Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722686

ABSTRACT

Group 3 innate lymphoid cells (ILC3s) are key players in intestinal homeostasis. Endoplasmic reticulum (ER) stress is linked to inflammatory bowel disease (IBD). Herein, we used cell culture, mouse models, and human specimens to examine if ER stress in ILC3s impacts IBD pathophysiology. We show that mouse intestinal ILC3s exhibited a 24h-rhythmic expression pattern of the master ER stress response regulator, IRE1α/XBP1. Proinflammatory cytokine IL-23 selectively stimulated IRE1α/XBP1 in mouse ILC3s through mitochondrial reactive oxygen species (mtROS). IRE1α/XBP1 was activated in ILC3s of mice exposed to experimental colitis and in inflamed human IBD specimens. Mice with Ire1α deletion in ILC3s (Ire1αΔRorc) showed reduced expression of ER stress response and cytokine genes including Il22 in ILC3s and were highly vulnerable to infections and colitis. Administration of IL-22 counteracted their colitis susceptibility. In human ILC3s, IRE1 inhibitors suppressed cytokine production, which was upregulated by an IRE1 activator. Moreover, the frequencies of intestinal XBP1s+ ILC3s in Crohn's disease patients before administration of ustekinumab, an anti-IL-12/IL-23 antibody, positively correlated with response to treatment. We demonstrate that a non-canonical mtROS-IRE1α/XBP1 pathway augments cytokine production by ILC3s and identify XBP1s+ ILC3s as a potential biomarker for predicting response to anti-IL-23 therapies in IBD.

2.
Sci Transl Med ; 16(741): eadj9052, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38569016

ABSTRACT

Microglia help limit the progression of Alzheimer's disease (AD) by constraining amyloid-ß (Aß) pathology, effected through a balance of activating and inhibitory intracellular signals delivered by distinct cell surface receptors. Human leukocyte Ig-like receptor B4 (LILRB4) is an inhibitory receptor of the immunoglobulin (Ig) superfamily that is expressed on myeloid cells and recognizes apolipoprotein E (ApoE) among other ligands. Here, we find that LILRB4 is highly expressed in the microglia of patients with AD. Using mice that accumulate Aß and carry a transgene encompassing a portion of the LILR region that includes LILRB4, we corroborated abundant LILRB4 expression in microglia wrapping around Aß plaques. Systemic treatment of these mice with an anti-human LILRB4 monoclonal antibody (mAb) reduced Aß load, mitigated some Aß-related behavioral abnormalities, enhanced microglia activity, and attenuated expression of interferon-induced genes. In vitro binding experiments established that human LILRB4 binds both human and mouse ApoE and that anti-human LILRB4 mAb blocks such interaction. In silico modeling, biochemical, and mutagenesis analyses identified a loop between the two extracellular Ig domains of LILRB4 required for interaction with mouse ApoE and further indicated that anti-LILRB4 mAb may block LILRB4-mApoE by directly binding this loop. Thus, targeting LILRB4 may be a potential therapeutic avenue for AD.


Subject(s)
Alzheimer Disease , Microglia , Humans , Mice , Animals , Microglia/metabolism , Antibodies/metabolism , Receptors, Cell Surface/metabolism , Amyloid/metabolism , Disease Models, Animal , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Apolipoproteins E , Leukocytes/metabolism , Mice, Transgenic , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism
3.
Cell Rep ; 42(4): 112293, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36952346

ABSTRACT

Demyelination is a hallmark of multiple sclerosis, leukoencephalopathies, cerebral vasculopathies, and several neurodegenerative diseases. The cuprizone mouse model is widely used to simulate demyelination and remyelination occurring in these diseases. Here, we present a high-resolution single-nucleus RNA sequencing (snRNA-seq) analysis of gene expression changes across all brain cells in this model. We define demyelination-associated oligodendrocytes (DOLs) and remyelination-associated MAFBhi microglia, as well as astrocytes and vascular cells with signatures of altered metabolism, oxidative stress, and interferon response. Furthermore, snRNA-seq provides insights into how brain cell types connect and interact, defining complex circuitries that impact demyelination and remyelination. As an explicative example, perturbation of microglia caused by TREM2 deficiency indirectly impairs the induction of DOLs. Altogether, this study provides a rich resource for future studies investigating mechanisms underlying demyelinating diseases.


Subject(s)
Demyelinating Diseases , Remyelination , Animals , Mice , Demyelinating Diseases/metabolism , Transcriptome/genetics , Brain/metabolism , Oligodendroglia/metabolism , Microglia/metabolism , Cuprizone/toxicity , Disease Models, Animal , Mice, Inbred C57BL , Myelin Sheath/metabolism
5.
Nat Immunol ; 24(3): 545-557, 2023 03.
Article in English | MEDLINE | ID: mdl-36658241

ABSTRACT

The TREM2-DAP12 receptor complex sustains microglia functions. Heterozygous hypofunctional TREM2 variants impair microglia, accelerating late-onset Alzheimer's disease. Homozygous inactivating variants of TREM2 or TYROBP-encoding DAP12 cause Nasu-Hakola disease (NHD), an early-onset dementia characterized by cerebral atrophy, myelin loss and gliosis. Mechanisms underpinning NHD are unknown. Here, single-nucleus RNA-sequencing analysis of brain specimens from DAP12-deficient NHD individuals revealed a unique microglia signature indicating heightened RUNX1, STAT3 and transforming growth factor-ß signaling pathways that mediate repair responses to injuries. This profile correlated with a wound healing signature in astrocytes and impaired myelination in oligodendrocytes, while pericyte profiles indicated vascular abnormalities. Conversely, single-nuclei signatures in mice lacking DAP12 signaling reflected very mild microglial defects that did not recapitulate NHD. We envision that DAP12 signaling in microglia attenuates wound healing pathways that, if left unchecked, interfere with microglial physiological functions, causing pathology in human. The identification of a dysregulated NHD microglia signature sparks potential therapeutic strategies aimed at resetting microglia signaling pathways.


Subject(s)
Dementia , Subacute Sclerosing Panencephalitis , Animals , Humans , Mice , Brain/metabolism , Dementia/metabolism , Dementia/pathology , Membrane Glycoproteins/metabolism , Microglia/metabolism , Receptors, Immunologic/metabolism , Subacute Sclerosing Panencephalitis/metabolism , Subacute Sclerosing Panencephalitis/pathology
6.
Cell ; 185(22): 4153-4169.e19, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36306735

ABSTRACT

Genetic studies have highlighted microglia as pivotal in orchestrating Alzheimer's disease (AD). Microglia that adhere to Aß plaques acquire a transcriptional signature, "disease-associated microglia" (DAM), which largely emanates from the TREM2-DAP12 receptor complex that transmits intracellular signals through the protein tyrosine kinase SYK. The human TREM2R47H variant associated with high AD risk fails to activate microglia via SYK. We found that SYK-deficient microglia cannot encase Aß plaques, accelerating brain pathology and behavioral deficits. SYK deficiency impaired the PI3K-AKT-GSK-3ß-mTOR pathway, incapacitating anabolic support required for attaining the DAM profile. However, SYK-deficient microglia proliferated and advanced to an Apoe-expressing prodromal stage of DAM; this pathway relied on the adapter DAP10, which also binds TREM2. Thus, microglial responses to Aß involve non-redundant SYK- and DAP10-pathways. Systemic administration of an antibody against CLEC7A, a receptor that directly activates SYK, rescued microglia activation in mice expressing the TREM2R47H allele, unveiling new options for AD immunotherapy.


Subject(s)
Alzheimer Disease , Microglia , Animals , Mice , Humans , Microglia/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/pathology , Plaque, Amyloid/metabolism , Brain/metabolism , Disease Models, Animal , Syk Kinase/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism
7.
Proc Natl Acad Sci U S A ; 119(17): e2106083119, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35446623

ABSTRACT

CD8 T cells mediate protection against intracellular pathogens and tumors. However, persistent antigen during chronic infections or cancer leads to T cell exhaustion, suboptimal functionality, and reduced protective capacity. Despite considerable work interrogating the transcriptional regulation of exhausted CD8 T cells (TEX), the posttranscriptional control of TEX remains poorly understood. Here, we interrogated the role of microRNAs (miRs) in CD8 T cells responding to acutely resolved or chronic viral infection and identified miR-29a as a key regulator of TEX. Enforced expression of miR-29a improved CD8 T cell responses during chronic viral infection and antagonized exhaustion. miR-29a inhibited exhaustion-driving transcriptional pathways, including inflammatory and T cell receptor signaling, and regulated ribosomal biogenesis. As a result, miR-29a fostered a memory-like CD8 T cell differentiation state during chronic infection. Thus, we identify miR-29a as a key regulator of TEX and define mechanisms by which miR-29a can divert exhaustion toward a more beneficial memory-like CD8 T cell differentiation state.


