1
|
Zhang W, Uyemura R, Zhong K, Guo R, Zhong L. Current Advances and Future Perspectives on Mesenchymal Stem Cell-Derived Extracellular Vesicles in Alzheimer's Disease. Aging Dis 2024; 15:2015-2027. [PMID: 38270122 PMCID: PMC11346404 DOI: 10.14336/ad.2023.1206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/06/2023] [Indexed: 01/26/2024] Open
Abstract
The incidence of Alzheimer's disease (AD) has been increasing in recent years as the world's population ages, which poses a significant challenge to public health. Due to the complexity of pathogenesis of AD, currently there is no effective treatment for it. In recent years, cell and gene therapy has attracted widespread attention in the treatment of neurodegenerative diseases. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) represent a novel cell-free therapy with numerous advantages over cell-based therapies owing to their low immunogenicity and high safety profile. We summarize recent progress in the application of EVs for treating AD and the specific mechanisms and outline the underlying mechanisms. We also explore various methods for optimizing the function of MSC-EVs, including gene editing, modifying stem cell culture conditions and peptide modification. In addition, we discuss the therapeutic potentials of MSC-EVs, as well as the obstacles that currently impede their clinical utilization.
Collapse
Affiliation(s)
- Wenjing Zhang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Russell Uyemura
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| | - Kun Zhong
- American Center of Stem Cell Research and Regenerative Medicine, Farmington Hills, Michigan 48336, USA
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Li Zhong
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| |
Collapse
|
2
|
Berki P, Cserép C, Környei Z, Pósfai B, Szabadits E, Domonkos A, Kellermayer A, Nyerges M, Wei X, Mody I, Kunihiko A, Beck H, Kaikai H, Ya W, Lénárt N, Wu Z, Jing M, Li Y, Gulyás AI, Dénes Á. Microglia contribute to neuronal synchrony despite endogenous ATP-related phenotypic transformation in acute mouse brain slices. Nat Commun 2024; 15:5402. [PMID: 38926390 PMCID: PMC11208608 DOI: 10.1038/s41467-024-49773-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Acute brain slices represent a workhorse model for studying the central nervous system (CNS) from nanoscale events to complex circuits. While slice preparation inherently involves tissue damage, it is unclear how microglia, the main immune cells and damage sensors of the CNS react to this injury and shape neuronal activity ex vivo. To this end, we investigated microglial phenotypes and contribution to network organization and functioning in acute brain slices. We reveal time-dependent microglial phenotype changes influenced by complex extracellular ATP dynamics through P2Y12R and CX3CR1 signalling, which is sustained for hours in ex vivo mouse brain slices. Downregulation of P2Y12R and changes of microglia-neuron interactions occur in line with alterations in the number of excitatory and inhibitory synapses over time. Importantly, functional microglia modulate synapse sprouting, while microglial dysfunction results in markedly impaired ripple activity both ex vivo and in vivo. Collectively, our data suggest that microglia are modulators of complex neuronal networks with important roles to maintain neuronal network integrity and activity. We suggest that slice preparation can be used to model time-dependent changes of microglia-neuron interactions to reveal how microglia shape neuronal circuits in physiological and pathological conditions.
Collapse
Affiliation(s)
- Péter Berki
- János Szentágothai Doctoral School of Neuroscience, Semmelweis University, Budapest, H-1083, Hungary
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Neuronal Network and Behaviour, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zsuzsanna Környei
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Eszter Szabadits
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Andor Domonkos
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Thalamus Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Anna Kellermayer
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Miklós Nyerges
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Xiaofei Wei
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Istvan Mody
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Araki Kunihiko
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - Heinz Beck
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - He Kaikai
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Wang Ya
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Miao Jing
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Attila I Gulyás
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary.
| |
Collapse
|
3
|
Sepulveda J, Kim JY, Binder J, Vicini S, Rebeck GW. APOE4 genotype and aging impair injury-induced microglial behavior in brain slices, including toward Aβ, through P2RY12. Mol Neurodegener 2024; 19:24. [PMID: 38468308 DOI: 10.1186/s13024-024-00714-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Microglia are highly dynamic cells that play a critical role in tissue homeostasis through the surveillance of brain parenchyma and response to cues associated with damage. Aging and APOE4 genotype are the strongest risk factors for Alzheimer's disease (AD), but how they affect microglial dynamics remains unclear. Using ex vivo confocal microscopy, we analyzed microglial dynamic behaviors in the entorhinal cortex (EC) and hippocampus CA1 of 6-, 12-, and 21-month-old mice APOE3 or APOE4 knock-in mice expressing GFP under the CX3CR1 promoter. To study microglia surveillance, we imaged microglia baseline motility for 20 min and measured the extension and retraction of processes. We found that APOE4 microglia exhibited significantly less brain surveillance (27%) compared to APOE3 microglia in 6-month-old mice; aging exacerbated this deficit. To measure microglia response to damage, we imaged process motility in response to ATP, an injury-associated signal, for 30 min. We found APOE4 microglia extended their processes significantly slower (0.9 µm/min, p < 0.005) than APOE3 microglia (1.1 μm/min) in 6-month-old animals. APOE-associated alterations in microglia motility were observed in 12- and 21-month-old animals, and this effect was exacerbated with aging in APOE4 microglia. We measured protein and mRNA levels of P2RY12, a core microglial receptor required for process movement in response to damage. We found that APOE4 microglia express significantly less P2RY12 receptors compared to APOE3 microglia despite no changes in P2RY12 transcripts. To examine if the effect of APOE4 on the microglial response to ATP also applied to amyloid β (Aβ), we infused locally Hi-Lyte Fluor 555-labeled Aβ in acute brain slices of 6-month-old mice and imaged microglia movement for 2 h. APOE4 microglia showed a significantly slower (p < 0.0001) process movement toward the Aβ, and less Aβ coverage at early time points after Aβ injection. To test whether P2RY12 is involved in process movement in response to Aβ, we treated acute brain slices with a P2RY12 antagonist before Aβ injection; microglial processes no longer migrated towards Aβ. These results provide mechanistic insights into the impact of APOE4 genotype and aging in dynamic microglial behaviors prior to gross Aβ pathology and could help explain how APOE4 brains are more susceptible to AD pathogenesis.
Collapse
Affiliation(s)
- Jordy Sepulveda
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, 20007, USA
| | - Jennifer Yejean Kim
- Department of Neuroscience, Georgetown University, Washington, DC, 20007, USA
| | - Joseph Binder
- Department of Neuroscience, Georgetown University, Washington, DC, 20007, USA
| | - Stefano Vicini
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, 20007, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University, Washington, DC, 20007, USA.
| |
Collapse
|
4
|
Sun MK, Alkon DL. Alzheimer's therapeutic development: shifting neurodegeneration to neuroregeneration. Trends Pharmacol Sci 2024; 45:197-209. [PMID: 38360510 PMCID: PMC10939773 DOI: 10.1016/j.tips.2024.01.012] [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: 12/14/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 02/17/2024]
Abstract
Alzheimer's disease (AD), similar to AD-related dementias, is characterized by impaired/lost neuronal structures and functions due to a long progression of neurodegeneration. Derailed endogenous signal pathways and disease processes have critical roles in neurodegeneration and are pharmacological targets in inducing neuroregeneration. Pharmacologically switching/shifting the brain status from neurodegeneration to neuroregeneration is emerging as a new therapeutic concept, one that is not only achievable, but also essential for effective therapy for AD. The results of the pharmacological-induced shift from neurodegeneration to neuroregeneration are twofold: arresting cognitive deterioration (and directing the brain toward cognitive recovery) in established AD, and preventing neurodegeneration through building up cognitive resilience in patients with preclinical or probable AD. In this review, we discuss these new developments in AD pharmacology and relevant clinical trials.
Collapse
Affiliation(s)
- Miao-Kun Sun
- Synaptogenix, Inc., 1185 Avenue of the Americas, 3rd Floor, New York, NY 10036, USA.
| | - Daniel L Alkon
- Synaptogenix, Inc., 1185 Avenue of the Americas, 3rd Floor, New York, NY 10036, USA
| |
Collapse
|
5
|
Kapate N, Liao R, Sodemann RL, Stinson T, Prakash S, Kumbhojkar N, Suja VC, Wang LLW, Flanz M, Rajeev R, Villafuerte D, Shaha S, Janes M, Park KS, Dunne M, Golemb B, Hone A, Adebowale K, Clegg J, Slate A, McGuone D, Costine-Bartell B, Mitragotri S. Backpack-mediated anti-inflammatory macrophage cell therapy for the treatment of traumatic brain injury. PNAS NEXUS 2024; 3:pgad434. [PMID: 38187808 PMCID: PMC10768983 DOI: 10.1093/pnasnexus/pgad434] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024]
Abstract
Traumatic brain injury (TBI) is a debilitating disease with no current therapies outside of acute clinical management. While acute, controlled inflammation is important for debris clearance and regeneration after injury, chronic, rampant inflammation plays a significant adverse role in the pathophysiology of secondary brain injury. Immune cell therapies hold unique therapeutic potential for inflammation modulation, due to their active sensing and migration abilities. Macrophages are particularly suited for this task, given the role of macrophages and microglia in the dysregulated inflammatory response after TBI. However, maintaining adoptively transferred macrophages in an anti-inflammatory, wound-healing phenotype against the proinflammatory TBI milieu is essential. To achieve this, we developed discoidal microparticles, termed backpacks, encapsulating anti-inflammatory interleukin-4, and dexamethasone for ex vivo macrophage attachment. Backpacks durably adhered to the surface of macrophages without internalization and maintained an anti-inflammatory phenotype of the carrier macrophage through 7 days in vitro. Backpack-macrophage therapy was scaled up and safely infused into piglets in a cortical impact TBI model. Backpack-macrophages migrated to the brain lesion site and reduced proinflammatory activation of microglia in the lesion penumbra of the rostral gyrus of the cortex and decreased serum concentrations of proinflammatory biomarkers. These immunomodulatory effects elicited a 56% decrease in lesion volume. The results reported here demonstrate, to the best of our knowledge, a potential use of a cell therapy intervention for a large animal model of TBI and highlight the potential of macrophage-based therapy. Further investigation is required to elucidate the neuroprotection mechanisms associated with anti-inflammatory macrophage therapy.
Collapse
Affiliation(s)
- Neha Kapate
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rick Liao
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Ryan Luke Sodemann
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Tawny Stinson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Supriya Prakash
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Ninad Kumbhojkar
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Vineeth Chandran Suja
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Lily Li-Wen Wang
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mikayla Flanz
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Rohan Rajeev
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Dania Villafuerte
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Suyog Shaha
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Morgan Janes
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kyung Soo Park
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Michael Dunne
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - Bryan Golemb
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alexander Hone
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kolade Adebowale
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| | - John Clegg
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Andrea Slate
- Center of Comparative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Declan McGuone
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Beth Costine-Bartell
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA 02115, USA
| | - Samir Mitragotri
- School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02134, USA
| |
Collapse
|
6
|
Nabizadeh F. sTREM2 is associated with attenuated tau aggregate accumulation in the presence of amyloid-β pathology. Brain Commun 2023; 5:fcad286. [PMID: 37942087 PMCID: PMC10629471 DOI: 10.1093/braincomms/fcad286] [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: 05/31/2023] [Revised: 08/16/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023] Open
Abstract
Triggering Receptor Expressed on Myeloid Cell 2 (TREM2) plays a crucial role in the transition of microglia from a state of homeostasis to a state associated with the disease. Mutations in TREM2 are strongly linked with a higher risk of developing neurodegenerative diseases, including Alzheimer's disease. There have been contradictory findings regarding the potential detrimental or protective effects of microglial activation and TREM2-related microglial responses in Alzheimer's disease. Although previous studies reported increased CSF soluble TREM2 (sTREM2) in different clinical stages of Alzheimer's disease, the exact association between Alzheimer's disease hallmarks such as amyloid-beta and tau pathology remains unclear. In the present study, I aimed to investigate the association between TREM2-related microglial responses and tau accumulation in the presence and absence of amyloid-beta pathology in order to give a better view of the role of microglial activation in Alzheimer's disease development. Imaging data of 178 non-demented participants including 107 amyloid-beta-negative participants, 71 amyloid-beta-positive were recruited from Alzheimer's disease Neuroimaging Initiative. The CSF sTREM2 was used as an in vivo indicator of microglial responses associated with TREM2. Furthermore, I used longitudinal tau-PET and resting-state functional MRI connectomes in order to investigate the association of TREM2-related microglial activation and tau spreading through functional connections. A higher level of sTREM2 was associated with slower tau aggregate accumulation in non-demented amyloid-beta-positive. Furthermore, measuring the tau spreading through inter-connected regions using functional MRI connectomes confirms that the TREM2-related microglial activity might be a protective factor against tau pathology in brain tissue. These findings demonstrate that in individuals with initial amyloid-beta abnormalities, TREM2-related microglial activation is linked to reduced regional accumulation of tau aggregates and also, spreading across inter-connected brain regions, as evaluated through functional MRI connectomes during the early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| |
Collapse
|
7
|
Zajac DJ, Simpson J, Zhang E, Parikh I, Estus S. Expression of INPP5D Isoforms in Human Brain: Impact of Alzheimer's Disease Neuropathology and Genetics. Genes (Basel) 2023; 14:763. [PMID: 36981033 PMCID: PMC10048252 DOI: 10.3390/genes14030763] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
The single nucleotide polymorphisms rs35349669 and rs10933431 within Inositol Polyphosphate-5-Phosphatase D (INPP5D) are strongly associated with Alzheimer's Disease risk. To better understand INPP5D expression in the brain, we investigated INPP5D isoform expression as a function of rs35349669 and rs10933431, as well as Alzheimer's disease neuropathology, by qPCR and isoform-specific primers. In addition, INPP5D allelic expression imbalance was evaluated relative to rs1141328 within exon 1. Expression of INPP5D isoforms associated with transcription start sites in exon 1 and intron 14 was increased in individuals with high Alzheimer's disease neuropathology. In addition, a novel variant with 47bp lacking from exon 12 increased expression in Alzheimer's Disease brains, accounting for 13% of total INPP5D expression, and was found to undergo nonsense-mediated decay. Although inter-individual variation obscured a possible polymorphism effect on INPP5D isoform expression as measured by qPCR, rs35349669 was associated with rs1141328 allelic expression imbalance, suggesting that rs35349669 is significantly associated with full-length INPP5D isoform expression. In summary, expression of INPP5D isoforms with start sites in exon 1 and intron 14 are increased in brains with high Alzheimer's Disease neuropathology, a novel isoform lacking the phosphatase domain was significantly increased with the disease, and the polymorphism rs35349669 correlates with allele-specific full-length INPP5D expression.
Collapse
Affiliation(s)
| | | | | | | | - Steven Estus
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40508, USA
| |
Collapse
|
8
|
Hou J, Chen Y, Grajales-Reyes G, Colonna M. TREM2 dependent and independent functions of microglia in Alzheimer's disease. Mol Neurodegener 2022; 17:84. [PMID: 36564824 PMCID: PMC9783481 DOI: 10.1186/s13024-022-00588-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/02/2022] [Indexed: 12/25/2022] Open
Abstract
Microglia are central players in brain innate immunity and have been the subject of extensive research in Alzheimer's disease (AD). In this review, we aim to summarize the genetic and functional discoveries that have advanced our understanding of microglia reactivity to AD pathology. Given the heightened AD risk posed by rare variants of the microglial triggering receptor expressed on myeloid cells 2 (TREM2), we will focus on the studies addressing the impact of this receptor on microglia responses to amyloid plaques, tauopathy and demyelination pathologies in mouse and human. Finally, we will discuss the implications of recent discoveries on microglia and TREM2 biology on potential therapeutic strategies for AD.
Collapse
Affiliation(s)
- Jinchao Hou
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Yun Chen
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA ,grid.4367.60000 0001 2355 7002Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Gary Grajales-Reyes
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Marco Colonna
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
9
|
Redefining microglia states: Lessons and limits of human and mouse models to study microglia states in neurodegenerative diseases. Semin Immunol 2022; 60:101651. [PMID: 36155944 DOI: 10.1016/j.smim.2022.101651] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/03/2022] [Indexed: 01/15/2023]
Abstract
Microglia are resident macrophages of the brain parenchyma and play an essential role in various aspects of brain development, plasticity, and homeostasis. With recent advances in single-cell RNA-sequencing, heterogeneous microglia transcriptional states have been identified in both animal models of neurodegenerative disorders and patients. However, the functional roles of these microglia states remain unclear; specifically, the question of whether individual states or combinations of states are protective or detrimental (or both) in the context of disease progression. To attempt to answer this, the field has largely relied on studies employing mouse models, human in vitro and chimeric models, and human post-mortem tissue, all of which have their caveats, but used in combination can enable new biological insight and validation of candidate disease pathways and mechanisms. In this review, we summarize our current understanding of disease-associated microglia states and phenotypes in neurodegenerative disorders, discuss important considerations when comparing mouse and human microglia states and functions, and identify areas of microglia biology where species differences might limit our understanding of microglia state.
Collapse
|
10
|
Taheri F, Taghizadeh E, Navashenaq JG, Rezaee M, Gheibihayat SM. The role of efferocytosis in neuro-degenerative diseases. Neurol Sci 2022; 43:1593-1603. [PMID: 35059903 DOI: 10.1007/s10072-021-05835-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/11/2021] [Indexed: 02/06/2023]
Abstract
Efferocytosis has a critical role in maintaining tissues and organs' homeostasis by removing apoptotic cells. It is essential for human health, and disturbances in efferocytosis may result indifferent illnesses. In case of inadequate clearance of the dead cells, the content in the cells would be released. In fact, it induces some damages to the tissue and leads to the prolonged inflammation, so unsuitable phagocytosis of the apoptotic cells is involved in occurrence as well as expansion of numerous human chronic inflammatory diseases. Studies have shown age dependence of the neuro-degenerative diseases, which are largely due to the neuro-inflammation and the loss of neurons and thus cause the brain's functional disorders. Efferocytosis is coupled to anti-inflammatory responses that contribute to the elimination of the dying neurons in neuro-degenerative diseases, so its disruption may make a risk factor in numerous human chronic inflammatory diseases such as multiple sclerosis, Alzheimer's disease, glioblastoma, and Rett syndrome. This study is a review of the efferocytosis molecular pathways and their role in neuro-degenerative diseases in order to discover a new treatment option to cure patients.
Collapse
Affiliation(s)
- Forough Taheri
- Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Eskandar Taghizadeh
- Department of Medical Genetic, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Mehdi Rezaee
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, P.O. Box: 8915173143, Yazd, Iran.
| |
Collapse
|
11
|
Ganz T, Fainstein N, Elad A, Lachish M, Goldfarb S, Einstein O, Ben-Hur T. Microbial pathogens induce neurodegeneration in Alzheimer's disease mice: protection by microglial regulation. J Neuroinflammation 2022; 19:5. [PMID: 34991645 PMCID: PMC8740456 DOI: 10.1186/s12974-021-02369-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neurodegeneration is considered the consequence of misfolded proteins' deposition. Little is known about external environmental effects on the neurodegenerative process. Infectious agent-derived pathogen-associated molecular patterns (PAMPs) activate microglia, key players in neurodegenerative diseases. We hypothesized that systemic microbial pathogens may accelerate neurodegeneration in Alzheimer's disease (AD) and that microglia play a central role in this process. METHODS We examined the effect of an infectious environment and of microbial Toll-like receptor (TLR) agonists on cortical neuronal loss and on microglial phenotype in wild type versus 5xFAD transgenic mice, carrying mutated genes associated with familial AD. RESULTS We examined the effect of a naturally bred environment on the neurodegenerative process. Earlier and accelerated cortical neuron loss occurred in 5xFAD mice housed in a natural ("dirty") environment than in a specific-pathogen-free (SPF) environment, without increasing the burden of Amyloid deposits and microgliosis. Neuronal loss occurred in a microglia-rich cortical region but not in microglia-poor CA regions of the hippocampus. Environmental exposure had no effect on cortical neuron density in wild-type mice. To model the neurodegenerative process caused by the natural infectious environment, we injected systemically the bacterial endotoxin lipopolysaccharide (LPS), a TLR4 agonist PAMP. LPS caused cortical neuronal death in 5xFAD, but not wt mice. We used the selective retinoic acid receptor α agonist Am580 to regulate microglial activation. In primary microglia isolated from 5xFAD mice, Am580 markedly attenuated TLR agonists-induced iNOS expression, without canceling their basic immune response. Intracerebroventricular delivery of Am580 in 5xFAD mice reduced significantly the fraction of (neurotoxic) iNOS + microglia and increased the fraction of (neuroprotective) TREM2 + microglia. Furthermore, intracerebroventricular delivery of Am580 prevented neurodegeneration induced by microbial TLR agonists. CONCLUSIONS Exposure to systemic infections causes neurodegeneration in brain regions displaying amyloid pathology and high local microglia density. AD brains exhibit increased susceptibility to microbial PAMPs' neurotoxicity, which accelerates neuronal death. Microglial modulation protects the brain from microbial TLR agonist PAMP-induced neurodegeneration.
Collapse
Affiliation(s)
- Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amit Elad
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Smadar Goldfarb
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ofira Einstein
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
12
|
Di Liberto G, Egervari K, Kreutzfeldt M, Schürch CM, Hewer E, Wagner I, Du Pasquier R, Merkler D. OUP accepted manuscript. Brain 2022; 145:2730-2741. [PMID: 35808999 PMCID: PMC9420019 DOI: 10.1093/brain/awac102] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 11/14/2022] Open
Abstract
Glial cell activation is a hallmark of several neurodegenerative and neuroinflammatory diseases. During HIV infection, neuroinflammation is associated with cognitive impairment, even during sustained long-term suppressive antiretroviral therapy. However, the cellular subsets contributing to neuronal damage in the CNS during HIV infection remain unclear. Using post-mortem brain samples from eight HIV patients and eight non-neurological disease controls, we identify a subset of CNS phagocytes highly enriched in LGALS3, CTSB, GPNMB and HLA-DR, a signature identified in the context of ageing and neurodegeneration. In HIV patients, the presence of this phagocyte phenotype was associated with synaptic stripping, suggesting an involvement in the pathogenesis of HIV-associated neurocognitive disorder. Taken together, our findings elucidate some of the molecular signatures adopted by CNS phagocytes in HIV-positive patients and contribute to the understanding of how HIV might pave the way to other forms of cognitive decline in ageing HIV patient populations.
Collapse
Affiliation(s)
- Giovanni Di Liberto
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Department of Clinical Neurosciences, Service of Neurology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kristof Egervari
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Christian M Schürch
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Ekkehard Hewer
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Renaud Du Pasquier
- Department of Clinical Neurosciences, Service of Neurology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Doron Merkler
- Correspondence to: Doron Merkler Centre Médical Universitaire (CMU) 1, rue Michel Servet 1211 Geneva, Switzerland E-mail:
| |
Collapse
|
13
|
Epigenetic Modulation of Microglia Function and Phenotypes in Neurodegenerative Diseases. Neural Plast 2021; 2021:9912686. [PMID: 34194489 PMCID: PMC8181095 DOI: 10.1155/2021/9912686] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/14/2021] [Indexed: 12/14/2022] Open
Abstract
Microglia-mediated neuroinflammation is one of the most remarkable hallmarks of neurodegenerative diseases (NDDs), including AD, PD, and ALS. Accumulating evidence indicates that microglia play both neuroprotective and detrimental roles in the onset and progression of NDDs. Yet, the specific mechanisms of action surrounding microglia are not clear. Modulation of microglia function and phenotypes appears to be a potential strategy to reverse NDDs. Until recently, research into the epigenetic mechanisms of diseases has been gradually developed, making it possible to elucidate the molecular mechanisms underlying the epigenetic regulation of microglia in NDDs. This review highlights the function and phenotypes of microglia, elucidates the relationship between microglia, epigenetic modifications, and NDDs, as well as the possible mechanisms underlying the epigenetic modulation of microglia in NDDs with a focus on potential intervention strategies.
Collapse
|
14
|
Mass E, Gentek R. Fetal-Derived Immune Cells at the Roots of Lifelong Pathophysiology. Front Cell Dev Biol 2021; 9:648313. [PMID: 33708774 PMCID: PMC7940384 DOI: 10.3389/fcell.2021.648313] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/05/2021] [Indexed: 12/12/2022] Open
Abstract
Tissue-resident innate immune cells exert a wide range of functions in both adult homeostasis and pathology. Our understanding of when and how these cellular networks are established has dramatically changed with the recognition that many lineages originate at least in part from fetal sources and self-maintain independently from hematopoietic stem cells. Indeed, fetal-derived immune cells are found in most organs and serous cavities of our body, where they reside throughout the entire lifespan. At the same time, there is a growing appreciation that pathologies manifesting in adulthood may be caused by adverse early life events, a concept known as “developmental origins of health and disease” (DOHaD). Yet, whether fetal-derived immune cells are mechanistically involved in DOHaD remains elusive. In this review, we summarize our knowledge of fetal hematopoiesis and its contribution to adult immune compartments, which results in a “layered immune system.” Based on their ontogeny, we argue that fetal-derived immune cells are prime transmitters of long-term consequences of prenatal adversities. In addition to increasing disease susceptibility, these may also directly cause inflammatory, degenerative, and metabolic disorders. We explore this notion for cells generated from erythro-myeloid progenitors (EMP) produced in the extra-embryonic yolk sac. Focusing on macrophages and mast cells, we present emerging evidence implicating them in lifelong disease by either somatic mutations or developmental programming events resulting from maternal and early environmental perturbations.
Collapse
Affiliation(s)
- Elvira Mass
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Rebecca Gentek
- Centre for Inflammation Research & Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
15
|
Baidya F, Bohra M, Datta A, Sarmah D, Shah B, Jagtap P, Raut S, Sarkar A, Singh U, Kalia K, Borah A, Wang X, Dave KR, Yavagal DR, Bhattacharya P. Neuroimmune crosstalk and evolving pharmacotherapies in neurodegenerative diseases. Immunology 2021; 162:160-178. [PMID: 32939758 PMCID: PMC7808166 DOI: 10.1111/imm.13264] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/20/2020] [Accepted: 08/29/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegeneration is characterized by gradual onset and limited availability of specific biomarkers. Apart from various aetiologies such as infection, trauma, genetic mutation, the interaction between the immune system and CNS is widely associated with neuronal damage in neurodegenerative diseases. The immune system plays a distinct role in disease progression and cellular homeostasis. It induces cellular and humoral responses, and enables tissue repair, cellular healing and clearance of cellular detritus. Aberrant and chronic activation of the immune system can damage healthy neurons. The pro-inflammatory mediators secreted by chief innate immune components, the complement system, microglia and inflammasome can augment cytotoxicity. Furthermore, these inflammatory mediators accelerate microglial activation resulting in progressive neuronal loss. Various animal studies have been carried out to unravel the complex pathology and ascertain biomarkers for these harmful diseases, but have had limited success. The present review will provide a thorough understanding of microglial activation, complement system and inflammasome generation, which lead the healthy brain towards neurodegeneration. In addition to this, possible targets of immune components to confer a strategic treatment regime for the alleviation of neuronal damage are also summarized.
Collapse
Affiliation(s)
- Falguni Baidya
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Mariya Bohra
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Aishika Datta
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Deepaneeta Sarmah
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Birva Shah
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Priya Jagtap
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Swapnil Raut
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Ankan Sarkar
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Upasna Singh
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Kiran Kalia
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| | - Anupom Borah
- Department of Life Science and BioinformaticsAssam UniversitySilcharAssamIndia
| | - Xin Wang
- Department of NeurosurgeryBrigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | - Kunjan R. Dave
- Department of NeurologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Dileep R. Yavagal
- Department of Neurology and NeurosurgeryUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Pallab Bhattacharya
- Department of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research, Ahmedabad (NIPER‐A)GandhinagarGujaratIndia
| |
Collapse
|
16
|
Cao D, Qiao H, He D, Qin X, Zhang Q, Zhou Y. Mesenchymal stem cells inhibited the inflammation and oxidative stress in LPS-activated microglial cells through AMPK pathway. J Neural Transm (Vienna) 2019; 126:1589-1597. [PMID: 31707461 DOI: 10.1007/s00702-019-02102-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022]
Abstract
Microglia are the resident mononuclear immune cells of the central nervous system (CNS) and the activation of microglia contributes to the production of excessive neurotoxic factors. In particular, the overproduction of neurotoxic factors has critical effects on the development of brain injuries and neurodegenerative diseases. The human bone marrow-derived mesenchymal stem cells (hBM-MSCs) have blossomed into an effective approach with great potential for the treatment of neurodegenerative diseases and gliomas. The present study aimed to investigate the mechanism behind the therapeutic effect of hBM-MSCs on the activation of microglia in vitro. Specifically, the hBM-MSCs significantly inhibited the proliferation of lipopolysaccharide-activated microglial cells (LPS)-activated microglial cells. Additionally, we investigated whether the adenosine-monophosphate-activated protein kinase signaling (AMPK) pathway was involved in this process. Our data demonstrated that hBM-MSCs significantly increased the phosphorylated AMPK in LPS-activated microglial cells. In addition, our study indicated the inhibitory effect of hBM-MSCs on the pro-inflammatory mediators and oxidative stress by the AMPK pathway in LPS-activated microglial cells. These results could shed light on the understanding of the molecular basis for the inhibition of hBM-MSCs on LPS-activated microglial cells and provide a molecular mechanism for the hBM-MSCs implication in brain injuries and neurodegenerative diseases.
Collapse
Affiliation(s)
- Dayong Cao
- Department of Burns, The First People's Hospital of Zhengzhou, Zhengzhou, 450000, Henan, People's Republic of China
| | - Haowen Qiao
- School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, People's Republic of China
| | - Dejiao He
- Department of Nephrology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Rd, Wuhan, 430060, People's Republic of China
| | - Xingping Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Qian Zhang
- Department of Oncology, The First People's Hospital of Zhengzhou, Zhengzhou, 450000, Henan, People's Republic of China
| | - Yu Zhou
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Road, Changsha, 410000, Hunan, People's Republic of China.
| |
Collapse
|
17
|
de la Rubia Ortí JE, Prado-Gascó V, Sancho Castillo S, Julián-Rochina M, Romero Gómez FJ, García-Pardo MP. Cortisol and IgA are Involved in the Progression of Alzheimer's Disease. A Pilot Study. Cell Mol Neurobiol 2019; 39:1061-1065. [PMID: 31203531 DOI: 10.1007/s10571-019-00699-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 06/04/2019] [Indexed: 12/29/2022]
Abstract
It is known that stress and immune systems are related with Alzheimer's disease (AD). However, the relationship of both systems in the progression of disease is not clearly demonstrated. Hair cortisol and salivary immunoglobulin A (IgA) were quantified in 49 patients with mild, moderate, and severe AD. A significant change was seen in both molecules as AD progressed from mild to moderate and severe. Low levels of cortisol were observed in mild AD patients compared with moderate and severe. However, IgA showed a contrary pattern. High levels were observed in mild AD patientes but low in moderate and severe AD subjects. The secretion of cortisol and IgA seems to be very different at the start compared with posterior development of AD suggesting that neuroinflammation can be involved. Both molecules could be used as possible therapeutical tools.
Collapse
Affiliation(s)
| | - Vicente Prado-Gascó
- Faculty of Psychology, University of Valencia, Av. Blasco Ibáñez, 21, 46010, Valencia, Spain
| | - Sandra Sancho Castillo
- Faculty of Nursing, Catholic University of Valencia, c/ Espartero, 7, 46007, Valencia, Spain
| | - Mariano Julián-Rochina
- Faculty of Nursing and Podology, University of Valencia, c/ Jaime Roig s/n, 46010, Valencia, Spain
| | | | - María Pilar García-Pardo
- Department of Psychology and Sociology, University of Zaragoza, Campus Ciudad Escolar, 44003, Teruel, Spain.
| |
Collapse
|
18
|
Biber K, Bhattacharya A, Campbell BM, Piro JR, Rohe M, Staal RGW, Talanian RV, Möller T. Microglial Drug Targets in AD: Opportunities and Challenges in Drug Discovery and Development. Front Pharmacol 2019; 10:840. [PMID: 31507408 PMCID: PMC6716448 DOI: 10.3389/fphar.2019.00840] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease (AD) is a large and increasing unmet medical need with no disease-modifying treatment currently available. Genetic evidence from genome-wide association studies (GWASs) and gene network analysis has clearly revealed a key role of the innate immune system in the brain, of which microglia are the most important element. Single-nucleotide polymorphisms (SNPs) in genes predominantly expressed in microglia have been associated with altered risk of developing AD. Furthermore, microglia-specific pathways are affected on the messenger RNA (mRNA) expression level in post-mortem AD tissue and in mouse models of AD. Together these findings have increased the interest in microglia biology, and numerous scientific reports have proposed microglial molecules and pathways as drug targets for AD. Target identification and validation are generally the first steps in drug discovery. Both target validation and drug lead identification for central nervous system (CNS) targets and diseases entail additional significant obstacles compared to peripheral targets and diseases. This makes CNS drug discovery, even with well-validated targets, challenging. In this article, we will illustrate the special challenges of AD drug discovery by discussing the viability/practicality of possible microglia drug targets including cluster of differentiation 33 (CD33), KCa3.1, kynurenines, ionotropic P2 receptor 7 (P2X7), programmed death-1 (PD-1), Toll-like receptors (TLRs), and triggering receptor expressed in myeloid cells 2 (TREM2).
