1
|
Schild H, Bopp T. [Immunological foundations of neurological diseases]. DER NERVENARZT 2024; 95:894-908. [PMID: 38953921 DOI: 10.1007/s00115-024-01696-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/07/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Neurodegenerative diseases represent an increasing challenge in ageing societies, as only limited treatment options are currently available. OBJECTIVE New research methods and interdisciplinary interaction of different disciplines have changed the way neurological disorders are viewed and paved the way for the comparatively new field of neuroimmunology, which was established in the early 1980s. Starting from neurological autoimmune diseases, such as multiple sclerosis, knowledge about the involvement of immunological processes in other contexts, such as stroke or traumatic brain injury, has been significantly expanded in recent years. MATERIAL AND METHODS This review article provides an overview of the role of the immune system and the resulting potential for novel treatment approaches. RESULTS The immune system plays a central role in fighting infections but is also able to react to the body's own signals under sterile conditions and cause inflammation and subsequent adaptive immune responses through the release of immune mediators and the recruitment and differentiation of certain immune cell types. This can be beneficial in initiating healing processes; however, chronic inflammatory conditions usually have destructive consequences for the tissue and the organism and must be interrupted. CONCLUSION It is now known that different cells of the immune system play an important role in neurological diseases. Regulatory mechanisms, which are mediated by regulatory T cells or Th2 cells, are usually associated with a good prognosis, whereas inflammatory processes and polarization towards Th1 or Th17 have a destructive character. Novel immunomodulators, which are also increasingly being used in cancer treatment, can now be used in a tissue-specific manner and therefore offer great potential for use in neurological diseases.
Collapse
Affiliation(s)
- Hansjörg Schild
- Institut für Immunologie, Universitätsmedizin Mainz, Langenbeckstraße 1, 55131, Mainz, Deutschland
| | - Tobias Bopp
- Institut für Immunologie, Universitätsmedizin Mainz, Langenbeckstraße 1, 55131, Mainz, Deutschland.
| |
Collapse
|
2
|
Chang Z, Hu Y, Liang X, Miao L, Chi X, Wang X, Chen H, Liu H, Liu L, Zhang Y, Han Z. Exploring the causal role of immune cells in vascular dementia: A bidirectional mendelian randomization study. J Neuroimmunol 2024; 394:578409. [PMID: 39088909 DOI: 10.1016/j.jneuroim.2024.578409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/01/2024] [Accepted: 07/21/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND The aim of this study was to explore the causal association between immune cells and VaD based on a two-sample bidirectional Mendelian randomization study. METHODS Bidirectional two-sample MR analyses based on pooled datasets from publicly available genome-wide association studies were performed using inverse variance weighted (IVW), weighted median (WE), and MR-Egger regressions to evaluate the causal relationships between immune cells and vascular dementia. Heterogeneity was assessed using Cochran's Q statistic. The reliability of the MR analysis results was verified by using the MR-PRESSO method for outlier detection, the MR-Egger method for horizontal multivariate analysis, and the leave-one-out method for sensitivity analysis. RESULTS Specifically, 27 immunophenotypes were associated with VaD pathogenesis, including Sw mem %lymphocyte (P = 0.043), CD38 on CD20- (P = 0.039), CD11c+ monocyte AC (P = 0.024), DC AC (P = 0.002), CCR2 on CD62L+ myeloid DC (P = 0.039), Resting Treg %CD4 (P = 0.042), Activated & resting Treg %CD4+ (P = 0.038), CD28+ CD45RA- CD8br %CD8br (P = 0.047), NK %CD3- lymphocyte (P = 0.042), CD45 on B cell (P = 0.029), FSC-A on NKT (P = 0.033), CD45 on CD33br HLA DR+ CD14- (P = 0.039) were significantly correlated with increased VaD risk. Additionally, four immune phenotypes, namely, CD19 on CD20-, Resting Treg %CD4, Activated & resting Treg %CD4+, and CD11c+ monocyte AC, showed bidirectional effects on VaD. CONCLUSIONS MR analysis revealed potential causal relationships between certain immune cells and VaD. Our preliminary exploration through immune cell infiltration analysis highlights the significant value of immune cells in VaD. Therefore, this study may provide a new perspective for the prevention and treatment of VaD.
Collapse
Affiliation(s)
- Ze Chang
- Xiyuan Hospital of China Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Yuli Hu
- Zhejiang Provincial People's Hospital, 310006 Hangzhou, China
| | - Xiao Liang
- Xiyuan Hospital of China Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Lina Miao
- Xiyuan Hospital of China Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Xiansu Chi
- Xiyuan Hospital of China Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Xie Wang
- The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China
| | - Hong Chen
- The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China
| | - Hongxi Liu
- Xiyuan Hospital of China Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Longtao Liu
- Xiyuan Hospital of China Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Yunling Zhang
- Xiyuan Hospital of China Academy of Traditional Chinese Medicine, Beijing 100091, China.
| | - Zhenyun Han
- Shenzhen Hospital of Beijing University of Chinese Medicine, Shenzhen 518000, Guangdong, China.
| |
Collapse
|
3
|
Kostic M, Zivkovic N, Cvetanovic A, Basic J, Stojanovic I. Dissecting the immune response of CD4 + T cells in Alzheimer's disease. Rev Neurosci 2024:revneuro-2024-0090. [PMID: 39238424 DOI: 10.1515/revneuro-2024-0090] [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: 07/04/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
The formation of amyloid-β (Aβ) plaques is a neuropathological hallmark of Alzheimer's disease (AD), however, these pathological aggregates can also be found in the brains of cognitively unimpaired elderly population. In that context, individual variations in the Aβ-specific immune response could be key factors that determine the level of Aβ-induced neuroinflammation and thus the propensity to develop AD. CD4+ T cells are the cornerstone of the immune response that coordinate the effector functions of both adaptive and innate immunity. However, despite intensive research efforts, the precise role of these cells during AD pathogenesis is still not fully elucidated. Both pathogenic and beneficial effects have been observed in various animal models of AD, as well as in humans with AD. Although this functional duality of CD4+ T cells in AD can be simply attributed to the vast phenotype heterogeneity of this cell lineage, disease stage-specific effect have also been proposed. Therefore, in this review, we summarized the current understanding of the role of CD4+ T cells in the pathophysiology of AD, from the aspect of their antigen specificity, activation, and phenotype characteristics. Such knowledge is of practical importance as it paves the way for immunomodulation as a therapeutic option for AD treatment, given that currently available therapies have not yielded satisfactory results.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ana Cvetanovic
- Department of Oncology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Jelena Basic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| |
Collapse
|
4
|
Sumida TS, Lincoln MR, He L, Park Y, Ota M, Oguchi A, Son R, Yi A, Stillwell HA, Leissa GA, Fujio K, Murakawa Y, Kulminski AM, Epstein CB, Bernstein BE, Kellis M, Hafler DA. An autoimmune transcriptional circuit drives FOXP3 + regulatory T cell dysfunction. Sci Transl Med 2024; 16:eadp1720. [PMID: 39196959 DOI: 10.1126/scitranslmed.adp1720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/02/2024] [Indexed: 08/30/2024]
Abstract
Autoimmune diseases, among the most common disorders of young adults, are mediated by genetic and environmental factors. Although CD4+FOXP3+ regulatory T cells (Tregs) play a central role in preventing autoimmunity, the molecular mechanism underlying their dysfunction is unknown. Here, we performed comprehensive transcriptomic and epigenomic profiling of Tregs in the autoimmune disease multiple sclerosis (MS) to identify critical transcriptional programs regulating human autoimmunity. We found that up-regulation of a primate-specific short isoform of PR domain zinc finger protein 1 (PRDM1-S) induces expression of serum and glucocorticoid-regulated kinase 1 (SGK1) independent from the evolutionarily conserved long PRDM1, which led to destabilization of forkhead box P3 (FOXP3) and Treg dysfunction. This aberrant PRDM1-S/SGK1 axis is shared among other autoimmune diseases. Furthermore, the chromatin landscape profiling in Tregs from individuals with MS revealed enriched activating protein-1 (AP-1)/interferon regulatory factor (IRF) transcription factor binding as candidate upstream regulators of PRDM1-S expression and Treg dysfunction. Our study uncovers a mechanistic model where the evolutionary emergence of PRDM1-S and epigenetic priming of AP-1/IRF may be key drivers of dysfunctional Tregs in autoimmune diseases.
Collapse
Affiliation(s)
- Tomokazu S Sumida
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Matthew R Lincoln
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON M6R 1B5, Canada
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, ON M6R 1B5, Canada
| | - Liang He
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02139, USA
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27705, USA
| | - Yongjin Park
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02139, USA
| | - Mineto Ota
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Akiko Oguchi
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto 606-8303, Japan
| | - Raku Son
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto 606-8303, Japan
| | - Alice Yi
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Helen A Stillwell
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Greta A Leissa
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Yasuhiro Murakawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto 606-8303, Japan
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC 27705, USA
| | | | - Bradley E Bernstein
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02139, USA
| | - David A Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
5
|
Hu D, Chen M, Li X, Morin P, Daley S, Han Y, Hemberg M, Weiner HL, Xia W. ApoE ε4-dependent alteration of CXCR3 + CD127 + CD4 + T cells is associated with elevated plasma neurofilament light chain in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596276. [PMID: 38853824 PMCID: PMC11160665 DOI: 10.1101/2024.05.28.596276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Recent findings indicate a correlation between the peripheral adaptive immune system and neuroinflammation in Alzheimer's disease (AD). To characterize the composition of adaptive immune cells in the peripheral blood of AD patients, we utilized single-cell mass cytometry (CyTOF) to profile peripheral blood mononuclear cells (PBMCs). Concurrently, we assessed the concentration of proteins associated with AD and neuroinflammation in the plasma of the same subjects. We found that the abundance of proinflammatory CXCR3 + CD127 + Type 1 T helper (Th1) cells in AD patients was negatively correlated with the abundance of neurofilament light chain (NfL) protein. This correlation is apolipoprotein E (ApoE) ε4-dependent. Analyzing public single-cell RNA-sequencing (scRNA-seq) data, we found that, contrary to the scenario in the peripheral blood, the cell frequency of CXCR3 + CD127 + Th1 cells in the cerebrospinal fluid (CSF) of AD patients was increased compared to healthy controls (HCs). Moreover, the proinflammatory capacity of CXCR3 + CD127 + Th1 cells in the CSF of AD patients was further increased compared to HCs. These results reveal an association of a peripheral T-cell change with neuroinflammation in AD and suggest that dysregulation of peripheral adaptive immune responses, particularly involving CXCR3 + CD127 + Th1 cells, may potentially be mediated by factors such as ApoE ε4 genotype. One sentence summary An apolipoprotein E (ApoE) ε4-dependent alteration of CD4 T cell subpopulation in peripheral blood is associated with neuroinflammation in patients with Alzheimer's disease.
Collapse
|
6
|
Zhang Q, Yang G, Luo Y, Jiang L, Chi H, Tian G. Neuroinflammation in Alzheimer's disease: insights from peripheral immune cells. Immun Ageing 2024; 21:38. [PMID: 38877498 PMCID: PMC11177389 DOI: 10.1186/s12979-024-00445-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024]
Abstract
Alzheimer's disease (AD) is a serious brain disorder characterized by the presence of beta-amyloid plaques, tau pathology, inflammation, neurodegeneration, and cerebrovascular dysfunction. The presence of chronic neuroinflammation, breaches in the blood-brain barrier (BBB), and increased levels of inflammatory mediators are central to the pathogenesis of AD. These factors promote the penetration of immune cells into the brain, potentially exacerbating clinical symptoms and neuronal death in AD patients. While microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in AD, recent evidence suggests the infiltration of cerebral vessels and parenchyma by peripheral immune cells, including neutrophils, T lymphocytes, B lymphocytes, NK cells, and monocytes in AD. These cells participate in the regulation of immunity and inflammation, which is expected to play a huge role in future immunotherapy. Given the crucial role of peripheral immune cells in AD, this article seeks to offer a comprehensive overview of their contributions to neuroinflammation in the disease. Understanding the role of these cells in the neuroinflammatory response is vital for developing new diagnostic markers and therapeutic targets to enhance the diagnosis and treatment of AD patients.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Laboratory Medicine, Southwest Medical University, Luzhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, USA
| | - Yuan Luo
- Department of Laboratory Medicine, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China.
| | - Gang Tian
- Department of Laboratory Medicine, Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University, Sichuan, 646000, China.
| |
Collapse
|
7
|
Gao Y, Lu Y, Liang X, Zhao M, Yu X, Fu H, Yang W. CD4 + T-Cell Senescence in Neurodegenerative Disease: Pathogenesis and Potential Therapeutic Targets. Cells 2024; 13:749. [PMID: 38727285 PMCID: PMC11083511 DOI: 10.3390/cells13090749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/07/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
With the increasing proportion of the aging population, neurodegenerative diseases have become one of the major health issues in society. Neurodegenerative diseases (NDs), including multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), are characterized by progressive neurodegeneration associated with aging, leading to a gradual decline in cognitive, emotional, and motor functions in patients. The process of aging is a normal physiological process in human life and is accompanied by the aging of the immune system, which is known as immunosenescence. T-cells are an important part of the immune system, and their senescence is the main feature of immunosenescence. The appearance of senescent T-cells has been shown to potentially lead to chronic inflammation and tissue damage, with some studies indicating a direct link between T-cell senescence, inflammation, and neuronal damage. The role of these subsets with different functions in NDs is still under debate. A growing body of evidence suggests that in people with a ND, there is a prevalence of CD4+ T-cell subsets exhibiting characteristics that are linked to senescence. This underscores the significance of CD4+ T-cells in NDs. In this review, we summarize the classification and function of CD4+ T-cell subpopulations, the characteristics of CD4+ T-cell senescence, the potential roles of these cells in animal models and human studies of NDs, and therapeutic strategies targeting CD4+ T-cell senescence.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (Y.G.); (Y.L.); (X.L.); (M.Z.); (X.Y.); (H.F.)
| |
Collapse
|
8
|
Zeng J, Liao Z, Yang H, Wang Q, Wu Z, Hua F, Zhou Z. T cell infiltration mediates neurodegeneration and cognitive decline in Alzheimer's disease. Neurobiol Dis 2024; 193:106461. [PMID: 38437992 DOI: 10.1016/j.nbd.2024.106461] [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: 11/05/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder with pathological features of β-amyloid (Aβ) and hyperphosphorylated tau protein accumulation in the brain, often accompanied by cognitive decline. So far, our understanding of the extent and role of adaptive immune responses in AD has been quite limited. T cells, as essential members of the adaptive immune system, exhibit quantitative and functional abnormalities in the brains of AD patients. Dysfunction of the blood-brain barrier (BBB) in AD is considered one of the factors leading to T cell infiltration. Moreover, the degree of neuronal loss in AD is correlated with the quantity of T cells. We first describe the differentiation and subset functions of peripheral T cells in AD patients and provide an overview of the key findings related to BBB dysfunction and how T cells infiltrate the brain parenchyma through the BBB. Furthermore, we emphasize the risk factors associated with AD, including Aβ, Tau protein, microglial cells, apolipoprotein E (ApoE), and neuroinflammation. We discuss their regulation of T cell activation and proliferation, as well as the connection between T cells, neurodegeneration, and cognitive decline. Understanding the innate immune response is crucial for providing comprehensive personalized therapeutic strategies for AD.
Collapse
Affiliation(s)
- Junjian Zeng
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Zhiqiang Liao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Hanqin Yang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Qiong Wang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Zhiyong Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China.
| | - Zhidong Zhou
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China.
| |
Collapse
|
9
|
Xue H, Chen J, Zeng L, Fan W. Causal relationship between circulating immune cells and the risk of Alzheimer's disease: A Mendelian randomization study. Exp Gerontol 2024; 187:112371. [PMID: 38301877 DOI: 10.1016/j.exger.2024.112371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Increasing evidence has shown a link between immune cells and Alzheimer's disease (AD). Comprehensive two-sample Mendelian randomization (MR) analysis was performed to determine the causal association between 731 immune cell signatures and AD in this study. METHODS We extracted genetic variants of 731 immune cell traits and AD from the publicly available GWAS dataset. The immune features included median fluorescence intensity (MFI), relative cellular (RC), absolute cellular (AC) and morphological parameters (MP). The inverse variance weighted (IVW) method was the main MR analysis method, and sensitivity analyses were used to validate the robustness, heterogeneity and horizontal pleiotropy of the results. RESULTS After FDR adjustment, seven immune phenotypes were found to be associated significantly with AD risk: HLA DR on CD33-HLA DR+ (OR = 0.938, PFDR = 0.001), Secreting Treg %CD4 (OR = 0.972, PFDR = 0.021), HLA DR+T cell AC (OR = 0.928, PFDR = 0.041), Activated & resting Treg % CD4 Treg (OR = 1.031, PFDR = 0.002), CD33 on CD33dim HLA DR+CD11b+ (OR = 1.025, PFDR = 0.025), CD33 on CD14+monocyte (OR = 1.026, PFDR = 0.027) and CD33 on CD66b++myeloid cell (OR = 1.027, PFDR = 0.036). CONCLUSIONS These findings demonstrated seven immune phenotypes were significantly associated with AD risk. This may provide researchers with a new perspective in exploring the biological mechanisms of AD and may lead to the exploration of earlier treatment.
Collapse
Affiliation(s)
- Hua Xue
- Department of Neurology, Sichuan Taikang Hospital, Chengdu, Sichuan, China.
| | - Jiajia Chen
- College of Chemical Engineering, Sichuan University of Science & Engineering, Zigong, China
| | - Li Zeng
- Department of Respiratory, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Wenhui Fan
- Department of Neurology, Sichuan Taikang Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
10
|
Graber DJ, Cook WJ, Sentman ML, Murad-Mabaera JM, Sentman CL. Human CD4+CD25+ T cells expressing a chimeric antigen receptor against aberrant superoxide dismutase 1 trigger antigen-specific immunomodulation. Cytotherapy 2024; 26:126-135. [PMID: 38043051 PMCID: PMC10872388 DOI: 10.1016/j.jcyt.2023.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/12/2023] [Accepted: 11/14/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND AIMS Amyotrophic lateral sclerosis (ALS) is a fatal disease associated with motor neuron degeneration, accumulation of aggregated misfolded proteins and neuroinflammation in motor regions of the central nervous system (CNS). Clinical trials using regulatory T cells (Tregs) are ongoing because of Tregs' immunomodulatory function, ability to traffic to the CNS, high numbers correlating with slower disease in ALS and disease-modifying activity in ALS mouse models. In the current study, a chimeric antigen receptor (CAR) was developed and characterized in human Tregs to enhance their immunomodulatory activity when in contact with an ALS protein aggregate. METHODS A CAR (DG05-28-3z) consisting of a human superoxide dismutase 1 (hSOD1)-binding single-chain variable fragment, CD28 hinge, transmembrane and co-stimulatory domain and CD3ζ signaling domain was created and expressed in human Tregs. Human Tregs were isolated by either magnetic enrichment for CD4+CD25hi cells (Enr-Tregs) or cell sorting for CD4+CD25hiCD127lo cells (FP-Tregs), transduced and expanded for 17 days. RESULTS The CAR bound preferentially to the ALS mutant G93A-hSOD1 protein relative to the wild-type hSOD1 protein. The CAR Tregs produced IL-10 when cultured with aggregated G93A-hSOD1 proteins or spinal cord explants from G93A-hSOD1 transgenic mice. Co-culturing DG05-28-3z CAR Tregs with human monocytes/macrophages inhibited production of tumor necrosis factor alpha and reactive oxygen species. Expanded FP-Tregs resulted in more robust Tregs compared with Enr-Tregs. FP-Tregs produced similar IL-10 and less interferon gamma, had lower Treg-specific demethylated region methylation and expressed higher FoxP3 and CD39. CONCLUSIONS Taken together, this study demonstrates that gene-modified Tregs can be developed to target an aggregated ALS-relevant protein to elicit CAR-mediated Treg effector functions and provides an approach for generating Treg therapies for ALS with the goal of enhanced disease site-specific immunomodulation.
Collapse
Affiliation(s)
- David J Graber
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA; Center for Synthetic Immunity, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - W James Cook
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA; Center for Synthetic Immunity, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Marie-Louise Sentman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA; Center for Synthetic Immunity, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | | | - Charles L Sentman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA; Center for Synthetic Immunity, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA.
| |
Collapse
|
11
|
Gakis G, Angelopoulos I, Panagoulias I, Mouzaki A. Current knowledge on multiple sclerosis pathophysiology, disability progression assessment and treatment options, and the role of autologous hematopoietic stem cell transplantation. Autoimmun Rev 2024; 23:103480. [PMID: 38008300 DOI: 10.1016/j.autrev.2023.103480] [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: 10/31/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) that affects nearly 2.8 million people each year. MS distinguishes three main types: relapsing-remitting MS (RRMS), secondary progressive MS (SPMS) and primary progressive MS (PPMS). RRMS is the most common type, with the majority of patients eventually progressing to SPMS, in which neurological development is constant, whereas PPMS is characterized by a progressive course from disease onset. New or additional insights into the role of effector and regulatory cells of the immune and CNS systems, Epstein-Barr virus (EBV) infection, and the microbiome in the pathophysiology of MS have emerged, which may lead to the development of more targeted therapies that can halt or reverse neurodegeneration. Depending on the type and severity of the disease, various disease-modifying therapies (DMTs) are currently used for RRMS/SPMS and PPMS. As a last resort, and especially in highly active RRMS that does not respond to DMTs, autologous hematopoietic stem cell transplantation (AHSCT) is performed and has shown good results in reducing neuroinflammation. Nevertheless, the question of its potential role in preventing disability progression remains open. The aim of this review is to provide a comprehensive update on MS pathophysiology, assessment of MS disability progression and current treatments, and to examine the potential role of AHSCT in preventing disability progression.
Collapse
Affiliation(s)
- Georgios Gakis
- Laboratory of Immunohematology, Medical School, University of Patras, Patras, Greece
| | - Ioannis Angelopoulos
- Laboratory of Immunohematology, Medical School, University of Patras, Patras, Greece
| | - Ioannis Panagoulias
- Laboratory of Immunohematology, Medical School, University of Patras, Patras, Greece
| | - Athanasia Mouzaki
- Laboratory of Immunohematology, Medical School, University of Patras, Patras, Greece.
| |
Collapse
|
12
|
Huang Z, Gong Z, Lin Y, Yang F, Chen W, Xiang S, Huang Y, Xiao H, Xu S, Duan J. Treatment with glatiramer acetate in APPswe/PS1dE9 mice at an early stage of Alzheimer's disease prior to amyloid-beta deposition delays the disease's pathological development and ameliorates cognitive decline. Front Aging Neurosci 2024; 16:1267780. [PMID: 38352237 PMCID: PMC10861656 DOI: 10.3389/fnagi.2024.1267780] [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: 07/27/2023] [Accepted: 01/11/2024] [Indexed: 02/16/2024] Open
Abstract
Background Alzheimer's disease (AD) is characterized by neuroinflammation, which is frequently accompanied by immune system dysfunction. Although the mechanism of neurodegenerative lesions is unclear, various clinical trials have highlighted that early intervention in AD is crucial to the success of treatment. In order to explore the potential of immunotherapy in the early period of AD, the present study evaluated whether application of glatiramer acetate (GA), an immunomodulatory agent approved for remitting-relapsing multiple sclerosis (RRMS), in the early stages of AD prior to amyloid beta (Aβ) deposition altered the Aβ pathology and cognitive impairments in APPswe/PSEN1dE9 (APP/PS1) transgenic mice. Methods We treated two cohorts of pre-depositing and amyloid-depositing (2- and 6-month-old) APP/PS1 mice with weekly-GA subcutaneous injection over a 12-week period. We then tested spatial learning and memory using the Morris water maze (MWM) and the Y maze. Immunohistochemistry staining was utilized to analyze Aβ burden in the brain as well as activated microglia. Furthermore, the inflammatory cytokine milieu within brains was estimated by quantitative real-time polymerase chain reaction, and the peripheral CD4+CD25+Foxp3+ regulatory T cells (Tregs) in the spleen were measured by flow cytometry. Results We found that early GA administration reduced Aβ burden and ameliorated cognitive decline. Meanwhile, the immune microenvironment had changed in the brain, with an increase in the production of anti-inflammatory cytokines and a decrease in microglial activation. Interestingly, early GA administration also modulated the peripheral immune system through the amplification of Tregs in the spleen. Conclusion Overall, our findings revealed that GA treatment might enhance the central and peripheral immune systems' protective capabilities in the early stages of AD, eventually improving cognitive deficits. Our research supports the advantages of immunomodulatory treatments for AD at an early stage.
