosphorylation state of this kinase. A response of microglial cells was noticed. These results point to the involvement of mitochondrial dysfunction in the development of the tangle-pathology and may suggest that interfering with mitochondrial dysfunction may have an anti-tangle therapeutic potential.
Collapse
Affiliation(s)
- Inbal Lahiani-Cohen
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Olga Touloumi
- B' Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Roza Lagoudaki
- B' Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | | | - Hanna Rosenmann
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
123
|
Ferguson SA, Varma V, Sloper D, Panos JJ, Sarkar S. Increased inflammation in BA21 brain tissue from African Americans with Alzheimer's disease. Metab Brain Dis 2020; 35:121-133. [PMID: 31823110 DOI: 10.1007/s11011-019-00512-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/25/2019] [Indexed: 12/21/2022]
Abstract
Chronic neuroinflammation is strongly associated with AD and altered peripheral and central levels of chemokines and cytokines have been frequently described in those with AD. Given the increasing evidence of ethnicity-related differences in AD, it was of interest to determine if those altered chemokine and cytokine levels are ethnicity-related. Because African Americans exhibit a higher incidence of AD and increased symptom severity, we explored chemokine and cytokine concentrations in post-mortem brain tissue from the BA21 region of African Americans and Caucasians with AD using multiplex assays. IL-1β, MIG, TRAIL, and FADD levels were significantly increased in African Americans while levels of IL-3 and IL-8 were significantly decreased. Those effects did not interact with gender; however, overall levels of CCL25, CCL26 and CX3CL1 were significantly decreased in women. The NLRP3 inflammasome is thought to be critically involved in AD. Increased activation of this inflammasome in African Americans is consistent with the current results.
Collapse
Affiliation(s)
- Sherry A Ferguson
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA.
| | - Vijayalakshmi Varma
- Division of Systems Biology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Daniel Sloper
- Division of Systems Biology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, 72079, USA
| | - John J Panos
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| |
Collapse
|
124
|
Li Y, Xu P, Shan J, Sun W, Ji X, Chi T, Liu P, Zou L. Interaction between hyperphosphorylated tau and pyroptosis in forskolin and streptozotocin induced AD models. Biomed Pharmacother 2020; 121:109618. [DOI: 10.1016/j.biopha.2019.109618] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/17/2019] [Accepted: 10/26/2019] [Indexed: 12/15/2022] Open
|
125
|
Balasubramaniam M, Parcon PA, Bose C, Liu L, Jones RA, Farlow MR, Mrak RE, Barger SW, Griffin WST. Interleukin-1β drives NEDD8 nuclear-to-cytoplasmic translocation, fostering parkin activation via NEDD8 binding to the P-ubiquitin activating site. J Neuroinflammation 2019; 16:275. [PMID: 31882005 PMCID: PMC6935243 DOI: 10.1186/s12974-019-1669-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/02/2019] [Indexed: 01/14/2023] Open
Abstract
Background Neuroinflammation, typified by elevated levels of interleukin-1 (IL-1) α and β, and deficits in proteostasis, characterized by accumulation of polyubiquitinated proteins and other aggregates, are associated with neurodegenerative disease independently and through interactions of the two phenomena. We investigated the influence of IL-1β on ubiquitination via its impact on activation of the E3 ligase parkin by either phosphorylated ubiquitin (P-Ub) or NEDD8. Methods Immunohistochemistry and Proximity Ligation Assay were used to assess colocalization of parkin with P-tau or NEDD8 in hippocampus from Alzheimer patients (AD) and controls. IL-1β effects on PINK1, P-Ub, parkin, P-parkin, and GSK3β—as well as phosphorylation of parkin by GSK3β—were assessed in cell cultures by western immunoblot, using two inhibitors and siRNA knockdown to suppress GSK3β. Computer modeling characterized the binding and the effects of P-Ub and NEDD8 on parkin. IL-1α, IL-1β, and parkin gene expression was assessed by RT-PCR in brains of 2- and 17-month-old PD-APP mice and wild-type littermates. Results IL-1α, IL-1β, and parkin mRNA levels were higher in PD-APP mice compared with wild-type littermates, and IL-1α-laden glia surrounded parkin- and P-tau-laden neurons in human AD. Such neurons showed a nuclear-to-cytoplasmic translocation of NEDD8 that was mimicked in IL-1β-treated primary neuronal cultures. These cultures also showed higher parkin levels and GSK3β-induced parkin phosphorylation; PINK1 levels were suppressed. In silico simulation predicted that binding of either P-Ub or NEDD8 at a singular position on parkin opens the UBL domain, exposing Ser65 for parkin activation. Conclusions The promotion of parkin- and NEDD8-mediated ubiquitination by IL-1β is consistent with an acute neuroprotective role. However, accumulations of P-tau and P-Ub and other elements of proteostasis, such as translocated NEDD8, in AD and in response to IL-1β suggest either over-stimulation or a proteostatic failure that may result from chronic IL-1β elevation, easily envisioned considering its early induction in Down’s syndrome and mild cognitive impairment. The findings further link autophagy and neuroinflammation, two important aspects of AD pathogenesis, which have previously been only loosely related.
Collapse
Affiliation(s)
| | - Paul A Parcon
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.,Department of Psychiatry, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Chhanda Bose
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Ling Liu
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Richard A Jones
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.,Geriatric Research Education and Clinical Center at the Central Arkansas Healthcare Veterans System, Little Rock, AR, 72205, USA
| | - Martin R Farlow
- Department of Neurology, Indiana Alzheimer Disease Center, Indiana University, Bloomington, USA
| | - Robert E Mrak
- Department of Pathology, University of Toledo Health Sciences Campus, Toledo, OH, 43614, USA
| | - Steven W Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.,Geriatric Research Education and Clinical Center at the Central Arkansas Healthcare Veterans System, Little Rock, AR, 72205, USA
| | - W Sue T Griffin
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA. .,Geriatric Research Education and Clinical Center at the Central Arkansas Healthcare Veterans System, Little Rock, AR, 72205, USA.
| |
Collapse
|
126
|
Chen X, Jiang H. Tau as a potential therapeutic target for ischemic stroke. Aging (Albany NY) 2019; 11:12827-12843. [PMID: 31841442 PMCID: PMC6949092 DOI: 10.18632/aging.102547] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022]
Abstract
Tau is a protein mainly expressed in adult human brain. It plays important roles both in neurodegenerative diseases and stroke. Stroke is an important cause of adult death and disability, ischemic stroke almost account for 80% in all cases. Abundant studies have proven that the increase of dysfunctional tau may act as a vital factor in pathological changes after ischemic stroke. However, the relationship between tau and ischemic stroke remains ununified. Based on present studies, we firstly introduced the structure and biological function of tau protein. Secondly, we summarized the potential regulatory mechanisms of tau protein in the process of ischemic stroke. Thirdly, we discussed about the findings in therapeutic researches of ischemic stroke. This review may be helpful in implementing new therapies for ischemic stroke and may be beneficial for the clinical and experimental studies.
