1
|
Hemmer BM, Ferreira AC, Philippi SM, Petridis SF, Phan A, Castellano JM. Youth-associated protein TIMP2 alters microglial state and function in the context of aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.20.655226. [PMID: 40475629 PMCID: PMC12139801 DOI: 10.1101/2025.05.20.655226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
There is little understanding of how aging serves as the strongest risk factor for several neurogenerative diseases. Specific neural cell types, such as microglia, undergo age-related maladaptive changes, including increased inflammation, impaired debris clearance, and cellular senescence, yet specific mediators that regulate these processes remain unclear. The aged brain is rejuvenated by youth-associated plasma factors, including tissue inhibitor of metalloproteinases 2 (TIMP2), which we have shown acts on the extracellular matrix (ECM) to regulate synaptic plasticity. Given emerging roles for microglia in these processes, we examined the impact of TIMP2 on microglial function. We show that TIMP2 deletion exacerbates microglial phenotypes associated with aging, including transcriptomic changes in cell activation, increased microgliosis, and increased levels of stress and inflammatory proteins measured in the brain extracellular space by in vivo microdialysis. Deleting specific cellular pools of TIMP2 in vivo increased microglial activation and altered myelin phagocytosis. Treating aged mice with TIMP2 reversed several phenotypes observed in our deletion models, resulting in decreased microglial activation, reduced proportions of proinflammatory microglia, and enhanced phagocytosis of physiological substrates. Our results identify TIMP2 as a key modulator of age-associated microglia dysfunction. Harnessing its activity may mitigate detrimental effects of age-associated insults on microglia function.
Collapse
|
2
|
Liu P, Wang Q, Wang S, Liu Y, Chen Q, Qin W, Liu X, Ye X, Jiao Y, Yuan H, Shao Z. Single-Cell RNA-Seq Reveals Aging-Related Impairment of Microglial Efferocytosis Contributing to Apoptotic Cells Accumulation After Retinal Injury. Aging Cell 2025:e70097. [PMID: 40374315 DOI: 10.1111/acel.70097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/17/2025] Open
Abstract
Aging is associated with increased retinal cell apoptosis, which contributes to decreases in retinal function. Apoptotic retinal cell clearance relies on microglial efferocytosis, but the impact of aging on this process has not been fully elucidated. In this study, we aimed to shed light on this by using single-cell RNA sequencing (sc-RNA-seq) to compare young and aged mouse retinal transcriptional profiles, in which 74,412 retinal cells from young and aged mice were classified into 10 transcriptionally distinct retinal cell types, and differentially expressed genes between young versus aged retinas were mainly associated with cellular senescence and apoptosis. Furthermore, ligand-receptor interactions (e.g., AXL-GAS6, MERTK-GAS6) between microglia and other retinal cells were strengthened in aged, compared to young retinas. Additionally, among microglia, Subcluster 4 was found under partial clustering to be associated with efferocytosis, of which aged microglia had downregulated efferocytosis-associated genes. The impact of aging on microglial efferocytosis was further verified in vitro by doxorubicin (DOX)-induced senescent BV2 microglia, and in vivo by a retinal ischemia/reperfusion (I/R) injury mouse model. In vitro, DOX-treated BV2 microglia had significantly lowered efferocytosis, as well as efferocytosis-related MerTK and Axl protein expression; this was also present in vivo in aged retinas post-I/R injury, with increased co-localization of ionized calcium-binding adapter molecule 1+ microglia with apoptotic retinal cells, along with reduced efferocytosis-related protein expression. Overall, microglial efferocytosis of apoptotic cells decreased with aging, suggesting that modulating this process could serve as a possible therapeutic target for age-related retinal diseases.
Collapse
Affiliation(s)
- Pan Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuimiao Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiqi Chen
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wanyun Qin
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinna Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinqi Ye
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yexuan Jiao
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huiping Yuan
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhengbo Shao
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Hardy J, Lewis P. Evidence suggesting that microglia make amyloid from neuronally expressed APP: a hypothesis. Mol Neurodegener 2025; 20:55. [PMID: 40346674 PMCID: PMC12065260 DOI: 10.1186/s13024-025-00847-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025] Open
Abstract
While APP is largely neuonally expressed, Aβ amyloid is largely produced by microglia as the clearance mechanisms for damaged membranes becomes overwhelmed.
Collapse
Affiliation(s)
- John Hardy
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.
- UK Dementia Research Institute at UCL, London, WC1N 3BG, UK.
- Hong Kong University of Science and Technology, Hong Kong, Hong Kong.
| | - Patrick Lewis
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, NW1 0TU, UK
| |
Collapse
|
4
|
Hours C, Vayssière P, Gressens P, Laforge M. Immunity in neuromodulation: probing neural and immune pathways in brain disorders. J Neuroinflammation 2025; 22:122. [PMID: 40296049 PMCID: PMC12038965 DOI: 10.1186/s12974-025-03440-4] [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: 02/20/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Immunity finely regulates brain function. It is directly involved in the pathological processes of neurodegenerative diseases such as Parkinson's and Alzheimer's disease, post-stroke conditions, multiple sclerosis, traumatic brain injury, and psychiatric disorders (mood disorders, major depressive disorder (MDD), anxiety disorders, psychosis disorders and schizophrenia, and neurodevelopmental disorders (NDD)). Neuromodulation is currently a leading therapeutic strategy for the treatment of these disorders, but little is yet known about its immune impact on neuronal function and its precise beneficial or harmful consequences. We review relevant clinical and preclinical studies and identify several specific immune modifications. These data not only provide insights into how neuromodulation acts to optimize immune-brain interactions, but also pave the way for a better understanding of these interactions in pathological processes.
Collapse
Affiliation(s)
- C Hours
- Université Paris Cité, NeuroDiderot, Inserm, Paris, France.
- Service de Neurochirurgie, Hôpital Fondation Adolphe de Rothschild, Paris, France.
| | - Pia Vayssière
- Service de Neurochirurgie, Hôpital Fondation Adolphe de Rothschild, Paris, France
| | - P Gressens
- Université Paris Cité, NeuroDiderot, Inserm, Paris, France
| | - M Laforge
- Université Paris Cité, NeuroDiderot, Inserm, Paris, France
| |
Collapse
|
5
|
Li F, Shi Y, Chen J, Sun J, Shi J, Sun K, Zheng B. LGR6 modulates intervertebral disc degeneration through regulation of macrophage efferocytosis. J Transl Med 2025; 23:475. [PMID: 40281518 PMCID: PMC12023656 DOI: 10.1186/s12967-025-06427-0] [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: 10/17/2024] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Intervertebral disc degeneration (IVDD) is a leading cause of chronic low back pain, characterized by extracellular matrix (ECM) degradation, excessive inflammation activation, and increased cell apoptosis. LGR6, a receptor known for its role in tissue regeneration, has recently been implicated in modulating macrophage efferocytosis, a process critical for clearing apoptotic cells and maintaining tissue homeostasis. This study aimed to investigate the role of LGR6 in regulating IVDD progression and to focus on its impact on macrophage efferocytosis, ECM regulation, and apoptosis in nucleus pulposus cells (NPCs). METHODS A comprehensive bioinformatic analysis was performed using datasets GSE56081 and GSE70362 to identify differentially expressed genes (DEGs) and gene modules associated with IVDD. Principal component analysis (PCA), volcano plots, and hierarchical clustering were utilized to assess gene expression patterns. Weighted Gene Co-Expression Network Analysis (WGCNA) was employed to identify gene modules correlated with IVDD, and integrative analysis pinpointed key genes and pathways. In vitro, LGR6 expression in macrophages was manipulated through shRNA interference and overexpression assay. The effects of LGR6 on macrophage efferocytosis, ECM synthesis, and apoptosis were assessed. An in vivo IVDD model was established in mice via disc puncture to evaluate the impact of LGR6 modulation on disc degeneration. RESULTS Bioinformatic analysis revealed distinct gene expression profiles between control and IVDD samples, with key gene modules identified by WGCNA showing strong correlations with IVDD. Integrative analysis highlighted critical pathways, including ECM-receptor interaction and efferocytosis, that are potentially regulated by several key genes including SERPINA1, THBS4, ELMO1, LGR6, and ITGB8. Of those genes, LGR6 appeared to be the gene most closely related to IVDD severity. In addition, the mRNA level and protein level of LGR6 in macrophages co-cultured with IL-1β-treated NPCs were raised significantly, compared to the control group. In vitro, LGR6 overexpression enhanced macrophage efferocytosis. Meanwhile, under co-culturing with IL-1β-treated NPCs, LGR6 overexpression in macrophages led to increased expression of ECM components such as COL2A1 and decreased expression of matrix-degrading enzymes like MMP13, indicating a protective effect against matrix degradation. Additionally, LGR6 overexpression inhibited IL-1β-induced apoptosis in NPCs by upregulating anti-apoptotic proteins (BCL2) and downregulating pro-apoptotic markers (cleaved caspase 3 and BAX). Conversely, LGR6 knockdown impaired macrophage efferocytosis and exacerbated NPCs apoptosis. In the mouse IVDD model, promoting efferocytosis resulted in improved ECM integrity and reduced apoptosis; and suppressing efferocytosis caused opposite effect, further supporting the protective role of LGR6-related efferocytosis in IVDD. CONCLUSIONS LGR6 significantly contributes to the protective effects on IVDD by modulating macrophage efferocytosis, enhancing ECM synthesis, and reducing apoptosis in NPCs. These findings highlight that LGR6 could be a promising therapeutic target for IVDD, with its dual role in regulating immune responses and preserving tissue integrity. Future studies are necessary to evaluate the clinical potential of LGR6-based therapies in treating degenerative disc diseases.
Collapse
Affiliation(s)
- Fudong Li
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Yangyang Shi
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jun Chen
- Department of Orthopaedics, Shanghai Jing'an District Zhabei Central Hospital, Shanghai, 200070, China
| | - Jingchuan Sun
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jiangang Shi
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| | - Kaiqiang Sun
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
- Department of Orthopedics, Naval Medical Center of PLA, Shanghai, 200052, China.
| | - Bing Zheng
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| |
Collapse
|
6
|
Tian A, Bhattacharya A, Muffat J, Li Y. Expanding the neuroimmune research toolkit with in vivo brain organoid technologies. Dis Model Mech 2025; 18:dmm052200. [PMID: 40231345 PMCID: PMC12032547 DOI: 10.1242/dmm.052200] [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] [Indexed: 04/16/2025] Open
Abstract
Human pluripotent stem cell-derived microglia-like cells (MLCs) and brain organoid systems have revolutionized the study of neuroimmune interactions, providing new opportunities to model human-specific brain development and disease. Over the past decade, advances in protocol design have improved the fidelity, reproducibility and scalability of MLC and brain organoid generation. Co-culturing of MLCs and brain organoids have enabled direct investigations of human microglial interactions in vitro, although opportunities remain to improve microglial maturation and long-term survival. To address these limitations, innovative xenotransplantation approaches have introduced MLCs, organoids or neuroimmune organoids into the rodent brain, providing a vascularized environment that supports prolonged development and potential behavioral readouts. These expanding in vitro and in vivo toolkits offer complementary strategies to study neuroimmune interactions in health and disease. In this Perspective, we discuss the strengths, limitations and synergies of these models, highlighting important considerations for their future applications.
Collapse
Affiliation(s)
- Ai Tian
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Afrin Bhattacharya
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Julien Muffat
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Yun Li
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
7
|
Zang H, Ji X, Yao W, Wan L, Zhang C, Zhu C, Liu T. Role of efferocytosis in chronic pain -- From molecular perspective. Neurobiol Dis 2025; 207:106857. [PMID: 40015655 DOI: 10.1016/j.nbd.2025.106857] [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: 12/27/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025] Open
Abstract
The complex nature of pain pathophysiology complicates the establishment of objective diagnostic criteria and targeted treatments. The heterogeneous manifestations of pain stemming from various primary diseases contribute to the complexity and diversity of underlying mechanisms, leading to challenges in treatment efficacy and undesirable side effects. Recent evidence suggests the presence of apoptotic cells at injury sites, the distal dorsal root ganglia (DRG), spinal cord, and certain brain regions, indicating a potential link between the ineffective clearance of dead cells and debris and pain persistence. This review highlights recent research findings indicating that efferocytosis plays a significant yet often overlooked role in lesion expansion while also representing a potentially reversible impairment that could be targeted therapeutically to mitigate chronic pain progression. We examine recent advances into how efferocytosis, a process by which phagocytes clear apoptotic cells without triggering inflammation, influences pain initiation and intensity in both human diseases and animal models. This review summarizes that efferocytosis contributes to pain progression from the perspective of defective and inefficient efferocytosis and its subsequent secondary necrocytosis, cascade inflammatory response, and the shift of phenotypic plasticity and metabolism. Additionally, we investigate the roles of newly discovered genetic alterations or modifications in biological signaling pathways in pain development and chronicity, providing insights into innovative treatment strategies that modulate efferocytosis, which are promising candidates and potential avenues for further research in pain management and prevention.
Collapse
Affiliation(s)
- Hu Zang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaoyu Ji
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Wenlong Yao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Li Wan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chuanhan Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chang Zhu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| | - Tongtong Liu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.
| |
Collapse
|
8
|
Koller EJ, McFarland KN, Angelle C, Howard J, Ryu D, Dillon KD, Erquizi A, Beheray M, De La Cruz EG, Cruz PE, Lewis J, Golde TE, Levites Y, Chakrabarty P. Antagonizing Il10 and Il4 signaling via intracerebral decoy receptor expression attenuates Aβ accumulation. Acta Neuropathol Commun 2025; 13:51. [PMID: 40055831 PMCID: PMC11887169 DOI: 10.1186/s40478-025-01968-3] [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: 11/30/2024] [Accepted: 02/20/2025] [Indexed: 05/13/2025] Open
Abstract
Multiple lines of evidence indicate that immune signaling can impact the pathological progression in Alzheimer's disease (AD), including amyloid deposition, tau aggregation, synaptic pathology and neurodegenerative trajectory. In earlier studies, we reported that intracerebral expression of the anti-inflammatory cytokines, Interleukin-10 (Il10) and Interleukin-4 (Il4), increased amyloid β (Aβ) burden in TgCRND8 mice, a preclinical model of AD-type amyloidosis. As both Interleukin-10 receptor (IL10R) and Interleukin-4 receptor (IL4R) are upregulated in an age-progressive manner in rodent models of AD and in specific regions of human AD brains, we hypothesized that a decoy receptor strategy specifically targeting Il10 and Il4 signaling could have a disease-modifying effect. We derivatized the ectodomains of mouse Il10R (sIl10R) and mouse Il4R (sIl4R) into corresponding recombinant solubilized receptor forms and delivered these intracranially into neonatal TgCRND8 mice or hippocampally into adult TgCRND8 mice with pre-existing Aβ deposits. AAV-mediated expression of sIl10R and sIl4R robustly attenuated Aβ burden in TgCRND8 mice when expressed neonatally while in the hippocampus injection cohort, AAV-sIl4R, but not sIl10R, reduced Aβ burden. sIl10R and sIl4R had opposing effects on microglial and astrocyte proliferation, with sIl10R generally reducing gliosis. RNAseq analysis showed that sIl10R likely acts as a microglial immune checkpoint inhibitor while both sIl10R and sIl4R expression show unexpected impacts on genes related to circadian rhythm. Notably, neither Il10 nor sIl10R expression altered tau pathology in two tau transgenic models, despite robust expression and impacts on glial proliferation. Together, these data reveal that decoy receptor mediated targeting of physiological Il10 or Il4 signaling can beneficially impact amyloid deposition and thus represent novel immunomodulatory approaches for AD therapy.
Collapse
Affiliation(s)
- Emily J Koller
- Department of Neuroscience, University of Florida, Gainesville, FL-32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL-32610, USA
- Medpace, 2120 West Walnut Hill Lane, Irving, TX, 75038, USA
| | - Karen N McFarland
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA
- Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Conner Angelle
- Department of Neuroscience, University of Florida, Gainesville, FL-32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL-32610, USA
| | - John Howard
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL-32610, USA
- Department of Physiological Sciences, University of Florida, Gainesville, FL-32610, USA
| | - Danny Ryu
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA
- Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Kristy D Dillon
- Department of Neuroscience, University of Florida, Gainesville, FL-32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL-32610, USA
| | - Aya Erquizi
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA
- Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Mihir Beheray
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA
- Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Elsa Gonzalez De La Cruz
- Department of Neuroscience, University of Florida, Gainesville, FL-32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL-32610, USA
| | - Pedro E Cruz
- Department of Neuroscience, University of Florida, Gainesville, FL-32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL-32610, USA
- Department of Pathology, University of Florida, Gainesville, FL-32610, USA
| | - Jada Lewis
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL-32610, USA
- Department of Physiological Sciences, University of Florida, Gainesville, FL-32610, USA
| | - Todd E Golde
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA
- Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Yona Levites
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA.
- Goizueta Brain Health Institute and Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA.
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida, Gainesville, FL-32610, USA.
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL-32610, USA.
| |
Collapse
|
9
|
Novel mRNA isoforms in human microglia refine genetic associations with neurodegeneration. Nat Genet 2025; 57:492-493. [PMID: 40055481 DOI: 10.1038/s41588-025-02112-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
|
10
|
Flury A, Aljayousi L, Park HJ, Khakpour M, Mechler J, Aziz S, McGrath JD, Deme P, Sandberg C, González Ibáñez F, Braniff O, Ngo T, Smith S, Velez M, Ramirez DM, Avnon-Klein D, Murray JW, Liu J, Parent M, Mingote S, Haughey NJ, Werneburg S, Tremblay MÈ, Ayata P. A neurodegenerative cellular stress response linked to dark microglia and toxic lipid secretion. Neuron 2025; 113:554-571.e14. [PMID: 39719704 DOI: 10.1016/j.neuron.2024.11.018] [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: 04/11/2024] [Revised: 10/22/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024]
Abstract
The brain's primary immune cells, microglia, are a leading causal cell type in Alzheimer's disease (AD). Yet, the mechanisms by which microglia can drive neurodegeneration remain unresolved. Here, we discover that a conserved stress signaling pathway, the integrated stress response (ISR), characterizes a microglia subset with neurodegenerative outcomes. Autonomous activation of ISR in microglia is sufficient to induce early features of the ultrastructurally distinct "dark microglia" linked to pathological synapse loss. In AD models, microglial ISR activation exacerbates neurodegenerative pathologies and synapse loss while its inhibition ameliorates them. Mechanistically, we present evidence that ISR activation promotes the secretion of toxic lipids by microglia, impairing neuron homeostasis and survival in vitro. Accordingly, pharmacological inhibition of ISR or lipid synthesis mitigates synapse loss in AD models. Our results demonstrate that microglial ISR activation represents a neurodegenerative phenotype, which may be sustained, at least in part, by the secretion of toxic lipids.
Collapse
Affiliation(s)
- Anna Flury
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Leen Aljayousi
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Hye-Jin Park
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | | | - Jack Mechler
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biochemistry, CUNY Graduate Center, New York, NY 10016, USA
| | - Siaresh Aziz
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Jackson D McGrath
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Michigan Medicine, Ann Arbor, MI 48105, USA
| | - Pragney Deme
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Colby Sandberg
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada
| | | | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada
| | - Thi Ngo
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Simira Smith
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Matthew Velez
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Denice Moran Ramirez
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Dvir Avnon-Klein
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - John W Murray
- Columbia Center for Human Development, Center for Stem Cell Therapies, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Martin Parent
- CERVO Brain Research Center, Québec City, QC G1E 1T2, Canada
| | - Susana Mingote
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sebastian Werneburg
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Michigan Medicine, Ann Arbor, MI 48105, USA; Michigan Neuroscience Institute, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada; Department of Molecular Medicine, Université Laval, Québec City, QC G1V 0A6, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC H3A 2B4, Canada; Canada Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 2A1, Canada; Centre for Advanced Materials and Related Technology and Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC V8N 5M8, Canada
| | - Pinar Ayata
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA; Graduate Program in Biochemistry, CUNY Graduate Center, New York, NY 10016, USA.
| |
Collapse
|
11
|
Wu X, Miller JA, Lee BTK, Wang Y, Ruedl C. Reducing microglial lipid load enhances β amyloid phagocytosis in an Alzheimer's disease mouse model. SCIENCE ADVANCES 2025; 11:eadq6038. [PMID: 39908361 PMCID: PMC11797491 DOI: 10.1126/sciadv.adq6038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025]
Abstract
Macrophages accumulate lipid droplets (LDs) under stress and inflammatory conditions. Despite the presence of LD-loaded macrophages in many tissues, including the brain, their contribution to neurodegenerative disorders remains elusive. This study investigated the role of lipid metabolism in Alzheimer's disease (AD) by assessing the contribution of LD-loaded brain macrophages, including microglia and border-associated macrophages (BAMs), in an AD mouse model. Particularly, BAMs and activated CD11c+ microglia localized near β amyloid (Aβ) plaques exhibited a pronounced lipid-associated gene signature and a high LD load. Having observed that elevated intracellular LD content correlated inversely with microglial phagocytic activities, we subsequently inhibited LD formation specifically in CX3CR1+ brain macrophages using an inducible APP-KI/Fit2iΔMφ transgenic mouse model. We demonstrated that reducing LD content in microglia and CX3CR1+ BAMs remarkably improved their phagocytic ability. Furthermore, lowering microglial LDs consistently enhanced their efferocytosis capacities and notably reduced Aβ deposition in the brain parenchyma. Therefore, mitigating LD accumulation in brain macrophages provides perspectives for AD treatment.
Collapse
Affiliation(s)
- Xiaoting Wu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - James Alastair Miller
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Bernett Teck Kwong Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yulan Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
12
|
Sewell M, Fialova N, Montagne A. Unraveling the transcriptomic landscape of brain vascular cells in dementia: A systematic review. Alzheimers Dement 2025; 21:e14512. [PMID: 39807599 PMCID: PMC11851133 DOI: 10.1002/alz.14512] [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: 10/10/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Cerebrovascular dysfunction plays a critical role in the pathogenesis of dementia and related neurodegenerative disorders. Recent omics-driven research has revealed associations between vascular abnormalities and transcriptomic alterations in brain vascular cells, particularly endothelial cells (ECs) and pericytes (PCs). However, the impact of these molecular changes on dementia remains unclear. METHODS We conducted a comparative analysis of gene expression in ECs and PCs across neurodegenerative conditions, including Alzheimer's disease (AD), Huntington's disease, and arteriovenous malformation, utilizing transcriptomic data from published postmortem human tissue studies. RESULTS We identified differentially expressed genes (DEGs) consistently dysregulated in ECs and PCs across these pathologies. Notably, several DEGs are linked to vascular cell zonation and genetic risks for AD and cerebral small vessel disease. DISCUSSION Our findings provide insights into the cellular and molecular mechanisms underlying vascular dysfunction in dementia, highlight the knowledge gaps, and suggest potential novel vascular therapeutic targets, including genes not previously investigated in this context. HIGHLIGHTS Systematic review of differentially expressed genes (DEGs) in vascular cells from neurodegenerative single-nuclear RNA-sequencing (snRNA-seq) studies. Identify overlapping DEGs in multiple vascular cell types across studies. Examine functional relevance and associations with genetic risk for common DEGs. Outline future directions for the vascular omics field.
Collapse
Affiliation(s)
- Michael Sewell
- UK Dementia Research Institute at the University of EdinburghEdinburghUK
- British Heart Foundation ‐ UK Dementia Research Institute Centre for Vascular Dementia Research at the University of EdinburghEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Nela Fialova
- UK Dementia Research Institute at the University of EdinburghEdinburghUK
- British Heart Foundation ‐ UK Dementia Research Institute Centre for Vascular Dementia Research at the University of EdinburghEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Axel Montagne
- UK Dementia Research Institute at the University of EdinburghEdinburghUK
- British Heart Foundation ‐ UK Dementia Research Institute Centre for Vascular Dementia Research at the University of EdinburghEdinburghUK
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
13
|
Guo JL, Braun D, Fitzgerald GA, Hsieh YT, Rougé L, Litvinchuk A, Steffek M, Propson NE, Heffner CM, Discenza C, Han SJ, Rana A, Skuja LL, Lin BQ, Sun EW, Davis SS, Balasundar S, Becerra I, Dugas JC, Ha C, Hsiao-Nakamoto J, Huang F, Jain S, Kung JE, Liau NPD, Mahon CS, Nguyen HN, Nguyen N, Samaddar M, Shi Y, Tatarakis D, Tian Y, Zhu Y, Suh JH, Sandmann T, Calvert MEK, Arguello A, Kane LA, Lewcock JW, Holtzman DM, Koth CM, Di Paolo G. Decreased lipidated ApoE-receptor interactions confer protection against pathogenicity of ApoE and its lipid cargoes in lysosomes. Cell 2025; 188:187-206.e26. [PMID: 39532095 PMCID: PMC11724755 DOI: 10.1016/j.cell.2024.10.027] [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: 01/05/2024] [Revised: 07/08/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
While apolipoprotein E (APOE) is the strongest genetic modifier for late-onset Alzheimer's disease (LOAD), the molecular mechanisms underlying isoform-dependent risk and the relevance of ApoE-associated lipids remain elusive. Here, we report that impaired low-density lipoprotein (LDL) receptor (LDLR) binding of lipidated ApoE2 (lipApoE2) avoids LDLR recycling defects observed with lipApoE3/E4 and decreases the uptake of cholesteryl esters (CEs), which are lipids linked to neurodegeneration. In human neurons, the addition of ApoE carrying polyunsaturated fatty acids (PUFAs)-CE revealed an allelic series (ApoE4 > ApoE3 > ApoE2) associated with lipofuscinosis, an age-related lysosomal pathology resulting from lipid peroxidation. Lipofuscin increased lysosomal accumulation of tau fibrils and was elevated in the APOE4 mouse brain with exacerbation by tau pathology. Intrahippocampal injection of PUFA-CE-lipApoE4 was sufficient to induce lipofuscinosis in wild-type mice. Finally, the protective Christchurch mutation also reduced LDLR binding and phenocopied ApoE2. Collectively, our data strongly suggest decreased lipApoE-LDLR interactions minimize LOAD risk by reducing the deleterious effects of endolysosomal targeting of ApoE and associated pathogenic lipids.
Collapse
Affiliation(s)
- Jing L Guo
- Denali Therapeutics Inc., South San Francisco, CA, USA.
| | - Dylan Braun
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | - Lionel Rougé
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Alexandra Litvinchuk
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Micah Steffek
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | - Suk Ji Han
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Anil Rana
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Lukas L Skuja
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Bi Qi Lin
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | | | - Jason C Dugas
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Connie Ha
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Fen Huang
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Shourya Jain
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | | | - Nathan Nguyen
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Yajuan Shi
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - Yuxi Tian
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Yuda Zhu
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | | | | | - Lesley A Kane
- Denali Therapeutics Inc., South San Francisco, CA, USA
| | | | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | |
Collapse
|
14
|
Yuan X, Gao L, Wang H, Wang J. A pathway-informed disease-related gene identification approach and its application to screen novel risk genes for Alzheimer's disease. J Alzheimers Dis Rep 2025; 9:25424823251343812. [PMID: 40406678 PMCID: PMC12095958 DOI: 10.1177/25424823251343812] [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: 02/10/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025] Open
Abstract
Background Alzheimer's disease (AD) is a severe neurodegenerative disorder, yet its molecular mechanisms remain incompletely understood. It is known that the joint action of a number of genetic and other factors is involved in the pathogenesis of this disorder. Objective In the past years, extensive research has focused on identifying novel AD predictors and genetic markers. However, our understanding of the molecular features of AD is still incomplete, and it is essential to discover novel genes and their interaction involved in the etiology and development of AD. Methods Here, we developed GRESA (Gene RElationship Sequence Analyzer), to predict novel genes related to disease by a combination of machine learning algorithm Skip-gram and Monte Carlo Tree Search (MCTS). First, we extracted gene association information contained in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database and generated gene relationship sequences. Then, Skip-gram neural network was trained and combined with MCTS to predict genes potentially associated with diseases like AD. Results The performance of GRESA was evaluated on breast cancer and gastric cancer. We further constructed the gene network underlying AD pathogenesis via GRESA, by which 164 potential gene candidates were predicted and their molecular function and biological features were investigated. Conclusions Results from this study provided insights for understanding the molecular feature underlying AD. As a useful systematic method, our approach can also be applied to construct gene networks for other complex diseases.
