1
|
Chen H, Fang Z, Lin SL, Schachner M. L1CAM mimetic compound duloxetine improves cognitive impairment in 5xFAD mice and protects Aβ1-42-damaged HT22 cells. Eur J Pharmacol 2025; 997:177476. [PMID: 40057157 DOI: 10.1016/j.ejphar.2025.177476] [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: 08/26/2024] [Revised: 01/08/2025] [Accepted: 03/04/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Synapse loss and damage are underlying causes of Alzheimer's disease. Duloxetine has been identified as a mimetic of neural adhesion molecule L1CAM, a neuronal synapse component, suggesting duloxetine could be therapeutic for Alzheimer's disease. METHODS Cognitive function in 5xFAD mice was evaluated by open field, novel object recognition, and Morris water maze tests. Hippocampal and cortical Aβ1-40, Aβ1-42 and amyloid plaque deposition were quantified by ELISA and immunohistochemistry. RT-qPCR and western blotting quantified the effects of duloxetine treatment on L1CAM levels and PI3K/Akt/CREB signaling pathway activation. Apoptosis markers Bcl-2 and Bax were also measured by RT-qPCR and western blotting. HT22 cell survival was measured by CCK8 assay. RESULTS Duloxetine preserved learning and memory abilities, but had no effect on locomotor performance of 5xFAD mice. Duloxetine decreased Aβ1-42 expression levels, increased Aβ1-40 levels, reduced amyloid plaque formation, and activated the PI3K/Akt/CREB signaling pathway in both cortices and hippocampi of 5xFAD mice. Moreover, duloxetine increased the expression of L1CAM and Bcl-2, and inhibited the expression of Bax, as well as prevented Aβ1-42 cytotoxicity in wild-type, but not L1CAM-knockdown HT22 cells, suggesting a feed-forward mechanism for duloxetine-mediated neuroprotection, whereby duloxetine induces and activates L1CAM to exert neuroprotective effects. CONCLUSIONS Our findings demonstrate that duloxetine plays a neuroprotective role in 5xFAD mice and HT22 cells through activating L1CAM, likely by regulating the PI3K/Akt/CREB signaling pathway. These results suggest that duloxetine may be a potential reagent for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Hanyu Chen
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Zhou Fang
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Stanley Li Lin
- Shantou Key Laboratory of Precision Diagnosis and Treatment of Women's Cancer, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, China; Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou, Guangdong, China.
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, 515041, China; Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers the State University of New Jersey, Piscataway, NJ, 08554, USA.
| |
Collapse
|
2
|
Zhu B, Feng J, Liang X, Fu Z, Liao M, Deng T, Wang K, Xie J, Chi J, Yang L, Gao Y, Nie K, Wang L, Zhang P, Zhang Y. TREM2 deficiency exacerbates cognitive impairment by aggravating α-Synuclein-induced lysosomal dysfunction in Parkinson's disease. Cell Death Discov 2025; 11:243. [PMID: 40393958 DOI: 10.1038/s41420-025-02538-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/27/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025] Open
Abstract
Cognitive impairment in Parkinson's disease (PD) is a widespread and rapidly progressive feature that impacts prognosis. Although TREM2 has been implicated in neuroprotection across various neurodegenerative diseases, its specific role in PD remains to be clarified. In this study, we first detected the hippocampus of human PD specimens and of the mutant A53T α-Synuclein transgenic mice (A53T mice), and found a significant increase in the number of TREM2+ microglia. To evaluate the effects of TREM2 deficiency, TREM2-deficient A53T mice (TREM2-/-/A53T mice) were generated. In these mice, exacerbated cognitive impairment, neurodegeneration, disruption of synaptic plasticity, and accumulation of pathological α-Synuclein (α-Syn) in the hippocampus were observed, without any detected motor dysfunction. Despite increased infiltration of activated microglia surrounding α-Syn aggregates, lysosomal dysfunction in microglia was aggravated in the TREM2-/-/A53T mice. In addition, transcriptional analyses and in vitro experiments further found that TREM2 knockdown inhibited the nuclear distribution of TFEB via the ERK1/2 pathway, exacerbating α-Syn-induced lysosomal dysfunction and causing more pathological α-Syn accumulation. Finally, HT22 cells were cocultured with TREM2 knockdown of BV-2 cells pretreated with recombinant human A53T α-Syn preformed fibrils (PFFs). The coculture experiments showed that TREM2 knockdown in BV-2 cells pretreated with PFFs enhanced the phosphorylation of α-Syn and promoted apoptosis in HT22 cells via inhibiting α-Syn degradation. In conclusion, TREM2 deficiency exacerbates cognitive impairment in PD by exacerbating α-Syn-induced microglial lysosomal dysfunction, identifying TREM2 as a potential therapeutic target.
Collapse
Affiliation(s)
- Baoyu Zhu
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Jiezhu Feng
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Xiaomei Liang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Zhongling Fu
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Mengshi Liao
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Tongtong Deng
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Kaicheng Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Jianwei Xie
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
- Department of Neurology, Longyan First Hospital, Fujian Province, China
| | - Jieshan Chi
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Lu Yang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Yuyuan Gao
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Kun Nie
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Lijuan Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Piao Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China.
| | - Yuhu Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China.
- Guangzhou Key Laboratory of Diagnosis and Treatment for Neurodegenerative Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China.
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China.
| |
Collapse
|
3
|
Villacampa N, Sarlus H, Martorell P, Bhalla K, Gomez-Castro S, Vieira-Saecker A, Slutzkin I, Händler K, Venegas C, McManus R, Ulas T, Beyer M, Segal E, Heneka MT. Proliferating Microglia Exhibit Unique Transcriptional and Functional Alterations in Alzheimer's Disease. ASN Neuro 2025; 17:2506406. [PMID: 40387894 DOI: 10.1080/17590914.2025.2506406] [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: 11/25/2024] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025] Open
Abstract
Proliferation of microglia represents a physiological process, which is accelerated in several neurodegenerative disorders including Alzheimer disease (AD). The effect of such neurodegeneration-associated microglial proliferation on function and disease progression remains unclear. Here, we show that proliferation results in profound alterations of cellular function by providing evidence that newly proliferated microglia show impaired beta-amyloid clearance in vivo. Through sorting of proliferating microglia of APP/PS1 mice and subsequent transcriptome analysis, we define unique proliferation-associated transcriptomic signatures that change with age and beta-amyloid accumulation and are characterized by enrichment of immune system-related pathways. Of note, we identify the DEAD-Box Helicase 3 X-Linked (DDX3X) as a key molecule to modulate microglia activation and cytokine secretion and it is expressed in the AD brain. Together, these results argue for a novel concept by which phenotypic and functional microglial changes occur longitudinally as a response to accelerated proliferation in a neurodegenerative environment.
Collapse
Affiliation(s)
- Nàdia Villacampa
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Heela Sarlus
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Paula Martorell
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Khushbu Bhalla
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
| | - Sergio Gomez-Castro
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Ana Vieira-Saecker
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Ilya Slutzkin
- Department of Computer Science and Applied Mathematic, Weizmann Institute of Science, Rehovot, Israel
| | - Kristian Händler
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Carmen Venegas
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Róisín McManus
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Thomas Ulas
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Marc Beyer
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Eran Segal
- Department of Computer Science and Applied Mathematic, Weizmann Institute of Science, Rehovot, Israel
| | - Michael T Heneka
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
4
|
Aathira NS, Kaur A, Kumar A, Dar GM, Nimisha, Sharma AK, Bera P, Mahajan B, Chatterjee A, Saluja SS. The genetic risk factors, molecular pathways, microRNAs, and the gut microbiome in Alzheimer's disease. Neuroscience 2025:S0306-4522(25)00373-2. [PMID: 40374065 DOI: 10.1016/j.neuroscience.2025.05.021] [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: 11/23/2024] [Revised: 04/25/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia worldwide. It is a multifaceted condition resulting from interplay of genetic mutations (e.g., APP, PSEN1, PSEN2) that account for less than 5% of cases, several genetic risk variants such as APOE4, TREM2, CD33, CLU, SORL1, and CR1 contribute to disease susceptibility and epigenetic factors, which may mediate the influence of environmental and lifestyle factors over time. Other critical contributors such as aging, protein misfolding and aggregation (amyloid-β and tau), molecular and transcriptomic dysregulation affecting neuronal function, and modifiable lifestyle factors like diet, physical activity, and environmental exposures presents challenges in accurate diagnosis and management. Research has predominantly focused on the diverse molecular pathways in the pathogenesis of AD, with particular attention given to the amyloidogenic pathways, tau pathology, calcium signalling, endolysosomal pathways, and others, whether they are directly or indirectly involved. Apart from these known molecular pathways, miRNAs are gaining attention as important regulators, which have been implicated in moderating the expression of mRNA targets involved in various processes associated with the clearance of pathogenic β-amyloid proteins. A mounting body of research suggests the possible role of gut microbiota in AD which regulates inflammation, neurotransmitters, and the blood-brain barrier. Gut dysbiosis can trigger neuroinflammation and amyloid-beta aggregation, making microbiome composition a potential early AD biomarker. This review aims to explore briefly the diverse risk encompassing genetic polymorphisms, altered molecular pathways implicated in AD pathogenesis, miRNA regulatory mechanisms, and the potential impact of gut microbiota on AD risk.
Collapse
Affiliation(s)
- N S Aathira
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Amanpreet Kaur
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Arun Kumar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Ghulam Mehdi Dar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Nimisha
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Abhay Kumar Sharma
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Pinki Bera
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Bhawna Mahajan
- Department of Biochemistry, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Atri Chatterjee
- Department of Neurology, VMMC and Safdarjung Hospital, New Delhi, India.
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of GI Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India.
| |
Collapse
|
5
|
Geraghty AC, Acosta-Alvarez L, Rotiroti MC, Dutton S, O'Dea MR, Kim W, Trivedi V, Mancusi R, Shamardani K, Malacon K, Woo PJ, Martinez-Velez N, Pham T, Reche-Ley NN, Otubu G, Castenada EH, Nwangwu K, Xu H, Mulinyawe SB, Zamler DB, Ni L, Cross K, Rustenhoven J, Kipnis J, Liddelow SA, Mackall CL, Majzner RG, Monje M. Immunotherapy-related cognitive impairment after CAR T cell therapy in mice. Cell 2025:S0092-8674(25)00391-5. [PMID: 40359942 DOI: 10.1016/j.cell.2025.03.041] [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: 01/25/2023] [Revised: 01/06/2025] [Accepted: 03/25/2025] [Indexed: 05/15/2025]
Abstract
Immunotherapies have revolutionized cancer care for many tumor types, but their potential long-term cognitive impacts are incompletely understood. Here, we demonstrated in mouse models that chimeric antigen receptor (CAR) T cell therapy for both central nervous system (CNS) and non-CNS cancers impaired cognitive function and induced a persistent CNS immune response characterized by white matter microglial reactivity, microglial chemokine expression, and elevated cerebrospinal fluid (CSF) cytokines and chemokines. Consequently, oligodendroglial homeostasis and hippocampal neurogenesis were disrupted. Single-nucleus sequencing studies of human frontal lobe from patients with or without previous CAR T cell therapy for brainstem tumors confirmed reactive states of microglia and oligodendrocytes following treatment. In mice, transient microglial depletion or CCR3 chemokine receptor blockade rescued oligodendroglial deficits and cognitive performance in a behavioral test of attention and short-term memory function following CAR T cell therapy. Taken together, these findings illustrate targetable neural-immune mechanisms underlying immunotherapy-related cognitive impairment.
Collapse
Affiliation(s)
- Anna C Geraghty
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Lehi Acosta-Alvarez
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Maria C Rotiroti
- Department of Pediatrics, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Selena Dutton
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michael R O'Dea
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Wonju Kim
- Department of Pediatrics, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Vrunda Trivedi
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Rebecca Mancusi
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Kiarash Shamardani
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Karen Malacon
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Pamelyn J Woo
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | | | - Theresa Pham
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Noemi N Reche-Ley
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Gabriel Otubu
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Enrique H Castenada
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Kamsi Nwangwu
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Haojun Xu
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Sara B Mulinyawe
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Daniel B Zamler
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Lijun Ni
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Kevin Cross
- Brain immunology and Glia (BIG) Center and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Justin Rustenhoven
- Brain immunology and Glia (BIG) Center and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Jonathan Kipnis
- Brain immunology and Glia (BIG) Center and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY 10016, USA; Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Crystal L Mackall
- Department of Pediatrics, Stanford School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cellular Therapy, Stanford School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robbie G Majzner
- Department of Pediatrics, Stanford School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cellular Therapy, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michelle Monje
- Department of Neurology and Neurosciences, Stanford School of Medicine, Stanford, CA 94305, USA; Department of Pediatrics, Stanford School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cellular Therapy, Stanford School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
6
|
Ye K, Zhao X, Liu L, Ge F, Zheng F, Liu Z, Tian M, Han X, Gao X, Xia Q, Wang D. Comparative Analysis of Human Brain RNA-seq Reveals the Combined Effects of Ferroptosis and Autophagy on Alzheimer's Disease in Multiple Brain Regions. Mol Neurobiol 2025; 62:6128-6149. [PMID: 39710824 DOI: 10.1007/s12035-024-04642-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024]
Abstract
Ferroptosis and autophagy are closely associated with Alzheimer's disease (AD). Elevated ferric ion levels can induce oxidative stress and chronic inflammatory responses, resulting in brain tissue damage and further neurological cell damage. Autophagy in Alzheimer's has a dual role. On one hand, it protects neurons by removing β-amyloid and cellular damage products caused by oxidative stress and inflammation. On the other hand, abnormal autophagy is linked to neuronal apoptosis and neurodegeneration. However, the intricate interplay between ferroptosis and autophagy in AD remains insufficiently explored. This study focuses on the roles of ferroptosis and autophagy in AD and their interconnection through bioinformatics analysis, shedding light on the disease. Ferroptosis and autophagy significantly correlate with the development and course of AD. Using PPI network analysis and unsupervised consistency clustering analysis, we uncovered a complex network of interactions between ferroptosis and autophagy during disease progression, demonstrating a significant congruence in their modification patterns. Functional analyses further demonstrated that ferroptosis and autophagy together affect the immunological status and synaptic regulation in hippocampal regions in patients with AD, which significantly impacts the start and progression of the disease.
Collapse
Affiliation(s)
- Ke Ye
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Xue Zhao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Lulu Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Fangliang Ge
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Feifei Zheng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Zijie Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Mengjie Tian
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Xinyu Han
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China.
- Key Laboratory of Heilongjiang Province for Genetically Modified Animals, Harbin Medical University, Harbin, 150000, Heilongjiang, China.
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150000, Heilongjiang, China.
| | - Qing Xia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Dayong Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, 150000, Heilongjiang, China.
| |
Collapse
|
7
|
Kimura K, Subramanian A, Yin Z, Khalilnezhad A, Wu Y, He D, Dixon KO, Chitta UK, Ding X, Adhikari N, Guzchenko I, Zhang X, Tang R, Pertel T, Myers SA, Aastha A, Nomura M, Eskandari-Sedighi G, Singh V, Liu L, Lambden C, Kleemann KL, Gupta N, Barry JL, Durao A, Cheng Y, Silveira S, Zhang H, Suhail A, Delorey T, Rozenblatt-Rosen O, Freeman GJ, Selkoe DJ, Weiner HL, Blurton-Jones M, Cruchaga C, Regev A, Suvà ML, Butovsky O, Kuchroo VK. Immune checkpoint TIM-3 regulates microglia and Alzheimer's disease. Nature 2025; 641:718-731. [PMID: 40205047 PMCID: PMC12079183 DOI: 10.1038/s41586-025-08852-z] [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: 01/29/2023] [Accepted: 02/28/2025] [Indexed: 04/11/2025]
Abstract
Microglia are the resident immune cells in the brain and have pivotal roles in neurodevelopment and neuroinflammation1,2. This study investigates the function of the immune-checkpoint molecule TIM-3 (encoded by HAVCR2) in microglia. TIM-3 was recently identified as a genetic risk factor for late-onset Alzheimer's disease3, and it can induce T cell exhaustion4. However, its specific function in brain microglia remains unclear. We demonstrate in mouse models that TGFβ signalling induces TIM-3 expression in microglia. In turn, TIM-3 interacts with SMAD2 and TGFBR2 through its carboxy-terminal tail, which enhances TGFβ signalling by promoting TGFBR-mediated SMAD2 phosphorylation, and this process maintains microglial homeostasis. Genetic deletion of Havcr2 in microglia leads to increased phagocytic activity and a gene-expression profile consistent with the neurodegenerative microglial phenotype (MGnD), also referred to as disease-associated microglia (DAM). Furthermore, microglia-targeted deletion of Havcr2 ameliorates cognitive impairment and reduces amyloid-β pathology in 5×FAD mice (a transgenic model of Alzheimer's disease). Single-nucleus RNA sequencing revealed a subpopulation of MGnD microglia in Havcr2-deficient 5×FAD mice characterized by increased pro-phagocytic and anti-inflammatory gene expression alongside reduced pro-inflammatory gene expression. These transcriptomic changes were corroborated by single-cell RNA sequencing data across most microglial clusters in Havcr2-deficient 5×FAD mice. Our findings reveal that TIM-3 mediates microglia homeostasis through TGFβ signalling and highlight the therapeutic potential of targeting microglial TIM-3 in Alzheimer's disease.
Collapse
Affiliation(s)
- Kimitoshi Kimura
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ayshwarya Subramanian
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zhuoran Yin
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Ahad Khalilnezhad
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yufan Wu
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Danyang He
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Karen O Dixon
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Udbhav Kasyap Chitta
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xiaokai Ding
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Niraj Adhikari
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Isabell Guzchenko
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xiaoming Zhang
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ruihan Tang
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas Pertel
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Samuel A Myers
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, San Diego, CA, USA
| | - Aastha Aastha
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Masashi Nomura
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ghazaleh Eskandari-Sedighi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | | | - Lei Liu
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Conner Lambden
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kilian L Kleemann
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Neha Gupta
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jen-Li Barry
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana Durao
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yiran Cheng
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sebastian Silveira
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Huiyuan Zhang
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Aamir Suhail
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Toni Delorey
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Howard L Weiner
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Mario L Suvà
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Oleg Butovsky
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Vijay K Kuchroo
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
8
|
Peralta Ramos JM, Castellani G, Kviatcovsky D, Croese T, Tsitsou-Kampeli A, Burgaletto C, Abellanas MA, Cahalon L, Phoebeluc Colaiuta S, Salame TM, Kuperman Y, Savidor A, Itkin M, Malitsky S, Ovadia S, Ferrera S, Kalfon L, Kadmani S, Samra N, Paz R, Rokach L, Furlan R, Aharon-Peretz J, Falik-Zaccai TC, Schwartz M. Targeting CD38 immunometabolic checkpoint improves metabolic fitness and cognition in a mouse model of Alzheimer's disease. Nat Commun 2025; 16:3736. [PMID: 40254603 PMCID: PMC12009998 DOI: 10.1038/s41467-025-58494-y] [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: 01/23/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Protective immunity, essential for brain maintenance and repair, may be compromised in Alzheimer's disease (AD). Here, using high-dimensional single-cell mass cytometry, we find a unique immunometabolic signature in circulating CD4+ T cells preceding symptom onset in individuals with familial AD, featured by the elevation of CD38 expression. Using female 5xFAD mice, a mouse model of AD, we show that treatment with an antibody directed to CD38 leads to restored metabolic fitness, improved cognitive performance, and attenuated local neuroinflammation. Comprehensive profiling across distinct immunological niches in 5xFAD mice, reveals a high level of disease-associated CD4+ T cells that produce IL-17A in the dural meninges, previously linked to cognitive decline. Targeting CD38 leads to abrogation of meningeal TH17 immunity and cortical IL-1β, breaking the negative feedback loop between these two compartments. Taken together, the present findings suggest CD38 as an immunometabolic checkpoint that could be adopted as a pre-symptomatic biomarker for early diagnosis of AD, and might also be therapeutically targeted alone or in combination with other immunotherapies for disease modification.
Collapse
Affiliation(s)
| | - Giulia Castellani
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | - Liora Cahalon
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Tomer-Meir Salame
- Department Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- The De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Maxim Itkin
- Department Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Sergey Malitsky
- Department Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Sharon Ovadia
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | | | - Limor Kalfon
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
| | - Shiran Kadmani
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
| | - Nadra Samra
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Rotem Paz
- Cognitive Neurology Institute, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Lior Rokach
- Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Judith Aharon-Peretz
- Cognitive Neurology Institute, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Tzipora C Falik-Zaccai
- Institute of Human Genetics, Galilee Medical Center, Nahariya, Israel
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
9
|
Wang X, Wang Y, Yang L, Zhang Y, Yang L. TREM2 + macrophages: a key role in disease development. Front Immunol 2025; 16:1550893. [PMID: 40242752 PMCID: PMC12000036 DOI: 10.3389/fimmu.2025.1550893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Triggering receptors expressed on myeloid cells 2 (TREM2), an immune receptor expressed on myeloid cells, has garnered considerable attention in recent years due to its role in unique signaling pathways and diverse biological functions, including phagocytosis, lipid metabolism, cell survival, and inflammatory responses. Although TREM2 is expressed in various cell types, such as macrophages, dendritic cells (DCs), osteoclasts, and others, where it exhibits context-dependent functional characteristics, it is mainly expressed in macrophages. Notably, TREM2 is implicated in the development and progression of multiple diseases, playing dual and often opposing roles in noncancerous diseases and cancers. This review aims to highlight the pivotal role of TREM2 in macrophages and immune-related diseases, elucidate its underlying mechanisms of action, explore its potential as a clinical diagnostic and prognostic marker, and propose therapeutic strategies targeting TREM2 based on current clinical trial data, providing comprehensive guidance and references for clinical practice.
Collapse
Affiliation(s)
- Xinxin Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunhan Wang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Yang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Biotherapy Center and Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, China
| |
Collapse
|
10
|
Xavier C, Pinto N. Navigating the blurred boundary: Neuropathologic changes versus clinical symptoms in Alzheimer's disease, and its consequences for research in genetics. J Alzheimers Dis 2025; 104:611-626. [PMID: 39956949 DOI: 10.1177/13872877251317543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
During decades scientists tried to unveil the genetic architecture of Alzheimer's disease (AD), recurring to increasingly larger sample numbers for genome-wide association studies (GWAS) in hope for higher statistical gains. Here, a retrospective look on the most prominent GWAS was performed, focusing on the quality of the diagnosis associated with the used data and databases. Different methods for AD diagnosis (or absence) carry different levels of accuracy and certainty applied to both subsets of cases and controls. Furthermore, the different phenotypes included in these databases were explored, as several incorporate other ageing comorbidities and might be encompassing many confounding agents as well. Age of the samples' donors and origin populations were also investigated as these could be biasing factors in posterior analyses. A tendency for looser diagnostic methods in more recent GWAS was observed, where greater datasets of individuals are analyzed, which may have been hampering the discovery of associated genetic variants. Specifically for AD, a diagnostic method conveying a clinical outcome may be distinct from the disease neuropathological assessment, since the first has a practical perspective that not necessarily needs a confirmation. Due to its properties and complex diagnosis, this work highlights the importance of the neuropathological confirmation of AD (or its absence) in the subjects considered for research purposes to avoid reaching statistically weak and/or misleading conclusions that may trigger further studies with powerless groundwork.
Collapse
Affiliation(s)
- Catarina Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Nádia Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- CMUP - Centro de Matemática da Universidade do Porto, Porto, Portugal
| |
Collapse
|
11
|
Ma X, Ding L, Li S, Fan Y, Wang X, Han Y, Yuan H, Sun L, He Q, Liu M. Druggable genome-wide Mendelian randomization identifies therapeutic targets for metabolic dysfunction-associated steatotic liver disease. Lipids Health Dis 2025; 24:113. [PMID: 40140823 PMCID: PMC11938603 DOI: 10.1186/s12944-025-02515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) affects > 25% of the global population, potentially leading to severe hepatic and extrahepatic complications, including metabolic dysfunction-associated steatohepatitis. Given that the pathophysiology of MASLD is incompletely understood, identifying therapeutic targets and optimizing treatment strategies are crucial for addressing this severe condition. METHODS Mendelian randomization (MR) analysis was conducted using two genome-wide association study datasets: a European meta-analysis (8,434 cases; 770,180 controls) and an additional study (3,954 cases; 355,942 controls), identifying therapeutic targets for MASLD. Of 4302 drug-target genes, 2,664 genetic instrument variables were derived from cis-expression quantitative trait loci (cis-eQTLs). Colocalization analyses assessed shared causal variants between MASLD-associated single nucleotide polymorphisms and eQTLs. Using the drug target gene cis-eQTL of liver tissue from the genotype-tissue expression project, we performed MR and summary MR to validate the significance of the gene results of the blood eQTL MR. RNA-sequencing data from liver biopsies were validated using immunohistochemistry and quantitative polymerase chain reaction (qPCR) tests to confirm gene expression findings. RESULT MR analysis across both datasets identified significant MR associations between MASLD and two drug targets-milk fat globule-EGF factor 8 (MFGE8) (odds ratio [OR] 0.89, 95% confidence interval [CI] 0.85-0.94; P = 2.15 × 10-6) and cluster of differentiation 33 (CD33) (OR 1.17, 95% CI 1.10-1.25; P = 1.39 × 10-6). Both targets exhibited strong colocalization with MASLD. Genetic manipulation indicating MFGE8 activation and CD33 inhibition did not increase the risk for other metabolic disorders. RNA-sequencing, qPCR, and immunohistochemistry validation demonstrated consistent differential expressions of MFGE8 and CD33 in MASLD. CONCLUSION CD33 inhibition can reduce MASLD risk, while MFGE8 activation may offer therapeutic benefits for MASLD treatment.
Collapse
Affiliation(s)
- Xiaohui Ma
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Department of Endocrinology and Metabolism, Baotou Central Hospital, Baotou, China
| | - Li Ding
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Shuo Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yu Fan
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xin Wang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yitong Han
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Hengjie Yuan
- Department of Pharmacy, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Longhao Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| |
Collapse
|
12
|
Chen K, Li F, Zhang S, Chen Y, Ikezu TC, Li Z, Martens YA, Qiao W, Meneses A, Zhu Y, Xhafkollari G, Bu G, Zhao N. Enhancing TREM2 expression activates microglia and modestly mitigates tau pathology and neurodegeneration. J Neuroinflammation 2025; 22:93. [PMID: 40122810 PMCID: PMC11931752 DOI: 10.1186/s12974-025-03420-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
TREM2, a microglia-specific receptor, is strongly associated with Alzheimer's disease (AD) risk, mediating microglial responses to amyloid pathology critical to AD development. However, its role in tau pathology and neurodegeneration remains unclear. Using the PS19 tauopathy mouse model with inducible overexpression of human wild-type TREM2 (TREM2-WT) or the R47H variant (TREM2-R47H), we show that increasing TREM2-WT expression modestly reduces soluble phosphorylated tau levels and mildly preserves neuronal integrity. Single-cell RNA sequencing reveals that TREM2-WT robustly enhances microglial activation, characterized by a disease-associated microglia (DAM) signature. In contrast, TREM2-R47H overexpression exhibits a loss-of-function phenotype, with no significant impact on tau levels, neurodegeneration, or microglial activation. These findings highlight the role of TREM2 in modulating microglial activity and its influence on tau pathology and neurodegeneration, providing important insights for the future development of therapies targeting TREM2 or microglial pathways in AD or other tauopathies.
Collapse
Affiliation(s)
- Kai Chen
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Fuyao Li
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Shuwen Zhang
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Tadafumi C Ikezu
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Zonghua Li
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Axel Meneses
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Clinical and Translational Science Graduate Program, Mayo Clinic, Jacksonville, FL, USA
| | - Yiyang Zhu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Gisela Xhafkollari
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
- Clinical and Translational Science Graduate Program, Mayo Clinic, Jacksonville, FL, USA.
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
13
|
McKinsey GL, Santander N, Zhang X, Kleemann KL, Tran L, Katewa A, Conant K, Barraza M, Waddell K, Lizama CO, La Russa M, Koo JH, Lee H, Mukherjee D, Paidassi H, Anton ES, Atabai K, Sheppard D, Butovsky O, Arnold TD. Radial glia integrin avb8 regulates cell autonomous microglial TGFβ1 signaling that is necessary for microglial identity. Nat Commun 2025; 16:2840. [PMID: 40121230 PMCID: PMC11929771 DOI: 10.1038/s41467-025-57684-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Microglial diversity arises from the interplay between inherent genetic programs and external environmental signals. However, the mechanisms by which these processes develop and interact within the growing brain are not yet fully understood. Here, we show that radial glia-expressed integrin beta 8 (ITGB8) activates microglia-expressed TGFβ1 to drive microglial development. Domain-restricted deletion of Itgb8 in these progenitors results in regionally restricted and developmentally arrested microglia that persist into adulthood. In the absence of autocrine TGFβ1 signaling, microglia adopt a similar phenotype, leading to neuromotor symptoms almost identical to Itgb8 mutant mice. In contrast, microglia lacking the canonical TGFβ signal transducers Smad2 and Smad3 have a less polarized dysmature phenotype and correspondingly less severe neuromotor dysfunction. Our study describes the spatio-temporal regulation of TGFβ activation and signaling in the brain necessary to promote microglial development, and provides evidence for the adoption of microglial developmental signaling pathways in brain injury or disease.
Collapse
Affiliation(s)
- Gabriel L McKinsey
- University of California San Francisco, Department of Pediatrics and Newborn Brain Research Institute, San Francisco, CA, USA.
| | - Nicolas Santander
- Instituto de Ciencias de la Salud, Universidad de O´Higgins, Rancagua, Chile
| | - Xiaoming Zhang
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Kilian L Kleemann
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lauren Tran
- University of California San Francisco, Department of Pediatrics and Newborn Brain Research Institute, San Francisco, CA, USA
| | - Aditya Katewa
- University of California San Francisco, Department of Pediatrics and Newborn Brain Research Institute, San Francisco, CA, USA
| | - Kaylynn Conant
- University of California San Francisco, Department of Pediatrics and Newborn Brain Research Institute, San Francisco, CA, USA
| | - Matthew Barraza
- Northwestern University, Department of Neuroscience, Chicago, IL, USA
| | - Kian Waddell
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Carlos O Lizama
- University of California San Francisco, Cardiovascular Research Institute, San Francisco, CA, USA
| | - Marie La Russa
- Stanford University, Department of Bioengineering, Stanford, CA, USA
| | - Ji Hyun Koo
- University of California San Francisco, Department of Pediatrics and Newborn Brain Research Institute, San Francisco, CA, USA
| | - Hyunji Lee
- University of California San Francisco, Department of Pediatrics and Newborn Brain Research Institute, San Francisco, CA, USA
| | - Dibyanti Mukherjee
- University of California San Francisco, Department of Pediatrics and Newborn Brain Research Institute, San Francisco, CA, USA
| | - Helena Paidassi
- CIRI Centre International de Recherche en Infectiologie, Univ Lyon Inserm U1111 Université Claude Bernard Lyon 1 CNRS UMR5308 ENS de Lyon, F-69007, Lyon, France
| | - E S Anton
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kamran Atabai
- University of California San Francisco, Cardiovascular Research Institute, San Francisco, CA, USA
| | - Dean Sheppard
- University of California San Francisco, Cardiovascular Research Institute, San Francisco, CA, USA
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas D Arnold
- University of California San Francisco, Department of Pediatrics and Newborn Brain Research Institute, San Francisco, CA, USA.
| |
Collapse
|
14
|
Li W, Yong-Yan X, Jia-Xin M, Shu-Chao G, Li-Ping H. Senescent microglia: The hidden culprits accelerating Alzheimer's disease. Brain Res 2025; 1851:149480. [PMID: 39884491 DOI: 10.1016/j.brainres.2025.149480] [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: 10/03/2024] [Revised: 12/07/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Ageing is a major risk factor for neurodegenerative diseases like Alzheimer's disease (AD). Microglia, as the principal innate immune cells within the brain, exert homeostatic and active immunological defense functions throughout human lifespan. The age-related dysfunction of microglia is currently recognized as a pivotal trigger for brain diseases associated with aging. In AD, microglia exhibit alterations in gene expression, cellular morphology, and functional behavior. By focusing on the immunomodulatory functions of factors secreted by senescent microglia, such as cytokines, chemokines, complement factors, and reactive oxygen species (ROS), we explore the diverse detrimental effects of microglia in aging and AD pathogenesis, including Aβ accumulation, Tau deposition, synaptic dysfunction, and neuroinflammation. These collectively contribute to hastening the progression of. In this review, we highlight the key role of senescent microglia in the pathological processes of AD. Then we propose that targeting senescent microglia holds great promise for therapeutic interventions in neurodegenerative diseases.
Collapse
Affiliation(s)
- Wu Li
- School of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, NanChang, China
| | - Xie Yong-Yan
- School of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, NanChang, China
| | - Mu Jia-Xin
- School of Pharmacy, Jiangxi University of Chinese Medicine, NanChang, China
| | - Ge Shu-Chao
- School of Pharmacy, Jiangxi University of Chinese Medicine, NanChang, China.
| | - Huang Li-Ping
- Jiangxi Provincial Key Laboratory of Pharmacology of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, NanChang, China; School of Pharmacy, Jiangxi University of Chinese Medicine, NanChang, China.
| |
Collapse
|
15
|
Sălcudean A, Popovici RA, Pitic DE, Sârbu D, Boroghina A, Jomaa M, Salehi MA, Kher AAM, Lica MM, Bodo CR, Enatescu VR. Unraveling the Complex Interplay Between Neuroinflammation and Depression: A Comprehensive Review. Int J Mol Sci 2025; 26:1645. [PMID: 40004109 PMCID: PMC11855341 DOI: 10.3390/ijms26041645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
The relationship between neuroinflammation and depression is a complex area of research that has garnered significant attention in recent years. Neuroinflammation, characterized by the activation of glial cells and the release of pro-inflammatory cytokines, has been implicated in the pathophysiology of depression. The relationship between neuroinflammation and depression is bidirectional; not only can inflammation contribute to the onset of depressive symptoms, but depression itself can also exacerbate inflammatory responses, creating a vicious cycle that complicates treatment and recovery. The present comprehensive review aimed to explore the current findings on the interplay between neuroinflammation and depression, as well as the mechanisms, risk factors, and therapeutic implications. The mechanisms by which neuroinflammation induces depressive-like behaviors are diverse. Neuroinflammation can increase pro-inflammatory cytokines, activate the hypothalamus-pituitary-adrenal (HPA) axis, and impair serotonin synthesis, all of which contribute to depressive symptoms. Furthermore, the activation of microglia has been linked to the release of inflammatory mediators that can disrupt neuronal function and contribute to mood disorders. Stress-induced neuroinflammatory responses can lead to the release of pro-inflammatory cytokines that not only affect brain function but also influence behavior and mood. Understanding these mechanisms is crucial for developing targeted therapies that can mitigate the effects of neuroinflammation on mood disorders.
Collapse
Affiliation(s)
- Andreea Sălcudean
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.S.); (M.M.L.); (C.R.B.)
| | - Ramona-Amina Popovici
- Department of Management and Communication in Dental Medicine, Department I, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania;
| | - Dana Emanuela Pitic
- Department of Management and Communication in Dental Medicine, Department I, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania;
| | - Diana Sârbu
- Doctoral School of Pharmacy, Victor Babes University of Medicine and Pharmacy of Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Adela Boroghina
- Doctoral School of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania; (A.B.); (M.J.); (M.A.S.); (A.A.M.K.)
| | - Mohammad Jomaa
- Doctoral School of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania; (A.B.); (M.J.); (M.A.S.); (A.A.M.K.)
| | - Matin Asad Salehi
- Doctoral School of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania; (A.B.); (M.J.); (M.A.S.); (A.A.M.K.)
| | - Alsayed Ahmad Mhd Kher
- Doctoral School of Dental Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 9 Revolutiei 1989 Bv., 300070 Timisoara, Romania; (A.B.); (M.J.); (M.A.S.); (A.A.M.K.)
| | - Maria Melania Lica
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.S.); (M.M.L.); (C.R.B.)
| | - Cristina Raluca Bodo
- Department of Ethics and Social Sciences, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.S.); (M.M.L.); (C.R.B.)
| | - Virgil Radu Enatescu
- Department of Psychiatry, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy of Timisoara, 300041 Timisoara, Romania;
| |
Collapse
|
16
|
Abdelhamed HG, Hassan AA, Sakraan AA, Al-Deeb RT, Mousa DM, Aboul Ezz HS, Noor NA, Khadrawy YA, Radwan NM. Brain interleukins and Alzheimer's disease. Metab Brain Dis 2025; 40:116. [PMID: 39891777 PMCID: PMC11787210 DOI: 10.1007/s11011-025-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
The central nervous system (CNS) is immune-privileged by several immuno-modulators as interleukins (ILs). ILs are cytokines secreted by immune cells for cell-cell signaling communications and affect the functions of the CNS. ILs were reported to orchestrate different molecular and cellular mechanisms of both physiological and pathological events, through overproduction or over-expression of their receptors. They interact with numerous receptors mediating pro-inflammatory and/or anti-inflammatory actions. Interleukins have been implicated to participate in neurodegenerative diseases. They play a critical role in Alzheimer's disease (AD) pathology which is characterized by the over-production of pro-inflammatory ILs. These may aggravate neurodegeneration, in addition to their contribution to detrimental mechanisms as oxidative stress, and excitotoxicity. However, recent research on the relation between ILs and AD revealed major discrepancies. Most of the major ILs were shown to play both pro- and anti-inflammatory roles in different experimental settings and models. The interactions between different ILs through shared pathways also add to the difficulty of drawing solid conclusions. In addition, targeting the different ILs has not yielded consistent results. The repeated failures of therapeutic drugs in treating AD necessitate the search for novel agents targeting multiple mechanisms of the disease pathology. In this context, the understanding of interleukins and their roles throughout the disease progression and interaction with other systems in the brain may provide promising therapeutic targets for the prevention or treatment of AD.
Collapse
Affiliation(s)
- Heba G Abdelhamed
- Department of Zoology and Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Arwa A Hassan
- Faculty of Pharmacy & Pharmaceutical Industries, Sinai University, Sinai, Egypt
| | - Alaa A Sakraan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Dalia M Mousa
- Department of Biotechnology, Faculty of Science, Cairo University, Giza, Egypt
| | - Heba S Aboul Ezz
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Neveen A Noor
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Center, Giza, Egypt
| | - Nasr M Radwan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
17
|
Zou J, Wang Y, Zhang X, Pan X, Fang T, Cai D, Guo L, Li Y, He Y, Cao X. CD33 Ameliorates Surgery-Induced Spatial Learning and Memory Impairments Through TREM2. Mol Neurobiol 2025; 62:2180-2190. [PMID: 39088031 DOI: 10.1007/s12035-024-04410-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/30/2024] [Indexed: 08/02/2024]
Abstract
Neuroinflammation is implicated in the onset of postoperative cognitive dysfunction (POCD), with CD33 and triggering receptor expressed on myeloid cells 2 (TREM2) playing crucial roles in immune response modulation and neuroinflammatory processes. A total of 96 aged male C57/BL6 mice (9-12 months) were randomly assigned to one of four groups, each receiving an siRNA injection into the lateral ventricle. Subsequently, the mice underwent partial hepatectomy under general anesthesia. To assess cognitive function, the Morris water maze tests were conducted both pre- and post-surgery. Following behavioral assessments, hippocampal tissues were swiftly harvested. The regulation of CD33 and TREM2 expression was achieved through siRNA in the BV2 microglia cell line. Expression levels of CD33 and TREM2 were evaluated both in vitro and in vivo using quantitative RT-PCR and western blot analyses. This study explored the impact of CD33 and TREM2 on POCD in aged mice and revealed that surgery and anesthesia increased CD33 expression, leading to spatial learning and memory impairments. Inhibiting CD33 expression via siRNA administration ameliorated cognitive deficits and mitigated the neuroinflammatory response triggered by surgery. Additionally, CD33 inhibition reversed the surgery-induced decrease in synaptic-related proteins, highlighting its role in preserving synaptic integrity. Moreover, our experiments suggest that CD33 may influence neuroinflammation and cognitive function through mechanisms involving TREM2. This is evidenced by the suppression of pro-inflammatory cytokines following CD33 knockdown in microglia and the reversal of these effects when both CD33 and TREM2 are concurrently knocked down. These findings imply that CD33 might promote neuroinflammation by inhibiting TREM2. This study highlights the potential of targeting CD33 as a promising therapeutic strategy for preventing and treating POCD. It provides valuable insights into the intricate mechanisms underlying cognitive dysfunction following surgical procedures.
Collapse
Affiliation(s)
- Jie Zou
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaxuan Wang
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China
| | - Xinyue Zhang
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China
| | - Xue Pan
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China
| | - Te Fang
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China
| | - Dasheng Cai
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China
| | - Lili Guo
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China
| | - Yu Li
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China
| | - Yi He
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China
| | - Xuezhao Cao
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
18
|
Farzan M, Saberi-Rounkian M, Asadi-Rizi A, Heidari Z, Farzan M, Fathi M, Aghaei A, Azadegan-Dehkordi F, Bagheri N. The emerging role of the microglia triggering receptor expressed on myeloid cells (TREM) 2 in multiple sclerosis. Exp Neurol 2025; 384:115071. [PMID: 39586397 DOI: 10.1016/j.expneurol.2024.115071] [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: 01/20/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND The chronic inflammatory condition known as multiple sclerosis (MS) causes inflammation and demyelination in the central nervous system (CNS). The activation of multiple cell types, including the CNS's resident immune cells called microglia, is a component of the immunological response in MS. Recently, the triggering receptor expressed on myeloid cells (TREM) family has emerged as a crucial player in modulating microglial function and subsequent neuroinflammation. Understanding the role of TREM receptors in MS pathogenesis could provide insightful information on how to develop new therapeutic approaches. MAIN BODY The TREM family consists of several receptors, including TREM-1 and TREM-2, which can be expressed on both immune cells, such as myeloid cells and microglia, and non-immune cells. These receptors interact with their respective ligands and regulate signaling pathways, ultimately leading to the control of microglial activation and inflammatory reactions. TREM-2, in particular, has garnered significant interest because of its connection with MS and other neurodegenerative diseases. The activation of microglia through TREM receptors in MS is thought to influence the equilibrium between helpful and detrimental inflammatory responses. TREM receptors can promote the phagocytosis of myelin debris and remove apoptotic cells, thus contributing to tissue repair and regeneration. However, excessive or dysregulated activation of microglia mediated by TREM receptors can lead to the release of pro-inflammatory cytokines and neurotoxic factors, exacerbating neuroinflammation and neurodegeneration in MS. CONCLUSION The emerging role of the TREM family in demyelinating diseases highlights the importance of microglia in disease pathogenesis. Understanding the mechanisms by which TREM receptors modulate microglial function can provide valuable insights into the development of targeted therapies for these disorders. By selectively targeting TREM receptors, it may be possible to harness their beneficial effects on tissue repair while dampening their detrimental pro-inflammatory responses. Further research is warranted to elucidate the precise signaling pathways and ligand interactions involved in TREM-mediated microglial activation, which could uncover novel therapeutic avenues for treating MS and other neuroinflammatory disorders.
Collapse
Affiliation(s)
- Mahan Farzan
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; Medical Plants Research Center, Basic Health Sciences institute, Shahrekord University of Medical sciences, Shahrekord, Iran
| | - Masoumeh Saberi-Rounkian
- Student Research committee, School of Paramedicine, Guilan University of Medical sciences, Rasht, Iran
| | - Atefeh Asadi-Rizi
- Young researchers and Elite club, Flavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Zahra Heidari
- Medical Plants Research Center, Basic Health Sciences institute, Shahrekord University of Medical sciences, Shahrekord, Iran
| | - Mahour Farzan
- Medical Plants Research Center, Basic Health Sciences institute, Shahrekord University of Medical sciences, Shahrekord, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ava Aghaei
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; Medical Plants Research Center, Basic Health Sciences institute, Shahrekord University of Medical sciences, Shahrekord, Iran
| | - Fatemeh Azadegan-Dehkordi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Nader Bagheri
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
19
|
Zheng Q, Wang X. Alzheimer's disease: insights into pathology, molecular mechanisms, and therapy. Protein Cell 2025; 16:83-120. [PMID: 38733347 PMCID: PMC11786724 DOI: 10.1093/procel/pwae026] [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: 03/04/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, is characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. This condition casts a significant shadow on global health due to its complex and multifactorial nature. In addition to genetic predispositions, the development of AD is influenced by a myriad of risk factors, including aging, systemic inflammation, chronic health conditions, lifestyle, and environmental exposures. Recent advancements in understanding the complex pathophysiology of AD are paving the way for enhanced diagnostic techniques, improved risk assessment, and potentially effective prevention strategies. These discoveries are crucial in the quest to unravel the complexities of AD, offering a beacon of hope for improved management and treatment options for the millions affected by this debilitating disease.
Collapse
Affiliation(s)
- Qiuyang Zheng
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Xin Wang
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| |
Collapse
|
20
|
Liu Z, Song SY. Genomic and Transcriptomic Approaches Advance the Diagnosis and Prognosis of Neurodegenerative Diseases. Genes (Basel) 2025; 16:135. [PMID: 40004464 PMCID: PMC11855287 DOI: 10.3390/genes16020135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS), represent a growing societal challenge due to their irreversible progression and significant impact on patients, caregivers, and healthcare systems. Despite advances in clinical and imaging-based diagnostics, these diseases are often detected at advanced stages, limiting the effectiveness of therapeutic interventions. Recent breakthroughs in genomic and transcriptomic technologies, including whole-genome sequencing, single-cell RNA sequencing (scRNA-seq), and CRISPR-based screens, have revolutionized the field, offering new avenues for early diagnosis and personalized prognosis. Genomic approaches have elucidated disease-specific genetic risk factors and molecular pathways, while transcriptomic studies have identified stage-specific biomarkers that correlate with disease progression and severity. Furthermore, genome-wide association studies (GWAS), polygenic risk scores (PRS), and spatial transcriptomics are enabling the stratification of patients based on their risk profiles and prognostic trajectories. Advances in functional genomics have uncovered actionable targets, such as ATXN2 in ALS and TREM2 in AD, paving the way for tailored therapeutic strategies. Despite these achievements, challenges remain in translating genomic discoveries into clinical practice due to disease heterogeneity and the complexity of neurodegenerative pathophysiology. Future integration of genetic technologies holds promise for transforming diagnostic and prognostic paradigms, offering hope for improved patient outcomes and precision medicine approaches.
Collapse
Affiliation(s)
- Zheng Liu
- Pathology Department, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Si-Yuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
21
|
Gao M, Kong W, Liu K, Wen G, Yu Y, Zhu Y, Jiang Z, Wei K. Exploring Brain Imaging and Genetic Risk Factors in Different Progression States of Alzheimer's Disease Through OSnetNMF-Based Methods. J Mol Neurosci 2025; 75:7. [PMID: 39815147 DOI: 10.1007/s12031-024-02274-8] [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: 01/07/2024] [Accepted: 09/29/2024] [Indexed: 01/18/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with no effective treatment, often preceded by mild cognitive impairment (MCI). Multimodal imaging genetics integrates imaging and genetic data to gain a deeper understanding of disease progression and individual variations. This study focuses on exploring the mechanisms that drive the transition from normal cognition to MCI and ultimately to AD. As an effective joint feature extraction and dimensionality reduction method, non-negative matrix factorization (NMF) and its improved variants, particularly the network-based non-negative matrix factorization (netNMF), have been widely used in multimodal analysis to mine brain imaging and genetic data by considering the interactions between different features. However, many of these methods overlook the importance of the coefficient matrix and do not address issues related to data accuracy and feature redundancy. To address these limitations, we propose an orthogonal sparse network non-negative matrix factorization (OSnetNMF) algorithm, which introduces orthogonal and sparse constraints based on netNMF. By establishing linear relationships between structural magnetic resonance imaging (sMRI) and corresponding gene expression data, OSnetNMF reduces feature redundancy and decreases correlation between data, resulting in more accurate and reliable biomarker extraction. Experiments demonstrate that the OSnetNMF algorithm can accurately identify risk regions of interest (ROIs) and key genes that characterize AD progression, revealing significant trends in ROI pairs such as l4thVen-HIF1A, rBst-MPO, and rBst-PTK2B. Comparative experiments show that the improved algorithm outperforms traditional methods, identifying more disease-related biomarkers and achieving better reconstruction performance.
Collapse
Affiliation(s)
- Min Gao
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, P. R. China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, P. R. China.
| | - Kun Liu
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, P. R. China
| | - Gen Wen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yaling Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yuemin Zhu
- CREATIS, University of Lyon, INSA Lyon, CNRS UMR 5220, Inserm U1294, Lyon, 69621, France
| | - Zhihan Jiang
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, P. R. China
| | - Kai Wei
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
22
|
Righi D, Manco C, Pardini M, Stufano A, Schino V, Pelagotti V, Massa F, Stefano ND, Plantone D. Investigating interleukin-8 in Alzheimer's disease: A comprehensive review. J Alzheimers Dis 2025; 103:38-55. [PMID: 39558604 DOI: 10.1177/13872877241298973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Several studies indicate that the development of Alzheimer's disease (AD) has strong interactions with immune mechanisms within the brain, indicating a close association between inflammation in the central nervous system and the progression of neurodegeneration. Despite considerable progress in understanding the inflammatory aspects of AD, several of them remain unresolved. Pro-inflammatory cytokines and microglia are pivotal components in the inflammatory cascade. Among these, the role of interleukin-8 (IL-8) in neurodegeneration seems complex and multifaceted, involving inflammation, neurotoxicity, blood-brain barrier disruption, and oxidative stress, and is still poorly characterized. We conducted a review to describe the evidence of IL-8 involvement in AD. IL-8 is a cytokine known for its proinflammatory properties and typically produced by macrophages, predominantly functions as a chemotactic signal for attracting neutrophils to inflamed sites in the bloodstream. Interestingly, IL-8 is also present in the brain, where it is primarily released by microglia in response to inflammatory signals. This review aims to provide a comprehensive overview of the structure, function, and regulatory mechanisms of IL-8 relevant to AD pathology.
Collapse
Affiliation(s)
- Delia Righi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Carlo Manco
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Angela Stufano
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Valentina Schino
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Virginia Pelagotti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genova, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Domenico Plantone
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| |
Collapse
|
23
|
Zhao Y, Guo Q, Tian J, Liu W, Wang X. TREM2 bridges microglia and extracellular microenvironment: Mechanistic landscape and therapeutical prospects on Alzheimer's disease. Ageing Res Rev 2025; 103:102596. [PMID: 39608728 DOI: 10.1016/j.arr.2024.102596] [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: 07/05/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
Neuroinflammation is closely related to the pathogenesis of Alzheimer's disease (AD). One of its prominent cellular components, microglia, is a potent coordinator of neuroinflammation in interplay with the characteristic AD pathological alterations including Aβ, tau, and neuronal defects, which constitute the AD-unique extracellular microenvironment. Mounting evidence implicates Triggering Receptors Expressed on Myeloid Cells 2 (TREM2) in the center of microglial activation, a vital event in the pathogenesis of AD. TREM2 is a pivotal microglial receptor that interacts with specific elements present in the AD microenvironment and induces microglial intracellular signallings contributing to phagocytosis, migration, cytokine production, metabolism, and survival, which shapes the microglial activation profile. It follows that TREM2 builds up a bridge between microglia and the extracellular microenvironment. This review illustrates how TREM2 modulates microglia to affect AD pathogenesis. Mainly presented facets in the review are i. the development of AD-specific microglial phenotypes (disease-associated microglia, DAM), ii. microglial interactions with major AD pathologies, and iii. the underlying intracellular signallings of microglial activation. Also, outstanding controversies regarding the nature of neuroinflammation are discussed. Through our illustration, we attempt to establish a TREM2-centered network of AD pathogenesis, in the hope as well to provide insights into the potential therapeutic strategies based on the underlying mechanisms.
Collapse
Affiliation(s)
- Yiheng Zhao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Guo
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jia Tian
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry/Hubei Province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| |
Collapse
|
24
|
Xu B, Tang C, Han R, Zhu C, Yang Y, Li H, Wu N, He D. Targeting the chemokine-microglia nexus: A novel strategy for modulating neuroinflammation in Alzheimer's disease. J Alzheimers Dis Rep 2025; 9:25424823251326044. [PMID: 40321241 PMCID: PMC12049630 DOI: 10.1177/25424823251326044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/07/2025] [Indexed: 05/08/2025] Open
Abstract
An increasing body of evidence suggests neuroinflammation has a prominent role in the pathogenesis of Alzheimer's disease (AD). The amyloid-β-tau-neurodegeneration (ATN) classification system is now being expanded toward an amyloid-β-tau neurodegeneration-neuroinflammation (ATN(I)) system. Activated microglia and reactive astrocytes are the key hubs for neuroinflammation in AD, and chemokines are recognized as pivotal modulators of microglial innate immune functions. In this review, based on the chemokine-microglia regulatory axis, we elucidate the mechanisms through which chemokines influence microglial function, potentially modulating neurotoxicity or neuroprotection in AD. The key chemokines that significantly affect microglial polarization, such as CCL2, CCL3, and CXCL1, are summarized, and their role in disease progression are elaborated. Additionally, we explore prospective therapeutic interventions centered on the chemokine-microglia regulatory axis, offering valuable perspectives on pathobiology of AD and avenues for pharmacological advancements.
Collapse
Affiliation(s)
- Bingyang Xu
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Chao Tang
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Rongshou Han
- Chemistry and Biochemistry Laboratory, Guizhou Medical University, Guiyang, Guizhou, China
| | - Chaomin Zhu
- Chemistry and Biochemistry Laboratory, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yuxuan Yang
- Chemistry and Biochemistry Laboratory, Guizhou Medical University, Guiyang, Guizhou, China
| | - Heyi Li
- Chemistry and Biochemistry Laboratory, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ning Wu
- Chemistry and Biochemistry Laboratory, Guizhou Medical University, Guiyang, Guizhou, China
| | - Dian He
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
25
|
Yao M, Miller GW, Vardarajan BN, Baccarelli AA, Guo Z, Liu Z. Deciphering proteins in Alzheimer's disease: A new Mendelian randomization method integrated with AlphaFold3 for 3D structure prediction. CELL GENOMICS 2024; 4:100700. [PMID: 39637861 DOI: 10.1016/j.xgen.2024.100700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/27/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Hidden confounding biases hinder identifying causal protein biomarkers for Alzheimer's disease in non-randomized studies. While Mendelian randomization (MR) can mitigate these biases using protein quantitative trait loci (pQTLs) as instrumental variables, some pQTLs violate core assumptions, leading to biased conclusions. To address this, we propose MR-SPI, a novel MR method that selects valid pQTL instruments using Leo Tolstoy's Anna Karenina principle and performs robust post-selection inference. Integrating MR-SPI with AlphaFold3, we developed a computational pipeline to identify causal protein biomarkers and predict 3D structural changes. Applied to genome-wide proteomics data from 54,306 UK Biobank participants and 455,258 subjects (71,880 cases and 383,378 controls) for a genome-wide association study of Alzheimer's disease, we identified seven proteins (TREM2, PILRB, PILRA, EPHA1, CD33, RET, and CD55) with structural alterations due to missense mutations. These findings offer insights into the etiology and potential drug targets for Alzheimer's disease.
Collapse
Affiliation(s)
- Minhao Yao
- Department of Statistics and Actuarial Science, The University of Hong Kong, Hong Kong SAR, China
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Badri N Vardarajan
- Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY, USA
| | - Andrea A Baccarelli
- Office of the Dean, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zijian Guo
- Department of Statistics, Rutgers University, Piscataway, NJ, USA.
| | - Zhonghua Liu
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA.
| |
Collapse
|
26
|
Wang H, Shi C, Jiang L, Liu X, Tang R, Tang M. Neuroimaging techniques, gene therapy, and gut microbiota: frontier advances and integrated applications in Alzheimer's Disease research. Front Aging Neurosci 2024; 16:1485657. [PMID: 39691161 PMCID: PMC11649678 DOI: 10.3389/fnagi.2024.1485657] [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: 08/24/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder marked by cognitive decline, for which effective treatments remain elusive due to complex pathogenesis. Recent advances in neuroimaging, gene therapy, and gut microbiota research offer new insights and potential intervention strategies. Neuroimaging enables early detection and staging of AD through visualization of biomarkers, aiding diagnosis and tracking of disease progression. Gene therapy presents a promising approach for modifying AD-related genetic expressions, targeting amyloid and tau pathology, and potentially repairing neuronal damage. Furthermore, emerging evidence suggests that the gut microbiota influences AD pathology through the gut-brain axis, impacting inflammation, immune response, and amyloid metabolism. However, each of these technologies faces significant challenges, including concerns about safety, efficacy, and ethical considerations. This article reviews the applications, advantages, and limitations of neuroimaging, gene therapy, and gut microbiota research in AD, with a particular focus on their combined potential for early diagnosis, mechanistic insights, and therapeutic interventions. We propose an integrated approach that leverages these tools to provide a multi-dimensional framework for advancing AD diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Haitao Wang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Chen Shi
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ling Jiang
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiaozhu Liu
- Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Rui Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxi Tang
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, China
- Department of Pathology, Yaan People’s Hospital (Yaan Hospital of West China Hospital of Sichuan University), Yaan, Sichuan, China
| |
Collapse
|
27
|
Hu Y, Tao W. Current perspectives on microglia-neuron communication in the central nervous system: Direct and indirect modes of interaction. J Adv Res 2024; 66:251-265. [PMID: 38195039 PMCID: PMC11674795 DOI: 10.1016/j.jare.2024.01.006] [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: 04/07/2023] [Revised: 10/05/2023] [Accepted: 01/06/2024] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND The incessant communication that takes place between microglia and neurons is essential the development, maintenance, and pathogenesis of the central nervous system (CNS). As mobile phagocytic cells, microglia serve a critical role in surveilling and scavenging the neuronal milieu to uphold homeostasis. AIM OF REVIEW This review aims to discuss the various mechanisms that govern the interaction between microglia and neurons, from the molecular to the organ system level, and to highlight the importance of these interactions in the development, maintenance, and pathogenesis of the CNS. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent research has revealed that microglia-neuron interaction is vital for regulating fundamental neuronal functions, such as synaptic pruning, axonal remodeling, and neurogenesis. The review will elucidate the intricate signaling pathways involved in these interactions, both direct and indirect, to provide a better understanding of the fundamental mechanisms of brain function. Furthermore, gaining insights into these signals could lead to the development of innovative therapies for neural disorders.
Collapse
Affiliation(s)
- Yue Hu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 220023, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weiwei Tao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 220023, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
28
|
Long H, Simmons A, Mayorga A, Burgess B, Nguyen T, Budda B, Rychkova A, Rhinn H, Tassi I, Ward M, Yeh F, Schwabe T, Paul R, Kenkare-Mitra S, Rosenthal A. Preclinical and first-in-human evaluation of AL002, a novel TREM2 agonistic antibody for Alzheimer's disease. Alzheimers Res Ther 2024; 16:235. [PMID: 39444037 PMCID: PMC11515656 DOI: 10.1186/s13195-024-01599-1] [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: 08/26/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Variants of the gene triggering receptor expressed on myeloid cells-2 (TREM2) increase the risk of Alzheimer's disease (AD) and other neurodegenerative disorders. Signaling by TREM2, an innate immune receptor expressed by microglia, is thought to enhance phagocytosis of amyloid beta (Aβ) and other damaged proteins, promote microglial proliferation, migration, and survival, and regulate inflammatory signaling. Thus, TREM2 activation has potential to alter the progression of AD. AL002 is an investigational, engineered, humanized monoclonal immunoglobulin G1 (IgG1) antibody designed to target TREM2. In AD mouse models, an AL002 murine variant has been previously shown to induce microglial proliferation and reduce filamentous Aβ plaques and neurite dystrophy. METHODS Preclinical studies assessed the safety, tolerability, pharmacokinetics, and pharmacodynamics of AL002 in cynomolgus monkeys. INVOKE-1 (NCT03635047) was a first-in-human phase 1, randomized, placebo-controlled, double-blind study assessing the safety, tolerability, PK, and PD of AL002 administered as single ascending doses (SAD) in healthy volunteers. RESULTS In cynomolgus monkeys, weekly intravenous injections of AL002 for 4 weeks were well tolerated, dose-dependently decreased soluble TREM2 (sTREM2) in cerebrospinal fluid (CSF) and total TREM2 in hippocampus and frontal cortex, and increased biomarkers of TREM2 signaling in CSF and brain. In the phase 1 study of 64 healthy volunteers, a single intravenous infusion of AL002 demonstrated brain target engagement based on a dose-dependent reduction of sTREM2 in CSF and parallel increases in biomarkers of TREM2 signaling and microglia recruitment. Single-dose AL002 showed central nervous system penetrance and was well tolerated, with no treatment-related serious adverse events over 12 weeks. CONCLUSIONS These findings support the continued clinical development of AL002 for AD and other neurodegenerative diseases in which TREM2 activation may be beneficial. AL002 is currently being tested in a phase 2, randomized, double-blind, placebo-controlled study in early AD. TRIAL REGISTRATION Clinicaltrials.gov, NCT03635047. Registered on August 15, 2018, https://www. CLINICALTRIALS gov/study/NCT03635047 .
Collapse
Affiliation(s)
- Hua Long
- Alector, Inc., South San Francisco, CA, 94080, USA
| | - Adam Simmons
- Alector, Inc., South San Francisco, CA, 94080, USA.
| | | | | | - Tuan Nguyen
- Alector, Inc., South San Francisco, CA, 94080, USA
| | | | | | - Herve Rhinn
- Alector, Inc., South San Francisco, CA, 94080, USA
- Leal Therapeutics, Worcester, MA, USA
| | - Ilaria Tassi
- Alector, Inc., South San Francisco, CA, 94080, USA
- Deep Apple Therapeutics, Inc., San Francisco, CA, USA
| | - Michael Ward
- Alector, Inc., South San Francisco, CA, 94080, USA
- Independent Consultant, San Francisco, CA, USA
| | - Felix Yeh
- Alector, Inc., South San Francisco, CA, 94080, USA
- Genentech, Inc., South San Francisco, CA, USA
| | - Tina Schwabe
- Alector, Inc., South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, USA
| | - Robert Paul
- Alector, Inc., South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
29
|
Zhu B, Wangzhou A, Yu D, Li T, Schmidt R, De Florencio SL, Chao L, Perez Y, Grinberg LT, Spina S, Ransohoff RM, Kriegstein AR, Seeley WW, Nowakowski T, Piao X. Adhesion G protein-coupled receptor ADGRG1 promotes protective microglial response in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618329. [PMID: 39464012 PMCID: PMC11507791 DOI: 10.1101/2024.10.15.618329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Germline genetic architecture of Alzheimer's disease (AD) indicates microglial mechanisms of disease susceptibility and outcomes. However, the mechanisms that enable microglia to mediate protective responses to AD pathology remain elusive. Adgrg1 is specifically expressed in yolk-sac-derived microglia. This study reveals the role of yolk-sac-derived microglia in AD pathology, highlighting the function of ADGRG1 in modulating microglial protective responses to amyloid deposition. Utilizing both constitutive and inducible microglial Adgrg1 knockout 5xFAD models, we demonstrate that Adgrg1 deficiency leads to increased amyloid deposition, exacerbated neuropathology, and accelerated cognitive impairment. Transcriptomic analyses reveal a distinct microglial state characterized by downregulated genes associated with homeostasis, phagocytosis, and lysosomal functions. Functional assays in mouse models and human embryonic stem cells-derived microglia support that microglial ADGRG1 is required for efficient Aβ phagocytosis. Together, these results uncover a GPCR-dependent microglial response to Aβ, pointing towards potential therapeutic strategies to alleviate disease progression by enhancing microglial functional competence.
Collapse
|
30
|
Doke R, Lamkhade GJ, Vinchurkar K, Singh S. Demystifying the Role of Neuroinflammatory Mediators as Biomarkers for Diagnosis, Prognosis, and Treatment of Alzheimer's Disease: A Review. ACS Pharmacol Transl Sci 2024; 7:2987-3003. [PMID: 39416969 PMCID: PMC11475310 DOI: 10.1021/acsptsci.4c00457] [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: 07/28/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Neuroinflammatory mediators play a pivotal role in the pathogenesis of Alzheimer's Disease (AD), influencing its onset, progression, and severity. The precise mechanisms behind AD are still not fully understood, leading current treatments to focus mainly on managing symptoms rather than preventing or curing the condition. The amyloid and tau hypotheses are the most widely accepted explanations for AD pathology; however, they do not completely account for the neuronal degeneration observed in AD. Growing evidence underscores the crucial role of neuroinflammation in the pathology of AD. The neuroinflammatory hypothesis presents a promising new approach to understanding the mechanisms driving AD. This review examines the importance of neuroinflammatory biomarkers in the diagnosis, prognosis, and treatment of AD. It delves into the mechanisms underlying neuroinflammation in AD, highlighting the involvement of various mediators such as cytokines, chemokines, and ROS. Additionally, this review discusses the potential of neuroinflammatory biomarkers as diagnostic tools, prognostic indicators, and therapeutic targets for AD management. By understanding the intricate interplay between neuroinflammation and AD pathology, this review aims to help in the development of efficient diagnostic and treatment plans to fight this debilitating neurological condition. Furthermore, it elaborates recent advancements in neuroimaging techniques and biofluid analysis for the identification and monitoring of neuroinflammatory biomarkers in AD patients.
Collapse
Affiliation(s)
- Rohit
R. Doke
- Jaihind
College of Pharmacy, Vadgaon Sahani, Pune, Maharashtra 412401, India
| | | | - Kuldeep Vinchurkar
- Krishna
School of Pharmacy, Kiran and Pallavi Patel
Global University, Vadodara, Gujarat 391243, India
| | - Sudarshan Singh
- Office
of Research Administration, Chiang Mai University, Chaing Mai 50200, Thailand
- Faculty
of Pharmacy, Chiang Mai University, Chaing Mai 50200, Thailand
| |
Collapse
|
31
|
Zhu M, Tian X, Han X, Ma Y, Fa W, Wang N, Liu R, Dong Y, Ren Y, Liu C, Tian N, Zhang H, Song L, Tang S, Cong L, Wang Y, Hou T, Qiu C, Du Y. Synergistic associations of CD33 variants and hypertension with brain and cognitive aging among dementia-free older adults: A population-based study. Alzheimers Dement 2024; 20:7193-7204. [PMID: 39215505 PMCID: PMC11485077 DOI: 10.1002/alz.14209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION CD33 rs3865444 and hypertension (HTN) are related to cognitive impairment, individually. However, little is known about their combined effects on cognitive function in older adults. METHODS This population-based study included 4368 dementia-free participants (age ≥65 years) in the Multimodal Interventions to Delay Dementia and Disability in Rural China (MIND-China), with data available in 1044 persons for gray matter volume and 85 persons for cerebral blood flow (CBF). We used general linear regression and mediation models to examine the associations of rs3865444 and HTN with cognition, brain atrophy, and CBF. RESULTS Among rs3865444 CC carriers, HTN and late-life HTN were significantly associated with impaired cognition. Midlife and late-life HTN were correlated with brain atrophy. CD33 rs3865444 CC moderated the mediation effect of gray matter volume on the association between HTN and global cognition. HTN was correlated with low CBF in rs3865444 CC carriers. DISCUSSION There are synergistic associations of CD33 rs3865444 and HTN with brain and cognitive aging in dementia-free older adults.
Collapse
Affiliation(s)
- Min Zhu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Xunyao Tian
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Xiaodong Han
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingP.R. China
| | - Yixun Ma
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Wenxin Fa
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Nan Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Rui Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Yi Dong
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Yifei Ren
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Cuicui Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Na Tian
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Heng Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Lin Song
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Shi Tang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Lin Cong
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Yongxiang Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Institute of Brain Science and Brain‐Inspired ResearchShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
- Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and SocietyKarolinska Institute‐Stockholm UniversityStockholmSweden
| | - Tingting Hou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
| | - Chengxuan Qiu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Institute of Brain Science and Brain‐Inspired ResearchShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
- Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and SocietyKarolinska Institute‐Stockholm UniversityStockholmSweden
| | - Yifeng Du
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP.R. China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial HospitalShandong UniversityJinanShandongP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanShandongP.R. China
- Institute of Brain Science and Brain‐Inspired ResearchShandong First Medical University & Shandong Academy of Medical SciencesJinanShandongP.R. China
| | | |
Collapse
|
32
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [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: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
33
|
Samuels JD, Lukens JR, Price RJ. Emerging roles for ITAM and ITIM receptor signaling in microglial biology and Alzheimer's disease-related amyloidosis. J Neurochem 2024; 168:3558-3573. [PMID: 37822118 PMCID: PMC11955997 DOI: 10.1111/jnc.15981] [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: 08/07/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023]
Abstract
Microglia are critical responders to amyloid beta (Aβ) plaques in Alzheimer's disease (AD). Therefore, the therapeutic targeting of microglia in AD is of high clinical interest. While previous investigation has focused on the innate immune receptors governing microglial functions in response to Aβ plaques, how microglial innate immune responses are regulated is not well understood. Interestingly, many of these microglial innate immune receptors contain unique cytoplasmic motifs, termed immunoreceptor tyrosine-based activating and inhibitory motifs (ITAM/ITIM), that are commonly known to regulate immune activation and inhibition in the periphery. In this review, we summarize the diverse functions employed by microglia in response to Aβ plaques and also discuss the innate immune receptors and intracellular signaling players that guide these functions. Specifically, we focus on the role of ITAM and ITIM signaling cascades in regulating microglia innate immune responses. A better understanding of how microglial innate immune responses are regulated in AD may provide novel therapeutic avenues to tune the microglial innate immune response in AD pathology.
Collapse
Affiliation(s)
- Joshua D. Samuels
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
| | - Richard J. Price
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
34
|
Cao C, Fu G, Xu R, Li N. Coupling of Alzheimer's Disease Genetic Risk Factors with Viral Susceptibility and Inflammation. Aging Dis 2024; 15:2028-2050. [PMID: 37962454 PMCID: PMC11346407 DOI: 10.14336/ad.2023.1017] [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: 06/30/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by persistent cognitive decline. Amyloid plaque deposition and neurofibrillary tangles are the main pathological features of AD brain, though mechanisms leading to the formation of lesions remain to be understood. Genetic efforts through genome-wide association studies (GWAS) have identified dozens of risk genes influencing the pathogenesis and progression of AD, some of which have been revealed in close association with increased viral susceptibilities and abnormal inflammatory responses in AD patients. In the present study, we try to present a list of AD candidate genes that have been shown to affect viral infection and inflammatory responses. Understanding of how AD susceptibility genes interact with the viral life cycle and potential inflammatory pathways would provide possible therapeutic targets for both AD and infectious diseases.
Collapse
Affiliation(s)
| | | | - Ruodan Xu
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Ning Li
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
35
|
Liu E, Zhang Y, Wang JZ. Updates in Alzheimer's disease: from basic research to diagnosis and therapies. Transl Neurodegener 2024; 13:45. [PMID: 39232848 PMCID: PMC11373277 DOI: 10.1186/s40035-024-00432-x] [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: 03/12/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, characterized pathologically by extracellular deposition of β-amyloid (Aβ) into senile plaques and intracellular accumulation of hyperphosphorylated tau (pTau) as neurofibrillary tangles. Clinically, AD patients show memory deterioration with varying cognitive dysfunctions. The exact molecular mechanisms underlying AD are still not fully understood, and there are no efficient drugs to stop or reverse the disease progression. In this review, we first provide an update on how the risk factors, including APOE variants, infections and inflammation, contribute to AD; how Aβ and tau become abnormally accumulated and how this accumulation plays a role in AD neurodegeneration. Then we summarize the commonly used experimental models, diagnostic and prediction strategies, and advances in periphery biomarkers from high-risk populations for AD. Finally, we introduce current status of development of disease-modifying drugs, including the newly officially approved Aβ vaccines, as well as novel and promising strategies to target the abnormal pTau. Together, this paper was aimed to update AD research progress from fundamental mechanisms to the clinical diagnosis and therapies.
Collapse
Affiliation(s)
- Enjie Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yao Zhang
- Department of Endocrine, Liyuan Hospital, Key Laboratory of Ministry of Education for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Jian-Zhi Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
36
|
Samuel Olajide T, Oyerinde TO, Omotosho OI, Okeowo OM, Olajide OJ, Ijomone OM. Microglial senescence in neurodegeneration: Insights, implications, and therapeutic opportunities. NEUROPROTECTION 2024; 2:182-195. [PMID: 39364217 PMCID: PMC11449118 DOI: 10.1002/nep3.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/03/2024] [Indexed: 10/05/2024]
Abstract
The existing literature on neurodegenerative diseases (NDDs) reveals a common pathological feature: the accumulation of misfolded proteins. However, the heterogeneity in disease onset mechanisms and the specific brain regions affected complicates the understanding of the diverse clinical manifestations of individual NDDs. Dementia, a hallmark symptom across various NDDs, serves as a multifaceted denominator, contributing to the clinical manifestations of these disorders. There is a compelling hypothesis that therapeutic strategies capable of mitigating misfolded protein accumulation and disrupting ongoing pathogenic processes may slow or even halt disease progression. Recent research has linked disease-associated microglia to their transition into a senescent state-characterized by irreversible cell cycle arrest-in aging populations and NDDs. Although senescent microglia are consistently observed in NDDs, few studies have utilized animal models to explore their role in disease pathology. Emerging evidence from experimental rat models suggests that disease-associated microglia exhibit characteristics of senescence, indicating that deeper exploration of microglial senescence could enhance our understanding of NDD pathogenesis and reveal novel therapeutic targets. This review underscores the importance of investigating microglial senescence and its potential contributions to the pathophysiology of NDDs, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Additionally, it highlights the potential of targeting microglial senescence through iron chelation and senolytic therapies as innovative approaches for treating age-related NDDs.
Collapse
Affiliation(s)
- Tobiloba Samuel Olajide
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Ondo, Nigeria
| | - Toheeb O. Oyerinde
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Ondo, Nigeria
| | - Omolabake I. Omotosho
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Ondo, Nigeria
| | - Oritoke M. Okeowo
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Ondo, Nigeria
- Department of Physiology, School of Basic Medical Science, Federal University of Technology, Akure, Ondo, Nigeria
| | - Olayemi J. Olajide
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
- Division of Neurobiology, Department of Anatomy, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara, Nigeria
| | - Omamuyouwi M. Ijomone
- Laboratory for Experimental and Translational Neurobiology, University of Medical Sciences, Ondo, Ondo, Nigeria
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
37
|
Shen Y, Liu F, Zhang M. Therapeutic potential of plant-derived natural compounds in Alzheimer's disease: Targeting microglia-mediated neuroinflammation. Biomed Pharmacother 2024; 178:117235. [PMID: 39094545 DOI: 10.1016/j.biopha.2024.117235] [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: 06/12/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
Microglia are resident immune cells of the central nervous system (CNS) with roles in sensing, housekeeping, and defense. Exploring the role of microglia in the occurrence and development of Alzheimer's disease (AD) and the possible therapeutic mechanism of plant-derived natural compounds (PDNCs) that regulate microglia-associated neuroinflammation may potentially help in elucidating the pathogenesis of AD and provide novel insights for its treatment. This review explores the role of abnormal microglial activation and its dominant neuroinflammatory response, as well as the activation of their target receptors and signaling pathways in AD pathogenesis. Additionally, we report an update on the potential pharmacological mechanisms of multiple PDNCs in modulating microglia-associated neuroinflammation in AD treatment. Dysregulated activation of microglial receptors and their downstream pathways impaired immune homeostasis in animal models of AD. Multiple signaling pathways, such as mitogen-activated protein kinase (MAPK), nuclear factor kappa light chain enhancer of activated B cells (NF-κB), and Toll-like receptors, play important roles in microglial activation and can exacerbate microglia-mediated neuroinflammation. PDNCs, such as magnolol, stigmasterol, matrine, naringenin, naringin, and resveratrol, can delay the progression of AD by inhibiting the proinflammatory receptors of microglia, activating its anti-inflammatory receptors, regulating the receptors related to β-amyloid (Aβ) clearance, reversing immune dysregulation, and maintaining the immune homeostasis of microglial downstream pathways. This review summarizes the mechanisms by which microglia cause chronic inflammation in AD and evaluates the beneficial effects of PDNCs on immune regulation in AD by regulating microglial receptors and their downstream pathways.
Collapse
Affiliation(s)
- Yanyan Shen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, China.
| | - Fang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, China
| | - Mingjie Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
38
|
Yin Y, Yang H, Li R, Wu G, Qin Q, Tang Y. A systematic review of the role of TREM2 in Alzheimer's disease. Chin Med J (Engl) 2024; 137:1684-1694. [PMID: 38915213 PMCID: PMC11268819 DOI: 10.1097/cm9.0000000000003000] [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: 12/30/2023] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Given the established genetic linkage between triggering receptors expressed on myeloid cells 2 (TREM2) and Alzheimer's disease (AD), an expanding research body has delved into the intricate role of TREM2 within the AD context. However, a conflicting landscape of outcomes has emerged from both in vivo and in vitro investigations. This study aimed to elucidate the multifaceted nuances and gain a clearer comprehension of the role of TREM2. METHODS PubMed database was searched spanning from its inception to January 2022. The search criteria took the form of ("Alzheimer's disease" OR "AD") AND ("transgenic mice model" OR "transgenic mouse model") AND ("Triggering receptor expressed on myeloid cells" OR "TREM2"). Inclusion criteria consisted of the following: (1) publication of original studies in English; (2) utilization of transgenic mouse models for AD research; and (3) reports addressing the subject of TREM2. RESULTS A total of 43 eligible articles were identified. Our analysis addresses four pivotal queries concerning the interrelation of TREM2 with microglial function, Aβ accumulation, tau pathology, and inflammatory processes. However, the diverse inquiries posed yielded inconsistent responses. Nevertheless, the inconsistent roles of TREM2 within these AD mouse models potentially hinge upon factors such as age, sex, brain region, model type, and detection methodologies. CONCLUSIONS This review substantiates the evolving understanding of TREM2's disease progression-dependent impacts. Furthermore, it reviews the interplay between TREM2 and its effects across diverse tissues and temporal stages.
Collapse
Affiliation(s)
- Yunsi Yin
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China
| | - Hanchen Yang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China
| | - Ruiyang Li
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China
| | - Guangshan Wu
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China
| | - Qi Qin
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China
| |
Collapse
|
39
|
Hudgins AD, Zhou S, Arey RN, Rosenfeld MG, Murphy CT, Suh Y. A systems biology-based identification and in vivo functional screening of Alzheimer's disease risk genes reveal modulators of memory function. Neuron 2024; 112:2112-2129.e4. [PMID: 38692279 PMCID: PMC11223975 DOI: 10.1016/j.neuron.2024.04.009] [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: 07/16/2022] [Revised: 10/18/2023] [Accepted: 04/08/2024] [Indexed: 05/03/2024]
Abstract
Genome-wide association studies (GWASs) have uncovered over 75 genomic loci associated with risk for late-onset Alzheimer's disease (LOAD), but identification of the underlying causal genes remains challenging. Studies of induced pluripotent stem cell (iPSC)-derived neurons from LOAD patients have demonstrated the existence of neuronal cell-intrinsic functional defects. Here, we searched for genetic contributions to neuronal dysfunction in LOAD using an integrative systems approach that incorporated multi-evidence-based gene mapping and network-analysis-based prioritization. A systematic perturbation screening of candidate risk genes in Caenorhabditis elegans (C. elegans) revealed that neuronal knockdown of the LOAD risk gene orthologs vha-10 (ATP6V1G2), cmd-1 (CALM3), amph-1 (BIN1), ephx-1 (NGEF), and pho-5 (ACP2) alters short-/intermediate-term memory function, the cognitive domain affected earliest during LOAD progression. These results highlight the impact of LOAD risk genes on evolutionarily conserved memory function, as mediated through neuronal endosomal dysfunction, and identify new targets for further mechanistic interrogation.
Collapse
Affiliation(s)
- Adam D Hudgins
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Shiyi Zhou
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Rachel N Arey
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Michael G Rosenfeld
- Department of Medicine, School of Medicine, University of California, La Jolla, CA, USA; Howard Hughes Medical Institute, University of California, La Jolla, CA, USA
| | - Coleen T Murphy
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; LSI Genomics, Princeton University, Princeton, NJ, USA.
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
40
|
Wong E, Malviya M, Jain T, Liao GP, Kehs Z, Chang JC, Studer L, Scheinberg DA, Li YM. HuM195 and its single-chain variable fragment increase Aβ phagocytosis in microglia via elimination of CD33 inhibitory signaling. Mol Psychiatry 2024; 29:2084-2094. [PMID: 38383769 PMCID: PMC11336028 DOI: 10.1038/s41380-024-02474-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/18/2024] [Accepted: 01/31/2024] [Indexed: 02/23/2024]
Abstract
CD33 is a transmembrane receptor expressed on cells of myeloid lineage and regulates innate immunity. CD33 is a risk factor for Alzheimer's disease (AD) and targeting CD33 has been a promising strategy drug development. However, the mechanism of CD33's action is poorly understood. Here we investigate the mechanism of anti-CD33 antibody HuM195 (Lintuzumab) and its single-chain variable fragment (scFv) and examine their therapeutic potential. Treatment with HuM195 full-length antibody or its scFv increased phagocytosis of β-amyloid 42 (Aβ42) in human microglia and monocytes. This activation of phagocytosis was driven by internalization and degradation of CD33, thereby downregulating its inhibitory signal. HumM195 transiently induced CD33 phosphorylation and its signaling via receptor dimerization. However, this signaling decayed with degradation of CD33. scFv binding to CD33 leads to a degradation of CD33 without detection of the CD33 dimerization and signaling. Moreover, we found that treatments with either HuM195 or scFv promotes the secretion of IL33, a cytokine implicated in microglia reprogramming. Importantly, recombinant IL33 potentiates the uptake of Aβ42 in monocytes. Collectively, our findings provide unanticipated mechanistic insight into the role of CD33 signaling in both monocytes and microglia and define a molecular basis for the development of CD33-based therapy of AD.
Collapse
Affiliation(s)
- Eitan Wong
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Manish Malviya
- Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - Tanya Jain
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
- Program of Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, 10021, USA
| | - George P Liao
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
- Program of Pharmacology Weill Graduate School of Medical Sciences of Cornell University, New York, NY, 10021, USA
| | - Zoe Kehs
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
- Program of Pharmacology Weill Graduate School of Medical Sciences of Cornell University, New York, NY, 10021, USA
| | - Jerry C Chang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, 10021, USA
| | - Lorenz Studer
- Developmental biology program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10021, USA
| | - David A Scheinberg
- Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA.
- Program of Pharmacology Weill Graduate School of Medical Sciences of Cornell University, New York, NY, 10021, USA.
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA.
- Program of Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, 10021, USA.
- Program of Pharmacology Weill Graduate School of Medical Sciences of Cornell University, New York, NY, 10021, USA.
| |
Collapse
|
41
|
Yan H, Wang W, Cui T, Shao Y, Li M, Fang L, Feng L. Advances in the Understanding of the Correlation Between Neuroinflammation and Microglia in Alzheimer's Disease. Immunotargets Ther 2024; 13:287-304. [PMID: 38881647 PMCID: PMC11180466 DOI: 10.2147/itt.s455881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease with a subtle and progressive onset and is the most common type of dementia. However, its etiology and pathogenesis have not yet been fully elucidated. The common pathological manifestations of AD include extraneuronal β-amyloid deposition (Aβ), intraneuronal tau protein phosphorylation leading to the formation of 'neurofibrillary tangles' (NFTs), neuroinflammation, progressive loss of brain neurons/synapses, and glucose metabolism disorders. Current treatment approaches for AD primarily focus on the 'Aβ cascade hypothesis and abnormal aggregation of hyperphosphorylation of tau proteins', but have shown limited efficacy. Therefore, there is an ongoing need to identify more effective treatment targets for AD. The central nervous system (CNS) inflammatory response plays a key role in the occurrence and development of AD. Neuroinflammation is an immune response activated by glial cells in the CNS that usually occurs in response to stimuli such as nerve injury, infection and toxins or in response to autoimmunity. Neuroinflammation ranks as the third most prominent pathological feature in AD, following Aβ and NFTs. In recent years, the focus on the role of neuroinflammation and microglia in AD has increased due to the advancements in genome-wide association studies (GWAS) and sequencing technology. Furthermore, research has validated the pivotal role of microglia-mediated neuroinflammation in the progression of AD. Therefore, this article reviews the latest research progress on the role of neuroinflammation triggered by microglia in AD in recent years, aiming to provide a new theoretical basis for further exploring the role of neuroinflammation in the process of AD occurrence and development.
Collapse
Affiliation(s)
- Huiying Yan
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Wei Wang
- Department of Intensive Care Unit, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Tingting Cui
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Yanxin Shao
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, People's Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Limei Fang
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| |
Collapse
|
42
|
Wang W, Wang Y, Wang F, Xie G, Liu S, Li Z, Wang P, Liu J, Lin L. Gastrodin regulates the TLR4/TRAF6/NF-κB pathway to reduce neuroinflammation and microglial activation in an AD model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155518. [PMID: 38552431 DOI: 10.1016/j.phymed.2024.155518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/25/2024] [Accepted: 03/07/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Gastrodia elata (Orchidaceae) is a medicinal plant used in traditional Chinese medicine. The rhizomes contain numerous active components, of which Gastrodin (p-hydroxymethylphenyl-B-D-glucopyranoside) forms the basis of the traditional medicine Gastrodiae Rhizoma. Gastrodin is also found in other medicinal plants and has neuroprotective, antioxidant, and anti-inflammatory effects. Neuroinflammation plays a crucial role in neurodegeneration. Research indicates that consuming meals and drinks containing Gastrodiaelata can enhance cognitive functioning and memory in elderly patients. The mechanisms relevant to the problem have not been completely understood. PURPOSE The aim was to examine the in vivo and in vitro anti-neuroinflammatory effects of Gastrodin. STUDY DESIGN The neuroprotective effects of Gastrodin on the TLR4/TRAF6/NF-κB pathway and Stat3 phosphorylation in LPS-treated C57BL/6 mice and BV-2 cells were investigated. METHODS 1. C57BL/6 mice were assigned to model, gastrodin, donepezil, and control groups (n = 10 per group). The Gastrodin group received 100 mg/kg/d for five days, and the Dopenezil group 1.3 mg/kg/d. A neuroinflammation model was established by administering intraperitoneal injections of 2 mg/kg LPS to all groups, excluding the control. To induce microglial activation in Gastrodin-treated mouse microglial BV-2 cells, 1 µg/ml LPS was introduced for 24 h Morris water mazes were utilized to evaluate learning and spatial memory. Expression and subcellular localization of TLR4/TRAF6/NF-κB axis-related proteins and p-Stat3, Iba-1, GFAP, iNOS, and CD206 were assessed by immunofluorescence, western blots, and ELISA. qRT-PCR was performed to determine and measure IL-1β, TNF-α, cell migration, and phagocytosis. Overexpression of TRAF6 was induced by transfection, and the effect of Gastrodin on IL-1β and p-NF-κB p65 levels was assessed. RESULTS 1. In mice, gastrodin treatment mitigated LPS-induced deficits in learning and spatial memory, as well as reducing neuroinflammation in the hippocampus, expression of TLR4/TRAF6/NF-κB pathway proteins, activation of microglia and astrocytes, and phosphorylation of Stat3. 2. Gastrodin pretreatment improved LPS-induced inflammation in vitro, reducing expression of TLR4/TRAF6/NF-κB-associated proteins and p-Stat3, inducing microglial transformation from M1 to M2, and inhibiting migration and phagocytosis. Overexpression of TRAF6 inhibited the Gastrodin-induced effects. CONCLUSION Gastrodin suppresses neuroinflammation and microglial activation by modifying the TLR4/TRAF6/NF-κB pathway and Stat3 phosphorylation.