Collapse
Affiliation(s)
- Xin Chen
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hua Jiang
- Department of Geriatrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
127
|
Fiery Cell Death: Pyroptosis in the Central Nervous System. Trends Neurosci 2019; 43:55-73. [PMID: 31843293 DOI: 10.1016/j.tins.2019.11.005] [Citation(s) in RCA: 228] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/01/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022]
Abstract
Pyroptosis ('fiery death') is an inflammatory type of regulated cell death (RCD), which occurs downstream of inflammasome activation. Pyroptosis is mediated directly by the recently identified family of pore-forming proteins known as gasdermins, the best characterized of which is gasdermin D (GSDMD). Recent investigations implicate pyroptosis in the pathogenesis of multiple neurological diseases. In this review, we discuss molecular mechanisms that drive pyroptosis, evidence for pyroptosis within the CNS, and emerging therapeutic strategies for its inhibition in the context of neurological disease.
Collapse
|
128
|
Cellular and Molecular Mediators of Neuroinflammation in Alzheimer Disease. Int Neurourol J 2019; 23:S54-62. [PMID: 31795604 PMCID: PMC6905206 DOI: 10.5213/inj.1938184.092] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/14/2019] [Indexed: 12/26/2022] Open
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder characterized by the loss of neuronal cells and the progressive decline of cognitive function. The major pathological culprit of AD is aggregation of amyloid-β (Aβ) and hyperphosphorylation of tau, eventually leading to progressive neuronal cell death and brain atrophy. However, the detailed molecular and cellular mechanisms underlying AD development as a result of neuronal cell death are little known. Although several hypotheses have been proposed regarding the development of AD, increasingly many studies suggest that the pathological progress of AD is not restricted to neuronal components such as Aβ and tau, but is also closely related to inflammatory responses in the brain. Abnormalities of Aβ and tau cause activity of pattern recognition receptors on the brain’s immune cells, including microglia and astrocytes, and trigger the innate immune system by releasing inflammatory mediators in the pathogenesis of AD. In this review, we present a basic overview of the current knowledge regarding inflammation and molecular mediators in the pathological progress of AD.
Collapse
|
129
|
Martini AC, Gomez-Arboledas A, Forner S, Rodriguez-Ortiz CJ, McQuade A, Danhash E, Phan J, Javonillo D, Ha JV, Tram M, Trujillo-Estrada L, da Cunha C, Ager RR, Davila JC, Kitazawa M, Blurton-Jones M, Gutierrez A, Baglietto-Vargas D, Medeiros R, LaFerla FM. Amyloid-beta impairs TOM1-mediated IL-1R1 signaling. Proc Natl Acad Sci U S A 2019; 116:21198-21206. [PMID: 31570577 PMCID: PMC6800331 DOI: 10.1073/pnas.1914088116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Defects in interleukin-1β (IL-1β)-mediated cellular responses contribute to Alzheimer's disease (AD). To decipher the mechanism associated with its pathogenesis, we investigated the molecular events associated with the termination of IL-1β inflammatory responses by focusing on the role played by the target of Myb1 (TOM1), a negative regulator of the interleukin-1β receptor-1 (IL-1R1). We first show that TOM1 steady-state levels are reduced in human AD hippocampi and in the brain of an AD mouse model versus respective controls. Experimentally reducing TOM1 affected microglia activity, substantially increased amyloid-beta levels, and impaired cognition, whereas enhancing its levels was therapeutic. These data show that reparation of the TOM1-signaling pathway represents a therapeutic target for brain inflammatory disorders such as AD. A better understanding of the age-related changes in the immune system will allow us to craft therapies to limit detrimental aspects of inflammation, with the broader purpose of sharply reducing the number of people afflicted by AD.
Collapse
Affiliation(s)
- Alessandra Cadete Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Angela Gomez-Arboledas
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Carlos J Rodriguez-Ortiz
- Center for Occupational and Environmental Health, School of Medicine, University of California, Irvine, CA 92697
| | - Amanda McQuade
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697
| | - Emma Danhash
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697
| | - Jimmy Phan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Dominic Javonillo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Jordan-Vu Ha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Melanie Tram
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - Celia da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Rahasson R Ager
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Jose C Davila
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - Masashi Kitazawa
- Center for Occupational and Environmental Health, School of Medicine, University of California, Irvine, CA 92697
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697
| | - Antonia Gutierrez
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
| | - Rodrigo Medeiros
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697;
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697;
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
| |
Collapse
|
130
|
Shi Y, Manis M, Long J, Wang K, Sullivan PM, Remolina Serrano J, Hoyle R, Holtzman DM. Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. J Exp Med 2019; 216:2546-2561. [PMID: 31601677 PMCID: PMC6829593 DOI: 10.1084/jem.20190980] [Citation(s) in RCA: 285] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 07/21/2019] [Accepted: 08/08/2019] [Indexed: 11/16/2022] Open
Abstract
Shi et al. find that microglia, instead of tau-induced direct neurotoxicity, are the driving force of neurodegeneration in a tauopathy mouse model. Microglia are also required for tau pathogenesis. In addition, apoE strongly regulates neurodegeneration in the setting of tauopathy predominantly by modulating microglial function. Chronic activation of brain innate immunity is a prominent feature of Alzheimer’s disease (AD) and primary tauopathies. However, to what degree innate immunity contributes to neurodegeneration as compared with pathological protein-induced neurotoxicity, and the requirement of a particular glial cell type in neurodegeneration, are still unclear. Here we demonstrate that microglia-mediated damage, rather than pathological tau-induced direct neurotoxicity, is the leading force driving neurodegeneration in a tauopathy mouse model. Importantly, the progression of ptau pathology is also driven by microglia. In addition, we found that APOE, the strongest genetic risk factor for AD, regulates neurodegeneration predominantly by modulating microglial activation, although a minor role of apoE in regulating ptau and insoluble tau formation independent of its immunomodulatory function was also identified. Our results suggest that therapeutic strategies targeting microglia may represent an effective approach to prevent disease progression in the setting of tauopathy.
Collapse
Affiliation(s)
- Yang Shi
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - Melissa Manis
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - Justin Long
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - Kairuo Wang
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | | | - Javier Remolina Serrano
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - Rosa Hoyle
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
131
|
Španić E, Langer Horvat L, Hof PR, Šimić G. Role of Microglial Cells in Alzheimer's Disease Tau Propagation. Front Aging Neurosci 2019; 11:271. [PMID: 31636558 PMCID: PMC6787141 DOI: 10.3389/fnagi.2019.00271] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/19/2019] [Indexed: 12/30/2022] Open
Abstract
Uncontrolled immune response in the brain contributes to the progression of all neurodegenerative disease, including Alzheimer's disease (AD). Recent investigations have documented the prion-like features of tau protein and the involvement of microglial changes with tau pathology. While it is still unclear what sequence of events is causal, it is likely that tau seeding potential and microglial contribution to tau propagation act together, and are essential for the development and progression of degenerative changes. Based on available evidence, targeting tau seeds and controlling some signaling pathways in a complex inflammation process could represent a possible new therapeutic approach for treating neurodegenerative diseases. Recent findings propose novel diagnostic assays and markers that may be used together with standard methods to complete and improve the diagnosis and classification of these diseases. In conclusion, a novel perspective on microglia-tau relations reveals new issues to investigate and imposes different approaches for developing therapeutic strategies for AD.
Collapse
Affiliation(s)
- Ena Španić
- Laboratory for Developmental Neuropathology, Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lea Langer Horvat
- Laboratory for Developmental Neuropathology, Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer’s Disease, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Šimić
- Laboratory for Developmental Neuropathology, Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
132
|
Nizami S, Hall‐Roberts H, Warrier S, Cowley SA, Di Daniel E. Microglial inflammation and phagocytosis in Alzheimer's disease: Potential therapeutic targets. Br J Pharmacol 2019; 176:3515-3532. [PMID: 30740661 PMCID: PMC6715590 DOI: 10.1111/bph.14618] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/19/2018] [Accepted: 01/15/2019] [Indexed: 12/20/2022] Open
Abstract
One of the largest unmet medical needs is a disease-modifying treatment for Alzheimer's disease (AD). Recently, the role of microglia in disease, particularly AD, has gained great interest, following the identification of several disease risk-associated genes that are highly expressed in microglia. Microglia play a critical homeostatic role in the brain, with neuroinflammatory and phagocytic mechanisms being of particular importance. Here, we review the role of NLRP3, the complement system, and the triggering receptor expressed in myeloid cells 2 (TREM2) in modulating microglial functions. We have reviewed the targets, their molecular pathways and the therapeutic interventions aimed at modulating these targets, in the hope of discovering a novel therapeutic approach for the treatment of AD. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Sohaib Nizami
- Alzheimer's Research UK Oxford Drug Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Hazel Hall‐Roberts
- Alzheimer's Research UK Oxford Drug Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- James Martin Stem Cell Facility, Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Sharat Warrier
- Alzheimer's Research UK Oxford Drug Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- James Martin Stem Cell Facility, Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Sally A. Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Elena Di Daniel
- Alzheimer's Research UK Oxford Drug Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
133
|
Kheiri G, Dolatshahi M, Rahmani F, Rezaei N. Role of p38/MAPKs in Alzheimer's disease: implications for amyloid beta toxicity targeted therapy. Rev Neurosci 2019; 30:9-30. [PMID: 29804103 DOI: 10.1515/revneuro-2018-0008] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 03/22/2018] [Indexed: 01/06/2023]
Abstract
A myriad of environmental and genetic factors, as well as the physiologic process of aging, contribute to Alzheimer's disease (AD) pathology. Neuroinflammation is and has been a focus of interest, as a common gateway for initiation of many of the underlying pathologies of AD. Amyloid beta (Aβ) toxicity, increasing RAGE expression, tau hyperphosphorylation, induction of apoptosis, and deregulated autophagy are among other mechanisms, partly entangled and being explained by activation of mitogen-activated protein kinase (MAPK) and MAPK signaling. p38 MAPK is the most essential regulator of Aβ induced toxicity from this family. p38 induces NF-κB activation, glutamate excitotoxicity, and disruption of synaptic plasticity, which are other implications of all justifying the p38 MAPK as a potential target to break the vicious Aβ toxicity cycle. Until recently, many in vivo and in vitro studies have investigated the effects of p38 MAPK inhibitors in AD. The pyridinyl imidazole compounds SB202190 and SB203580 have shown promising anti-apoptotic results in vivo. MW108 inhibits activation of p38 and is able to postpone cognitive decline in animal models. The PD169316, with anti-inflammatory, anti-oxidative, and anti-apoptotic features, has improved spatial memory in vivo. Natural compounds from Camellia sinensis (green tea), polyphenols from olive oil, pinocembrin from propolis, and the puerarine extract isoflavones, have shown strong anti-apoptotic features, mediated by p38 MAPK inhibition. Use of these drug targets is limited due to central nervous system side effects or cross-reactivity with other kinases, predicting the low efficacy of these drugs in clinical trials.
Collapse
Affiliation(s)
- Ghazaleh Kheiri
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, 1416753955 Tehran, Iran.,NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), 19166 Tehran, Iran
| | - Mahsa Dolatshahi
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, 1416753955 Tehran, Iran.,NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), 19166 Tehran, Iran
| | - Farzaneh Rahmani
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, 1416753955 Tehran, Iran.,NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), 19166 Tehran, Iran
| | - Nima Rezaei
- NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), 19166 Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran 14194, Iran
| |
Collapse
|
134
|
Shi Y, Holtzman DM. Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat Rev Immunol 2019; 18:759-772. [PMID: 30140051 DOI: 10.1038/s41577-018-0051-1] [Citation(s) in RCA: 399] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer disease is more than a pure proteopathy. Chronic neuroinflammation stands out during the pathogenesis of the disease and in turn modulates disease progression. The central nervous system (CNS) is separated from the blood circulation by the blood-brain barrier. In Alzheimer disease, neuroinflammation heavily relies on innate immune responses that are primarily mediated by CNS-resident microglia. APOE (which encodes apolipoprotein E) is the strongest genetic risk factor for Alzheimer disease, and APOE was recently shown to affect the disease in part through its immunomodulatory function. This function of APOE is likely linked to triggering receptor expressed on myeloid cells 2 (TREM2), which is expressed by microglia in the CNS. Here, we review the rapidly growing literature on the role of disease-associated microglia, TREM2 and APOE in the pathogenesis of Alzheimer disease and present an integrated view of innate immune function in Alzheimer disease.
Collapse
Affiliation(s)
- Yang Shi
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
135
|
Xu M, Huang Y, Song P, Huang Y, Huang W, Zhang HT, Hu Y. AAV9-Mediated Cdk5 Inhibitory Peptide Reduces Hyperphosphorylated Tau and Inflammation and Ameliorates Behavioral Changes Caused by Overexpression of p25 in the Brain. J Alzheimers Dis 2019; 70:573-585. [PMID: 31256130 DOI: 10.3233/jad-190099] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Miaojing Xu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yingwei Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Pingping Song
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Yaowei Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Wei Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
- Department of Neurology, the First People’s Hospital of Shunde, Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Han-Ting Zhang
- Department of Behavioral Medicine and Psychiatry and Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
136
|
Vogels T, Murgoci AN, Hromádka T. Intersection of pathological tau and microglia at the synapse. Acta Neuropathol Commun 2019; 7:109. [PMID: 31277708 PMCID: PMC6612163 DOI: 10.1186/s40478-019-0754-y] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
Tauopathies are a heterogenous class of diseases characterized by cellular accumulation of aggregated tau and include diseases such as Alzheimer’s disease (AD), progressive supranuclear palsy and chronic traumatic encephalopathy. Tau pathology is strongly linked to neurodegeneration and clinical symptoms in tauopathy patients. Furthermore, synapse loss is an early pathological event in tauopathies and is the strongest correlate of cognitive decline. Tau pathology is additionally associated with chronic neuroinflammatory processes, such as reactive microglia, astrocytes, and increased levels of pro-inflammatory molecules (e.g. complement proteins, cytokines). Recent studies show that as the principal immune cells of the brain, microglia play a particularly important role in the initiation and progression of tau pathology and associated neurodegeneration. Furthermore, AD risk genes such as Triggering receptor expressed on myeloid cells 2 (TREM2) and Apolipoprotein E (APOE) are enriched in the innate immune system and modulate the neuroinflammatory response of microglia to tau pathology. Microglia can play an active role in synaptic dysfunction by abnormally phagocytosing synaptic compartments of neurons with tau pathology. Furthermore, microglia are involved in synaptic spreading of tau – a process which is thought to underlie the progressive nature of tau pathology propagation through the brain. Spreading of pathological tau is also the predominant target for tau-based immunotherapy. Active tau vaccines, therapeutic tau antibodies and other approaches targeting the immune system are actively explored as treatment options for AD and other tauopathies. This review describes the role of microglia in the pathobiology of tauopathies and the mechanism of action of potential therapeutics targeting the immune system in tauopathies.