Collapse
Affiliation(s)
- Xin Yuan
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Lihui Gao
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Hao Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Ju Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| |
Collapse
|
15
|
Navarro E, Montesinos J. Mitochondria-Associated Endoplasmic Reticulum Membranes in Microglia: One Contact Site to Rule Them all. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2025; 8:25152564241312807. [PMID: 39881949 PMCID: PMC11775980 DOI: 10.1177/25152564241312807] [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: 09/24/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025]
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in maintaining tissue homeostasis by monitoring and responding to environmental changes through processes such as phagocytosis, cytokine production or synapse remodeling. Their dynamic nature and diverse functions are supported by the regulation of multiple metabolic pathways, enabling microglia to efficiently adapt to fluctuating signals. A key aspect of this regulation occurs at mitochondria-associated ER membranes (MAM), specialized contact sites between the ER and mitochondria. These structures facilitate the exchange of calcium, lipids, and metabolites and serve as metabolic and signaling hubs. This review synthesizes current research on how MAM influence microglial physiology, with an emphasis on their role in immunometabolism, offering new insights into the integration of metabolic and immune functions in the CNS and its impact in the context of neurodegeneration.
Collapse
Affiliation(s)
- Elisa Navarro
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Neurochemistry Research Institute, Complutense University of Madrid, Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Jorge Montesinos
- Department of Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040 Madrid, Spain
| |
Collapse
|
16
|
Navarro E, Esteras N. Multitarget Effects of Nrf2 Signalling in the Brain: Common and Specific Functions in Different Cell Types. Antioxidants (Basel) 2024; 13:1502. [PMID: 39765831 PMCID: PMC11673142 DOI: 10.3390/antiox13121502] [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: 11/20/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a crucial regulator of cellular defence mechanisms, essential for maintaining the brain's health. Nrf2 supports mitochondrial function and protects against oxidative damage, which is vital for meeting the brain's substantial energy and antioxidant demands. Furthermore, Nrf2 modulates glial inflammatory responses, playing a pivotal role in preventing neuroinflammation. This review explores these multifaceted functions of Nrf2 within the central nervous system, focusing on its activity across various brain cell types, including neurons, astrocytes, microglia, and oligodendrocytes. Due to the brain's vulnerability to oxidative stress and metabolic challenges, Nrf2 is emerging as a key therapeutic target to enhance resilience against oxidative stress, inflammation, mitochondrial dysfunction, and demyelination, which are central to many neurodegenerative diseases.
Collapse
Affiliation(s)
- Elisa Navarro
- Neurochemistry Research Institute, Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Noemí Esteras
- Neurochemistry Research Institute, Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28040 Madrid, Spain
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
17
|
Podlesny-Drabiniok A, Romero-Molina C, Patel T, See WY, Liu Y, Marcora E, Goate AM. Cytokine-induced reprogramming of human macrophages toward Alzheimer's disease-relevant molecular and cellular phenotypes in vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.619910. [PMID: 39554174 PMCID: PMC11565805 DOI: 10.1101/2024.10.24.619910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Myeloid cells including brain-resident (microglia) and peripheral macrophages play a crucial role in various pathological conditions, including neurodegenerative disorders like Alzheimer's disease (AD). They respond to disruption of tissue homeostasis associated with disease conditions by acquiring various transcriptional and functional states. Experimental investigation of these states is hampered by the lack of tools that enable accessible and robust reprogramming of human macrophages toward Alzheimer's disease-relevant molecular and cellular phenotypes in vitro. In this study, we investigated the ability of a cytokine mix, including interleukin-4 (IL4), colony stimulating factor 1 (CSF1/MCSF), interleukin 34 (IL34) and transforming growth factor beta (TGFβ), to induce reprogramming of cultured human THP-1 macrophages. Our results indicate this treatment led to significant transcriptomic changes, driving THP-1 macrophages towards a transcriptional state reminiscent of disease-associated microglia (DAM) and lipid-associated macrophages (LAM) collectively referred to as DLAM. Transcriptome profiling revealed gene expression changes related to oxidative phosphorylation, lysosome function, and lipid metabolism. Single-cell RNA sequencing revealed an increased proportion of DLAM clusters in cytokine mix-treated THP-1 macrophages. Functional assays demonstrated alterations in cell motility, phagocytosis, lysosomal activity, and metabolic and energetic profiles. Our findings provide insights into the cytokine-mediated reprogramming of macrophages towards disease-relevant states, highlighting their role in neurodegenerative diseases and potential for therapeutic development.
Collapse
Affiliation(s)
- Anna Podlesny-Drabiniok
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Carmen Romero-Molina
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Tulsi Patel
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Wen Yi See
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Yiyuan Liu
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Edoardo Marcora
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Alison M. Goate
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
18
|
Kloske CM, Belloy ME, Blue EE, Bowman GR, Carrillo MC, Chen X, Chiba‐Falek O, Davis AA, Paolo GD, Garretti F, Gate D, Golden LR, Heinecke JW, Herz J, Huang Y, Iadecola C, Johnson LA, Kanekiyo T, Karch CM, Khvorova A, Koppes‐den Hertog SJ, Lamb BT, Lawler PE, Guen YL, Litvinchuk A, Liu C, Mahinrad S, Marcora E, Marino C, Michaelson DM, Miller JJ, Morganti JM, Narayan PS, Naslavsky MS, Oosthoek M, Ramachandran KV, Ramakrishnan A, Raulin A, Robert A, Saleh RNM, Sexton C, Shah N, Shue F, Sible IJ, Soranno A, Strickland MR, TCW J, Thierry M, Tsai L, Tuckey RA, Ulrich JD, van der Kant R, Wang N, Wellington CL, Weninger SC, Yassine HN, Zhao N, Bu G, Goate AM, Holtzman DM. Advancements in APOE and dementia research: Highlights from the 2023 AAIC Advancements: APOE conference. Alzheimers Dement 2024; 20:6590-6605. [PMID: 39031528 PMCID: PMC11497726 DOI: 10.1002/alz.13877] [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: 02/02/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 07/22/2024]
Abstract
INTRODUCTION The apolipoprotein E gene (APOE) is an established central player in the pathogenesis of Alzheimer's disease (AD), with distinct apoE isoforms exerting diverse effects. apoE influences not only amyloid-beta and tau pathologies but also lipid and energy metabolism, neuroinflammation, cerebral vascular health, and sex-dependent disease manifestations. Furthermore, ancestral background may significantly impact the link between APOE and AD, underscoring the need for more inclusive research. METHODS In 2023, the Alzheimer's Association convened multidisciplinary researchers at the "AAIC Advancements: APOE" conference to discuss various topics, including apoE isoforms and their roles in AD pathogenesis, progress in apoE-targeted therapeutic strategies, updates on disease models and interventions that modulate apoE expression and function. RESULTS This manuscript presents highlights from the conference and provides an overview of opportunities for further research in the field. DISCUSSION Understanding apoE's multifaceted roles in AD pathogenesis will help develop targeted interventions for AD and advance the field of AD precision medicine. HIGHLIGHTS APOE is a central player in the pathogenesis of Alzheimer's disease. APOE exerts a numerous effects throughout the brain on amyloid-beta, tau, and other pathways. The AAIC Advancements: APOE conference encouraged discussions and collaborations on understanding the role of APOE.
Collapse
Affiliation(s)
| | - Michael E. Belloy
- Department of Neurology and Neurological SciencesStanford University, StanfordPalo AltoCaliforniaUSA
- NeuroGenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
- Department of NeurologyWashington University School of Medicine, St. Louis, MissouriSt. LouisMissouriUSA
| | - Elizabeth E. Blue
- Division of Medical GeneticsDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
- Institute for Public Health GeneticsUniversity of WashingtonSeattleWashingtonUSA
| | - Gregory R. Bowman
- Departments of Biochemistry & Biophysics and BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - Xiaoying Chen
- Department of NeurologyHope Center for Neurological DisordersKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Ornit Chiba‐Falek
- Division of Translational Brain SciencesDepartment of NeurologyDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Albert A. Davis
- Department of Neurology Hope Center for Neurological Disorders Washington University School of MedicineSt. LouisMissouriUSA
| | | | - Francesca Garretti
- Ronald M. Loeb Center for Alzheimer's DiseaseNew YorkNew YorkUSA
- Department of Genetics & Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - David Gate
- The Ken & Ruth Davee Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Lesley R. Golden
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Jay W. Heinecke
- Department of MedicineUniversity of Washington, UV MedicineSeattleWashingtonUSA
| | - Joachim Herz
- Center for Translational Neurodegeneration ResearchUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Yadong Huang
- Gladstone Institute of Neurological DiseaseGladstone InstitutesSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research InstituteWeill Cornell MedicineNew YorkNew YorkUSA
| | - Lance A. Johnson
- Department of PhysiologyUniversity of KentuckyLexingtonKentuckyUSA
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
| | - Takahisa Kanekiyo
- Department of NeuroscienceMayo Clinic JacksonvilleJacksonvilleFloridaUSA
| | - Celeste M. Karch
- Department of PsychiatryWashington University in St LouisSt. LouisMissouriUSA
| | - Anastasia Khvorova
- RNA Therapeutic InstituteUMass Chan Medical SchoolWorcesterMassachusettsUSA
| | - Sascha J. Koppes‐den Hertog
- Department of Functional GenomicsCenter for Neurogenomics and Cognitive Research (CNCR)VU University AmsterdamAmsterdamUSA
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam Neuroscience, Amsterdam University Medical CenterAmsterdamUSA
| | - Bruce T. Lamb
- Stark Neurosciences Research Institute Indiana University School of MedicineIndianapolisIndianaUSA
| | - Paige E. Lawler
- Department of NeurologyWashington University School of Medicine, St. Louis, MissouriSt. LouisMissouriUSA
- The Tracy Family SILQ CenterWashington University School of MedicineIndianapolisIndianaUSA
| | - Yann Le Guen
- Department of Neurology and Neurological SciencesStanford UniversityPalo AltoCaliforniaUSA
- Institut du Cerveau–Paris Brain Institute–ICMParisFrance
| | - Alexandra Litvinchuk
- Department of NeurologyHope Center for Neurological DisordersKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Chia‐Chen Liu
- Department of NeuroscienceMayo Clinic JacksonvilleJacksonvilleFloridaUSA
| | | | - Edoardo Marcora
- Department of Genetics and Genomic SciencesNash Family Department of NeuroscienceIcahn Genomics Institute; Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Claudia Marino
- Schepens Eye Research Institute of Mass Eye and Ear and Department of Ophthalmology at Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Justin J. Miller
- Departments of Biochemistry & Biophysics and BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biochemistry and Molecular BiophysicsWashington University School of MedicineSt. LouisMissouriUSA
| | - Josh M. Morganti
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKentuckyUSA
- Department of NeuroscienceUniversity of KentuckyLexingtonKentuckyUSA
| | - Priyanka S. Narayan
- Genetics and Biochemistry BranchNational Institute of Diabetes and Digestive and Kidney DiseasesNational Institute of Neurological Disorders and StrokeCenter for Alzheimer's and Related Dementias (CARD)National Institutes of HealthMarylandUSA
| | - Michel S. Naslavsky
- Human Genome and Stem‐cell Research CenterBiosciences InstituteUniversity of São PauloRua do MataoSão PauloBrazil
- Hospital Israelita Albert EinsteinAvenida Albert EinsteinSão PauloBrazil
| | - Marlies Oosthoek
- Neurochemistry LaboratoryDepartment of Laboratory MedicineVrije Universiteit Amsterdam, Amsterdam UMCAmsterdamNetherlands
| | - Kapil V. Ramachandran
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Department of NeuroscienceColumbia University Vagelos College of Physicians and SurgeonsNew YorkUSA
| | - Abhirami Ramakrishnan
- The Ken & Ruth Davee Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | | | - Aiko Robert
- Department of Functional GenomicsCenter for Neurogenomics and Cognitive Research (CNCR)VU University AmsterdamAmsterdamUSA
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam Neuroscience, Amsterdam University Medical CenterAmsterdamUSA
| | - Rasha N. M. Saleh
- Norwich Medical SchoolUniversity of East Anglia, UK Clinical and Chemical PathologyNorfolkUK
- Faculty of MedicineAlexandria UniversityAlexandria GovernorateEgypt
| | | | | | | | | | - Andrea Soranno
- Washington University in Saint Louis, St. Louis, Missouri, USASt. LouisMissouriUSA
| | - Michael R. Strickland
- Department of NeurologyWashington University School of Medicine, St. Louis, MissouriSt. LouisMissouriUSA
| | - Julia TCW
- Department of PharmacologyPhysiology & BiophysicsChobanian and Avedisian School of MedicineBoston UniversityBostonMassachusettsUSA
- Bioinformatics ProgramFaculty of Computing & Data SciencesBoston UniversityBostonMassachusettsUSA
| | - Manon Thierry
- Center for Cognitive NeurologyDepartment of NeurologyNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Li‐Huei Tsai
- Picower Institute for Learning and MemoryDepartment of Brain and Cognitive SciencesMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Ryan A. Tuckey
- Department of NeurologyCenter for Neurodegeneration and Experimental TherapeuticsMedical Scientist Training ProgramUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Jason D. Ulrich
- Department of NeurologyHope Center for Neurological DisordersKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Rik van der Kant
- Department of Functional GenomicsCenter for Neurogenomics and Cognitive Research (CNCR)VU University AmsterdamAmsterdamUSA
- Alzheimer Center AmsterdamDepartment of NeurologyAmsterdam Neuroscience, Amsterdam University Medical CenterAmsterdamUSA
| | - Na Wang
- Mayo Clinic RochesterRochesterMinnesotaUSA
| | - Cheryl L. Wellington
- Djavad Mowafaghian Centre for Brain Health Department of Pathology and Laboratory Medicine International Collaboration on Repair Discoveries School of Biomedical Engineering University of British ColumbiaVancouverCanada
| | | | - Hussein N. Yassine
- Department of NeurologyKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Na Zhao
- Department of NeuroscienceMayo Clinic JacksonvilleJacksonvilleFloridaUSA
| | - Guojun Bu
- Division of Life ScienceHong Kong University of Science and TechnologyClear Water BayKowloonHong Kong
| | - Alison M. Goate
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's diseaseIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - David M. Holtzman
- Department of NeurologyHope Center for Neurological DisordersKnight Alzheimer's Disease Research CenterWashington University School of MedicineSt. LouisMissouriUSA
| |
