Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106.978
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Annu Rev Immunol ; 40: 323-348, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35113729

RESUMO

The diverse biological activity of interleukin-6 (IL-6) contributes to the maintenance of homeostasis. Emergent infection or tissue injury induces rapid production of IL-6 and activates host defense through augmentation of acute-phase proteins and immune responses. However, excessive IL-6 production and uncontrolled IL-6 receptor signaling are critical to pathogenesis. Over the years, therapeutic agents targeting IL-6 signaling, such as tocilizumab, a humanized anti-IL-6 receptor antibody, have shown remarkable efficacy for rheumatoid arthritis, Castleman disease, and juvenile idiopathic arthritis, and their efficacy in other diseases is continually being reported. Emerging evidence has demonstrated the benefit of tocilizumab for several types of acute inflammatory diseases, including cytokine storms induced by chimeric antigen receptor T cell therapy and coronavirus disease 2019 (COVID-19). Here, we refocus attention on the biology of IL-6 and summarize the distinct pathological roles of IL-6 signaling in several acute and chronic inflammatory diseases.


Assuntos
Artrite Reumatoide , COVID-19 , Animais , Artrite Reumatoide/terapia , COVID-19/terapia , Humanos , Imunoterapia Adotiva , Interleucina-6/metabolismo , Transdução de Sinais
2.
Annu Rev Immunol ; 37: 439-456, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31026415

RESUMO

Monocytes are innate blood cells that maintain vascular homeostasis and are early responders to pathogens in acute infections. There are three well-characterized classes of monocytes: classical (CD14+CD16- in humans and Ly6Chi in mice), intermediate (CD14+CD16+ in humans and Ly6C+Treml4+ in mice), and nonclassical (CD14-CD16+ in humans and Ly6Clo in mice). Classical monocytes are critical for the initial inflammatory response. Classical monocytes can differentiate into macrophages in tissue and can contribute to chronic disease. Nonclassical monocytes have been widely viewed as anti-inflammatory, as they maintain vascular homeostasis. They are a first line of defense in recognition and clearance of pathogens. However, their roles in chronic disease are less clear. They have been shown to be protective as well as positively associated with disease burden. This review focuses on the state of the monocyte biology field and the functions of monocytes, particularly nonclassical monocytes, in health and disease.


Assuntos
Artrite Reumatoide/imunologia , Aterosclerose/imunologia , Vasos Sanguíneos/fisiologia , Monócitos/imunologia , Infarto do Miocárdio/imunologia , Animais , Autoimunidade , Hematopoese , Homeostase , Humanos , Inflamação , Camundongos
3.
Nat Immunol ; 24(7): 1200-1210, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37277655

RESUMO

Inflammation of non-barrier immunologically quiescent tissues is associated with a massive influx of blood-borne innate and adaptive immune cells. Cues from the latter are likely to alter and expand activated states of the resident cells. However, local communications between immigrant and resident cell types in human inflammatory disease remain poorly understood. Here, we explored drivers of fibroblast-like synoviocyte (FLS) heterogeneity in inflamed joints of patients with rheumatoid arthritis using paired single-cell RNA and ATAC sequencing, multiplexed imaging and spatial transcriptomics along with in vitro modeling of cell-extrinsic factor signaling. These analyses suggest that local exposures to myeloid and T cell-derived cytokines, TNF, IFN-γ, IL-1ß or lack thereof, drive four distinct FLS states some of which closely resemble fibroblast states in other disease-affected tissues including skin and colon. Our results highlight a role for concurrent, spatially distributed cytokine signaling within the inflamed synovium.


Assuntos
Artrite Reumatoide , Humanos , Células Cultivadas , Artrite Reumatoide/genética , Membrana Sinovial , Citocinas/metabolismo , Fibroblastos
4.
Nat Immunol ; 23(9): 1330-1341, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35999392

RESUMO

Fibroblasts, the most abundant structural cells, exert homeostatic functions but also drive disease pathogenesis. Single-cell technologies have illuminated the shared characteristics of pathogenic fibroblasts in multiple diseases including autoimmune arthritis, cancer and inflammatory colitis. However, the molecular mechanisms underlying the disease-associated fibroblast phenotypes remain largely unclear. Here, we identify ETS1 as the key transcription factor governing the pathological tissue-remodeling programs in fibroblasts. In arthritis, ETS1 drives polarization toward tissue-destructive fibroblasts by orchestrating hitherto undescribed regulatory elements of the osteoclast differentiation factor receptor activator of nuclear factor-κB ligand (RANKL) as well as matrix metalloproteinases. Fibroblast-specific ETS1 deletion resulted in ameliorated bone and cartilage damage under arthritic conditions without affecting the inflammation level. Cross-tissue fibroblast single-cell data analyses and genetic loss-of-function experiments lent support to the notion that ETS1 defines the perturbation-specific fibroblasts shared among various disease settings. These findings provide a mechanistic basis for pathogenic fibroblast polarization and have important therapeutic implications.


Assuntos
Artrite Reumatoide , Fibroblastos , Proteína Proto-Oncogênica c-ets-1 , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Metaloproteinases da Matriz/metabolismo , Proteína Proto-Oncogênica c-ets-1/genética , Proteína Proto-Oncogênica c-ets-1/metabolismo , Ligante RANK/genética , Fatores de Transcrição/metabolismo
5.
Nat Immunol ; 23(9): 1342-1354, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35995859

RESUMO

Appropriate regulation of B cell differentiation into plasma cells is essential for humoral immunity while preventing antibody-mediated autoimmunity; however, the underlying mechanisms, especially those with pathological consequences, remain unclear. Here, we found that the expression of Jmjd1c, a member of JmjC domain histone demethylase, in B cells but not in other immune cells, protected mice from rheumatoid arthritis (RA). In humans with RA, JMJD1C expression levels in B cells were negatively associated with plasma cell frequency and disease severity. Mechanistically, Jmjd1c demethylated STAT3, rather than histone substrate, to restrain plasma cell differentiation. STAT3 Lys140 hypermethylation caused by Jmjd1c deletion inhibited the interaction with phosphatase Ptpn6 and resulted in abnormally sustained STAT3 phosphorylation and activity, which in turn promoted plasma cell generation. Germinal center B cells devoid of Jmjd1c also acquired strikingly increased propensity to differentiate into plasma cells. STAT3 Lys140Arg point mutation completely abrogated the effect caused by Jmjd1c loss. Mice with Jmjd1c overexpression in B cells exhibited opposite phenotypes to Jmjd1c-deficient mice. Overall, our study revealed Jmjd1c as a critical regulator of plasma cell differentiation and RA and also highlighted the importance of demethylation modification for STAT3 in B cells.


Assuntos
Artrite Reumatoide , Histona Desmetilases com o Domínio Jumonji , Animais , Diferenciação Celular , Hematopoese , Histonas/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Oxirredutases N-Desmetilantes/química , Oxirredutases N-Desmetilantes/genética , Oxirredutases N-Desmetilantes/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
6.
Nat Immunol ; 22(1): 10-18, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33257900

RESUMO

The immunopathogenesis of rheumatoid arthritis (RA) spans decades, beginning with the production of autoantibodies against post-translationally modified proteins (checkpoint 1). After years of asymptomatic autoimmunity and progressive immune system remodeling, tissue tolerance erodes and joint inflammation ensues as tissue-invasive effector T cells emerge and protective joint-resident macrophages fail (checkpoint 2). The transition of synovial stromal cells into autoaggressive effector cells converts synovitis from acute to chronic destructive (checkpoint 3). The loss of T cell tolerance derives from defective DNA repair, causing abnormal cell cycle dynamics, telomere fragility and instability of mitochondrial DNA. Mitochondrial and lysosomal anomalies culminate in the generation of short-lived tissue-invasive effector T cells. This differentiation defect builds on a metabolic platform that shunts glucose away from energy generation toward the cell building and motility programs. The next frontier in RA is the development of curative interventions, for example, reprogramming T cell defects during the period of asymptomatic autoimmunity.


Assuntos
Artrite Reumatoide/imunologia , Animais , Artrite Reumatoide/etiologia , Autoimunidade , Reparo do DNA , Humanos , Inflamação/imunologia , Tolerância a Antígenos Próprios , Sinovite/imunologia , Linfócitos T/imunologia
7.
Nat Immunol ; 22(12): 1551-1562, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34811544

RESUMO

Misdirected immunity gives rise to the autoimmune tissue inflammation of rheumatoid arthritis, in which excess production of the cytokine tumor necrosis factor (TNF) is a central pathogenic event. Mechanisms underlying the breakdown of self-tolerance are unclear, but T cells in the arthritic joint have a distinctive metabolic signature of ATPlo acetyl-CoAhi proinflammatory effector cells. Here we show that a deficiency in the production of mitochondrial aspartate is an important abnormality in these autoimmune T cells. Shortage of mitochondrial aspartate disrupted the regeneration of the metabolic cofactor nicotinamide adenine dinucleotide, causing ADP deribosylation of the endoplasmic reticulum (ER) sensor GRP78/BiP. As a result, ribosome-rich ER membranes expanded, promoting co-translational translocation and enhanced biogenesis of transmembrane TNF. ERrich T cells were the predominant TNF producers in the arthritic joint. Transfer of intact mitochondria into T cells, as well as supplementation of exogenous aspartate, rescued the mitochondria-instructed expansion of ER membranes and suppressed TNF release and rheumatoid tissue inflammation.


Assuntos
Artrite Reumatoide/metabolismo , Ácido Aspártico/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Mitocôndrias/metabolismo , Membrana Sinovial/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , ADP-Ribosilação , Transferência Adotiva , Animais , Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Autoimunidade , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD4-Positivos/ultraestrutura , Estudos de Casos e Controles , Células Cultivadas , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Chaperona BiP do Retículo Endoplasmático/metabolismo , Feminino , Humanos , Masculino , Camundongos , Mitocôndrias/imunologia , Mitocôndrias/transplante , Mitocôndrias/ultraestrutura , Membrana Sinovial/imunologia , Membrana Sinovial/ultraestrutura , Fator de Necrose Tumoral alfa/genética
8.
Immunity ; 56(5): 1046-1063.e7, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-36948194

RESUMO

Immunoglobulin G (IgG) antibodies are major drivers of inflammation during infectious and autoimmune diseases. In pooled serum IgG (IVIg), however, antibodies have a potent immunomodulatory and anti-inflammatory activity, but how this is mediated is unclear. We studied IgG-dependent initiation of resolution of inflammation in cytokine- and autoantibody-driven models of rheumatoid arthritis and found IVIg sialylation inhibited joint inflammation, whereas inhibition of osteoclastogenesis was sialic acid independent. Instead, IVIg-dependent inhibition of osteoclastogenesis was abrogated in mice lacking receptors Dectin-1 or FcγRIIb. Atomistic molecular dynamics simulations and super-resolution microscopy revealed that Dectin-1 promoted FcγRIIb membrane conformations that allowed productive IgG binding and enhanced interactions with mouse and human IgG subclasses. IVIg reprogrammed monocytes via FcγRIIb-dependent signaling that required Dectin-1. Our data identify a pathogen-independent function of Dectin-1 as a co-inhibitory checkpoint for IgG-dependent inhibition of mouse and human osteoclastogenesis. These findings may have implications for therapeutic targeting of autoantibody and cytokine-driven inflammation.


Assuntos
Artrite Reumatoide , Imunoglobulinas Intravenosas , Lectinas Tipo C , Receptores de IgG , Animais , Humanos , Camundongos , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Membrana Celular/metabolismo , Imunoglobulinas Intravenosas/administração & dosagem , Lectinas Tipo C/metabolismo , Camundongos Endogâmicos C57BL , Osteoclastos/metabolismo , Processamento de Proteína Pós-Traducional , Receptores de IgG/metabolismo
9.
Nat Immunol ; 20(3): 313-325, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30718913

RESUMO

N-myristoyltransferase (NMT) attaches the fatty acid myristate to the N-terminal glycine of proteins to sort them into soluble and membrane-bound fractions. Function of the energy-sensing AMP-activated protein kinase, AMPK, is myristoylation dependent. In rheumatoid arthritis (RA), pathogenic T cells shift glucose away from adenosine tri-phosphate production toward synthetic and proliferative programs, promoting proliferation, cytokine production, and tissue invasion. We found that RA T cells had a defect in NMT1 function, which prevented AMPK activation and enabled unopposed mTORC1 signaling. Lack of the myristate lipid tail disrupted the lysosomal translocation and activation of AMPK. Instead, myristoylation-incompetent RA T cells hyperactivated the mTORC1 pathway and differentiated into pro-inflammatory TH1 and TH17 helper T cells. In vivo, NMT1 loss caused robust synovial tissue inflammation, whereas forced NMT1 overexpression rescued AMPK activation and suppressed synovitis. Thus, NMT1 has tissue-protective functions by facilitating lysosomal recruitment of AMPK and dampening mTORC1 signaling.


Assuntos
Proteínas Quinases Ativadas por AMP/imunologia , Aciltransferases/imunologia , Artrite Reumatoide/imunologia , Sinovite/imunologia , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Aciltransferases/genética , Aciltransferases/metabolismo , Adulto , Animais , Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Células Cultivadas , Ativação Enzimática/imunologia , Feminino , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Pessoa de Meia-Idade , Interferência de RNA , Sinovite/genética , Sinovite/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Adulto Jovem
10.
Nat Immunol ; 20(2): 141-151, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30643265

RESUMO

Rheumatoid arthritis is characterized by progressive joint inflammation and affects ~1% of the human population. We noted single-nucleotide polymorphisms (SNPs) in the apoptotic cell-engulfment genes ELMO1, DOCK2, and RAC1 linked to rheumatoid arthritis. As ELMO1 promotes cytoskeletal reorganization during engulfment, we hypothesized that ELMO1 loss would worsen inflammatory arthritis. Surprisingly, Elmo1-deficient mice showed reduced joint inflammation in acute and chronic arthritis models. Genetic and cell-biology studies revealed that ELMO1 associates with receptors linked to neutrophil function in arthritis and regulates activation and early neutrophil recruitment to the joints, without general inhibition of inflammatory responses. Further, neutrophils from the peripheral blood of human donors that carry the SNP in ELMO1 associated with arthritis display increased migratory capacity, whereas ELMO1 knockdown reduces human neutrophil migration to chemokines linked to arthritis. These data identify 'noncanonical' roles for ELMO1 as an important cytoplasmic regulator of specific neutrophil receptors and promoter of arthritis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Neutrófilos/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose/imunologia , Artrite Experimental/diagnóstico , Artrite Experimental/genética , Artrite Experimental/patologia , Artrite Reumatoide/diagnóstico , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Quimiotaxia/genética , Quimiotaxia/imunologia , Colágeno/imunologia , Complemento C5a/imunologia , Complemento C5a/metabolismo , Citoplasma/imunologia , Citoplasma/metabolismo , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Voluntários Saudáveis , Humanos , Microscopia Intravital , Articulações/citologia , Articulações/imunologia , Leucotrieno B4/imunologia , Leucotrieno B4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Polimorfismo de Nucleotídeo Único , Proteômica , Índice de Gravidade de Doença , Transdução de Sinais/imunologia , Imagem com Lapso de Tempo
11.
Nat Immunol ; 20(4): 458-470, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30890796

RESUMO

The cytokine IL-6 controls the survival, proliferation and effector characteristics of lymphocytes through activation of the transcription factors STAT1 and STAT3. While STAT3 activity is an ever-present feature of IL-6 signaling in CD4+ T cells, prior activation via the T cell antigen receptor limits IL-6's control of STAT1 in effector and memory populations. Here we found that phosphorylation of STAT1 in response to IL-6 was regulated by the tyrosine phosphatases PTPN2 and PTPN22 expressed in response to the activation of naïve CD4+ T cells. Transcriptomics and chromatin immunoprecipitation-sequencing (ChIP-seq) of IL-6 responses in naïve and effector memory CD4+ T cells showed how the suppression of STAT1 activation shaped the functional identity and effector characteristics of memory CD4+ T cells. Thus, tyrosine phosphatases induced by the activation of naïve T cells determine the way activated or memory CD4+ T cells sense and interpret cytokine signals.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Animais , Artrite Reumatoide/enzimologia , Artrite Reumatoide/patologia , Linfócitos T CD4-Positivos/enzimologia , Células CHO , Células Cultivadas , Cricetulus , Regulação da Expressão Gênica , Humanos , Memória Imunológica , Interleucina-6/fisiologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Interleucina-6/fisiologia , Membrana Sinovial/imunologia , Transcrição Gênica
12.
Nat Immunol ; 20(7): 928-942, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31061532

RESUMO

To define the cell populations that drive joint inflammation in rheumatoid arthritis (RA), we applied single-cell RNA sequencing (scRNA-seq), mass cytometry, bulk RNA sequencing (RNA-seq) and flow cytometry to T cells, B cells, monocytes, and fibroblasts from 51 samples of synovial tissue from patients with RA or osteoarthritis (OA). Utilizing an integrated strategy based on canonical correlation analysis of 5,265 scRNA-seq profiles, we identified 18 unique cell populations. Combining mass cytometry and transcriptomics revealed cell states expanded in RA synovia: THY1(CD90)+HLA-DRAhi sublining fibroblasts, IL1B+ pro-inflammatory monocytes, ITGAX+TBX21+ autoimmune-associated B cells and PDCD1+ peripheral helper T (TPH) cells and follicular helper T (TFH) cells. We defined distinct subsets of CD8+ T cells characterized by GZMK+, GZMB+, and GNLY+ phenotypes. We mapped inflammatory mediators to their source cell populations; for example, we attributed IL6 expression to THY1+HLA-DRAhi fibroblasts and IL1B production to pro-inflammatory monocytes. These populations are potentially key mediators of RA pathogenesis.


Assuntos
Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Perfilação da Expressão Gênica , Membrana Sinovial/metabolismo , Transcriptoma , Artrite Reumatoide/patologia , Autoimunidade/genética , Biomarcadores , Biologia Computacional/métodos , Estudos Transversais , Citocinas/metabolismo , Fibroblastos/metabolismo , Citometria de Fluxo , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Leucócitos/imunologia , Leucócitos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Transdução de Sinais , Análise de Célula Única/métodos , Membrana Sinovial/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Fluxo de Trabalho
13.
Nat Immunol ; 20(12): 1631-1643, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31740799

RESUMO

Osteoclasts have a unique bone-destroying capacity, playing key roles in steady-state bone remodeling and arthritic bone erosion. Whether the osteoclasts in these different tissue settings arise from the same precursor states of monocytoid cells is presently unknown. Here, we show that osteoclasts in pannus originate exclusively from circulating bone marrow-derived cells and not from locally resident macrophages. We identify murine CX3CR1hiLy6CintF4/80+I-A+/I-E+ macrophages (termed here arthritis-associated osteoclastogenic macrophages (AtoMs)) as the osteoclast precursor-containing population in the inflamed synovium, comprising a subset distinct from conventional osteoclast precursors in homeostatic bone remodeling. Tamoxifen-inducible Foxm1 deletion suppressed the capacity of AtoMs to differentiate into osteoclasts in vitro and in vivo. Furthermore, synovial samples from human patients with rheumatoid arthritis contained CX3CR1+HLA-DRhiCD11c+CD80-CD86+ cells that corresponded to mouse AtoMs, and human osteoclastogenesis was inhibited by the FoxM1 inhibitor thiostrepton, constituting a potential target for rheumatoid arthritis treatment.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Células da Medula Óssea/fisiologia , Proteína Forkhead Box M1/metabolismo , Macrófagos/fisiologia , Osteoclastos/fisiologia , Animais , Receptor 1 de Quimiocina CX3C/metabolismo , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Proteína Forkhead Box M1/antagonistas & inibidores , Proteína Forkhead Box M1/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Osteogênese , Tioestreptona/farmacologia
14.
Immunity ; 55(12): 2255-2270, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36516818

RESUMO

Significant recent progress in understanding rheumatoid arthritis (RA) pathogenesis has led to improved treatment and quality of life. The introduction of targeted-biologic and -synthetic disease modifying anti-rheumatic drugs (DMARDs) has also transformed clinical outcomes. Despite this, RA remains a life-long disease without a cure. Unmet needs include partial response and non-response to treatment in many patients, failure to achieve immune homeostasis or drug free remission, and inability to repair damaged tissues. RA is now recognized as the end of a multi-year prodromal phase in which systemic immune dysregulation, likely beginning in mucosal surfaces, is followed by a symptomatic clinical phase. Inflammation and immune reactivity are primarily localized to the synovium leading to pain and articular damage, but is also associated with a broader series of comorbidities. Here, we review recently described immunologic mechanisms that drive breach of tolerance, chronic synovitis, and remission.


Assuntos
Antirreumáticos , Artrite Reumatoide , Sinovite , Humanos , Qualidade de Vida , Artrite Reumatoide/tratamento farmacológico , Antirreumáticos/uso terapêutico , Membrana Sinovial
15.
Nat Immunol ; 24(3): 377, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36829072
16.
Nat Immunol ; 19(9): 942-953, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30111894

RESUMO

The sensing of microbial genetic material by leukocytes often elicits beneficial pro-inflammatory cytokines, but dysregulated responses can cause severe pathogenesis. Genome-wide association studies have linked the gene encoding phospholipase D3 (PLD3) to Alzheimer's disease and have linked PLD4 to rheumatoid arthritis and systemic sclerosis. PLD3 and PLD4 are endolysosomal proteins whose functions are obscure. Here, PLD4-deficient mice were found to have an inflammatory disease, marked by elevated levels of interferon-γ (IFN-γ) and splenomegaly. These phenotypes were traced to altered responsiveness of PLD4-deficient dendritic cells to ligands of the single-stranded DNA sensor TLR9. Macrophages from PLD3-deficient mice also had exaggerated TLR9 responses. Although PLD4 and PLD3 were presumed to be phospholipases, we found that they are 5' exonucleases, probably identical to spleen phosphodiesterase, that break down TLR9 ligands. Mice deficient in both PLD3 and PLD4 developed lethal liver inflammation in early life, which indicates that both enzymes are needed to regulate inflammatory cytokine responses via the degradation of nucleic acids.


Assuntos
Células Dendríticas/fisiologia , Endossomos/metabolismo , Exonucleases/metabolismo , Hepatite/genética , Macrófagos/fisiologia , Glicoproteínas de Membrana/metabolismo , Fosfolipase D/metabolismo , Doença de Alzheimer/genética , Animais , Artrite Reumatoide/genética , DNA de Cadeia Simples/imunologia , Exonucleases/genética , Estudo de Associação Genômica Ampla , Humanos , Interferon gama/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfolipase D/genética , Escleroderma Sistêmico/genética , Transdução de Sinais , Receptor Toll-Like 9/metabolismo
17.
Immunity ; 54(5): 1002-1021.e10, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33761330

RESUMO

Arthritis typically involves recurrence and progressive worsening at specific predilection sites, but the checkpoints between remission and persistence remain unknown. Here, we defined the molecular and cellular mechanisms of this inflammation-mediated tissue priming. Re-exposure to inflammatory stimuli caused aggravated arthritis in rodent models. Tissue priming developed locally and independently of adaptive immunity. Repeatedly stimulated primed synovial fibroblasts (SFs) exhibited enhanced metabolic activity inducing functional changes with intensified migration, invasiveness and osteoclastogenesis. Meanwhile, human SF from patients with established arthritis displayed a similar primed phenotype. Transcriptomic and epigenomic analyses as well as genetic and pharmacological targeting demonstrated that inflammatory tissue priming relies on intracellular complement C3- and C3a receptor-activation and downstream mammalian target of rapamycin- and hypoxia-inducible factor 1α-mediated metabolic SF invigoration that prevents activation-induced senescence, enhances NLRP3 inflammasome activity, and in consequence sensitizes tissue for inflammation. Our study suggests possibilities for therapeutic intervention abrogating tissue priming without immunosuppression.


Assuntos
Proteínas do Sistema Complemento/imunologia , Fibroblastos/imunologia , Inflamação/imunologia , Membrana Sinovial/imunologia , Imunidade Adaptativa/imunologia , Animais , Artrite Reumatoide/imunologia , Linhagem Celular , Cães , Humanos , Mediadores da Inflamação/imunologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Ratos Wistar , Transdução de Sinais/imunologia
18.
Immunity ; 54(8): 1883-1900.e5, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34331874

RESUMO

Mononuclear phagocytes (MNPs) encompass dendritic cells, monocytes, and macrophages (MoMac), which exhibit antimicrobial, homeostatic, and immunoregulatory functions. We integrated 178,651 MNPs from 13 tissues across 41 datasets to generate a MNP single-cell RNA compendium (MNP-VERSE), a publicly available tool to map MNPs and define conserved gene signatures of MNP populations. Next, we generated a MoMac-focused compendium that revealed an array of specialized cell subsets widely distributed across multiple tissues. Specific pathological forms were expanded in cancer and inflammation. All neoplastic tissues contained conserved tumor-associated macrophage populations. In particular, we focused on IL4I1+CD274(PD-L1)+IDO1+ macrophages, which accumulated in the tumor periphery in a T cell-dependent manner via interferon-γ (IFN-γ) and CD40/CD40L-induced maturation from IFN-primed monocytes. IL4I1_Macs exhibited immunosuppressive characteristics through tryptophan degradation and promoted the entry of regulatory T cell into tumors. This integrated analysis provides a robust online-available platform for uniform annotation and dissection of specific macrophage functions in healthy and pathological states.


Assuntos
Células Dendríticas/imunologia , Expressão Gênica/imunologia , Monócitos/imunologia , Transcriptoma/genética , Macrófagos Associados a Tumor/imunologia , Artrite Reumatoide/imunologia , COVID-19/imunologia , Expressão Gênica/genética , Perfilação da Expressão Gênica , Humanos , Interferon gama/imunologia , L-Aminoácido Oxidase/metabolismo , Cirrose Hepática/imunologia , Macrófagos/imunologia , Neoplasias/imunologia , RNA Citoplasmático Pequeno/genética , Análise de Célula Única , Linfócitos T Reguladores/imunologia , Transcriptoma/imunologia
19.
Nature ; 633(8029): 442-450, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39143217

RESUMO

Regulation of neutrophil activation is critical for disease control. Neutrophil extracellular traps (NETs), which are web-like structures composed of DNA and neutrophil-derived proteins, are formed following pro-inflammatory signals; however, if this process is uncontrolled, NETs contribute to disease pathogenesis, exacerbating inflammation and host tissue damage1,2. Here we show that myeloid inhibitory C-type lectin-like (MICL), an inhibitory C-type lectin receptor, directly recognizes DNA in NETs; this interaction is vital to regulate neutrophil activation. Loss or inhibition of MICL functionality leads to uncontrolled NET formation through the ROS-PAD4 pathway and the development of an auto-inflammatory feedback loop. We show that in the context of rheumatoid arthritis, such dysregulation leads to exacerbated pathology in both mouse models and in human patients, where autoantibodies to MICL inhibit key functions of this receptor. Of note, we also detect similarly inhibitory anti-MICL autoantibodies in patients with other diseases linked to aberrant NET formation, including lupus and severe COVID-19. By contrast, dysregulation of NET release is protective during systemic infection with the fungal pathogen Aspergillus fumigatus. Together, we show that the recognition of NETs by MICL represents a fundamental autoregulatory pathway that controls neutrophil activity and NET formation.


Assuntos
Artrite Reumatoide , Armadilhas Extracelulares , Ativação de Neutrófilo , Neutrófilos , Animais , Feminino , Humanos , Masculino , Camundongos , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Artrite Reumatoide/metabolismo , Aspergillus fumigatus/imunologia , Aspergillus fumigatus/patogenicidade , Autoanticorpos/imunologia , Autoanticorpos/farmacologia , COVID-19/imunologia , COVID-19/virologia , Modelos Animais de Doenças , DNA/metabolismo , DNA/imunologia , Armadilhas Extracelulares/metabolismo , Armadilhas Extracelulares/imunologia , Retroalimentação Fisiológica , Inflamação/imunologia , Inflamação/metabolismo , Lectinas Tipo C/antagonistas & inibidores , Lectinas Tipo C/deficiência , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Neutrófilos/metabolismo , Proteína-Arginina Desiminase do Tipo 4/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Mitogênicos/antagonistas & inibidores , Receptores Mitogênicos/deficiência , Receptores Mitogênicos/imunologia , Receptores Mitogênicos/metabolismo
20.
Nat Immunol ; 18(6): 612-621, 2017 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-28518156

RESUMO

Increased understanding of the biology of interleukin 17 (IL-17) has revealed that this cytokine is a central player in immunity at the sites most exposed to microorganisms. Although it has been strongly associated with immunopathology, IL-17 also has an important role in host defense. The regulation of IL-17 secretion seems to be shared among various cell types, each of which can concomitantly secrete additional products. IL-17 has only modest activity on its own; its impact in immunity arises from its synergistic action with other factors, its self-sustaining feedback loop and, in some cases, its role as a counterpart of interferon-γ (IFN-γ). Together these attributes provide a robust response against microorganisms, but they can equally contribute to immune pathology. Here we focus on a discussion of the role of IL-17 during infection.


Assuntos
Imunidade Adaptativa/imunologia , Artrite Reumatoide/imunologia , Imunidade Inata/imunologia , Infecções/imunologia , Interleucina-17/imunologia , Neoplasias/imunologia , Psoríase/imunologia , Animais , Retroalimentação , Humanos , Interferon gama/imunologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA