Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.659
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 40: 323-348, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35113729

ABSTRACT

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.


Subject(s)
Arthritis, Rheumatoid , COVID-19 , Animals , Arthritis, Rheumatoid/therapy , COVID-19/therapy , Humans , Immunotherapy, Adoptive , Interleukin-6/metabolism , Signal Transduction
2.
Cell ; 186(2): 382-397.e24, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36669473

ABSTRACT

Blood and lymphatic vessels form a versatile transport network and provide inductive signals to regulate tissue-specific functions. Blood vessels in bone regulate osteogenesis and hematopoiesis, but current dogma suggests that bone lacks lymphatic vessels. Here, by combining high-resolution light-sheet imaging and cell-specific mouse genetics, we demonstrate presence of lymphatic vessels in mouse and human bones. We find that lymphatic vessels in bone expand during genotoxic stress. VEGF-C/VEGFR-3 signaling and genotoxic stress-induced IL6 drive lymphangiogenesis in bones. During lymphangiogenesis, secretion of CXCL12 from proliferating lymphatic endothelial cells is critical for hematopoietic and bone regeneration. Moreover, lymphangiocrine CXCL12 triggers expansion of mature Myh11+ CXCR4+ pericytes, which differentiate into bone cells and contribute to bone and hematopoietic regeneration. In aged animals, such expansion of lymphatic vessels and Myh11-positive cells in response to genotoxic stress is impaired. These data suggest lymphangiogenesis as a therapeutic avenue to stimulate hematopoietic and bone regeneration.


Subject(s)
Bone Regeneration , Lymphatic Vessels , Aged , Animals , Humans , Mice , Endothelial Cells , Lymphangiogenesis
3.
Cell ; 186(18): 3882-3902.e24, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37597510

ABSTRACT

Inflammation can trigger lasting phenotypes in immune and non-immune cells. Whether and how human infections and associated inflammation can form innate immune memory in hematopoietic stem and progenitor cells (HSPC) has remained unclear. We found that circulating HSPC, enriched from peripheral blood, captured the diversity of bone marrow HSPC, enabling investigation of their epigenomic reprogramming following coronavirus disease 2019 (COVID-19). Alterations in innate immune phenotypes and epigenetic programs of HSPC persisted for months to 1 year following severe COVID-19 and were associated with distinct transcription factor (TF) activities, altered regulation of inflammatory programs, and durable increases in myelopoiesis. HSPC epigenomic alterations were conveyed, through differentiation, to progeny innate immune cells. Early activity of IL-6 contributed to these persistent phenotypes in human COVID-19 and a mouse coronavirus infection model. Epigenetic reprogramming of HSPC may underlie altered immune function following infection and be broadly relevant, especially for millions of COVID-19 survivors.


Subject(s)
COVID-19 , Epigenetic Memory , Post-Acute COVID-19 Syndrome , Animals , Humans , Mice , Cell Differentiation , COVID-19/immunology , Disease Models, Animal , Hematopoietic Stem Cells , Inflammation/genetics , Trained Immunity , Monocytes/immunology , Post-Acute COVID-19 Syndrome/genetics , Post-Acute COVID-19 Syndrome/immunology , Post-Acute COVID-19 Syndrome/pathology
4.
Cell ; 182(2): 372-387.e14, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32610084

ABSTRACT

Acute psychological stress has long been known to decrease host fitness to inflammation in a wide variety of diseases, but how this occurs is incompletely understood. Using mouse models, we show that interleukin-6 (IL-6) is the dominant cytokine inducible upon acute stress alone. Stress-inducible IL-6 is produced from brown adipocytes in a beta-3-adrenergic-receptor-dependent fashion. During stress, endocrine IL-6 is the required instructive signal for mediating hyperglycemia through hepatic gluconeogenesis, which is necessary for anticipating and fueling "fight or flight" responses. This adaptation comes at the cost of enhancing mortality to a subsequent inflammatory challenge. These findings provide a mechanistic understanding of the ontogeny and adaptive purpose of IL-6 as a bona fide stress hormone coordinating systemic immunometabolic reprogramming. This brain-brown fat-liver axis might provide new insights into brown adipose tissue as a stress-responsive endocrine organ and mechanistic insight into targeting this axis in the treatment of inflammatory and neuropsychiatric diseases.


