Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Cell ; 185(14): 2452-2468.e16, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35768006

ABSTRACT

COVID survivors frequently experience lingering neurological symptoms that resemble cancer-therapy-related cognitive impairment, a syndrome for which white matter microglial reactivity and consequent neural dysregulation is central. Here, we explored the neurobiological effects of respiratory SARS-CoV-2 infection and found white-matter-selective microglial reactivity in mice and humans. Following mild respiratory COVID in mice, persistently impaired hippocampal neurogenesis, decreased oligodendrocytes, and myelin loss were evident together with elevated CSF cytokines/chemokines including CCL11. Systemic CCL11 administration specifically caused hippocampal microglial reactivity and impaired neurogenesis. Concordantly, humans with lasting cognitive symptoms post-COVID exhibit elevated CCL11 levels. Compared with SARS-CoV-2, mild respiratory influenza in mice caused similar patterns of white-matter-selective microglial reactivity, oligodendrocyte loss, impaired neurogenesis, and elevated CCL11 at early time points, but after influenza, only elevated CCL11 and hippocampal pathology persisted. These findings illustrate similar neuropathophysiology after cancer therapy and respiratory SARS-CoV-2 infection which may contribute to cognitive impairment following even mild COVID.


Subject(s)
COVID-19 , Influenza, Human , Neoplasms , Animals , Humans , Influenza, Human/pathology , Mice , Microglia/pathology , Myelin Sheath , Neoplasms/pathology , SARS-CoV-2
2.
Cell ; 181(7): 1445-1449, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32533917

ABSTRACT

The COVID19 crisis has magnified the issues plaguing academic science, but it has also provided the scientific establishment with an unprecedented opportunity to reset. Shoring up the foundation of academic science will require a concerted effort between funding agencies, universities, and the public to rethink how we support scientists, with a special emphasis on early career researchers.


Subject(s)
Career Mobility , Research Personnel/trends , Research/trends , Achievement , Biomedical Research , Humans , Research Personnel/education , Science/education , Science/trends , Universities
3.
Cell ; 176(1-2): 43-55.e13, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30528430

ABSTRACT

Chemotherapy results in a frequent yet poorly understood syndrome of long-term neurological deficits. Neural precursor cell dysfunction and white matter dysfunction are thought to contribute to this debilitating syndrome. Here, we demonstrate persistent depletion of oligodendrocyte lineage cells in humans who received chemotherapy. Developing a mouse model of methotrexate chemotherapy-induced neurological dysfunction, we find a similar depletion of white matter OPCs, increased but incomplete OPC differentiation, and a persistent deficit in myelination. OPCs from chemotherapy-naive mice similarly exhibit increased differentiation when transplanted into the microenvironment of previously methotrexate-exposed brains, indicating an underlying microenvironmental perturbation. Methotrexate results in persistent activation of microglia and subsequent astrocyte activation that is dependent on inflammatory microglia. Microglial depletion normalizes oligodendroglial lineage dynamics, myelin microstructure, and cognitive behavior after methotrexate chemotherapy. These findings indicate that methotrexate chemotherapy exposure is associated with persistent tri-glial dysregulation and identify inflammatory microglia as a therapeutic target to abrogate chemotherapy-related cognitive impairment. VIDEO ABSTRACT.


Subject(s)
Cognitive Dysfunction/chemically induced , Methotrexate/adverse effects , Oligodendroglia/drug effects , Animals , Brain/metabolism , Cell Differentiation , Cell Lineage , Cognitive Dysfunction/metabolism , Disease Models, Animal , Drug Therapy , Drug-Related Side Effects and Adverse Reactions , Humans , Methotrexate/pharmacology , Mice , Microglia/metabolism , Myelin Sheath/metabolism , Nerve Fibers, Myelinated , Neurogenesis/physiology , Neuroglia/metabolism , Neurons/drug effects , Oligodendroglia/metabolism , White Matter/metabolism
4.
Physiol Rev ; 103(1): 919-956, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36173801

ABSTRACT

Studies of the choroid plexus lag behind those of the more widely known blood-brain barrier, despite a much longer history. This review has two overall aims. The first is to outline long-standing areas of research where there are unanswered questions, such as control of cerebrospinal fluid (CSF) secretion and blood flow. The second aim is to review research over the past 10 years where the focus has shifted to the idea that there are choroid plexuses located in each of the brain's ventricles that make specific contributions to brain development and function through molecules they generate for delivery via the CSF. These factors appear to be particularly important for aspects of normal brain growth. Most research carried out during the twentieth century dealt with the choroid plexus, a brain barrier interface making critical contributions to the composition and stability of the brain's internal environment throughout life. More recent research in the twenty-first century has shown the importance of choroid plexus-generated CSF in neurogenesis, influence of sex and other hormones on choroid plexus function, and choroid plexus involvement in circadian rhythms and sleep. The advancement of technologies to facilitate delivery of brain-specific therapies via the CSF to treat neurological disorders is a rapidly growing area of research. Conversely, understanding the basic mechanisms and implications of how maternal drug exposure during pregnancy impacts the developing brain represents another key area of research.


Subject(s)
Blood-Brain Barrier , Choroid Plexus , Humans , Blood-Brain Barrier/physiology , Brain , Biological Transport/physiology , Cerebral Ventricles
5.
Immunity ; 54(4): 614-616, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852828

ABSTRACT

Microbiome-induced interferon signaling through gut-derived natural killer cells is integral to minimize peripheral inflammatory responses in the brain and spinal cord. In a recent issue of Nature, Sanmarco, Wheeler, et al. define how interferon signaling induces LAMP1+TRAIL+ astrocytes, which cause death of inflammatory T cells, mitigating degeneration in a mouse model of demyealination.


Subject(s)
Astrocytes/immunology , Inflammation/immunology , T-Lymphocytes/immunology , Animals , Brain/immunology , Disease Models, Animal , Interferons/immunology , Killer Cells, Natural/immunology , Mice , Signal Transduction/immunology , Spinal Cord/immunology
6.
Immunity ; 54(2): 211-224, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33567261

ABSTRACT

Astrocytes play both physiological and pathological roles in maintaining central nervous system (CNS) function. Here, we review the varied functions of astrocytes and how these might change in subsets of reactive astrocytes. We review the current understanding of astrocyte interactions with microglia and the vasculature and protective barriers in the central nervous system as well as highlight recent insights into physiologic and reactive astrocyte sub-states identified by transcriptional profiling. Our goal is to stimulate inquiry into how these molecular identifiers link to specific functional changes in astrocytes and to define the implications of these heterogeneous molecular and functional changes in brain function and pathology. Defining these complex interactions has the potential to yield new therapies in CNS injury, infection, and disease.


Subject(s)
Astrocytes/immunology , Brain/pathology , Central Nervous System Diseases/immunology , Infections/immunology , Inflammation/immunology , Animals , Cell Communication , Humans , Immunity, Cellular , Neuroimmunomodulation
7.
Cell ; 162(5): 1170-1170.e1, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317476

ABSTRACT

Astrocytes are central nervous system (CNS) glial cells with many important functions for normal development and neural functioning. They help control extracellular ion and neurotransmitter concentrations; provide neurotrophic support; are implicated in synapse formation, function, and pruning; and help maintain the blood-brain barrier. Following injury and in disease, they undergo rapid and chronic alterations in function that can either promote or hinder recovery, depending on the disease.


Subject(s)
Astrocytes/metabolism , Central Nervous System Diseases/pathology , Animals , Humans , Neuroglia/metabolism
8.
Nature ; 601(7893): 404-409, 2022 01.
Article in English | MEDLINE | ID: mdl-34912118

ABSTRACT

During neurogenesis, mitotic progenitor cells lining the ventricles of the embryonic mouse brain undergo their final rounds of cell division, giving rise to a wide spectrum of postmitotic neurons and glia1,2. The link between developmental lineage and cell-type diversity remains an open question. Here we used massively parallel tagging of progenitors to track clonal relationships and transcriptomic signatures during mouse forebrain development. We quantified clonal divergence and convergence across all major cell classes postnatally, and found diverse types of GABAergic neuron that share a common lineage. Divergence of GABAergic clones occurred during embryogenesis upon cell-cycle exit, suggesting that differentiation into subtypes is initiated as a lineage-dependent process at the progenitor cell level.


Subject(s)
Brain , Cell Lineage , GABAergic Neurons , Neural Stem Cells , Neurogenesis , Animals , Brain/cytology , Cell Differentiation , Embryonic Development , GABAergic Neurons/cytology , Mice , Mitosis , Neural Stem Cells/cytology , Neurogenesis/genetics , Transcriptome
9.
Nat Immunol ; 21(5): 495-497, 2020 05.
Article in English | MEDLINE | ID: mdl-32284595

Subject(s)
Oxidative Stress
10.
Nature ; 599(7883): 102-107, 2021 11.
Article in English | MEDLINE | ID: mdl-34616039

ABSTRACT

Astrocytes regulate the response of the central nervous system to disease and injury and have been hypothesized to actively kill neurons in neurodegenerative disease1-6. Here we report an approach to isolate one component of the long-sought astrocyte-derived toxic factor5,6. Notably, instead of a protein, saturated lipids contained in APOE and APOJ lipoparticles mediate astrocyte-induced toxicity. Eliminating the formation of long-chain saturated lipids by astrocyte-specific knockout of the saturated lipid synthesis enzyme ELOVL1 mitigates astrocyte-mediated toxicity in vitro as well as in a model of acute axonal injury in vivo. These results suggest a mechanism by which astrocytes kill cells in the central nervous system.


Subject(s)
Astrocytes/chemistry , Astrocytes/metabolism , Cell Death/drug effects , Lipids/chemistry , Lipids/toxicity , Animals , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/toxicity , Fatty Acid Elongases/deficiency , Fatty Acid Elongases/genetics , Fatty Acid Elongases/metabolism , Female , Gene Knockout Techniques , Male , Mice , Mice, Knockout , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurotoxins/chemistry , Neurotoxins/toxicity
11.
Immunity ; 46(6): 957-967, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28636962

ABSTRACT

Astrocytes constitute approximately 30% of the cells in the mammalian central nervous system (CNS). They are integral to brain and spinal-cord physiology and perform many functions important for normal neuronal development, synapse formation, and proper propagation of action potentials. We still know very little, however, about how these functions change in response to immune attack, chronic neurodegenerative disease, or acute trauma. In this review, we summarize recent studies that demonstrate that different initiating CNS injuries can elicit at least two types of "reactive" astrocytes with strikingly different properties, one type being helpful and the other harmful. We will also discuss new methods for purifying and investigating reactive-astrocyte functions and provide an overview of new markers for delineating these different states of reactive astrocytes. The discovery that astrocytes have different types of reactive states has important implications for the development of new therapies for CNS injury and diseases.


Subject(s)
Astrocytes/physiology , Biological Therapy/trends , Brain/immunology , Central Nervous System/immunology , Neurodegenerative Diseases/immunology , Animals , Humans , Neurodegenerative Diseases/therapy , Neurons/physiology
12.
Nature ; 588(7838): 459-465, 2020 12.
Article in English | MEDLINE | ID: mdl-32866962