Collapse
Affiliation(s)
- Wensheng Wang
- Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, No.16 of Huangjia Lake Western Road, Hong Shan District, Wuhan 430065, China; Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China
| | - Yu Wang
- Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, No.16 of Huangjia Lake Western Road, Hong Shan District, Wuhan 430065, China
| | - Fengjie Wang
- Department of Medicine, Hubei Minzu University, Enshi 445000, China
| | - Guangjing Xie
- Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China
| | - Shangzhi Liu
- Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China
| | - Zefei Li
- Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China
| | - Ping Wang
- Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China.
| | - Junfeng Liu
- Key Laboratory of TCM Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, No.16 of Huangjia Lake Western Road, Hong Shan District, Wuhan 430065, China.
| | - Li Lin
- Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, No.16 of Huangjia Lake Western Road, Hong Shan District, Wuhan 430065, China.
| |
Collapse
|
43
|
Chen Z, Balachandran YL, Chong WP, Chan KWY. Roles of Cytokines in Alzheimer's Disease. Int J Mol Sci 2024; 25:5803. [PMID: 38891990 PMCID: PMC11171747 DOI: 10.3390/ijms25115803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
The neuroimmune system is a collection of immune cells, cytokines, and the glymphatic system that plays a pivotal role in the pathogenesis and progression of Alzheimer's disease (AD). Of particular focus are cytokines, a group of immune signaling molecules that facilitate communication among immune cells and contribute to inflammation in AD. Extensive research has shown that the dysregulated secretion of certain cytokines (IL-1β, IL-17, IL-12, IL-23, IL-6, and TNF-α) promotes neuroinflammation and exacerbates neuronal damage in AD. However, anti-inflammatory cytokines (IL-2, IL-3, IL-33, and IL-35) are also secreted during AD onset and progression, thereby preventing neuroinflammation. This review summarizes the involvement of pro- and anti-inflammatory cytokines in AD pathology and discusses their therapeutic potential.
Collapse
Affiliation(s)
- Zilin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
| | - Yekkuni L. Balachandran
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
| | - Wai Po Chong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Kannie W. Y. Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; (Z.C.); (Y.L.B.)
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, China
- Shenzhen Research Institute, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
44
|
Jiang S, Cai G, Yang Z, Shi H, Zeng H, Ye Q, Hu Z, Wang Z. Biomimetic Nanovesicles as a Dual Gene Delivery System for the Synergistic Gene Therapy of Alzheimer's Disease. ACS NANO 2024; 18:11753-11768. [PMID: 38649866 DOI: 10.1021/acsnano.3c13150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
The association between dysfunctional microglia and amyloid-β (Aβ) is a fundamental pathological event and increases the speed of Alzheimer's disease (AD). Additionally, the pathogenesis of AD is intricate and a single drug may not be enough to achieve a satisfactory therapeutic outcome. Herein, we reported a facile and effective gene therapy strategy for the modulation of microglia function and intervention of Aβ anabolism by ROS-responsive biomimetic exosome-liposome hybrid nanovesicles (designated as TSEL). The biomimetic nanovesicles codelivery β-site amyloid precursor protein cleaving enzyme-1 (BACE1) siRNA (siBACE1) and TREM2 plasmid (pTREM2) gene drug efficiently penetrate the blood-brain barrier and enhance the drug accumulation at AD lesions with the help of exosomes homing ability and angiopep-2 peptides. Specifically, an upregulation of TREM2 expression can reprogram microglia from a pro-inflammatory M1 phenotype to an anti-inflammatory M2 phenotype while also restoring its capacity to phagocytose Aβ and its nerve repair function. In addition, siRNA reduces the production of Aβ plaques at the source by knocking out the BACE1 gene, which is expected to further enhance the therapeutic effect of AD. The in vivo study suggests that TSEL through the synergistic effect of two gene drugs can ameliorate APP/PS1 mice cognitive impairment by regulating the activated microglial phenotype, reducing the accumulation of Aβ, and preventing the retriggering of neuroinflammation. This strategy employs biomimetic nanovesicles for the delivery of dual nucleic acids, achieving synergistic gene therapy for AD, thus offering more options for the treatment of AD.
Collapse
Affiliation(s)
- Sujun Jiang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Guoen Cai
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhimin Yang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Haoyuan Shi
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Huajie Zeng
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Qinyong Ye
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhiyuan Hu
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- School of Nanoscience and Technology, Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, China
| | - Zihua Wang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| |
Collapse
|
45
|
Kapasi A, Yu L, Leurgans SE, Agrawal S, Boyle PA, Bennett DA, Schneider JA. Association between hippocampal microglia, AD and LATE-NC, and cognitive decline in older adults. Alzheimers Dement 2024; 20:3193-3202. [PMID: 38494787 PMCID: PMC11095444 DOI: 10.1002/alz.13780] [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: 10/26/2023] [Accepted: 01/29/2024] [Indexed: 03/19/2024]
Abstract
INTRODUCTION This study investigates the relationship between microglia inflammation in the hippocampus, brain pathologies, and cognitive decline. METHODS Participants underwent annual clinical evaluations and agreed to brain donation. Neuropathologic evaluations quantified microglial burden in the hippocampus, amyloid beta (Aβ), tau tangles, and limbic age-related transactive response DNA-binding protein 43 (TDP-43) encephalopathy neuropathologic changes (LATE-NC), and other common brain pathologies. Mixed-effect and linear regression models examined the association of microglia with a decline in global and domain-specific cognitive measures, and separately with brain pathologies. Path analyses estimated direct and indirect effects of microglia on global cognition. RESULT Hippocampal microglia were associated with a faster decline in global cognition, specifically in episodic memory, semantic memory, and perceptual speed. Tau tangles and LATE-NC were independently associated with microglia. Other pathologies, including Aβ, were not related. Regional hippocampal burden of tau tangles and TDP-43 accounted for half of the association of microglia with cognitive decline. DISCUSSION Microglia inflammation in the hippocampus contributes to cognitive decline. Tau tangles and LATE-NC partially mediate this association.
Collapse
Affiliation(s)
- Alifiya Kapasi
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
| | - Lei Yu
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Sue E Leurgans
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Sonal Agrawal
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
| | - Patricia A Boyle
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Psychiatry and Behavioral SciencesRush University Medical CenterChicagoIllinoisUSA
| | - David A Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Julie A Schneider
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
46
|
Adamu A, Li S, Gao F, Xue G. The role of neuroinflammation in neurodegenerative diseases: current understanding and future therapeutic targets. Front Aging Neurosci 2024; 16:1347987. [PMID: 38681666 PMCID: PMC11045904 DOI: 10.3389/fnagi.2024.1347987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/25/2024] [Indexed: 05/01/2024] Open
Abstract
Neuroinflammation refers to a highly complicated reaction of the central nervous system (CNS) to certain stimuli such as trauma, infection, and neurodegenerative diseases. This is a cellular immune response whereby glial cells are activated, inflammatory mediators are liberated and reactive oxygen and nitrogen species are synthesized. Neuroinflammation is a key process that helps protect the brain from pathogens, but inappropriate, or protracted inflammation yields pathological states such as Parkinson's disease, Alzheimer's, Multiple Sclerosis, and other neurodegenerative disorders that showcase various pathways of neurodegeneration distributed in various parts of the CNS. This review reveals the major neuroinflammatory signaling pathways associated with neurodegeneration. Additionally, it explores promising therapeutic avenues, such as stem cell therapy, genetic intervention, and nanoparticles, aiming to regulate neuroinflammation and potentially impede or decelerate the advancement of these conditions. A comprehensive understanding of the intricate connection between neuroinflammation and these diseases is pivotal for the development of future treatment strategies that can alleviate the burden imposed by these devastating disorders.
Collapse
Affiliation(s)
| | | | | | - Guofang Xue
- Department of Neurology, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
47
|
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
|
48
|
Oshima T, Kater MSJ, Huffels CFM, Wesseling EM, Middeldorp J, Hol EM, Verheijen MHG, Smit AB, Boddeke EWGM, Eggen BJL. Early amyloid-induced changes in microglia gene expression in male APP/PS1 mice. J Neurosci Res 2024; 102:e25295. [PMID: 38515329 DOI: 10.1002/jnr.25295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/04/2023] [Accepted: 01/12/2024] [Indexed: 03/23/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and the most common cause of dementia, characterized by deposition of extracellular amyloid-beta (Aβ) aggregates and intraneuronal hyperphosphorylated Tau. Many AD risk genes, identified in genome-wide association studies (GWAS), are expressed in microglia, the innate immune cells of the central nervous system. Specific subtypes of microglia emerged in relation to AD pathology, such as disease-associated microglia (DAMs), which increased in number with age in amyloid mouse models and in human AD cases. However, the initial transcriptional changes in these microglia in response to amyloid are still unknown. Here, to determine early changes in microglia gene expression, hippocampal microglia from male APPswe/PS1dE9 (APP/PS1) mice and wild-type littermates were isolated and analyzed by RNA sequencing (RNA-seq). By bulk RNA-seq, transcriptomic changes were detected in hippocampal microglia from 6-months-old APP/PS1 mice. By performing single-cell RNA-seq of CD11c-positive and negative microglia from 6-months-old APP/PS1 mice and analysis of the transcriptional trajectory from homeostatic to CD11c-positive microglia, we identified a set of genes that potentially reflect the initial response of microglia to Aβ.
Collapse
Affiliation(s)
- Takuya Oshima
- Department of Biomedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Mandy S J Kater
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Christiaan F M Huffels
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Evelyn M Wesseling
- Department of Biomedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jinte Middeldorp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Department of Neurobiology & Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Mark H G Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Erik W G M Boddeke
- Department of Biomedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Bart J L Eggen
- Department of Biomedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
49
|
Wang H, Shen Z, Wu CS, Ji P, Noh JY, Geoffroy CG, Kim S, Threadgill D, Li J, Zhou Y, Xiao X, Zheng H, Sun Y. Neuronal ablation of GHSR mitigates diet-induced depression and memory impairment via AMPK-autophagy signaling-mediated inflammation. Front Immunol 2024; 15:1339937. [PMID: 38464534 PMCID: PMC10920242 DOI: 10.3389/fimmu.2024.1339937] [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: 11/17/2023] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
Obesity is associated with chronic inflammation in the central nervous system (CNS), and neuroinflammation has been shown to have detrimental effects on mood and cognition. The growth hormone secretagogue receptor (GHSR), the biologically relevant receptor of the orexigenic hormone ghrelin, is primarily expressed in the brain. Our previous study showed that neuronal GHSR deletion prevents high-fat diet-induced obesity (DIO). Here, we investigated the effect of neuronal GHSR deletion on emotional and cognitive functions in DIO. The neuron-specific GHSR-deficient mice exhibited reduced depression and improved spatial memory compared to littermate controls under DIO. We further examined the cortex and hippocampus, the major regions regulating cognitive and emotional behaviors, and found that the neuronal deletion of GHSR reduced DIO-induced neuroinflammation by suppressing proinflammatory chemokines/cytokines and decreasing microglial activation. Furthermore, our data showed that neuronal GHSR deletion suppresses neuroinflammation by downregulating AMPK-autophagy signaling in neurons. In conclusion, our data reveal that neuronal GHSR inhibition protects against DIO-induced depressive-like behavior and spatial cognitive dysfunction, at least in part, through AMPK-autophagy signaling-mediated neuroinflammation.
Collapse
Affiliation(s)
- Hongying Wang
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Department of Endocrinology, Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zheng Shen
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Chia-Shan Wu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Pengfei Ji
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Ji Yeon Noh
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| | - Cédric G. Geoffroy
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University, College Station, TX, United States
| | - Sunja Kim
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, United States
| | - David Threadgill
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, United States
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, United States
| | - Jianrong Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Yu Zhou
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, United States
| | - Yuxiang Sun
- Department of Nutrition, Texas A&M University, College Station, TX, United States
| |
Collapse
|
50
|
Ifediora N, Canoll P, Hargus G. Human stem cell transplantation models of Alzheimer's disease. Front Aging Neurosci 2024; 16:1354164. [PMID: 38450383 PMCID: PMC10915253 DOI: 10.3389/fnagi.2024.1354164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024] Open
Abstract
Alzheimer's disease (AD) is the most frequent form of dementia. It is characterized by pronounced neuronal degeneration with formation of neurofibrillary tangles and deposition of amyloid β throughout the central nervous system. Animal models have provided important insights into the pathogenesis of AD and they have shown that different brain cell types including neurons, astrocytes and microglia have important functions in the pathogenesis of AD. However, there are difficulties in translating promising therapeutic observations in mice into clinical application in patients. Alternative models using human cells such as human induced pluripotent stem cells (iPSCs) may provide significant advantages, since they have successfully been used to model disease mechanisms in neurons and in glial cells in neurodegenerative diseases in vitro and in vivo. In this review, we summarize recent studies that describe the transplantation of human iPSC-derived neurons, astrocytes and microglial cells into the forebrain of mice to generate chimeric transplantation models of AD. We also discuss opportunities, challenges and limitations in using differentiated human iPSCs for in vivo disease modeling and their application for biomedical research.
Collapse
Affiliation(s)
- Nkechime Ifediora
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States
| |
Collapse
|