Subject(s)
Adipose Tissue, Brown/metabolism , Interleukin-6/metabolism , Stress, Psychological , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Brain/metabolism , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Gluconeogenesis , Hyperglycemia/metabolism , Hyperglycemia/pathology , Interleukin-6/blood , Interleukin-6/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adrenergic, beta-3/metabolism , Receptors, Interleukin-6/metabolism , Uncoupling Protein 1/deficiency , Uncoupling Protein 1/genetics
5.
Cell ; 181(5): 1036-1045.e9, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32416070

ABSTRACT

Viral pandemics, such as the one caused by SARS-CoV-2, pose an imminent threat to humanity. Because of its recent emergence, there is a paucity of information regarding viral behavior and host response following SARS-CoV-2 infection. Here we offer an in-depth analysis of the transcriptional response to SARS-CoV-2 compared with other respiratory viruses. Cell and animal models of SARS-CoV-2 infection, in addition to transcriptional and serum profiling of COVID-19 patients, consistently revealed a unique and inappropriate inflammatory response. This response is defined by low levels of type I and III interferons juxtaposed to elevated chemokines and high expression of IL-6. We propose that reduced innate antiviral defenses coupled with exuberant inflammatory cytokine production are the defining and driving features of COVID-19.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , RNA Viruses/immunology , Animals , COVID-19 , Cells, Cultured , Chemokines/genetics , Chemokines/immunology , Coronavirus Infections/genetics , Disease Models, Animal , Host-Pathogen Interactions , Humans , Immunity, Innate , Inflammation/virology , Interferons/genetics , Interferons/immunology , Pandemics , Pneumonia, Viral/genetics , RNA Viruses/classification , SARS-CoV-2 , Transcription, Genetic
6.
Immunity ; 56(5): 979-997.e11, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37100060

ABSTRACT

Immune cell trafficking constitutes a fundamental component of immunological response to tissue injury, but the contribution of intrinsic RNA nucleotide modifications to this response remains elusive. We report that RNA editor ADAR2 exerts a tissue- and stress-specific regulation of endothelial responses to interleukin-6 (IL-6), which tightly controls leukocyte trafficking in IL-6-inflamed and ischemic tissues. Genetic ablation of ADAR2 from vascular endothelial cells diminished myeloid cell rolling and adhesion on vascular walls and reduced immune cell infiltration within ischemic tissues. ADAR2 was required in the endothelium for the expression of the IL-6 receptor subunit, IL-6 signal transducer (IL6ST; gp130), and subsequently, for IL-6 trans-signaling responses. ADAR2-induced adenosine-to-inosine RNA editing suppressed the Drosha-dependent primary microRNA processing, thereby overwriting the default endothelial transcriptional program to safeguard gp130 expression. This work demonstrates a role for ADAR2 epitranscriptional activity as a checkpoint in IL-6 trans-signaling and immune cell trafficking to sites of tissue injury.


Subject(s)
Interleukin-6 , RNA , Endothelial Cells/metabolism , Cytokine Receptor gp130 , Endothelium/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism
7.
Cell ; 169(1): 132-147.e16, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28340339

ABSTRACT

The accumulation of irreparable cellular damage restricts healthspan after acute stress or natural aging. Senescent cells are thought to impair tissue function, and their genetic clearance can delay features of aging. Identifying how senescent cells avoid apoptosis allows for the prospective design of anti-senescence compounds to address whether homeostasis can also be restored. Here, we identify FOXO4 as a pivot in senescent cell viability. We designed a FOXO4 peptide that perturbs the FOXO4 interaction with p53. In senescent cells, this selectively causes p53 nuclear exclusion and cell-intrinsic apoptosis. Under conditions where it was well tolerated in vivo, this FOXO4 peptide neutralized doxorubicin-induced chemotoxicity. Moreover, it restored fitness, fur density, and renal function in both fast aging XpdTTD/TTD and naturally aged mice. Thus, therapeutic targeting of senescent cells is feasible under conditions where loss of health has already occurred, and in doing so tissue homeostasis can effectively be restored.


Subject(s)
Aging/pathology , Antibiotics, Antineoplastic/adverse effects , Cell-Penetrating Peptides/pharmacology , Doxorubicin/adverse effects , Aging/drug effects , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Cell Cycle Proteins , Cell Line , Cell Survival , Cellular Senescence/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Female , Fibroblasts/cytology , Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/metabolism , Humans , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Kidney/drug effects , Kidney/physiology , Liver/drug effects , Liver/physiology , Male , Mice , Trichothiodystrophy Syndromes/drug therapy , Tumor Suppressor Protein p53/metabolism
8.
Immunity ; 55(6): 1032-1050.e14, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35704993

ABSTRACT

Conventional dendritic cells (cDCs), cDC1 and cDC2, act both to initiate immunity and maintain self-tolerance. The tryptophan metabolic enzyme indoleamine 2,3-dioxygenase 1 (IDO1) is used by cDCs in maintaining tolerance, but its role in different subsets remains unclear. At homeostasis, only mature CCR7+ cDC1 expressed IDO1 that was dependent on IRF8. Lipopolysaccharide treatment induced maturation and IDO1-dependent tolerogenic activity in isolated immature cDC1, but not isolated cDC2. However, both human and mouse cDC2 could induce IDO1 and acquire tolerogenic function when co-cultured with mature cDC1 through the action of cDC1-derived l-kynurenine. Accordingly, cDC1-specific inactivation of IDO1 in vivo exacerbated disease in experimental autoimmune encephalomyelitis. This study identifies a previously unrecognized metabolic communication in which IDO1-expressing cDC1 cells extend their immunoregulatory capacity to the cDC2 subset through their production of tryptophan metabolite l-kynurenine. This metabolic axis represents a potential therapeutic target in treating autoimmune demyelinating diseases.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase , Kynurenine , Animals , Dendritic Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Mice , Signal Transduction , Tryptophan/metabolism
9.
Mol Cell ; 83(11): 1872-1886.e5, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37172591

ABSTRACT

Deregulated inflammation is a critical feature driving the progression of tumors harboring mutations in the liver kinase B1 (LKB1), yet the mechanisms linking LKB1 mutations to deregulated inflammation remain undefined. Here, we identify deregulated signaling by CREB-regulated transcription coactivator 2 (CRTC2) as an epigenetic driver of inflammatory potential downstream of LKB1 loss. We demonstrate that LKB1 mutations sensitize both transformed and non-transformed cells to diverse inflammatory stimuli, promoting heightened cytokine and chemokine production. LKB1 loss triggers elevated CRTC2-CREB signaling downstream of the salt-inducible kinases (SIKs), increasing inflammatory gene expression in LKB1-deficient cells. Mechanistically, CRTC2 cooperates with the histone acetyltransferases CBP/p300 to deposit histone acetylation marks associated with active transcription (i.e., H3K27ac) at inflammatory gene loci, promoting cytokine expression. Together, our data reveal a previously undefined anti-inflammatory program, regulated by LKB1 and reinforced through CRTC2-dependent histone modification signaling, that links metabolic and epigenetic states to cell-intrinsic inflammatory potential.


Subject(s)
Histones , Protein Serine-Threonine Kinases , Humans , Histones/genetics , Histones/metabolism , Acetylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Cytokines/metabolism , Inflammation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Immunity ; 54(12): 2877-2892.e7, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34852217

ABSTRACT

Adjuvants are critical for improving the quality and magnitude of adaptive immune responses to vaccination. Lipid nanoparticle (LNP)-encapsulated nucleoside-modified mRNA vaccines have shown great efficacy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but the mechanism of action of this vaccine platform is not well-characterized. Using influenza virus and SARS-CoV-2 mRNA and protein subunit vaccines, we demonstrated that our LNP formulation has intrinsic adjuvant activity that promotes induction of strong T follicular helper cell, germinal center B cell, long-lived plasma cell, and memory B cell responses that are associated with durable and protective antibodies in mice. Comparative experiments demonstrated that this LNP formulation outperformed a widely used MF59-like adjuvant, AddaVax. The adjuvant activity of the LNP relies on the ionizable lipid component and on IL-6 cytokine induction but not on MyD88- or MAVS-dependent sensing of LNPs. Our study identified LNPs as a versatile adjuvant that enhances the efficacy of traditional and next-generation vaccine platforms.


Subject(s)
B-Lymphocytes/immunology , COVID-19 Vaccines/immunology , COVID-19/immunology , Germinal Center/immunology , SARS-CoV-2/physiology , T-Lymphocytes, Helper-Inducer/immunology , mRNA Vaccines/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adjuvants, Immunologic , Animals , HEK293 Cells , Humans , Immunity, Humoral , Interleukin-6/genetics , Interleukin-6/metabolism , Liposomes/administration & dosage , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Protein Subunits/genetics , mRNA Vaccines/genetics
11.
Immunity ; 54(6): 1186-1199.e7, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33915108

ABSTRACT

A cardinal feature of COVID-19 is lung inflammation and respiratory failure. In a prospective multi-country cohort of COVID-19 patients, we found that increased Notch4 expression on circulating regulatory T (Treg) cells was associated with disease severity, predicted mortality, and declined upon recovery. Deletion of Notch4 in Treg cells or therapy with anti-Notch4 antibodies in conventional and humanized mice normalized the dysregulated innate immunity and rescued disease morbidity and mortality induced by a synthetic analog of viral RNA or by influenza H1N1 virus. Mechanistically, Notch4 suppressed the induction by interleukin-18 of amphiregulin, a cytokine necessary for tissue repair. Protection by Notch4 inhibition was recapitulated by therapy with Amphiregulin and, reciprocally, abrogated by its antagonism. Amphiregulin declined in COVID-19 subjects as a function of disease severity and Notch4 expression. Thus, Notch4 expression on Treg cells dynamically restrains amphiregulin-dependent tissue repair to promote severe lung inflammation, with therapeutic implications for COVID-19 and related infections.


Subject(s)
Host-Pathogen Interactions , Immunity, Cellular , Pneumonia, Viral/etiology , Pneumonia, Viral/metabolism , Receptor, Notch4/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Amphiregulin/pharmacology , Animals , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Immunohistochemistry , Immunomodulation/drug effects , Inflammation Mediators/metabolism , Influenza A virus/physiology , Lung/immunology , Lung/metabolism , Lung/pathology , Lung/virology , Mice , Mice, Transgenic , Pneumonia, Viral/pathology , Receptor, Notch4/antagonists & inhibitors , Receptor, Notch4/genetics , Severity of Illness Index
12.
Immunity ; 54(11): 2611-2631.e8, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758338

ABSTRACT

Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.


Subject(s)
Hippocampus/metabolism , Interleukin-6/biosynthesis , Maternal Exposure , Neurons/metabolism , Prenatal Exposure Delayed Effects , Synapses/metabolism , Animals , Cytokines/biosynthesis , Disease Models, Animal , Disease Susceptibility , Female , Hippocampus/physiopathology , Inflammation Mediators/metabolism , Mice , Pregnancy , Signal Transduction , Synaptic Transmission
13.
Immunity ; 54(7): 1463-1477.e11, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34115964

ABSTRACT

Acute respiratory distress syndrome (ARDS), an inflammatory condition with high mortality rates, is common in severe COVID-19, whose risk is reduced by metformin rather than other anti-diabetic medications. Detecting of inflammasome assembly in post-mortem COVID-19 lungs, we asked whether and how metformin inhibits inflammasome activation while exerting its anti-inflammatory effect. We show that metformin inhibited NLRP3 inflammasome activation and interleukin (IL)-1ß production in cultured and alveolar macrophages along with inflammasome-independent IL-6 secretion, thus attenuating lipopolysaccharide (LPS)- and SARS-CoV-2-induced ARDS. By targeting electron transport chain complex 1 and independently of AMP-activated protein kinase (AMPK) or NF-κB, metformin blocked LPS-induced and ATP-dependent mitochondrial (mt) DNA synthesis and generation of oxidized mtDNA, an NLRP3 ligand. Myeloid-specific ablation of LPS-induced cytidine monophosphate kinase 2 (CMPK2), which is rate limiting for mtDNA synthesis, reduced ARDS severity without a direct effect on IL-6. Thus, inhibition of ATP and mtDNA synthesis is sufficient for ARDS amelioration.


Subject(s)
Adenosine Triphosphate/metabolism , DNA, Mitochondrial/biosynthesis , Inflammasomes/drug effects , Metformin/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pneumonia/prevention & control , Animals , COVID-19/metabolism , COVID-19/prevention & control , Cytokines/genetics , Cytokines/metabolism , DNA, Mitochondrial/metabolism , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Metformin/therapeutic use , Mice , Nucleoside-Phosphate Kinase/metabolism , Pneumonia/metabolism , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/prevention & control , SARS-CoV-2/pathogenicity
14.
Immunity ; 53(3): 685-696.e3, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32783921

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic poses a current world-wide public health threat. However, little is known about its hallmarks compared to other infectious diseases. Here, we report the single-cell transcriptional landscape of longitudinally collected peripheral blood mononuclear cells (PBMCs) in both COVID-19- and influenza A virus (IAV)-infected patients. We observed increase of plasma cells in both COVID-19 and IAV patients and XIAP associated factor 1 (XAF1)-, tumor necrosis factor (TNF)-, and FAS-induced T cell apoptosis in COVID-19 patients. Further analyses revealed distinct signaling pathways activated in COVID-19 (STAT1 and IRF3) versus IAV (STAT3 and NFκB) patients and substantial differences in the expression of key factors. These factors include relatively increase of interleukin (IL)6R and IL6ST expression in COVID-19 patients but similarly increased IL-6 concentrations compared to IAV patients, supporting the clinical observations of increased proinflammatory cytokines in COVID-19 patients. Thus, we provide the landscape of PBMCs and unveil distinct immune response pathways in COVID-19 and IAV patients.


Subject(s)
Coronavirus Infections/immunology , Cytokines/immunology , Influenza, Human/immunology , Leukocytes, Mononuclear/immunology , Pneumonia, Viral/immunology , Signal Transduction/immunology , Betacoronavirus/immunology , COVID-19 , Humans , Influenza A Virus, H1N1 Subtype/immunology , Pandemics , SARS-CoV-2
15.
Immunity ; 50(4): 1007-1023, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995492

ABSTRACT

Interleukin-6 (IL-6) is a pleiotropic cytokine with roles in immunity, tissue regeneration, and metabolism. Rapid production of IL-6 contributes to host defense during infection and tissue injury, but excessive synthesis of IL-6 and dysregulation of IL-6 receptor signaling is involved in disease pathology. Therapeutic agents targeting the IL-6 axis are effective in rheumatoid arthritis, and applications are being extended to other settings of acute and chronic inflammation. Recent studies reveal that selective blockade of different modes of IL-6 receptor signaling has different outcomes on disease pathology, suggesting novel strategies for therapeutic intervention. However, some inflammatory diseases do not seem to respond to IL-6 blockade. Here, we review the current state of IL-6-targeting approaches in the clinic and discuss how to apply the growing understanding of the immunobiology of IL-6 to clinical decisions.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Inflammation/drug therapy , Interleukin-6/antagonists & inhibitors , Molecular Targeted Therapy , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Clinical Trials as Topic , Cytokine Receptor gp130/antagonists & inhibitors , Cytokine Receptor gp130/immunology , Humans , Inflammation/immunology , Interleukin-6/biosynthesis , Interleukin-6/deficiency , Interleukin-6/immunology , Janus Kinases/antagonists & inhibitors , Janus Kinases/physiology , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , Receptors, Interleukin-6/immunology , Ribonucleases/deficiency , STAT3 Transcription Factor/physiology , Suppressor of Cytokine Signaling 1 Protein/physiology , Suppressor of Cytokine Signaling 3 Protein/physiology
16.
Development ; 151(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39250533