Collapse
Affiliation(s)
- Knut Biber
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Ludwigshafen, Germany
| | | | | | - Justin R Piro
- AbbVie Foundational Neuroscience Center, Cambridge, MA, United States
| | - Michael Rohe
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Ludwigshafen, Germany
| | | | - Robert V Talanian
- AbbVie Foundational Neuroscience Center, Cambridge, MA, United States
| | - Thomas Möller
- AbbVie Foundational Neuroscience Center, Cambridge, MA, United States
| |
Collapse
|
19
|
Hammond TR, Marsh SE, Stevens B. Immune Signaling in Neurodegeneration. Immunity 2019; 50:955-974. [PMID: 30995509 PMCID: PMC6822103 DOI: 10.1016/j.immuni.2019.03.016] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/17/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases of the central nervous system progressively rob patients of their memory, motor function, and ability to perform daily tasks. Advances in genetics and animal models are beginning to unearth an unexpected role of the immune system in disease onset and pathogenesis; however, the role of cytokines, growth factors, and other immune signaling pathways in disease pathogenesis is still being examined. Here we review recent genetic risk and genome-wide association studies and emerging mechanisms for three key immune pathways implicated in disease, the growth factor TGF-β, the complement cascade, and the extracellular receptor TREM2. These immune signaling pathways are important under both healthy and neurodegenerative conditions, and recent work has highlighted new functional aspects of their signaling. Finally, we assess future directions for immune-related research in neurodegeneration and potential avenues for immune-related therapies.
Collapse
Affiliation(s)
- Timothy R Hammond
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samuel E Marsh
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beth Stevens
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
20
|
Bijland S, Thomson G, Euston M, Michail K, Thümmler K, Mücklisch S, Crawford CL, Barnett SC, McLaughlin M, Anderson TJ, Linington C, Brown ER, Kalkman ER, Edgar JM. An in vitro model for studying CNS white matter: functional properties and experimental approaches. F1000Res 2019; 8:117. [PMID: 31069065 PMCID: PMC6489523 DOI: 10.12688/f1000research.16802.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/15/2019] [Indexed: 12/23/2022] Open
Abstract
The normal development and maintenance of CNS white matter, and its responses to disease and injury, are defined by synergies between axons, oligodendrocytes, astrocytes and microglia, and further influenced by peripheral components such as the gut microbiome and the endocrine and immune systems. Consequently, mechanistic insights, therapeutic approaches and safety tests rely ultimately on in vivo models and clinical trials. However, in vitro models that replicate the cellular complexity of the CNS can inform these approaches, reducing costs and minimising the use of human material or experimental animals; in line with the principles of the 3Rs. Using electrophysiology, pharmacology, time-lapse imaging, and immunological assays, we demonstrate that murine spinal cord-derived myelinating cell cultures recapitulate spinal-like electrical activity and innate CNS immune functions, including responses to disease-relevant myelin debris and pathogen associated molecular patterns (PAMPs). Further, we show they are (i) amenable to siRNA making them suitable for testing gene-silencing strategies; (ii) can be established on microelectrode arrays (MEAs) for electrophysiological studies; and (iii) are compatible with multi-well microplate formats for semi-high throughput screens, maximising information output whilst further reducing animal use. We provide protocols for each of these. Together, these advances increase the utility of this in vitro tool for studying normal and pathological development and function of white matter, and for screening therapeutic molecules or gene targets for diseases such as multiple sclerosis, motor neuron disease or spinal cord injury, whilst avoiding in vivo approaches on experimental animals.
Collapse
Affiliation(s)
- Silvia Bijland
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Gemma Thomson
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Matthew Euston
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Kyriakos Michail
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot Watt University, Edinburgh, EH14 4AS, UK
| | - Katja Thümmler
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Steve Mücklisch
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Colin L Crawford
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Mark McLaughlin
- School of Veterinary Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - T James Anderson
- School of Veterinary Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Christopher Linington
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Euan R Brown
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot Watt University, Edinburgh, EH14 4AS, UK
| | - Eric R Kalkman
- Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Julia M Edgar
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| |
Collapse
|