1
|
Xiang G, Sui M, Jiang N, Luo R, Xia J, Wei X, Lin Y, Li X, Cai Z, Lin J, Li S, Chen W, Zhao Y, Yang L. The progress in epidemiological, diagnosis and treatment of primary hemifacial spasm. Heliyon 2024; 10:e38600. [PMID: 39430510 PMCID: PMC11490810 DOI: 10.1016/j.heliyon.2024.e38600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/22/2024] Open
Abstract
Hemifacial Spasm is a neurological disorder characterized by persistent and rhythmic spasms of the facial muscles, significantly affecting the patient's quality of life. This condition can be classified into primary and secondary types; this article focuses on the characteristics of primary hemifacial spasm. Epidemiological studies indicate that the condition is more common in women, older adults, and individuals with posterior fossa stenosis or uneven blood flow dynamics, and is associated with gene expression related to demyelinating lesions. In terms of diagnosis, magnetic resonance imaging can show the location of arterial or venous compression on the facial nerve on a macroscopic level and reveal white matter lesions on a microscopic level. Additionally, optimized electrophysiological techniques can determine the type of neural excitation disorder from both central and peripheral perspectives, thereby improving detection rates. There are numerous treatment options available. Although early oral medications may have limited effectiveness, botulinum toxin injections can provide temporary relief. Future considerations include balancing injection costs with long-term efficacy. Microvascular decompression remains the preferred treatment approach and can be further optimized with endoscopic techniques. For refractory cases, alternative therapies such as facial nerve massage, radiofrequency techniques, rhizotomy, or acupuncture may be considered.
Collapse
Affiliation(s)
- Guangfa Xiang
- Zunyi Medical University Campus Zhuhai, Zhuhai, Guangdong, China
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, China
| | - Minghong Sui
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Naifu Jiang
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, China
| | - Rui Luo
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Jianwei Xia
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- School of Special Education and Rehabilitation, Binzhou Medical University, Yantai, Shandong, China
| | - Xinling Wei
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yifeng Lin
- Zunyi Medical University Campus Zhuhai, Zhuhai, Guangdong, China
| | - Xingyu Li
- Zunyi Medical University Campus Zhuhai, Zhuhai, Guangdong, China
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Zixiang Cai
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- College of Health Science, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Junxia Lin
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- College of Health Science, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Shipei Li
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- College of Health Science, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Wanyi Chen
- Department of Rehabilitation Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
- College of Health Science, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yang Zhao
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, China
- Standard Robots Co.,Ltd, Room 405, Building D, Huafeng International Robot Fusen Industrial Park, Hangcheng Avenue, Guxing Community, Xixiang Street, Baoan District, Shenzhen, Guangdong, China
| | - Lin Yang
- Zunyi Medical University Campus Zhuhai, Zhuhai, Guangdong, China
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Sekyi MT, Feri M, Desfor S, Atkinson KC, Golestany B, Beltran F, Tiwari-Woodruff SK. Demyelination and neurodegeneration early in experimental autoimmune encephalomyelitis contribute to functional deficits in the anterior visual pathway. Sci Rep 2024; 14:24048. [PMID: 39402114 PMCID: PMC11473523 DOI: 10.1038/s41598-024-73792-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/20/2024] [Indexed: 10/17/2024] Open
Abstract
Impaired visual function is a prevalent feature of optic neuritis (ON) in multiple sclerosis (MS). Abnormal visual evoked potential (VEP) findings of increased latencies, reduced amplitudes and abnormal waveforms as well as decreased retinal nerve fiber layer (RNFL) assessed by optical coherence tomography (OCT) are hallmarks of ON-induced visual dysfunction. Here we utilized the experimental autoimmune encephalomyelitis (EAE) mouse model of MS to investigate the functional and pathological progression during early (before any clinical symptoms), peak (initial maximal clinical symptoms), and late (chronic disease for > 3 weeks) disease stages. Demyelination and initial stages of axon damage were observed in early EAE. Significant demyelination, inflammation, increased axon damage and impaired P1/N2 amplitudes and latencies by VEP were seen in middle and late EAE groups. A decrease in RNFL thickness by OCT was observed only during late EAE. NanoString analysis of optic nerves from late EAE indicated elevated inflammation-related genes, reduced myelin-related genes, and changes in axon degeneration-related genes. Early inflammatory demyelination and functional deficits of the visual pathway, if untreated, may lead to severe irrecoverable axon damage in EAE. These studies potentially help explain the progression of visual dysfunction during MS.
Collapse
Affiliation(s)
- Maria T Sekyi
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Micah Feri
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Shane Desfor
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Kelley C Atkinson
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Batis Golestany
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Fernando Beltran
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA.
| |
Collapse
|
3
|
Cousineau JP, Dawe AM, Alpaugh M. Investigating the Interplay between Cardiovascular and Neurodegenerative Disease. BIOLOGY 2024; 13:764. [PMID: 39452073 PMCID: PMC11505144 DOI: 10.3390/biology13100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024]
Abstract
Neurological diseases, including neurodegenerative diseases (NDDs), are the primary cause of disability worldwide and the second leading cause of death. The chronic nature of these conditions and the lack of disease-modifying therapies highlight the urgent need for developing effective therapies. To accomplish this, effective models of NDDs are required to increase our understanding of underlying pathophysiology and for evaluating treatment efficacy. Traditionally, models of NDDs have focused on the central nervous system (CNS). However, evidence points to a relationship between systemic factors and the development of NDDs. Cardiovascular disease and related risk factors have been shown to modify the cerebral vasculature and the risk of developing Alzheimer's disease. These findings, combined with reports of changes to vascular density and blood-brain barrier integrity in other NDDs, such as Huntington's disease and Parkinson's disease, suggest that cardiovascular health may be predictive of brain function. To evaluate this, we explore evidence for disruptions to the circulatory system in murine models of NDDs, evidence of disruptions to the CNS in cardiovascular disease models and summarize models combining cardiovascular disruption with models of NDDs. In this study, we aim to increase our understanding of cardiovascular disease and neurodegeneration interactions across multiple disease states and evaluate the utility of combining model systems.
Collapse
Affiliation(s)
| | | | - Melanie Alpaugh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (J.P.C.); (A.M.D.)
| |
Collapse
|
4
|
Xu F, Gu P, Yuan H, Jiang L, Xie Y, Shi Q, Zhan Y. Analysis of risk factors related to the progression rate of hemifacial spasm. Front Neurol 2024; 15:1357280. [PMID: 38606273 PMCID: PMC11007217 DOI: 10.3389/fneur.2024.1357280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Introduction Although there have been many researches on the etiology and risk factors with the onset of hemifacial spasm, researches on the risk factors related to progression rate are limited. This study aims to analyze the risk factors related to the progression rate of hemifacial spasm. Methods The study enrolled 142 patients who underwent microvascular decompression for hemifacial spasm. Based on the duration and severity of symptoms, patients were classified into rapid progression group and slow progression group. To analyze risk factors, univariate and multivariate logistic regression analyses were conducted. Of 142 patients with hemifacial spasm, 90(63.3%) were classified as rapid progression group, 52(36.7%) were classified as slow progression group. Results In the univariate analysis, there were significant statistical differences between the two groups in terms of age of onset (P = 0.021), facial nerve angle (P < 0.01), hypertension (P = 0.01), presence of APOE ε4 expression (P < 0.01) and different degrees of brainstem compression in the Root Entry Zone (P < 0.01). In the multivariable analyses, there were significant statistical differences between the two groups in terms of age of symptom onset (P < 0.01 OR = 6.591), APOE ε4 (P < 0.01 OR = 5.691), brainstem compression (P = 0.006 OR = 5.620), and facial nerve angle (P < 0.01 OR = 5.758). Furthermore, we found no significant correlation between the severity of facial spasms and the progression rate of the disease (t = 2.47, P = 0.12>0.05). Conclusion According to our study, patients with facial nerve angle ≤ 96.5°, severer compression of the brainstem by offending vessels, an onset age > 45 years and positive expression of APOE ε4, may experience faster progression of hemifacial spasm.
Collapse
|
5
|
Wei W, Sun H, Yang B, Song E, Song Y. Coronal ApoE Protein Combines with LRP1 to Inactivate GSK3β That Mitigates Silica Nanoparticle-Induced Brain Lesion. ACS Chem Neurosci 2024; 15:808-815. [PMID: 38315060 DOI: 10.1021/acschemneuro.3c00728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024] Open
Abstract
Silica nanoparticles (SiO2 NPs) are widely used engineered materials that warrant their obvious environmental exposure risk. Our previous study has shown that different routes of SiO2 NP exposure on the glycogen synthase kinase 3 beta (GSK3β) activity were related to the serum proteins enriched on the surface of SiO2 NPs, which implied that a particular protein in the serum changed the inherent toxic behavior of SiO2 NPs and inhibited the activation of GSK3β by SiO2 NPs. Here, we identified that the SiO2 NP surface enriched a large amount of apolipoprotein E (ApoE), and the ApoE protein corona bound to the lipoprotein receptor-related protein 1 (LRP1) to inactivate GSK3β, thereby reducing the damage of SiO2 NPs to the brain. This work presented the first evidence that specific biocorona reduced the toxicity of SiO2 NPs at the molecular level, which helped to elucidate the role of specific corona components on nanotoxicity.
Collapse
Affiliation(s)
- Wei Wei
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Hang Sun
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Bingwei Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Rd, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| |
Collapse
|
6
|
Yi W, Lv D, Sun Y, Mu J, Lu X. Role of APOE in glaucoma. Biochem Biophys Res Commun 2024; 694:149414. [PMID: 38145596 DOI: 10.1016/j.bbrc.2023.149414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
Glaucoma is a chronic blinding eye disease caused by the progressive loss of retinal ganglion cells (RGCs). Currently, no clinically approved treatment can directly improve the survival rate of RGCs. The Apolipoprotein E (APOE) gene is closely related to the genetic risk of numerous neurodegenerative diseases and has become a hot topic in the field of neurodegenerative disease research in recent years. The optic nerve and retina are extensions of the brain's nervous system. The pathogenesis of retinal degenerative diseases is closely related to the degenerative diseases of the nerves in the brain. APOE consists of three alleles, ε4, ε3, and ε2, in a single locus. They have varying degrees of risk for glaucoma. APOE4 and the APOE gene deletion (APOE-/-) can reduce RGC loss. By contrast, APOE3 and the overall presence of APOE genes (APOE+/+) result in significant loss of RGC bodies and axons, increasing the risk of glaucoma RGCs death. Currently, there is no clear literature indicating that APOE2 is beneficial or harmful to glaucoma. This study summarises the mechanism of different APOE genes in glaucoma and speculates that APOE targeted intervention may be a promising method for protecting against RGCs loss in glaucoma.
Collapse
Affiliation(s)
- Wenhua Yi
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China.
| | - De Lv
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, China.
| | - Yue Sun
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China.
| | - Jingyu Mu
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China.
| | - Xuejing Lu
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China; Ineye Hospital of Chengdu University of TCM, Chengdu City, Sichuan province, China; Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu City, Sichuan province, China; Retinal Image Technology and Chronic Vascular Disease Prevention&Control and Collaborative Innovation Center, Chengdu City, Sichuan province, China.
| |
Collapse
|
7
|
Vigne S, Duc D, Peter B, Rebeaud J, Yersin Y, Ruiz F, Bressoud V, Collet TH, Pot C. Lowering blood cholesterol does not affect neuroinflammation in experimental autoimmune encephalomyelitis. J Neuroinflammation 2022; 19:42. [PMID: 35130916 PMCID: PMC8822860 DOI: 10.1186/s12974-022-02409-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 02/01/2022] [Indexed: 01/07/2023] Open
Abstract
Background Multiple sclerosis (MS) is a chronic disabling disease of the central nervous system (CNS) commonly affecting young adults. There is increasing evidence that environmental factors are important in the development and course of MS. The metabolic syndrome (MetS) which comprises dyslipidemia has been associated with a worse outcome in MS disease. Furthermore, the lipid-lowering drug class of statins has been proposed to improve MS disease course. However, cholesterol is also rate-limiting for myelin biogenesis and promotes remyelination in MS animal models. Thus, the impact of circulating blood cholesterol levels during the disease remains debated and controversial. Methods We assessed the role of circulating cholesterol on the murine model of MS, the experimental autoimmune encephalomyelitis (EAE) disease using two different approaches: (1) the mouse model of familial hypercholesterolemia induced by low-density lipoprotein receptor (LDLr) deficiency, and (2) the use of the monoclonal anti-PCSK9 neutralizing antibody alirocumab, which reduces LDLr degradation and consequently lowers blood levels of cholesterol. Results Elevated blood cholesterol levels induced by LDLr deficiency did not worsen clinical symptoms of mice during EAE. In addition, we observed that the anti-PCSK9 antibody alirocumab did not influence EAE disease course, nor modulate the immune response in EAE. Conclusions These findings suggest that blood cholesterol level has no direct role in neuro-inflammatory diseases and that the previously shown protective effects of statins in MS are not related to circulating cholesterol. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02409-x.
Collapse
Affiliation(s)
- Solenne Vigne
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Donovan Duc
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Benjamin Peter
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Jessica Rebeaud
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Yannick Yersin
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Florian Ruiz
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Valentine Bressoud
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Tinh-Hai Collet
- Service of Endocrinology, Diabetes, Nutrition and Therapeutic Education, Department of Medicine, Geneva University Hospitals (HUG), Rue Gabrielle-Perret-Gentil 4, 1211, Geneva 14, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Neuroscience Research Center and Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| |
Collapse
|
8
|
Mishra A, Wang Y, Yin F, Vitali F, Rodgers KE, Soto M, Mosconi L, Wang T, Brinton RD. A tale of two systems: Lessons learned from female mid-life aging with implications for Alzheimer's prevention & treatment. Ageing Res Rev 2022; 74:101542. [PMID: 34929348 PMCID: PMC8884386 DOI: 10.1016/j.arr.2021.101542] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 12/05/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
Neurological aging is frequently viewed as a linear process of decline, whereas in reality, it is a dynamic non-linear process. The dynamic nature of neurological aging is exemplified during midlife in the female brain. To investigate fundamental mechanisms of midlife aging that underlie risk for development of Alzheimer's disease (AD) in late life, we investigated the brain at greatest risk for the disease, the aging female brain. Outcomes of our research indicate that mid-life aging in the female is characterized by the emergence of three phases: early chronological (pre-menopause), endocrinological (peri-menopause) and late chronological (post-menopause) aging. The endocrinological aging program is sandwiched between early and late chronological aging. Throughout the three stages of midlife aging, two systems of biology, metabolic and immune, are tightly integrated through a network of signaling cascades. The network of signaling between these two systems of biology underlie an orchestrated sequence of adaptative starvation responses that shift the brain from near exclusive dependence on a single fuel, glucose, to utilization of an auxiliary fuel derived from lipids, ketone bodies. The dismantling of the estrogen control of glucose metabolism during mid-life aging is a critical contributor to the shift in fuel systems and emergence of dynamic neuroimmune phenotype. The shift in fuel reliance, puts the largest reservoir of local fatty acids, white matter, at risk for catabolism as a source of lipids to generate ketone bodies through astrocytic beta oxidation. APOE4 genotype accelerates the tipping point for emergence of the bioenergetic crisis. While outcomes derived from research conducted in the female brain are not directly translatable to the male brain, the questions addressed in a female centric program of research are directly applicable to investigation of the male brain. Like females, males with AD exhibit deficits in the bioenergetic system of the brain, activation of the immune system and hallmark Alzheimer's pathologies. The drivers and trajectory of mechanisms underlying neurodegeneration in the male brain will undoubtedly share common aspects with the female in addition to factors unique to the male. Preclinical and clinical evidence indicate that midlife endocrine aging can also be a transitional bridge to autoimmune disorders. Collectively, the data indicate that endocrinological aging is a critical period "tipping point" in midlife which can initiate emergence of the prodromal stage of late-onset-Alzheimer's disease. Interventions that target both immune and metabolic shifts that occur during midlife aging have the potential to alter the trajectory of Alzheimer's risk in late life. Further, to achieve precision medicine for AD, chromosomal sex is a critical variable to consider along with APOE genotype, other genetic risk factors and stage of disease.
Collapse
Affiliation(s)
- Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Fei Yin
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Francesca Vitali
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Kathleen E Rodgers
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Maira Soto
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Tian Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Roberta D Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA.
| |
Collapse
|
9
|
Salamero-Boix A, Schulz M, Sevenich L. Activation of Microglia and Macrophages in Neurodegenerative Diseases. NEUROMETHODS 2022:11-38. [DOI: 10.1007/978-1-0716-1712-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Zhou J, Jiang L, Yuan S, Huang J, Shi Q, Xie Y, Deng B, Zhan Y. Association Study of Apolipoprotein E Gene Polymorphism With Incidence and Delayed Resolution of Hemifacial Spasm. Front Neurol 2021; 12:760126. [PMID: 34975724 PMCID: PMC8714662 DOI: 10.3389/fneur.2021.760126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/18/2021] [Indexed: 11/26/2022] Open
Abstract
Objective: This study investigates the correlation between Apolipoprotein E gene (APOE) polymorphism and the incidence and delayed resolution of hemifacial spasms. Methods: The APOE genotypes of 151 patients with hemifacial spasm and 73 control cases were determined by cleaved amplification polymorphism sequence-tagged sites. The distribution of three APOE alleles (ε2, ε3, and ε4) in two groups and the delayed resolution rate in 6 genotypes were calculated and statistically analyzed. Results: The proportion of patients with APOE ε3/ε4 genotype in the hemifacial spasm group (25.17%) was significantly higher than that in the control group (12.33%) (P = 0.027). In terms of allele frequency, the proportion of the APOE ε4 allele in the hemifacial spasm group (15.56%) was significantly higher than that in the control group (6.85%) (P = 0.009). Meanwhile, the proportion of APOE ε4 allele carriers in the hemifacial spasm group (29.80%) was significantly higher than that in the control group (13.7%) (P = 0.009). Logistic regression analysis showed that the ε4 allele significantly increased the incidence of hemifacial spasm (OR 2.675, 95%CI 1.260-5.678, P = 0.010). Among the 32 patients with a delayed resolution, the ε3/ε3 and ε3/ε4 had the highest proportion in 6 genotypes. The delayed resolution rate of APOE ε3/ε4 (34.21%) was significantly higher than APOE ε3/ε3 (17.78%) (P < 0.05). The delayed resolution rate of APOE ε4 carriers was the highest (33.33%) in the 3 allele carriers, but there was no significant difference among the 3 allele carriers (P = 0.065). Conclusion: The polymorphism of APOE is relevant to the incidence rate of hemifacial spasms. APOE ε4 allele increases the incidence of hemifacial spasm. The APOE ε4 allele may promote the occurrence of delayed resolution.
Collapse
|
11
|
Gong K, Chen Y, Liu W, Wang Z. Global research trends of Apolipoprotein E in central nervous system: A scientometric analysis. Int Immunopharmacol 2021; 98:107919. [PMID: 34217139 DOI: 10.1016/j.intimp.2021.107919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/25/2021] [Accepted: 06/21/2021] [Indexed: 10/21/2022]
Abstract
Apolipoprotein E (apoE, protein; APOE, gene) involves in cholesterol recycling and redistribution by mediating lipoprotein pathways unique to central nervous system (CNS), which is a potential therapeutic target for diseases. We visually analyzed the research hotspots of APOE related to CNS in this work, by scientometric analysis from the Web of Science Core Collection (WOSCC) database over the past two decades. A total of 25,719 references of "APOE" and 836 references of "APOE in CNS" were retrieved from the WOSCC on October 26, 2020, and then VOSviewer 1.6.15, Citespace 5.7.R2 were used for visual analysis. Over the last two decades, the research on the field of APOE in CNS is not faddish. Although many funds, organizations, and scholars were affiliated in this field, organizations and scholars, especially the top teams in this field, still lacked close cooperation with other teams around the world. Few articles with high citations had been published in the last decade, but recent studies still lacked scale and breakthrough, and the keywords associated with APOE appeared more outdated. However, the current researches have not fully elucidated the crosstalk between APOE and neuroinflammation in CNS, some new ideas may rekindle the research enthusiasm of scholars. Although the field of APOE in CNS appeared more outdated. Based on keyword analysis, we hypothesized new ideas for further investigation of neuroinflammation would light the interest of APOE in CNS for the scholars. The crosstalk between ApoE and inflammasome may be the focus of future researches. How APOE modulates the time course or intensity of the inflammasome activation, inflammatory response (proinflammatory or anti-inflammatory), and pathological process of CNS disease deserves future attention in both basic and clinical studies. More apoE/APOE-targeted pharmacological interventions will be available for preclinical experiments and clinical trials and bring hope for patients with CNS diseases.
Collapse
Affiliation(s)
- Kai Gong
- Trauma Center, First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China; Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China
| | - Yuhua Chen
- Trauma Center, First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China; Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China
| | - Wei Liu
- Trauma Center, First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China; Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China.
| | - Zhanxiang Wang
- Trauma Center, First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China; Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, 55 Zhenhai Rd, Xiamen ,361003, Fujian, China.
| |
Collapse
|
12
|
Davis AA, Inman CE, Wargel ZM, Dube U, Freeberg BM, Galluppi A, Haines JN, Dhavale DD, Miller R, Choudhury FA, Sullivan PM, Cruchaga C, Perlmutter JS, Ulrich JD, Benitez BA, Kotzbauer PT, Holtzman DM. APOE genotype regulates pathology and disease progression in synucleinopathy. Sci Transl Med 2021; 12:12/529/eaay3069. [PMID: 32024799 DOI: 10.1126/scitranslmed.aay3069] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Apolipoprotein E (APOE) ε4 genotype is associated with increased risk of dementia in Parkinson's disease (PD), but the mechanism is not clear, because patients often have a mixture of α-synuclein (αSyn), amyloid-β (Aβ), and tau pathologies. APOE ε4 exacerbates brain Aβ pathology, as well as tau pathology, but it is not clear whether APOE genotype independently regulates αSyn pathology. In this study, we generated A53T αSyn transgenic mice (A53T) on Apoe knockout (A53T/EKO) or human APOE knockin backgrounds (A53T/E2, E3, and E4). At 12 months of age, A53T/E4 mice accumulated higher amounts of brainstem detergent-insoluble phosphorylated αSyn compared to A53T/EKO and A53T/E3; detergent-insoluble αSyn in A53T/E2 mice was undetectable. By immunohistochemistry, A53T/E4 mice displayed a higher burden of phosphorylated αSyn and reactive gliosis compared to A53T/E2 mice. A53T/E2 mice exhibited increased survival and improved motor performance compared to other APOE genotypes. In a complementary model of αSyn spreading, striatal injection of αSyn preformed fibrils induced greater accumulation of αSyn pathology in the substantia nigra of A53T/E4 mice compared to A53T/E2 and A53T/EKO mice. In two separate cohorts of human patients with PD, APOE ε4/ε4 individuals showed the fastest rate of cognitive decline over time. Our results demonstrate that APOE genotype directly regulates αSyn pathology independent of its established effects on Aβ and tau, corroborate the finding that APOE ε4 exacerbates pathology, and suggest that APOE ε2 may protect against αSyn aggregation and neurodegeneration in synucleinopathies.
Collapse
Affiliation(s)
- Albert A Davis
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA. .,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Casey E Inman
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Zachary M Wargel
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Umber Dube
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
| | - Brittany M Freeberg
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Alexander Galluppi
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Jessica N Haines
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Dhruva D Dhavale
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Rebecca Miller
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Fahim A Choudhury
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Patrick M Sullivan
- Department of Medicine, Duke University Medical Center, Durham VAMC and Geriatric Research Clinical Center, Durham, NC 27705, USA
| | - Carlos Cruchaga
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA.,Departments of Neuroscience and Radiology, Programs in Physical and Occupational Therapy, Washington University, St. Louis, MO 63110, USA
| | - Jason D Ulrich
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Bruno A Benitez
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University, St. Louis, MO 63110, USA
| | - Paul T Kotzbauer
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA.,Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110, USA. .,Department of Neurology, Washington University, St. Louis, MO 63110, USA.,Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
13
|
Vergallo A, Lista S, Lemercier P, Chiesa PA, Zetterberg H, Blennow K, Potier MC, Habert MO, Baldacci F, Cavedo E, Caraci F, Dubois B, Hampel H. Association of plasma YKL-40 with brain amyloid-β levels, memory performance, and sex in subjective memory complainers. Neurobiol Aging 2020; 96:22-32. [PMID: 32920471 DOI: 10.1016/j.neurobiolaging.2020.07.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 06/28/2020] [Accepted: 07/09/2020] [Indexed: 02/08/2023]
Abstract
Neuroinflammation, a key early pathomechanistic alteration of Alzheimer's disease, may represent either a detrimental or a compensatory mechanism or both (according to the disease stage). YKL-40, a glycoprotein highly expressed in differentiated glial cells, is a candidate biomarker for in vivo tracking neuroinflammation in humans. We performed a longitudinal study in a monocentric cohort of cognitively healthy individuals at risk for Alzheimer's disease exploring whether age, sex, and the apolipoprotein E ε4 allele affect plasma YKL-40 concentrations. We investigated whether YKL-40 is associated with brain amyloid-β (Aβ) deposition, neuronal activity, and neurodegeneration as assessed via neuroimaging biomarkers. Finally, we investigated whether YKL-40 may predict cognitive performance. We found an age-associated increase of YKL-40 and observed that men display higher concentrations than women, indicating a potential sexual dimorphism. Moreover, YKL-40 was positively associated with memory performance and negatively associated with brain Aβ deposition (but not with metabolic signal). Consistent with translational studies, our results suggest a potentially protective effect of glia on incipient brain Aβ accumulation and neuronal homeostasis.
Collapse
Affiliation(s)
- Andrea Vergallo
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U1127, CNRS UMR 7225, Paris, France; Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France.
| | - Simone Lista
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U1127, CNRS UMR 7225, Paris, France; Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Pablo Lemercier
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U1127, CNRS UMR 7225, Paris, France; Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Patrizia A Chiesa
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U1127, CNRS UMR 7225, Paris, France; Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - 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 Neurodegenerative Disease, UCL Institute of Neurology, London, UK; UK Dementia Research Institute, 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
| | - Marie-Claude Potier
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Marie-Odile Habert
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France; Centre pour l'Acquisition et le Traitement des Images, Paris, France; Département de Médecine Nucléaire, AP-HP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Filippo Baldacci
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U1127, CNRS UMR 7225, Paris, France; Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France; Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Enrica Cavedo
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U1127, CNRS UMR 7225, Paris, France; Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy; Oasi Research Institute - IRCCS, Troina, Italy
| | - Bruno Dubois
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute (ICM), INSERM U1127, CNRS UMR 7225, Paris, France; Department of Neurology, Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Harald Hampel
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | | | | | | |
Collapse
|
14
|
Rui W, Li S, Xiao H, Xiao M, Shi J. Baicalein Attenuates Neuroinflammation by Inhibiting NLRP3/caspase-1/GSDMD Pathway in MPTP Induced Mice Model of Parkinson's Disease. Int J Neuropsychopharmacol 2020; 23:pyaa060. [PMID: 32761175 PMCID: PMC7745250 DOI: 10.1093/ijnp/pyaa060] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/12/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammasome-induced neuroinflammation is a major pathogenic mechanism underlying the degeneration of nigral dopaminergic neurons in Parkinson's disease (PD). Baicalein is a flavonoid isolated from the traditional Chinese medicinal herbal Scutellaria baicalensis Georgi with known anti-inflammatory and neuroprotective efficacy in models of neurodegenerative diseases, including PD. However, its effects on inflammasome-induced neuroinflammation during PD remain unclear. METHODS We used N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to induce PD-like pathology in mice. Behavioral assessments including the pole test, rotarod test and open filed test were conducted to evaluate the effects of baicalein on MPTP-induced motor dysfunction. The efficacies of baicalein against MPTP-induced dopaminergic neuron loss and glial cell activation in the substantia nigra compact (SNc) were examined by immunohistochemistry, effects on proinflammatory cytokines by qPCR and enzyme-linked immunosorbent assay (ELISA), effects on inflammasome pathway activation by immunoblotting and flow cytometry. RESULTS Administration of baicalein reversed MPTP-induced motor dysfunction, loss of dopaminergic neurons, and pro-inflammatory cytokine elevation. Baicalein also inhibited NLRP3 and caspase-1 activation and suppressed gasdermin D (GSDMD)-dependent pyroptosis. Additionally, baicalein inhibited the activation and proliferation of disease-associated proinflammatory microglia. CONCLUSIONS These findings suggest that baicalein can reverse MPTP-induced neuroinflammation in mice by suppressing NLRP3/caspase-1/GSDMD pathway. Our study provides potential insight of baicalein in PD therapy.
Collapse
Affiliation(s)
- Wenjuan Rui
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neuro-Psychiatric Institute, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sheng Li
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hong Xiao
- Department of Neuro-Psychiatric Institute, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingping Shi
- Department of Neurology, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
15
|
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: 377] [Impact Index Per Article: 62.8] [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
|
16
|
Abstract
BACKGROUND The growing body of evidence indicating the heterogeneity of Alzheimer's disease (AD), coupled with disappointing clinical studies directed at a fit-for-all therapy, suggest that the development of a single magic cure suitable for all cases may not be possible. This calls for a shift in paradigm where targeted treatment is developed for specific AD subpopulations that share distinct genetic or pathological properties. Apolipoprotein E4 (apoE4), the most prevalent genetic risk factor of AD, is expressed in more than half of AD patients and is thus an important possible AD therapeutic target. REVIEW This review focuses initially on the pathological effects of apoE4 in AD, as well as on the corresponding cellular and animal models and the suggested cellular and molecular mechanisms which mediate them. The second part of the review focuses on recent apoE4-targeted (from the APOE gene to the apoE protein and its interactors) therapeutic approaches that have been developed in animal models and are ready to be translated to human. Further, the issue of whether the pathological effects of apoE4 are due to loss of protective function or due to gain of toxic function is discussed herein. It is possible that both mechanisms coexist, with certain constituents of the apoE4 molecule and/or its downstream signaling mediating a toxic effect, while others are associated with a loss of protective function. CONCLUSION ApoE4 is a promising AD therapeutic target that remains understudied. Recent studies are now paving the way for effective apoE4-directed AD treatment approaches.
Collapse
|
17
|
Tay TL, Sagar, Dautzenberg J, Grün D, Prinz M. Unique microglia recovery population revealed by single-cell RNAseq following neurodegeneration. Acta Neuropathol Commun 2018; 6:87. [PMID: 30185219 PMCID: PMC6123921 DOI: 10.1186/s40478-018-0584-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/11/2018] [Indexed: 11/17/2022] Open
Abstract
Microglia are brain immune cells that constantly survey their environment to maintain homeostasis. Enhanced microglial reactivity and proliferation are typical hallmarks of neurodegenerative diseases. Whether specific disease-linked microglial subsets exist during the entire course of neurodegeneration, including the recovery phase, is currently unclear. Taking a single-cell RNA-sequencing approach in a susceptibility gene-free model of nerve injury, we identified a microglial subpopulation that upon acute neurodegeneration shares a conserved gene regulatory profile compared to previously reported chronic and destructive neurodegeneration transgenic mouse models. Our data also revealed rapid shifts in gene regulation that defined microglial subsets at peak and resolution of neurodegeneration. Finally, our discovery of a unique transient microglial subpopulation at the onset of recovery may provide novel targets for modulating microglia-mediated restoration of brain health.
Collapse
|
18
|
Ulrich JD, Ulland TK, Mahan TE, Nyström S, Nilsson KP, Song WM, Zhou Y, Reinartz M, Choi S, Jiang H, Stewart FR, Anderson E, Wang Y, Colonna M, Holtzman DM. ApoE facilitates the microglial response to amyloid plaque pathology. J Exp Med 2018; 215:1047-1058. [PMID: 29483128 PMCID: PMC5881464 DOI: 10.1084/jem.20171265] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 12/28/2017] [Accepted: 02/07/2018] [Indexed: 01/22/2023] Open
Abstract
Increasing evidence suggests that apoE influences the innate immune response in neurodegeneration. Here, Ulrich et al. report that apoE influences amyloid plaque morphology and the microglial response to amyloid plaques, along with plaque-associated neuronal toxicity. One of the hallmarks of Alzheimer’s disease is the presence of extracellular diffuse and fibrillar plaques predominantly consisting of the amyloid-β (Aβ) peptide. Apolipoprotein E (ApoE) influences the deposition of amyloid pathology through affecting the clearance and aggregation of monomeric Aβ in the brain. In addition to influencing Aβ metabolism, increasing evidence suggests that apoE influences microglial function in neurodegenerative diseases. Here, we characterize the impact that apoE has on amyloid pathology and the innate immune response in APPPS1ΔE9 and APPPS1-21 transgenic mice. We report that Apoe deficiency reduced fibrillar plaque deposition, consistent with previous studies. However, fibrillar plaques in Apoe-deficient mice exhibited a striking reduction in plaque compaction. Hyperspectral fluorescent imaging using luminescent conjugated oligothiophenes identified distinct Aβ morphotypes in Apoe-deficient mice. We also observed a significant reduction in fibrillar plaque–associated microgliosis and activated microglial gene expression in Apoe-deficient mice, along with significant increases in dystrophic neurites around fibrillar plaques. Our results suggest that apoE is critical in stimulating the innate immune response to amyloid pathology.
Collapse
Affiliation(s)
- Jason D Ulrich
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - Tyler K Ulland
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Thomas E Mahan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - Sofie Nyström
- Department of Chemistry, IFM, Linköping University, Linköping, Sweden
| | - K Peter Nilsson
- Department of Chemistry, IFM, Linköping University, Linköping, Sweden
| | - Wilbur M Song
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Mariska Reinartz
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Radboud University Nijmegen, Nijmegen, Netherlands
| | - Seulah Choi
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - Hong Jiang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - Floy R Stewart
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - Elise Anderson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - Yaming Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO.,Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO .,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
19
|
The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity 2017; 47:566-581.e9. [PMID: 28930663 DOI: 10.1016/j.immuni.2017.08.008] [Citation(s) in RCA: 1696] [Impact Index Per Article: 212.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/06/2017] [Accepted: 08/17/2017] [Indexed: 12/12/2022]
Abstract
Microglia play a pivotal role in the maintenance of brain homeostasis but lose homeostatic function during neurodegenerative disorders. We identified a specific apolipoprotein E (APOE)-dependent molecular signature in microglia from models of amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and Alzheimer's disease (AD) and in microglia surrounding neuritic β-amyloid (Aβ)-plaques in the brains of people with AD. The APOE pathway mediated a switch from a homeostatic to a neurodegenerative microglia phenotype after phagocytosis of apoptotic neurons. TREM2 (triggering receptor expressed on myeloid cells 2) induced APOE signaling, and targeting the TREM2-APOE pathway restored the homeostatic signature of microglia in ALS and AD mouse models and prevented neuronal loss in an acute model of neurodegeneration. APOE-mediated neurodegenerative microglia had lost their tolerogenic function. Our work identifies the TREM2-APOE pathway as a major regulator of microglial functional phenotype in neurodegenerative diseases and serves as a novel target that could aid in the restoration of homeostatic microglia.
Collapse
|
20
|
Expression profiles of cholesterol metabolism-related genes are altered during development of experimental autoimmune encephalomyelitis in the rat spinal cord. Sci Rep 2017; 7:2702. [PMID: 28578430 PMCID: PMC5457442 DOI: 10.1038/s41598-017-02638-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/12/2017] [Indexed: 12/04/2022] Open
Abstract
Increased evidence suggests that dysregulation of cholesterol metabolism may be a key event contributing to progression of multiple sclerosis (MS). Using an experimental autoimmune encephalomyelitis (EAE) model of MS we revealed specific changes in the mRNA and protein expression of key molecules involved in the maintaining of cholesterol homeostasis in the rat spinal cord: 3-hydroxy-3-methylglutaryl-coenzyme-A reductase (HMGCR), apolipoprotein E (ApoE) and cholesterol 24-hydroxylase (CYP46A1) during the course of disease. The presence of myelin lipid debris was seen only at the peak of EAE in demyelination loci being efficiently removed during the recovery period. Since CYP46A1 is responsible for removal of cholesterol excess, we performed a detailed profiling of CYP46A1 expression and revealed regional and temporal specificities in its distribution. Double immunofluorescence staining demonstrated CYP46A1 localization with neurons, infiltrated macrophages, microglia and astrocytes in the areas of demyelination, suggesting that these cells play a role in cholesterol turnover in EAE. We propose that alterations in the regulation of cholesterol metabolism at the onset and peak of EAE may add to the progression of disease, while during the recovery period may have beneficial effects contributing to the regeneration of myelin sheath and restoration of neuronal function.
Collapse
|
21
|
Leichner TM, Satake A, Harrison VS, Tanaka Y, Archambault AS, Kim BS, Siracusa MC, Leonard WJ, Naji A, Wu GF, Artis D, Kambayashi T. Skin-derived TSLP systemically expands regulatory T cells. J Autoimmun 2017; 79:39-52. [PMID: 28126203 PMCID: PMC5386815 DOI: 10.1016/j.jaut.2017.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 12/16/2022]
Abstract
Regulatory T cells (Tregs) are a subset of CD4+ T cells with suppressive function and are critical for limiting inappropriate activation of T cells. Hence, the expansion of Tregs is an attractive strategy for the treatment of autoimmune diseases. Here, we demonstrate that the skin possesses the remarkable capacity to systemically expand Treg numbers by producing thymic stromal lymphopoietin (TSLP) in response to vitamin D receptor stimulation. An ∼2-fold increase in the proportion and absolute number of Tregs was observed in mice treated topically but not systemically with the Vitamin D3 analog MC903. This expansion of Tregs was dependent on TSLP receptor signaling but not on VDR signaling in hematopoietic cells. However, TSLP receptor expression by Tregs was not required for their proliferation. Rather, skin-derived TSLP promoted Treg expansion through dendritic cells. Importantly, treatment of skin with MC903 significantly lowered the incidence of autoimmune diabetes in non-obese diabetic mice and attenuated disease score in experimental autoimmune encephalomyelitis. Together, these data demonstrate that the skin has the remarkable potential to control systemic immune responses and that Vitamin D-mediated stimulation of skin could serve as a novel strategy to therapeutically modulate the systemic immune system for the treatment of autoimmunity.
Collapse
MESH Headings
- Animals
- Biomarkers
- Cholecalciferol/analogs & derivatives
- Cholecalciferol/pharmacology
- Cytokines/metabolism
- Cytokines/pharmacology
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Mice
- Mice, Knockout
- Mice, Transgenic
- Models, Biological
- Signal Transduction/drug effects
- Skin/immunology
- Skin/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Thymic Stromal Lymphopoietin
Collapse
Affiliation(s)
- Theresa M Leichner
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, United States
| | - Atsushi Satake
- First Department of Internal Medicine, Kansai Medical University, Japan
| | | | - Yukinori Tanaka
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, United States
| | - Angela S Archambault
- Department of Neurology, Washington University School of Medicine, United States
| | - Brian S Kim
- Division of Dermatology, Department of Medicine, Department of Anesthesiology, Department of Pathology and Immunology, Center for the Study of Itch, United States
| | - Mark C Siracusa
- Center for Immunity and Inflammation, Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, United States
| | | | - Ali Naji
- Department of Surgery, University of Pennsylvania, United States
| | - Gregory F Wu
- Department of Neurology, Washington University School of Medicine, United States
| | - David Artis
- Department of Medicine, Weill Cornell Medical College, United States
| | - Taku Kambayashi
- Department of Pathology & Laboratory Medicine, University of Pennsylvania, United States.
| |
Collapse
|
22
|
Geraldes R, Esiri MM, DeLuca GC, Palace J. Age-related small vessel disease: a potential contributor to neurodegeneration in multiple sclerosis. Brain Pathol 2017; 27:707-722. [PMID: 27864848 DOI: 10.1111/bpa.12460] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 11/17/2016] [Indexed: 12/18/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disorder of the central nervous system wherein, after an initial phase of transient neurological defects, slow neurological deterioration due to progressive neuronal loss ensues. Age is a major determinant of MS progression onset and disability. Over the past years, several mechanisms have been proposed to explain the key drivers of neurodegeneration and disability accumulation in MS. However, the effect of commonly encountered age-related cerebral vessel disease, namely small vessel disease (SVD), has been largely neglected and constitutes the aim of this review. SVD shares some features with MS, that is, white matter demyelination and brain atrophy, and has been shown to contribute to the neuronal damage seen in vascular cognitive impairment. Several lines of evidence suggest that an interaction between MS and SVD may influence MS-related neurodegeneration. SVD may contribute to hypoperfusion, reduced vascular reactivity and tissue hypoxia, features seen in MS. Venule and endothelium abnormalities have been documented in MS but the role of arterioles and of other neurovascular unit structures, such as the pericyte, has not been explored. Vascular risk factors (VRF) have recently been associated with faster progression in MS, though the mechanisms are unclear since very few studies have addressed the impact of VRF and SVD on MS imaging and pathology outcomes. Therapeutic agents targeting the microvasculature and the neurovascular unit may impact both SVD and MS and may benefit patients with dual pathology.
Collapse
Affiliation(s)
- Ruth Geraldes
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| | - Margaret M Esiri
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
23
|
Neuroprotective pentapeptide CN-105 improves functional and histological outcomes in a murine model of intracerebral hemorrhage. Sci Rep 2016; 6:34834. [PMID: 27713572 PMCID: PMC5054364 DOI: 10.1038/srep34834] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/24/2016] [Indexed: 01/18/2023] Open
Abstract
Presently, no pharmacological treatments have been demonstrated to improve long-term functional outcomes following intracerebral hemorrhage (ICH). Clinical evidence associates apolipoprotein E (apoE) genotype with ICH incidence and outcome. While apoE modifies neuroinflammatory responses through its adaptive role in glial downregulation, intact apoE holoprotein is too large to cross the blood-brain barrier (BBB). Therefore, we developed a 5-amino acid peptide – CN-105 – that mimics the polar face of the apoE helical domain involved in receptor interactions. In the current study, we investigated the therapeutic potential of CN-105 in a mouse model of ICH. Three doses of CN-105 (0.05 mg/kg) was administered by tail vein injection within 24 hours after ICH induction. Functional assessment showed durable improvement in vestibulomotor performance after CN-105 treatment, as quantified by increased Rotarod latencies on Days 1–5 post-ICH, and long-term improvement in neurocognitive performance, as quantified by reduced Morris water maze latencies on Days 29–32 post-ICH. Further, brain water content was significantly reduced, neuroinflammation was decreased and hippocampal CA3 neuronal survival was increased, although hemorrhage volume was not affected by CN-105. We concluded, therefore, that pentapeptide CN-105 improved short- and long-term neurobehavioral outcomes in a murine model of ICH, suggesting therapeutic potential for patients with acute ICH.
Collapse
|
24
|
Schrewe L, Lill CM, Liu T, Salmen A, Gerdes LA, Guillot-Noel L, Akkad DA, Blaschke P, Graetz C, Hoffjan S, Kroner A, Demir S, Böhme A, Rieckmann P, ElAli A, Hagemann N, Hermann DM, Cournu-Rebeix I, Zipp F, Kümpfel T, Buttmann M, Zettl UK, Fontaine B, Bertram L, Gold R, Chan A. Investigation of sex-specific effects of apolipoprotein E on severity of EAE and MS. J Neuroinflammation 2015; 12:234. [PMID: 26669675 PMCID: PMC4681148 DOI: 10.1186/s12974-015-0429-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/04/2015] [Indexed: 12/20/2022] Open
Abstract
Background Despite pleiotropic immunomodulatory effects of apolipoprotein E (apoE) in vitro, its effects on the clinical course of experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS) are still controversial. As sex hormones modify immunomodulatory apoE functions, they may explain contentious findings. This study aimed to investigate sex-specific effects of apoE on disease course of EAE and MS. Methods MOG35-55 induced EAE in female and male apoE-deficient mice was assessed clinically and histopathologically. apoE expression was investigated by qPCR. The association of the MS severity score (MSSS) and APOE rs429358 and rs7412 was assessed across 3237 MS patients using linear regression analyses. Results EAE disease course was slightly attenuated in male apoE-deficient (apoE−/−) mice compared to wildtype mice (cumulative median score: apoE−/− = 2 [IQR 0.0–4.5]; wildtype = 4 [IQR 1.0–5.0]; n = 10 each group, p = 0.0002). In contrast, EAE was more severe in female apoE−/− mice compared to wildtype mice (cumulative median score: apoE−/− = 3 [IQR 2.0–4.5]; wildtype = 3 [IQR 0.0–4.0]; n = 10, p = 0.003). In wildtype animals, apoE expression during the chronic EAE phase was increased in both females and males (in comparison to naïve animals; p < 0.001). However, in MS, we did not observe a significant association between MSSS and rs429358 or rs7412, neither in the overall analyses nor upon stratification for sex. Conclusions apoE exerts moderate sex-specific effects on EAE severity. However, the results in the apoE knock-out model are not comparable to effects of polymorphic variants in the human APOE gene, thus pinpointing the challenge of translating findings from the EAE model to the human disease. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0429-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- L Schrewe
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstr. 56, Bochum, 44791, Germany.
| | - C M Lill
- Platform for Genome Analytics, Institutes of Neurogenetics & Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany. .,Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany. .,Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany.
| | - T Liu
- Max Planck Institute for Human Development, Berlin, Germany.
| | - A Salmen
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstr. 56, Bochum, 44791, Germany.
| | - L A Gerdes
- Institute for Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University, Munich, Germany.
| | - L Guillot-Noel
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France.
| | - D A Akkad
- Department of Human Genetics, Ruhr-University Bochum, Bochum, Germany.
| | - P Blaschke
- Department of Neurology, University of Rostock, Rostock, Germany.
| | - C Graetz
- Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - S Hoffjan
- Department of Human Genetics, Ruhr-University Bochum, Bochum, Germany.
| | - A Kroner
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada. .,Department of Neurology, University of Würzburg, Würzburg, Germany.
| | - S Demir
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstr. 56, Bochum, 44791, Germany.
| | - A Böhme
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstr. 56, Bochum, 44791, Germany.
| | - P Rieckmann
- Department of Neurology, University of Würzburg, Würzburg, Germany.
| | - A ElAli
- Neuroscience Axis, Research Center of CHU de Québec-CHUL, Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec City, QC, Canada. .,Department of Vascular Neurology and Dementia, University of Duisburg-Essen, Essen, Germany.
| | - N Hagemann
- Department of Vascular Neurology and Dementia, University of Duisburg-Essen, Essen, Germany.
| | - D M Hermann
- Department of Vascular Neurology and Dementia, University of Duisburg-Essen, Essen, Germany.
| | - I Cournu-Rebeix
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France.
| | - F Zipp
- Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| | - T Kümpfel
- Institute for Clinical Neuroimmunology, Medical Campus Grosshadern, Ludwig Maximilian University, Munich, Germany.
| | - M Buttmann
- Department of Neurology, University of Würzburg, Würzburg, Germany.
| | - U K Zettl
- Department of Neurology, University of Rostock, Rostock, Germany.
| | - B Fontaine
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France. .,AP-HP, Hôpital de la Pitié Salpêtrière, Département des maladies du système nerveux, F-75013, Paris, France.
| | - L Bertram
- Platform for Genome Analytics, Institutes of Neurogenetics & Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany. .,Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany. .,School of Public Health, Faculty of Medicine, Imperial College of Science, Technology and Medicine, London, UK.
| | - R Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstr. 56, Bochum, 44791, Germany.
| | - A Chan
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstr. 56, Bochum, 44791, Germany.
| |
Collapse
|
25
|
Parker Harp CR, Archambault AS, Sim J, Ferris ST, Mikesell RJ, Koni PA, Shimoda M, Linington C, Russell JH, Wu GF. B cell antigen presentation is sufficient to drive neuroinflammation in an animal model of multiple sclerosis. THE JOURNAL OF IMMUNOLOGY 2015; 194:5077-84. [PMID: 25895531 DOI: 10.4049/jimmunol.1402236] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 03/20/2015] [Indexed: 11/19/2022]
Abstract
B cells are increasingly regarded as integral to the pathogenesis of multiple sclerosis, in part as a result of the success of B cell-depletion therapy. Multiple B cell-dependent mechanisms contributing to inflammatory demyelination of the CNS have been explored using experimental autoimmune encephalomyelitis (EAE), a CD4 T cell-dependent animal model for multiple sclerosis. Although B cell Ag presentation was suggested to regulate CNS inflammation during EAE, direct evidence that B cells can independently support Ag-specific autoimmune responses by CD4 T cells in EAE is lacking. Using a newly developed murine model of in vivo conditional expression of MHC class II, we reported previously that encephalitogenic CD4 T cells are incapable of inducing EAE when B cells are the sole APC. In this study, we find that B cells cooperate with dendritic cells to enhance EAE severity resulting from myelin oligodendrocyte glycoprotein (MOG) immunization. Further, increasing the precursor frequency of MOG-specific B cells, but not the addition of soluble MOG-specific Ab, is sufficient to drive EAE in mice expressing MHCII by B cells alone. These data support a model in which expansion of Ag-specific B cells during CNS autoimmunity amplifies cognate interactions between B and CD4 T cells and have the capacity to independently drive neuroinflammation at later stages of disease.
Collapse
Affiliation(s)
- Chelsea R Parker Harp
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Angela S Archambault
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Julia Sim
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Stephen T Ferris
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Robert J Mikesell
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Pandelakis A Koni
- Cancer Immunology, Inflammation, and Tolerance Program, Cancer Center and Department of Medicine, Georgia Regents University, Augusta, GA 30912
| | - Michiko Shimoda
- Department of Dermatology, University of California at Davis School of Medicine, Sacramento, CA 95817; and
| | | | - John H Russell
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gregory F Wu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
| |
Collapse
|
26
|
Schmitz K, Barthelmes J, Stolz L, Beyer S, Diehl O, Tegeder I. "Disease modifying nutricals" for multiple sclerosis. Pharmacol Ther 2014; 148:85-113. [PMID: 25435020 DOI: 10.1016/j.pharmthera.2014.11.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/20/2014] [Indexed: 12/26/2022]
Abstract
The association between vitamin D and multiple sclerosis has (re)-opened new interest in nutrition and natural compounds in the prevention and treatment of this neuroinflammatory disease. The dietary amount and type of fat, probiotics and biologicals, salmon proteoglycans, phytoestrogens and protease inhibitor of soy, sodium chloride and trace elements, and fat soluble vitamins including D, A and E were all considered as disease-modifying nutraceuticals. Studies in experimental autoimmune encephalomyelitis mice suggest that poly-unsaturated fatty acids and their 'inflammation-resolving' metabolites and the gut microflora may reduce auto-aggressive immune cells and reduce progression or risk of relapse, and infection with whipworm eggs may positively change the gut-brain communication. Encouraged by the recent interest in multiple sclerosis-nutrition nature's pharmacy has been searched for novel compounds with anti-inflammatory, immune-modifying and antioxidative properties, the most interesting being the scorpion toxins that inhibit specific potassium channels of T cells and antioxidative compounds including the green tea flavonoid epigallocatechin-3-gallate, curcumin and the mustard oil glycoside from e.g. broccoli and sulforaphane. They mostly also inhibit pro-inflammatory signaling through NF-κB or toll-like receptors and stabilize the blood brain barrier. Disease modifying functions may also complement analgesic and anti-spastic effects of cannabis, its constituents, and of 'endocannabinoid enhancing' drugs or nutricals like inhibitors of fatty acid amide hydrolase. Nutricals will not solve multiple sclerosis therapeutic challenges but possibly support pharmacological interventions or unearth novel structures.
Collapse
Affiliation(s)
- Katja Schmitz
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Julia Barthelmes
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Leonie Stolz
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Susanne Beyer
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Olaf Diehl
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany
| | - Irmgard Tegeder
- The MS Study Group of the TRIP-Graduate School, Goethe-University Frankfurt, Germany.
| |
Collapse
|