Collapse
|
19
|
Kouri N, Frankenhauser I, Peng Z, Labuzan SA, Boon BDC, Moloney CM, Pottier C, Wickland DP, Caetano-Anolles K, Corriveau-Lecavalier N, Tranovich JF, Wood AC, Hinkle KM, Lincoln SJ, Spychalla AJ, Senjem ML, Przybelski SA, Engelberg-Cook E, Schwarz CG, Kwan RS, Lesser ER, Crook JE, Carter RE, Ross OA, Lachner C, Ertekin-Taner N, Ferman TJ, Fields JA, Machulda MM, Ramanan VK, Nguyen AT, Reichard RR, Jones DT, Graff-Radford J, Boeve BF, Knopman DS, Petersen RC, Jack CR, Kantarci K, Day GS, Duara R, Graff-Radford NR, Dickson DW, Lowe VJ, Vemuri P, Murray ME. Clinicopathologic Heterogeneity and Glial Activation Patterns in Alzheimer Disease. JAMA Neurol 2024; 81:619-629. [PMID: 38619853 PMCID: PMC11019448 DOI: 10.1001/jamaneurol.2024.0784] [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: 10/17/2023] [Accepted: 01/05/2024] [Indexed: 04/16/2024]
Abstract
Importance Factors associated with clinical heterogeneity in Alzheimer disease (AD) lay along a continuum hypothesized to associate with tangle distribution and are relevant for understanding glial activation considerations in therapeutic advancement. Objectives To examine clinicopathologic and neuroimaging characteristics of disease heterogeneity in AD along a quantitative continuum using the corticolimbic index (CLix) to account for individuality of spatially distributed tangles found at autopsy. Design, Setting, and Participants This cross-sectional study was a retrospective medical record review performed on the Florida Autopsied Multiethnic (FLAME) cohort accessioned from 1991 to 2020. Data were analyzed from December 2022 to December 2023. Structural magnetic resonance imaging (MRI) and tau positron emission tomography (PET) were evaluated in an independent neuroimaging group. The FLAME cohort includes 2809 autopsied individuals; included in this study were neuropathologically diagnosed AD cases (FLAME-AD). A digital pathology subgroup of FLAME-AD cases was derived for glial activation analyses. Main Outcomes and Measures Clinicopathologic factors of heterogeneity that inform patient history and neuropathologic evaluation of AD; CLix score (lower, relative cortical predominance/hippocampal sparing vs higher, relative cortical sparing/limbic predominant cases); neuroimaging measures (ie, structural MRI and tau-PET). Results Of the 2809 autopsied individuals in the FLAME cohort, 1361 neuropathologically diagnosed AD cases were evaluated. A digital pathology subgroup included 60 FLAME-AD cases. The independent neuroimaging group included 93 cases. Among the 1361 FLAME-AD cases, 633 were male (47%; median [range] age at death, 81 [54-96] years) and 728 were female (53%; median [range] age at death, 81 [53-102] years). A younger symptomatic onset (Spearman ρ = 0.39, P < .001) and faster decline on the Mini-Mental State Examination (Spearman ρ = 0.27; P < .001) correlated with a lower CLix score in FLAME-AD series. Cases with a nonamnestic syndrome had lower CLix scores (median [IQR], 13 [9-18]) vs not (median [IQR], 21 [15-27]; P < .001). Hippocampal MRI volume (Spearman ρ = -0.45; P < .001) and flortaucipir tau-PET uptake in posterior cingulate and precuneus cortex (Spearman ρ = -0.74; P < .001) inversely correlated with CLix score. Although AD cases with a CLix score less than 10 had higher cortical tangle count, we found lower percentage of CD68-activated microglia/macrophage burden (median [IQR], 0.46% [0.32%-0.75%]) compared with cases with a CLix score of 10 to 30 (median [IQR], 0.75% [0.51%-0.98%]) and on par with a CLix score of 30 or greater (median [IQR], 0.40% [0.32%-0.57%]; P = .02). Conclusions and Relevance Findings show that AD heterogeneity exists along a continuum of corticolimbic tangle distribution. Reduced CD68 burden may signify an underappreciated association between tau accumulation and microglia/macrophages activation that should be considered in personalized therapy for immune dysregulation.
Collapse
Affiliation(s)
- Naomi Kouri
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Isabelle Frankenhauser
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
- Paracelsus Medical Private University, Salzburg, Austria
| | - Zhongwei Peng
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | | | | | | | - Cyril Pottier
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Daniel P. Wickland
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | | | - Nick Corriveau-Lecavalier
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | - Ashley C. Wood
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Kelly M. Hinkle
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | | | | | | | - Scott A. Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | | | | | - Rain S. Kwan
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Elizabeth R. Lesser
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Julia E. Crook
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Rickey E. Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Christian Lachner
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, Florida
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
- Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | - Tanis J. Ferman
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, Florida
| | - Julie A. Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
| | - Mary M. Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
| | | | - Aivi T. Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - David T. Jones
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | - Ranjan Duara
- Wien Center for Alzheimer’s Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, Florida
| | | | | | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | |
Collapse
|
20
|
Sarazin M, Lagarde J, El Haddad I, de Souza LC, Bellier B, Potier MC, Bottlaender M, Dorothée G. The path to next-generation disease-modifying immunomodulatory combination therapies in Alzheimer's disease. NATURE AGING 2024; 4:761-770. [PMID: 38839924 DOI: 10.1038/s43587-024-00630-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/09/2024] [Indexed: 06/07/2024]
Abstract
The cautious optimism following recent anti-amyloid therapeutic trials for Alzheimer's disease (AD) provides a glimmer of hope after years of disappointment. Although these encouraging results represent discernible progress, they also highlight the need to enhance further the still modest clinical efficacy of current disease-modifying immunotherapies. Here, we highlight crucial milestones essential for advancing precision medicine in AD. These include reevaluating the choice of therapeutic targets by considering the key role of both central neuroinflammation and peripheral immunity in disease pathogenesis, refining patient stratification by further defining the inflammatory component within the forthcoming ATN(I) (amyloid, tau and neurodegeneration (and inflammation)) classification of AD biomarkers and defining more accurate clinical outcomes and prognostic biomarkers that better reflect disease heterogeneity. Next-generation immunotherapies will need to go beyond the current antibody-only approach by simultaneously targeting pathological proteins together with innate neuroinflammation and/or peripheral-central immune crosstalk. Such innovative immunomodulatory combination therapy approaches should be evaluated in appropriately redesigned clinical therapeutic trials, which must carefully integrate the neuroimmune component.
Collapse
Affiliation(s)
- Marie Sarazin
- Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte-Anne, Paris, France.
- Université Paris-Cité, Paris, France.
- Université Paris-Saclay, BioMaps, Service Hospitalier Frédéric Joliot, CEA, CNRS, Inserm, Orsay, France.
| | - Julien Lagarde
- Department of Neurology of Memory and Language, GHU Paris Psychiatrie & Neurosciences, Hôpital Sainte-Anne, Paris, France
- Université Paris-Cité, Paris, France
- Université Paris-Saclay, BioMaps, Service Hospitalier Frédéric Joliot, CEA, CNRS, Inserm, Orsay, France
| | - Inès El Haddad
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, Paris, France
| | - Leonardo Cruz de Souza
- Grupo de Pesquisa em Neurologia Cognitiva e do Comportamento, Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Programa de Pós-Graduação em Neurociências, UFMG, Belo Horizonte, Brazil
- Departamento de Clínica Médica, Faculdade de Medicina, UFMG, Belo Horizonte, Brazil
| | - Bertrand Bellier
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, Paris, France
| | - Marie-Claude Potier
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Michel Bottlaender
- Université Paris-Saclay, BioMaps, Service Hospitalier Frédéric Joliot, CEA, CNRS, Inserm, Orsay, France
- Université Paris-Saclay, UNIACT, Neurospin, Joliot Institute, CEA, Gif-sur-Yvette, France
| | - Guillaume Dorothée
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Immune System and Neuroinflammation Laboratory, Hôpital Saint-Antoine, Paris, France.
| |
Collapse
|
21
|
Sheng Y, Hu W, Chen S, Zhu X. Efferocytosis by macrophages in physiological and pathological conditions: regulatory pathways and molecular mechanisms. Front Immunol 2024; 15:1275203. [PMID: 38779685 PMCID: PMC11109379 DOI: 10.3389/fimmu.2024.1275203] [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: 08/09/2023] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
Efferocytosis is defined as the highly effective phagocytic removal of apoptotic cells (ACs) by professional or non-professional phagocytes. Tissue-resident professional phagocytes ("efferocytes"), such as macrophages, have high phagocytic capacity and are crucial to resolve inflammation and aid in homeostasis. Recently, numerous exciting discoveries have revealed divergent (and even diametrically opposite) findings regarding metabolic immune reprogramming associated with efferocytosis by macrophages. In this review, we highlight the key metabolites involved in the three phases of efferocytosis and immune reprogramming of macrophages under physiological and pathological conditions. The next decade is expected to yield further breakthroughs in the regulatory pathways and molecular mechanisms connecting immunological outcomes to metabolic cues as well as avenues for "personalized" therapeutic intervention.
Collapse
Affiliation(s)
- Yan−Ran Sheng
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Wen−Ting Hu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Siman Chen
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiao−Yong Zhu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Ahmed H, Wang Y, Griffiths WJ, Levey AI, Pikuleva I, Liang SH, Haider A. Brain cholesterol and Alzheimer's disease: challenges and opportunities in probe and drug development. Brain 2024; 147:1622-1635. [PMID: 38301270 PMCID: PMC11068113 DOI: 10.1093/brain/awae028] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 12/20/2023] [Accepted: 01/13/2024] [Indexed: 02/03/2024] Open
Abstract
Cholesterol homeostasis is impaired in Alzheimer's disease; however, attempts to modulate brain cholesterol biology have not translated into tangible clinical benefits for patients to date. Several recent milestone developments have substantially improved our understanding of how excess neuronal cholesterol contributes to the pathophysiology of Alzheimer's disease. Indeed, neuronal cholesterol was linked to the formation of amyloid-β and neurofibrillary tangles through molecular pathways that were recently delineated in mechanistic studies. Furthermore, remarkable advances in translational molecular imaging have now made it possible to probe cholesterol metabolism in the living human brain with PET, which is an important prerequisite for future clinical trials that target the brain cholesterol machinery in Alzheimer's disease patients-with the ultimate aim being to develop disease-modifying treatments. This work summarizes current concepts of how the biosynthesis, transport and clearance of brain cholesterol are affected in Alzheimer's disease. Further, current strategies to reverse these alterations by pharmacotherapy are critically discussed in the wake of emerging translational research tools that support the assessment of brain cholesterol biology not only in animal models but also in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Hazem Ahmed
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, 8093 Zurich, Switzerland
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - William J Griffiths
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Irina Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ahmed Haider
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
23
|
Benzow K, Karanjeet K, Oblak AL, Carter GW, Sasner M, Koob MD. Gene replacement-Alzheimer's disease (GR-AD): Modeling the genetics of human dementias in mice. Alzheimers Dement 2024; 20:3080-3087. [PMID: 38343132 PMCID: PMC11032548 DOI: 10.1002/alz.13730] [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: 11/20/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Genetic studies conducted over the past four decades have provided us with a detailed catalog of genes that play critical roles in the etiology of Alzheimer's disease (AD) and related dementias (ADRDs). Despite this progress, as a field we have had only limited success in incorporating this rich complexity of human AD/ADRD genetics findings into our animal models of these diseases. Our primary goal for the gene replacement (GR)-AD project is to develop mouse lines that model the genetics of AD/ADRD as closely as possible. METHODS To do this, we are generating mouse lines in which the genes of interest are precisely and completely replaced in the mouse genome by their full human orthologs. RESULTS Each model set consists of a control line with a wild-type human allele and variant lines that precisely match the human genomic sequence in the control line except for a high-impact pathogenic mutation or risk variant.
Collapse
Affiliation(s)
- Kellie Benzow
- Laboratory Medicine and Pathology, and Institute for Translational Neuroscience University of MinnesotaMinneapolisMinnesotaUSA
| | - Kul Karanjeet
- Laboratory Medicine and Pathology, and Institute for Translational Neuroscience University of MinnesotaMinneapolisMinnesotaUSA
| | | | | | | | - Michael D. Koob
- Laboratory Medicine and Pathology, and Institute for Translational Neuroscience University of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
24
|
Guan X, Wang Y, Li W, Mu W, Tang Y, Wang M, Seyam A, Yang Y, Pan L, Hou T. The Role of Macrophage Efferocytosis in the Pathogenesis of Apical Periodontitis. Int J Mol Sci 2024; 25:3854. [PMID: 38612664 PMCID: PMC11011522 DOI: 10.3390/ijms25073854] [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: 02/24/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Macrophages (Mφs) play a crucial role in the homeostasis of the periapical immune micro-environment caused by bacterial infection. Mφ efferocytosis has been demonstrated to promote the resolution of multiple infected diseases via accelerating Mφ polarization into M2 type. However, the Mφ efferocytosis-apical periodontitis (AP) relationship has not been elucidated yet. This study aimed to explore the role of Mφ efferocytosis in the pathogenesis of AP. Clinical specimens were collected to determine the involvement of Mφ efferocytosis in the periapical region via immunohistochemical and immunofluorescence staining. For a further understanding of the moderator effect of Mφ efferocytosis in the pathogenesis of AP, both an in vitro AP model and in vivo AP model were treated with ARA290, a Mφ efferocytosis agonist. Histological staining, micro-ct, flow cytometry, RT-PCR and Western blot analysis were performed to detect the inflammatory status, alveolar bone loss and related markers in AP models. The data showed that Mφ efferocytosis is observed in the periapical tissues and enhancing the Mφ efferocytosis ability could effectively promote AP resolution via facilitating M2 Mφ polarization. Collectively, our study demonstrates the functional importance of Mφ efferocytosis in AP pathology and highlights that accelerating Mφ efferocytosis via ARA290 could serve as an adjuvant therapeutic strategy for AP.
Collapse
Affiliation(s)
- Xiaoyue Guan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| | - Yuting Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| | - Wenlan Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| | - Wenli Mu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| | - Yifei Tang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| | - Mingfei Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| | - Abdelrahman Seyam
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| | - Yao Yang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| | - Lifei Pan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| | - Tiezhou Hou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China (A.S.)
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
- Department of Cariology and Endodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| |
Collapse
|
25
|
Podleśny-Drabiniok A, Novikova G, Liu Y, Dunst J, Temizer R, Giannarelli C, Marro S, Kreslavsky T, Marcora E, Goate AM. BHLHE40/41 regulate microglia and peripheral macrophage responses associated with Alzheimer's disease and other disorders of lipid-rich tissues. Nat Commun 2024; 15:2058. [PMID: 38448474 PMCID: PMC10917780 DOI: 10.1038/s41467-024-46315-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Genetic and experimental evidence suggests that Alzheimer's disease (AD) risk alleles and genes may influence disease susceptibility by altering the transcriptional and cellular responses of macrophages, including microglia, to damage of lipid-rich tissues like the brain. Recently, sc/nRNA sequencing studies identified similar transcriptional activation states in subpopulations of macrophages in aging and degenerating brains and in other diseased lipid-rich tissues. We collectively refer to these subpopulations of microglia and peripheral macrophages as DLAMs. Using macrophage sc/nRNA-seq data from healthy and diseased human and mouse lipid-rich tissues, we reconstructed gene regulatory networks and identified 11 strong candidate transcriptional regulators of the DLAM response across species. Loss or reduction of two of these transcription factors, BHLHE40/41, in iPSC-derived microglia and human THP-1 macrophages as well as loss of Bhlhe40/41 in mouse microglia, resulted in increased expression of DLAM genes involved in cholesterol clearance and lysosomal processing, increased cholesterol efflux and storage, and increased lysosomal mass and degradative capacity. These findings provide targets for therapeutic modulation of macrophage/microglial function in AD and other disorders affecting lipid-rich tissues.
Collapse
Affiliation(s)
- Anna Podleśny-Drabiniok
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gloriia Novikova
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- OMNI Bioinformatics Department, Genentech, Inc., South San Francisco, CA, USA
| | - Yiyuan Liu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Josefine Dunst
- Department of Medicine, Division of Immunology and Allergy, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rose Temizer
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chiara Giannarelli
- Department of Medicine, Division of Cardiology, NYU Cardiovascular Research Center, New York University School of Medicine, New York, NY, USA
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Samuele Marro
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Taras Kreslavsky
- Department of Medicine, Division of Immunology and Allergy, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Edoardo Marcora
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Alison Mary Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
26
|
García-Culebras A, Cuartero MI, Peña-Martínez C, Moraga A, Vázquez-Reyes S, de Castro-Millán FJ, Cortes-Canteli M, Lizasoain I, Moro MÁ. Myeloid cells in vascular dementia and Alzheimer's disease: Possible therapeutic targets? Br J Pharmacol 2024; 181:777-798. [PMID: 37282844 DOI: 10.1111/bph.16159] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/10/2023] [Accepted: 05/20/2023] [Indexed: 06/08/2023] Open
Abstract
Growing evidence supports the suggestion that the peripheral immune system plays a role in different pathologies associated with cognitive impairment, such as vascular dementia (VD) or Alzheimer's disease (AD). The aim of this review is to summarize, within the peripheral immune system, the implications of different types of myeloid cells in AD and VD, with a special focus on post-stroke cognitive impairment and dementia (PSCID). We will review the contributions of the myeloid lineage, from peripheral cells (neutrophils, platelets, monocytes and monocyte-derived macrophages) to central nervous system (CNS)-associated cells (perivascular macrophages and microglia). Finally, we will evaluate different potential strategies for pharmacological modulation of pathological processes mediated by myeloid cell subsets, with an emphasis on neutrophils, their interaction with platelets and the process of immunothrombosis that triggers neutrophil-dependent capillary stall and hypoperfusion, as possible effector mechanisms that may pave the way to novel therapeutic avenues to stop dementia, the epidemic of our time. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
- Alicia García-Culebras
- Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Departamento de Biología Celular, Facultad de Medicina, UCM, Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, UCM, Madrid, Spain
| | - María Isabel Cuartero
- Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, UCM, Madrid, Spain
| | - Carolina Peña-Martínez
- Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, UCM, Madrid, Spain
| | - Ana Moraga
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Departamento de Biología Celular, Facultad de Medicina, UCM, Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, UCM, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Sandra Vázquez-Reyes
- Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, UCM, Madrid, Spain
| | - Francisco Javier de Castro-Millán
- Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, UCM, Madrid, Spain
| | - Marta Cortes-Canteli
- Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, UCM, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - María Ángeles Moro
- Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, UCM, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
27
|
Romero-Molina C, Neuner SM, Ryszawiec M, Pébay A, Dominantly Inherited Alzheimer Network, Marcora E, Goate A. Autosomal Dominant Alzheimer's Disease Mutations in Human Microglia Are Not Sufficient to Trigger Amyloid Pathology in WT Mice but Might Affect Pathology in 5XFAD Mice. Int J Mol Sci 2024; 25:2565. [PMID: 38473822 PMCID: PMC10932392 DOI: 10.3390/ijms25052565] [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: 01/30/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 03/14/2024] Open
Abstract
Several genetic variants that affect microglia function have been identified as risk factors for Alzheimer's Disease (AD), supporting the importance of this cell type in disease progression. However, the effect of autosomal dominant mutations in the amyloid precursor protein (APP) or the presenilin (PSEN1/2) genes has not been addressed in microglia in vivo. We xenotransplanted human microglia derived from non-carriers and carriers of autosomal dominant AD (ADAD)-causing mutations in the brain of hCSF1 WT or 5XFAD mice. We observed that ADAD mutations in microglia are not sufficient to trigger amyloid pathology in WT mice. In 5XFAD mice, we observed a non-statistically significant increase in amyloid plaque volume and number of dystrophic neurites, coupled with a reduction in plaque-associated microglia in the brain of mice xenotransplanted with ADAD human microglia compared to mice xenotransplanted with non-ADAD microglia. In addition, we observed a non-statistically significant impairment in working and contextual memory in 5XFAD mice xenotransplanted with ADAD microglia compared to those xenotransplanted with non-ADAD-carrier microglia. We conclude that, although not sufficient to initiate amyloid pathology in the healthy brain, mutations in APP and PSEN1 in human microglia might cause mild changes in pathological and cognitive outcomes in 5XFAD mice in a manner consistent with increased AD risk.
Collapse
Affiliation(s)
- Carmen Romero-Molina
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| | - Sarah M. Neuner
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| | - Marcelina Ryszawiec
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| | - Alice Pébay
- Department of Anatomy and Physiology, Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3010, Australia;
| | | | - Edoardo Marcora
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| | - Alison Goate
- Ronald M. Loeb Center for Alzheimer’s Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA; (C.R.-M.); (S.M.N.); (M.R.); (E.M.)
| |
Collapse
|
28
|
Litvinchuk A, Suh JH, Guo JL, Lin K, Davis SS, Bien-Ly N, Tycksen E, Tabor GT, Remolina Serrano J, Manis M, Bao X, Lee C, Bosch M, Perez EJ, Yuede CM, Cashikar AG, Ulrich JD, Di Paolo G, Holtzman DM. Amelioration of Tau and ApoE4-linked glial lipid accumulation and neurodegeneration with an LXR agonist. Neuron 2024; 112:384-403.e8. [PMID: 37995685 PMCID: PMC10922706 DOI: 10.1016/j.neuron.2023.10.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Apolipoprotein E (APOE) is a strong genetic risk factor for late-onset Alzheimer's disease (LOAD). APOE4 increases and APOE2 decreases risk relative to APOE3. In the P301S mouse model of tauopathy, ApoE4 increases tau pathology and neurodegeneration when compared with ApoE3 or the absence of ApoE. However, the role of ApoE isoforms and lipid metabolism in contributing to tau-mediated degeneration is unknown. We demonstrate that in P301S tau mice, ApoE4 strongly promotes glial lipid accumulation and perturbations in cholesterol metabolism and lysosomal function. Increasing lipid efflux in glia via an LXR agonist or Abca1 overexpression strongly attenuates tau pathology and neurodegeneration in P301S/ApoE4 mice. We also demonstrate reductions in reactive astrocytes and microglia, as well as changes in cholesterol biosynthesis and metabolism in glia of tauopathy mice in response to LXR activation. These data suggest that promoting efflux of glial lipids may serve as a therapeutic approach to ameliorate tau and ApoE4-linked neurodegeneration.
Collapse
Affiliation(s)
- Alexandra Litvinchuk
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Jing L Guo
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Karin Lin
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Sonnet S Davis
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Nga Bien-Ly
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Eric Tycksen
- Genome Technology Access Center, McDonnell Genome Institute, St. Louis, MO 63110, USA
| | - G Travis Tabor
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Javier Remolina Serrano
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Melissa Manis
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Xin Bao
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Choonghee Lee
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Megan Bosch
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Enmanuel J Perez
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Carla M Yuede
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Anil G Cashikar
- Department of Psychiatry, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jason D Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | | | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA.
| |
Collapse
|
29
|
Gorijala P, Aslam MM, Dang LT, Xicota L, Fernandez MV, Sung YJ, Fan K, Feingold E, Surace EI, Chhatwal JP, Hom CL, Dominantly Inherited Alzheimer Network (DIAN), the Alzheimer's Disease Neuroimaging Initiative (ADNI), the NIA‐LOAD family study, for the Alzheimer's Biomarkers Consortium–Down Syndrome (ABC‐DS) Investigators, Hartley SL, Hassenstab J, Perrin RJ, Mapstone M, Zaman SH, Ances BM, Kamboh MI, Lee JH, Cruchaga C. Alzheimer's polygenic risk scores are associated with cognitive phenotypes in Down syndrome. Alzheimers Dement 2024; 20:1038-1049. [PMID: 37855447 PMCID: PMC10916941 DOI: 10.1002/alz.13506] [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: 05/03/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
INTRODUCTION This study aimed to investigate the influence of the overall Alzheimer's disease (AD) genetic architecture on Down syndrome (DS) status, cognitive measures, and cerebrospinal fluid (CSF) biomarkers. METHODS AD polygenic risk scores (PRS) were tested for association with DS-related traits. RESULTS The AD risk PRS was associated with disease status in several cohorts of sporadic late- and early-onset and familial late-onset AD, but not in familial early-onset AD or DS. On the other hand, lower DS Mental Status Examination memory scores were associated with higher PRS, independent of intellectual disability and APOE (PRS including APOE, PRSAPOE , p = 2.84 × 10-4 ; PRS excluding APOE, PRSnonAPOE , p = 1.60 × 10-2 ). PRSAPOE exhibited significant associations with Aβ42, tTau, pTau, and Aβ42/40 ratio in DS. DISCUSSION These data indicate that the AD genetic architecture influences cognitive and CSF phenotypes in DS adults, supporting common pathways that influence memory decline in both traits. HIGHLIGHTS Examination of the polygenic risk of AD in DS presented here is the first of its kind. AD PRS influences memory aspects in DS individuals, independently of APOE genotype. These results point to an overlap between the genes and pathways that leads to AD and those that influence dementia and memory decline in the DS population. APOE ε4 is linked to DS cognitive decline, expanding cognitive insights in adults.
Collapse
Affiliation(s)
- Priyanka Gorijala
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
- Neurogenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - M. Muaaz Aslam
- Department of Human GeneticsUniversity of PittsburghSchool of Public HealthPittsburghPennsylvaniaUSA
| | - Lam‐Ha T. Dang
- Department of EpidemiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Sergievsky CenterTaub Institute for Research on Alzheimer's Disease and the Aging Brainand Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - L. Xicota
- Sergievsky CenterTaub Institute for Research on Alzheimer's Disease and the Aging Brainand Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Maria V. Fernandez
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
- Neurogenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
| | - Yun Ju Sung
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
- Neurogenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
- Division of BiostatisticsWashington University School of MedicineSt. LouisMissouriUSA
| | - Kang‐Hsien Fan
- Department of Human GeneticsUniversity of PittsburghSchool of Public HealthPittsburghPennsylvaniaUSA
| | - Eleanor Feingold
- Department of Human GeneticsUniversity of PittsburghSchool of Public HealthPittsburghPennsylvaniaUSA
| | - Ezequiel I. Surace
- Laboratory of Neurodegenerative Diseases ‐ Institute of Neurosciences (INEU‐Fleni‐ CONICET)Buenos AiresArgentina
| | - Jasmeer P Chhatwal
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Christy L. Hom
- Dept. of Psychiatry and Human BehaviorUniversity of CaliforniaIrvine School of MedicineCaliforniaUSA
| | | | | | - Sigan L. Hartley
- Waisman Center and School of Human EcologyUniversity of Wisconsin‐ MadisonMadisonWisconsinUSA
| | - Jason Hassenstab
- Department of Neurology and Psychological & Brain SciencesWashington UniversitySt. LouisMissouriUSA
| | - Richard J. Perrin
- Hope Center for Neurologic DiseasesWashington UniversitySt. LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Mark Mapstone
- Department of NeurologyUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Shahid H Zaman
- Cambridge Intellectual and Developmental Disabilities Research GroupDepartment of PsychiatryUniversity of CambridgeDouglas HouseCambridgeUK
- Cambridgeshire and Peterborough NHS Foundation TrustElizabeth HouseFulbourn HospitalFulbournCambridgeUK
| | - Beau M Ances
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - M. Ilyas Kamboh
- Department of Human GeneticsUniversity of PittsburghSchool of Public HealthPittsburghPennsylvaniaUSA
| | - Joseph H Lee
- Department of EpidemiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
- Sergievsky CenterTaub Institute for Research on Alzheimer's Disease and the Aging Brainand Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
- Neurogenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
- Hope Center for Neurologic DiseasesWashington UniversitySt. LouisMissouriUSA
| |
Collapse
|
30
|
Abstract
Cells can die as a consequence of being phagocytosed by other cells - a form of cell death that has been called phagotrophy, cell cannibalism, programmed cell removal and primary phagocytosis. However, these are all different manifestations of cell death by phagocytosis (termed 'phagoptosis' for short). The engulfed cells die as a result of cytotoxic oxidants, peptides and degradative enzymes within acidic phagolysosomes. Cell death by phagocytosis was discovered by Metchnikov in the 1880s, but was neglected until recently. It is now known to contribute to developmental cell death in nematodes, Drosophila and mammals, and is central to innate and adaptive immunity against pathogens. Cell death by phagocytosis mediates physiological turnover of erythrocytes and other leucocytes, making it the most abundant form of cell death in the mammalian body. Immunity against cancer is also partly mediated by macrophage phagocytosis of cancer cells, but cancer cells can also phagocytose host cells and other cancer cells in order to survive. Recent evidence indicates neurodegeneration and other neuropathologies can be mediated by microglial phagocytosis of stressed neurons. Thus, despite cell death by phagocytosis being poorly recognized, it is one of the oldest, commonest and most important forms of cell death.
Collapse
Affiliation(s)
- Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
31
|
Fisher DW, Dunn JT, Dong H. Distinguishing features of depression in dementia from primary psychiatric disease. DISCOVER MENTAL HEALTH 2024; 4:3. [PMID: 38175420 PMCID: PMC10767128 DOI: 10.1007/s44192-023-00057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024]
Abstract
Depression is a common and devastating neuropsychiatric symptom in the elderly and in patients with dementia. In particular, nearly 80% of patients with Alzheimer's Disease dementia experience depression during disease development and progression. However, it is unknown whether the depression in patients with dementia shares the same molecular mechanisms as depression presenting as primary psychiatric disease or occurs and persists through alternative mechanisms. In this review, we discuss how the clinical presentation and treatment differ between depression in dementia and as a primary psychiatric disease, with a focus on major depressive disorder. Then, we hypothesize several molecular mechanisms that may be unique to depression in dementia such as neuropathological changes, inflammation, and vascular events. Finally, we discuss existing issues and future directions for investigation and treatment of depression in dementia.
Collapse
Affiliation(s)
- Daniel W Fisher
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL, 60611, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, 1959 NE Pacific Street, Box 356560, Seattle, WA, 98195, USA
| | - Jeffrey T Dunn
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL, 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL, 60611, USA.
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL, 60611, USA.
| |
Collapse
|
32
|
Moon HS, Mahzarnia A, Stout J, Anderson RJ, Strain M, Tremblay JT, Han ZY, Niculescu A, MacFarlane A, King J, Ashley-Koch A, Clark D, Lutz MW, Badea A. Multivariate investigation of aging in mouse models expressing the Alzheimer's protective APOE2 allele: integrating cognitive metrics, brain imaging, and blood transcriptomics. Brain Struct Funct 2024; 229:231-249. [PMID: 38091051 PMCID: PMC11082910 DOI: 10.1007/s00429-023-02731-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/03/2023] [Indexed: 01/31/2024]
Abstract
APOE allelic variation is critical in brain aging and Alzheimer's disease (AD). The APOE2 allele associated with cognitive resilience and neuroprotection against AD remains understudied. We employed a multipronged approach to characterize the transition from middle to old age in mice with APOE2 allele, using behavioral assessments, image-derived morphometry and diffusion metrics, structural connectomics, and blood transcriptomics. We used sparse multiple canonical correlation analyses (SMCCA) for integrative modeling, and graph neural network predictions. Our results revealed brain sub-networks associated with biological traits, cognitive markers, and gene expression. The cingulate cortex emerged as a critical region, demonstrating age-associated atrophy and diffusion changes, with higher fractional anisotropy in males and middle-aged subjects. Somatosensory and olfactory regions were consistently highlighted, indicating age-related atrophy and sex differences. The hippocampus exhibited significant volumetric changes with age, with differences between males and females in CA3 and CA1 regions. SMCCA underscored changes in the cingulate cortex, somatosensory cortex, olfactory regions, and hippocampus in relation to cognition and blood-based gene expression. Our integrative modeling in aging APOE2 carriers revealed a central role for changes in gene pathways involved in localization and the negative regulation of cellular processes. Our results support an important role of the immune system and response to stress. This integrative approach offers novel insights into the complex interplay among brain connectivity, aging, and sex. Our study provides a foundation for understanding the impact of APOE2 allele on brain aging, the potential for detecting associated changes in blood markers, and revealing novel therapeutic intervention targets.
Collapse
Affiliation(s)
- Hae Sol Moon
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Quantitative Imaging and Analysis Laboratory, Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Ali Mahzarnia
- Quantitative Imaging and Analysis Laboratory, Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Jacques Stout
- Brain Imaging and Analysis Center, Duke University School of Medicine, Durham, NC, USA
| | - Robert J Anderson
- Quantitative Imaging and Analysis Laboratory, Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Madison Strain
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Jessica T Tremblay
- Quantitative Imaging and Analysis Laboratory, Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Zay Yar Han
- Quantitative Imaging and Analysis Laboratory, Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Andrei Niculescu
- Quantitative Imaging and Analysis Laboratory, Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Anna MacFarlane
- Department of Neuroscience, Duke University, Durham, NC, USA
| | - Jasmine King
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Allison Ashley-Koch
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Darin Clark
- Quantitative Imaging and Analysis Laboratory, Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Michael W Lutz
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Alexandra Badea
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Quantitative Imaging and Analysis Laboratory, Department of Radiology, Duke University School of Medicine, Durham, NC, USA.
- Brain Imaging and Analysis Center, Duke University School of Medicine, Durham, NC, USA.
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
33
|
Tijms BM, Vromen EM, Mjaavatten O, Holstege H, Reus LM, van der Lee S, Wesenhagen KEJ, Lorenzini L, Vermunt L, Venkatraghavan V, Tesi N, Tomassen J, den Braber A, Goossens J, Vanmechelen E, Barkhof F, Pijnenburg YAL, van der Flier WM, Teunissen CE, Berven FS, Visser PJ. Cerebrospinal fluid proteomics in patients with Alzheimer's disease reveals five molecular subtypes with distinct genetic risk profiles. NATURE AGING 2024; 4:33-47. [PMID: 38195725 PMCID: PMC10798889 DOI: 10.1038/s43587-023-00550-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/29/2023] [Indexed: 01/11/2024]
Abstract
Alzheimer's disease (AD) is heterogenous at the molecular level. Understanding this heterogeneity is critical for AD drug development. Here we define AD molecular subtypes using mass spectrometry proteomics in cerebrospinal fluid, based on 1,058 proteins, with different levels in individuals with AD (n = 419) compared to controls (n = 187). These AD subtypes had alterations in protein levels that were associated with distinct molecular processes: subtype 1 was characterized by proteins related to neuronal hyperplasticity; subtype 2 by innate immune activation; subtype 3 by RNA dysregulation; subtype 4 by choroid plexus dysfunction; and subtype 5 by blood-brain barrier impairment. Each subtype was related to specific AD genetic risk variants, for example, subtype 1 was enriched with TREM2 R47H. Subtypes also differed in clinical outcomes, survival times and anatomical patterns of brain atrophy. These results indicate molecular heterogeneity in AD and highlight the need for personalized medicine.
Collapse
Affiliation(s)
- Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands.
| | - Ellen M Vromen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Olav Mjaavatten
- Proteomics Unit at the University of Bergen, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Henne Holstege
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Clinical Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Lianne M Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sven van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Kirsten E J Wesenhagen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Luigi Lorenzini
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neuroimaging, Amsterdam, the Netherlands
| | - Lisa Vermunt
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Department of Laboratory Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Vikram Venkatraghavan
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Niccoló Tesi
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
| | - Jori Tomassen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | | | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Epidemiology & Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Neurochemistry Laboratory, Department of Laboratory Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Frode S Berven
- Proteomics Unit at the University of Bergen, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Schlepckow K, Morenas-Rodríguez E, Hong S, Haass C. Stimulation of TREM2 with agonistic antibodies-an emerging therapeutic option for Alzheimer's disease. Lancet Neurol 2023; 22:1048-1060. [PMID: 37863592 DOI: 10.1016/s1474-4422(23)00247-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 10/22/2023]
Abstract
Neurodegenerative disorders, including Alzheimer's disease, are associated with microgliosis. Microglia have long been considered to have detrimental roles in Alzheimer's disease. However, functional analyses of genes encoding risk factors that are linked to late-onset Alzheimer's disease, and that are enriched or exclusively expressed in microglia, have revealed unexpected protective functions. One of the major risk genes for Alzheimer's disease is TREM2. Risk variants of TREM2 are loss-of-function mutations affecting chemotaxis, phagocytosis, lipid and energy metabolism, and survival and proliferation. Agonistic anti-TREM2 antibodies have been developed to boost these protective functions in patients with intact TREM2 alleles. Several anti-TREM2 antibodies are in early clinical trials, and current efforts aim to achieve more efficient transport of these antibodies across the blood-brain barrier. PET imaging could be used to monitor target engagement. Data from animal models, and biomarker studies in patients, further support a rationale for boosting TREM2 functions during the preclinical stage of Alzheimer's disease.
Collapse
Affiliation(s)
- Kai Schlepckow
- German Centre for Neurodegenerative Diseases, Munich, Germany
| | - Estrella Morenas-Rodríguez
- Memory Unit, Department of Neurology, Hospital Universitario 12 de Octubre, Madrid, Spain; Group of Neurogenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Christian Haass
- German Centre for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Centre, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
35
|
Essayan-Perez S, Südhof TC. Neuronal γ-secretase regulates lipid metabolism, linking cholesterol to synaptic dysfunction in Alzheimer's disease. Neuron 2023; 111:3176-3194.e7. [PMID: 37543038 PMCID: PMC10592349 DOI: 10.1016/j.neuron.2023.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/16/2023] [Accepted: 07/10/2023] [Indexed: 08/07/2023]
Abstract
Presenilin mutations that alter γ-secretase activity cause familial Alzheimer's disease (AD), whereas ApoE4, an apolipoprotein for cholesterol transport, predisposes to sporadic AD. Both sporadic and familial AD feature synaptic dysfunction. Whether γ-secretase is involved in cholesterol metabolism and whether such involvement impacts synaptic function remains unknown. Here, we show that in human neurons, chronic pharmacological or genetic suppression of γ-secretase increases synapse numbers but decreases synaptic transmission by lowering the presynaptic release probability without altering dendritic or axonal arborizations. In search of a mechanism underlying these synaptic impairments, we discovered that chronic γ-secretase suppression robustly decreases cholesterol levels in neurons but not in glia, which in turn stimulates neuron-specific cholesterol-synthesis gene expression. Suppression of cholesterol levels by HMG-CoA reductase inhibitors (statins) impaired synaptic function similar to γ-secretase inhibition. Thus, γ-secretase enables synaptic function by maintaining cholesterol levels, whereas the chronic suppression of γ-secretase impairs synapses by lowering cholesterol levels.
Collapse
Affiliation(s)
- Sofia Essayan-Perez
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
36
|
Gamache J, Gingerich D, Shwab EK, Barrera J, Garrett ME, Hume C, Crawford GE, Ashley-Koch AE, Chiba-Falek O. Integrative single-nucleus multi-omics analysis prioritizes candidate cis and trans regulatory networks and their target genes in Alzheimer's disease brains. Cell Biosci 2023; 13:185. [PMID: 37789374 PMCID: PMC10546724 DOI: 10.1186/s13578-023-01120-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/30/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND The genetic underpinnings of late-onset Alzheimer's disease (LOAD) are yet to be fully elucidated. Although numerous LOAD-associated loci have been discovered, the causal variants and their target genes remain largely unknown. Since the brain is composed of heterogenous cell subtypes, it is imperative to study the brain on a cell subtype specific level to explore the biological processes underlying LOAD. METHODS Here, we present the largest parallel single-nucleus (sn) multi-omics study to simultaneously profile gene expression (snRNA-seq) and chromatin accessibility (snATAC-seq) to date, using nuclei from 12 normal and 12 LOAD brains. We identified cell subtype clusters based on gene expression and chromatin accessibility profiles and characterized cell subtype-specific LOAD-associated differentially expressed genes (DEGs), differentially accessible peaks (DAPs) and cis co-accessibility networks (CCANs). RESULTS Integrative analysis defined disease-relevant CCANs in multiple cell subtypes and discovered LOAD-associated cell subtype-specific candidate cis regulatory elements (cCREs), their candidate target genes, and trans-interacting transcription factors (TFs), some of which, including ELK1, JUN, and SMAD4 in excitatory neurons, were also LOAD-DEGs. Finally, we focused on a subset of cell subtype-specific CCANs that overlap known LOAD-GWAS regions and catalogued putative functional SNPs changing the affinities of TF motifs within LOAD-cCREs linked to LOAD-DEGs, including APOE and MYO1E in a specific subtype of microglia and BIN1 in a subpopulation of oligodendrocytes. CONCLUSIONS To our knowledge, this study represents the most comprehensive systematic interrogation to date of regulatory networks and the impact of genetic variants on gene dysregulation in LOAD at a cell subtype resolution. Our findings reveal crosstalk between epigenetic, genomic, and transcriptomic determinants of LOAD pathogenesis and define catalogues of candidate genes, cCREs, and variants involved in LOAD genetic etiology and the cell subtypes in which they act to exert their pathogenic effects. Overall, these results suggest that cell subtype-specific cis-trans interactions between regulatory elements and TFs, and the genes dysregulated by these networks contribute to the development of LOAD.
Collapse
Affiliation(s)
- Julia Gamache
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, DUMC Box 2900, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Daniel Gingerich
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, DUMC Box 2900, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - E Keats Shwab
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, DUMC Box 2900, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Julio Barrera
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, DUMC Box 2900, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, DUMC Box 104775, Durham, NC, 27701, USA
| | - Cordelia Hume
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, DUMC Box 2900, Durham, NC, 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Gregory E Crawford
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA.
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical Center, DUMC Box 3382, Durham, NC, 27708, USA.
- Center for Advanced Genomic Technologies, Duke University Medical Center, Durham, NC, 27708, USA.
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, DUMC Box 104775, Durham, NC, 27701, USA.
- Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA.
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, DUMC Box 2900, Durham, NC, 27710, USA.
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA.
| |
Collapse
|
37
|
Signal B, Pérez Suárez TG, Taberlay PC, Woodhouse A. Cellular specificity is key to deciphering epigenetic changes underlying Alzheimer's disease. Neurobiol Dis 2023; 186:106284. [PMID: 37683959 DOI: 10.1016/j.nbd.2023.106284] [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: 05/01/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Different cell types in the brain play distinct roles in Alzheimer's disease (AD) progression. Late onset AD (LOAD) is a complex disease, with a large genetic component, but many risk loci fall in non-coding genome regions. Epigenetics implicates the non-coding genome with control of gene expression. The epigenome is highly cell-type specific and dynamically responds to the environment. Therefore, epigenetic mechanisms are well placed to explain genetic and environmental factors that are associated with AD. However, given this cellular specificity, purified cell populations or single cells need to be profiled to avoid effect masking. Here we review the current state of cell-type specific genome-wide profiling in LOAD, covering DNA methylation (CpG, CpH, and hydroxymethylation), histone modifications, and chromatin changes. To date, these data reveal that distinct cell types contribute and react differently to AD progression through epigenetic alterations. This review addresses the current gap in prior bulk-tissue derived work by spotlighting cell-specific changes that govern the complex interplay of cells throughout disease progression and are critical in understanding and developing effective treatments for AD.
Collapse
Affiliation(s)
- Brandon Signal
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia.
| | | | - Phillippa C Taberlay
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
38
|
Pesämaa I, Müller SA, Robinson S, Darcher A, Paquet D, Zetterberg H, Lichtenthaler SF, Haass C. A microglial activity state biomarker panel differentiates FTD-granulin and Alzheimer's disease patients from controls. Mol Neurodegener 2023; 18:70. [PMID: 37775827 PMCID: PMC10543321 DOI: 10.1186/s13024-023-00657-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/13/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND With the emergence of microglia-modulating therapies there is an urgent need for reliable biomarkers to evaluate microglial activation states. METHODS Using mouse models and human induced pluripotent stem cell-derived microglia (hiMGL), genetically modified to yield the most opposite homeostatic (TREM2-knockout) and disease-associated (GRN-knockout) states, we identified microglia activity-dependent markers. Non-targeted mass spectrometry was used to identify proteomic changes in microglia and cerebrospinal fluid (CSF) of Grn- and Trem2-knockout mice. Additionally, we analyzed the proteome of GRN- and TREM2-knockout hiMGL and their conditioned media. Candidate marker proteins were tested in two independent patient cohorts, the ALLFTD cohort (GRN mutation carriers versus non-carriers), as well as the proteomic data set available from the EMIF-AD MBD study. RESULTS We identified proteomic changes between the opposite activation states in mouse microglia and CSF, as well as in hiMGL cell lysates and conditioned media. For further verification, we analyzed the CSF proteome of heterozygous GRN mutation carriers suffering from frontotemporal dementia (FTD). We identified a panel of six proteins (FABP3, MDH1, GDI1, CAPG, CD44, GPNMB) as potential indicators for microglial activation. Moreover, we confirmed three of these proteins (FABP3, GDI1, MDH1) to be significantly elevated in the CSF of Alzheimer's (AD) patients. Remarkably, each of these markers differentiated amyloid-positive cases with mild cognitive impairment (MCI) from amyloid-negative individuals. CONCLUSIONS The identified candidate proteins reflect microglia activity and may be relevant for monitoring the microglial response in clinical practice and clinical trials modulating microglial activity and amyloid deposition. Moreover, the finding that three of these markers differentiate amyloid-positive from amyloid-negative MCI cases in the AD cohort suggests that these proteins associate with a very early immune response to seeded amyloid. This is consistent with our previous findings in the Dominantly Inherited Alzheimer's Disease Network (DIAN) cohort, where soluble TREM2 increases as early as 21 years before symptom onset. Moreover, in mouse models for amyloidogenesis, seeding of amyloid is limited by physiologically active microglia further supporting their early protective role. The biological functions of some of our main candidates (FABP3, CD44, GPNMB) also further emphasize that lipid dysmetabolism may be a common feature of neurodegenerative disorders.
Collapse
Affiliation(s)
- Ida Pesämaa
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Psychiatry & Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Sophie Robinson
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Ludwig-Maximilians-University Munich, Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital Munich, Ludwig-Maximilians- University Munich, Munich, Germany
| | - Alana Darcher
- Epileptology, University Hospital Bonn, Bonn, Germany
| | - Dominik Paquet
- Institute for Stroke and Dementia Research, University Hospital Munich, Ludwig-Maximilians- University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| | - Henrik Zetterberg
- Department of Psychiatry & Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany.
- Biomedical Centre (BMC), Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
39
|
Sirkis DW, Yokoyama JS. Role for cell death pathway in Alzheimer's disease. Science 2023; 381:1156-1157. [PMID: 37708264 DOI: 10.1126/science.adk2009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Human neurons transplanted into mice with amyloid plaques die by necroptosis.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
40
|
Tsai AP, Dong C, Lin PBC, Oblak AL, Viana Di Prisco G, Wang N, Hajicek N, Carr AJ, Lendy EK, Hahn O, Atkins M, Foltz AG, Patel J, Xu G, Moutinho M, Sondek J, Zhang Q, Mesecar AD, Liu Y, Atwood BK, Wyss-Coray T, Nho K, Bissel SJ, Lamb BT, Landreth GE. Genetic variants of phospholipase C-γ2 alter the phenotype and function of microglia and confer differential risk for Alzheimer's disease. Immunity 2023; 56:2121-2136.e6. [PMID: 37659412 PMCID: PMC10564391 DOI: 10.1016/j.immuni.2023.08.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/20/2023] [Accepted: 08/14/2023] [Indexed: 09/04/2023]
Abstract
Genetic association studies have demonstrated the critical involvement of the microglial immune response in Alzheimer's disease (AD) pathogenesis. Phospholipase C-gamma-2 (PLCG2) is selectively expressed by microglia and functions in many immune receptor signaling pathways. In AD, PLCG2 is induced uniquely in plaque-associated microglia. A genetic variant of PLCG2, PLCG2P522R, is a mild hypermorph that attenuates AD risk. Here, we identified a loss-of-function PLCG2 variant, PLCG2M28L, that confers an increased AD risk. PLCG2P522R attenuated disease in an amyloidogenic murine AD model, whereas PLCG2M28L exacerbated the plaque burden associated with altered phagocytosis and Aβ clearance. The variants bidirectionally modulated disease pathology by inducing distinct transcriptional programs that identified microglial subpopulations associated with protective or detrimental phenotypes. These findings identify PLCG2M28L as a potential AD risk variant and demonstrate that PLCG2 variants can differentially orchestrate microglial responses in AD pathogenesis that can be therapeutically targeted.
Collapse
Affiliation(s)
- Andy P Tsai
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Chuanpeng Dong
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Peter Bor-Chian Lin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adrian L Oblak
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gonzalo Viana Di Prisco
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nian Wang
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nicole Hajicek
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam J Carr
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emma K Lendy
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Oliver Hahn
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Micaiah Atkins
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Aulden G Foltz
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Jheel Patel
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Guixiang Xu
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Miguel Moutinho
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John Sondek
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Qisheng Zhang
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew D Mesecar
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brady K Atwood
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Kwangsik Nho
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephanie J Bissel
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bruce T Lamb
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gary E Landreth
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
41
|
Self WK, Holtzman DM. Emerging diagnostics and therapeutics for Alzheimer disease. Nat Med 2023; 29:2187-2199. [PMID: 37667136 DOI: 10.1038/s41591-023-02505-2] [Citation(s) in RCA: 146] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/18/2023] [Indexed: 09/06/2023]
Abstract
Alzheimer disease (AD) is the most common contributor to dementia in the world, but strategies that slow or prevent its clinical progression have largely remained elusive, until recently. This Review highlights the latest advances in biomarker technologies and therapeutic development to improve AD diagnosis and treatment. We review recent results that enable pathological staging of AD with neuroimaging and fluid-based biomarkers, with a particular emphasis on the role of amyloid, tau and neuroinflammation in disease pathogenesis. We discuss the lessons learned from randomized controlled trials, including some supporting the proposal that certain anti-amyloid antibodies slow cognitive decline during the mildly symptomatic phase of AD. In addition, we highlight evidence for newly identified therapeutic targets that may be able to modify AD pathogenesis and progression. Collectively, these recent discoveries-and the research directions that they open-have the potential to move AD clinical care toward disease-modifying treatment strategies with maximal benefits for patients.
Collapse
Affiliation(s)
- Wade K Self
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
42
|
Lutz MW, Chiba-Falek O. Bioinformatics pipeline to guide post-GWAS studies in Alzheimer's: A new catalogue of disease candidate short structural variants. Alzheimers Dement 2023; 19:4094-4109. [PMID: 37253165 PMCID: PMC10524333 DOI: 10.1002/alz.13168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/27/2023] [Accepted: 05/08/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Short structural variants (SSVs), including insertions/deletions (indels), are common in the human genome and impact disease risk. The role of SSVs in late-onset Alzheimer's disease (LOAD) has been understudied. In this study, we developed a bioinformatics pipeline of SSVs within LOAD-genome-wide association study (GWAS) regions to prioritize regulatory SSVs based on the strength of their predicted effect on transcription factor (TF) binding sites. METHODS The pipeline utilized publicly available functional genomics data sources including candidate cis-regulatory elements (cCREs) from ENCODE and single-nucleus (sn)RNA-seq data from LOAD patient samples. RESULTS We catalogued 1581 SSVs in candidate cCREs in LOAD GWAS regions that disrupted 737 TF sites. That included SSVs that disrupted the binding of RUNX3, SPI1, and SMAD3, within the APOE-TOMM40, SPI1, and MS4A6A LOAD regions. CONCLUSIONS The pipeline developed here prioritized non-coding SSVs in cCREs and characterized their putative effects on TF binding. The approach integrates multiomics datasets for validation experiments using disease models.
Collapse
Affiliation(s)
- Michael W. Lutz
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
43
|
Comerota MM, Gedam M, Xiong W, Jin F, Deng L, Wang MC, Wang J, Zheng H. Oleoylethanolamide facilitates PPARα and TFEB signaling and attenuates Aβ pathology in a mouse model of Alzheimer's disease. Mol Neurodegener 2023; 18:56. [PMID: 37580742 PMCID: PMC10426131 DOI: 10.1186/s13024-023-00648-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/08/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Age is the strongest risk factor for the development of Alzheimer's disease (AD). Besides the pathological hallmarks of β-amyloid (Aβ) plaques and neurofibrillary tangles, emerging evidence demonstrates a critical role of microglia and neuroinflammation in AD pathogenesis. Oleoylethanolamide (OEA) is an endogenous lipid amide that has been shown to promote lifespan and healthspan in C. elegans through regulation of lysosome-to-nucleus signaling and cellular metabolism. The goal of our study was to determine the role of OEA in the mediation of microglial activity and AD pathology using its stable analog, KDS-5104. METHODS We used primary microglial cultures and genetic and pharmacological approaches to examine the signaling mechanisms and functional roles of OEA in mediating Aβ phagocytosis and clearance, lipid metabolism and inflammasome formation. Further, we tested the effect of OEA in vivo in acute LPS-induced neuroinflammation and by chronic treatment of 5xFAD mice. RESULTS We found that OEA activates PPARα signaling and its downstream cell-surface receptor CD36 activity. In addition, OEA promotes TFEB lysosomal function in a PPARα-dependent but mTORC1-independent manner, the combination of which leads to enhanced microglial Aβ uptake and clearance. These are associated with the suppression of LPS-induced lipid droplet accumulation and inflammasome activation. Chronic treatment of 5xFAD mice with KDS-5104 restored dysregulated lipid profiles, reduced reactive gliosis and Aβ pathology and rescued cognitive impairments. CONCLUSION Together, our study provides support that augmenting OEA-mediated lipid signaling may offer therapeutic benefit against aging and AD through modulating lipid metabolism and microglia phagocytosis and clearance.
Collapse
Affiliation(s)
- Michele M Comerota
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Manasee Gedam
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Translational Biology and Molecular Medicine Graduate Program, Houston, TX, USA
| | - Wen Xiong
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Feng Jin
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Pharmacology and Chemical Biology, Houston, TX, USA
| | - Lisheng Deng
- Department of Pharmacology and Chemical Biology, Houston, TX, USA
| | - Meng C Wang
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
- HHMI Janelia Research Campus, Ashburn, VA, USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Houston, TX, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Translational Biology and Molecular Medicine Graduate Program, Houston, TX, USA.
- Department of Molecular and Human Genetics, Houston, TX, USA.
| |
Collapse
|
44
|
Paseban T, Alavi MS, Etemad L, Roohbakhsh A. The role of the ATP-Binding Cassette A1 (ABCA1) in neurological disorders: a mechanistic review. Expert Opin Ther Targets 2023; 27:531-552. [PMID: 37428709 DOI: 10.1080/14728222.2023.2235718] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/09/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION Cholesterol homeostasis is critical for normal brain function. It is tightly controlled by various biological elements. ATP-binding cassette transporter A1 (ABCA1) is a membrane transporter that effluxes cholesterol from cells, particularly astrocytes, into the extracellular space. The recent studies pertaining to ABCA1's role in CNS disorders were included in this study. AREAS COVERED In this comprehensive literature review, preclinical and human studies showed that ABCA1 has a significant role in the following diseases or disorders: Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, neuropathy, anxiety, depression, psychosis, epilepsy, stroke, and brain ischemia and trauma. EXPERT OPINION ABCA1 via modulating normal and aberrant brain functions such as apoptosis, phagocytosis, BBB leakage, neuroinflammation, amyloid β efflux, myelination, synaptogenesis, neurite outgrowth, and neurotransmission promotes beneficial effects in aforementioned diseases. ABCA1 is a key molecule in the CNS. By boosting its expression or function, some CNS disorders may be resolved. In preclinical studies, liver X receptor agonists have shown promise in treating CNS disorders via ABCA1 and apoE enhancement.
Collapse
Affiliation(s)
- Tahere Paseban
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
45
|
Pesämaa I, Müller SA, Robinson S, Darcher A, Paquet D, Zetterberg H, Lichtenthaler SF, Haass C. A MICROGLIAL ACTIVITY STATE BIOMARKER PANEL DIFFERENTIATES FTD-GRANULIN AND ALZHEIMER'S DISEASE PATIENTS FROM CONTROLS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.15.545187. [PMID: 37398209 PMCID: PMC10312678 DOI: 10.1101/2023.06.15.545187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background With the emergence of microglia-modulating therapies there is an urgent need for reliable biomarkers to evaluate microglial activation states. Methods Using mouse models and human induced pluripotent stem cell-derived microglia (hiMGL), which were genetically modified to yield the most opposite homeostatic ( TREM2- knockout) and disease-associated ( GRN -knockout) states, we identified microglia activity-dependent markers. Non-targeted mass spectrometry was used to identify changes in microglial and cerebrospinal (CSF) proteome of Grn - and Trem2 -knockout mice. Additionally, we analyzed the proteome of GRN - and TREM2 -knockout hiMGL and their conditioned media. Candidate marker proteins were tested in two independent patient cohorts, the ALLFTD cohort with 11 GRN mutation carriers and 12 non-carriers, as well as the proteomic data set available from the European Medical Information Framework Alzheimer's Disease Multimodal Biomarker Discovery (EMIF-AD MBD). Findings We identified proteomic changes between the opposite activation states in mouse microglia and cerebrospinal fluid (CSF), as well as in hiMGL cell lysates and conditioned media. For further verification, we analyzed the CSF proteome of heterozygous GRN mutation carriers suffering from frontotemporal dementia (FTD). We identified a panel of six proteins (FABP3, MDH1, GDI1, CAPG, CD44, GPNMB) as potential indicators for microglial activation. Moreover, we confirmed three of these proteins (FABP3, GDI1, MDH1) to be significantly elevated in the CSF of AD patients. In AD, these markers differentiated amyloid-positive cases with mild cognitive impairment (MCI) from amyloid-negative individuals. Interpretation The identified candidate proteins reflect microglia activity and may be relevant for monitoring the microglial response in clinical practice and clinical trials modulating microglial activity and amyloid deposition. Moreover, the finding that three of these markers differentiate amyloid-positive from amyloid-negative MCI cases in the AD cohort suggests that these marker proteins associate with a very early immune response to seeded amyloid. This is consistent with our previous findings in the DIAN (Dominantly Inherited Alzheimer's Disease Network) cohort, where soluble TREM2 increases as early as 21 years before symptom onset. Moreover, in mouse models for amyloidogenesis, seeding of amyloid is limited by physiologically active microglia further supporting their early protective role. The biological functions of some of our main candidates (FABP3, CD44, GPNMB) also further emphasize that lipid dysmetabolism may be a common feature of neurodegenerative disorders. Funding This work was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany's Excellence Strategy within the framework of the Munich Cluster for Systems Neurology (EXC 2145 SyNergy - ID 390857198 to CH, SFL and DP) and a Koselleck Project HA1737/16-1 (to CH).
Collapse
|
46
|
Temple S. Advancing cell therapy for neurodegenerative diseases. Cell Stem Cell 2023; 30:512-529. [PMID: 37084729 PMCID: PMC10201979 DOI: 10.1016/j.stem.2023.03.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/23/2023]
Abstract
Cell-based therapies are being developed for various neurodegenerative diseases that affect the central nervous system (CNS). Concomitantly, the roles of individual cell types in neurodegenerative pathology are being uncovered by genetic and single-cell studies. With a greater understanding of cellular contributions to health and disease and with the arrival of promising approaches to modulate them, effective therapeutic cell products are now emerging. This review examines how the ability to generate diverse CNS cell types from stem cells, along with a deeper understanding of cell-type-specific functions and pathology, is advancing preclinical development of cell products for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| |
Collapse
|
47
|
Andrews SJ, Renton AE, Fulton-Howard B, Podlesny-Drabiniok A, Marcora E, Goate AM. The complex genetic architecture of Alzheimer's disease: novel insights and future directions. EBioMedicine 2023; 90:104511. [PMID: 36907103 PMCID: PMC10024184 DOI: 10.1016/j.ebiom.2023.104511] [Citation(s) in RCA: 162] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disorder and the most common form of dementia. AD is highly heritable, with heritability estimates of ∼70% from twin studies. Progressively larger genome-wide association studies (GWAS) have continued to expand our knowledge of AD/dementia genetic architecture. Until recently these efforts had identified 39 disease susceptibility loci in European ancestry populations. RECENT DEVELOPMENTS Two new AD/dementia GWAS have dramatically expanded the sample sizes and the number of disease susceptibility loci. The first increased total sample size to 1,126,563-with an effective sample size of 332,376-by predominantly including new biobank and population-based dementia datasets. The second, expands on an earlier GWAS from the International Genomics of Alzheimer's Project (IGAP) by increasing the number of clinically-defined AD cases/controls in addition to incorporating biobank dementia datasets, resulting in a total sample size to 788,989 and an effective sample size of 382,472. Collectively both GWAS identified 90 independent variants across 75 AD/dementia susceptibility loci, including 42 novel loci. Pathway analyses indicate the susceptibility loci are enriched for genes involved in amyloid plaque and neurofibrillary tangle formation, cholesterol metabolism, endocytosis/phagocytosis, and the innate immune system. Gene prioritization efforts for the novel loci identified 62 candidate causal genes. Many of the candidate genes from known and newly discovered loci play key roles in macrophages and highlight phagocytic clearance of cholesterol-rich brain tissue debris by microglia (efferocytosis) as a core pathogenetic hub and putative therapeutic target for AD. WHERE NEXT?: While GWAS in European ancestry populations have substantially enhanced our understanding of AD genetic architecture, heritability estimates from population based GWAS cohorts are markedly smaller than those from twin studies. While this missing heritability is likely due to a combination of factors, it highlights that our understanding of AD genetic architecture and genetic risk mechanisms remains incomplete. These knowledge gaps result from several underexplored areas in AD research. First, rare variants remain understudied due to methodological issues in identifying them and the cost of generating sufficiently powered whole exome/genome sequencing datasets. Second, sample sizes of non-European ancestry populations in AD GWAS remain small. Third, GWAS of AD neuroimaging and cerebrospinal fluid endophenotypes remains limited due to low compliance and high costs associated with measuring amyloid-β and tau levels and other disease-relevant biomarkers. Studies generating sequencing data, including diverse populations, and incorporating blood-based AD biomarkers are set to substantially improve our knowledge of AD genetic architecture.
Collapse
Affiliation(s)
- Shea J Andrews
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA.
| | - Alan E Renton
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Fulton-Howard
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Podlesny-Drabiniok
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edoardo Marcora
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison M Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
48
|
Podlesny-Drabiniok A, Novikova G, Liu Y, Dunst J, Temizer R, Giannarelli C, Marro S, Kreslavsky T, Marcora E, Goate AM. BHLHE40/41 regulate macrophage/microglia responses associated with Alzheimer's disease and other disorders of lipid-rich tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528372. [PMID: 36824752 PMCID: PMC9948946 DOI: 10.1101/2023.02.13.528372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Background Genetic and experimental evidence strongly implicates myeloid cells in the etiology of AD and suggests that AD-associated alleles and genes may modulate disease risk by altering the transcriptional and cellular responses of macrophages (like microglia) to damage of lipid-rich tissues (like the brain). Specifically, recent single-cell/nucleus RNA sequencing (sc/nRNA-seq) studies identified a transcriptionally distinct state of subsets of macrophages in aging or degenerating brains (usually referred to as disease-associated microglia or DAM) and in other diseased lipid-rich tissues (e.g., obese adipose tissue, fatty liver, and atherosclerotic plaques). We collectively refer to these subpopulations as lipid-associated macrophages or LAMs. Importantly, this particular activation state is characterized by increased expression of genes involved in the phagocytic clearance of lipid-rich cellular debris (efferocytosis), including several AD risk genes. Methods We used sc/nRNA-seq data from human and mouse microglia from healthy and diseased brains and macrophages from other lipid-rich tissues to reconstruct gene regulatory networks and identify transcriptional regulators whose regulons are enriched for LAM response genes (LAM TFs) across species. We then used gene knock-down/knock-out strategies to validate some of these LAM TFs in human THP-1 macrophages and iPSC-derived microglia in vitro, as well as mouse microglia in vivo. Results We nominate 11 strong candidate LAM TFs shared across human and mouse networks (BHLHE41, HIF1A, ID2, JUNB, MAF, MAFB, MEF2A, MEF2C, NACA, POU2F2 and SPI1). We also demonstrate a strong enrichment of AD risk alleles in the cistrome of BHLHE41 (and its close homolog BHLHE40), thus implicating its regulon in the modulation of disease susceptibility. Loss or reduction of BHLHE40/41 expression in human THP-1 macrophages and iPSC-derived microglia, as well as loss of Bhlhe40/41 in mouse microglia led to increased expression of LAM response genes, specifically those involved in cholesterol clearance and lysosomal processing, with a concomitant increase in cholesterol efflux and storage, as well as lysosomal mass and degradative capacity. Conclusions Taken together, this study nominates transcriptional regulators of the LAM response, experimentally validates BHLHE40/41 in human and mouse macrophages/microglia, and provides novel targets for therapeutic modulation of macrophage/microglia function in AD and other disorders of lipid-rich tissues.
Collapse
Affiliation(s)
- Anna Podlesny-Drabiniok
- Department of Genetics and Genomic Sciences, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gloriia Novikova
- Department of Genetics and Genomic Sciences, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
- OMNI Bioinformatics Department and Neuroscience Department, Genentech, Inc., South San Francisco, CA, USA
| | - Yiyuan Liu
- Department of Genetics and Genomic Sciences, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josefine Dunst
- Department of Medicine, Division of Immunology and Allergy, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rose Temizer
- Department of Genetics and Genomic Sciences, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Giannarelli
- Department of Medicine (C.G.), Cardiology, NYU Grossman School of Medicine
- Department of Pathology (C.G.), Cardiology, NYU Grossman School of Medicine
| | - Samuele Marro
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Taras Kreslavsky
- Department of Medicine, Division of Immunology and Allergy, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Edoardo Marcora
- Department of Genetics and Genomic Sciences, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alison Mary Goate
- Department of Genetics and Genomic Sciences, New York, NY, USA; Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
49
|
Comerota M, Gedam M, Xiong W, Jin F, Deng L, Wang M, Wang J, Zheng H. Oleoylethanolamide facilitates PPARa and TFEB signaling and attenuates Ab pathology in a mouse model of Alzheimer's disease. RESEARCH SQUARE 2023:rs.3.rs-2484513. [PMID: 36711875 PMCID: PMC9882642 DOI: 10.21203/rs.3.rs-2484513/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Emerging evidence implicates impaired microglia function and dysregulation of lipid metabolism in Alzheimer's disease (AD). Oleoylethanolamide (OEA), an endogenous lipid and PPARα agonist, has been shown to promote longevity in C. elegans through regulation of lysosome-to-nucleus signaling and cellular metabolism. Using a stable OEA analog, KDS-5104, we found that OEA-PPARα signaling promotes TFEB lysosomal activity independent of mTORC1 and upregulates cell-surface receptor CD36, leading to enhanced microglial Aβ uptake and clearance. These are associated with the suppression of LPS-induced lipid droplet accumulation and inflammasome activation. Chronic treatment of the 5xFAD mice with KDS-5104 restored dysregulated profiles, reduced reactive gliosis and Aβ pathology and rescued cognitive impairments. Together, our study provides support that augmenting OEA-mediated lipid signaling may offer therapeutic benefit against aging and AD through modulating lipid metabolism and microglia phagocytosis and clearance.
Collapse
|
50
|
Rosenberry TL, Zhou HX, Stagg SM, Paravastu AK. Oligomer Formation by Amyloid-β42 in a Membrane-Mimicking Environment in Alzheimer's Disease. Molecules 2022; 27:8804. [PMID: 36557940 PMCID: PMC9781152 DOI: 10.3390/molecules27248804] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
The brains of Alzheimer's disease (AD) patients contain numerous amyloid plaques that are diagnostic of the disease. The plaques are primarily composed of the amyloidogenic peptides proteins Aβ40 and Aβ42, which are derived by the processing of the amyloid pre-cursor protein (APP) by two proteases called β-secretase and γ-secretase. Aβ42 differs from Aβ40 in having two additional hydrophobic amino acids, ILE and ALA, at the C-terminus. A small percentage of AD is autosomal dominant (ADAD) and linked either to the genes for the presenilins, which are part of γ-secretase, or APP. Because ADAD shares most pathogenic features with widespread late-onset AD, Aβ peptides have become the focus of AD research. Fibrils formed by the aggregation of these peptides are the major component of plaques and were initially targeted in AD therapy. However, the fact that the abundance of plaques does not correlate well with cognitive decline in AD patients has led investigators to examine smaller Aβ aggregates called oligomers. The low levels and heterogeneity of Aβ oligomers have made the determination of their structures difficult, but recent structure determinations of oligomers either formed or initiated in detergents have been achieved. We report here on the structures of these oligomers and suggest how they may be involved in AD.
Collapse
Affiliation(s)
- Terrone L. Rosenberry
- The Departments of Neuroscience and Pharmacology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Huan-Xiang Zhou
- Departments of Chemistry and Physics, University of Illinois Chicago, Chicago, IL 60608, USA
| | - Scott M. Stagg
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
- Department of Biological Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Anant K. Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, GA 30332, USA
| |
Collapse
|