51
|
Prevention of L-Dopa-Induced Dyskinesias by MPEP Blockade of Metabotropic Glutamate Receptor 5 Is Associated with Reduced Inflammation in the Brain of Parkinsonian Monkeys. Cells 2022; 11:cells11040691. [PMID: 35203338 PMCID: PMC8870609 DOI: 10.3390/cells11040691] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
Proinflammatory markers were found in brains of Parkinson’s disease (PD) patients. After years of L-Dopa symptomatic treatment, most PD patients develop dyskinesias. The relationship between inflammation and L-Dopa-induced dyskinesias (LID) is still unclear. We previously reported that MPEP (a metabotropic glutamate receptor 5 antagonist) reduced the development of LID in de novo MPTP-lesioned monkeys. We thus investigated if MPEP reduced the brain inflammatory response in these MPTP-lesioned monkeys and the relationship to LID. The panmacrophage/microglia marker Iba1, the phagocytosis-related receptor CD68, and the astroglial protein GFAP were measured by Western blots. The L-Dopa-treated dyskinetic MPTP monkeys had increased Iba1 content in the putamen, substantia nigra, and globus pallidus, which was prevented by MPEP cotreatment; similar findings were observed for CD68 contents in the putamen and globus pallidus. There was a strong positive correlation between dyskinesia scores and microglial markers in these regions. GFAP contents were elevated in MPTP + L-Dopa-treated monkeys among these brain regions and prevented by MPEP in the putamen and subthalamic nucleus. In conclusion, these results showed increased inflammatory markers in the basal ganglia associated with LID and revealed that MPEP inhibition of glutamate activity reduced LID and levels of inflammatory markers.
Collapse
|
52
|
Li KL, Huang HY, Ren H, Yang XL. Role of exosomes in the pathogenesis of inflammation in Parkinson's disease. Neural Regen Res 2022; 17:1898-1906. [PMID: 35142665 PMCID: PMC8848593 DOI: 10.4103/1673-5374.335143] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Inflammatory responses, including glial cell activation and peripheral immune cell infiltration, are involved in the pathogenesis of Parkinson’s disease (PD). These inflammatory responses appear to be closely related to the release of extracellular vesicles, such as exosomes. However, the relationships among different forms of glial cell activation, synuclein dysregulation, mitochondrial dysfunction, and exosomes are complicated. This review discusses the multiple roles played by exosomes in PD-associated inflammation and concludes that exosomes can transport toxic α-synuclein oligomers to immature neurons and into the extracellular environment, inducing the oligomerization of α-synuclein in normal neurons. Misfolded α-synuclein causes microglia and astrocytes to activate and secrete exosomes. Glial cell-derived exosomes participate in communications between glial cells and neurons, triggering anti-stress and anti-inflammatory responses, in addition to axon growth. The production and release of mitochondrial vesicles and exosomes establish a new mechanism for linking mitochondrial dysfunction to systemic inflammation associated with PD. Given the relevance of exosomes as mediators of neuron-glia communication in neuroinflammation and neuropathogenesis, new targeted treatment strategies are currently being developed that use these types of extracellular vesicles as drug carriers. Exosome-mediated inflammation may be a promising target for intervention in PD patients.
Collapse
Affiliation(s)
- Ke-Lu Li
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Hong-Yan Huang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hui Ren
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xing-Long Yang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
53
|
Garcia P, Jürgens‐Wemheuer W, Uriarte Huarte O, Michelucci A, Masuch A, Brioschi S, Weihofen A, Koncina E, Coowar D, Heurtaux T, Glaab E, Balling R, Sousa C, Kaoma T, Nicot N, Pfander T, Schulz‐Schaeffer W, Allouche A, Fischer N, Biber K, Kleine‐Borgmann F, Mittelbronn M, Ostaszewski M, Schmit KJ, Buttini M. Neurodegeneration and neuroinflammation are linked, but independent of alpha‐synuclein inclusions, in a seeding/spreading mouse model of Parkinson's disease. Glia 2022; 70:935-960. [PMID: 35092321 PMCID: PMC9305192 DOI: 10.1002/glia.24149] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/07/2022] [Accepted: 01/13/2022] [Indexed: 12/16/2022]
Abstract
A key pathological process in Parkinson's disease (PD) is the transneuronal spreading of α‐synuclein. Alpha‐synuclein (α‐syn) is a presynaptic protein that, in PD, forms pathological inclusions. Other hallmarks of PD include neurodegeneration and microgliosis in susceptible brain regions. Whether it is primarily transneuronal spreading of α‐syn particles, inclusion formation, or other mechanisms, such as inflammation, that cause neurodegeneration in PD is unclear. We used a model of spreading of α‐syn induced by striatal injection of α‐syn preformed fibrils into the mouse striatum to address this question. We performed quantitative analysis for α‐syn inclusions, neurodegeneration, and microgliosis in different brain regions, and generated gene expression profiles of the ventral midbrain, at two different timepoints after disease induction. We observed significant neurodegeneration and microgliosis in brain regions not only with, but also without α‐syn inclusions. We also observed prominent microgliosis in injured brain regions that did not correlate with neurodegeneration nor with inclusion load. Using longitudinal gene expression profiling, we observed early gene expression changes, linked to neuroinflammation, that preceded neurodegeneration, indicating an active role of microglia in this process. Altered gene pathways overlapped with those typical of PD. Our observations indicate that α‐syn inclusion formation is not the major driver in the early phases of PD‐like neurodegeneration, but that microglia, activated by diffusible, oligomeric α‐syn, may play a key role in this process. Our findings uncover new features of α‐syn induced pathologies, in particular microgliosis, and point to the necessity for a broader view of the process of α‐syn spreading.
Collapse
Affiliation(s)
- Pierre Garcia
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
- Luxembourg Center of Neuropathology Dudelange Luxembourg
| | - Wiebke Jürgens‐Wemheuer
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
- Institute of Neuropathology Saarland University Clinic (UKS) Homburg Germany
| | - Oihane Uriarte Huarte
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
- Luxembourg Center of Neuropathology Dudelange Luxembourg
| | - Alessandro Michelucci
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
- Department of Cancer Research Luxembourg Institute of Health Strassen Luxembourg
| | - Annette Masuch
- Department of Psychiatry University of Freiburg Medical Center Freiburg Germany
| | - Simone Brioschi
- Department of Psychiatry University of Freiburg Medical Center Freiburg Germany
| | | | - Eric Koncina
- Department of Life Science and Medicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
| | - Djalil Coowar
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
| | - Tony Heurtaux
- Luxembourg Center of Neuropathology Dudelange Luxembourg
- Department of Life Science and Medicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
| | - Carole Sousa
- Department of Cancer Research Luxembourg Institute of Health Strassen Luxembourg
| | - Tony Kaoma
- Department of Cancer Research Luxembourg Institute of Health Strassen Luxembourg
| | - Nathalie Nicot
- Department of Cancer Research Luxembourg Institute of Health Strassen Luxembourg
| | - Tatjana Pfander
- Institute of Neuropathology Saarland University Clinic (UKS) Homburg Germany
| | | | | | | | - Knut Biber
- Department of Psychiatry University of Freiburg Medical Center Freiburg Germany
| | - Felix Kleine‐Borgmann
- Luxembourg Center of Neuropathology Dudelange Luxembourg
- Department of Cancer Research Luxembourg Institute of Health Strassen Luxembourg
- Faculty of Science, Technology and Medicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
| | - Michel Mittelbronn
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
- Luxembourg Center of Neuropathology Dudelange Luxembourg
- Department of Cancer Research Luxembourg Institute of Health Strassen Luxembourg
- Department of Life Science and Medicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
- Faculty of Science, Technology and Medicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
| | - Kristopher J. Schmit
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
- Luxembourg Center of Neuropathology Dudelange Luxembourg
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine University of Luxembourg Esch‐sur‐Alzette Luxembourg
- Luxembourg Center of Neuropathology Dudelange Luxembourg
| |
Collapse
|
54
|
Li S, Bi G, Han S, Huang R. MicroRNAs Play a Role in Parkinson’s Disease by Regulating Microglia Function: From Pathogenetic Involvement to Therapeutic Potential. Front Mol Neurosci 2022; 14:744942. [PMID: 35126050 PMCID: PMC8814625 DOI: 10.3389/fnmol.2021.744942] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/14/2021] [Indexed: 12/31/2022] Open
Abstract
Parkinson’s disease (PD) is a clinically common neurodegenerative disease of the central nervous system (CNS) characterized by loss of dopamine neurons in the substantia nigra. Microglia (MG), as an innate immune cell in the CNS, are involved in a variety of immunity and inflammatory responses in the CNS. A number of studies have shown that the overactivation of MG is one of the critical pathophysiological mechanisms underlying PD. MicroRNAs (miRNAs) are considered to be an important class of gene expression regulators and are involved in a variety of physiological and pathological mechanisms, including immunity and inflammation. In addition, miRNAs can affect the progress of PD by regulating the expression of various MG genes and the polarization state of the MG. Here, we summarize recent articles and describe the important role of MG pathological polarization in the progression of PD, the diverse mechanisms responsible for how miRNAs regulate MG, and the potential therapeutic prospects of miRNAs for PD. We also propose that the regulation of miRNAs may be a novel protective approach against the pathogenesis of PD.
Collapse
Affiliation(s)
- Silu Li
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guorong Bi
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shunchang Han
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Rui Huang,
| |
Collapse
|
55
|
Gao A, McCoy HM, Zaman V, Shields DC, Banik NL, Haque A. Calpain activation and progression of inflammatory cycles in Parkinson's disease. FRONT BIOSCI-LANDMRK 2022; 27:20. [PMID: 35090325 PMCID: PMC9723550 DOI: 10.31083/j.fbl2701020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/01/2021] [Accepted: 12/14/2021] [Indexed: 07/27/2023]
Abstract
Parkinson's disease (PD) is a progressive, neurodegenerative condition of the central nervous system (CNS) affecting 6.3 million people worldwide with no curative treatments. Current therapies aim to mitigate PD's effects and offer symptomatic relief for patients. Multiple pathways are involved in the pathogenesis of PD, leading to neuroinflammation and the destruction of dopaminergic neurons in the CNS. This review focuses on PD pathology and the role of calpain, a neutral protease, as a regulator of various immune cells such as T-cells, microglia and astrocytes which lead to persistent neuroinflammatory responses and neuronal loss in both the brain and spinal cord (SC). Calpain plays a significant role in the cleavage and aggregation of toxic α-synuclein (α-syn), a presynaptic neural protein, and other organelles, contributing to mitochondrial dysfunction and oxidative stress. α-Syn aggregation results in the formation of Lewy bodies (LB) that further contribute to neuronal damage through lipid bilayer penetration, calcium ion (Ca2+) influx, oxidative stress and damage to the blood brain barrier (BBB). Dysfunctional mitochondria destabilize cytosolic Ca2+ concentrations, raising intracellular Ca2+; this leads to excessive calpain activation and persistent inflammatory responses. α-Syn aggregation also results in the disruption of dopamine synthesis through phosphorylation of tyrosine hydroxylase (TH), a key enzyme involved in the conversion of tyrosine to levodopa (L-DOPA), the amino acid precursor to dopamine. Decreased dopamine levels result in altered dopamine receptor (DR) signaling, ultimately activating pro-inflammatory T-cells to further contribute to the inflammatory response. All of these processes, together, result in neuroinflammation, degeneration and ultimately neuronal death seen in PD. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP-a prodrug to the neurotoxin 1-methyl-4-phenylpyridinium (MPP+)), rotenone (an environmental neurotoxin), and 6-hydroxydopamine (6-OHDA - a neurotoxic synthetic organic compound) induce PD-like conditions when injected into rodents. All three agents work through similar mechanisms and lead to degeneration of dopaminergic neurons in the substantia nigra (SN) and more recently discovered in motor neurons of the spinal cord (SC). These neurotoxins also increase calpain activity, furthering the neuroinflammatory response. Hence, calpain inhibitors have been posited as potential therapeutics for PD to prevent calpain-related inflammation and neurodegenerative responses in not only the SN but the SC as well.
Collapse
Affiliation(s)
- Andrew Gao
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Hannah M. McCoy
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Vandana Zaman
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29401, USA
| | - Donald C. Shields
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Naren L. Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29401, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29401, USA
| |
Collapse
|
56
|
Jaffa AA, Jaffa MA, Moussa M, Ahmed IA, Karam M, Aldeen KS, Al Sayegh R, El-Achkar GA, Nasrallah L, Yehya Y, Habib A, Ziyadeh FN, Eid AH, Kobeissy FH, Jaffa AA. Modulation of Neuro-Inflammatory Signals in Microglia by Plasma Prekallikrein and Neuronal Cell Debris. Front Pharmacol 2021; 12:743059. [PMID: 34867349 PMCID: PMC8636058 DOI: 10.3389/fphar.2021.743059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/28/2021] [Indexed: 11/13/2022] Open
Abstract
Microglia, the resident phagocytes of the central nervous system and one of the key modulators of the innate immune system, have been shown to play a major role in brain insults. Upon activation in response to neuroinflammation, microglia promote the release of inflammatory mediators as well as promote phagocytosis. Plasma prekallikrein (PKall) has been recently implicated as a mediator of neuroinflammation; nevertheless, its role in mediating microglial activation has not been investigated yet. In the current study, we evaluate the mechanisms through which PKall contributes to microglial activation and release of inflammatory cytokines assessing PKall-related receptors and their dynamics. Murine N9-microglial cells were exposed to PKall (2.5 ng/ml), lipopolysaccharide (100 ng/ml), bradykinin (BK, 0.1 μM), and neuronal cell debris (16.5 μg protein/ml). Gene expression of bradykinin 2 receptor (B2KR), protease-activated receptor 2 (PAR-2), along with cytokines and fibrotic mediators were studied. Bioinformatic analysis was conducted to correlate altered protein changes with microglial activation. To assess receptor dynamics, HOE-140 (1 μM) and GB-83 (2 μM) were used to antagonize the B2KR and PAR-2 receptors, respectively. Also, the role of autophagy in modulating microglial response was evaluated. Data from our work indicate that PKall, LPS, BK, and neuronal cell debris resulted in the activation of microglia and enhanced expression/secretion of inflammatory mediators. Elevated increase in inflammatory mediators was attenuated in the presence of HOE-140 and GB-83, implicating the engagement of these receptors in the activation process coupled with an increase in the expression of B2KR and PAR-2. Finally, the inhibition of autophagy significantly enhanced the release of the cytokine IL-6 which were validated via bioinformatics analysis demonstrating the role of PKall in systematic and brain inflammatory processes. Taken together, we demonstrated that PKall can modulate microglial activation via the engagement of PAR-2 and B2KR where PKall acts as a neuromodulator of inflammatory processes.