Collapse
Affiliation(s)
- Zengyong Huang
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
- Shantou Central Hospital, Shantou, China
| | - Zhuo Gong
- Shantou Central Hospital, Shantou, China
| | - Yongtai Lin
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Fan Yang
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Weiping Chen
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Shaotong Xiang
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Yuedong Huang
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Hao Xiao
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Shuwen Xu
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Jinhai Duan
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Garmendia JV, De Sanctis CV, Das V, Annadurai N, Hajduch M, De Sanctis JB. Inflammation, Autoimmunity and Neurodegenerative Diseases, Therapeutics and Beyond. Curr Neuropharmacol 2024; 22:1080-1109. [PMID: 37898823 PMCID: PMC10964103 DOI: 10.2174/1570159x22666231017141636] [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: 06/05/2023] [Revised: 07/13/2023] [Accepted: 08/03/2023] [Indexed: 10/30/2023] Open
Abstract
Neurodegenerative disease (ND) incidence has recently increased due to improved life expectancy. Alzheimer's (AD) or Parkinson's disease (PD) are the most prevalent NDs. Both diseases are poly genetic, multifactorial and heterogenous. Preventive medicine, a healthy diet, exercise, and controlling comorbidities may delay the onset. After the diseases are diagnosed, therapy is needed to slow progression. Recent studies show that local, peripheral and age-related inflammation accelerates NDs' onset and progression. Patients with autoimmune disorders like inflammatory bowel disease (IBD) could be at higher risk of developing AD or PD. However, no increase in ND incidence has been reported if the patients are adequately diagnosed and treated. Autoantibodies against abnormal tau, β amyloid and α- synuclein have been encountered in AD and PD and may be protective. This discovery led to the proposal of immune-based therapies for AD and PD involving monoclonal antibodies, immunization/ vaccines, pro-inflammatory cytokine inhibition and anti-inflammatory cytokine addition. All the different approaches have been analysed here. Future perspectives on new therapeutic strategies for both disorders are concisely examined.
Collapse
Affiliation(s)
- Jenny Valentina Garmendia
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
| | - Claudia Valentina De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
| | - Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
- The Czech Advanced Technology and Research Institute (Catrin), Palacky University, Olomouc, The Czech Republic
| | - Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
| | - Marián Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
- The Czech Advanced Technology and Research Institute (Catrin), Palacky University, Olomouc, The Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, The Czech Republic
- The Czech Advanced Technology and Research Institute (Catrin), Palacky University, Olomouc, The Czech Republic
| |
Collapse
|
14
|
Arneth B. Regulatory T Cells in Multiple Sclerosis Diagnostics-What Do We Know So Far? J Pers Med 2023; 14:29. [PMID: 38248730 PMCID: PMC10821144 DOI: 10.3390/jpm14010029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/10/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune disorder that affects the central nervous system (CNS) through inflammation. MS symptoms become acute if the disease progresses to the relapsing phase. AIM This review aimed to evaluate the role played by regulatory T cells (Tregs) in the pathogenesis of MS. METHODS This review used scholarly journal articles obtained from PubMed, PsycINFO, and CINAHL with different search parameters such as 'regulatory T cells', 'multiple sclerosis', and 'current knowledge'. The process of searching for articles was limited to those that had publication dates falling between 2010 and 2020. RESULTS Tregs play a role in the pathogenesis of MS. This conclusion is supported by animal disease models and environmental factors that can underlie Treg alterations in MS. Despite the knowledge of the role played by Tregs in MS pathogenesis, the specific subsets of Tregs involved in MS development remain incompletely understood. DISCUSSION This review provides an essential link between Tregs and MS activity. Targeting Tregs could be an efficient way to establish new treatment methods for MS management. CONCLUSION MS is a complex condition affecting many people worldwide. Research has shown that Tregs can influence MS development and progression. More investigations are needed to understand how Tregs affect the pathogenesis of MS.
Collapse
Affiliation(s)
- Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Philipps University Marburg, 35043 Marburg, Germany;
- Institute of Laboratory Medicine and Pathobiochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
- Hospital of the Universities of Giessen and Marburg, 35392 Giessen, Germany
| |
Collapse
|
15
|
Yeapuri P, Machhi J, Lu Y, Abdelmoaty MM, Kadry R, Patel M, Bhattarai S, Lu E, Namminga KL, Olson KE, Foster EG, Mosley RL, Gendelman HE. Amyloid-β specific regulatory T cells attenuate Alzheimer's disease pathobiology in APP/PS1 mice. Mol Neurodegener 2023; 18:97. [PMID: 38111016 PMCID: PMC10729469 DOI: 10.1186/s13024-023-00692-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) maintain immune tolerance. While Treg-mediated neuroprotective activities are now well-accepted, the lack of defined antigen specificity limits their therapeutic potential. This is notable for neurodegenerative diseases where cell access to injured brain regions is required for disease-specific therapeutic targeting and improved outcomes. To address this need, amyloid-beta (Aβ) antigen specificity was conferred to Treg responses by engineering the T cell receptor (TCR) specific for Aβ (TCRAβ). The TCRAb were developed from disease-specific T cell effector (Teff) clones. The ability of Tregs expressing a transgenic TCRAβ (TCRAβ -Tregs) to reduce Aβ burden, transform effector to regulatory cells, and reverse disease-associated neurotoxicity proved beneficial in an animal model of Alzheimer's disease. METHODS TCRAβ -Tregs were generated by CRISPR-Cas9 knockout of endogenous TCR and consequent incorporation of the transgenic TCRAb identified from Aβ reactive Teff monoclones. Antigen specificity was confirmed by MHC-Aβ-tetramer staining. Adoptive transfer of TCRAβ-Tregs to mice expressing a chimeric mouse-human amyloid precursor protein and a mutant human presenilin-1 followed measured behavior, immune, and immunohistochemical outcomes. RESULTS TCRAβ-Tregs expressed an Aβ-specific TCR. Adoptive transfer of TCRAβ-Tregs led to sustained immune suppression, reduced microglial reaction, and amyloid loads. 18F-fluorodeoxyglucose radiolabeled TCRAβ-Treg homed to the brain facilitating antigen specificity. Reduction in amyloid load was associated with improved cognitive functions. CONCLUSIONS TCRAβ-Tregs reduced amyloid burden, restored brain homeostasis, and improved learning and memory, supporting the increased therapeutic benefit of antigen specific Treg immunotherapy for AD.
Collapse
Affiliation(s)
- Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mai Mohamed Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Rana Kadry
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Milankumar Patel
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Eugene Lu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Krista L Namminga
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Emma G Foster
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
16
|
Asamu MO, Oladipo OO, Abayomi OA, Adebayo AA. Alzheimer's disease: The role of T lymphocytes in neuroinflammation and neurodegeneration. Brain Res 2023; 1821:148589. [PMID: 37734576 DOI: 10.1016/j.brainres.2023.148589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/03/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Alzheimer's disease, the leading cause of progressive cognitive decline globally, has been reported to be enhanced by neuroinflammation. Brain-resident innate immune cells and adaptive immune cells work together to produce neuroinflammation. Studies over the past decade have established the neuroimmune axis present in Alzheimer's disease; the crosstalk between adaptive and innate immune cells within and outside the brain is crucial to the onset and progression of Alzheimer's disease. Although the role of the adaptive immune system in Alzheimer's disease is not fully understood, it has been hypothesized that the brain's immune homeostasis is significantly disrupted, which greatly contributes to neuroinflammation. Brain-infiltrating T cells possess proinflammatory phenotypes and activities that directly contribute to neuroinflammation. The pro-inflammatory activities of the adaptive immune system in Alzheimer's disease are characterized by the upregulation of effector T cell activities and the downregulation of regulatory T cell activities in the brain, blood, and cerebrospinal fluid. In this review, we discuss the major impact of T lymphocytes on the pathogenesis and progression of Alzheimer's disease. Understanding the role and mechanism of action of T cells in Alzheimer's disease would significantly contribute to the identification of novel biomarkers for diagnosing and monitoring the progression of the disease. This knowledge could also be crucial to the development of immunotherapies for Alzheimer's disease.
Collapse
Affiliation(s)
- Moses O Asamu
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oladapo O Oladipo
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | - Oluseun A Abayomi
- College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; Olabisi Onabanjo University Teaching Hospital (OOUTH), Sagamu, Ogun State, Nigeria
| | - Afeez A Adebayo
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
17
|
Srivastava P, Nishiyama S, Zhou F, Lin SH, Srivastava A, Su C, Xu Y, Peng W, Levy M, Schwarzschild M, Chen X. Peripheral MC1R Activation Modulates Immune Responses and is Neuroprotective in a Mouse Model of Parkinson's Disease. J Neuroimmune Pharmacol 2023; 18:704-717. [PMID: 38110615 PMCID: PMC10769915 DOI: 10.1007/s11481-023-10094-7] [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: 06/18/2023] [Accepted: 10/17/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Melanocortin 1 receptor (MC1R) is a key pigmentation gene, and loss-of-function of MC1R variants that produce red hair may be associated with Parkinson's disease (PD). We previously reported compromised dopaminergic neuron survival in Mc1r mutant mice and dopaminergic neuroprotective effects of local injection of a MC1R agonist to the brain or a systemically administered MC1R agonist with appreciable central nervous system (CNS) permeability. Beyond melanocytes and dopaminergic neurons, MC1R is expressed in other peripheral tissues and cell types, including immune cells. The present study investigates the impact of NDP-MSH, a synthetic melanocortin receptor (MCR) agonist that does not cross BBB, on the immune system and the nigrostriatal dopaminergic system in mouse model of PD. METHODS C57BL/6 mice were treated systemically with MPTP.HCl (20 mg/kg) and LPS (1 mg/kg) from day 1 to day 4 and NDP-MSH (400 µg/kg) or vehicle from day 1 to day 12 following which the mice were sacrificed. Peripheral and CNS immune cells were phenotyped and inflammatory markers were measured. The nigrostriatal dopaminergic system was assessed behaviorally, chemically, immunologically, and pathologically. To understand the role of regulatory T cells (Tregs) in this model, CD25 monoclonal antibody was used to deplete CD25 + Tregs. RESULTS Systemic NDP-MSH administration significantly attenuated striatal dopamine depletion and nigral dopaminergic neuron loss induced by MPTP + LPS. It improved the behavioral outcomes in the pole test. Mc1r mutant mice injected with NDP-MSH in the MPTP and LPS paradigm showed no changes in striatal dopamine levels suggesting that the NDP-MSH acts through the MC1R pathway. Although no NDP-MSH was detected in the brain, peripheral, NDP-MSH attenuated neuroinflammation as observed by diminished microglial activation in the nigral region, along with reduced TNF-α and IL1β levels in the ventral midbrain. Depletion of Tregs was associated with diminished neuroprotective effects of NDP-MSH. CONCLUSIONS Our study demonstrates that peripherally acting NDP-MSH confers protection on dopaminergic nigrostriatal neurons and reduces hyperactivated microglia. NDP-MSH modulates peripheral immune responses, and Tregs may be involved in the neuroprotective effect of NDP-MSH.
