Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Trends Pharmacol Sci ; 44(10): 674-688, 2023 10.
Article in English | MEDLINE | ID: mdl-37657967

ABSTRACT

Iron accumulation has been associated with the etiology and progression of multiple neurodegenerative diseases (NDDs). The exact role of iron in these diseases is not fully understood, but an iron-dependent form of regulated cell death called ferroptosis could be key. Although there is substantial preclinical and clinical evidence that ferroptosis plays a role in NDD, there are still questions regarding how to target ferroptosis therapeutically, including which proteins to target, identification of clinically relevant biomarkers, and which patients might benefit most. Clinical trials of iron- and ferroptosis-targeted therapies are beginning to provide some answers, but there is growing interest in developing new ferroptosis inhibitors. We describe newly identified ferroptosis targets, opportunities, and challenges in NDD, as well as key considerations for progressing new therapeutics to the clinic.


Subject(s)
Ferroptosis , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/drug therapy , Iron
2.
Immunity ; 56(9): 1985-1987, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37703828

ABSTRACT

PLCγ2 is genetically linked to Alzheimer's disease (AD), but it is unclear how PLCγ2 contributes to pathology. Tsai et al. demonstrate that AD-associated PLCG2 variants bidirectionally orchestrate microglial responses to plaques and impact neural function in an AD mouse model. This positions PLCγ2 as a key microglial signaling node and shows that targeting PLCγ2 could have therapeutic benefits in AD.


Subject(s)
Microglia , Plaque, Amyloid , Animals , Mice , Phospholipase C gamma/genetics , Disease Models, Animal
3.
Nat Neurosci ; 26(1): 12-26, 2023 01.
Article in English | MEDLINE | ID: mdl-36536241

ABSTRACT

Iron dysregulation has been implicated in multiple neurodegenerative diseases, including Parkinson's disease (PD). Iron-loaded microglia are frequently found in affected brain regions, but how iron accumulation influences microglia physiology and contributes to neurodegeneration is poorly understood. Here we show that human induced pluripotent stem cell-derived microglia grown in a tri-culture system are highly responsive to iron and susceptible to ferroptosis, an iron-dependent form of cell death. Furthermore, iron overload causes a marked shift in the microglial transcriptional state that overlaps with a transcriptomic signature found in PD postmortem brain microglia. Our data also show that this microglial response contributes to neurodegeneration, as removal of microglia from the tri-culture system substantially delayed iron-induced neurotoxicity. To elucidate the mechanisms regulating iron response in microglia, we performed a genome-wide CRISPR screen and identified novel regulators of ferroptosis, including the vesicle trafficking gene SEC24B. These data suggest a critical role for microglia iron overload and ferroptosis in neurodegeneration.


Subject(s)
Ferroptosis , Induced Pluripotent Stem Cells , Iron Overload , Parkinson Disease , Humans , Induced Pluripotent Stem Cells/metabolism , Iron/metabolism , Iron Overload/metabolism , Microglia/metabolism , Parkinson Disease/genetics
4.
Neurotherapeutics ; 19(3): 864-873, 2022 04.
Article in English | MEDLINE | ID: mdl-35378684

ABSTRACT

Chronic inflammatory demyelinating polyneuropathy (CIDP) is the most common, heterogeneous, immune-mediated neuropathy, characterized by predominant demyelination of motor and sensory nerves. CIDP follows a relapsing-remitting or a progressive course and causes substantial disability. The pathogenesis of CIDP involves a complex interplay of multiple aberrant immune responses, creating a pro-inflammatory environment, subsequently inflicting damage on the myelin sheath. Though the exact triggers are unclear, diverse immune mechanisms encompassing cellular and humoral pathways are implicated. The complement system appears to play a role in promoting macrophage-mediated demyelination. Complement deposition in sural nerve biopsies, as well as signs of increased complement activation in serum and CSF of patients with CIDP, suggest complement involvement in CIDP pathogenesis. Here, we present a comprehensive overview of the preclinical and clinical evidence supporting the potential role of the complement system in CIDP. This understanding furnishes a strong rationale for targeting the complement system to develop new therapies that could serve the unmet needs of patients affected by CIDP, particularly in those refractory to standard therapies.


Subject(s)
Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Biopsy , Humans , Macrophages/pathology , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnosis , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/drug therapy
5.
Nat Neurosci ; 25(3): 306-316, 2022 03.
Article in English | MEDLINE | ID: mdl-35260865

ABSTRACT

A key aspect of nearly all single-cell sequencing experiments is dissociation of intact tissues into single-cell suspensions. While many protocols have been optimized for optimal cell yield, they have often overlooked the effects that dissociation can have on ex vivo gene expression. Here, we demonstrate that use of enzymatic dissociation on brain tissue induces an aberrant ex vivo gene expression signature, most prominently in microglia, which is prevalent in published literature and can substantially confound downstream analyses. To address this issue, we present a rigorously validated protocol that preserves both in vivo transcriptional profiles and cell-type diversity and yield across tissue types and species. We also identify a similar signature in postmortem human brain single-nucleus RNA-sequencing datasets, and show that this signature is induced in freshly isolated human tissue by exposure to elevated temperatures ex vivo. Together, our results provide a methodological solution for preventing artifactual gene expression changes during fresh tissue digestion and a reference for future deeper analysis of the potential confounding states present in postmortem human samples.


Subject(s)
Neuroglia , Transcriptome , Brain , Humans , Microglia/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
7.
Nat Genet ; 53(6): 861-868, 2021 06.
Article in English | MEDLINE | ID: mdl-34083789

ABSTRACT

Microglia, the tissue-resident macrophages of the central nervous system (CNS), play critical roles in immune defense, development and homeostasis. However, isolating microglia from humans in large numbers is challenging. Here, we profiled gene expression variation in primary human microglia isolated from 141 patients undergoing neurosurgery. Using single-cell and bulk RNA sequencing, we identify how age, sex and clinical pathology influence microglia gene expression and which genetic variants have microglia-specific functions using expression quantitative trait loci (eQTL) mapping. We follow up one of our findings using a human induced pluripotent stem cell-based macrophage model to fine-map a candidate causal variant for Alzheimer's disease at the BIN1 locus. Our study provides a population-scale transcriptional map of a critically important cell for human CNS development and disease.


Subject(s)
Gene Expression Regulation , Microglia/metabolism , Transcription, Genetic , Alzheimer Disease/genetics , Humans , Models, Genetic , Quantitative Trait Loci/genetics , Sequence Analysis, RNA , Single-Cell Analysis
8.
Immunity ; 50(4): 955-974, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995509

ABSTRACT

Neurodegenerative diseases of the central nervous system progressively rob patients of their memory, motor function, and ability to perform daily tasks. Advances in genetics and animal models are beginning to unearth an unexpected role of the immune system in disease onset and pathogenesis; however, the role of cytokines, growth factors, and other immune signaling pathways in disease pathogenesis is still being examined. Here we review recent genetic risk and genome-wide association studies and emerging mechanisms for three key immune pathways implicated in disease, the growth factor TGF-ß, the complement cascade, and the extracellular receptor TREM2. These immune signaling pathways are important under both healthy and neurodegenerative conditions, and recent work has highlighted new functional aspects of their signaling. Finally, we assess future directions for immune-related research in neurodegeneration and potential avenues for immune-related therapies.


Subject(s)
Neurodegenerative Diseases/immunology , Signal Transduction/immunology , Aging/immunology , Animals , Complement Activation , Disease Progression , Genetic Predisposition to Disease , Genome-Wide Association Study , Gliosis/immunology , Gliosis/pathology , Humans , Immunity, Innate , Inflammation/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Mice, Transgenic , Microglia/immunology , Models, Immunological , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy , Protein Aggregation, Pathological/immunology , Receptors, Immunologic/immunology , Transforming Growth Factor beta/immunology
9.
Immunity ; 50(1): 253-271.e6, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30471926

ABSTRACT

Microglia, the resident immune cells of the brain, rapidly change states in response to their environment, but we lack molecular and functional signatures of different microglial populations. Here, we analyzed the RNA expression patterns of more than 76,000 individual microglia in mice during development, in old age, and after brain injury. Our analysis uncovered at least nine transcriptionally distinct microglial states, which expressed unique sets of genes and were localized in the brain using specific markers. The greatest microglial heterogeneity was found at young ages; however, several states-including chemokine-enriched inflammatory microglia-persisted throughout the lifespan or increased in the aged brain. Multiple reactive microglial subtypes were also found following demyelinating injury in mice, at least one of which was also found in human multiple sclerosis lesions. These distinct microglia signatures can be used to better understand microglia function and to identify and manipulate specific subpopulations in health and disease.


Subject(s)
Aging/immunology , Brain Injuries/immunology , Brain/physiology , Microglia/physiology , Multiple Sclerosis/immunology , Adaptation, Physiological , Aging/genetics , Animals , Brain Injuries/genetics , Cell Differentiation , Demyelinating Diseases , Humans , Longevity , Mice , Mice, Inbred C57BL , Sequence Analysis, RNA , Single-Cell Analysis
10.
Annu Rev Cell Dev Biol ; 34: 523-544, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30089221

ABSTRACT

An explosion of findings driven by powerful new technologies has expanded our understanding of microglia, the resident immune cells of the central nervous system (CNS). This wave of discoveries has fueled a growing interest in the roles that these cells play in the development of the CNS and in the neuropathology of a diverse array of disorders. In this review, we discuss the crucial roles that microglia play in shaping the brain-from their influence on neurons and glia within the developing CNS to their roles in synaptic maturation and brain wiring-as well as some of the obstacles to overcome when assessing their contributions to normal brain development. Furthermore, we examine how normal developmental functions of microglia are perturbed or remerge in neurodevelopmental and neurodegenerative disease.


Subject(s)
Brain/growth & development , Central Nervous System/growth & development , Microglia/metabolism , Neurons/metabolism , Animals , Brain/metabolism , Brain/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Humans , Microglia/pathology , Neurodegenerative Diseases , Neuroglia/metabolism , Neuroglia/pathology , Neurons/pathology , Signal Transduction/genetics
11.
Elife ; 62017 03 13.
Article in English | MEDLINE | ID: mdl-28244870

ABSTRACT

The medial subnucleus of the amygdala (MeA) plays a central role in processing sensory cues required for innate behaviors. However, whether there is a link between developmental programs and the emergence of inborn behaviors remains unknown. Our previous studies revealed that the telencephalic preoptic area (POA) embryonic niche is a novel source of MeA destined progenitors. Here, we show that the POA is comprised of distinct progenitor pools complementarily marked by the transcription factors Dbx1 and Foxp2. As determined by molecular and electrophysiological criteria this embryonic parcellation predicts postnatal MeA inhibitory neuronal subtype identity. We further find that Dbx1-derived and Foxp2+ cells in the MeA are differentially activated in response to innate behavioral cues in a sex-specific manner. Thus, developmental transcription factor expression is predictive of MeA neuronal identity and sex-specific neuronal responses, providing a potential developmental logic for how innate behaviors could be processed by different MeA neuronal subtypes.


Subject(s)
Corticomedial Nuclear Complex/embryology , Corticomedial Nuclear Complex/physiology , Forkhead Transcription Factors/analysis , Homeodomain Proteins/analysis , Instinct , Neurons/physiology , Repressor Proteins/analysis , Animals , Cues , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Sex Factors
13.
Nature ; 530(7589): 177-83, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26814963

ABSTRACT

Schizophrenia is a heritable brain illness with unknown pathogenic mechanisms. Schizophrenia's strongest genetic association at a population level involves variation in the major histocompatibility complex (MHC) locus, but the genes and molecular mechanisms accounting for this have been challenging to identify. Here we show that this association arises in part from many structurally diverse alleles of the complement component 4 (C4) genes. We found that these alleles generated widely varying levels of C4A and C4B expression in the brain, with each common C4 allele associating with schizophrenia in proportion to its tendency to generate greater expression of C4A. Human C4 protein localized to neuronal synapses, dendrites, axons, and cell bodies. In mice, C4 mediated synapse elimination during postnatal development. These results implicate excessive complement activity in the development of schizophrenia and may help explain the reduced numbers of synapses in the brains of individuals with schizophrenia.


Subject(s)
Complement C4/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Schizophrenia/genetics , Alleles , Amino Acid Sequence , Animals , Axons/metabolism , Base Sequence , Brain/metabolism , Brain/pathology , Complement C4/chemistry , Complement Pathway, Classical , Dendrites/metabolism , Gene Dosage/genetics , Gene Expression Regulation/genetics , Haplotypes/genetics , Humans , Major Histocompatibility Complex/genetics , Mice , Models, Animal , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Polymorphism, Single Nucleotide/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Risk Factors , Schizophrenia/pathology , Synapses/metabolism
14.
Cell Rep ; 13(10): 2090-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26628380

ABSTRACT

Reactive astrogliosis is an essential and ubiquitous response to CNS injury, but in some cases, aberrant activation of astrocytes and their release of inhibitory signaling molecules can impair endogenous neural repair processes. Our lab previously identified a secreted intercellular signaling molecule, called endothelin-1 (ET-1), which is expressed at high levels by reactive astrocytes in multiple sclerosis (MS) lesions and limits repair by delaying oligodendrocyte progenitor cell (OPC) maturation. However, as ET receptors are widely expressed on neural cells, the cell- and receptor-specific mechanisms of OPC inhibition by ET-1 action remain undefined. Using pharmacological approaches and cell-specific endothelin receptor (EDNR) ablation, we show that ET-1 acts selectively through EDNRB on astrocytes--and not OPCs--to indirectly inhibit remyelination. These results demonstrate that targeting specific pathways in reactive astrocytes represents a promising therapeutic target in diseases with extensive reactive astrogliosis, including MS.


Subject(s)
Astrocytes/metabolism , Demyelinating Diseases/metabolism , Neural Stem Cells/metabolism , Receptor, Endothelin B/metabolism , Animals , Cell Differentiation/physiology , Disease Models, Animal , Endothelin-1/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Electron, Transmission , Neural Stem Cells/cytology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Regeneration/physiology
15.
Neuron ; 81(3): 588-602, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24507193

ABSTRACT

Oligodendrocyte progenitor cells (OPCs) can repair demyelinated lesions by maturing into myelin-producing oligodendrocytes. However, the OPC potential to differentiate can be prevented by inhibitory signals present in the pathological lesion environment. Identification of these signals is essential to promote OPC differentiation and lesion repair. We identified an endogenous inhibitor of remyelination, Endothelin-1 (ET-1), which is highly expressed in reactive astrocytes of demyelinated lesions. Using both gain- and loss-of-function approaches, we demonstrate that ET-1 drastically reduces the rate of remyelination. We also discovered that ET-1 acts mechanistically by promoting Notch activation in OPCs during remyelination through induction of Jagged1 expression in reactive astrocytes. Pharmacological inhibition of ET signaling prevented Notch activation in demyelinated lesions and accelerated remyelination. These findings reveal that ET-1 is a negative regulator of OPC differentiation and remyelination and is potentially a therapeutic target to promote lesion repair in demyelinated tissue.


Subject(s)
Astrocytes/metabolism , Demyelinating Diseases/pathology , Endothelin-1/metabolism , Gene Expression Regulation/physiology , Receptors, Notch/metabolism , Animals , Astrocytes/drug effects , Astrocytes/ultrastructure , Calcium-Binding Proteins/metabolism , Cell Count , Cell Differentiation/drug effects , Demyelinating Diseases/chemically induced , Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Disease Models, Animal , Drug Delivery Systems , Endothelin-1/adverse effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/genetics , Green Fluorescent Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Lipopolysaccharides/pharmacology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/pharmacology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Serrate-Jagged Proteins , Stem Cells/drug effects , Stem Cells/physiology
16.
Nature ; 506(7487): 230-4, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24390343

ABSTRACT

There are no clinically relevant treatments available that improve function in the growing population of very preterm infants (less than 32 weeks' gestation) with neonatal brain injury. Diffuse white matter injury (DWMI) is a common finding in these children and results in chronic neurodevelopmental impairments. As shown recently, failure in oligodendrocyte progenitor cell maturation contributes to DWMI. We demonstrated previously that the epidermal growth factor receptor (EGFR) has an important role in oligodendrocyte development. Here we examine whether enhanced EGFR signalling stimulates the endogenous response of EGFR-expressing progenitor cells during a critical period after brain injury, and promotes cellular and behavioural recovery in the developing brain. Using an established mouse model of very preterm brain injury, we demonstrate that selective overexpression of human EGFR in oligodendrocyte lineage cells or the administration of intranasal heparin-binding EGF immediately after injury decreases oligodendroglia death, enhances generation of new oligodendrocytes from progenitor cells and promotes functional recovery. Furthermore, these interventions diminish ultrastructural abnormalities and alleviate behavioural deficits on white-matter-specific paradigms. Inhibition of EGFR signalling with a molecularly targeted agent used for cancer therapy demonstrates that EGFR activation is an important contributor to oligodendrocyte regeneration and functional recovery after DWMI. Thus, our study provides direct evidence that targeting EGFR in oligodendrocyte progenitor cells at a specific time after injury is clinically feasible and potentially applicable to the treatment of premature children with white matter injury.


Subject(s)
Brain Injuries/congenital , Brain Injuries/drug therapy , Epidermal Growth Factor/pharmacology , Epidermal Growth Factor/therapeutic use , Oligodendroglia/drug effects , Administration, Intranasal , Animals , Animals, Newborn , Brain Injuries/pathology , Brain Injuries/prevention & control , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Lineage/drug effects , Cell Survival/drug effects , Demyelinating Diseases/congenital , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Demyelinating Diseases/prevention & control , Disease Models, Animal , Epidermal Growth Factor/administration & dosage , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia/physiopathology , Infant, Premature, Diseases/drug therapy , Infant, Premature, Diseases/metabolism , Infant, Premature, Diseases/pathology , Male , Mice , Molecular Targeted Therapy , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Regeneration/drug effects , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...