Collapse
Affiliation(s)
- Aneese A Jaffa
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Miran A Jaffa
- Epidemiology and Population Health Department, Faculty of Health Sciences, American University of Beirut, Beirut, Lebanon
| | - Mayssam Moussa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ibrahim A Ahmed
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mia Karam
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Kawthar Sharaf Aldeen
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rola Al Sayegh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,INSERM-UMR1149, Centre de Recherche sur l'Inflammation, and Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Universite de Paris, Paris, France
| | - Ghewa A El-Achkar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Leila Nasrallah
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yara Yehya
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Aida Habib
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,INSERM-UMR1149, Centre de Recherche sur l'Inflammation, and Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Universite de Paris, Paris, France
| | - Fuad N Ziyadeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Firas H Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
57
|
Karagyaur M, Dzhauari S, Basalova N, Aleksandrushkina N, Sagaradze G, Danilova N, Malkov P, Popov V, Skryabina M, Efimenko A, Tkachuk V. MSC Secretome as a Promising Tool for Neuroprotection and Neuroregeneration in a Model of Intracerebral Hemorrhage. Pharmaceutics 2021; 13:2031. [PMID: 34959314 PMCID: PMC8707464 DOI: 10.3390/pharmaceutics13122031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/11/2021] [Accepted: 11/23/2021] [Indexed: 01/17/2023] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are considered to be critical contributors to injured tissue repair and regeneration, and MSC-based therapeutic approaches have been applied to many peripheral and central neurologic disorders. It has been demonstrated that the beneficial effects of MSC are mainly mediated by the components of their secretome. In the current study, we have explored the neuroprotective potential of the MSC secretome in a rat model of intracerebral hemorrhage and shown that a 10-fold concentrated secretome of human MSC and its combination with the brain-derived neurotrophic factor (BDNF) provided a better survival and neurological outcome of rats within 14 days of intracerebral hemorrhage compared to the negative (non-treated) and positive (BDNF) control groups. We found that it was due to the ability of MSC secretome to stimulate neuron survival under conditions of glutamate-induced neurotoxicity. However, the lesion volume did not shrink in these rats, and this also correlated with prominent microglia activation. We hypothesize that this could be caused by the species-specificity of the used MSC secretome and provide evidence to confirm this. Thus, we have found that allogenic rat MSC secretome was more effective than xenogenic human MSC secretome in the rat intracerebral hemorrhage model: it reduced the volume of the lesion and promoted excellent survival and neurological outcome of the treated rats.
Collapse
Affiliation(s)
- Maxim Karagyaur
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10 Lomonosovsky Ave, 119192 Moscow, Russia; (N.B.); (N.A.); (G.S.); (V.P.); (A.E.); (V.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| | - Stalik Dzhauari
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| | - Nataliya Basalova
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10 Lomonosovsky Ave, 119192 Moscow, Russia; (N.B.); (N.A.); (G.S.); (V.P.); (A.E.); (V.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| | - Natalia Aleksandrushkina
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10 Lomonosovsky Ave, 119192 Moscow, Russia; (N.B.); (N.A.); (G.S.); (V.P.); (A.E.); (V.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| | - Georgy Sagaradze
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10 Lomonosovsky Ave, 119192 Moscow, Russia; (N.B.); (N.A.); (G.S.); (V.P.); (A.E.); (V.T.)
| | - Natalia Danilova
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| | - Pavel Malkov
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| | - Vladimir Popov
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10 Lomonosovsky Ave, 119192 Moscow, Russia; (N.B.); (N.A.); (G.S.); (V.P.); (A.E.); (V.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| | - Mariya Skryabina
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10 Lomonosovsky Ave, 119192 Moscow, Russia; (N.B.); (N.A.); (G.S.); (V.P.); (A.E.); (V.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| | - Vsevolod Tkachuk
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10 Lomonosovsky Ave, 119192 Moscow, Russia; (N.B.); (N.A.); (G.S.); (V.P.); (A.E.); (V.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1 Lomonosovsky Ave, 119192 Moscow, Russia; (S.D.); (N.D.); (P.M.); (M.S.)
| |
Collapse
|
58
|
Shin TH, Manavalan B, Lee DY, Basith S, Seo C, Paik MJ, Kim SW, Seo H, Lee JY, Kim JY, Kim AY, Chung JM, Baik EJ, Kang SH, Choi DK, Kang Y, Maral Mouradian M, Lee G. Silica-coated magnetic-nanoparticle-induced cytotoxicity is reduced in microglia by glutathione and citrate identified using integrated omics. Part Fibre Toxicol 2021; 18:42. [PMID: 34819099 PMCID: PMC8614058 DOI: 10.1186/s12989-021-00433-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/25/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Nanoparticles have been utilized in brain research and therapeutics, including imaging, diagnosis, and drug delivery, owing to their versatile properties compared to bulk materials. However, exposure to nanoparticles leads to their accumulation in the brain, but drug development to counteract this nanotoxicity remains challenging. To date, concerns have risen about the potential toxicity to the brain associated with nanoparticles exposure via penetration of the brain blood barrier to address this issue. METHODS Here the effect of silica-coated-magnetic nanoparticles containing the rhodamine B isothiocyanate dye [MNPs@SiO2(RITC)] were assessed on microglia through toxicological investigation, including biological analysis and integration of transcriptomics, proteomics, and metabolomics. MNPs@SiO2(RITC)-induced biological changes, such as morphology, generation of reactive oxygen species, intracellular accumulation of MNPs@SiO2(RITC) using transmission electron microscopy, and glucose uptake efficiency, were analyzed in BV2 murine microglial cells. Each omics data was collected via RNA-sequencing-based transcriptome analysis, liquid chromatography-tandem mass spectrometry-based proteome analysis, and gas chromatography- tandem mass spectrometry-based metabolome analysis. The three omics datasets were integrated and generated as a single network using a machine learning algorithm. Nineteen compounds were screened and predicted their effects on nanotoxicity within the triple-omics network. RESULTS Intracellular reactive oxygen species production, an inflammatory response, and morphological activation of cells were greater, but glucose uptake was lower in MNPs@SiO2(RITC)-treated BV2 microglia and primary rat microglia in a dose-dependent manner. Expression of 121 genes (from 41,214 identified genes), and levels of 45 proteins (from 5918 identified proteins) and 17 metabolites (from 47 identified metabolites) related to the above phenomena changed in MNPs@SiO2(RITC)-treated microglia. A combination of glutathione and citrate attenuated nanotoxicity induced by MNPs@SiO2(RITC) and ten other nanoparticles in vitro and in the murine brain, protecting mostly the hippocampus and thalamus. CONCLUSIONS Combination of glutathione and citrate can be one of the candidates for nanotoxicity alleviating drug against MNPs@SiO2(RITC) induced detrimental effect, including elevation of intracellular reactive oxygen species level, activation of microglia, and reduction in glucose uptake efficiency. In addition, our findings indicate that an integrated triple omics approach provides useful and sensitive toxicological assessment for nanoparticles and screening of drug for nanotoxicity.
Collapse
Affiliation(s)
- Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Balachandran Manavalan
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Da Yeon Lee
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Chan Seo
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon, 57922 Republic of Korea
| | - Man Jeong Paik
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon, 57922 Republic of Korea
| | - Sang-Wook Kim
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Haewoon Seo
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Ju Yeon Lee
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanji-ro, Cheongju, 28119 Republic of Korea
| | - Jin Young Kim
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanji-ro, Cheongju, 28119 Republic of Korea
| | - A Young Kim
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Jee Min Chung
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Eun Joo Baik
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Seong Ho Kang
- Department of Chemistry, Graduate School, Kyung Hee University, Yongin-si, Gyeonggi-do 17104 Republic of Korea
- Department of Applied Chemistry and Institute of Natural Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104 Republic of Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, 268 Chungwondaero, Chungju, 27478 Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - M. Maral Mouradian
- RWJMS Institute for Neurological Therapeutics, Rutgers Biomedical and Health Sciences, and Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854 USA
| | - Gwang Lee
- Department of Molecular Science and Technology, Ajou University, Suwon-si, Gyeonggi-do 16499 Republic of Korea
- Department of Physiology, Ajou University School of Medicine, Suwon-si, Gyeonggi-do 16499 Republic of Korea
| |
Collapse
|
59
|
Badanjak K, Mulica P, Smajic S, Delcambre S, Tranchevent LC, Diederich N, Rauen T, Schwamborn JC, Glaab E, Cowley SA, Antony PMA, Pereira SL, Venegas C, Grünewald A. iPSC-Derived Microglia as a Model to Study Inflammation in Idiopathic Parkinson's Disease. Front Cell Dev Biol 2021; 9:740758. [PMID: 34805149 PMCID: PMC8602578 DOI: 10.3389/fcell.2021.740758] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with unknown cause in the majority of patients, who are therefore considered "idiopathic" (IPD). PD predominantly affects dopaminergic neurons in the substantia nigra pars compacta (SNpc), yet the pathology is not limited to this cell type. Advancing age is considered the main risk factor for the development of IPD and greatly influences the function of microglia, the immune cells of the brain. With increasing age, microglia become dysfunctional and release pro-inflammatory factors into the extracellular space, which promote neuronal cell death. Accordingly, neuroinflammation has also been described as a feature of PD. So far, studies exploring inflammatory pathways in IPD patient samples have primarily focused on blood-derived immune cells or brain sections, but rarely investigated patient microglia in vitro. Accordingly, we decided to explore the contribution of microglia to IPD in a comparative manner using, both, iPSC-derived cultures and postmortem tissue. Our meta-analysis of published RNAseq datasets indicated an upregulation of IL10 and IL1B in nigral tissue from IPD patients. We observed increased expression levels of these cytokines in microglia compared to neurons using our single-cell midbrain atlas. Moreover, IL10 and IL1B were upregulated in IPD compared to control microglia. Next, to validate these findings in vitro, we generated IPD patient microglia from iPSCs using an established differentiation protocol. IPD microglia were more readily primed as indicated by elevated IL1B and IL10 gene expression and higher mRNA and protein levels of NLRP3 after LPS treatment. In addition, IPD microglia had higher phagocytic capacity under basal conditions-a phenotype that was further exacerbated upon stimulation with LPS, suggesting an aberrant microglial function. Our results demonstrate the significance of microglia as the key player in the neuroinflammation process in IPD. While our study highlights the importance of microglia-mediated inflammatory signaling in IPD, further investigations will be needed to explore particular disease mechanisms in these cells.
Collapse
Affiliation(s)
- Katja Badanjak
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Patrycja Mulica
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Semra Smajic
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Sylvie Delcambre
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | | | - Nico Diederich
- Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Thomas Rauen
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Jens C. Schwamborn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Sally A. Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Paul M. A. Antony
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
- Disease Modeling and Screening Platform (DMSP), Luxembourg Institute of Systems Biomedicine, University of Luxembourg and Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Sandro L. Pereira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Carmen Venegas
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
60
|
Mishra A, Bandopadhyay R, Singh PK, Mishra PS, Sharma N, Khurana N. Neuroinflammation in neurological disorders: pharmacotherapeutic targets from bench to bedside. Metab Brain Dis 2021; 36:1591-1626. [PMID: 34387831 DOI: 10.1007/s11011-021-00806-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
Neuroinflammation is one of the host defensive mechanisms through which the nervous system protects itself from pathogenic and or infectious insults. Moreover, neuroinflammation occurs as one of the most common pathological outcomes in various neurological disorders, makes it the promising target. The present review focuses on elaborating the recent advancement in understanding molecular mechanisms of neuroinflammation and its role in the etiopathogenesis of various neurological disorders, especially Alzheimer's disease (AD), Parkinson's disease (PD), and Epilepsy. Furthermore, the current status of anti-inflammatory agents in neurological diseases has been summarized in light of different preclinical and clinical studies. Finally, possible limitations and future directions for the effective use of anti-inflammatory agents in neurological disorders have been discussed.
Collapse
Affiliation(s)
- Awanish Mishra
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India.
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, 781101, India.
| | - Ritam Bandopadhyay
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Prabhakar Kumar Singh
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Pragya Shakti Mishra
- Department of Nuclear Medicine, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGIMS), Raebareli Road, Lucknow, 226014, India
| | - Neha Sharma
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Navneet Khurana
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| |
Collapse
|
61
|
Ahuja S, Lazar IM. Systems-Level Proteomics Evaluation of Microglia Response to Tumor-Supportive Anti-Inflammatory Cytokines. Front Immunol 2021; 12:646043. [PMID: 34566949 PMCID: PMC8458581 DOI: 10.3389/fimmu.2021.646043] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/06/2021] [Indexed: 12/24/2022] Open
Abstract
Background Microglia safeguard the CNS against injuries and pathogens, and in the presence of certain harmful stimuli are capable of inducing a disease-dependent inflammatory response. When exposed to anti-inflammatory cytokines, however, these cells possess the ability to switch from an inflammatory to an immunosuppressive phenotype. Cancer cells exploit this property to evade the immune system, and elicit an anti-inflammatory microenvironment that facilitates tumor attachment and growth. Objective The tumor-supportive biological processes that are activated in microglia cells in response to anti-inflammatory cytokines released from cancer cells were explored with mass spectrometry and proteomic technologies. Methods Serum-depleted and non-depleted human microglia cells (HMC3) were treated with a cocktail of IL-4, IL-13, IL-10, TGFβ, and CCL2. The cellular protein extracts were analyzed by LC-MS/MS. Using functional annotation clustering tools, statistically significant proteins that displayed a change in abundance between cytokine-treated and non-treated cells were mapped to their biological networks and pathways. Results The proteomic analysis of HMC3 cells enabled the identification of ~10,000 proteins. Stimulation with anti-inflammatory cytokines resulted in the activation of distinct, yet integrated clusters of proteins that trigger downstream a number of tumor-promoting biological processes. The observed changes could be classified into four major categories, i.e., mitochondrial gene expression, ECM remodeling, immune response, and impaired cell cycle progression. Intracellular immune activation was mediated mainly by the transducers of MAPK, STAT, TGFβ, NFKB, and integrin signaling pathways. Abundant collagen formation along with the expression of additional receptors, matrix components, growth factors, proteases and protease inhibitors, was indicative of ECM remodeling processes supportive of cell-cell and cell-matrix adhesion. Overexpression of integrins and their modulators was reflective of signaling processes that link ECM reorganization with cytoskeletal re-arrangements supportive of cell migration. Antigen processing/presentation was represented by HLA class I histocompatibility antigens, and correlated with upregulated proteasomal subunits, vesicular/viral transport, and secretory processes. Immunosuppressive and proangiogenic chemokines, as well as anti-angiogenic factors, were detectable in low abundance. Pronounced pro-inflammatory, chemotactic or phagocytic trends were not observed, however, the expression of certain receptors, signaling and ECM proteins indicated the presence of such capabilities. Conclusions Comprehensive proteomic profiling of HMC3 cells stimulated with anti-inflammatory cytokines revealed a spectrum of microglia phenotypes supportive of cancer development in the brain via microenvironment-dependent biological mechanisms.
Collapse
Affiliation(s)
- Shreya Ahuja
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Iulia M. Lazar
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
- Carilion School of Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
62
|
Maguire E, Menzies GE, Phillips T, Sasner M, Williams HM, Czubala MA, Evans N, Cope EL, Sims R, Howell GR, Lloyd-Evans E, Williams J, Allen ND, Taylor PR. PIP2 depletion and altered endocytosis caused by expression of Alzheimer's disease-protective variant PLCγ2 R522. EMBO J 2021; 40:e105603. [PMID: 34254352 PMCID: PMC8408593 DOI: 10.15252/embj.2020105603] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/06/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Variants identified in genome-wide association studies have implicated immune pathways in the development of Alzheimer's disease (AD). Here, we investigated the mechanistic basis for protection from AD associated with PLCγ2 R522, a rare coding variant of the PLCG2 gene. We studied the variant's role in macrophages and microglia of newly generated PLCG2-R522-expressing human induced pluripotent cell lines (hiPSC) and knockin mice, which exhibit normal endogenous PLCG2 expression. In all models, cells expressing the R522 mutation show a consistent non-redundant hyperfunctionality in the context of normal expression of other PLC isoforms. This manifests as enhanced release of cellular calcium ion stores in response to physiologically relevant stimuli like Fc-receptor ligation or exposure to Aβ oligomers. Expression of the PLCγ2-R522 variant resulted in increased stimulus-dependent PIP2 depletion and reduced basal PIP2 levels in vivo. Furthermore, it was associated with impaired phagocytosis and enhanced endocytosis. PLCγ2 acts downstream of other AD-related factors, such as TREM2 and CSF1R, and alterations in its activity directly impact cell function. The inherent druggability of enzymes such as PLCγ2 raises the prospect of PLCγ2 manipulation as a future therapeutic approach in AD.
Collapse
Affiliation(s)
- Emily Maguire
- UK Dementia Research Institute at Cardiff, Cardiff, UK
| | - Georgina E Menzies
- UK Dementia Research Institute at Cardiff, Cardiff, UK.,School of Biosciences, Cardiff University, Cardiff, UK
| | | | | | | | | | - Neil Evans
- UK Dementia Research Institute at Cardiff, Cardiff, UK
| | - Emma L Cope
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Rebecca Sims
- MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff, UK
| | | | | | - Julie Williams
- UK Dementia Research Institute at Cardiff, Cardiff, UK.,MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff, UK
| | | | - Philip R Taylor
- UK Dementia Research Institute at Cardiff, Cardiff, UK.,Systems Immunity University Research Institute, Cardiff, UK
| |
Collapse
|
63
|
Ren J, Dai C, Zhou X, Barnes JA, Chen X, Wang Y, Yuan L, Shingu T, Heimberger AB, Chen Y, Hu J. Qki is an essential regulator of microglial phagocytosis in demyelination. J Exp Med 2021; 218:191206. [PMID: 33045062 PMCID: PMC7543092 DOI: 10.1084/jem.20190348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 08/07/2019] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
The mechanism underpinning the regulation of microglial phagocytosis in demyelinating diseases is unclear. Here, we showed that the Quaking protein (Qki) in microglia was greatly induced by demyelination in the brains of both mice and humans. Deletion of the Quaking gene (Qk) in microglia severely impaired the clearance of myelin debris. Transcriptomic profiling indicated that depletion of Qki impaired total RNA levels and splicing of the genes involved in phagosome formation and maturation. RNA immunoprecipitation (RIP) confirmed the physical interactions between the Qki protein and the mRNAs of Qki targets that are involved in phagocytosis, indicating that Qki regulates their RNA stability. Both Qki depletion and inhibition of Qki target Cd36 greatly reduced the phagocytic activity of microglia and macrophages. The defective uptake and degradation of myelin debris caused by Qki depletion in microglia resulted in unresolved myelin debris that impaired axon integrity, oligodendrocyte maturation, and subsequent remyelination. Thus, our results demonstrate that Qki is an essential regulator of microglia’s phagocytic activity under demyelinating conditions.
Collapse
Affiliation(s)
- Jiangong Ren
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Congxin Dai
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Neurosurgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xin Zhou
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Joseph A Barnes
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX.,The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX
| | - Xi Chen
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yunfei Wang
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Liang Yuan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Graduate School of Biomedical Sciences, Tufts University, Boston, MA
| | - Takashi Shingu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yiwen Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jian Hu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX.,The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX
| |
Collapse
|
64
|
Li L, Fan S, Zhang W, Li D, Yang Z, Zhuang P, Han J, Guo H, Zhang Y. Duzhong Fang Attenuates the POMC-Derived Neuroinflammation in Parkinsonian Mice. J Inflamm Res 2021; 14:3261-3276. [PMID: 34326654 PMCID: PMC8315774 DOI: 10.2147/jir.s316314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/01/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Neuroinflammation and microglia reactivity are now recognized to be features of Parkinson's disease (PD). Thus, microglia phenotype is a potential new target for developing treatments against PD. Duzhong Fang (DZF) is a traditional Chinese medicine (TCM) prescription. The theory of TCM argues that Duzhong Fang, nourishing yin and tonifying yang, may treat PD. However, its modern pharmacological studies and the underlying mechanisms are unclear. METHODS First, MPTP was used to establish a parkinsonian mouse model, and behavioral testing was used to evaluate the locomotor dysfunction. Then, HPLC, immunohistochemical staining, and Western blot assays were performed to evaluate the survival of dopaminergic neurons. Molecular biological and immunofluorescence staining were used to evaluate the neuroinflammation and microglial activation. In addition, RNA-seq transcriptomics was used to analyze differentially expressed genes and verify by RT-PCR. RESULTS In the present study, we first confirmed that DZF can alleviate neuroinflammation and ameliorate dyskinesia in parkinsonian mice. Then, further studies found that DZF can regulate microglial morphology and reactivity and act on the POMC gene. POMC is an upstream target for regulating inflammation and proinflammatory cytokines, and DZF can directly inhibit the POMC level and restore the homeostatic signature of microglia in parkinsonian mice. CONCLUSION This study found that POMC may have a potential role as a therapeutic target for PD. DZF may inhibit neuroinflammation and play an anti-PD effect by down-regulating the expression of POMC.
Collapse
Affiliation(s)
- Lili Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Shanshan Fan
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Wenqi Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Dongna Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Zhen Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Pengwei Zhuang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Juan Han
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Hong Guo
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Yanjun Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| |
Collapse
|
65
|
Thomsen BB, Madsen C, Krohn KT, Thygesen C, Schütt T, Metaxas A, Darvesh S, Agerholm JS, Wirenfeldt M, Berendt M, Finsen B. Mild Microglial Responses in the Cortex and Perivascular Macrophage Infiltration in Subcortical White Matter in Dogs with Age-Related Dementia Modelling Prodromal Alzheimer's Disease. J Alzheimers Dis 2021; 82:575-592. [PMID: 34057083 PMCID: PMC8385501 DOI: 10.3233/jad-210040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Microglia contribute to Alzheimer’s disease (AD) pathogenesis by clearing amyloid-β (Aβ) and driving neuroinflammation. Domestic dogs with age-related dementia (canine cognitive dysfunction (CCD)) develop cerebral amyloidosis like humans developing AD, and studying such dogs can provide novel information about microglial response in prodromal AD. Objective: The aim was to investigate the microglial response in the cortical grey and the subcortical white matter in dogs with CCD versus age-matched cognitively normal dogs. Methods: Brains from aged dogs with CCD and age-matched controls without dementia were studied. Cases were defined by dementia rating score. Brain sections were stained for Aβ, thioflavin S, hyperphosphorylated tau, and the microglial-macrophage ionized calcium binding adaptor molecule 1 (Iba1). Results were correlated to dementia rating score and tissue levels of Aβ. Results: Microglial numbers were higher in the Aβ plaque-loaded deep cortical layers in CCD versus control dogs, while the coverage by microglial processes were comparable. Aβ plaques were of the diffuse type and without microglial aggregation. However, a correlation was found between the %Iba1 area and insoluble Aβ 42 and N-terminal pyroglutamate modified Aβ(N3pE)-42. The %Iba1 area was higher in white matter, showing phosphorylation of S396 tau, versus grey matter. Perivascular macrophage infiltrates were abundant in the white matter particularly in CDD dogs. Conclusion: The results from this study of the microglial-macrophage response in dogs with CCD are suggestive of relatively mild microglial responses in the Aβ plaque-loaded deep cortical layers and perivascular macrophage infiltrates in the subcortical white matter, in prodromal AD.
Collapse
Affiliation(s)
- Barbara Blicher Thomsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Madsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Katrine Tækker Krohn
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Camilla Thygesen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Trine Schütt
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Athanasios Metaxas
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,School of Science, Department of Life Sciences, European University Cyprus, Nicosia, Cyprus
| | - Sultan Darvesh
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada.,Division of Neurology and Geriatric Medicine, Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Jørgen Steen Agerholm
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Wirenfeldt
- BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Institute of Clinical Science, Odense University Hospital, Odense, Denmark
| | - Mette Berendt
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE: Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
66
|
Sinha A, Kushwaha R, Molesworth K, Mychko O, Makarava N, Baskakov IV. Phagocytic Activities of Reactive Microglia and Astrocytes Associated with Prion Diseases Are Dysregulated in Opposite Directions. Cells 2021; 10:1728. [PMID: 34359897 PMCID: PMC8304827 DOI: 10.3390/cells10071728] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 01/11/2023] Open
Abstract
Phagocytosis is one of the most important physiological functions of the glia directed at maintaining a healthy, homeostatic environment in the brain. Under a homeostatic environment, the phagocytic activities of astrocytes and microglia are tightly coordinated in time and space. In neurodegenerative diseases, both microglia and astrocytes contribute to neuroinflammation and disease pathogenesis, however, whether their phagocytic activities are up- or downregulated in reactive states is not known. To address this question, this current study isolated microglia and astrocytes from C57BL/6J mice infected with prions and tested their phagocytic activities in live-cell imaging assays that used synaptosomes and myelin debris as substrates. The phagocytic uptake by the reactive microglia was found to be significantly upregulated, whereas that of the reactive astrocytes was strongly downregulated. The up- and downregulation of phagocytosis by the two cell types were observed irrespective of whether disease-associated synaptosomes, normal synaptosomes, or myelin debris were used in the assays, indicating that dysregulations are dictated by cell reactive states, not substrates. Analysis of gene expression confirmed dysregulation of phagocytic functions in both cell types. Immunostaining of animal brains infected with prions revealed that at the terminal stage of disease, neuronal cell bodies were subject to engulfment by reactive microglia. This study suggests that imbalance in the phagocytic activities of the reactive microglia and astrocytes, which are dysregulated in opposite directions, is likely to lead to excessive microglia-mediated neuronal death on the one hand, and the inability of astrocytes to clear cell debris on the other hand, contributing to the neurotoxic effects of glia as a whole.
Collapse
Affiliation(s)
- Anshuman Sinha
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (R.K.); (K.M.); (O.M.); (N.M.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rajesh Kushwaha
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (R.K.); (K.M.); (O.M.); (N.M.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kara Molesworth
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (R.K.); (K.M.); (O.M.); (N.M.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Olga Mychko
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (R.K.); (K.M.); (O.M.); (N.M.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (R.K.); (K.M.); (O.M.); (N.M.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ilia V. Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.S.); (R.K.); (K.M.); (O.M.); (N.M.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
67
|
Heras-Garvin A, Refolo V, Schmidt C, Malfertheiner K, Wenning GK, Bradbury M, Stamler D, Stefanova N. ATH434 Reduces α-Synuclein-Related Neurodegeneration in a Murine Model of Multiple System Atrophy. Mov Disord 2021; 36:2605-2614. [PMID: 34236731 DOI: 10.1002/mds.28714] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 06/02/2021] [Accepted: 06/14/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Multiple system atrophy (MSA) is a fatal neurodegenerative disorder characterized by aggregated α-synuclein (α-syn) in oligodendrocytes and accompanied by striatonigral and olivopontocerebellar degeneration and motor symptoms. Key features of MSA are replicated in the PLP-α-syn transgenic mouse, including progressive striatonigral degeneration and motor deterioration. There are currently no approved treatments for MSA. ATH434 is a novel, orally bioavailable brain penetrant small molecule inhibitor of α-syn aggregation. OBJECTIVES To characterize ATH434 for disease modification in a mouse model of MSA. METHODS Six-month-old PLP-α-syn mice (MSA mice) were ATH434-treated (ATH434 in food) or untreated (normal food) for 6 months. Motor behavior and numbers of nigral and striatal neurons were evaluated. α-syn aggregates and oligomers were quantified by immunohistochemical and western blot analyses. Microglial activation and neuroinflammation were assessed by histological and molecular analyses. Ferric iron in the Substantia nigra was evaluated with the Perls method. RESULTS ATH434-treated mice demonstrated preservation of motor performance in MSA mice that was associated with neuroprotection of nigral and striatal neurons. The rescue of the phenotype correlated with the reduction of α-syn inclusions and oligomers in animals receiving ATH434. ATH434-treated mice exhibited significantly increased lysosomal activity of microglia without increased pro-inflammatory markers, suggesting a role in α-syn clearing. ATH434-treatment was associated with lower intracellular nigral iron levels. CONCLUSIONS Our findings demonstrate the beneficial disease-modifying effect of ATH434 in oligodendroglial α-synucleinopathy on both the motor phenotype and neurodegenerative pathology in the PLP-α-syn transgenic mouse and support the development of ATH434 for MSA. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Antonio Heras-Garvin
- Laboratory for Translational Neurodegeneration Research, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Violetta Refolo
- Laboratory for Translational Neurodegeneration Research, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudio Schmidt
- Laboratory for Translational Neurodegeneration Research, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Katja Malfertheiner
- Laboratory for Translational Neurodegeneration Research, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor K Wenning
- Laboratory for Translational Neurodegeneration Research, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | | | - Nadia Stefanova
- Laboratory for Translational Neurodegeneration Research, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
68
|
Jeon MT, Kim KS, Kim ES, Lee S, Kim J, Hoe HS, Kim DG. Emerging pathogenic role of peripheral blood factors following BBB disruption in neurodegenerative disease. Ageing Res Rev 2021; 68:101333. [PMID: 33774194 DOI: 10.1016/j.arr.2021.101333] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/03/2021] [Accepted: 03/19/2021] [Indexed: 12/15/2022]
Abstract
The responses of central nervous system (CNS) cells such as neurons and glia in neurodegenerative diseases (NDs) suggest that regulation of neuronal and glial functions could be a strategy for ND prevention and/or treatment. However, attempts to develop such therapeutics for NDs have been hindered by the challenge of blood-brain barrier (BBB) permeability and continued constitutive neuronal loss. These limitations indicate the need for additional perspectives for the prevention/treatment of NDs. In particular, the disruption of the blood-brain barrier (BBB) that accompanies NDs allows brain infiltration by peripheral factors, which may stimulate innate immune responses involved in the progression of neurodegeneration. The accumulation of blood factors like thrombin, fibrinogen, c-reactive protein (CRP) and complement components in the brain has been observed in NDs and may activate the innate immune system in the CNS. Thus, strengthening the integrity of the BBB may enhance its protective role to attenuate ND progression and functional loss. In this review, we describe the innate immune system in the CNS and the contribution of blood factors to the role of the CNS immune system in neurodegeneration and neuroprotection.
Collapse
Affiliation(s)
- Min-Tae Jeon
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea
| | - Kyu-Sung Kim
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Republic of Korea
| | - Eun Seon Kim
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Republic of Korea
| | - Suji Lee
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, 16 De Crespigny Park, London, SE5 8AF, UK
| | - Jieun Kim
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea
| | - Hyang-Sook Hoe
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Republic of Korea.
| | - Do-Geun Kim
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea.
| |
Collapse
|
69
|
Pascual M, Calvo-Rodriguez M, Núñez L, Villalobos C, Ureña J, Guerri C. Toll-like receptors in neuroinflammation, neurodegeneration, and alcohol-induced brain damage. IUBMB Life 2021; 73:900-915. [PMID: 34033211 DOI: 10.1002/iub.2510] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/05/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) or pattern recognition receptors respond to pathogen-associated molecular patterns (PAMPs) or internal damage-associated molecular patterns (DAMPs). TLRs are integral membrane proteins with both extracellular leucine-rich and cytoplasmic domains that initiate downstream signaling through kinases by activating transcription factors like AP-1 and NF-κB, which lead to the release of various inflammatory cytokines and immune modulators. In the central nervous system, different TLRs are expressed mainly in microglia and astroglial cells, although some TLRs are also expressed in oligodendroglia and neurons. Activation of TLRs triggers signaling cascades by the host as a defense mechanism against invaders to repair damaged tissue. However, overactivation of TLRs disrupts the sustained immune homeostasis-induced production of pro-inflammatory molecules, such as cytokines, miRNAs, and inflammatory components of extracellular vesicles. These inflammatory mediators can, in turn, induce neuroinflammation, and neural tissue damage associated with many neurodegenerative diseases. This review discusses the critical role of TLRs response in Alzheimer's disease, Parkinson's disease, ischemic stroke, amyotrophic lateral sclerosis, and alcohol-induced brain damage and neurodegeneration.
Collapse
Affiliation(s)
- María Pascual
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Maria Calvo-Rodriguez
- Alzheimer's Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Lucía Núñez
- Institute of Biology and Molecular Genetics (IBGM), University of Valladolid and National Research Council (CSIC), Valladolid, Spain.,Department of Biochemistry and Molecular Biology and Physiology, University of Valladolid, Valladolid, Spain
| | - Carlos Villalobos
- Institute of Biology and Molecular Genetics (IBGM), University of Valladolid and National Research Council (CSIC), Valladolid, Spain
| | - Juan Ureña
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain
| |
Collapse
|
70
|
Wyatt-Johnson SK, Sommer AL, Shim KY, Brewster AL. Suppression of Microgliosis With the Colony-Stimulating Factor 1 Receptor Inhibitor PLX3397 Does Not Attenuate Memory Defects During Epileptogenesis in the Rat. Front Neurol 2021; 12:651096. [PMID: 34149593 PMCID: PMC8209304 DOI: 10.3389/fneur.2021.651096] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/11/2021] [Indexed: 12/03/2022] Open
Abstract
Events of status epilepticus (SE) trigger the development of temporal lobe epilepsy (TLE), a type of focal epilepsy that is commonly drug-resistant and is highly comorbid with cognitive deficits. While SE-induced hippocampal injury, accompanied by gliosis and neuronal loss, typically disrupts cognitive functions resulting in memory defects, it is not definitively known how. Our previous studies revealed extensive hippocampal microgliosis that peaked between 2 and 3 weeks after SE and paralleled the development of cognitive impairments, suggesting a role for reactive microglia in this pathophysiology. Microglial survival and proliferation are regulated by the colony-stimulating factor 1 receptor (CSF1R). The CSF1R inhibitor PLX3397 has been shown to reduce/deplete microglial populations and improve cognitive performance in models of neurodegenerative disorders. Therefore, we hypothesized that suppression of microgliosis with PLX3397 during epileptogenesis may attenuate the hippocampal-dependent spatial learning and memory deficits in the rat pilocarpine model of SE and acquired TLE. Different groups of control and SE rats were fed standard chow (SC) or chow with PLX3397 starting immediately after SE and for 3 weeks. Novel object recognition (NOR) and Barnes maze (BM) were performed to determine memory function between 2 and 3 weeks after SE. Then microglial populations were assessed using immunohistochemistry. Control rats fed with either SC or PLX3397 performed similarly in both NOR and BM tests, differentiating novel vs. familiar objects in NOR, and rapidly learning the location of the hidden platform in BM. In contrast, both SE groups (SC and PLX3397) showed significant deficits in both NOR and BM tests compared to controls. Both PLX3397-treated control and SE groups had significantly decreased numbers of microglia in the hippocampus (60%) compared to those in SC. In parallel, we found that PLX3397 treatment also reduced SE-induced hippocampal astrogliosis. Thus, despite drastic reductions in microglial cells, memory was unaffected in the PLX3397-treated groups compared to those in SC, suggesting that remaining microglia may be sufficient to help maintain hippocampal functions. In sum, PLX3397 did not improve or worsen the memory deficits in rats that sustained pilocarpine-induced SE. Further research is required to determine whether microglia play a role in cognitive decline during epileptogenesis.
Collapse
Affiliation(s)
- Season K Wyatt-Johnson
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, United States
| | - Alexandra L Sommer
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, United States
| | - Kevin Y Shim
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, United States
| | - Amy L Brewster
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, United States.,Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
71
|
Tran AP, Warren PM, Silver J. New insights into glial scar formation after spinal cord injury. Cell Tissue Res 2021; 387:319-336. [PMID: 34076775 PMCID: PMC8975767 DOI: 10.1007/s00441-021-03477-w] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
Severe spinal cord injury causes permanent loss of function and sensation throughout the body. The trauma causes a multifaceted torrent of pathophysiological processes which ultimately act to form a complex structure, permanently remodeling the cellular architecture and extracellular matrix. This structure is traditionally termed the glial/fibrotic scar. Similar cellular formations occur following stroke, infection, and neurodegenerative diseases of the central nervous system (CNS) signifying their fundamental importance to preservation of function. It is increasingly recognized that the scar performs multiple roles affecting recovery following traumatic injury. Innovative research into the properties of this structure is imperative to the development of treatment strategies to recover motor function and sensation following CNS trauma. In this review, we summarize how the regeneration potential of the CNS alters across phyla and age through formation of scar-like structures. We describe how new insights from next-generation sequencing technologies have yielded a more complex portrait of the molecular mechanisms governing the astrocyte, microglial, and neuronal responses to injury and development, especially of the glial component of the scar. Finally, we discuss possible combinatorial therapeutic approaches centering on scar modulation to restore function after severe CNS injury.
Collapse
Affiliation(s)
- Amanda Phuong Tran
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Philippa Mary Warren
- Wolfson Centre for Age Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Guy's Campus, London Bridge, London, UK
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
72
|
Bocharova O, Pandit NP, Molesworth K, Fisher A, Mychko O, Makarava N, Baskakov IV. Alzheimer's disease-associated β-amyloid does not protect against herpes simplex virus 1 infection in the mouse brain. J Biol Chem 2021; 297:100845. [PMID: 34052228 PMCID: PMC8214219 DOI: 10.1016/j.jbc.2021.100845] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/13/2021] [Accepted: 05/26/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating fatal neurodegenerative disease. An alternative to the amyloid cascade hypothesis is that a viral infection is key to the etiology of late-onset AD, with β-amyloid (Aβ) peptides playing a protective role. In the current study, young 5XFAD mice that overexpress mutant human amyloid precursor protein with the Swedish, Florida, and London familial AD mutations were infected with one of two strains of herpes simplex virus 1 (HSV-1), 17syn+ and McKrae, at three different doses. Contrary to previous work, 5XFAD genotype failed to protect mice against HSV-1 infection. The region- and cell-specific tropisms of HSV-1 were not affected by the 5XFAD genotype, indicating that host–pathogen interactions were not altered. Seven- to ten-month-old 5XFAD animals in which extracellular Aβ aggregates were abundant showed slightly better survival rate relative to their wild-type (WT) littermates, although the difference was not statistically significant. In these 5XFAD mice, HSV-1 replication centers were partially excluded from the brain areas with high densities of Aβ aggregates. Aβ aggregates were free of HSV-1 viral particles, and the limited viral invasion to areas with a high density of Aβ aggregates was attributed to phagocytic activity of reactive microglia. In the oldest mice (12–15 months old), the survival rate did not differ between 5XFAD and WT littermates. While the current study questions the antiviral role of Aβ, it neither supports nor refutes the viral etiology hypothesis of late-onset AD.
Collapse
Affiliation(s)
- Olga Bocharova
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Narayan P Pandit
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kara Molesworth
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Aidan Fisher
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Olga Mychko
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ilia V Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
73
|
The Potential of Induced Pluripotent Stem Cells to Treat and Model Alzheimer's Disease. Stem Cells Int 2021; 2021:5511630. [PMID: 34122554 PMCID: PMC8172295 DOI: 10.1155/2021/5511630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/20/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022] Open
Abstract
An estimated 6.2 million Americans aged 65 or older are currently living with Alzheimer's disease (AD), a neurodegenerative disease that disrupts an individual's ability to function independently through the degeneration of key regions in the brain, including but not limited to the hippocampus, the prefrontal cortex, and the motor cortex. The cause of this degeneration is not known, but research has found two proteins that undergo posttranslational modifications: tau, a protein concentrated in the axons of neurons, and amyloid precursor protein (APP), a protein concentrated near the synapse. Through mechanisms that have yet to be elucidated, the accumulation of these two proteins in their abnormal aggregate forms leads to the neurodegeneration that is characteristic of AD. Until the invention of induced pluripotent stem cells (iPSCs) in 2006, the bulk of research was carried out using transgenic animal models that offered little promise in their ability to translate well from benchtop to bedside, creating a bottleneck in the development of therapeutics. However, with iPSC, patient-specific cell cultures can be utilized to create models based on human cells. These human cells have the potential to avoid issues in translatability that have plagued animal models by providing researchers with a model that closely resembles and mimics the neurons found in humans. By using human iPSC technology, researchers can create more accurate models of AD ex vivo while also focusing on regenerative medicine using iPSC in vivo. The following review focuses on the current uses of iPSC and how they have the potential to regenerate damaged neuronal tissue, in the hopes that these technologies can assist in getting through the bottleneck of AD therapeutic research.
Collapse
|
74
|
Carotenoid Extract Derived from Euglena gracilis Overcomes Lipopolysaccharide-Induced Neuroinflammation in Microglia: Role of NF-κB and Nrf2 Signaling Pathways. Mol Neurobiol 2021; 58:3515-3528. [PMID: 33745115 PMCID: PMC8257518 DOI: 10.1007/s12035-021-02353-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/09/2021] [Indexed: 01/18/2023]
Abstract
Activation of microglia results in the increased production and release of a series of inflammatory and neurotoxic mediators, which play essential roles in structural and functional neuronal damage and in the development and progression of a number of neurodegenerative diseases. The microalga Euglena gracilis (Euglena), rich in vitamins, minerals, and other nutrients, has gained increasing attention due to its antimicrobial, anti-viral, antitumor, and anti-inflammatory activities. In particular, anti-inflammatory properties of Euglena could exert neuroprotective functions in different neurodegenerative diseases related to inflammation. However, the mechanisms underlying the anti-inflammatory effect of Euglena are not fully understood. In this study, we investigated whether Euglena could attenuate microglia activation and we also studied the mechanism of its anti-inflammatory activity. Our results showed that non-cytotoxic concentrations of a Euglena acetone extract (EAE) downregulated the mRNA expression levels and release of pro-inflammatory mediators, including NO, IL-1β, and TNF-α in LPS-stimulated microglia. EAE also significantly blocked the LPS-induced nuclear translocation of NF-κB p65 subunit and increased the mRNA expression of nuclear factor erythroid 2–related factor (Nrf2) and heme oxygenase-1 (HO-1). Furthermore, the release of pro-inflammatory mediators and NF-κB activation were also blocked by EAE in the presence of ML385, a specific Nrf2 inhibitor. Together, these results show that EAE overcomes LPS-induced microglia pro-inflammatory responses through downregulation of NF-κB and activation of Nrf2 signaling pathways, although the two pathways seem to get involved in an independent manner.
Collapse
|
75
|
Mapping of Microglial Brain Region, Sex and Age Heterogeneity in Obesity. Int J Mol Sci 2021; 22:ijms22063141. [PMID: 33808700 PMCID: PMC8003547 DOI: 10.3390/ijms22063141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/27/2022] Open
Abstract
The prevalence of obesity has increased rapidly in recent years and has put a huge burden on healthcare worldwide. Obesity is associated with an increased risk for many comorbidities, such as cardiovascular diseases, type 2 diabetes and hypertension. The hypothalamus is a key brain region involved in the regulation of food intake and energy expenditure. Research on experimental animals has shown neuronal loss, as well as microglial activation in the hypothalamus, due to dietary-induced obesity. Microglia, the resident immune cells in the brain, are responsible for maintaining the brain homeostasis and, thus, providing an optimal environment for neuronal function. Interestingly, in obesity, microglial cells not only get activated in the hypothalamus but in other brain regions as well. Obesity is also highly associated with changes in hippocampal function, which could ultimately result in cognitive decline and dementia. Moreover, changes have also been reported in the striatum and cortex. Microglial heterogeneity is still poorly understood, not only in the context of brain region but, also, age and sex. This review will provide an overview of the currently available data on the phenotypic differences of microglial innate immunity in obesity, dependent on brain region, sex and age.
Collapse
|
76
|
Butler CA, Popescu AS, Kitchener EJA, Allendorf DH, Puigdellívol M, Brown GC. Microglial phagocytosis of neurons in neurodegeneration, and its regulation. J Neurochem 2021; 158:621-639. [PMID: 33608912 DOI: 10.1111/jnc.15327] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/13/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
There is growing evidence that excessive microglial phagocytosis of neurons and synapses contributes to multiple brain pathologies. RNA-seq and genome-wide association (GWAS) studies have linked multiple phagocytic genes to neurodegenerative diseases, and knock-out of phagocytic genes has been found to protect against neurodegeneration in animal models, suggesting that excessive microglial phagocytosis contributes to neurodegeneration. Here, we review recent evidence that microglial phagocytosis of live neurons and synapses causes neurodegeneration in animal models of Alzheimer's disease and other tauopathies, Parkinson's disease, frontotemporal dementias, multiple sclerosis, retinal degeneration and neurodegeneration induced by ischaemia, infection or ageing. We also review factors regulating microglial phagocytosis of neurons, including: nucleotides, frackalkine, phosphatidylserine, calreticulin, UDP, CD47, sialylation, complement, galectin-3, Apolipoprotein E, phagocytic receptors, Siglec receptors, cytokines, microglial epigenetics and expression profile. Some of these factors may be potential treatment targets to prevent neurodegeneration mediated by excessive microglial phagocytosis of live neurons and synapses.
Collapse
Affiliation(s)
- Claire A Butler
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Alma S Popescu
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - Mar Puigdellívol
- Department of Biochemistry, University of Cambridge, Cambridge, UK.,Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
77
|
Campagno KE, Mitchell CH. The P2X 7 Receptor in Microglial Cells Modulates the Endolysosomal Axis, Autophagy, and Phagocytosis. Front Cell Neurosci 2021; 15:645244. [PMID: 33790743 PMCID: PMC8005553 DOI: 10.3389/fncel.2021.645244] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/22/2021] [Indexed: 01/18/2023] Open
Abstract
Microglial cells regulate neural homeostasis by coordinating both immune responses and clearance of debris, and the P2X7 receptor for extracellular ATP plays a central role in both functions. The P2X7 receptor is primarily known in microglial cells for its immune signaling and NLRP3 inflammasome activation. However, the receptor also affects the clearance of extracellular and intracellular debris through modifications of lysosomal function, phagocytosis, and autophagy. In the absence of an agonist, the P2X7 receptor acts as a scavenger receptor to phagocytose material. Transient receptor stimulation induces autophagy and increases LC3-II levels, likely through calcium-dependent phosphorylation of AMPK, and activates microglia to an M1 or mixed M1/M2 state. We show an increased expression of Nos2 and Tnfa and a decreased expression of Chil3 (YM1) from primary cultures of brain microglia exposed to high levels of ATP. Sustained stimulation can reduce lysosomal function in microglia by increasing lysosomal pH and slowing autophagosome-lysosome fusion. P2X7 receptor stimulation can also cause lysosomal leakage, and the subsequent rise in cytoplasmic cathepsin B activates the NLRP3 inflammasome leading to caspase-1 cleavage and IL-1β maturation and release. Support for P2X7 receptor activation of the inflammasome following lysosomal leakage comes from data on primary microglia showing IL-1β release following receptor stimulation is inhibited by cathepsin B blocker CA-074. This pathway bridges endolysosomal and inflammatory roles and may provide a key mechanism for the increased inflammation found in age-dependent neurodegenerations characterized by excessive lysosomal accumulations. Regardless of whether the inflammasome is activated via this lysosomal leakage or the better-known K+-efflux pathway, the inflammatory impact of P2X7 receptor stimulation is balanced between the autophagic reduction of inflammasome components and their increase following P2X7-mediated priming. In summary, the P2X7 receptor modulates clearance of extracellular debris by microglial cells and mediates lysosomal damage that can activate the NLRP3 inflammasome. A better understanding of how the P2X7 receptor alters phagocytosis, lysosomal health, inflammation, and autophagy can lead to therapies that balance the inflammatory and clearance roles of microglial cells.
Collapse
Affiliation(s)
- Keith E Campagno
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Claire H Mitchell
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, United States.,Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, United States.,Department of Physiology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
78
|
Wang K, Li J, Zhang Y, Huang Y, Chen D, Shi Z, Smith AD, Li W, Gao Y. Central nervous system diseases related to pathological microglial phagocytosis. CNS Neurosci Ther 2021; 27:528-539. [PMID: 33650762 PMCID: PMC8025646 DOI: 10.1111/cns.13619] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 12/18/2022] Open
Abstract
Microglia are important phagocytes of the central nervous system (CNS). They play an important role in protecting the CNS by clearing necrotic tissue and apoptotic cells in many CNS diseases. However, recent studies have found that microglia can phagocytose parts of neurons excessively, such as the neuronal cell body, synapse, or myelin sheaths, before or after the onset of CNS diseases, leading to aggravated injury and impaired tissue repair. Meanwhile, reduced phagocytosis of synapses and myelin results in abnormal circuit connections and inhibition of remyelination, respectively. Previous studies focused primarily on the positive effects of microglia phagocytosis, whereas only a few studies have focused on the negative effects. In this review, we use the term "pathological microglial phagocytosis" to refer to excessive or reduced phagocytosis by microglia that leads to structural or functional abnormalities in target cells and brain tissue. The classification of pathological microglial phagocytosis, the composition, and activation of related signaling pathways, as well as the process of pathological phagocytosis in various kinds of CNS diseases, are described in this review. We hypothesize that pathological microglial phagocytosis leads to aggravation of tissue damage and negative functional outcome. For example, excessive microglial phagocytosis of synapses can be observed in Alzheimer's disease and schizophrenia, leading to significant synapse loss and memory impairment. In Parkinson's disease, ischemic stroke, and traumatic brain injury, excessive microglial phagocytosis of neuronal cell bodies causes impaired gray matter recovery and sensory dysfunction. We therefore believe that more studies should focus on the mechanism of pathological microglial phagocytosis and activation to uncover potential targets of therapeutic intervention.
Collapse
Affiliation(s)
- Ke Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yue Zhang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Di Chen
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ziyu Shi
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Amanda D Smith
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Wei Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
79
|
Morini R, Bizzotto M, Perrucci F, Filipello F, Matteoli M. Strategies and Tools for Studying Microglial-Mediated Synapse Elimination and Refinement. Front Immunol 2021; 12:640937. [PMID: 33708226 PMCID: PMC7940197 DOI: 10.3389/fimmu.2021.640937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/01/2021] [Indexed: 01/14/2023] Open
Abstract
The role of microglia in controlling synapse homeostasis is becoming increasingly recognized by the scientific community. In particular, the microglia-mediated elimination of supernumerary synapses during development lays the basis for the correct formation of neuronal circuits in adulthood, while the possible reactivation of this process in pathological conditions, such as schizophrenia or Alzheimer's Disease, provides a promising target for future therapeutic strategies. The methodological approaches to investigate microglial synaptic engulfment include different in vitro and in vivo settings. Basic in vitro assays, employing isolated microglia and microbeads, apoptotic membranes, liposomes or synaptosomes allow the quantification of the microglia phagocytic abilities, while co-cultures of microglia and neurons, deriving from either WT or genetically modified mice models, provide a relatively manageable setting to investigate the involvement of specific molecular pathways. Further detailed analysis in mice brain is then mandatory to validate the in vitro assays as representative for the in vivo situation. The present review aims to dissect the main technical approaches to investigate microglia-mediated phagocytosis of neuronal and synaptic substrates in critical developmental time windows.
Collapse
Affiliation(s)
- Raffaella Morini
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Matteo Bizzotto
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Fabio Perrucci
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Fabia Filipello
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Consiglio Nazionale Delle Ricerche (CNR), Institute of Neuroscience - URT Humanitas, Rozzano, Italy
| |
Collapse
|
80
|
Bogale TA, Faustini G, Longhena F, Mitola S, Pizzi M, Bellucci A. Alpha-Synuclein in the Regulation of Brain Endothelial and Perivascular Cells: Gaps and Future Perspectives. Front Immunol 2021; 12:611761. [PMID: 33679750 PMCID: PMC7933041 DOI: 10.3389/fimmu.2021.611761] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/27/2021] [Indexed: 12/27/2022] Open
Abstract
Misfolded proteins, inflammation, and vascular alterations are common pathological hallmarks of neurodegenerative diseases. Alpha-synuclein is a small synaptic protein that was identified as a major component of Lewy bodies and Lewy neurites in the brain of patients affected by Parkinson's disease (PD), Lewy body dementia (LBD), and other synucleinopathies. It is mainly involved in the regulation of synaptic vesicle trafficking but can also control mitochondrial/endoplasmic reticulum (ER) homeostasis, lysosome/phagosome function, and cytoskeleton organization. Recent evidence supports that the pathological forms of α-synuclein can also reduce the release of vasoactive and inflammatory mediators from endothelial cells (ECs) and modulates the expression of tight junction (TJ) proteins important for maintaining the blood-brain barrier (BBB). This hints that α-synuclein deposition can affect BBB integrity. Border associated macrophages (BAMs) are brain resident macrophages found in association with the vasculature (PVMs), meninges (MAMs), and choroid plexus (CPMs). Recent findings indicate that these cells play distinct roles in stroke and neurodegenerative disorders. Although many studies have addressed how α-synuclein may modulate microglia, its effect on BAMs has been scarcely investigated. This review aims at summarizing the main findings supporting how α-synuclein can affect ECs and/or BAMs function as well as their interplay and effect on other cells in the brain perivascular environment in physiological and pathological conditions. Gaps of knowledge and new perspectives on how this protein can contribute to neurodegeneration by inducing BBB homeostatic changes in different neurological conditions are highlighted.
Collapse
Affiliation(s)
- Tizibt Ashine Bogale
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Longhena
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Stefania Mitola
- Biotechnology Division, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Laboratory for Preventive and Personalized Medicine, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Laboratory for Preventive and Personalized Medicine, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
81
|
Desale SE, Chinnathambi S. α-Linolenic acid induces clearance of Tau seeds via Actin-remodeling in Microglia. MOLECULAR BIOMEDICINE 2021; 2:4. [PMID: 35006402 PMCID: PMC8607384 DOI: 10.1186/s43556-021-00028-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 01/14/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is known by characteristic features, extracellular burden of amyloid-β and intracellular neuronal Tau. Microglia, the innate immune cell of the brain has the ability to clear the burden of accumulated proteins via phagocytosis. But the excessive proinflammatory cytokine production, altered cellular signaling and actin remodeling hampers the process of migration and phagocytosis by microglia. Actin remodeling is necessary to initiate the chemotactic migration of microglia towards the target and engulf it. The formation of lamellipodia, filopodia, membrane ruffling and rapid turnover of F-actin is necessary to sense the extracellular target by the cells. Omega-3 fatty acids, are known to impose anti-inflammatory phenotype of microglia by enhancing its ability for migration and phagocytosis. But the role of omega-3 fatty acids in cellular actin remodeling, which is the basis of cellular functions such as migration and phagocytosis, is not well understood. Here, we have focused on the effect of dietary supplement of α-linolenic acid (ALA) on extracellular Tau internalization and assisted actin polymerization for the process. ALA is found to induce membrane ruffling and phagocytic cup formation along with cytoskeletal rearrangement. ALA also enhances the localization of Arp2/3 complex at the leading edge and its colocalization with F-actin to induce the actin polymerization. The excessive actin polymerization might help the cell to protrude forward and perform its migration. The results suggest that dietary supplement of ALA could play a neuroprotective role and slow down the AD pathology.
Collapse
Affiliation(s)
- Smita Eknath Desale
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
82
|
Trapecar M, Wogram E, Svoboda D, Communal C, Omer A, Lungjangwa T, Sphabmixay P, Velazquez J, Schneider K, Wright CW, Mildrum S, Hendricks A, Levine S, Muffat J, Lee MJ, Lauffenburger DA, Trumper D, Jaenisch R, Griffith LG. Human physiomimetic model integrating microphysiological systems of the gut, liver, and brain for studies of neurodegenerative diseases. SCIENCE ADVANCES 2021; 7:7/5/eabd1707. [PMID: 33514545 PMCID: PMC7846169 DOI: 10.1126/sciadv.abd1707] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/11/2020] [Indexed: 05/11/2023]
Abstract
Slow progress in the fight against neurodegenerative diseases (NDs) motivates an urgent need for highly controlled in vitro systems to investigate organ-organ- and organ-immune-specific interactions relevant for disease pathophysiology. Of particular interest is the gut/microbiome-liver-brain axis for parsing out how genetic and environmental factors contribute to NDs. We have developed a mesofluidic platform technology to study gut-liver-cerebral interactions in the context of Parkinson's disease (PD). It connects microphysiological systems (MPSs) of the primary human gut and liver with a human induced pluripotent stem cell-derived cerebral MPS in a systemically circulated common culture medium containing CD4+ regulatory T and T helper 17 cells. We demonstrate this approach using a patient-derived cerebral MPS carrying the PD-causing A53T mutation, gaining two important findings: (i) that systemic interaction enhances features of in vivo-like behavior of cerebral MPSs, and (ii) that microbiome-associated short-chain fatty acids increase expression of pathology-associated pathways in PD.
Collapse
Affiliation(s)
- Martin Trapecar
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emile Wogram
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Devon Svoboda
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Catherine Communal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Attya Omer
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | | | - Pierre Sphabmixay
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jason Velazquez
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kirsten Schneider
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charles W Wright
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samuel Mildrum
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- MIT BioMicro Center, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Austin Hendricks
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- MIT BioMicro Center, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stuart Levine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- MIT BioMicro Center, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Julien Muffat
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Meelim Jasmine Lee
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David Trumper
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
83
|
Cserép C, Pósfai B, Dénes Á. Shaping Neuronal Fate: Functional Heterogeneity of Direct Microglia-Neuron Interactions. Neuron 2020; 109:222-240. [PMID: 33271068 DOI: 10.1016/j.neuron.2020.11.007] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/13/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
The functional contribution of microglia to normal brain development, healthy brain function, and neurological disorders is increasingly recognized. However, until recently, the nature of intercellular interactions mediating these effects remained largely unclear. Recent findings show microglia establishing direct contact with different compartments of neurons. Although communication between microglia and neurons involves intermediate cells and soluble factors, direct membrane contacts enable a more precisely regulated, dynamic, and highly effective form of interaction for fine-tuning neuronal responses and fate. Here, we summarize the known ultrastructural, molecular, and functional features of direct microglia-neuron interactions and their roles in brain disease.
Collapse
Affiliation(s)
- Csaba Cserép
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary
| | - Balázs Pósfai
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary; Szentágothai János Doctoral School of Neurosciences, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary.
| |
Collapse
|
84
|
Kubick N, Henckell Flournoy PC, Klimovich P, Manda G, Mickael M. What has single-cell RNA sequencing revealed about microglial neuroimmunology? Immun Inflamm Dis 2020; 8:825-839. [PMID: 33085226 PMCID: PMC7654416 DOI: 10.1002/iid3.362] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 12/22/2022] Open
Abstract
The use of single-cell RNA sequencing (scRNA-seq) in microglial research is increasing rapidly. The basic workflow of this approach consists of isolating single cells, followed by sequencing. scRNA-seq is capable of examining microglial heterogeneity on a cellular level. However, the results gained from applying this technique suffer from discrepancies due to differences between applied methods characteristics such as the number of cells sequenced and the depth of sequencing. This review aims to shed more light on the recent developments that happened in this field and how they are related to the methods used. To do that, we track the progress and limitations of various scRNA-seq methods currently available. The review then summarizes the current knowledge gained using scRNA-seq in the field of microglia, including novel subpopulations associated with function and development under homeostasis as well during several pathological conditions such as Alzheimer, lipopolysaccharide response, and HIV in relation to the methods employed. Our review points out that despite major developments found using this technique, current scRNA-seq methods suffer from high cost, low yields, and nonstandardization of generated data. Additional development of scRNA-seq methods will raise our awareness of microglia's heterogeneity and plasticity under healthy and pathological conditions.
Collapse
Affiliation(s)
- Norwin Kubick
- Department of Biochemistry and Molecular Cell Biology (IBMZ)University Medical Center Hamburg‐EppendorfHamburgGermany
| | | | - Pavel Klimovich
- Department of ImmunologyPM Research CenterStockholmEkeröSweden
| | - Gina Manda
- Department of RadiologyVictor Babes National Institute of PathologyBucharestRomania
| | - Michel‐Edwar Mickael
- Department of ImmunologyPM Research CenterStockholmEkeröSweden
- Department of PathologyBevill Biomedical SciencesBirminghamAlabamaUSA
- Department of Experimental Genomics, Neuroimmunology Group, Institute of Genetics and Animal BiotechnologyPolish Academy of ScienceMagdalenkaPoland
| |
Collapse
|
85
|
Boi L, Pisanu A, Palmas MF, Fusco G, Carboni E, Casu MA, Satta V, Scherma M, Janda E, Mocci I, Mulas G, Ena A, Spiga S, Fadda P, De Simone A, Carta AR. Modeling Parkinson's Disease Neuropathology and Symptoms by Intranigral Inoculation of Preformed Human α-Synuclein Oligomers. Int J Mol Sci 2020; 21:E8535. [PMID: 33198335 PMCID: PMC7696693 DOI: 10.3390/ijms21228535] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022] Open
Abstract
The accumulation of aggregated α-synuclein (αSyn) is a hallmark of Parkinson's disease (PD). Current evidence indicates that small soluble αSyn oligomers (αSynOs) are the most toxic species among the forms of αSyn aggregates, and that size and topological structural properties are crucial factors for αSynOs-mediated toxicity, involving the interaction with either neurons or glial cells. We previously characterized a human αSynO (H-αSynO) with specific structural properties promoting toxicity against neuronal membranes. Here, we tested the neurotoxic potential of these H-αSynOs in vivo, in relation to the neuropathological and symptomatic features of PD. The H-αSynOs were unilaterally infused into the rat substantia nigra pars compacta (SNpc). Phosphorylated αSyn (p129-αSyn), reactive microglia, and cytokine levels were measured at progressive time points. Additionally, a phagocytosis assay in vitro was performed after microglia pre-exposure to αsynOs. Dopaminergic loss, motor, and cognitive performances were assessed. H-αSynOs triggered p129-αSyn deposition in SNpc neurons and microglia and spread to the striatum. Early and persistent neuroinflammatory responses were induced in the SNpc. In vitro, H-αSynOs inhibited the phagocytic function of microglia. H-αsynOs-infused rats displayed early mitochondrial loss and abnormalities in SNpc neurons, followed by a gradual nigrostriatal dopaminergic loss, associated with motor and cognitive impairment. The intracerebral inoculation of structurally characterized H-αSynOs provides a model of progressive PD neuropathology in rats, which will be helpful for testing neuroprotective therapies.
Collapse
Affiliation(s)
- Laura Boi
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | | | - Maria Francesca Palmas
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Giuliana Fusco
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, UK;
| | - Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Maria Antonietta Casu
- CNR Institute of Translational Pharmacology, 09010 Cagliari, Italy; (M.A.C.); (I.M.)
| | - Valentina Satta
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Maria Scherma
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Elzbieta Janda
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Ignazia Mocci
- CNR Institute of Translational Pharmacology, 09010 Cagliari, Italy; (M.A.C.); (I.M.)
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, 09126 Cagliari, Italy; (G.M.); (S.S.)
| | - Anna Ena
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, 09126 Cagliari, Italy; (G.M.); (S.S.)
| | - Paola Fadda
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
- CNR Institute of Neuroscience, 09042 Cagliari, Italy;
- Italian Neuroscience Institute (INN), 10126 Torino, Italy
| | - Alfonso De Simone
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Anna R. Carta
- Department of Biomedical Sciences, University of Cagliari, 09042 Cagliari, Italy; (L.B.); (M.F.P.); (E.C.); (V.S.); (M.S.); (A.E.); (P.F.)
- Italian Neuroscience Institute (INN), 10126 Torino, Italy
| |
Collapse
|
86
|
Iannucci J, Rao HV, Grammas P. High Glucose and Hypoxia-Mediated Damage to Human Brain Microvessel Endothelial Cells Induces an Altered, Pro-Inflammatory Phenotype in BV-2 Microglia In Vitro. Cell Mol Neurobiol 2020; 42:985-996. [PMID: 33136275 PMCID: PMC8942976 DOI: 10.1007/s10571-020-00987-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/22/2020] [Indexed: 01/13/2023]
Abstract
Diabetes is strongly linked to the development of Alzheimer’s disease (AD), though the mechanisms for this enhanced risk are unclear. Because vascular inflammation is a consistent feature of both diabetes and AD, the cerebral microcirculation could be a key target for the effects of diabetes in the brain. The goal of this study is to explore whether brain endothelial cells, injured by diabetes-related insults, glucose and hypoxia, can affect inflammatory and activation processes in microglia in vitro. Human brain microvascular endothelial cells (HBMVECs) were either treated with 5 mM glucose (control), 30 mM glucose (high glucose), exposed to hypoxia, or exposed to hypoxia plus high glucose. HBMVEC-conditioned medium was then used to treat BV-2 microglia. Alterations in microglia phenotype were assessed through measurement of nitric oxide (NO), cytokine production, microglial activation state markers, and microglial phagocytosis. HBMVECs were injured by exposure to glucose and/or hypoxia, as assessed by release of LDH, interleukin (IL)-1β, and reactive oxygen species (ROS). HBMVECs injured by glucose and hypoxia induced increases in microglial production of NO, tumor necrosis factor-α (TNFα) and matrix metalloproteinase (MMP)-9. Injured HBMVECs significantly increased microglial expression of CD11c and CLEC7A, and decreased expression of the homeostatic marker P2RY12. Finally, bead uptake by BV-2 cells, an index of phagocytic ability, was elevated by conditioned media from injured HBMVECs. The demonstration that injury to brain endothelial cells by diabetic-associated insults, glucose and hypoxia, promotes microglial inflammation supports the idea that the cerebral microcirculation is a critical locus for the deleterious effects of diabetes in the AD brain.
Collapse
Affiliation(s)
- Jaclyn Iannucci
- The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, United States. .,Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA.
| | - Haripriya Vittal Rao
- The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, United States.,Wake Forest Baptist Medical Center, Winston-Salem, Wake Forest, NC, 27101, USA
| | - Paula Grammas
- The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, 130 Flagg Road, Kingston, RI, 02881, United States.,Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| |
Collapse
|
87
|
Lyu J, Jiang X, Leak RK, Shi Y, Hu X, Chen J. Microglial Responses to Brain Injury and Disease: Functional Diversity and New Opportunities. Transl Stroke Res 2020; 12:474-495. [PMID: 33128703 DOI: 10.1007/s12975-020-00857-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/27/2022]
Abstract
As an integral part of the innate immune system of the brain, resident microglia must react rapidly to the onset of brain injury and neurological disease. These dynamic cells then continue to shift their phenotype along a multidimensional continuum with overlapping pro- and anti-inflammatory states, allowing them to adapt to microenvironmental changes during the progression of brain disorders. However, the ability of microglia to shift phenotype through nimble molecular, structural, and functional changes comes at a cost, as the extreme pro-inflammatory states may prevent these professional phagocytes from clearing toxic debris and secreting tissue-repairing neurotrophic factors. Evolution has strongly favored heterogeneity in microglia in both the spatial and temporal dimensions-they can assume diverse roles in different brain regions, throughout the course of brain development and aging, and during the spatiotemporal progression of brain injuries and neurological diseases. Age and sex differences add further diversity to microglia functional status under physiological and pathological conditions. This article reviews recent advances in our knowledge of microglia with emphases on molecular mediators of phenotype shifts and functional diversity. We describe microglia-targeted therapeutic opportunities, including pharmacologic modulation of phenotype and repopulation of the brain with fresh microglia. With the advent of powerful new tools, research on microglia has recently accelerated in pace and may translate into potential therapeutics against brain injury and neurological disease.
Collapse
Affiliation(s)
- Junxuan Lyu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xiaoyan Jiang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, 15282, USA
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Xiaoming Hu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
88
|
Morales-Ropero JM, Arroyo-Urea S, Neubrand VE, Martín-Oliva D, Marín-Teva JL, Cuadros MA, Vangheluwe P, Navascués J, Mata AM, Sepúlveda MR. The endoplasmic reticulum Ca 2+ -ATPase SERCA2b is upregulated in activated microglia and its inhibition causes opposite effects on migration and phagocytosis. Glia 2020; 69:842-857. [PMID: 33105046 DOI: 10.1002/glia.23931] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022]
Abstract
Activation of microglia is an early immune response to damage in the brain. Although a key role for Ca2+ as trigger of microglial activation has been considered, little is known about the molecular scenario for regulating Ca2+ homeostasis in these cells. Taking into account the importance of the endoplasmic reticulum as a cellular Ca2+ store, the sarco(endo)plasmic reticulum Ca2+ -ATPase (SERCA2b) is an interesting target to modulate intracellular Ca2+ dynamics. We found upregulation of SERCA2b in activated microglia of human brain with Alzheimer's disease and we further studied the participation of SERCA2b in microglial functions by using the BV2 murine microglial cell line and primary microglia isolated from mouse brain. To trigger microglia activation, we used the bacterial lipopolysaccharide (LPS), which is known to induce an increase of cytosolic Ca2+ . Our results showed an upregulated expression of SERCA2b in LPS-induced activated microglia likely associated to an attempt to restore the increased cytosolic Ca2+ concentration. We analyzed SERCA2b contribution in microglial migration by using the specific SERCA inhibitor thapsigargin in scratch assays. Microglial migration was strongly stimulated with thapsigargin, even more than with LPS-induction, but delayed in time. However, phagocytic capacity of microglia was blocked in the presence of the SERCA inhibitor, indicating the importance of a tight control of cytosolic Ca2+ in these processes. All together, these results provide for the first time compelling evidence for SERCA2b as a major player regulating microglial functions, affecting migration and phagocytosis in an opposite manner.
Collapse
Affiliation(s)
- Juan M Morales-Ropero
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Sandra Arroyo-Urea
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Veronika E Neubrand
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - David Martín-Oliva
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - José L Marín-Teva
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Miguel A Cuadros
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Julio Navascués
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Ana M Mata
- Department of Biochemistry and Molecular Biology and Genetics, Faculty of Sciences, University of Extremadura, Badajoz, Spain
| | - M Rosario Sepúlveda
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| |
Collapse
|
89
|
Berglund R, Guerreiro-Cacais AO, Adzemovic MZ, Zeitelhofer M, Lund H, Ewing E, Ruhrmann S, Nutma E, Parsa R, Thessen-Hedreul M, Amor S, Harris RA, Olsson T, Jagodic M. Microglial autophagy-associated phagocytosis is essential for recovery from neuroinflammation. Sci Immunol 2020; 5:5/52/eabb5077. [PMID: 33067381 DOI: 10.1126/sciimmunol.abb5077] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 09/24/2020] [Indexed: 01/07/2023]
Abstract
Multiple sclerosis (MS) is a leading cause of incurable progressive disability in young adults caused by inflammation and neurodegeneration in the central nervous system (CNS). The capacity of microglia to clear tissue debris is essential for maintaining and restoring CNS homeostasis. This capacity diminishes with age, and age strongly associates with MS disease progression, although the underlying mechanisms are still largely elusive. Here, we demonstrate that the recovery from CNS inflammation in a murine model of MS is dependent on the ability of microglia to clear tissue debris. Microglia-specific deletion of the autophagy regulator Atg7, but not the canonical macroautophagy protein Ulk1, led to increased intracellular accumulation of phagocytosed myelin and progressive MS-like disease. This impairment correlated with a microglial phenotype previously associated with neurodegenerative pathologies. Moreover, Atg7-deficient microglia showed notable transcriptional and functional similarities to microglia from aged wild-type mice that were also unable to clear myelin and recover from disease. In contrast, induction of autophagy in aged mice using the disaccharide trehalose found in plants and fungi led to functional myelin clearance and disease remission. Our results demonstrate that a noncanonical form of autophagy in microglia is responsible for myelin degradation and clearance leading to recovery from MS-like disease and that boosting this process has a therapeutic potential for age-related neuroinflammatory conditions.
Collapse
Affiliation(s)
- Rasmus Berglund
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Andre Ortlieb Guerreiro-Cacais
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Milena Z Adzemovic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Manuel Zeitelhofer
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Solna, Sweden
| | - Harald Lund
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Ewoud Ewing
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Sabrina Ruhrmann
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Erik Nutma
- Department of Pathology, Amsterdam UMC, Location VUmc, De Boelelaan 1117, 1081 HV Amsterdam, Netherlands
| | - Roham Parsa
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Melanie Thessen-Hedreul
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, Location VUmc, De Boelelaan 1117, 1081 HV Amsterdam, Netherlands.,Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden.
| |
Collapse
|
90
|
Feng Z, Zhu Z, Chen W, Bai Y, Hu D, Cheng J. Chloride intracellular channel 4 participate in the protective effect of Ginkgolide B in MPP+ injured MN9D cells: insight from proteomic analysis. Clin Proteomics 2020; 17:32. [PMID: 32944011 PMCID: PMC7487930 DOI: 10.1186/s12014-020-09295-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/30/2020] [Indexed: 11/18/2022] Open
Abstract
Background Ginkgolide B (GB), the extract of G. biloba leaves, has been shown to be protective against many neurological disorders, including Parkinson’s disease (PD). Efforts have been made to synthesized ginkgolides analogs and derivatives with more targeted and smaller molecular weight. In the present study, four GB derivatives (GBHC-1-GBHC-4) were synthesized, and their protective roles in N-methyl-4-phenylpyridinium (MPP +) injured MN9D dopaminergic neuronal cell line were evaluated. Also, cell response mechanisms upon these GB derivatives treatment were analyzed by iTRAQ proteomics. Methods MN9D cells were treated with MPP + to induce in vitro cell models of PD. Four GB derivatives (GBHC-1-GBHC-4) were synthesized, and their protective roles on cell viability and apoptosis in in vitro PD model cells were evaluated by CCK8 assay, fluorescence-activated cell sorting and DAPI staining, respectively. The proteomic profiles of MPP+ injured MN9D cells pretreated with or without GB and GB derivatives were detected using the isobaric tags for relative and absolute quantification (iTRAQ) labeling technique. Results Pretreatment with GBHC-1-GBHC-4 noticeably increased cell viability and attenuated cell apoptosis in MPP+ -injured MN9D cells. Using proteomic analysis, we identified differentially expressed proteins upon GB and GB derivatives treatment. Chloride intracellular channel 4 (CLIC4) and “protein processing in endoplasmic reticulum” pathways participated in the protective roles of GB and GBHC-4. GB and GBHC-4 pretreatment could significantly reverse MPP+ -induced CLIC4 expression and translocation from cytoplasm to nucleus of MN9D cells. Conclusions Quantitative comparative proteomic analysis identified differentially expressed proteins associated with GB and GB derivatives. We further verified the expression of CLIC4 by western blotting and immunocytochemistry assay. This bio-information on the identified pathways and differentially expressed proteins such as CLIC4 provide more targeted directions for the synthesis of more effective and targeted GB derivatives for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Zili Feng
- School of Bioscience and Engeering, Shaanxi University of Technology, No. 1 Donghuan 1st Road, Hanzhong, 732001 Shaanxi People's Republic of China
| | - Zhibin Zhu
- School of Bioscience and Engeering, Shaanxi University of Technology, No. 1 Donghuan 1st Road, Hanzhong, 732001 Shaanxi People's Republic of China
| | - Wang Chen
- School of Bioscience and Engeering, Shaanxi University of Technology, No. 1 Donghuan 1st Road, Hanzhong, 732001 Shaanxi People's Republic of China
| | - Yu Bai
- School of Bioscience and Engeering, Shaanxi University of Technology, No. 1 Donghuan 1st Road, Hanzhong, 732001 Shaanxi People's Republic of China
| | - Daihua Hu
- School of Bioscience and Engeering, Shaanxi University of Technology, No. 1 Donghuan 1st Road, Hanzhong, 732001 Shaanxi People's Republic of China
| | - Jia Cheng
- School of Bioscience and Engeering, Shaanxi University of Technology, No. 1 Donghuan 1st Road, Hanzhong, 732001 Shaanxi People's Republic of China
| |
Collapse
|
91
|
Zhao N, Francis NL, Calvelli HR, Moghe PV. Microglia-targeting nanotherapeutics for neurodegenerative diseases. APL Bioeng 2020; 4:030902. [PMID: 32923843 PMCID: PMC7481010 DOI: 10.1063/5.0013178] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
Advances in nanotechnology have enabled the design of nanotherapeutic platforms that could address the challenges of targeted delivery of active therapeutic agents to the central nervous system (CNS). While the majority of previous research studies on CNS nanotherapeutics have focused on neurons and endothelial cells, the predominant resident immune cells of the CNS, microglia, are also emerging as a promising cellular target for neurodegeneration considering their prominent role in neuroinflammation. Under normal physiological conditions, microglia protect neurons by removing pathological agents. However, long-term exposure of microglia to stimulants will cause sustained activation and lead to neuronal damage due to the release of pro-inflammatory agents, resulting in neuroinflammation and neurodegeneration. This Perspective highlights criteria to be considered when designing microglia-targeting nanotherapeutics for the treatment of neurodegenerative disorders. These criteria include conjugating specific microglial receptor-targeting ligands or peptides to the nanoparticle surface to achieve targeted delivery, leveraging microglial phagocytic properties, and utilizing biocompatible and biodegradable nanomaterials with low immune reactivity and neurotoxicity. In addition, certain therapeutic agents for the controlled inhibition of toxic protein aggregation and for modulation of microglial activation pathways can also be incorporated within the nanoparticle structure without compromising stability. Overall, considering the multifaceted disease mechanisms of neurodegeneration, microglia-targeted nanodrugs and nanotherapeutic particles may have the potential to resolve multiple pathological determinants of the disease and to guide a shift in the microglial phenotype spectrum toward a more neuroprotective state.
Collapse
Affiliation(s)
- Nanxia Zhao
- Department of Chemical and Biochemical Engineering, 98 Brett Rd., Rutgers University, Piscataway, New Jersey 08854, USA
| | - Nicola L. Francis
- Department of Biomedical Engineering, 599 Taylor Rd., Rutgers University, Piscataway, New Jersey 08854, USA
| | - Hannah R. Calvelli
- Department of Molecular Biology and Biochemistry, 604 Allison Rd., Rutgers University, Piscataway, New Jersey 08854, USA
| | | |
Collapse
|
92
|
Doxorubicin generates senescent microglia that exhibit altered proteomes, higher levels of cytokine secretion, and a decreased ability to internalize amyloid β. Exp Cell Res 2020; 395:112203. [PMID: 32738344 DOI: 10.1016/j.yexcr.2020.112203] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022]
Abstract
Cellular senescence is defined by irreversible cell-cycle arrest and is an evolutionarily conserved hallmark of aging. In this study, we generate senescent microglial cells via exposure to the chemotherapy drug doxorubicin. Compared to control cells, doxorubicin-treated microglia exhibited an altered morphology characterized by an enlarged cell size, a flattened appearance, and the development of prominent filaments. Senescent cells harbored elevated levels of senescence associated-β-galactosidase, p16Ink4a, and γ-H2AX. Senescent microglia were also less efficient at internalizing amyloid β and pHrodo bioparticles. A detailed proteomic analysis using SWATH-MS identified 201 proteins that were significantly downregulated and 127 that were significantly upregulated in doxorubicin-treated microglia. Proteins involved in processes such as protein synthesis, RNA damage and repair, and protein degradation were largely downregulated while those compromising the integrity of the cell were predominantly upregulated. Various proteins involved in proteasomal processing were among the most significantly downregulated in senescent cells. Relevant to the deleterious senescence-associated secretory phenotype, senescent cells secreted higher levels of the inflammatory cytokines IL-6, IL-8, TNF-α, and GRO-α. Our data suggest that symptoms of brain aging and age-related neurodegenerative disease may be partially caused by defective phagocytosis, impaired proteasomal processing, and elevated cytokine secretion of senescent microglia.
Collapse
|
93
|
Terzi A, Suter DM. The role of NADPH oxidases in neuronal development. Free Radic Biol Med 2020; 154:33-47. [PMID: 32370993 DOI: 10.1016/j.freeradbiomed.2020.04.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) are critical for maintaining cellular homeostasis and function when produced in physiological ranges. Important sources of cellular ROS include NADPH oxidases (Nox), which are evolutionary conserved multi-subunit transmembrane proteins. Nox-mediated ROS regulate variety of biological processes including hormone synthesis, calcium signaling, cell migration, and immunity. ROS participate in intracellular signaling by introducing post-translational modifications to proteins and thereby altering their functions. The central nervous system (CNS) expresses different Nox isoforms during both development and adulthood. Here, we review the role of Nox-mediated ROS during CNS development. Specifically, we focus on how individual Nox isoforms contribute to signaling in neural stem cell maintenance and neuronal differentiation, as well as neurite outgrowth and guidance. We also discuss how ROS regulates the organization and dynamics of the actin cytoskeleton in the neuronal growth cone. Finally, we review recent evidence that Nox-derived ROS modulate axonal regeneration upon nervous system injury.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA; Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
94
|
Burns JC, Cotleur B, Walther DM, Bajrami B, Rubino SJ, Wei R, Franchimont N, Cotman SL, Ransohoff RM, Mingueneau M. Differential accumulation of storage bodies with aging defines discrete subsets of microglia in the healthy brain. eLife 2020; 9:e57495. [PMID: 32579115 PMCID: PMC7367682 DOI: 10.7554/elife.57495] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/21/2020] [Indexed: 12/19/2022] Open
Abstract
To date, microglia subsets in the healthy CNS have not been identified. Utilizing autofluorescence (AF) as a discriminating parameter, we identified two novel microglia subsets in both mice and non-human primates, termed autofluorescence-positive (AF+) and negative (AF-). While their proportion remained constant throughout most adult life, the AF signal linearly and specifically increased in AF+ microglia with age and correlated with a commensurate increase in size and complexity of lysosomal storage bodies, as detected by transmission electron microscopy and LAMP1 levels. Post-depletion repopulation kinetics revealed AF- cells as likely precursors of AF+ microglia. At the molecular level, the proteome of AF+ microglia showed overrepresentation of endolysosomal, autophagic, catabolic, and mTOR-related proteins. Mimicking the effect of advanced aging, genetic disruption of lysosomal function accelerated the accumulation of storage bodies in AF+ cells and led to impaired microglia physiology and cell death, suggestive of a mechanistic convergence between aging and lysosomal storage disorders.
Collapse
Affiliation(s)
- Jeremy Carlos Burns
- Multiple Sclerosis & Neurorepair Research Unit, BiogenCambridgeUnited States
- Department of Pharmacology & Experimental Therapeutics, Boston University School of MedicineBostonUnited States
| | - Bunny Cotleur
- Emerging Neurosciences Research Unit, BiogenCambridgeUnited States
| | | | - Bekim Bajrami
- Chemical Biology and ProteomicsCambridgeUnited States
| | - Stephen J Rubino
- Multiple Sclerosis & Neurorepair Research Unit, BiogenCambridgeUnited States
| | - Ru Wei
- Chemical Biology and ProteomicsCambridgeUnited States
| | | | - Susan L Cotman
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical SchoolBostonUnited States
| | | | - Michael Mingueneau
- Multiple Sclerosis & Neurorepair Research Unit, BiogenCambridgeUnited States
| |
Collapse
|
95
|
Ramesha S, Rayaprolu S, Rangaraju S. Flow Cytometry Approach to Characterize Phagocytic Properties of Acutely-Isolated Adult Microglia and Brain Macrophages In Vitro. JOURNAL OF VISUALIZED EXPERIMENTS : JOVE 2020:10.3791/61467. [PMID: 32658196 PMCID: PMC9888024 DOI: 10.3791/61467] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Microglia and central nervous system (CNS)-infiltrating macrophages, collectively called CNS mononuclear phagocytes (CNS-MPs), play central roles in neurological diseases including neurodegeneration and stroke. CNS-MPs are involved in phagocytic clearance of pathological proteins, debris and neuronal synapses, each with distinct underlying molecular pathways. Characterizing these phagocytic properties can provide a functional readout that compliments molecular profiling of microglia using traditional flow cytometry, transcriptomics and proteomics approaches. Phagocytic profiling of microglia has relied on microscopic visualization and in vitro cultures of mouse neonatal microglia. The former approach suffers from limited sampling while the latter approach is inherently poorly reflective of the true in vivo state of adult CNS-MPs. This paper describes optimized protocols to phenotype phagocytic properties of acutely-isolated mouse CNS-MPs by flow cytometry. CNS-MPs are acutely isolated from adult mouse brain using mechanical dissociation followed by density gradient centrifugation, incubated with fluorescent microspheres or fluorescent Aβ fibrils, washed, and then labeled with panels of antibodies against surface markers (CD11b, CD45). Using this approach, it is possible to compare phagocytic properties of brain-resident microglia with CNS-infiltrating macrophages and then assess the effect of aging and disease pathology on these phagocytic phenotypes. This rapid method also holds potential to functionally phenotype acutely-isolated human CNS-MPs from post-mortem or surgical brain specimens. Additionally, specific mechanisms of phagocytosis by CNS-MP subsets can be investigated by inhibiting select phagocytic pathways.
Collapse
|
96
|
Belloli S, Morari M, Murtaj V, Valtorta S, Moresco RM, Gilardi MC. Translation Imaging in Parkinson's Disease: Focus on Neuroinflammation. Front Aging Neurosci 2020; 12:152. [PMID: 32581765 PMCID: PMC7289967 DOI: 10.3389/fnagi.2020.00152] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the appearance of α-synuclein insoluble aggregates known as Lewy bodies. Neurodegeneration is accompanied by neuroinflammation mediated by cytokines and chemokines produced by the activated microglia. Several studies demonstrated that such an inflammatory process is an early event, and contributes to oxidative stress and mitochondrial dysfunctions. α-synuclein fibrillization and aggregation activate microglia and contribute to disease onset and progression. Mutations in different genes exacerbate the inflammatory phenotype in the monogenic compared to sporadic forms of PD. Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) with selected radiopharmaceuticals allow in vivo imaging of molecular modifications in the brain of living subjects. Several publications showed a reduction of dopaminergic terminals and dopamine (DA) content in the basal ganglia, starting from the early stages of the disease. Moreover, non-dopaminergic neuronal pathways are also affected, as shown by in vivo studies with serotonergic and glutamatergic radiotracers. The role played by the immune system during illness progression could be investigated with PET ligands that target the microglia/macrophage Translocator protein (TSPO) receptor. These agents have been used in PD patients and rodent models, although often without attempting correlations with other molecular or functional parameters. For example, neurodegeneration and brain plasticity can be monitored using the metabolic marker 2-Deoxy-2-[18F]fluoroglucose ([18F]-FDG), while oxidative stress can be probed using the copper-labeled diacetyl-bis(N-methyl-thiosemicarbazone) ([Cu]-ATSM) radioligand, whose striatal-specific binding ratio in PD patients seems to correlate with a disease rating scale and motor scores. Also, structural and functional modifications during disease progression may be evaluated by Magnetic Resonance Imaging (MRI), using different parameters as iron content or cerebral volume. In this review article, we propose an overview of in vivo clinical and non-clinical imaging research on neuroinflammation as an emerging marker of early PD. We also discuss how multimodal-imaging approaches could provide more insights into the role of the inflammatory process and related events in PD development.
Collapse
Affiliation(s)
- Sara Belloli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Michele Morari
- Section of Pharmacology, Department of Medical Sciences, National Institute for Neuroscience, University of Ferrara, Ferrara, Italy
| | - Valentina Murtaj
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,PhD Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Silvia Valtorta
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Rosa Maria Moresco
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| | - Maria Carla Gilardi
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.,Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
97
|
Loving BA, Bruce KD. Lipid and Lipoprotein Metabolism in Microglia. Front Physiol 2020; 11:393. [PMID: 32411016 PMCID: PMC7198855 DOI: 10.3389/fphys.2020.00393] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/02/2020] [Indexed: 12/25/2022] Open
Abstract
Microglia, once viewed as static bystanders with limited homeostatic functions, are now considered key players in the development of neuroinflammatory and neurodegenerative diseases. Microglial activation is a salient feature of neuroinflammation involving a dynamic process that generates multitudinous microglial phenotypes that can respond to a variety of situational cues in the central nervous system. Recently, a flurry of single cell RNA-sequencing studies have defined microglial phenotypes in unprecedented detail, and have highlighted robust changes in the expression of genes involved in lipid and lipoprotein metabolism. Increased expression of genes such as Apolipoprotein E (ApoE), Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) and Lipoprotein Lipase (LPL) in microglia during development, damage, and disease, suggest that increased lipid metabolism is needed to fuel protective cellular functions such as phagocytosis. This review describes our current understanding of lipid and lipoprotein metabolism in microglia, and highlights microglial lipid metabolism as a modifiable target for the treatment of neurodegenerative diseases such as Alzheimer's disease and multiple sclerosis.
Collapse
Affiliation(s)
- Bailey A. Loving
- School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| | - Kimberley D. Bruce
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
98
|
Poewe W, Seppi K, Marini K, Mahlknecht P. New hopes for disease modification in Parkinson's Disease. Neuropharmacology 2020; 171:108085. [PMID: 32298705 DOI: 10.1016/j.neuropharm.2020.108085] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/31/2020] [Indexed: 12/11/2022]
Abstract
To date, despite numerous clinical trials, no intervention has been demonstrated to modify the progression of Parkinson's disease (PD). However, over the past decades encouraging progress has been made towards a better understanding of molecular pathways relevant for the neurodegenerative process in PD. This is also based on new insights into the genetic architecture of the disease, revealing multiple novel targets for potentially disease-modifying interventions. Important achievements have also been made in the field of risk markers and combinations thereof, in the form of risk algorithms, will hopefully soon provide the possibility to identify affected individuals at yet prediagnostic or prodromal stages of the illness. Such phases of the disease would provide an ideal window for neuroprotection trials. Taken together, these developments offer hope that a breakthrough towards modifying the course of PD might be reached. In this article we summarize various approaches currently pursued in this quest. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- Werner Poewe
- Department of Neurology, Medical University Innsbruck, Austria.
| | - Klaus Seppi
- Department of Neurology, Medical University Innsbruck, Austria
| | - Kathrin Marini
- Department of Neurology, Medical University Innsbruck, Austria
| | | |
Collapse
|
99
|
Altered features of monocytes in adult onset leukoencephalopathy with axonal spheroids and pigmented glia: A clue to the pathomechanism of microglial dyshomeostasis. Neurobiol Dis 2020; 140:104867. [PMID: 32276111 DOI: 10.1016/j.nbd.2020.104867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 01/21/2020] [Accepted: 04/05/2020] [Indexed: 01/03/2023] Open
Abstract
Adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) is an autosomal-dominant type of leukoencephalopathy caused by gene mutation of colony stimulating factor 1 receptor, which is expressed mainly on monocyte lineage cells such as monocytes in the peripheral blood and microglia in the brain. Hence, microglial dysfunction is regarded as critical in the pathogenesis of ALSP. However, functional changes in these cells have not been elucidated. In this study, we report the phenotypic and functional alterations of monocytes in four patients with ALSP. Flow cytometric analysis revealed altered expression of antigen presentation- and migration-related molecules, an inflammatory shift in cytokine production and phagocytic impairment in ALSP monocytes. We speculate that the observed altered features of monocytes are mostly shared by microglial cells, leading to the clinical history and pathological characteristics of ALSP. Our analysis of PB monocytes provides novel insights into the pathogenesis of ALSP.
Collapse
|
100
|
Schölwer I, Habib P, Voelz C, Rolfes L, Beyer C, Slowik A. NLRP3 Depletion Fails to Mitigate Inflammation but Restores Diminished Phagocytosis in BV-2 Cells After In Vitro Hypoxia. Mol Neurobiol 2020; 57:2588-2599. [PMID: 32239449 DOI: 10.1007/s12035-020-01909-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/23/2020] [Indexed: 12/30/2022]
Abstract
Post-hypoxic/ischemic neuroinflammation is selectively driven by sterile inflammation, which implies the interplay of brain-intrinsic immune cells with other neural cells and immigrated peripheral immune cells. The resultant inflammatory cascade evolves extra- and intracellular pathogen and danger-associated receptors. The latter interacts with multiprotein complexes termed inflammasomes. The NLRP3 inflammasome is one of the best-described inflammasomes. However, its impact on post-ischemic neuroinflammation and its role in neuroprotection after ischemic stroke are still under debate. Microglial cells are known to be the main source of neuroinflammation; hence, we depleted NLRP3 in BV-2 microglial cells using shRNA to investigate its role in IL-1β maturation and phagocytosis after hypoxia (oxygen-glucose-deprivation (OGD)). We also examined the expression profiles of other inflammasomes (NLRC4, AIM2, ASC) and caspase-1 activity after OGD. OGD triggered caspase-1 activity and increased IL-1β secretion in BV-2 cells with no alteration after NLRP3 depletion. The expression of the AIM2 inflammasome was significantly higher after OGD in NLRP3-depleted cells, whereas NLRC4 was unaltered in all groups. Interestingly, OGD induced a complete inactivation of phagocytic activity in wild-type cells, while in NLRP3-depleted BV-2, this inactivity was restored after hypoxia. Our findings indicate a minor role of NLRP3 in the inflammatory response after hypoxic/ischemic stimulus. However, NLRP3 seems to play a pivotal role in the regulation of post-ischemic phagocytosis. This might be a prerequisite for the putative neuroprotective effect.
Collapse
Affiliation(s)
- Isabelle Schölwer
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Clara Voelz
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Leoni Rolfes
- Neurology Clinic and Institute for Translational Neurology, University of Muenster, Münster, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany
- JARA-Brain, RWTH Aachen University, Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|