ABSTRACT

Aberrant aggregation of the RNA-binding protein TDP-43 in neurons is a hallmark of frontotemporal lobar degeneration caused by haploinsufficiency in the gene encoding progranulin1,2. However, the mechanism leading to TDP-43 proteinopathy remains unclear. Here we use single-nucleus RNA sequencing to show that progranulin deficiency promotes microglial transition from a homeostatic to a disease-specific state that causes endolysosomal dysfunction and neurodegeneration in mice. These defects persist even when Grn-/- microglia are cultured ex vivo. In addition, single-nucleus RNA sequencing reveals selective loss of excitatory neurons at disease end-stage, which is characterized by prominent nuclear and cytoplasmic TDP-43 granules and nuclear pore defects. Remarkably, conditioned media from Grn-/- microglia are sufficient to promote TDP-43 granule formation, nuclear pore defects and cell death in excitatory neurons via the complement activation pathway. Consistent with these results, deletion of the genes encoding C1qa and C3 mitigates microglial toxicity and rescues TDP-43 proteinopathy and neurodegeneration. These results uncover previously unappreciated contributions of chronic microglial toxicity to TDP-43 proteinopathy during neurodegeneration.


Subject(s)
Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Progranulins/deficiency , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology , Aging/genetics , Aging/pathology , Animals , Cell Nucleus/genetics , Cell Nucleus/pathology , Complement Activation/drug effects , Complement Activation/immunology , Complement C1q/antagonists & inhibitors , Complement C1q/immunology , Complement C3b/antagonists & inhibitors , Complement C3b/immunology , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Male , Mice , Nuclear Pore/metabolism , Nuclear Pore/pathology , Progranulins/genetics , RNA-Seq , Single-Cell Analysis , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , Thalamus/metabolism , Thalamus/pathology , Transcriptome
13.
Glia ; 72(3): 625-642, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38031883

ABSTRACT

Astrocytes are a heterogeneous population of central nervous system glial cells that respond to pathological insults and injury by undergoing a transformation called "reactivity." Reactive astrocytes exhibit distinct and context-dependent cellular, molecular, and functional state changes that can either support or disturb tissue homeostasis. We recently identified a reactive astrocyte sub-state defined by interferon-responsive genes like Igtp, Ifit3, Mx1, and others, called interferon-responsive reactive astrocytes (IRRAs). To further this transcriptomic definition of IRRAs, we wanted to define the proteomic changes that occur in this reactive sub-state. We induced IRRAs in immunopanned rodent astrocytes and human iPSC-differentiated astrocytes using TNF, IL1α, C1Q, and IFNß and characterized their proteomic profile (both cellular and secreted) using unbiased quantitative proteomics. We identified 2335 unique cellular proteins, including IFIT2/3, IFITM3, OASL1/2, MX1/2/3, and STAT1. We also report that rodent and human IRRAs secrete PAI1, a serine protease inhibitor which may influence reactive states and functions of nearby cells. Finally, we evaluated how IRRAs are distinct from neurotoxic reactive astrocytes (NRAs). While NRAs are described by expression of the complement protein C3, it was not upregulated in IRRAs. Instead, we found ~90 proteins unique to IRRAs not identified in NRAs, including OAS1A, IFIT3, and MX1. Interferon signaling in astrocytes is critical for the antiviral immune response and for regulating synaptic plasticity and glutamate transport mechanisms. How IRRAs contribute to these functions is unknown. This study provides the basis for future experiments to define the functional roles of IRRAs in the context of neurodegenerative disorders.


Subject(s)
Astrocytes , Interferons , Animals , Humans , Astrocytes/metabolism , Interferons/metabolism , Rodentia/metabolism , Proteomics , Central Nervous System/metabolism , Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism
14.
Glia ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38924630

ABSTRACT

Perinatal white matter injury (WMI) is the leading cause of long-term neurological morbidity in infants born preterm. Neuroinflammation during a critical window of early brain development plays a key role in WMI disease pathogenesis. The mechanisms linking inflammation with the long-term myelination failure that characterizes WMI, however, remain unknown. Here, we investigate the role of astrocyte reactivity in WMI. In an experimental mouse model of WMI, we demonstrate that WMI disease outcomes are improved in mutant mice lacking secretion of inflammatory molecules TNF-α, IL-1α, and C1q known, in addition to other roles, to induce the formation of a neuroinflammatory reactive astrocyte substate. We show that astrocytes express molecular signatures of the neuroinflammatory reactive astrocyte substate in both our WMI mouse model and human tissue affected by WMI, and that this gene expression pattern is dampened in injured mutant mice. Our data provide evidence that a neuroinflammatory reactive astrocyte substate correlates with adverse WMI disease outcomes, thus highlighting the need for further investigation of these cells as potential causal players in WMI pathology.

15.
Trends Immunol ; 41(9): 820-835, 2020 09.
Article in English | MEDLINE | ID: mdl-32819809

ABSTRACT

Microglia-astrocyte interactions represent a delicate balance affecting neural cell functions in health and disease. Tightly controlled to maintain homeostasis during physiological conditions, rapid and prolonged departures during disease, infection, and following trauma drive multiple outcomes: both beneficial and detrimental. Recent sequencing studies at the bulk and single-cell level in humans and rodents provide new insight into microglia-astrocyte communication in homeostasis and disease. However, the complex changing ways these two cell types functionally interact has been a barrier to understanding disease initiation, progression, and disease mechanisms. Single cell sequencing is providing new insights; however, many questions remain. Here, we discuss how to bridge transcriptional states to specific functions so we can develop therapies to mediate negative effects of altered microglia-astrocyte interactions.


Subject(s)
Astrocytes , Microglia , Animals , Astrocytes/immunology , Cell Communication/immunology , Homeostasis/immunology , Humans , Microglia/immunology , Nervous System Diseases/immunology , Neurons/cytology , Neurons/immunology
16.
Nature ; 541(7638): 481-487, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28099414

ABSTRACT

Reactive astrocytes are strongly induced by central nervous system (CNS) injury and disease, but their role is poorly understood. Here we show that a subtype of reactive astrocytes, which we termed A1, is induced by classically activated neuroinflammatory microglia. We show that activated microglia induce A1 astrocytes by secreting Il-1α, TNF and C1q, and that these cytokines together are necessary and sufficient to induce A1 astrocytes. A1 astrocytes lose the ability to promote neuronal survival, outgrowth, synaptogenesis and phagocytosis, and induce the death of neurons and oligodendrocytes. Death of axotomized CNS neurons in vivo is prevented when the formation of A1 astrocytes is blocked. Finally, we show that A1 astrocytes are abundant in various human neurodegenerative diseases including Alzheimer's, Huntington's and Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. Taken together these findings help to explain why CNS neurons die after axotomy, strongly suggest that A1 astrocytes contribute to the death of neurons and oligodendrocytes in neurodegenerative disorders, and provide opportunities for the development of new treatments for these diseases.


Subject(s)
Astrocytes/classification , Astrocytes/pathology , Cell Death , Central Nervous System/pathology , Microglia/pathology , Neurons/pathology , Animals , Astrocytes/metabolism , Axotomy , Cell Culture Techniques , Cell Survival , Complement C1q/metabolism , Disease Progression , Humans , Inflammation/pathology , Interleukin-1alpha/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neurodegenerative Diseases/pathology , Oligodendroglia/pathology , Phagocytosis , Phenotype , Rats , Rats, Sprague-Dawley , Synapses/pathology , Toxins, Biological/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Nature ; 549(7673): 523-527, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28959956

ABSTRACT

APOE4 is the strongest genetic risk factor for late-onset Alzheimer disease. ApoE4 increases brain amyloid-ß pathology relative to other ApoE isoforms. However, whether APOE independently influences tau pathology, the other major proteinopathy of Alzheimer disease and other tauopathies, or tau-mediated neurodegeneration, is not clear. By generating P301S tau transgenic mice on either a human ApoE knock-in (KI) or ApoE knockout (KO) background, here we show that P301S/E4 mice have significantly higher tau levels in the brain and a greater extent of somatodendritic tau redistribution by three months of age compared with P301S/E2, P301S/E3, and P301S/EKO mice. By nine months of age, P301S mice with different ApoE genotypes display distinct phosphorylated tau protein (p-tau) staining patterns. P301S/E4 mice develop markedly more brain atrophy and neuroinflammation than P301S/E2 and P301S/E3 mice, whereas P301S/EKO mice are largely protected from these changes. In vitro, E4-expressing microglia exhibit higher innate immune reactivity after lipopolysaccharide treatment. Co-culturing P301S tau-expressing neurons with E4-expressing mixed glia results in a significantly higher level of tumour-necrosis factor-α (TNF-α) secretion and markedly reduced neuronal viability compared with neuron/E2 and neuron/E3 co-cultures. Neurons co-cultured with EKO glia showed the greatest viability with the lowest level of secreted TNF-α. Treatment of P301S neurons with recombinant ApoE (E2, E3, E4) also leads to some neuronal damage and death compared with the absence of ApoE, with ApoE4 exacerbating the effect. In individuals with a sporadic primary tauopathy, the presence of an ε4 allele is associated with more severe regional neurodegeneration. In individuals who are positive for amyloid-ß pathology with symptomatic Alzheimer disease who usually have tau pathology, ε4-carriers demonstrate greater rates of disease progression. Our results demonstrate that ApoE affects tau pathogenesis, neuroinflammation, and tau-mediated neurodegeneration independently of amyloid-ß pathology. ApoE4 exerts a 'toxic' gain of function whereas the absence of ApoE is protective.


Subject(s)
Apolipoprotein E4/metabolism , Apolipoprotein E4/toxicity , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/metabolism , Alleles , Animals , Apolipoprotein E4/deficiency , Apolipoprotein E4/genetics , Cell Survival/drug effects , Coculture Techniques , Disease Models, Animal , Disease Progression , Gene Knock-In Techniques , Genotype , Humans , Immunity, Innate , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phosphoproteins/analysis , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Tauopathies/genetics , Tumor Necrosis Factor-alpha/metabolism , tau Proteins/genetics
18.
Alzheimers Dement ; 19(6): 2239-2252, 2023 06.
Article in English | MEDLINE | ID: mdl-36448627

ABSTRACT

INTRODUCTION: The inositol polyphosphate-5-phosphatase D (INPP5D) gene encodes a dual-specificity phosphatase that can dephosphorylate both phospholipids and phosphoproteins. Single nucleotide polymorphisms in INPP5D impact risk for developing late onset sporadic Alzheimer's disease (LOAD). METHODS: To assess the consequences of inducible Inpp5d knockdown in microglia of APPKM670/671NL /PSEN1Δexon9 (PSAPP) mice, we injected 3-month-old Inpp5dfl/fl /Cx3cr1CreER/+ and PSAPP/Inpp5dfl/fl /Cx3cr1CreER/+ mice with either tamoxifen (TAM) or corn oil (CO) to induce recombination. RESULTS: At age 6 months, we found that the percent area of 6E10+ deposits and plaque-associated microglia in Inpp5d knockdown mice were increased compared to controls. Spatial transcriptomics identified a plaque-specific expression profile that was extensively altered by Inpp5d knockdown. DISCUSSION: These results demonstrate that conditional Inpp5d downregulation in the PSAPP mouse increases plaque burden and recruitment of microglia to plaques. Spatial transcriptomics highlighted an extended gene expression signature associated with plaques and identified CST7 (cystatin F) as a novel marker of plaques. HIGHLIGHTS: Inpp5d knockdown increases plaque burden and plaque-associated microglia number. Spatial transcriptomics identifies an expanded plaque-specific gene expression profile. Plaque-induced gene expression is altered by Inpp5d knockdown in microglia. Our plaque-associated gene signature overlaps with human Alzheimer's disease gene networks.


Subject(s)
Alzheimer Disease , Mice , Humans , Animals , Infant , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Microglia/metabolism , Mice, Transgenic , Plaque, Amyloid/metabolism , Disease Models, Animal , Amyloid beta-Peptides/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism
19.
Alzheimers Dement ; 19(6): 2677-2696, 2023 06.
Article in English | MEDLINE | ID: mdl-36975090

ABSTRACT

INTRODUCTION: At the Alzheimer's Association's APOE and Immunity virtual conference, held in October 2021, leading neuroscience experts shared recent research advances on and inspiring insights into the various roles that both the apolipoprotein E gene (APOE) and facets of immunity play in neurodegenerative diseases, including Alzheimer's disease and other dementias. METHODS: The meeting brought together more than 1200 registered attendees from 62 different countries, representing the realms of academia and industry. RESULTS: During the 4-day meeting, presenters illuminated aspects of the cross-talk between APOE and immunity, with a focus on the roles of microglia, triggering receptor expressed on myeloid cells 2 (TREM2), and components of inflammation (e.g., tumor necrosis factor α [TNFα]). DISCUSSION: This manuscript emphasizes the importance of diversity in current and future research and presents an integrated view of innate immune functions in Alzheimer's disease as well as related promising directions in drug development.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/pathology , Microglia/pathology , Inflammation , Apolipoproteins E/genetics
20.
Glia ; 70(10): 1950-1970, 2022 10.
Article in English | MEDLINE | ID: mdl-35809238

ABSTRACT

Multiple sclerosis (MS) is a central nervous system (CNS) autoimmune disease characterized by inflammation, demyelination, and neurodegeneration. The ideal MS therapy would both specifically inhibit the underlying autoimmune response and promote repair/regeneration of myelin as well as maintenance of axonal integrity. Currently approved MS therapies consist of non-specific immunosuppressive molecules/antibodies which block activation or CNS homing of autoreactive T cells, but there are no approved therapies for stimulation of remyelination nor maintenance of axonal integrity. In an effort to repurpose an FDA-approved medication for myelin repair, we chose to examine the effectiveness of digoxin, a cardiac glycoside (Na+ /K+ ATPase inhibitor), originally identified as pro-myelinating in an in vitro screen. We found that digoxin regulated multiple genes in oligodendrocyte progenitor cells (OPCs) essential for oligodendrocyte (OL) differentiation in vitro, promoted OL differentiation both in vitro and in vivo in female naïve C57BL/6J (B6) mice, and stimulated recovery of myelinated axons in B6 mice following demyelination in the corpus callosum induced by cuprizone and spinal cord demyelination induced by lysophosphatidylcholine (LPC), respectively. More relevant to treatment of MS, we show that digoxin treatment of mice with established MOG35-55 -induced Th1/Th17-mediated chronic EAE combined with tolerance induced by the i.v. infusion of biodegradable poly(lactide-co-glycolide) nanoparticles coupled with MOG35-55 (PLG-MOG35-55 ) completely ameliorated clinical disease symptoms and stimulated recovery of OL lineage cell numbers. These findings provide critical pre-clinical evidence supporting future clinical trials of myelin-specific tolerance with myelin repair/regeneration drugs, such as digoxin, in MS patients.


Subject(s)
Cardiac Glycosides , Demyelinating Diseases , Multiple Sclerosis , Animals , Cardiac Glycosides/adverse effects , Cell Differentiation , Cuprizone , Demyelinating Diseases/chemically induced , Digoxin/adverse effects , Disease Models, Animal , Drug Repositioning , Female , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Myelin Sheath/physiology , Oligodendroglia/physiology
SELECTION OF CITATIONS
SEARCH DETAIL