Collapse
|
137
|
Šimić G, Španić E, Langer Horvat L, Hof PR. Blood-brain barrier and innate immunity in the pathogenesis of Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 168:99-145. [PMID: 31699331 DOI: 10.1016/bs.pmbts.2019.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pathogenesis of Alzheimer's disease (AD) is only partly understood. This is the probable reason why significant efforts to treat or prevent AD have been unsuccessful. In fact, as of April 2019, there have been 2094 studies registered for AD on the clinicaltrials.gov U.S. National Library of Science web page, of which only a few are still ongoing. In AD, abnormal accumulation of amyloid and tau proteins in the brain are thought to begin 10-20 years before the onset of overt symptoms, suggesting that interventions designed to prevent pathological amyloid and tau accumulation may be more effective than attempting to reverse a pathology once it is established. However, to be successful, such early interventions need to be selectively administered to individuals who will likely develop the disease long before the symptoms occur. Therefore, it is critical to identify early biomarkers that are strongly predictive of AD. Currently, patients are diagnosed on the basis of a variety of clinical scales, neuropsychological tests, imaging and laboratory modalities, but definitive diagnosis can be made only by postmortem assessment of underlying neuropathology. People suffering from AD thus may be misdiagnosed clinically with other primary causes of dementia, and vice versa, thereby also reducing the power of clinical trials. The amyloid cascade hypothesis fits well for the familial cases of AD with known mutations, but is not sufficient to explain sporadic, late-onset AD (LOAD) that accounts for over 95% of all cases. Since the earliest descriptions of AD there have been neuropathological features described other than amyloid plaques (AP) and neurofibrillary tangles (NFT), most notably gliosis and neuroinflammation. However, it is only recently that genetic and experimental studies have implicated microglial dysfunction as a causal factor for AD, as opposed to a merely biological response of its accumulation around AP. Additionally, many studies have suggested the importance of changes in blood-brain barrier (BBB) permeability in the pathogenesis of AD. Here we suggest how these less investigated aspects of the disease that have gained increased attention in recent years may contribute mechanistically to the development of lesions and symptoms of AD.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.
| | - Ena Španić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lea Langer Horvat
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
138
|
Teter B, Morihara T, Lim GP, Chu T, Jones MR, Zuo X, Paul RM, Frautschy SA, Cole GM. Curcumin restores innate immune Alzheimer's disease risk gene expression to ameliorate Alzheimer pathogenesis. Neurobiol Dis 2019; 127:432-448. [PMID: 30951849 PMCID: PMC8092921 DOI: 10.1016/j.nbd.2019.02.015] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 02/15/2019] [Accepted: 02/20/2019] [Indexed: 01/28/2023] Open
Abstract
Alzheimer's disease (AD) genetics implies a causal role for innate immune genes, TREM2 and CD33, products that oppose each other in the downstream Syk tyrosine kinase pathway, activating microglial phagocytosis of amyloid (Aβ). We report effects of low (Curc-lo) and high (Curc-hi) doses of curcumin on neuroinflammation in APPsw transgenic mice. Results showed that Curc-lo decreased CD33 and increased TREM2 expression (predicted to decrease AD risk) and also increased TyroBP, which controls a neuroinflammatory gene network implicated in AD as well as phagocytosis markers CD68 and Arg1. Curc-lo coordinately restored tightly correlated relationships between these genes' expression levels, and decreased expression of genes characteristic of toxic pro-inflammatory M1 microglia (CD11b, iNOS, COX-2, IL1β). In contrast, very high dose curcumin did not show these effects, failed to clear amyloid plaques, and dysregulated gene expression relationships. Curc-lo stimulated microglial migration to and phagocytosis of amyloid plaques both in vivo and in ex vivo assays of sections of human AD brain and of mouse brain. Curcumin also reduced levels of miR-155, a micro-RNA reported to drive a neurodegenerative microglial phenotype. In conditions without amyloid (human microglial cells in vitro, aged wild-type mice), Curc-lo similarly decreased CD33 and increased TREM2. Like curcumin, anti-Aβ antibody (also reported to engage the Syk pathway, increase CD68, and decrease amyloid burden in human and mouse brain) increased TREM2 in APPsw mice and decreased amyloid in human AD sections ex vivo. We conclude that curcumin is an immunomodulatory treatment capable of emulating anti-Aβ vaccine in stimulating phagocytic clearance of amyloid by reducing CD33 and increasing TREM2 and TyroBP, while restoring neuroinflammatory networks implicated in neurodegenerative diseases.
Collapse
Affiliation(s)
- B Teter
- Departments of Neurology, Geriatric Research Education and Clinical Centerand, University of California, Los Angeles (UCLA), United States of America; Departments of Veterans Affairs Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center, University of California, Los Angeles (UCLA), United States of America; Alzheimer's Translational Center, Veterans Administration (Research 151), Bldg. 114, Rm. 114-1, 11301 Wilshire Blvd, Los Angeles, CA 90073, United States of America.
| | - T Morihara
- Departments of Neurology, Geriatric Research Education and Clinical Centerand, University of California, Los Angeles (UCLA), United States of America; Departments of Veterans Affairs Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center, University of California, Los Angeles (UCLA), United States of America.
| | - G P Lim
- Departments of Neurology, Geriatric Research Education and Clinical Centerand, University of California, Los Angeles (UCLA), United States of America; Departments of Veterans Affairs Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center, University of California, Los Angeles (UCLA), United States of America
| | - T Chu
- Departments of Neurology, Geriatric Research Education and Clinical Centerand, University of California, Los Angeles (UCLA), United States of America; Departments of Veterans Affairs Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center, University of California, Los Angeles (UCLA), United States of America
| | - M R Jones
- Departments of Neurology, Geriatric Research Education and Clinical Centerand, University of California, Los Angeles (UCLA), United States of America
| | - X Zuo
- Departments of Neurology, Geriatric Research Education and Clinical Centerand, University of California, Los Angeles (UCLA), United States of America; Departments of Veterans Affairs Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center, University of California, Los Angeles (UCLA), United States of America
| | - R M Paul
- Departments of Neurology, Geriatric Research Education and Clinical Centerand, University of California, Los Angeles (UCLA), United States of America; Departments of Medicine, University of California, Los Angeles (UCLA), United States of America
| | - S A Frautschy
- Departments of Neurology, Geriatric Research Education and Clinical Centerand, University of California, Los Angeles (UCLA), United States of America; Departments of Medicine, University of California, Los Angeles (UCLA), United States of America; Departments of Veterans Affairs Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center, University of California, Los Angeles (UCLA), United States of America.
