Try a new search

Format these results:

Searched for:

person:liddes01

in-biosketch:yes

Total Results:

87


Neurotoxic reactive astrocytes are induced by activated microglia

Liddelow, Shane A; Guttenplan, Kevin A; Clarke, Laura E; Bennett, Frederick C; Bohlen, Christopher J; Schirmer, Lucas; Bennett, Mariko L; Munch, Alexandra E; Chung, Won-Suk; Peterson, Todd C; Wilton, Daniel K; Frouin, Arnaud; Napier, Brooke A; Panicker, Nikhil; Kumar, Manoj; Buckwalter, Marion S; Rowitch, David H; Dawson, Valina L; Dawson, Ted M; Stevens, Beth; Barres, Ben A
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-1alpha, 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.
PMCID:5404890
PMID: 28099414
ISSN: 1476-4687
CID: 2743332

Normal aging induces A1-like astrocyte reactivity

Clarke, Laura E; Liddelow, Shane A; Chakraborty, Chandrani; Münch, Alexandra E; Heiman, Myriam; Barres, Ben A
The decline of cognitive function occurs with aging, but the mechanisms responsible are unknown. Astrocytes instruct the formation, maturation, and elimination of synapses, and impairment of these functions has been implicated in many diseases. These findings raise the question of whether astrocyte dysfunction could contribute to cognitive decline in aging. We used the Bac-Trap method to perform RNA sequencing of astrocytes from different brain regions across the lifespan of the mouse. We found that astrocytes have region-specific transcriptional identities that change with age in a region-dependent manner. We validated our findings using fluorescence in situ hybridization and quantitative PCR. Detailed analysis of the differentially expressed genes in aging revealed that aged astrocytes take on a reactive phenotype of neuroinflammatory A1-like reactive astrocytes. Hippocampal and striatal astrocytes up-regulated a greater number of reactive astrocyte genes compared with cortical astrocytes. Moreover, aged brains formed many more A1 reactive astrocytes in response to the neuroinflammation inducer lipopolysaccharide. We found that the aging-induced up-regulation of reactive astrocyte genes was significantly reduced in mice lacking the microglial-secreted cytokines (IL-1α, TNF, and C1q) known to induce A1 reactive astrocyte formation, indicating that microglia promote astrocyte activation in aging. Since A1 reactive astrocytes lose the ability to carry out their normal functions, produce complement components, and release a toxic factor which kills neurons and oligodendrocytes, the aging-induced up-regulation of reactive genes by astrocytes could contribute to the cognitive decline in vulnerable brain regions in normal aging and contribute to the greater vulnerability of the aged brain to injury.
PMCID:5828643
PMID: 29437957
ISSN: 1091-6490
CID: 2958252

ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy

Shi, Yang; Yamada, Kaoru; Liddelow, Shane Antony; Smith, Scott T; Zhao, Lingzhi; Luo, Wenjie; Tsai, Richard M; Spina, Salvatore; Grinberg, Lea T; Rojas, Julio C; Gallardo, Gilbert; Wang, Kairuo; Roh, Joseph; Robinson, Grace; Finn, Mary Beth; Jiang, Hong; Sullivan, Patrick M; Baufeld, Caroline; Wood, Michael W; Sutphen, Courtney; McCue, Lena; Xiong, Chengjie; Del-Aguila, Jorge L; Morris, John C; Cruchaga, Carlos; Fagan, Anne M; Miller, Bruce L; Boxer, Adam L; Seeley, William W; Butovsky, Oleg; Barres, Ben A; Paul, Steven M; Holtzman, David M
APOE4 is the strongest genetic risk factor for late-onset Alzheimer disease. ApoE4 increases brain amyloid-beta 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-alpha (TNF-alpha) 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-alpha. 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 epsilon4 allele is associated with more severe regional neurodegeneration. In individuals who are positive for amyloid-beta pathology with symptomatic Alzheimer disease who usually have tau pathology, epsilon4-carriers demonstrate greater rates of disease progression. Our results demonstrate that ApoE affects tau pathogenesis, neuroinflammation, and tau-mediated neurodegeneration independently of amyloid-beta pathology. ApoE4 exerts a 'toxic' gain of function whereas the absence of ApoE is protective.
PMCID:5641217
PMID: 28959956
ISSN: 1476-4687
CID: 2743932

Proteomic profiling of interferon-responsive reactive astrocytes in rodent and human

Prakash, Priya; Erdjument-Bromage, Hediye; O'Dea, Michael R; Munson, Christy N; Labib, David; Fossati, Valentina; Neubert, Thomas A; Liddelow, Shane A
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 IFNβ 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.
PMID: 38031883
ISSN: 1098-1136
CID: 5616902

Reactive Astrocytes and Emerging Roles in Central Nervous System (CNS) Disorders

Liddelow, Shane A; Olsen, Michelle L; Sofroniew, Michael V
In addition to their many functions in the healthy central nervous system (CNS), astrocytes respond to CNS damage and disease through a process called "reactivity." Recent evidence reveals that astrocyte reactivity is a heterogeneous spectrum of potential changes that occur in a context-specific manner. These changes are determined by diverse signaling events and vary not only with the nature and severity of different CNS insults but also with location in the CNS, genetic predispositions, age, and potentially also with "molecular memory" of previous reactivity events. Astrocyte reactivity can be associated with both essential beneficial functions as well as with harmful effects. The available information is rapidly expanding and much has been learned about molecular diversity of astrocyte reactivity. Emerging functional associations point toward central roles for astrocyte reactivity in determining the outcome in CNS disorders.
PMID: 38316554
ISSN: 1943-0264
CID: 5632792

Author Correction: Longitudinal scRNA-seq analysis in mouse and human informs optimization of rapid mouse astrocyte differentiation protocols

Frazel, Paul W; Labib, David; Fisher, Theodore; Brosh, Ran; Pirjanian, Nicolette; Marchildon, Anne; Boeke, Jef D; Fossati, Valentina; Liddelow, Shane A
PMID: 37996532
ISSN: 1546-1726
CID: 5608832

Longitudinal scRNA-seq analysis in mouse and human informs optimization of rapid mouse astrocyte differentiation protocols

Frazel, Paul W; Labib, David; Fisher, Theodore; Brosh, Ran; Pirjanian, Nicolette; Marchildon, Anne; Boeke, Jef D; Fossati, Valentina; Liddelow, Shane A
Macroglia (astrocytes and oligodendrocytes) are required for normal development and function of the central nervous system, yet many questions remain about their emergence during the development of the brain and spinal cord. Here we used single-cell/single-nucleus RNA sequencing (scRNA-seq/snRNA-seq) to analyze over 298,000 cells and nuclei during macroglia differentiation from mouse embryonic and human-induced pluripotent stem cells. We computationally identify candidate genes involved in the fate specification of glia in both species and report heterogeneous expression of astrocyte surface markers across differentiating cells. We then used our transcriptomic data to optimize a previous mouse astrocyte differentiation protocol, decreasing the overall protocol length and complexity. Finally, we used multi-omic, dual single-nuclei (sn)RNA-seq/snATAC-seq analysis to uncover potential genomic regulatory sites mediating glial differentiation. These datasets will enable future optimization of glial differentiation protocols and provide insight into human glial differentiation.
PMID: 37697111
ISSN: 1546-1726
CID: 5593902

Determinants of motor neuron functional subtypes important for locomotor speed

D'Elia, Kristen P; Hameedy, Hanna; Goldblatt, Dena; Frazel, Paul; Kriese, Mercer; Zhu, Yunlu; Hamling, Kyla R; Kawakami, Koichi; Liddelow, Shane A; Schoppik, David; Dasen, Jeremy S
Locomotion requires precise control of the strength and speed of muscle contraction and is achieved by recruiting functionally distinct subtypes of motor neurons (MNs). MNs are essential to movement and differentially susceptible in disease, but little is known about how MNs acquire functional subtype-specific features during development. Using single-cell RNA profiling in embryonic and larval zebrafish, we identify novel and conserved molecular signatures for MN functional subtypes and identify genes expressed in both early post-mitotic and mature MNs. Assessing MN development in genetic mutants, we define a molecular program essential for MN functional subtype specification. Two evolutionarily conserved transcription factors, Prdm16 and Mecom, are both functional subtype-specific determinants integral for fast MN development. Loss of prdm16 or mecom causes fast MNs to develop transcriptional profiles and innervation similar to slow MNs. These results reveal the molecular diversity of vertebrate axial MNs and demonstrate that functional subtypes are specified through intrinsic transcriptional codes.
PMCID:10600875
PMID: 37676768
ISSN: 2211-1247
CID: 5607632

Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration

Patani, Rickie; Hardingham, Giles E; Liddelow, Shane A
Despite advances in uncovering the mechanisms that underlie neuroinflammation and neurodegenerative disease, therapies that prevent neuronal loss remain elusive. Targeting of disease-defining markers in conditions such as Alzheimer disease (amyloid-β and tau) or Parkinson disease (α-synuclein) has been met with limited success, suggesting that these proteins do not act in isolation but form part of a pathological network. This network could involve phenotypic alteration of multiple cell types in the CNS, including astrocytes, which have a major neurosupportive, homeostatic role in the healthy CNS but adopt reactive states under acute or chronic adverse conditions. Transcriptomic studies in human patients and disease models have revealed the co-existence of many putative reactive sub-states of astrocytes. Inter-disease and even intra-disease heterogeneity of reactive astrocytic sub-states are well established, but the extent to which specific sub-states are shared across different diseases is unclear. In this Review, we highlight how single-cell and single-nuclei RNA sequencing and other 'omics' technologies can enable the functional characterization of defined reactive astrocyte states in various pathological scenarios. We provide an integrated perspective, advocating cross-modal validation of key findings to define functionally important sub-states of astrocytes and their triggers as tractable therapeutic targets with cross-disease relevance.
PMID: 37308616
ISSN: 1759-4766
CID: 5538362

Microglial INPP5D limits plaque formation and glial reactivity in the PSAPP mouse model of Alzheimer's disease

Castranio, Emilie L; Hasel, Philip; Haure-Mirande, Jean-Vianney; Ramirez Jimenez, Angie V; Hamilton, B Wade; Kim, Rachel D; Glabe, Charles G; Wang, Minghui; Zhang, Bin; Gandy, Sam; Liddelow, Shane A; Ehrlich, Michelle E
INTRODUCTION/BACKGROUND: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:mice with either tamoxifen (TAM) or corn oil (CO) to induce recombination. RESULTS: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/CONCLUSIONS: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/CONCLUSIONS: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.
PMID: 36448627
ISSN: 1552-5279
CID: 5383642