Collapse
Affiliation(s)
- Pranay Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Shuhei Nishiyama
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Fang Zhou
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Sonia H Lin
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Akriti Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Chienwen Su
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Yuehang Xu
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Weiyi Peng
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Michael Levy
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Michael Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
18
|
Jafarzadeh A, Sheikhi A, Jafarzadeh Z, Nemati M. Differential roles of regulatory T cells in Alzheimer's disease. Cell Immunol 2023; 393-394:104778. [PMID: 37907046 DOI: 10.1016/j.cellimm.2023.104778] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/02/2023]
Abstract
Regulatory T (Treg) cells interact with a variety of resident cells and infiltrated immune cells in the central nervous system (CNS) to modulate neuroinflammation and neurodegeneration. Extracellular amyloid-β (Aβ) peptide deposition and secondary persistent inflammation due to activation of microglia, astrocytes, and infiltrated immune cells contribute to Alzheimer's disease (AD)-related neurodegeneration. The majority of evidence supports the neuroprotective effects of Treg cells in AD. In the early stages of AD, appropriate Treg cell activity is required for the induction of microglia and astrocyte phagocytic activity in order to clear A deposits and prevent neuroinflammation. Such neuroprotective impacts were in part attributed to the ability of Treg cells to suppress deleterious and/or boost beneficial functions of microglia/astrocytes. In the later stages of AD, an effective Treg cell activity needs to prevent neurotoxicity and neurodegeneration. Treg cells can exert preventive effects on Th1-, and Th17 cell-related pathologic responses, whilst potentiating Th2-mediated protective activity. The impaired Treg cell-related immunomodulatory mechanisms have been described in AD patients and in related animal models which can contribute to the onset and progression of AD. This review aimed to provide a comprehensive figure regarding the role of Treg cells in AD while highlighting potential therapeutic approaches.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Abdolkarim Sheikhi
- Department of Immunology, Faculty of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Zahra Jafarzadeh
- Student Research Committee, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
19
|
Lopes JR, Zhang X, Mayrink J, Tatematsu BK, Guo L, LeServe DS, Abou-El-Hassan H, Rong F, Dalton MJ, Oliveira MG, Lanser TB, Liu L, Butovsky O, Rezende RM, Weiner HL. Nasal administration of anti-CD3 monoclonal antibody ameliorates disease in a mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 2023; 120:e2309221120. [PMID: 37669383 PMCID: PMC10500187 DOI: 10.1073/pnas.2309221120] [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: 06/04/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Emerging evidence suggests that dysregulation of neuroinflammation, particularly that orchestrated by microglia, plays a significant role in the pathogenesis of Alzheimer's disease (AD). Danger signals including dead neurons, dystrophic axons, phosphorylated tau, and amyloid plaques alter the functional phenotype of microglia from a homeostatic (M0) to a neurodegenerative or disease-associated phenotype, which in turn drives neuroinflammation and promotes disease. Thus, therapies that target microglia activation constitute a unique approach for treating AD. Here, we report that nasally administered anti-CD3 monoclonal antibody in the 3xTg AD mouse model reduced microglial activation and improved cognition independent of amyloid beta deposition. In addition, gene expression analysis demonstrated decreased oxidative stress, increased axogenesis and synaptic organization, and metabolic changes in the hippocampus and cortex of nasal anti-CD3 treated animals. The beneficial effect of nasal anti-CD3 was associated with the accumulation of T cells in the brain where they were in close contact with microglial cells. Taken together, our findings identify nasal anti-CD3 as a unique form of immunotherapy to treat Alzheimer's disease independent of amyloid beta targeting.
Collapse
Affiliation(s)
- Juliana R. Lopes
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Xiaoming Zhang
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Julia Mayrink
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Bruna K. Tatematsu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Lydia Guo
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Danielle S. LeServe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Hadi Abou-El-Hassan
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Felipe Rong
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Maria J. Dalton
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Marilia G. Oliveira
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Toby B. Lanser
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Lei Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Rafael M. Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Howard L. Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| |
Collapse
|
20
|
Lutshumba J, Wilcock DM, Monson NL, Stowe AM. Sex-based differences in effector cells of the adaptive immune system during Alzheimer's disease and related dementias. Neurobiol Dis 2023; 184:106202. [PMID: 37330146 PMCID: PMC10481581 DOI: 10.1016/j.nbd.2023.106202] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023] Open
Abstract
Neurological conditions such as Alzheimer's disease (AD) and related dementias (ADRD) present with many challenges due to the heterogeneity of the related disease(s), making it difficult to develop effective treatments. Additionally, the progression of ADRD-related pathologies presents differently between men and women. With two-thirds of the population affected with ADRD being women, ADRD has presented itself with a bias toward the female population. However, studies of ADRD generally do not incorporate sex-based differences in investigating the development and progression of the disease, which is detrimental to understanding and treating dementia. Additionally, recent implications for the adaptive immune system in the development of ADRD bring in new factors to be considered as part of the disease, including sex-based differences in immune response(s) during ADRD development. Here, we review the sex-based differences of pathological hallmarks of ADRD presentation and progression, sex-based differences in the adaptive immune system and how it changes with ADRD, and the importance of precision medicine in the development of a more targeted and personalized treatment for this devastating and prevalent neurodegenerative condition.
Collapse
Affiliation(s)
- Jenny Lutshumba
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America; Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Nancy L Monson
- Department of Neurology and Immunology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Ann M Stowe
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, United States of America; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, United States of America.
| |
Collapse
|
21
|
Srivastava P, Nishiyama S, Lin SH, Srivastava A, Su C, Peng W, Levy M, Schwarzschild M, Xu Y, Chen X. Peripheral MC1R activation modulates immune responses and is neuroprotective in a mouse model of Parkinson's disease. RESEARCH SQUARE 2023:rs.3.rs-3042571. [PMID: 37398302 PMCID: PMC10312952 DOI: 10.21203/rs.3.rs-3042571/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background Melanocortin 1 receptor (MC1R) is a key pigmentation gene, and loss-of-function of MC1R variants that produce red hair may be associated with Parkinson's disease (PD). We previously reported compromised dopaminergic neuron survival in Mc1r mutant mice and dopaminergic neuroprotective effects of local injection of a MC1R agonist to the brain or a systemically administered MC1R agonist with appreciable CNS permeability. Beyond melanocytes and dopaminergic neurons, MC1R is expressed in other peripheral tissues and cell types, including immune cells. The present study investigates the impact of NDP-MSH, a synthetic melanocortin receptor (MCR) agonist that does not cross BBB, on the immune system and the nigrostriatal dopaminergic system in mouse model of PD. Methods C57BL/6 mice were treated systemically with MPTP.HCl (20 mg/kg) and LPS (1 mg/kg) from day 1 to day 4 and NDP-MSH (400 μg/kg) or vehicle from day 1 to day 12 following which the mice were sacrificed. Peripheral and CNS immune cells were phenotyped and inflammatory markers were measured. The nigrostriatal dopaminergic system was assessed behaviorally, chemically, immunologically, and pathologically. To understand the role of regulatory T cells (Tregs) in this model, CD25 monoclonal antibody was used to deplete CD25+ Tregs. Results Systemic NDP-MSH administration significantly attenuated striatal dopamine depletion and nigral dopaminergic neuron loss induced by MPTP+LPS. It improved the behavioral outcomes in the pole test. Mc1r mutant mice injected with NDP-MSH in the MPTP and LPS paradigm showed no changes in striatal dopamine levels suggesting that the NDP-MSH acts through the MC1R pathway. Although no NDP-MSH was detected in the brain, peripheral, NDP-MSH attenuated neuroinflammation as observed by diminished microglial activation in the nigral region, along with reduced TNF-α and IL1β levels in the ventral midbrain. Depletion of Tregs limited the neuroprotective effects of NDP-MSH. Conclusions Our study demonstrates that peripherally acting NDP-MSH confers protection on dopaminergic nigrostriatal neurons and reduces hyperactivated microglia. NDP-MSH modulates peripheral immune responses, and Tregs may be involved in the neuroprotective effect of NDP-MSH.
Collapse
Affiliation(s)
- Pranay Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Shuhei Nishiyama
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Sonia H Lin
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Akriti Srivastava
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Chienwen Su
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston
| | - Michael Levy
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Michael Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Yuehang Xu
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School
| |
Collapse
|
22
|
Cheru N, Hafler DA, Sumida TS. Regulatory T cells in peripheral tissue tolerance and diseases. Front Immunol 2023; 14:1154575. [PMID: 37197653 PMCID: PMC10183596 DOI: 10.3389/fimmu.2023.1154575] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/13/2023] [Indexed: 05/19/2023] Open
Abstract
Maintenance of peripheral tolerance by CD4+Foxp3+ regulatory T cells (Tregs) is essential for regulating autoreactive T cells. The loss of function of Foxp3 leads to autoimmune disease in both animals and humans. An example is the rare, X-linked recessive disorder known as IPEX (Immune Dysregulation, Polyendocrinopathy, Enteropathy X-linked) syndrome. In more common human autoimmune diseases, defects in Treg function are accompanied with aberrant effector cytokines such as IFNγ. It has recently become appreciated that Tregs plays an important role in not only maintaining immune homeostasis but also in establishing the tissue microenvironment and homeostasis of non-lymphoid tissues. Tissue resident Tregs show profiles that are unique to their local environments which are composed of both immune and non-immune cells. Core tissue-residence gene signatures are shared across different tissue Tregs and are crucial to homeostatic regulation and maintaining the tissue Treg pool in a steady state. Through interaction with immunocytes and non-immunocytes, tissue Tregs exert a suppressive function via conventional ways involving contact dependent and independent processes. In addition, tissue resident Tregs communicate with other tissue resident cells which allows Tregs to adopt to their local microenvironment. These bidirectional interactions are dependent on the specific tissue environment. Here, we summarize the recent advancements of tissue Treg studies in both human and mice, and discuss the molecular mechanisms that maintain tissue homeostasis and prevent pathogenesis.
Collapse
Affiliation(s)
- Nardos Cheru
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - David A. Hafler
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Tomokazu S. Sumida
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
23
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
24
|
Poppell M, Hammel G, Ren Y. Immune Regulatory Functions of Macrophages and Microglia in Central Nervous System Diseases. Int J Mol Sci 2023; 24:5925. [PMID: 36982999 PMCID: PMC10059890 DOI: 10.3390/ijms24065925] [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: 01/31/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Macrophages can be characterized as a very multifunctional cell type with a spectrum of phenotypes and functions being observed spatially and temporally in various disease states. Ample studies have now demonstrated a possible causal link between macrophage activation and the development of autoimmune disorders. How these cells may be contributing to the adaptive immune response and potentially perpetuating the progression of neurodegenerative diseases and neural injuries is not fully understood. Within this review, we hope to illustrate the role that macrophages and microglia play as initiators of adaptive immune response in various CNS diseases by offering evidence of: (1) the types of immune responses and the processes of antigen presentation in each disease, (2) receptors involved in macrophage/microglial phagocytosis of disease-related cell debris or molecules, and, finally, (3) the implications of macrophages/microglia on the pathogenesis of the diseases.
Collapse
Affiliation(s)
| | | | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
25
|
Olson KE, Mosley RL, Gendelman HE. The potential for treg-enhancing therapies in nervous system pathologies. Clin Exp Immunol 2023; 211:108-121. [PMID: 36041453 PMCID: PMC10019130 DOI: 10.1093/cei/uxac084] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/28/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
While inflammation may not be the cause of disease, it is well known that it contributes to disease pathogenesis across a multitude of peripheral and central nervous system disorders. Chronic and overactive inflammation due to an effector T-cell-mediated aberrant immune response ultimately leads to tissue damage and neuronal cell death. To counteract peripheral and neuroinflammatory responses, research is being focused on regulatory T cell enhancement as a therapeutic target. Regulatory T cells are an immunosuppressive subpopulation of CD4+ T helper cells essential for maintaining immune homeostasis. The cells play pivotal roles in suppressing immune responses to maintain immune tolerance. In so doing, they control T cell proliferation and pro-inflammatory cytokine production curtailing autoimmunity and inflammation. For nervous system pathologies, Treg are known to affect the onset and tempo of neural injuries. To this end, we review recent findings supporting Treg's role in disease, as well as serving as a therapeutic agent in multiple sclerosis, myasthenia gravis, Guillain-Barre syndrome, Parkinson's and Alzheimer's diseases, and amyotrophic lateral sclerosis. An ever-broader role for Treg in the control of neurologic disease has been shown for traumatic brain injury, stroke, neurotrophic pain, epilepsy, and psychiatric disorders. To such ends, this review serves to examine the role played by Tregs in nervous system diseases with a focus on harnessing their functional therapeutic role(s).
Collapse
Affiliation(s)
- Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - R L Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| |
Collapse
|
26
|
Suzzi S, Croese T, Ravid A, Gold O, Clark AR, Medina S, Kitsberg D, Adam M, Vernon KA, Kohnert E, Shapira I, Malitsky S, Itkin M, Brandis A, Mehlman T, Salame TM, Colaiuta SP, Cahalon L, Slyper M, Greka A, Habib N, Schwartz M. N-acetylneuraminic acid links immune exhaustion and accelerated memory deficit in diet-induced obese Alzheimer's disease mouse model. Nat Commun 2023; 14:1293. [PMID: 36894557 PMCID: PMC9998639 DOI: 10.1038/s41467-023-36759-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 02/16/2023] [Indexed: 03/11/2023] Open
Abstract
Systemic immunity supports lifelong brain function. Obesity posits a chronic burden on systemic immunity. Independently, obesity was shown as a risk factor for Alzheimer's disease (AD). Here we show that high-fat obesogenic diet accelerated recognition-memory impairment in an AD mouse model (5xFAD). In obese 5xFAD mice, hippocampal cells displayed only minor diet-related transcriptional changes, whereas the splenic immune landscape exhibited aging-like CD4+ T-cell deregulation. Following plasma metabolite profiling, we identified free N-acetylneuraminic acid (NANA), the predominant sialic acid, as the metabolite linking recognition-memory impairment to increased splenic immune-suppressive cells in mice. Single-nucleus RNA-sequencing revealed mouse visceral adipose macrophages as a potential source of NANA. In vitro, NANA reduced CD4+ T-cell proliferation, tested in both mouse and human. In vivo, NANA administration to standard diet-fed mice recapitulated high-fat diet effects on CD4+ T cells and accelerated recognition-memory impairment in 5xFAD mice. We suggest that obesity accelerates disease manifestation in a mouse model of AD via systemic immune exhaustion.
Collapse
Grants
- R01 DK095045 NIDDK NIH HHS
- R01 DK099465 NIDDK NIH HHS
- the Vera and John Schwartz Family Center for Metabolic Biology.
- the National Institutes of Health (NIH) grants DK095045 and DK099465, the Cure Alzheimer’s Fund, the Chan Zuckerberg Foundation, and the Carlos Slim Foundation.
- the Israel Science Foundation (ISF) research grant no. 1709/19, the European Research Council grant 853409, the MOST-IL-China research grant no. 3-15687, and the Myers Foundation. N.H. holds the Goren-Khazzam chair in neuroscience.
- the Advanced European Research Council grants 232835 and 741744, the European Seventh Framework Program HEALTH-2011 (279017), the Israel Science Foundation (ISF)-research grant no. 991/16, the ISF-Legacy Heritage Bio-medical Science Partnership research grant no. 1354/15, and the Thompson Foundation and Adelis Foundation.
Collapse
Affiliation(s)
- Stefano Suzzi
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel.
| | - Tommaso Croese
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Adi Ravid
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Or Gold
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Abbe R Clark
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sedi Medina
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Daniel Kitsberg
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Miriam Adam
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Katherine A Vernon
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Eva Kohnert
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Inbar Shapira
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel
| | - Sergey Malitsky
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Maxim Itkin
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Alexander Brandis
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Tevie Mehlman
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Tomer M Salame
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot, Israel
| | - Sarah P Colaiuta
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Liora Cahalon
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel
| | - Michal Slyper
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Naomi Habib
- The Hebrew University of Jerusalem, Edmond & Lily Safra Center for Brain Sciences, Jerusalem, Israel.
| | - Michal Schwartz
- Weizmann Institute of Science, Department of Brain Sciences, Rehovot, Israel.
| |
Collapse
|
27
|
Xu QQ, Zhang XL, Luo B, Li T, Yuan G, Shao SY. Role of Peripheral Regulatory T Lymphocytes in Patients with Thyroid Associated Ophthalmopathy During Systemic Glucocorticoid Treatment: A Prospective Observational Study. Curr Med Sci 2023; 43:130-138. [PMID: 36459302 DOI: 10.1007/s11596-022-2671-8] [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: 04/07/2022] [Accepted: 08/31/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVE Thyroid-associated ophthalmopathy (TAO) is an autoimmune disorder involving the orbital tissue. This study aimed to understand the role of regulatory T cells (Tregs) in TAO during 12-week systemic glucocorticoid (GC) treatment. METHODS Thirty-two moderate-severe TAO patients with a clinical activity score (CAS) ≥3/7 or with prolonged T2 relaxation time (T2RT) on at least one side of extraocular muscle (EOM) were enrolled. The percentage of the peripheral CD4+CD25(high)CD127(-/low) Tregs was analyzed using flow cytometry before and after the GC treatment. The activity and severity of TAO, T2RT, and the clinical outcomes after the GC treatment were assessed. Their correlation with the peripheral Tregs was investigated. RESULTS There was no significant association between the baseline Treg fraction and the activity and severity of TAO or the treatment response. A significant reduction of Tregs was observed after the GC therapy merely in patients without any clinical improvement. CONCLUSION Treg reduction after systemic GC therapy is indicative of a poor therapeutic response. Accordingly, dynamic alterations of Tregs could help to evaluate the effectiveness of the GC treatment.
Collapse
Affiliation(s)
- Qin-Qin Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Branch of the National Clinical Research Center for Metabolic Diseases, Wuhan, 430030, China
| | - Xiao-Ling Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Branch of the National Clinical Research Center for Metabolic Diseases, Wuhan, 430030, China
| | - Ban Luo
- Depatment of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tao Li
- Depatment of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gang Yuan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Branch of the National Clinical Research Center for Metabolic Diseases, Wuhan, 430030, China
| | - Shi-Ying Shao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Branch of the National Clinical Research Center for Metabolic Diseases, Wuhan, 430030, China.
| |
Collapse
|
28
|
Canto-Gomes J, Da Silva-Ferreira S, Silva CS, Boleixa D, Martins da Silva A, González-Suárez I, Cerqueira JJ, Correia-Neves M, Nobrega C. People with Primary Progressive Multiple Sclerosis Have a Lower Number of Central Memory T Cells and HLA-DR + Tregs. Cells 2023; 12:439. [PMID: 36766781 PMCID: PMC9913799 DOI: 10.3390/cells12030439] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/13/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
The importance of circulating immune cells to primary progressive multiple sclerosis (PPMS) pathophysiology is still controversial because most immunotherapies were shown to be ineffective in treating people with PPMS (pwPPMS). Yet, although controversial, data exist describing peripheral immune system alterations in pwPPMS. This study aims to investigate which alterations might be present in pwPPMS free of disease-modifying drugs (DMD) in comparison to age- and sex-matched healthy controls. A multicentric cross-sectional study was performed using 23 pwPPMS and 23 healthy controls. The phenotype of conventional CD4+ and CD8+ T cells, regulatory T cells (Tregs), B cells, natural killer (NK) T cells and NK cells was assessed. Lower numbers of central memory CD4+ and CD8+ T cells and activated HLA-DR+ Tregs were observed in pwPPMS. Regarding NK and NKT cells, pwPPMS presented higher percentages of CD56dimCD57+ NK cells expressing NKp46 and of NKT cells expressing KIR2DL2/3 and NKp30. Higher disease severity scores and an increasing time since diagnosis was correlated with lower numbers of inhibitory NK cells subsets. Our findings contribute to reinforcing the hypotheses that alterations in peripheral immune cells are present in pwPPMS and that changes in NK cell populations are the strongest correlate of disease severity.
Collapse
Affiliation(s)
- João Canto-Gomes
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Sara Da Silva-Ferreira
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Carolina S. Silva
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Division of Infectious Diseases, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, 17176 Stockholm, Sweden
| | | | - Ana Martins da Silva
- Porto University Hospital Center, 4099-001 Porto, Portugal
- Multidisciplinary Unit for Biomedical Research (UMIB)—ICBAS, University of Porto, 4050-346 Porto, Portugal
| | - Inés González-Suárez
- University Hospital Complex of Vigo, 36312 Vigo, Spain
- Álvaro Cunqueiro Hospital, 36312 Vigo, Spain
| | - João J. Cerqueira
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Hospital of Braga, 4710-243 Braga, Portugal
- Clinical Academic Centre, Hospital of Braga, 4710-243 Braga, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Division of Infectious Diseases, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Claudia Nobrega
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
29
|
Andronie-Cioara FL, Ardelean AI, Nistor-Cseppento CD, Jurcau A, Jurcau MC, Pascalau N, Marcu F. Molecular Mechanisms of Neuroinflammation in Aging and Alzheimer's Disease Progression. Int J Mol Sci 2023; 24:ijms24031869. [PMID: 36768235 PMCID: PMC9915182 DOI: 10.3390/ijms24031869] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/01/2023] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Aging is the most prominent risk factor for late-onset Alzheimer's disease. Aging associates with a chronic inflammatory state both in the periphery and in the central nervous system, the evidence thereof and the mechanisms leading to chronic neuroinflammation being discussed. Nonetheless, neuroinflammation is significantly enhanced by the accumulation of amyloid beta and accelerates the progression of Alzheimer's disease through various pathways discussed in the present review. Decades of clinical trials targeting the 2 abnormal proteins in Alzheimer's disease, amyloid beta and tau, led to many failures. As such, targeting neuroinflammation via different strategies could prove a valuable therapeutic strategy, although much research is still needed to identify the appropriate time window. Active research focusing on identifying early biomarkers could help translating these novel strategies from bench to bedside.
Collapse
Affiliation(s)
- Felicia Liana Andronie-Cioara
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Adriana Ioana Ardelean
- Department of Preclinical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Carmen Delia Nistor-Cseppento
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
- Correspondence: (C.D.N.-C.); (N.P.)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | | | - Nicoleta Pascalau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
- Correspondence: (C.D.N.-C.); (N.P.)
| | - Florin Marcu
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
30
|
Deng G, Tang Y, Xiao J, Chen X, Chu YH, Shang K, Zhou LQ, Qin C, Wang F, Tian DS. Naïve-memory regulatory T cells ratio is a prognostic biomarker for patients with acute ischemic stroke. Front Aging Neurosci 2023; 15:1072980. [PMID: 36909948 PMCID: PMC9995800 DOI: 10.3389/fnagi.2023.1072980] [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: 10/18/2022] [Accepted: 02/03/2023] [Indexed: 02/25/2023] Open
Abstract
Background Regulatory T cells (Treg) have been identified as a key modulator of neuroinflammation in stroke. However, little is known about the association of Treg subpopulations with clinical outcome in patients with acute ischemic stroke (AIS). Methods Patients within 1 week from stroke onset were prospectively enrolled in this study. Healthy controls were sex-and age-matched 1:1 to AIS patients. The frequencies of Treg and Treg subsets were analyzed by flow cytometry and compared with nonstroke control. Univariate and multivariate logistic regression analysis was performed to investigate the prognostic value of Treg subsets in stroke outcomes. Results A total of 328 patients and 328 controls were included in the study. Compared with controls, patients with AIS had higher levels of Treg frequency and memory Treg (mTreg) frequency, but lower levels of naïve Treg (nTreg) frequency and nTreg/mTreg ratio. One hundred twenty-six (38.4%) patients experienced unfavorable outcome (modified Rankin score 2-6). Multivariate regression analysis showed that nTreg/mTreg ratio was negatively associated with unfavorable 90-day outcome (the highest tertile versus the lowest tertile: odds ratio 0.13, 95% confidential interval [CI] 0.05-0.35). The risk estimation of unfavorable 90 day outcome can be significantly improved by adding nTreg/mTreg ratio to the conventional clinical parameters (continuous net reclassification improvement 91.26, 95% CI 69.04-113.5%, p < 0.001; integrated discrimination improvement 22.38, 95% CI 17.16-27.59%, p < 0.001). Conclusion This study showed that patients with AIS had elevated Treg frequency and mTreg frequency, but reduced nTreg frequency and nTreg/mTreg ratio. Admission nTreg/mTreg ratio was an independent predictor of unfavorable 90 day outcome in AIS. However, large sample-size cohort studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Gang Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xiao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Tatara Y, Yamazaki H, Katsuoka F, Chiba M, Saigusa D, Kasai S, Nakamura T, Inoue J, Aoki Y, Shoji M, Motoike IN, Tamada Y, Hashizume K, Shoji M, Kinoshita K, Murashita K, Nakaji S, Yamamoto M, Itoh K. Multiomics and artificial intelligence enabled peripheral blood-based prediction of amnestic mild cognitive impairment. Curr Res Transl Med 2023; 71:103367. [PMID: 36446162 DOI: 10.1016/j.retram.2022.103367] [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: 03/28/2022] [Revised: 08/03/2022] [Accepted: 10/20/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Since dementia is preventable with early interventions, biomarkers that assist in diagnosing early stages of dementia, such as mild cognitive impairment (MCI), are urgently needed. METHODS Multiomics analysis of amnestic MCI (aMCI) peripheral blood (n = 25) was performed covering the transcriptome, microRNA, proteome, and metabolome. Validation analysis for microRNAs was conducted in an independent cohort (n = 12). Artificial intelligence was used to identify the most important features for predicting aMCI. FINDINGS We found that hsa-miR-4455 is the best biomarker in all omics analyses. The diagnostic index taking a ratio of hsa-miR-4455 to hsa-let-7b-3p predicted aMCI patients against healthy subjects with 97% overall accuracy. An integrated review of multiomics data suggested that a subset of T cells and the GCN (general control nonderepressible) pathway are associated with aMCI. INTERPRETATION The multiomics approach has enabled aMCI biomarkers with high specificity and illuminated the accompanying changes in peripheral blood. Future large-scale studies are necessary to validate candidate biomarkers for clinical use.
Collapse
Affiliation(s)
- Yota Tatara
- Department of Stress Response Science, Center for Advanced Medical Research, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Hiromi Yamazaki
- Department of Stress Response Science, Center for Advanced Medical Research, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Fumiki Katsuoka
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Mitsuru Chiba
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, 66-1 Hon-cho, Hirosaki, Aomori 036-8564, Japan
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Shuya Kasai
- Department of Stress Response Science, Center for Advanced Medical Research, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Tomohiro Nakamura
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Division of Personalized Prevention and Epidemiology, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Jin Inoue
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Yuichi Aoki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Miho Shoji
- Human Metabolome Technologies, Inc., 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan
| | - Ikuko N Motoike
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Yoshinori Tamada
- Innovation Center for Health Promotion, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Katsuhito Hashizume
- Human Metabolome Technologies, Inc., 246-2 Mizukami, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan
| | - Mikio Shoji
- Department of Neurology, Gunma University Hospital, 3-39-15 Showamachi, Maebashi, Gunma 371-8511, Japan; Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, 3-26-8 Ootomo-machi, Maebashi, Gunma 371-0847 Japan; COI Research Initiatives Organization, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8216, Japan
| | - Kengo Kinoshita
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan; Department of Applied Information Sciences, Graduate School of Information Sciences, Tohoku University, 6-6-05 Aramaki Aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan; Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Koichi Murashita
- COI Research Initiatives Organization, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8216, Japan
| | - Shigeyuki Nakaji
- Department of Social Medicine, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8216, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan; Department of Medical Biochemistry, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Ken Itoh
- Department of Stress Response Science, Center for Advanced Medical Research, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan.
| |
Collapse
|
32
|
Lai Y, Lin P, Lin F, Chen M, Lin C, Lin X, Wu L, Zheng M, Chen J. Identification of immune microenvironment subtypes and signature genes for Alzheimer's disease diagnosis and risk prediction based on explainable machine learning. Front Immunol 2022; 13:1046410. [PMID: 36569892 PMCID: PMC9773397 DOI: 10.3389/fimmu.2022.1046410] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Background Using interpretable machine learning, we sought to define the immune microenvironment subtypes and distinctive genes in AD. Methods ssGSEA, LASSO regression, and WGCNA algorithms were used to evaluate immune state in AD patients. To predict the fate of AD and identify distinctive genes, six machine learning algorithms were developed. The output of machine learning models was interpreted using the SHAP and LIME algorithms. For external validation, four separate GEO databases were used. We estimated the subgroups of the immunological microenvironment using unsupervised clustering. Further research was done on the variations in immunological microenvironment, enhanced functions and pathways, and therapeutic medicines between these subtypes. Finally, the expression of characteristic genes was verified using the AlzData and pan-cancer databases and RT-PCR analysis. Results It was determined that AD is connected to changes in the immunological microenvironment. WGCNA revealed 31 potential immune genes, of which the greenyellow and blue modules were shown to be most associated with infiltrated immune cells. In the testing set, the XGBoost algorithm had the best performance with an AUC of 0.86 and a P-R value of 0.83. Following the screening of the testing set by machine learning algorithms and the verification of independent datasets, five genes (CXCR4, PPP3R1, HSP90AB1, CXCL10, and S100A12) that were closely associated with AD pathological biomarkers and allowed for the accurate prediction of AD progression were found to be immune microenvironment-related genes. The feature gene-based nomogram may provide clinical advantages to patients. Two immune microenvironment subgroups for AD patients were identified, subtype2 was linked to a metabolic phenotype, subtype1 belonged to the immune-active kind. MK-866 and arachidonyltrifluoromethane were identified as the top treatment agents for subtypes 1 and 2, respectively. These five distinguishing genes were found to be intimately linked to the development of the disease, according to the Alzdata database, pan-cancer research, and RT-PCR analysis. Conclusion The hub genes associated with the immune microenvironment that are most strongly associated with the progression of pathology in AD are CXCR4, PPP3R1, HSP90AB1, CXCL10, and S100A12. The hypothesized molecular subgroups might offer novel perceptions for individualized AD treatment.
Collapse
Affiliation(s)
- Yongxing Lai
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Peiqiang Lin
- Department of Neurology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Fan Lin
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Manli Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Chunjin Lin
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Xing Lin
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Lijuan Wu
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Mouwei Zheng
- Department of Geriatric Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Fuzhou, Fujian, China,*Correspondence: Jianhao Chen, ; Mouwei Zheng,
| | - Jianhao Chen
- Department of Rehabilitation Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China,*Correspondence: Jianhao Chen, ; Mouwei Zheng,
| |
Collapse
|
33
|
Watchmaker PB, Colton M, Pineo-Cavanaugh PL, Okada H. Future development of chimeric antigen receptor T cell therapies for patients suffering from malignant glioma. Curr Opin Oncol 2022; 34:661-669. [PMID: 35855503 PMCID: PMC9560977 DOI: 10.1097/cco.0000000000000877] [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] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Chimeric antigen receptor (CAR) T cell therapy has been successful in some haematologic malignancies, but the central nervous system (CNS) presents unique obstacles to its use against tumours arising therein. This review discusses recent improvements in the delivery and design of these cells to improve the efficacy and safety of this treatment against malignant gliomas. RECENT FINDINGS The immunosuppressive environment of the CNS affects the functionality of CAR T cells, but recent developments using metabolic manipulation and cytokine delivery have shown that the performance of CAR T cells can be improved in this environment. Emerging techniques can improve the delivery of CAR T cells to the CNS parenchyma, which is normally well protected from peripheral immune cells. The implementation of novel antigens and CAR-expression regulation strategies will improve the specificity and efficacy of these cells. Finally, although autologous T cells have historically been the standard, recent developments have made the use of allogeneic T cells or natural killer (NK) cells more clinically feasible. SUMMARY The discoveries highlighted in this review will aid the development of CAR cells that are safer, more resilient against immunosuppressive signals in the CNS, and able to specifically target intracranial tumour cells.
Collapse
Affiliation(s)
| | - Maggie Colton
- Department of Neurosurgery, University of California, San Francisco
| | | | - Hideho Okada
- Department of Neurosurgery, University of California, San Francisco
- Parker Institute for Cancer Immunotherapy
| |
Collapse
|
34
|
Georgoulias G, Zaravinos A. Genomic landscape of the immunogenicity regulation in skin melanomas with diverse tumor mutation burden. Front Immunol 2022; 13:1006665. [PMID: 36389735 PMCID: PMC9650672 DOI: 10.3389/fimmu.2022.1006665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/10/2022] [Indexed: 08/27/2023] Open
Abstract
Skin melanoma cells are tightly interconnected with their tumor microenvironment (TME), which influences their initiation, progression, and sensitivity/resistance to therapeutic interventions. An immune-active TME favors patient response to immune checkpoint inhibition (ICI), but not all patients respond to therapy. Here, we assessed differential gene expression in primary and metastatic tumors from the TCGA-SKCM dataset, compared to normal skin samples from the GTEx project and validated key findings across 4 independent GEO datasets, as well as using immunohistochemistry in independent patient cohorts. We focused our attention on examining the expression of various immune receptors, immune-cell fractions, immune-related signatures and mutational signatures across cutaneous melanomas with diverse tumor mutation burdens (TMB). Globally, the expression of most immunoreceptors correlated with patient survival, but did not differ between TMBhigh and TMBlow tumors. Melanomas were enriched in "naive T-cell", "effector memory T-cell", "exhausted T-cell", "resting Treg T-cell" and "Th1-like" signatures, irrespective of their BRAF, NF1 or RAS mutational status. Somatic mutations in IDO1 and HLA-DRA were frequent and could be involved in hindering patient response to ICI therapies. We finally analyzed transcriptome profiles of ICI-treated patients and associated their response with high levels of IFNγ, Merck18, CD274, CD8, and low levels of myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs) and M2 macrophages, irrespective of their TMB status. Overall, our findings highlight the importance of pre-existing T-cell immunity in ICI therapeutic outcomes in skin melanoma and suggest that TMBlow patients could also benefit from such therapies.
Collapse
Affiliation(s)
- George Georgoulias
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
- Cancer Genetics, Genomics and Systems Biology laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus
| |
Collapse
|
35
|
Princiotta Cariddi L, Mauri M, Cosentino M, Versino M, Marino F. Alzheimer's Disease: From Immune Homeostasis to Neuroinflammatory Condition. Int J Mol Sci 2022; 23:13008. [PMID: 36361799 PMCID: PMC9658357 DOI: 10.3390/ijms232113008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's Disease is the most common cause in the world of progressive cognitive decline. Although many modifiable and non-modifiable risk factors have been proposed, in recent years, neuroinflammation has been hypothesized to be an important contributing factor of Alzheimer's Disease pathogenesis. Neuroinflammation can occur through the combined action of the Central Nervous System resident immune cells and adaptive peripheral immune system. In the past years, immunotherapies for neurodegenerative diseases have focused wrongly on targeting protein aggregates Aβ plaques and NFT treatment. The role of both innate and adaptive immune cells has not been fully clarified, but several data suggest that immune system dysregulation plays a key role in neuroinflammation. Recent studies have focused especially on the role of the adaptive immune system and have shown that inflammatory markers are characterized by increased CD4+ Teff cells' activities and reduced circulating CD4+ Treg cells. In this review, we discuss the key role of both innate and adaptive immune systems in the degeneration and regeneration mechanisms in the pathogenesis of Alzheimer's Disease, with a focus on how the crosstalk between these two systems is able to sustain brain homeostasis or shift it to a neurodegenerative condition.
Collapse
Affiliation(s)
- Lucia Princiotta Cariddi
- PhD Program in Clinical and Experimental Medicine and Medical Humanities, University of Insubria, 21100 Varese, Italy
- Neurology and Stroke Unit, ASST Sette Laghi Hospital, 21100 Varese, Italy
| | - Marco Mauri
- Neurology and Stroke Unit, ASST Sette Laghi Hospital, 21100 Varese, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Marco Cosentino
- Center of Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy
| | - Maurizio Versino
- Neurology and Stroke Unit, ASST Sette Laghi Hospital, 21100 Varese, Italy
- Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| | - Franca Marino
- Center of Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
36
|
Mason HD, McGavern DB. How the immune system shapes neurodegenerative diseases. Trends Neurosci 2022; 45:733-748. [PMID: 36075783 PMCID: PMC9746609 DOI: 10.1016/j.tins.2022.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/10/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022]
Abstract
Neurodegenerative diseases are a major cause of death and disability worldwide and are influenced by many factors including age, genetics, and injuries. While these diseases are often thought to result from the accumulation and spread of aberrant proteins, recent studies have demonstrated that they can be shaped by the innate and adaptive immune system. Resident myeloid cells typically mount a sustained response to the degenerating CNS, but peripheral leukocytes such as T and B cells can also alter disease trajectories. Here, we review the sometimes-dichotomous roles played by immune cells during neurodegenerative diseases and explore how brain trauma can serve as a disease initiator or accelerant. We also offer insights into how failure to properly resolve a CNS injury might promote the development of a neurodegenerative disease.
Collapse
Affiliation(s)
- Hannah D Mason
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
37
|
Ashraf H, Solla P, Sechi LA. Current Advancement of Immunomodulatory Drugs as Potential Pharmacotherapies for Autoimmunity Based Neurological Diseases. Pharmaceuticals (Basel) 2022; 15:ph15091077. [PMID: 36145298 PMCID: PMC9504155 DOI: 10.3390/ph15091077] [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: 07/27/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Dramatic advancement has been made in recent decades to understand the basis of autoimmunity-mediated neurological diseases. These diseases create a strong influence on the central nervous system (CNS) and the peripheral nervous system (PNS), leading to various clinical manifestations and numerous symptoms. Multiple sclerosis (MS) is the most prevalent autoimmune neurological disease while NMO spectrum disorder (NMOSD) is less common. Furthermore, evidence supports the presence of autoimmune mechanisms contributing to the pathogenesis of amyotrophic lateral sclerosis (ALS), which is a neurodegenerative disorder characterized by the progressive death of motor neurons. Additionally, autoimmunity is believed to be involved in the basis of Alzheimer’s and Parkinson’s diseases. In recent years, the prevalence of autoimmune-based neurological disorders has been elevated and current findings strongly suggest the role of pharmacotherapies in controlling the progression of autoimmune diseases. Therefore, this review focused on the current advancement of immunomodulatory drugs as novel approaches in the management of autoimmune neurological diseases and their future outlook.
Collapse
Affiliation(s)
- Hajra Ashraf
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Paolo Solla
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Leonardo Atonio Sechi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Complex Structure of Microbology and Virology, AOU Sassari, 07100 Sassari, Italy
- Correspondence:
| |
Collapse
|
38
|
Pelagic distribution of plastic debris (> 500 µm) and marine organisms in the upper layer of the North Atlantic Ocean. Sci Rep 2022; 12:13465. [PMID: 35953623 PMCID: PMC9372048 DOI: 10.1038/s41598-022-17742-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/29/2022] [Indexed: 11/28/2022] Open
Abstract
At present, the distribution of plastic debris in the ocean water column remains largely unknown. Such information, however, is required to assess the exposure of marine organisms to plastic pollution as well as to calculate the ocean plastic mass balance. Here, we provide water column profiles (0–300 m water depth) of plastic (0.05–5 cm in size) concentration and key planktonic species from the eastern North Atlantic Ocean. The amount of plastic decreases rapidly in the upper few meters, from ~ 1 item/m3 (~ 1000 µg/m3) at the sea surface to values of ~ 0.001–0.01 items/m3 (~ 0.1–10 µg/m3) at 300 m depth. Ratios of plastic to plankton varied between ~ 10–5 and 1 plastic particles per individual with highest ratios typically found in the surface waters. We further observed that pelagic ratios were generally higher in the water column below the subtropical gyre compared to those in more coastal ecosystems. Lastly, we show plastic to (non-gelatinous) plankton ratios could be as high as ~ 102–107 plastic particles per individual when considering reported concentrations of small microplastics < 100 μm. Plastic pollution in our oceans may therefore soon exceed estimated safe concentrations for many pelagic species.
Collapse
|
39
|
Schlöder J, Shahneh F, Schneider FJ, Wieschendorf B. Boosting regulatory T cell function for the treatment of autoimmune diseases – That’s only half the battle! Front Immunol 2022; 13:973813. [PMID: 36032121 PMCID: PMC9400058 DOI: 10.3389/fimmu.2022.973813] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/18/2022] [Indexed: 01/04/2023] Open
Abstract
Regulatory T cells (Treg) represent a subset of specialized T cells that are essential for the regulation of immune responses and maintenance of peripheral tolerance. Once activated, Treg exert powerful immunosuppressive properties, for example by inhibiting T cell-mediated immune responses against self-antigens, thereby protecting our body from autoimmunity. Autoimmune diseases such as multiple sclerosis, rheumatoid arthritis or systemic lupus erythematosus, exhibit an immunological imbalance mainly characterized by a reduced frequency and impaired function of Treg. In addition, there has been increasing evidence that – besides Treg dysfunction – immunoregulatory mechanisms fail to control autoreactive T cells due to a reduced responsiveness of T effector cells (Teff) for the suppressive properties of Treg, a process termed Treg resistance. In order to efficiently treat autoimmune diseases and thus fully induce immunological tolerance, a combined therapy aimed at both enhancing Treg function and restoring Teff responsiveness could most likely be beneficial. This review provides an overview of immunomodulating drugs that are currently used to treat various autoimmune diseases in the clinic and have been shown to increase Treg frequency as well as Teff sensitivity to Treg-mediated suppression. Furthermore, we discuss strategies on how to boost Treg activity and function, and their potential use in the treatment of autoimmunity. Finally, we present a humanized mouse model for the preclinical testing of Treg-activating substances in vivo.
Collapse
Affiliation(s)
- Janine Schlöder
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- *Correspondence: Janine Schlöder,
| | - Fatemeh Shahneh
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Franz-Joseph Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Björn Wieschendorf
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- ActiTrexx GmbH, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
40
|
Zhou J, Benoit M, Sharoar MG. Recent advances in pre-clinical diagnosis of Alzheimer's disease. Metab Brain Dis 2022; 37:1703-1725. [PMID: 33900524 DOI: 10.1007/s11011-021-00733-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is the most common dementia with currently no known cures or disease modifying treatments (DMTs), despite much time and effort from the field. Diagnosis and intervention of AD during the early pre-symptomatic phase of the disease is thought to be a more effective strategy. Therefore, the detection of biomarkers has emerged as a critical tool for monitoring the effect of new AD therapies, as well as identifying patients most likely to respond to treatment. The establishment of the amyloid/tau/neurodegeneration (A/T/N) framework in 2018 has codified the contexts of use of AD biomarkers in neuroimaging and bodily fluids for research and diagnostic purposes. Furthermore, a renewed drive for novel AD biomarkers and innovative methods of detection has emerged with the goals of adding additional insight to disease progression and discovery of new therapeutic targets. The use of biomarkers has accelerated the development of AD drugs and will bring new therapies to patients in need. This review highlights recent methods utilized to diagnose antemortem AD.
Collapse
Affiliation(s)
- John Zhou
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
- Molecular Medicine Program, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Marc Benoit
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Md Golam Sharoar
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, 06030, USA.
| |
Collapse
|
41
|
Samuels H, Malov M, Saha Detroja T, Ben Zaken K, Bloch N, Gal-Tanamy M, Avni O, Polis B, Samson AO. Autoimmune Disease Classification Based on PubMed Text Mining. J Clin Med 2022; 11:4345. [PMID: 35893435 PMCID: PMC9369164 DOI: 10.3390/jcm11154345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/15/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022] Open
Abstract
Autoimmune diseases (AIDs) are often co-associated, and about 25% of patients with one AID tend to develop other comorbid AIDs. Here, we employ the power of datamining to predict the comorbidity of AIDs based on their normalized co-citation in PubMed. First, we validate our technique in a test dataset using earlier-reported comorbidities of seven knowns AIDs. Notably, the prediction correlates well with comorbidity (R = 0.91) and validates our methodology. Then, we predict the association of 100 AIDs and classify them using principal component analysis. Our results are helpful in classifying AIDs into one of the following systems: (1) gastrointestinal, (2) neuronal, (3) eye, (4) cutaneous, (5) musculoskeletal, (6) kidneys and lungs, (7) cardiovascular, (8) hematopoietic, (9) endocrine, and (10) multiple. Our classification agrees with experimentally based taxonomy and ranks AID according to affected systems and gender. Some AIDs are unclassified and do not associate well with other AIDs. Interestingly, Alzheimer's disease correlates well with other AIDs such as multiple sclerosis. Finally, our results generate a network classification of autoimmune diseases based on PubMed text mining and help map this medical universe. Our results are expected to assist healthcare workers in diagnosing comorbidity in patients with an autoimmune disease, and to help researchers in identifying common genetic, environmental, and autoimmune mechanisms.
Collapse
Affiliation(s)
- Hadas Samuels
- Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel; (H.S.); (M.M.); (T.S.D.); (K.B.Z.); (N.B.); (M.G.-T.); (O.A.)
| | - Malki Malov
- Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel; (H.S.); (M.M.); (T.S.D.); (K.B.Z.); (N.B.); (M.G.-T.); (O.A.)
| | - Trishna Saha Detroja
- Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel; (H.S.); (M.M.); (T.S.D.); (K.B.Z.); (N.B.); (M.G.-T.); (O.A.)
| | - Karin Ben Zaken
- Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel; (H.S.); (M.M.); (T.S.D.); (K.B.Z.); (N.B.); (M.G.-T.); (O.A.)
| | - Naamah Bloch
- Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel; (H.S.); (M.M.); (T.S.D.); (K.B.Z.); (N.B.); (M.G.-T.); (O.A.)
| | - Meital Gal-Tanamy
- Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel; (H.S.); (M.M.); (T.S.D.); (K.B.Z.); (N.B.); (M.G.-T.); (O.A.)
| | - Orly Avni
- Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel; (H.S.); (M.M.); (T.S.D.); (K.B.Z.); (N.B.); (M.G.-T.); (O.A.)
| | - Baruh Polis
- School of Medicine, Yale University, New Haven, CT 06520, USA;
| | - Abraham O. Samson
- Azrieli Faculty of Medicine, Bar Ilan University, Safed 1311502, Israel; (H.S.); (M.M.); (T.S.D.); (K.B.Z.); (N.B.); (M.G.-T.); (O.A.)
| |
Collapse
|
42
|
van Olst L, Coenen L, Nieuwland JM, Rodriguez-Mogeda C, de Wit NM, Kamermans A, Middeldorp J, de Vries HE. Crossing borders in Alzheimer's disease: A T cell's perspective. Adv Drug Deliv Rev 2022; 188:114398. [PMID: 35780907 DOI: 10.1016/j.addr.2022.114398] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia affecting millions of people worldwide. While different immunotherapies are imminent, currently only disease-modifying medications are available and a cure is lacking. Over the past decade, immunological interfaces of the central nervous system (CNS) and their role in neurodegenerative diseases received increasing attention. Specifically, emerging evidence shows that subsets of circulating CD8+ T cells cross the brain barriers and associate with AD pathology. To gain more insight into how the adaptive immune system is involved in disease pathogenesis, we here provide a comprehensive overview of the contribution of T cells to AD pathology, incorporating changes at the brain barriers. In addition, we review studies that provide translation of these findings by targeting T cells to combat AD pathology and cognitive decline. Importantly, these data show that immunological changes in AD are not confined to the CNS and that AD-associated systemic immune changes appear to affect brain homeostasis.
Collapse
Affiliation(s)
- L van Olst
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - L Coenen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands; Department of Neurobiology and Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - J M Nieuwland
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands; Department of Neurobiology and Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - C Rodriguez-Mogeda
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - N M de Wit
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - A Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - J Middeldorp
- Department of Neurobiology and Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - H E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Liu W, Fan M, Lu W, Zhu W, Meng L, Lu S. Emerging Roles of T Helper Cells in Non-Infectious Neuroinflammation: Savior or Sinner. Front Immunol 2022; 13:872167. [PMID: 35844577 PMCID: PMC9280647 DOI: 10.3389/fimmu.2022.872167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/30/2022] [Indexed: 12/03/2022] Open
Abstract
CD4+ T cells, also known as T helper (Th) cells, contribute to the adaptive immunity both in the periphery and in the central nervous system (CNS). At least seven subsets of Th cells along with their signature cytokines have been identified nowadays. Neuroinflammation denotes the brain’s immune response to inflammatory conditions. In recent years, various CNS disorders have been related to the dysregulation of adaptive immunity, especially the process concerning Th cells and their cytokines. However, as the functions of Th cells are being discovered, it’s also found that their roles in different neuroinflammatory conditions, or even the participation of a specific Th subset in one CNS disorder may differ, and sometimes contrast. Based on those recent and contradictory evidence, the conflicting roles of Th cells in multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, epilepsy, traumatic brain injury as well as some typical mental disorders will be reviewed herein. Research progress, limitations and novel approaches concerning different neuroinflammatory conditions will also be mentioned and compared.