Subject(s)
MicroRNAs , Neoplasms , CD8-Positive T-Lymphocytes , Humans , Immunotherapy/methods , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/metabolism , Persistent Infection
8.
Sci Immunol ; 6(58)2021 04 23.
Article in English | MEDLINE | ID: mdl-33893172

ABSTRACT

Human cytomegalovirus (CMV) infection can stimulate robust human leukocyte antigen (HLA)-E-restricted CD8+ T cell responses. These T cells recognize a peptide from UL40, which differs by as little as a single methyl group from self-peptides that also bind HLA-E, challenging their capacity to avoid self-reactivity. Unexpectedly, we showed that the UL40/HLA-E T cell receptor (TCR) repertoire included TCRs that had high affinities for HLA-E/self-peptide. However, paradoxically, lower cytokine responses were observed from UL40/HLA-E T cells bearing TCRs with high affinity for HLA-E. RNA sequencing and flow cytometric analysis revealed that these T cells were marked by the expression of inhibitory natural killer cell receptors (NKRs) KIR2DL1 and KIR2DL2/L3. On the other hand, UL40/HLA-E T cells bearing lower-affinity TCRs expressed the activating receptor NKG2C. Activation of T cells bearing higher-affinity TCRs was regulated by the interaction between KIR2D receptors and HLA-C. These findings identify a role for NKR signaling in regulating self/non-self discrimination by HLA-E-restricted T cells, allowing for antiviral responses while avoiding contemporaneous self-reactivity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , Receptors, Natural Killer Cell/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cytomegalovirus/immunology , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/virology , Humans , Killer Cells, Natural/metabolism , Receptors, Antigen, T-Cell/metabolism , Viral Proteins/immunology , Viral Proteins/metabolism , HLA-E Antigens
9.
Cell ; 184(5): 1262-1280.e22, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33636129

ABSTRACT

Improving effector activity of antigen-specific T cells is a major goal in cancer immunotherapy. Despite the identification of several effector T cell (TEFF)-driving transcription factors (TFs), the transcriptional coordination of TEFF biology remains poorly understood. We developed an in vivo T cell CRISPR screening platform and identified a key mechanism restraining TEFF biology through the ETS family TF, Fli1. Genetic deletion of Fli1 enhanced TEFF responses without compromising memory or exhaustion precursors. Fli1 restrained TEFF lineage differentiation by binding to cis-regulatory elements of effector-associated genes. Loss of Fli1 increased chromatin accessibility at ETS:RUNX motifs, allowing more efficient Runx3-driven TEFF biology. CD8+ T cells lacking Fli1 provided substantially better protection against multiple infections and tumors. These data indicate that Fli1 safeguards the developing CD8+ T cell transcriptional landscape from excessive ETS:RUNX-driven TEFF cell differentiation. Moreover, genetic deletion of Fli1 improves TEFF differentiation and protective immunity in infections and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Proto-Oncogene Protein c-fli-1/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CRISPR-Cas Systems , Cell Differentiation , Chronic Disease , Core Binding Factor Alpha 3 Subunit/metabolism , Epigenesis, Genetic , Gene Regulatory Networks , Infections/immunology , Mice , Neoplasms/immunology
10.
Sci Immunol ; 6(55)2021 01 15.
Article in English | MEDLINE | ID: mdl-33452106

ABSTRACT

The developmental origins of memory T cells remain incompletely understood. During the expansion phase of acute viral infection, we identified a distinct subset of virus-specific CD8+ T cells that possessed distinct characteristics including expression of CD62L, T cell factor 1 (TCF-1), and Eomesodermin; relative quiescence; expression of activation markers; and features of limited effector differentiation. These cells were a quantitatively minor subpopulation of the TCF-1+ pool and exhibited self-renewal, heightened DNA damage surveillance activity, and preferential long-term recall capacity. Despite features of memory and somewhat restrained proliferation during the expansion phase, this subset displayed evidence of stronger TCR signaling than other responding CD8+ T cells, coupled with elevated expression of multiple inhibitory receptors including programmed cell death 1 (PD-1), lymphocyte activating gene 3 (LAG-3), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), CD5, and CD160. Genetic ablation of PD-1 and LAG-3 compromised the formation of this CD62Lhi TCF-1+ subset and subsequent CD8+ T cell memory. Although central memory phenotype CD8+ T cells were formed in the absence of these cells, subsequent memory CD8+ T cell recall responses were compromised. Together, these results identify an important link between genome integrity maintenance and CD8+ T cell memory. Moreover, the data indicate a role for inhibitory receptors in preserving key memory CD8+ T cell precursors during initial activation and differentiation. Identification of this rare subpopulation within the memory CD8+ T cell precursor pool may help reconcile models of the developmental origin of long-term CD8+ T cell memory.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Listeriosis/immunology , Lymphocytic Choriomeningitis/immunology , Memory T Cells/immunology , Precursor Cells, T-Lymphoid/immunology , Animals , Antigens, CD/genetics , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , DNA Damage/immunology , Disease Models, Animal , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Immunologic Memory/genetics , Listeria monocytogenes/immunology , Listeriosis/microbiology , Lymphocyte Activation , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Male , Memory T Cells/metabolism , Mice , Mice, Knockout , Precursor Cells, T-Lymphoid/metabolism , Programmed Cell Death 1 Receptor/genetics , Lymphocyte Activation Gene 3 Protein
11.
Immunity ; 51(5): 840-855.e5, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31606264

ABSTRACT

TCF-1 is a key transcription factor in progenitor exhausted CD8 T cells (Tex). Moreover, this Tex cell subset mediates responses to PD-1 checkpoint pathway blockade. However, the role of the transcription factor TCF-1 in early fate decisions and initial generation of Tex cells is unclear. Single-cell RNA sequencing (scRNA-seq) and lineage tracing identified a TCF-1+Ly108+PD-1+ CD8 T cell population that seeds development of mature Tex cells early during chronic infection. TCF-1 mediated the bifurcation between divergent fates, repressing development of terminal KLRG1Hi effectors while fostering KLRG1Lo Tex precursor cells, and PD-1 stabilized this TCF-1+ Tex precursor cell pool. TCF-1 mediated a T-bet-to-Eomes transcription factor transition in Tex precursors by promoting Eomes expression and drove c-Myb expression that controlled Bcl-2 and survival. These data define a role for TCF-1 in early-fate-bifurcation-driving Tex precursor cells and also identify PD-1 as a protector of this early TCF-1 subset.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Gene Regulatory Networks , T Cell Transcription Factor 1/metabolism , Transcription, Genetic , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Chronic Disease , Gene Expression Profiling , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Mice , Programmed Cell Death 1 Receptor/metabolism , T Cell Transcription Factor 1/genetics , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/virology
12.
Cell Rep ; 23(7): 2142-2156, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768211

ABSTRACT

Persistent viral infections and tumors drive development of exhausted T (TEX) cells. In these settings, TEX cells establish an important host-pathogen or host-tumor stalemate. However, TEX cells erode over time, leading to loss of pathogen or cancer containment. We identified microRNA (miR)-155 as a key regulator of sustained TEX cell responses during chronic lymphocytic choriomeningitis virus (LCMV) infection. Genetic deficiency of miR-155 ablated CD8 T cell responses during chronic infection. Conversely, enhanced miR-155 expression promoted expansion and long-term persistence of TEX cells. However, rather than strictly antagonizing exhaustion, miR-155 promoted a terminal TEX cell subset. Transcriptional profiling identified coordinated control of cell signaling and transcription factor pathways, including the key AP-1 family member Fosl2. Overexpression of Fosl2 reversed the miR-155 effects, identifying a link between miR-155 and the AP-1 transcriptional program in regulating TEX cells. Thus, we identify a mechanism of miR-155 regulation of TEX cells and a key role for Fosl2 in T cell exhaustion.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Communicable Diseases/genetics , Communicable Diseases/immunology , MicroRNAs/metabolism , Animals , Cell Differentiation , Cell Proliferation/genetics , Chronic Disease , Communicable Diseases/pathology , Fos-Related Antigen-2/metabolism , Gene Expression Regulation , Lymphocyte Subsets/immunology , Mice, Inbred C57BL , MicroRNAs/genetics , Phenotype , Time Factors , Transcription Factor AP-1/metabolism , Transcription, Genetic
13.
Cell Rep ; 20(11): 2584-2597, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28903040

ABSTRACT

MicroRNAs play an important role in T cell responses. However, how microRNAs regulate CD8 T cell memory remains poorly defined. Here, we found that miR-150 negatively regulates CD8 T cell memory in vivo. Genetic deletion of miR-150 disrupted the balance between memory precursor and terminal effector CD8 T cells following acute viral infection. Moreover, miR-150-deficient memory CD8 T cells were more protective upon rechallenge. A key circuit whereby miR-150 repressed memory CD8 T cell development through the transcription factor c-Myb was identified. Without miR-150, c-Myb was upregulated and anti-apoptotic targets of c-Myb, such as Bcl-2 and Bcl-xL, were also increased, suggesting a miR-150-c-Myb survival circuit during memory CD8 T cell development. Indeed, overexpression of non-repressible c-Myb rescued the memory CD8 T cell defects caused by overexpression of miR-150. Overall, these results identify a key role for miR-150 in memory CD8 T cells through a c-Myb-controlled enhanced survival circuit.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Immunologic Memory , MicroRNAs/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Animals , Apoptosis/genetics , Female , Immunity , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...