| | - G M Cole
- Departments of Neurology, Geriatric Research Education and Clinical Centerand, University of California, Los Angeles (UCLA), United States of America; Departments of Medicine, University of California, Los Angeles (UCLA), United States of America; Departments of Veterans Affairs Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center, University of California, Los Angeles (UCLA), United States of America.
| |
Collapse
|
139
|
Lachen-Montes M, González-Morales A, Palomino M, Ausin K, Gómez-Ochoa M, Zelaya MV, Ferrer I, Pérez-Mediavilla A, Fernández-Irigoyen J, Santamaría E. Early-Onset Molecular Derangements in the Olfactory Bulb of Tg2576 Mice: Novel Insights Into the Stress-Responsive Olfactory Kinase Dynamics in Alzheimer's Disease. Front Aging Neurosci 2019; 11:141. [PMID: 31244650 PMCID: PMC6579864 DOI: 10.3389/fnagi.2019.00141] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 05/23/2019] [Indexed: 11/13/2022] Open
Abstract
The olfactory bulb (OB) is the first processing station in the olfactory pathway. Despite smell impairment, which is considered an early event in Alzheimer’s disease (AD), little is known about the initial molecular disturbances that accompany the AD development at olfactory level. We have interrogated the time-dependent OB molecular landscape in Tg2576 AD mice prior to the appearance of neuropathological amyloid plaques (2-, and 6-month-old), using combinatorial omics analysis. The metabolic modulation induced by overproduction of human mutated amyloid precursor protein (APP) clearly differs between both time points. Besides the progressive perturbation of the APP interactome, functional network analysis unveiled an inverse regulation of downstream extracellular signal-regulated kinase (ERK1/2), and p38 mitogen-activated protein kinase (MAPK) routes in 2-month-old Tg2576 mice with respect to wild-type (WT) mice. In contrast, Akt and MAPK kinase 4 (SEK1)/ stress-activated protein kinase (SAPK) axis were parallel activated in the OB of 6-months-old-Tg2576 mice. Furthermore, a survival kinome profiling performed during the aging process (2-, 6-, and 18-month-old) revealed that olfactory APP overexpression leads to changes in the activation dynamics of protein kinase A (PKA), and SEK1/MKK4-SAPK/JNK between 6 and 18 months of age, when memory deficits appear and AD pathology is well established in transgenic mice. Interestingly, both olfactory pathways were differentially activated in a stage-dependent manner in human sporadic AD subjects with different neuropathological grading. Taken together, our data reflect the early impact of mutated APP on the OB molecular homeostasis, highlighting the progressive modulation of specific signaling pathways during the olfactory amyloidogenic pathology.
Collapse
Affiliation(s)
- Mercedes Lachen-Montes
- Clinical Neuroproteomics Group, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,Proteored-ISCIII Proteomics Unit, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Andrea González-Morales
- Clinical Neuroproteomics Group, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,Proteored-ISCIII Proteomics Unit, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Maialen Palomino
- Clinical Neuroproteomics Group, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Karina Ausin
- Proteored-ISCIII Proteomics Unit, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Marta Gómez-Ochoa
- Department of Pathology, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - María Victoria Zelaya
- Clinical Neuroproteomics Group, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Department of Pathology, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Isidro Ferrer
- Institut de Neuropatologia, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Alberto Pérez-Mediavilla
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Neurobiology of Alzheimer's Disease, Department of Biochemistry, Center for Applied Medical Research (CIMA), Neurosciences Division, University of Navarra, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Group, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,Proteored-ISCIII Proteomics Unit, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Group, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,Proteored-ISCIII Proteomics Unit, NavarraBiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain
| |
Collapse
|
140
|
Patrick KL, Bell SL, Weindel CG, Watson RO. Exploring the "Multiple-Hit Hypothesis" of Neurodegenerative Disease: Bacterial Infection Comes Up to Bat. Front Cell Infect Microbiol 2019; 9:138. [PMID: 31192157 PMCID: PMC6546885 DOI: 10.3389/fcimb.2019.00138] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/16/2019] [Indexed: 12/23/2022] Open
Abstract
Despite major strides in personalized genomics, it remains poorly understood why neurodegenerative diseases occur in only a fraction of individuals with a genetic predisposition and conversely, why individuals with no genetic risk of a disorder develop one. Chronic diseases like Alzheimer's, Parkinson's, and Multiple sclerosis are speculated to result from a combination of genetic and environmental factors, a concept commonly referred to as the “multiple hit hypothesis.” A number of bacterial infections have been linked to increased risk of neurodegeneration, and in some cases, clearance of bacterial pathogens has been correlated with amelioration of central nervous system (CNS) deficits. Additionally, mutations in several genes known to contribute to CNS disorders like Parkinson's Disease have repeatedly been implicated in susceptibility to intracellular bacterial infection. Recent data has begun to demonstrate roles for these genes (PARK2, PINK1, and LRRK2) in modulating innate immune outcomes, suggesting that immune dysregulation may play an even more important role in neurodegeneration than previously appreciated. This review will broadly explore the connections between bacterial infection, immune dysregulation, and CNS disorders. Understanding this interplay and how bacterial pathogenesis contributes to the “multiple-hit hypothesis” of neurodegeneration will be crucial to develop therapeutics to effectively treat both neurodegeneration and infection.
Collapse
Affiliation(s)
- Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, United States
| | - Samantha L Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, United States
| | - Chi G Weindel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, United States
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, United States
| |
Collapse
|
141
|
Jian M, Kwan JSC, Bunting M, Ng RCL, Chan KH. Adiponectin suppresses amyloid-β oligomer (AβO)-induced inflammatory response of microglia via AdipoR1-AMPK-NF-κB signaling pathway. J Neuroinflammation 2019; 16:110. [PMID: 31128596 PMCID: PMC6535190 DOI: 10.1186/s12974-019-1492-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/30/2019] [Indexed: 01/07/2023] Open
Abstract
Background Microglia-mediated neuroinflammation is important in Alzheimer’s disease (AD) pathogenesis. Extracellular deposition of β-amyloid (Aβ), a major pathological hallmark of AD, can induce microglia activation. Adiponectin (APN), an adipocyte-derived adipokine, exerts anti-inflammatory effects in the periphery and brain. Chronic APN deficiency leads to cognitive impairment and AD-like pathologies in aged mice. Here, we aim to study the role of APN in regulating microglia-mediated neuroinflammation in AD. Methods Inflammatory response of cultured microglia (BV2 cells) to AβO and effects of APN were studied by measuring levels of proinflammatory cytokines (tumor necrosis factor α [TNFα] and interleukin-1β [IL-1β]) in cultured medium before and after exposure to AβO, with and without APN pretreatment. Adiponectin receptor 1 (AdipoR1) and receptor 2 (AdipoR2) were targeted by small interference RNA. To study the neuroprotective effect of APN, cultured HT-22 hippocampal cells were treated with conditioned medium of AβO-exposed BV2 cells or were co-cultured with BV2 cells in transwells. The cytotoxicity of HT-22 hippocampal cells was assessed by MTT reduction. We generated APN-deficient AD mice (APN−/−5xFAD) by crossing APN-knockout mice with 5xFAD mice to determine the effects of APN deficiency on microglia-mediated neuroinflammation in AD. Results AdipoR1 and AdipoR2 were expressed in BV2 cells and microglia of mice. Pretreatment with APN for 2 h suppressed TNFα and IL-1β release induced by AβO in BV2 cells. Additionally, APN rescued the decrease of AMPK phosphorylation and suppressed nuclear translocation of nuclear factor kappa B (NF-κB) induced by AβO. Compound C, an inhibitor of AMPK, abolished these effects of APN. Knockdown of AdipoR1, but not AdipoR2 in BV2 cells, inhibited the ability of APN to suppress proinflammatory cytokine release induced by AβO. Moreover, pretreatment with APN inhibited the cytotoxicity of HT-22 cells co-cultured with AβO-exposed BV2 cells. Lastly, APN deficiency exacerbated microglia activation in 9-month-old APN−/−5xFAD mice associated with upregulation of TNFα and IL-1β in the cortex and hippocampus. Conclusions Our findings demonstrate that APN inhibits inflammatory response of microglia to AβO via AdipoR1-AMPK-NF-κB signaling, and APN deficiency aggravates microglia activation and neuroinflammation in AD mice. APN may be a novel therapeutic agent for inhibiting neuroinflammation in AD. Electronic supplementary material The online version of this article (10.1186/s12974-019-1492-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Jian
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China
| | - Jason Shing-Cheong Kwan
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China.,Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China
| | - Myriam Bunting
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China
| | - Roy Chun-Laam Ng
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China. .,Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China.
| | - Koon Ho Chan
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China. .,Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China. .,Research Center of Heart, Brain, Hormone and Healthy Aging, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China. .,Hong Kong University Alzheimer's Disease Research Network, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China. .,Department of Medicine, The University of Hong Kong, 4/F, Professorial Block, Queen Mary Hospital, 102 Pokfulam Road, Pokfulam, Hong Kong, Special Administrative Region of China.
| |
Collapse
|
142
|
Sheppard O, Coleman MP, Durrant CS. Lipopolysaccharide-induced neuroinflammation induces presynaptic disruption through a direct action on brain tissue involving microglia-derived interleukin 1 beta. J Neuroinflammation 2019; 16:106. [PMID: 31103036 PMCID: PMC6525970 DOI: 10.1186/s12974-019-1490-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/29/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Systemic inflammation has been linked to synapse loss and cognitive decline in human patients and animal models. A role for microglial release of pro-inflammatory cytokines has been proposed based on in vivo and primary culture studies. However, mechanisms are hard to study in vivo as specific microglial ablation is challenging and the extracellular fluid cannot be sampled without invasive methods. Primary cultures have different limitations as the intricate multicellular architecture in the brain is not fully reproduced. It is essential to confirm proposed brain-specific mechanisms of inflammatory synapse loss directly in brain tissue. Organotypic hippocampal slice cultures (OHSCs) retain much of the in vivo neuronal architecture, synaptic connections and diversity of cell types whilst providing convenient access to manipulate and sample the culture medium and observe cellular reactions. METHODS OHSCs were generated from P6-P9 C57BL/6 mice. Inflammation was induced via addition of lipopolysaccharide (LPS), and cultures were analysed for changes in synaptic proteins, gene expression and protein secretion. Microglia were selectively depleted using clodronate, and the effect of IL1β was assessed using a specific neutralising monoclonal antibody. RESULTS LPS treatment induced loss of the presynaptic protein synaptophysin without altering PSD95 or Aβ protein levels. Depletion of microglia prior to LPS application prevented the loss of synaptophysin, whilst microglia depletion after the inflammatory insult was partially effective, although less so than pre-emptive treatment, indicating a time-critical window in which microglia can induce synaptic damage. IL1β protein and mRNA were increased after LPS addition, with these effects also prevented by microglia depletion. Direct application of IL1β to OHSCs resulted in synaptophysin loss whilst pre-treatment with IL1β neutralising antibody prior to LPS addition prevented a significant loss of synaptophysin but may also impact basal synaptic levels. CONCLUSIONS The loss of synaptophysin in this system confirms LPS can act directly within brain tissue to disrupt synapses, and we show that microglia are the relevant cellular target when all major CNS cell types are present. By overcoming limitations of primary culture and in vivo work, our study strengthens the evidence for a key role of microglia-derived IL1β in synaptic dysfunction after inflammatory insult.
Collapse
Affiliation(s)
- Olivia Sheppard
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Michael P Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK.,Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Claire S Durrant
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK. .,Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
143
|
Song YN, Li JQ, Tan CC, Wang HF, Tan MS, Cao XP, Yu JT, Tan L. TREML2 Mutation Mediate Alzheimer's Disease Risk by Altering Neuronal Degeneration. Front Neurosci 2019; 13:455. [PMID: 31156362 PMCID: PMC6529571 DOI: 10.3389/fnins.2019.00455] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 04/23/2019] [Indexed: 11/26/2022] Open
Abstract
A coding missense mutation (rs3747742) in triggering receptor expressed on myeloid cell-like 2 (TREML2) has been recently proposed as an important protective factor against Alzheimer’s disease (AD). However, the link between TREML2 and AD pathology remains unclear. Therefore, we explored the association of TREML2 rs3747742 with cognitive function, neuroimaging biomarkers and cerebrospinal fluid (CSF) biomarkers related to AD, including CSF total-tau (T-tau), phosphor-tau (P-tau), and amyloid-β (Aβ1-42). As for cognitive function, related cognitive scores of Clinical Dementia Rating Sum of Boxes (CDRSB), Alzheimer’s Disease Assessment Scale-cognitive section 11 (ADAS-cog 11), Mini-Mental State Examination (MMSE), and Rey Auditory-Verbal Learning Test (RAVLT) were extracted. We used a multiple linear regression model to examine the association of TREML2 rs3747742 with the baseline variables. Furthermore, we also calculated the change rate of above variables influenced by TREML2 rs3747742 via applying a mixed-effects model over a 4-year follow-up. In this analysis, a total of 1,306 individuals from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database were included. Finally, we observed that only in AD patients, but not in normal controls or mild cognitive impairment (MCI) individuals, TREML2 rs3747742 exhibited a strong association with CSF total-tau levels at baseline (β = -22.1210, p = 0.0166) and 4-year follow-up (β = -0.3961, p = 0.0115). Furthermore, no associations were found with CSF Aβ1-42 levels, P-tau levels, neuroimaging biomarkers and cognitive function neither for baseline variables nor for longitudinal data. Thus, this study indicated that TREML2 mediated the risk of AD through influencing AD-related neurodegeneration (abnormal T-tau levels) but not P-tau levels and Aβ pathology.
Collapse
Affiliation(s)
- Ya-Nan Song
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jie-Qiong Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hui-Fu Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Meng-Shan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.,Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | | |
Collapse
|
144
|
Dionisio-Santos DA, Olschowka JA, O'Banion MK. Exploiting microglial and peripheral immune cell crosstalk to treat Alzheimer's disease. J Neuroinflammation 2019; 16:74. [PMID: 30953557 PMCID: PMC6449993 DOI: 10.1186/s12974-019-1453-0] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/18/2019] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation is considered one of the cardinal features of Alzheimer’s disease (AD). Neuritic plaques composed of amyloid β and neurofibrillary tangle-laden neurons are surrounded by reactive astrocytes and microglia. Exposure of microglia, the resident myeloid cell of the CNS, to amyloid β causes these cells to acquire an inflammatory phenotype. While these reactive microglia are important to contain and phagocytose amyloid plaques, their activated phenotype impacts CNS homeostasis. In rodent models, increased neuroinflammation promoted by overexpression of proinflammatory cytokines can cause an increase in hyperphosphorylated tau and a decrease in hippocampal function. The peripheral immune system can also play a detrimental or beneficial role in CNS inflammation. Systemic inflammation can increase the risk of developing AD dementia, and chemokines released directly by microglia or indirectly by endothelial cells can attract monocytes and T lymphocytes to the CNS. These peripheral immune cells can aid in amyloid β clearance or modulate microglia responses, depending on the cell type. As such, several groups have targeted the peripheral immune system to modulate chronic neuroinflammation. In this review, we focus on the interplay of immunomodulating factors and cell types that are being investigated as possible therapeutic targets for the treatment or prevention of AD.
Collapse
Affiliation(s)
- Dawling A Dionisio-Santos
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 603, Rochester, NY, 14642, USA
| | - John A Olschowka
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 603, Rochester, NY, 14642, USA
| | - M Kerry O'Banion
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box 603, Rochester, NY, 14642, USA.
| |
Collapse
|
145
|
Ren QG, Gong WG, Zhou H, Shu H, Wang YJ, Zhang ZJ. Spatial Training Ameliorates Long-Term Alzheimer's Disease-Like Pathological Deficits by Reducing NLRP3 Inflammasomes in PR5 Mice. Neurotherapeutics 2019; 16:450-464. [PMID: 30560481 PMCID: PMC6554388 DOI: 10.1007/s13311-018-00698-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recent studies have suggested that cognitive training could delay memory loss in Alzheimer's disease (AD). However, whether and how cognitive training produces long-term benefits remains unclear. Here, 10-month-old PR5 mice were spatially trained in a water maze for 4 consecutive weeks. The novel object recognition test (NORT), Western blots, Golgi staining, and ELISA were used to examine behavioral, biochemical, and pathological measures immediately after training and 3 months later. Immediately after training, we found that spatial training significantly improved cognitive performance; reduced tau neuropathology; increased the expression level of synaptophysin, PSD93, and PSD95 in the hippocampus; and increased the number of dendritic spines in PR5 mice. The expression levels of NLRP3, caspase-1, and interleukin (IL)-1β, which were significantly elevated in PR5 mice, were reversed by spatial training. Interestingly, these effects persisted 3 months later. To further detect the role of NLRP3 in spatial training, PR5/NLRP3-/- mice and PR5/NLRP3+/- mice were also used in our study. PR5/NLRP3-/- mice showed better cognitive performance than PR5 mice. After 1 week of spatial training, these changes (including those in expression levels of synaptophysin, PSD93, and PSD95; the number of dendritic spines; and caspase-1 and IL-1β content in PR5 mice) could be totally reversed in PR5/NLRP3-/- and PR5/NLRP3+/- mice. In addition, there was a positive correlation between NLRP3 content and the expression levels of caspase-1 and IL-1β. These results show an important role for the NLRP3/caspase-1/IL-1β axis in ameliorating the effect of spatial training on cognitive impairment in PR5 mice.
Collapse
Affiliation(s)
- Qing-Guo Ren
- Department of Neuropsychiatry, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Wei-Gang Gong
- Department of Neuropsychiatry, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Hong Zhou
- Department of Neuropsychiatry, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hao Shu
- Department of Neuropsychiatry, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yan-Juan Wang
- Department of Neuropsychiatry, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhi-Jun Zhang
- Department of Neuropsychiatry, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
146
|
Aggregated Tau activates NLRP3-ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded Tau pathology in vivo. Acta Neuropathol 2019; 137:599-617. [PMID: 30721409 PMCID: PMC6426830 DOI: 10.1007/s00401-018-01957-y] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/28/2018] [Accepted: 12/28/2018] [Indexed: 12/18/2022]
Abstract
Brains of Alzheimer's disease patients are characterized by the presence of amyloid plaques and neurofibrillary tangles, both invariably associated with neuroinflammation. A crucial role for NLRP3-ASC inflammasome [NACHT, LRR and PYD domains-containing protein 3 (NLRP3)-Apoptosis-associated speck-like protein containing a CARD (ASC)] in amyloid-beta (Aβ)-induced microgliosis and Aβ pathology has been unequivocally identified. Aβ aggregates activate NLRP3-ASC inflammasome (Halle et al. in Nat Immunol 9:857-865, 2008) and conversely NLRP3-ASC inflammasome activation exacerbates amyloid pathology in vivo (Heneka et al. in Nature 493:674-678, 2013), including by prion-like ASC-speck cross-seeding (Venegas et al. in Nature 552:355-361, 2017). However, the link between inflammasome activation, as crucial sensor of innate immunity, and Tau remains unexplored. Here, we analyzed whether Tau aggregates acting as prion-like Tau seeds can activate NLRP3-ASC inflammasome. We demonstrate that Tau seeds activate NLRP3-ASC-dependent inflammasome in primary microglia, following microglial uptake and lysosomal sorting of Tau seeds. Next, we analyzed the role of inflammasome activation in prion-like or templated seeding of Tau pathology and found significant inhibition of exogenously seeded Tau pathology by ASC deficiency in Tau transgenic mice. We furthermore demonstrate that chronic intracerebral administration of the NLRP3 inhibitor, MCC950, inhibits exogenously seeded Tau pathology. Finally, ASC deficiency also decreased non-exogenously seeded Tau pathology in Tau transgenic mice. Overall our findings demonstrate that Tau-seeding competent, aggregated Tau activates the ASC inflammasome through the NLRP3-ASC axis, and we demonstrate an exacerbating role of the NLRP3-ASC axis on exogenously and non-exogenously seeded Tau pathology in Tau mice in vivo. The NLRP3-ASC inflammasome, which is an important sensor of innate immunity and intensively explored for its role in health and disease, hence presents as an interesting therapeutic approach to target three crucial pathogenetic processes in AD, including prion-like seeding of Tau pathology, Aβ pathology and neuroinflammation.
Collapse
|
147
|
Inhibition of oxaliplatin-induced neurotoxicity by silymarin through increased expression of brain-derived neurotrophic factor and inhibition of p38-MAPK. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0018-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
148
|
Sarkar SN, Russell AE, Engler-Chiurazzi EB, Porter KN, Simpkins JW. MicroRNAs and the Genetic Nexus of Brain Aging, Neuroinflammation, Neurodegeneration, and Brain Trauma. Aging Dis 2019; 10:329-352. [PMID: 31011481 PMCID: PMC6457055 DOI: 10.14336/ad.2018.0409] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex and integrated gradual deterioration of cellular activities in specific organs of the body, which is associated with increased mortality. This deterioration is the primary risk factor for major human pathologies, including cancer, diabetes, cardiovascular disorders, neurovascular disorders, and neurodegenerative diseases. There are nine tentative hallmarks of aging. In addition, several of these hallmarks are increasingly being associated with acute brain injury conditions. In this review, we consider the genes and their functional pathways involved in brain aging as a means of developing new strategies for therapies targeted to the neuropathological processes themselves, but also as targets for many age-related brain diseases. A single microRNA (miR), which is a short, non-coding RNA species, has the potential for targeting many genes simultaneously and, like practically all other cellular processes, genes associated with many features of brain aging and injury are regulated by miRs. We highlight how certain miRs can mediate deregulation of genes involved in neuroinflammation, acute neuronal injury and chronic neurodegenerative diseases. Finally, we review the recent progress in the development of effective strategies to block specific miR functions and discuss future approaches with the prediction that anti-miR drugs may soon be used in the clinic.
Collapse
Affiliation(s)
- Saumyendra N Sarkar
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Ashley E Russell
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Elizabeth B Engler-Chiurazzi
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Keyana N Porter
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - James W Simpkins
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
149
|
Bettcher BM, Johnson SC, Fitch R, Casaletto KB, Heffernan KS, Asthana S, Zetterberg H, Blennow K, Carlsson CM, Neuhaus J, Bendlin BB, Kramer JH. Cerebrospinal Fluid and Plasma Levels of Inflammation Differentially Relate to CNS Markers of Alzheimer's Disease Pathology and Neuronal Damage. J Alzheimers Dis 2019; 62:385-397. [PMID: 29439331 DOI: 10.3233/jad-170602] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammatory markers have been shown to predict neurocognitive outcomes in aging adults; however, the degree to which peripheral markers mirror the central nervous system remains unknown. We investigated the association between plasma and cerebrospinal fluid (CSF) markers of inflammation, and explored whether these markers independently predict CSF indicators of Alzheimer's disease (AD) pathology or neuronal damage. Plasma and CSF samples were analyzed for inflammatory markers in a cohort of asymptomatic older adults (n = 173). CSF samples were analyzed for markers of AD pathology (Aβ42, phosphorylated tau [p-tau], sAβPPβ) or neuronal damage (total tau; neurofilament light chain) (n = 147). Separate linear models for each analyte were conducted with CSF and plasma levels entered simultaneously as predictors and markers of AD pathology or neuronal damage as outcome measures. Strong associations were noted between CSF and plasma MIP-1β levels, and modest associations were observed for remaining analytes. With respect to AD pathology, higher levels of plasma and CSF IL-8, CSF MIP-1β, and CSF IP-10 were associated with higher levels of p-tau. Higher levels of CSF IL-8 were associated with higher levels of CSF Aβ42. Higher CSF sAβPPβ levels were associated with higher plasma markers only (IL-8; MCP-1). In terms of neuronal injury, higher levels of plasma and CSF IL-8, CSF IP-10, and CSF MIP-1β were associated with higher levels of CSF total tau. Exploratory analyses indicated that CSF Aβ42 modifies the relationship between plasma inflammatory levels and CSF tau levels. Results suggest that both plasma and CSF inflammatory markers independently relay integral information about AD pathology and neuronal damage.
Collapse
Affiliation(s)
- Brianne M Bettcher
- Departments of Neurosurgery and Neurology, University of Colorado Anschutz Medical Campus, Rocky Mountain Alzheimer's Disease Center, Aurora, CO, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ryan Fitch
- Department of Neurology, University of California San Francisco, Memory and Aging Center, San Francisco, CA, USA
| | - Kaitlin B Casaletto
- Department of Neurology, University of California San Francisco, Memory and Aging Center, San Francisco, CA, USA
| | - Kate S Heffernan
- Departments of Neurosurgery and Neurology, University of Colorado Anschutz Medical Campus, Rocky Mountain Alzheimer's Disease Center, Aurora, CO, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, Wm. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - John Neuhaus
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Joel H Kramer
- Department of Neurology, University of California San Francisco, Memory and Aging Center, San Francisco, CA, USA
| |
Collapse
|
150
|
Noailles A, Kutsyr O, Maneu V, Ortuño-Lizarán I, Campello L, de Juan E, Gómez-Vicente V, Cuenca N, Lax P. The Absence of Toll-Like Receptor 4 Mildly Affects the Structure and Function in the Adult Mouse Retina. Front Cell Neurosci 2019; 13:59. [PMID: 30873007 PMCID: PMC6401850 DOI: 10.3389/fncel.2019.00059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/05/2019] [Indexed: 12/26/2022] Open
Abstract
The innate immune Toll-like receptor (TLR) family plays essential roles in cell proliferation, survival and function of the central nervous system. However, the way in which TLRs contribute to the development and maintenance of proper retinal structure and function remains uncertain. In this work, we assess the effect of genetic TLR4 deletion on the morphology and function of the retina in mice. Visual acuity and retinal responsiveness were evaluated in TLR4 knockout and wild type C57BL/6J control mice by means of an optomotor test and electroretinography, respectively, from P20 to P360. Retinal structure was also analyzed in both strains using confocal and electron microscopy. ERG data showed impaired retinal responsiveness in TLR4 KO mice, in comparison to wild type animals. The amplitudes of the scotopic a-waves were less pronounced in TLR4-deficient mice than in wild-type animals from P30 to P360, and TLR4 KO mice presented scotopic b-wave amplitudes smaller than those of age-matched control mice at all ages studied (P20 to P360). Visual acuity was also relatively poorer in TLR4 KO as compared to C57BL/6J mice from P20 to P360, with significant differences at P30 and P60. Immunohistochemical analysis of retinal vertical sections showed no differences between TLR4 KO and C57BL/6J mice, in terms of either photoreceptor number or photoreceptor structure. Horizontal cells also demonstrated no morphological differences between TLR4 KO and wild-type mice. However, TLR4 KO mice exhibited a lower density of bipolar cells (15% less at P30) and thus fewer bipolar cell dendrites than the wild type control mouse, even though both confocal and electron microscopy images showed no morphologic abnormalities in the synaptic contacts between the photoreceptors and second order neurons. Microglial cell density was significantly lower (26% less at P30) in TLR4 KO mice as compared to wild-type control mice. These results suggest that TLR4 deletion causes functional alterations in terms of visual response and acuity, probably through the loss of bipolar cells and microglia, but this receptor is not essential for the processing of visual information in the retina.
Collapse
Affiliation(s)
- Agustina Noailles
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Isabel Ortuño-Lizarán
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Emilio de Juan
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Violeta Gómez-Vicente
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain.,Institute Ramón Margalef, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| |
Collapse
|