Collapse
Affiliation(s)
- Wenbin Liu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Department of Neurosurgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Meiyang Fan
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wen Lu
- Department of Psychiatry, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wenhua Zhu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| | - Liesu Meng
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| | - Shemin Lu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
| |
Collapse
|
44
|
Liston A, Dooley J, Yshii L. Brain-resident regulatory T cells and their role in health and disease. Immunol Lett 2022; 248:26-30. [PMID: 35697195 DOI: 10.1016/j.imlet.2022.06.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 01/01/2023]
Abstract
Regulatory T cells (Tregs) control inflammation and maintain immune homeostasis. The well-characterised circulatory population of CD4+Foxp3+ Tregs is effective at preventing autoimmunity and constraining the immune response, through direct and indirect restraint of conventional T cell activation. Recent advances in Treg cell biology have identified tissue-resident Tregs, with tissue-specific functions that contribute to the maintenance of tissue homeostasis and repair. A population of brain-resident Tregs, characterised as CD69+, has recently been identified in the healthy brain of mice and humans, with rapid population expansion observed under a number of neuroinflammatory conditions. During neuroinflammation, brain-resident Tregs have been proposed to control astrogliosis through the production of amphiregulin, polarize microglia into neuroprotective states, and restrain inflammatory responses by releasing IL-10. While protective effects for Tregs have been demonstrated in a number of neuroinflammatory pathologies, a clear demarcation between the role of circulatory and brain-resident Tregs has been difficult to achieve. Here we review the state-of-the-art for brain-resident Treg population, and describe their potential utilization as a therapeutic target across different neuroinflammatory conditions.
Collapse
Affiliation(s)
- Adrian Liston
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT United Kingdom.
| | - James Dooley
- Immunology Programme, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT United Kingdom
| | - Lidia Yshii
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven 3000, Belgium.
| |
Collapse
|
45
|
Canto-Gomes J, Silva CS, Rb-Silva R, Boleixa D, da Silva AM, Cheynier R, Costa P, González-Suárez I, Correia-Neves M, Cerqueira JJ, Nobrega C. Low Memory T Cells Blood Counts and High Naïve Regulatory T Cells Percentage at Relapsing Remitting Multiple Sclerosis Diagnosis. Front Immunol 2022; 13:901165. [PMID: 35711452 PMCID: PMC9196633 DOI: 10.3389/fimmu.2022.901165] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/22/2022] [Indexed: 11/30/2022] Open
Abstract
Objective The aim of this study is to assess the peripheral immune system of newly diagnosed patients with relapsing remitting multiple sclerosis (RRMS) and compare it to healthy controls (HC). Methods This cross-sectional study involves 30 treatment-naïve newly diagnosed patients with RRMS and 33 sex- and age-matched HC. Peripheral blood mononuclear cells were analyzed regarding: i) thymic function surrogates [T cell receptor excision circles (TRECs) and recent thymic emigrants (RTEs)]; ii) naïve and memory CD4+ and CD8+ T cells subsets; iii) T helper (Th) phenotype and chemokine receptors expression on CD8+ T cells subsets; iv) regulatory T cell (Tregs) phenotype; and exclude expression of activating/inhibitory receptors by natural killer (NK) and NKT cells. Analyses were controlled for age, sex, and human cytomegalovirus (HCMV) IgG seroprevalence. Results Newly diagnosed patients with RRMS and HC have equivalent thymic function as determined by similar numbers of RTEs and levels of sjTRECs, DJβTRECs, and sj/DJβTREC ratio. In the CD8+ T cells compartment, patients with RRMS have a higher naive to memory ratio and lower memory cell counts in blood, specifically of effector memory and TemRA CD8+ T cells. Interestingly, higher numbers and percentages of central memory CD8+ T cells are associated with increasing time from the relapse. Among CD4+ T cells, lower blood counts of effector memory cells are found in patients upon controlling for sex, age, and anti-HCMV IgG seroprevalence. Higher numbers of CD4+ T cells (both naïve and memory) and of Th2 cells are associated with increasing time from the relapse; lower numbers of Th17 cells are associated with higher MS severity scores (MSSS). Patients with RRMS have a higher percentage of naïve Tregs compared with HC, and lower percentages of these cells are associated with higher MSSS. Percentages of immature CD56bright NK cells expressing the inhibitory receptor KLRG1 and of mature CD56dimCD57+ NK cells expressing NKp30 are higher in patients. No major alterations are observed on NKT cells. Conclusion Characterization of the peripheral immune system of treatment-naïve newly diagnosed patients with RRMS unveiled immune features present at clinical onset including lower memory T cells blood counts, particularly among CD8+ T cells, higher percentage of naïve Tregs and altered percentages of NK cells subsets expressing inhibitory or activating receptors. These findings might set the basis to better understand disease pathogenesis.
Collapse
Affiliation(s)
- João Canto-Gomes
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
| | - Carolina S. Silva
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
- Division of Infectious Diseases and Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Rita Rb-Silva
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
- Laboratory of Histology and Embryology, Department of Microscopy, ICBAS, University of Porto, Porto, Portugal
| | | | - Ana Martins da Silva
- Porto University Hospital Center, Porto, Portugal
- Multidisciplinary Unit for Biomedical Research (UMIB) - Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Rémi Cheynier
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Patrício Costa
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
| | - Inés González-Suárez
- University Hospital Complex of Vigo, Vigo, Spain
- Álvaro Cunqueiro Hospital, Vigo, Spain
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
- Division of Infectious Diseases and Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - João J. Cerqueira
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
- Hospital of Braga, Braga, Portugal
- Clinical Academic Centre, Hospital of Braga, Braga, Portugal
| | - Claudia Nobrega
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, Braga, Portugal
- *Correspondence: Claudia Nobrega,
| |
Collapse
|
46
|
Fohner AE, Sitlani CM, Buzkova P, Doyle MF, Liu X, Bis JC, Fitzpatrick A, Heckbert SR, Huber SA, Kuller L, Longstreth WT, Feinstein MJ, Freiberg M, Olson NC, Seshadri S, Lopez O, Odden MC, Tracy RP, Psaty BM, Delaney JA, Floyd JS. Association of Peripheral Lymphocyte Subsets with Cognitive Decline and Dementia: The Cardiovascular Health Study. J Alzheimers Dis 2022; 88:7-15. [PMID: 35527553 PMCID: PMC9277688 DOI: 10.3233/jad-220091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Alison E. Fohner
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Colleen M. Sitlani
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Petra Buzkova
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Margaret F. Doyle
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Xiaojuan Liu
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Annette Fitzpatrick
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Family Medicine and Global Health, University of Washington, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Sally A. Huber
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Lewis Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - William T. Longstreth
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Matthew J. Feinstein
- Departments of Medicine, Preventive Medicine and Pathology, Northwestern University, Evanston, IL, USA
| | - Matthew Freiberg
- Department of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | - Nels C. Olson
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, TX, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Oscar Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michelle C. Odden
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
| | - Russell P. Tracy
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Joseph A. Delaney
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- College of Pharmacy, University of Manitoba, Winnipeg, MB, Canada
| | - James S. Floyd
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
47
|
McManus RM. The Role of Immunity in Alzheimer's Disease. Adv Biol (Weinh) 2022; 6:e2101166. [PMID: 35254006 DOI: 10.1002/adbi.202101166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 02/03/2022] [Indexed: 01/27/2023]
Abstract
With the increase in the aging population, age-related conditions such as dementia and Alzheimer's disease will become ever more prevalent in society. As there is no cure for dementia and extremely limited therapeutic options, researchers are examining the mechanisms that contribute to the progression of cognitive decline in hopes of developing better therapies and even an effective, long-lasting treatment for this devastating condition. This review will provide an updated perspective on the role of immunity in triggering the changes that lead to the development of dementia. It will detail the latest findings on Aβ- and tau-induced microglial activation, including the role of the inflammasome. The contribution of the adaptive immune system, specifically T cells, will be discussed. Finally, whether the innate and adaptive immune system can be modulated to protect against dementia will be examined, along with an assessment of the prospective candidates for these that are currently in clinical trials.
Collapse
Affiliation(s)
- Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
| |
Collapse
|
48
|
Amarillo D, Brugnini A, Trías N, Rodriguez Sande V, Salisbury S, Cuello M, Lens D. Circulating T regulatory cell subsets in patients with untreated lung cancer. Clin Transl Oncol 2022; 24:1755-1763. [PMID: 35445283 DOI: 10.1007/s12094-022-02827-6] [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: 09/28/2021] [Accepted: 03/12/2022] [Indexed: 12/09/2022]
Abstract
INTRODUCTION Regulatory T Cells (Tregs) play an important role in carcinogenesis and tumor immunoediting by preventing the development of effective antitumor immunity. Several reports showed that circulating Tregs are increased in patients with solid tumors, including lung cancer. Treg population could be categorized into "naive," "effector," and "memory" subtypes, bearing potential unique functions. However, the data regarding the prognostic impact of these Tregs subtypes is limited in lung cancer. The aim of this study was to investigate the frequency of different circulating Tregs subtypes in lung cancer and their correlation with clinical outcomes. METHODS We analyzed the frequency of circulating CD4, CD8 and, Tregs lymphocytes in 66 patients with lung cancer and 32 healthy controls using flow cytometry. Circulating Tregs subtypes: naïve (CD3+ , CD4+ , CCR4+ , CD25+ and CD127low, CD45RO-), memory (CD3+ , CD4+ , CCR4+ , CD25+ and CD127low, CD45RO+) and the expression of the activation marker HLA-DR were correlated with overall survival. RESULTS The percentage and the absolute number of total, memory and activated Tregs was significantly higher in lung cancer patients than healthy controls. Patients with a Tregs percentage higher than 5.4% and higher than 20% of HLA-DR + Tregs had worse overall survival than those with lower levels. CONCLUSIONS Circulating Tregs and activated Tregs are a potential prognostic factor in patients with lung cancer treated with conventional therapy and could be considered a predictive biomarker in patients not eligible for immune blockade treatments. Additionally, it will be interesting to study these Tregs subsets for immune treatments in future clinical trials.
Collapse
Affiliation(s)
- Dahiana Amarillo
- Departamento Básico de Medicina, Hospital de Clínicas Dr. Manuel Quintela, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Servicio de Oncología Médica, Hospital de Clínicas Dr. Manuel Quintela, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Andreina Brugnini
- Departamento Básico de Medicina, Hospital de Clínicas Dr. Manuel Quintela, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Natalia Trías
- Departamento Básico de Medicina, Hospital de Clínicas Dr. Manuel Quintela, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Virginia Rodriguez Sande
- Departamento Básico de Medicina, Hospital de Clínicas Dr. Manuel Quintela, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Siul Salisbury
- Servicio de Cirugía Torácica, Hospital de Clínicas Dr. Manuel Quintela, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mauricio Cuello
- Servicio de Oncología Médica, Hospital de Clínicas Dr. Manuel Quintela, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Daniela Lens
- Departamento Básico de Medicina, Hospital de Clínicas Dr. Manuel Quintela, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
49
|
Creswell R, Dombrowski Y. Innate and adaptive immune mechanisms regulating central nervous system remyelination. Curr Opin Pharmacol 2022; 63:102175. [DOI: 10.1016/j.coph.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 11/03/2022]
|
50
|
Perdaens O, van Pesch V. Molecular Mechanisms of Immunosenescene and Inflammaging: Relevance to the Immunopathogenesis and Treatment of Multiple Sclerosis. Front Neurol 2022; 12:811518. [PMID: 35281989 PMCID: PMC8913495 DOI: 10.3389/fneur.2021.811518] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/27/2021] [Indexed: 12/18/2022] Open
Abstract
Aging is characterized, amongst other features, by a complex process of cellular senescence involving both innate and adaptive immunity, called immunosenescence and associated to inflammaging, a low-grade chronic inflammation. Both processes fuel each other and partially explain increasing incidence of cancers, infections, age-related autoimmunity, and vascular disease as well as a reduced response to vaccination. Multiple sclerosis (MS) is a lifelong disease, for which considerable progress in disease-modifying therapies (DMTs) and management has improved long-term survival. However, disability progression, increasing with age and disease duration, remains. Neurologists are now involved in caring for elderly MS patients, with increasing comorbidities. Aging of the immune system therefore has relevant implications for MS pathogenesis, response to DMTs and the risks mediated by these treatments. We propose to review current evidence regarding markers and molecular mechanisms of immunosenescence and their relevance to understanding MS pathogenesis. We will focus on age-related changes in the innate and adaptive immune system in MS and other auto-immune diseases, such as systemic lupus erythematosus and rheumatoid arthritis. The consequences of these immune changes on MS pathology, in interaction with the intrinsic aging process of central nervous system resident cells will be discussed. Finally, the impact of immunosenescence on disease evolution and on the safety and efficacy of current DMTs will be presented.
Collapse
Affiliation(s)
- Océane Perdaens
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Vincent van Pesch
- Laboratory of Neurochemistry, Institute of Neuroscience, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Neurology, Cliniques universitaires Saint-Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- *Correspondence: Vincent van Pesch
| |
Collapse
|