ABSTRACT

The cell-intrinsic mechanisms underlying the decision of a stem/progenitor cell to either proliferate or differentiate remain incompletely understood. Here, we identify the transmembrane protein Lrig1 as a physiological homeostatic regulator of FGF2-driven proliferation and self-renewal of neural progenitors at early-to-mid embryonic stages of cortical development. We show that Lrig1 is expressed in cortical progenitors (CPs), and its ablation caused expansion and increased proliferation of radial/apical progenitors and of neurogenic transit-amplifying Tbr2+ intermediate progenitors. Notably, our findings identify a previously unreported EGF-independent mechanism through which Lrig1 negatively regulates neural progenitor proliferation by modulating the FGF2-induced IL6/Jak2/Stat3 pathway, a molecular cascade that plays a pivotal role in the generation and maintenance of CPs. Consistently, Lrig1 knockout mice showed a significant increase in the density of pyramidal glutamatergic neurons placed in superficial layers 2 and 3 of the postnatal neocortex. Together, these results support a model in which Lrig1 regulates cortical neurogenesis by influencing the cycling activity of a set of progenitors that are temporally specified to produce upper layer glutamatergic neurons.


Subject(s)
Janus Kinase 2 , Membrane Glycoproteins , Mice, Knockout , Neural Stem Cells , Neurogenesis , Neurons , STAT3 Transcription Factor , Signal Transduction , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Janus Kinase 2/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mice , Neurogenesis/genetics , Neurons/metabolism , Neurons/cytology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Cell Proliferation , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cell Differentiation , Fibroblast Growth Factors/metabolism , Nerve Tissue Proteins
17.
Immunity ; 48(4): 745-759.e6, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669252

ABSTRACT

It is unclear how quiescence is enforced in naive T cells, but activation by foreign antigens and self-antigens is allowed, despite the presence of inhibitory signals. We showed that active transforming growth factor ß (TGF-ß) signaling was present in naive T cells, and T cell receptor (TCR) engagement reduced TGF-ß signaling during T cell activation by downregulating TGF-ß type 1 receptor (TßRI) through activation of caspase recruitment domain-containing protein 11 (CARD11) and nuclear factor κB (NF-κB). TGF-ß prevented TCR-mediated TßRI downregulation, but this was abrogated by interleukin-6 (IL-6). Mitigation of TCR-mediated TßRI downregulation through overexpression of TßRI in naive and activated T cells rendered T cells less responsive and suppressed autoimmunity. Naive T cells in autoimmune patients exhibited reduced TßRI expression and increased TCR-driven proliferation compared to healthy subjects. Thus, TCR-mediated regulation of TßRI-TGF-ß signaling acts as a crucial criterion to determine T cell quiescence and activation.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , CD4-Positive T-Lymphocytes/immunology , Guanylate Cyclase/metabolism , Lymphocyte Activation/immunology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptors, Antigen, T-Cell/immunology , Transforming Growth Factor beta1/metabolism , Animals , Autoimmunity/immunology , CARD Signaling Adaptor Proteins/genetics , Cell Line , Cell Proliferation , Colitis/immunology , Colitis/pathology , Disease Models, Animal , Down-Regulation/immunology , Guanylate Cyclase/genetics , HEK293 Cells , Humans , Interleukin-6/immunology , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Receptor, Transforming Growth Factor-beta Type I/biosynthesis , Signal Transduction/immunology , Transforming Growth Factor beta1/biosynthesis
18.
Proc Natl Acad Sci U S A ; 121(2): e2315898120, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38165930

ABSTRACT

Protection against endothelial damage is recognized as a frontline approach to preventing the progression of cytokine release syndrome (CRS). Accumulating evidence has demonstrated that interleukin-6 (IL-6) promotes vascular endothelial damage during CRS, although the molecular mechanisms remain to be fully elucidated. Targeting IL-6 receptor signaling delays CRS progression; however, current options are limited by persistent inhibition of the immune system. Here, we show that endothelial IL-6 trans-signaling promoted vascular damage and inflammatory responses via hypoxia-inducible factor-1α (HIF1α)-induced glycolysis. Using pharmacological inhibitors targeting HIF1α activity or mice with the genetic ablation of gp130 in the endothelium, we found that inhibition of IL-6R (IL-6 receptor)-HIF1α signaling in endothelial cells protected against vascular injury caused by septic damage and provided survival benefit in a mouse model of sepsis. In addition, we developed a short half-life anti-IL-6R antibody (silent anti-IL-6R antibody) and found that it was highly effective at augmenting survival for sepsis and severe burn by strengthening the endothelial glycocalyx and reducing cytokine storm, and vascular leakage. Together, our data advance the role of endothelial IL-6 trans-signaling in the progression of CRS and indicate a potential therapeutic approach for burns and sepsis.


Subject(s)
Cytokine Receptor gp130 , Hypoxia-Inducible Factor 1, alpha Subunit , Interleukin-6 , Receptors, Interleukin-6 , Sepsis , Animals , Mice , Cytokine Receptor gp130/genetics , Cytokine Release Syndrome , Endothelial Cells , Receptors, Interleukin-6/genetics , Sepsis/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
19.
Trends Immunol ; 44(6): 399-407, 2023 06.
Article in English | MEDLINE | ID: mdl-37100645

ABSTRACT

Conventional dendritic cells (cDCs) can integrate multiple stimuli from the environment and provide three separate outputs in terms of antigen presentation, costimulation, and cytokine production; this guides the activation, expansion, and differentiation of distinct functional T helper subsets. Accordingly, the current dogma posits that T helper cell specification requires these three signals in sequence. Data show that T helper 2 (Th2) cell differentiation requires antigen presentation and costimulation from cDCs but does not require polarizing cytokines. In this opinion article, we propose that the 'third signal' driving Th2 cell responses is, in fact, the absence of polarizing cytokines; indeed, the secretion of the latter is actively suppressed in cDCs, concomitant with acquired pro-Th2 functions.


Subject(s)
Cytokines , Th2 Cells , Humans , T-Lymphocytes, Helper-Inducer , Cell Differentiation , Th1 Cells
20.
Immunity ; 46(2): 220-232, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228280

ABSTRACT

Fibroblasts are major contributors to and regulators of inflammation and dominant producers of interleukin-6 (IL-6) in inflammatory diseases like rheumatoid arthritis. Yet, compared to leukocytes, the regulation of inflammatory pathways in fibroblasts is largely unknown. Here, we report that analyses of genes coordinately upregulated with IL-6 pointed to STAT4 and leukemia inhibitory factor (LIF) as potentially linked. Gene silencing revealed that STAT4 was required for IL-6 transcription. STAT4 was recruited to the IL-6 promoter after fibroblast activation, and LIF receptor (LIFR) and STAT4 formed a molecular complex that, together with JAK1 and TYK2 kinases, controlled STAT4 activation. Importantly, a positive feedback loop involving autocrine LIF, LIFR, and STAT4 drove sustained IL-6 transcription. Besides IL-6, this autorine loop also drove the production of other key inflammatory factors including IL-8, granulocyte-colony stimulating factor (G-CSF), IL-33, IL-11, IL-1α, and IL-1ß. These findings define the transcriptional regulation of fibroblast-mediated inflammation as distinct from leukocytes.


Subject(s)
Autocrine Communication/immunology , Fibroblasts/immunology , Gene Expression Regulation/immunology , Leukemia Inhibitory Factor/immunology , Receptors, OSM-LIF/immunology , Arthritis, Rheumatoid/immunology , Cells, Cultured , Cytokines/biosynthesis , Gene Expression Profiling , Humans , Inflammation/immunology , Interleukin-6/immunology , STAT4 Transcription Factor/immunology , Synovial Membrane/immunology , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL