Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35.632
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 347-373, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941603

ABSTRACT

Plasmacytoid dendritic cells (pDCs) represent a unique cell type within the innate immune system. Their defining property is the recognition of pathogen-derived nucleic acids through endosomal Toll-like receptors and the ensuing production of type I interferon and other soluble mediators, which orchestrate innate and adaptive responses. We review several aspects of pDC biology that have recently come to the fore. We discuss emerging questions regarding the lineage affiliation and origin of pDCs and argue that these cells constitute an integral part of the dendritic cell lineage. We emphasize the specific function of pDCs as innate sentinels of virus infection, particularly their recognition of and distinct response to virus-infected cells. This essential evolutionary role of pDCs has been particularly important for the control of coronaviruses, as demonstrated by the recent COVID-19 pandemic. Finally, we highlight the key contribution of pDCs to systemic lupus erythematosus, in which therapeutic targeting of pDCs is currently underway.


Subject(s)
COVID-19 , Dendritic Cells , Immunity, Innate , Lupus Erythematosus, Systemic , SARS-CoV-2 , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , COVID-19/immunology , Animals , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Lupus Erythematosus, Systemic/immunology , Toll-Like Receptors/metabolism , Cell Differentiation , Cell Lineage
2.
Annu Rev Immunol ; 41: 1-15, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37126416

ABSTRACT

I have been a scientific grasshopper throughout my career, moving from question to question within the domain of lupus. This has proven to be immensely gratifying. Scientific exploration is endlessly fascinating, and succeeding in studies you care about with colleagues and trainees leads to strong and lasting bonds. Science isn't easy; being a woman in science presents challenges, but the drive to understand a disease remains strong.


Subject(s)
Career Choice , Lupus Erythematosus, Systemic , Female , Humans , Biomedical Research
3.
Annu Rev Immunol ; 35: 313-336, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28142323

ABSTRACT

Protective immune responses to viral infection are initiated by innate immune sensors that survey extracellular and intracellular space for foreign nucleic acids. The existence of these sensors raises fundamental questions about self/nonself discrimination because of the abundance of self-DNA and self-RNA that occupy these same compartments. Recent advances have revealed that enzymes that metabolize or modify endogenous nucleic acids are essential for preventing inappropriate activation of the innate antiviral response. In this review, we discuss rare human diseases caused by dysregulated nucleic acid sensing, focusing primarily on intracellular sensors of nucleic acids. We summarize lessons learned from these disorders, we rationalize the existence of these diseases in the context of evolution, and we propose that this framework may also apply to a number of more common autoimmune diseases for which the underlying genetics and mechanisms are not yet fully understood.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , Autoimmunity , Lupus Erythematosus, Systemic/immunology , Nervous System Malformations/immunology , Nucleic Acids/immunology , Virus Diseases/immunology , Animals , Humans , Immunity, Innate , Interferon Type I/metabolism , Toll-Like Receptors/metabolism
4.
Cell ; 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39013470

ABSTRACT

Allogeneic chimeric antigen receptor (CAR)-T cells hold great promise for expanding the accessibility of CAR-T therapy, whereas the risks of allograft rejection have hampered its application. Here, we genetically engineered healthy-donor-derived, CD19-targeting CAR-T cells using CRISPR-Cas9 to address the issue of immune rejection and treated one patient with refractory immune-mediated necrotizing myopathy and two patients with diffuse cutaneous systemic sclerosis with these cells. This study was registered at ClinicalTrials.gov (NCT05859997). The infused cells persisted for over 3 months, achieving complete B cell depletion within 2 weeks of treatment. During the 6-month follow-up, we observed deep remission without cytokine release syndrome or other serious adverse events in all three patients, primarily shown by the significant improvement in the clinical response index scores for the two diseases, respectively, and supported by the observations of reversal of inflammation and fibrosis. Our results demonstrate the high safety and promising immune modulatory effect of the off-the-shelf CAR-T cells in treating severe refractory autoimmune diseases.

5.
Cell ; 187(17): 4637-4655.e26, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39043180

ABSTRACT

The medical burden of stroke extends beyond the brain injury itself and is largely determined by chronic comorbidities that develop secondarily. We hypothesized that these comorbidities might share a common immunological cause, yet chronic effects post-stroke on systemic immunity are underexplored. Here, we identify myeloid innate immune memory as a cause of remote organ dysfunction after stroke. Single-cell sequencing revealed persistent pro-inflammatory changes in monocytes/macrophages in multiple organs up to 3 months after brain injury, notably in the heart, leading to cardiac fibrosis and dysfunction in both mice and stroke patients. IL-1ß was identified as a key driver of epigenetic changes in innate immune memory. These changes could be transplanted to naive mice, inducing cardiac dysfunction. By neutralizing post-stroke IL-1ß or blocking pro-inflammatory monocyte trafficking with a CCR2/5 inhibitor, we prevented post-stroke cardiac dysfunction. Such immune-targeted therapies could potentially prevent various IL-1ß-mediated comorbidities, offering a framework for secondary prevention immunotherapy.


Subject(s)
Brain Injuries , Immunity, Innate , Immunologic Memory , Inflammation , Interleukin-1beta , Mice, Inbred C57BL , Monocytes , Animals , Mice , Interleukin-1beta/metabolism , Brain Injuries/immunology , Humans , Male , Monocytes/metabolism , Monocytes/immunology , Inflammation/immunology , Macrophages/immunology , Macrophages/metabolism , Stroke/complications , Stroke/immunology , Heart Diseases/immunology , Female , Receptors, CCR2/metabolism , Fibrosis , Epigenesis, Genetic , Trained Immunity
6.
Cell ; 187(12): 3024-3038.e14, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38781969

ABSTRACT

Plants frequently encounter wounding and have evolved an extraordinary regenerative capacity to heal the wounds. However, the wound signal that triggers regenerative responses has not been identified. Here, through characterization of a tomato mutant defective in both wound-induced defense and regeneration, we demonstrate that in tomato, a plant elicitor peptide (Pep), REGENERATION FACTOR1 (REF1), acts as a systemin-independent local wound signal that primarily regulates local defense responses and regenerative responses in response to wounding. We further identified PEPR1/2 ORTHOLOG RECEPTOR-LIKE KINASE1 (PORK1) as the receptor perceiving REF1 signal for plant regeneration. REF1-PORK1-mediated signaling promotes regeneration via activating WOUND-INDUCED DEDIFFERENTIATION 1 (WIND1), a master regulator of wound-induced cellular reprogramming in plants. Thus, REF1-PORK1 signaling represents a conserved phytocytokine pathway to initiate, amplify, and stabilize a signaling cascade that orchestrates wound-triggered organ regeneration. Application of REF1 provides a simple method to boost the regeneration and transformation efficiency of recalcitrant crops.


Subject(s)
Plant Proteins , Regeneration , Signal Transduction , Solanum lycopersicum , Plant Proteins/metabolism , Plant Proteins/genetics , Solanum lycopersicum/metabolism , Gene Expression Regulation, Plant , Peptides/metabolism
7.
Cell ; 186(17): 3606-3618.e16, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37480850

ABSTRACT

Injury induces systemic responses, but their functions remain elusive. Mechanisms that can rapidly synchronize wound responses through long distances are also mostly unknown. Using planarian flatworms capable of whole-body regeneration, we report that injury induces extracellular signal-regulated kinase (Erk) activity waves to travel at a speed 10-100 times faster than those in other multicellular tissues. This ultrafast propagation requires longitudinal body-wall muscles, elongated cells forming dense parallel tracks running the length of the organism. The morphological properties of muscles allow them to act as superhighways for propagating and disseminating wound signals. Inhibiting Erk propagation prevents tissues distant to the wound from responding and blocks regeneration, which can be rescued by a second injury to distal tissues shortly after the first injury. Our findings provide a mechanism for long-range signal propagation in large, complex tissues to coordinate responses across cell types and highlight the function of feedback between spatially separated tissues during whole-body regeneration.


Subject(s)
Planarians , Regeneration , Animals , MAP Kinase Signaling System , Muscles , Phosphorylation , Planarians/physiology , Protein Processing, Post-Translational
8.
Cell ; 185(8): 1373-1388.e20, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35381199

ABSTRACT

Systemic sclerosis (scleroderma, SSc) is an incurable autoimmune disease with high morbidity and mortality rates. Here, we conducted a population-scale single-cell genomic analysis of skin and blood samples of 56 healthy controls and 97 SSc patients at different stages of the disease. We found immune compartment dysfunction only in a specific subtype of diffuse SSc patients but global dysregulation of the stromal compartment, particularly in a previously undefined subset of LGR5+-scleroderma-associated fibroblasts (ScAFs). ScAFs are perturbed morphologically and molecularly in SSc patients. Single-cell multiome profiling of stromal cells revealed ScAF-specific markers, pathways, regulatory elements, and transcription factors underlining disease development. Systematic analysis of these molecular features with clinical metadata associates specific ScAF targets with disease pathogenesis and SSc clinical traits. Our high-resolution atlas of the sclerodermatous skin spectrum will enable a paradigm shift in the understanding of SSc disease and facilitate the development of biomarkers and therapeutic strategies.


Subject(s)
Scleroderma, Systemic , Cells, Cultured , Fibroblasts/metabolism , Fibrosis , Humans , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/genetics , Skin/metabolism
9.
Cell ; 185(18): 3375-3389.e21, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35998627

ABSTRACT

Systemic lupus erythematosus (SLE) is a complex autoimmune disease involving multiple immune cells. To elucidate SLE pathogenesis, it is essential to understand the dysregulated gene expression pattern linked to various clinical statuses with a high cellular resolution. Here, we conducted a large-scale transcriptome study with 6,386 RNA sequencing data covering 27 immune cell types from 136 SLE and 89 healthy donors. We profiled two distinct cell-type-specific transcriptomic signatures: disease-state and disease-activity signatures, reflecting disease establishment and exacerbation, respectively. We then identified candidate biological processes unique to each signature. This study suggested the clinical value of disease-activity signatures, which were associated with organ involvement and therapeutic responses. However, disease-activity signatures were less enriched around SLE risk variants than disease-state signatures, suggesting that current genetic studies may not well capture clinically vital biology. Together, we identified comprehensive gene signatures of SLE, which will provide essential foundations for future genomic and genetic studies.


Subject(s)
Lupus Erythematosus, Systemic , Transcriptome , Humans , Lupus Erythematosus, Systemic/genetics , Sequence Analysis, RNA
10.
Cell ; 184(17): 4464-4479.e19, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34384544

ABSTRACT

Emerging evidence supports that mitochondrial dysfunction contributes to systemic lupus erythematosus (SLE) pathogenesis. Here we show that programmed mitochondrial removal, a hallmark of mammalian erythropoiesis, is defective in SLE. Specifically, we demonstrate that during human erythroid cell maturation, a hypoxia-inducible factor (HIF)-mediated metabolic switch is responsible for the activation of the ubiquitin-proteasome system (UPS), which precedes and is necessary for the autophagic removal of mitochondria. A defect in this pathway leads to accumulation of red blood cells (RBCs) carrying mitochondria (Mito+ RBCs) in SLE patients and in correlation with disease activity. Antibody-mediated internalization of Mito+ RBCs induces type I interferon (IFN) production through activation of cGAS in macrophages. Accordingly, SLE patients carrying both Mito+ RBCs and opsonizing antibodies display the highest levels of blood IFN-stimulated gene (ISG) signatures, a distinctive feature of SLE.


Subject(s)
Interferon Type I/metabolism , Lupus Erythematosus, Systemic/metabolism , Mitochondria/metabolism , Myeloid Cells/metabolism , Adolescent , Basic Helix-Loop-Helix Transcription Factors/metabolism , Child , Child, Preschool , Erythroblasts/metabolism , Erythroblasts/ultrastructure , Erythrocytes/metabolism , Erythropoiesis , Humans , Mitophagy , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
11.
Cell ; 184(11): 3006-3021.e17, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33930287

ABSTRACT

Genetic studies have revealed many variant loci that are associated with immune-mediated diseases. To elucidate the disease pathogenesis, it is essential to understand the function of these variants, especially under disease-associated conditions. Here, we performed a large-scale immune cell gene-expression analysis, together with whole-genome sequence analysis. Our dataset consists of 28 distinct immune cell subsets from 337 patients diagnosed with 10 categories of immune-mediated diseases and 79 healthy volunteers. Our dataset captured distinctive gene-expression profiles across immune cell types and diseases. Expression quantitative trait loci (eQTL) analysis revealed dynamic variations of eQTL effects in the context of immunological conditions, as well as cell types. These cell-type-specific and context-dependent eQTLs showed significant enrichment in immune disease-associated genetic variants, and they implicated the disease-relevant cell types, genes, and environment. This atlas deepens our understanding of the immunogenetic functions of disease-associated variants under in vivo disease conditions.


Subject(s)
Gene Expression Regulation/genetics , Gene Expression/immunology , Immune System Diseases/genetics , Adult , Female , Gene Expression/genetics , Gene Expression Regulation/immunology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Humans , Immune System/cytology , Immune System/metabolism , Immune System Diseases/metabolism , Immune System Diseases/physiopathology , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Quantitative Trait Loci/immunology , Transcriptome/genetics , Whole Genome Sequencing/methods
12.
Annu Rev Cell Dev Biol ; 38: 419-446, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36201298

ABSTRACT

The peripheral nervous system (PNS) endows animals with the remarkable ability to sense and respond to a dynamic world. Emerging evidence shows the PNS also participates in tissue homeostasis and repair by integrating local changes with organismal and environmental changes. Here, we provide an in-depth summary of findings delineating the diverse roles of peripheral nerves in modulating stem cell behaviors and immune responses under steady-state conditions and in response to injury and duress, with a specific focus on the skin and the hematopoietic system. These examples showcase how elucidating neuro-stem cell and neuro-immune cell interactions provides a conceptual framework that connects tissue biology and local immunity with systemic bodily changes to meet varying demands. They also demonstrate how changes in these interactions can manifest in stress, aging, cancer, and inflammation, as well as how these findings can be harnessed to guide the development of new therapeutics.


Subject(s)
Neurobiology , Neuroimmunomodulation , Animals , Homeostasis , Inflammation , Stem Cells
13.
Cell ; 182(5): 1093-1108.e18, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32810437

ABSTRACT

In plants, pathogen effector-triggered immunity (ETI) often leads to programmed cell death, which is restricted by NPR1, an activator of systemic acquired resistance. However, the biochemical activities of NPR1 enabling it to promote defense and restrict cell death remain unclear. Here we show that NPR1 promotes cell survival by targeting substrates for ubiquitination and degradation through formation of salicylic acid-induced NPR1 condensates (SINCs). SINCs are enriched with stress response proteins, including nucleotide-binding leucine-rich repeat immune receptors, oxidative and DNA damage response proteins, and protein quality control machineries. Transition of NPR1 into condensates is required for formation of the NPR1-Cullin 3 E3 ligase complex to ubiquitinate SINC-localized substrates, such as EDS1 and specific WRKY transcription factors, and promote cell survival during ETI. Our analysis of SINCs suggests that NPR1 is centrally integrated into the cell death or survival decisions in plant immunity by modulating multiple stress-responsive processes in this quasi-organelle.


Subject(s)
Arabidopsis Proteins/immunology , Arabidopsis Proteins/metabolism , Cell Survival/immunology , Plant Immunity/immunology , Arabidopsis/immunology , Arabidopsis/metabolism , Gene Expression Regulation, Plant/immunology , Salicylic Acid/immunology , Salicylic Acid/metabolism , Ubiquitination/immunology
14.
Annu Rev Cell Dev Biol ; 37: 311-340, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34375534

ABSTRACT

Calcium (Ca2+) is a unique mineral that serves as both a nutrient and a signal in all eukaryotes. To maintain Ca2+ homeostasis for both nutrition and signaling purposes, the tool kit for Ca2+ transport has expanded across kingdoms of eukaryotes to encode specific Ca2+ signals referred to as Ca2+ signatures. In parallel, a large array of Ca2+-binding proteins has evolved as specific sensors to decode Ca2+ signatures. By comparing these coding and decoding mechanisms in fungi, animals, and plants, both unified and divergent themes have emerged, and the underlying complexity will challenge researchers for years to come. Considering the scale and breadth of the subject, instead of a literature survey, in this review we focus on a conceptual framework that aims to introduce readers to the principles and mechanisms of Ca2+ signaling. We finish with several examples of Ca2+-signaling pathways, including polarized cell growth, immunity and symbiosis, and systemic signaling, to piece together specific coding and decoding mechanisms in plants versus animals.


Subject(s)
Calcium Signaling , Calcium , Animals , Calcium/metabolism , Homeostasis , Plants/genetics , Plants/metabolism
15.
Cell ; 177(6): 1448-1462.e14, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150621

ABSTRACT

Mammals rely on a network of circadian clocks to control daily systemic metabolism and physiology. The central pacemaker in the suprachiasmatic nucleus (SCN) is considered hierarchically dominant over peripheral clocks, whose degree of independence, or tissue-level autonomy, has never been ascertained in vivo. Using arrhythmic Bmal1-null mice, we generated animals with reconstituted circadian expression of BMAL1 exclusively in the liver (Liver-RE). High-throughput transcriptomics and metabolomics show that the liver has independent circadian functions specific for metabolic processes such as the NAD+ salvage pathway and glycogen turnover. However, although BMAL1 occupies chromatin at most genomic targets in Liver-RE mice, circadian expression is restricted to ∼10% of normally rhythmic transcripts. Finally, rhythmic clock gene expression is lost in Liver-RE mice under constant darkness. Hence, full circadian function in the liver depends on signals emanating from other clocks, and light contributes to tissue-autonomous clock function.


Subject(s)
ARNTL Transcription Factors/physiology , Circadian Clocks/genetics , Liver/metabolism , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Circadian Clocks/physiology , Circadian Rhythm/genetics , Female , Gene Expression Regulation , Homeostasis , Light , Male , Mice , Mice, Knockout , Models, Animal , Organ Specificity/physiology , Photoperiod , Suprachiasmatic Nucleus/metabolism
16.
Cell ; 177(4): 865-880.e21, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31031002

ABSTRACT

Circular RNAs (circRNAs) produced from back-splicing of exons of pre-mRNAs are widely expressed, but current understanding of their functions is limited. These RNAs are stable in general and are thought to have unique structural conformations distinct from their linear RNA cognates. Here, we show that endogenous circRNAs tend to form 16-26 bp imperfect RNA duplexes and act as inhibitors of double-stranded RNA (dsRNA)-activated protein kinase (PKR) related to innate immunity. Upon poly(I:C) stimulation or viral infection, circRNAs are globally degraded by RNase L, a process required for PKR activation in early cellular innate immune responses. Augmented PKR phosphorylation and circRNA reduction are found in peripheral blood mononuclear cells (PBMCs) derived from patients with autoimmune disease systemic lupus erythematosus (SLE). Importantly, overexpression of the dsRNA-containing circRNA in PBMCs or T cells derived from SLE can alleviate the aberrant PKR activation cascade, thus providing a connection between circRNAs and SLE.


Subject(s)
RNA, Circular/metabolism , RNA, Circular/physiology , eIF-2 Kinase/metabolism , Adolescent , Adult , Autoimmune Diseases/genetics , Cell Line , Endoribonucleases/metabolism , Female , Humans , Immunity, Innate/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/genetics , Middle Aged , Phosphorylation , RNA/metabolism , RNA Splicing/genetics , RNA Stability/physiology , RNA, Circular/genetics , RNA, Double-Stranded/metabolism , Virus Diseases/metabolism , eIF-2 Kinase/immunology
17.
Immunity ; 57(7): 1603-1617.e7, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38761804

ABSTRACT

Recent evidence reveals hyper T follicular helper (Tfh) cell responses in systemic lupus erythematosus (SLE); however, molecular mechanisms responsible for hyper Tfh cell responses and whether they cause SLE are unclear. We found that SLE patients downregulated both ubiquitin ligases, casitas B-lineage lymphoma (CBL) and CBLB (CBLs), in CD4+ T cells. T cell-specific CBLs-deficient mice developed hyper Tfh cell responses and SLE, whereas blockade of Tfh cell development in the mutant mice was sufficient to prevent SLE. ICOS was upregulated in SLE Tfh cells, whose signaling increased BCL6 by attenuating BCL6 degradation via chaperone-mediated autophagy (CMA). Conversely, CBLs restrained BCL6 expression by ubiquitinating ICOS. Blockade of BCL6 degradation was sufficient to enhance Tfh cell responses. Thus, the compromised expression of CBLs is a prevalent risk trait shared by SLE patients and causative to hyper Tfh cell responses and SLE. The ICOS-CBLs axis may be a target to treat SLE.


Subject(s)
Adaptor Proteins, Signal Transducing , Inducible T-Cell Co-Stimulator Protein , Lupus Erythematosus, Systemic , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6 , Proto-Oncogene Proteins c-cbl , T Follicular Helper Cells , Animals , Female , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Autophagy/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Inducible T-Cell Co-Stimulator Protein/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Mice, Inbred C57BL , Proteolysis , Proto-Oncogene Proteins c-bcl-6/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/deficiency , Signal Transduction/immunology , T Follicular Helper Cells/immunology , T-Lymphocytes, Helper-Inducer/immunology , Ubiquitination
18.
Cell ; 173(2): 456-469.e16, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29576453

ABSTRACT

Following a previous microbial inoculation, plants can induce broad-spectrum immunity to pathogen infection, a phenomenon known as systemic acquired resistance (SAR). SAR establishment in Arabidopsis thaliana is regulated by the Lys catabolite pipecolic acid (Pip) and flavin-dependent-monooxygenase1 (FMO1). Here, we show that elevated Pip is sufficient to induce an FMO1-dependent transcriptional reprogramming of leaves that is reminiscent of SAR. In planta and in vitro analyses demonstrate that FMO1 functions as a pipecolate N-hydroxylase, catalyzing the biochemical conversion of Pip to N-hydroxypipecolic acid (NHP). NHP systemically accumulates in plants after microbial attack. When exogenously applied, it overrides the defect of NHP-deficient fmo1 in acquired resistance and acts as a potent inducer of plant immunity to bacterial and oomycete infection. Our work has identified a pathogen-inducible L-Lys catabolic pathway in plants that generates the N-hydroxylated amino acid NHP as a critical regulator of systemic acquired resistance to pathogen infection.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Oxygenases/metabolism , Pipecolic Acids/metabolism , Plant Immunity/drug effects , Arabidopsis/enzymology , Arabidopsis/immunology , Arabidopsis Proteins/genetics , Gas Chromatography-Mass Spectrometry , Lysine/metabolism , Oomycetes/pathogenicity , Oxygenases/genetics , Pipecolic Acids/analysis , Pipecolic Acids/pharmacology , Plant Leaves/enzymology , Plant Leaves/immunology , Plant Leaves/metabolism , Pseudomonas syringae/pathogenicity , Transaminases/genetics , Transaminases/metabolism
19.
Cell ; 170(5): 913-926.e19, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28841417

ABSTRACT

Germinal centers (GCs) are the primary sites of clonal B cell expansion and affinity maturation, directing the production of high-affinity antibodies. This response is a central driver of pathogenesis in autoimmune diseases, such as systemic lupus erythematosus (SLE), but the natural history of autoreactive GCs remains unclear. Here, we present a novel mouse model where the presence of a single autoreactive B cell clone drives the TLR7-dependent activation, expansion, and differentiation of other autoreactive B cells in spontaneous GCs. Once tolerance was broken for one self-antigen, autoreactive GCs generated B cells targeting other self-antigens. GCs became independent of the initial clone and evolved toward dominance of individual clonal lineages, indicating affinity maturation. This process produced serum autoantibodies to a breadth of self-antigens, leading to antibody deposition in the kidneys. Our data provide insight into the maturation of the self-reactive B cell response, contextualizing the epitope spreading observed in autoimmune disease.


Subject(s)
B-Lymphocytes/immunology , Clonal Evolution , Germinal Center/cytology , Germinal Center/immunology , Immune Tolerance , Animals , Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases/immunology , B-Lymphocytes/cytology , Chimera/immunology , Epitopes/immunology , Kidney/immunology , Mice , Mice, Inbred C57BL
20.
Mol Cell ; 84(1): 131-141, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38103555

ABSTRACT

Nonexpressor of pathogenesis-related genes 1 (NPR1) was discovered in Arabidopsis as an activator of salicylic acid (SA)-mediated immune responses nearly 30 years ago. How NPR1 confers resistance against a variety of pathogens and stresses has been extensively studied; however, only in recent years have the underlying molecular mechanisms been uncovered, particularly NPR1's role in SA-mediated transcriptional reprogramming, stress protein homeostasis, and cell survival. Structural analyses ultimately defined NPR1 and its paralogs as SA receptors. The SA-bound NPR1 dimer induces transcription by bridging two TGA transcription factor dimers, forming an enhanceosome. Moreover, NPR1 orchestrates its multiple functions through the formation of distinct nuclear and cytoplasmic biomolecular condensates. Furthermore, NPR1 plays a central role in plant health by regulating the crosstalk between SA and other defense and growth hormones. In this review, we focus on these recent advances and discuss how NPR1 can be utilized to engineer resistance against biotic and abiotic stresses.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Salicylic Acid/chemistry , Salicylic Acid/metabolism , Salicylic Acid/pharmacology , Transcription Factors/metabolism , Stress, Physiological , Gene Expression Regulation, Plant
SELECTION OF CITATIONS
SEARCH DETAIL