Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Nat Immunol ; 25(4): 693-702, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38486019

ABSTRACT

The inflammasome components NLRP3 and ASC are cytosolic proteins, which upon sensing endotoxins or danger cues, form multimeric complexes to process interleukin (IL)-1ß for secretion. Here we found that antigen (Ag)-triggered degranulation of IgE-sensitized mast cells (MCs) was mediated by NLRP3 and ASC. IgE-Ag stimulated NEK7 and Pyk2 kinases in MCs to induce the deposition of NLRP3 and ASC on granules and form a distinct protein complex (granulosome) that chaperoned the granules to the cell surface. MCs deficient in NLRP3 or ASC did not form granulosomes, degranulated poorly in vitro and did not evoke systemic anaphylaxis in mice. IgE-Ag-triggered anaphylaxis was prevented by an NLRP3 inhibitor. In endotoxin-primed MCs, pro-IL-1ß was rapidly packaged into granules after IgE-Ag stimulation and processed within granule remnants by proteases after degranulation, causing lethal anaphylaxis in mice. During IgE-Ag-mediated degranulation of endotoxin-primed MCs, granulosomes promoted degranulation, combined with exteriorization and processing of IL-1ß, resulting in severe inflammation.


Subject(s)
Anaphylaxis , Inflammasomes , Mice , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mast Cells , Anaphylaxis/metabolism , Immunoglobulin E/metabolism , Endotoxins/metabolism , Cell Degranulation
2.
Nature ; 623(7986): 415-422, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37914939

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with high resistance to therapies1. Inflammatory and immunomodulatory signals co-exist in the pancreatic tumour microenvironment, leading to dysregulated repair and cytotoxic responses. Tumour-associated macrophages (TAMs) have key roles in PDAC2, but their diversity has prevented therapeutic exploitation. Here we combined single-cell and spatial genomics with functional experiments to unravel macrophage functions in pancreatic cancer. We uncovered an inflammatory loop between tumour cells and interleukin-1ß (IL-1ß)-expressing TAMs, a subset of macrophages elicited by a local synergy between prostaglandin E2 (PGE2) and tumour necrosis factor (TNF). Physical proximity with IL-1ß+ TAMs was associated with inflammatory reprogramming and acquisition of pathogenic properties by a subset of PDAC cells. This occurrence was an early event in pancreatic tumorigenesis and led to persistent transcriptional changes associated with disease progression and poor outcomes for patients. Blocking PGE2 or IL-1ß activity elicited TAM reprogramming and antagonized tumour cell-intrinsic and -extrinsic inflammation, leading to PDAC control in vivo. Targeting the PGE2-IL-1ß axis may enable preventive or therapeutic strategies for reprogramming of immune dynamics in pancreatic cancer.


Subject(s)
Inflammation , Interleukin-1beta , Pancreatic Neoplasms , Tumor-Associated Macrophages , Humans , Carcinogenesis , Carcinoma, Pancreatic Ductal/complications , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Dinoprostone/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Tumor Necrosis Factors/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
3.
Mol Ther ; 29(2): 489-504, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33221437

ABSTRACT

Gene transfer into autologous hematopoietic stem progenitor cells (HSPCs) has the potential to cure monogenic inherited disorders caused by an altered development and/or function of the blood system, such as immune deficiencies and red blood cell and platelet disorders. Gene-corrected HSPCs and their progeny can also be exploited as cell vehicles to deliver molecules into the circulation and tissues, including the central nervous system. In this review, we focus on the progress of clinical development of medicinal products based on HSPCs engineered and modified by integrating viral vectors for the treatment of monogenic blood disorders and metabolic diseases. Two products have reached the stage of market approval in the EU, and more are foreseen to be approved in the near future. Despite these achievements, several challenges remain for HSPC gene therapy (HSPC-GT) precluding a wider application of this type of gene therapy to a wider set of diseases while gene-editing approaches are entering the clinical arena.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/therapy , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Animals , Disease Management , Disease Susceptibility , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Humans , Lentivirus/genetics , Transduction, Genetic
4.
Mol Ther ; 29(1): 86-102, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33010230

ABSTRACT

Chronic granulomatous disease (CGD) is a rare inherited disorder due to loss-of-function mutations in genes encoding the NADPH oxidase subunits. Hematopoietic stem and progenitor cell (HSPC) gene therapy (GT) using regulated lentiviral vectors (LVs) has emerged as a promising therapeutic option for CGD patients. We performed non-clinical Good Laboratory Practice (GLP) and laboratory-grade studies to assess the safety and genotoxicity of LV targeting myeloid-specific Gp91phox expression in X-linked chronic granulomatous disease (XCGD) mice. We found persistence of gene-corrected cells for up to 1 year, restoration of Gp91phox expression and NADPH oxidase activity in XCGD phagocytes, and reduced tissue inflammation after LV-mediated HSPC GT. Although most of the mice showed no hematological or biochemical toxicity, a small subset of XCGD GT mice developed T cell lymphoblastic lymphoma (2.94%) and myeloid leukemia (5.88%). No hematological malignancies were identified in C57BL/6 mice transplanted with transduced XCGD HSPCs. Integration pattern analysis revealed an oligoclonal composition with rare dominant clones harboring vector insertions near oncogenes in mice with tumors. Collectively, our data support the long-term efficacy of LV-mediated HSPC GT in XCGD mice and provide a safety warning because the chronic inflammatory XCGD background may contribute to oncogenesis.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/therapy , Hematologic Neoplasms/etiology , Lentivirus/genetics , Animals , Disease Models, Animal , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Granulomatous Disease, Chronic/genetics , Humans , Mice , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , Time Factors , Treatment Outcome
5.
Int J Mol Sci ; 22(12)2021 Jun 14.
Article in English | MEDLINE | ID: mdl-34198548

ABSTRACT

Inflammation in the tumor microenvironment has been shown to promote disease progression in pancreatic ductal adenocarcinoma (PDAC); however, the role of macrophage metabolism in promoting inflammation is unclear. Using an orthotopic mouse model of PDAC, we demonstrate that macrophages from tumor-bearing mice exhibit elevated glycolysis. Macrophage-specific deletion of Glucose Transporter 1 (GLUT1) significantly reduced tumor burden, which was accompanied by increased Natural Killer and CD8+ T cell activity and suppression of the NLRP3-IL1ß inflammasome axis. Administration of mice with a GLUT1-specific inhibitor reduced tumor burden, comparable with gemcitabine, the current standard-of-care. In addition, we observe that intra-tumoral macrophages from human PDAC patients exhibit a pronounced glycolytic signature, which reliably predicts poor survival. Our data support a key role for macrophage metabolism in tumor immunity, which could be exploited to improve patient outcomes.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cytoprotection , Glycolysis , Macrophages/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/immunology , Animals , Carcinoma, Pancreatic Ductal/immunology , Cell Proliferation/drug effects , Cytoprotection/drug effects , Drug Resistance, Neoplasm/drug effects , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Humans , Hydroxybenzoates/pharmacology , Inflammation/pathology , Interleukin-1beta/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Macrophages/drug effects , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pancreatic Neoplasms/immunology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/drug effects , Pancreatic Neoplasms
6.
J Immunol ; 198(1): 196-204, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27895176

ABSTRACT

Despite a long history and extensive usage of insoluble aluminum salts (alum) as vaccine adjuvants, the molecular mechanisms underpinning Ag-specific immunity upon vaccination remain unclear. Dendritic cells (DCs) are crucial initiators of immune responses, but little is known about the molecular pathways used by DCs to sense alum and, in turn, activate T and B cells. In this article, we show that alum adjuvanticity requires IL-2 specifically released by DCs, even when T cell secretion of IL-2 is intact. We demonstrate that alum, as well as other sterile particulates, such as uric acid crystals, induces DCs to produce IL-2 following initiation of actin-mediated phagocytosis that leads to Src and Syk kinase activation, Ca2+ mobilization, and calcineurin-dependent activation of NFAT, the master transcription factor regulating IL-2 expression. Using chimeric mice, we show that DC-derived IL-2 is required for maximal Ag-specific proliferation of CD4+ T cells and optimal humoral responses following alum-adjuvanted immunization. These data identify DC-derived IL-2 as a key mediator of alum adjuvanticity in vivo and the Src-Syk pathway as a potential leverage point in the rational design of novel adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Dendritic Cells/immunology , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukin-2/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Models, Animal , NFATC Transcription Factors/immunology , Signal Transduction/drug effects , Syk Kinase/immunology
7.
J Immunol ; 193(2): 496-509, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24943219

ABSTRACT

The Blomia tropicalis dust mite is prevalent in tropical and subtropical regions of the world. Although it is a leading cause of asthma, little is known how it induces allergy. Using a novel murine asthma model induced by intranasal exposure to B. tropicalis, we observed that a single intranasal sensitization to B. tropicalis extract induces strong Th2 priming in the lung draining lymph node. Resident CD11b(+) dendritic cells (DCs) preferentially transport Ag from the lung to the draining lymph node and are crucial for the initiation of Th2 CD4(+) T cell responses. As a consequence, mice selectively deficient in CD11b(+) DCs exhibited attenuated Th2 responses and more importantly did not develop any allergic inflammation. Conversely, mice deficient in CD103(+) DCs and CCR2-dependent monocyte-derived DCs exhibited similar allergic inflammation compared with their wild-type counterparts. We also show that CD11b(+) DCs constitutively express higher levels of GM-CSF receptor compared with CD103(+) DCs and are thus selectively licensed by lung epithelial-derived GM-CSF to induce Th2 immunity. Taken together, our study identifies GM-CSF-licensed CD11b(+) lung DCs as a key component for induction of Th2 responses and represents a potential target for therapeutic intervention in allergy.


Subject(s)
Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Lung/immunology , Mites/immunology , Th2 Cells/immunology , Administration, Intranasal , Adoptive Transfer , Animals , Asthma/immunology , Asthma/metabolism , CD11b Antigen/immunology , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunization/methods , Interleukin-4/immunology , Interleukin-4/metabolism , Lung/metabolism , Lung/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mites/metabolism , Ovalbumin/immunology , Th2 Cells/metabolism , Tissue Extracts/administration & dosage , Tissue Extracts/immunology
8.
J Pathol ; 233(4): 321-30, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24740681

ABSTRACT

Within the NOD-like receptor (NLR) family, there are several NLRP (NLR family, pyrin domain-containing) proteins that are involved in the formation of inflammasomes. These multi-protein complexes are a key part of the network of cellular events required for secretion of the pro-inflammatory cytokines IL-1ß and IL-18. The NLRP3 inflammasome is the best-characterized member of the family and has recently been implicated in gut homeostasis and determining the severity of inflammation in inflammatory bowel disease (IBD) and inflammation-associated colorectal cancer. This led to the discovery that NLRP6 and NLRP12 also contribute to the maintenance of intestinal homeostasis and modulation of the gut microbiota, which in turn influences the intestine and distant organs. In this review, we bring together the latest data on the potential roles of NLRP family members in gut health and disease and identify the most pressing questions that remain to be answered to further our understanding of human diseases including IBD, inflammation-associated cancers, and metabolic syndromes linked with obesity.


Subject(s)
Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiology , Homeostasis/physiology , Intracellular Signaling Peptides and Proteins/physiology , Microbiota/physiology , Animals , Colitis/physiopathology , Colorectal Neoplasms/physiopathology , Cytokines/metabolism , Humans , Inflammasomes/physiology , Inflammatory Bowel Diseases/physiopathology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Metabolic Syndrome/physiopathology , Mice , Mice, Knockout
9.
J Immunol ; 191(3): 1006-10, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23817414

ABSTRACT

The complement system is a potent component of the innate immune response, promoting inflammation and orchestrating defense against pathogens. However, dysregulation of complement is critical to several autoimmune and inflammatory syndromes. Elevated expression of the proinflammatory cytokine IL-1ß is often linked to such diseases. In this study, we reveal the mechanistic link between complement and IL-1ß secretion using murine dendritic cells. IL-1ß secretion occurs following intracellular caspase-1 activation by inflammasomes. We show that complement elicits secretion of both IL-1ß and IL-18 in vitro and in vivo via the NLRP3 inflammasome. This effect depends on the inflammasome components NLRP3 and ASC, as well as caspase-1 activity. Interestingly, sublethal complement membrane attack complex formation, but not the anaphylatoxins C3a and C5a, activated the NLRP3 inflammasome in vivo. These findings provide insight into the molecular processes underlying complement-mediated inflammation and highlight the possibility of targeting IL-1ß to control complement-induced disease and pathological inflammation.


Subject(s)
Carrier Proteins/metabolism , Caspase 1/metabolism , Dendritic Cells/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Animals , Bone Marrow Cells , Carrier Proteins/genetics , Cells, Cultured , Complement C6/deficiency , Complement C6/genetics , Complement System Proteins/immunology , Dendritic Cells/metabolism , Enzyme Activation , Inflammation/immunology , Interleukin-18/biosynthesis , Interleukin-18/immunology , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Receptors, Complement/deficiency , Receptors, Complement/genetics , Signal Transduction
10.
Infect Immun ; 82(3): 1112-22, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24366254

ABSTRACT

Acinetobacter baumannii is a major extensively drug-resistant lethal human nosocomial bacterium. However, the host innate immune mechanisms controlling A. baumannii are not well understood. Although viewed as an extracellular pathogen, A. baumannii can also invade and survive intracellularly. However, whether host innate immune pathways sensing intracellular bacteria contribute to immunity against A. baumannii is not known. Here, we provide evidence for the first time that intracellular antibacterial innate immune receptors Nod1 and Nod2, and their adaptor Rip2, play critical roles in the sensing and clearance of A. baumannii by human airway epithelial cells in vitro. A. baumannii infection upregulated Rip2 expression. Silencing of Nod1, Nod2, and Rip2 expression profoundly increased intracellular invasion and prolonged the multiplication and survival of A. baumannii in lung epithelial cells. Notably, the Nod1/2-Rip2 axis did not contribute to the control of A. baumannii infection of human macrophages, indicating that they play cell type-specific roles. The Nod1/2-Rip2 axis was needed for A. baumannii infection-induced activation of NF-κB but not mitogen-activated protein kinases. Moreover, the Nod1/2-Rip2 axis was critical to induce optimal cytokine and chemokine responses to A. baumannii infection. Mechanistic studies showed that the Nod1/2 pathway contributed to the innate control of A. baumannii infection through the production of ß-defensin 2 by airway epithelial cells. This study revealed new insights into the immune control of A. baumannii and may contribute to the development of effective immune therapeutics and vaccines against A. baumannii.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Immunity, Innate/immunology , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Acinetobacter Infections/genetics , Acinetobacter Infections/microbiology , Cell Line , Chemokines/genetics , Chemokines/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , HEK293 Cells , Humans , Immunity, Innate/genetics , Lung/immunology , Lung/microbiology , Macrophages/immunology , Macrophages/microbiology , NF-kappa B/genetics , NF-kappa B/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation/genetics , Up-Regulation/immunology , beta-Defensins/genetics , beta-Defensins/immunology
11.
Eur J Immunol ; 43(9): 2240-5, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24037676

ABSTRACT

Inflammasomes are large multiprotein platforms that mediate the processing of caspase-1, which in turn promotes the maturation and release of IL-1ß and IL-18 in response to microbial and danger signals. While the canonical pathway of inflammasome activation has been known for some time, a novel mechanism of noncanonical inflammasome activation mediated by caspase-11 was more recently identified. This pathway engages caspase-11 to trigger both caspase-1-dependent and -independent production of the inflammatory cytokines IL-1ß, IL-18, and IL-1α, as well as to promote pyroptosis, a form of genetically programmed cell death that is associated with the release of such cytokines. In this review, we gather together studies on both the mechanisms and implications of caspase-11-mediated noncanonical inflammasome activation, and discuss the emerging importance of this pathway in regulating host defense against intracellular bacterial pathogens.


Subject(s)
Bacterial Infections/immunology , Bacterial Infections/metabolism , Caspases/metabolism , Inflammasomes/metabolism , Animals , Apoptosis , Caspase 1/metabolism , Caspases, Initiator , Humans , Interleukin-18/biosynthesis , Interleukin-18/metabolism , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/metabolism , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice
12.
Eur J Immunol ; 43(8): 2126-37, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23619996

ABSTRACT

The NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is a cytoplasmic protein complex that mediates inflammatory responses to a broad array of danger signals. The inflammasome drives caspase-1 activation and promotes secretion of the pro-inflammatory cytokines IL-1ß and IL-18, and might also participate in other cellular processes. Here, we tried to identify new pathways regulated by the NLRP3 inflammasome in murine dendritic cells (DCs) in response to monosodium urate (MSU) crystals. Using a transcriptomic approach, we found that DCs from Nlrp3(-/-) mice responded to MSU with differential expression of genes involved in the DNA damage response and apoptosis. Upon exposure to MSU or other ROS-mobilizing stimuli (rotenone and γ-radiation), DNA fragmentation was markedly ameliorated in Nlrp3(-/-) and casp-1(-/-) DCs compared with WT DCs. Moreover, Nlrp3(-/-) DCs experienced significantly less oxidative DNA damage mediated by ROS. A significant decrease of the expression of several genes involved in double-strand and base-excision DNA repair was observed in WT DCs. Basal DNA repair capacity in WT DCs resulted in activation and stabilization of p53 in vitro and in vivo, which resulted in increased cell death compared with that in Nlrp3(-/-) DCs. These data provide the first evidence for the involvement of the NLRP3 inflammasome in DNA damage responses induced by cellular stress.


Subject(s)
Carrier Proteins/metabolism , DNA Damage/drug effects , DNA Repair/genetics , Dendritic Cells/metabolism , Inflammasomes/immunology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Carrier Proteins/genetics , Caspase 1/genetics , Cell Survival , Cells, Cultured , DNA Repair/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Enzyme Activation , Inflammation/chemically induced , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Peritonitis/chemically induced , Peritonitis/immunology , Reactive Oxygen Species , Rotenone/pharmacology , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Suppressor Protein p53/metabolism , Uncoupling Agents/pharmacology , Uric Acid/pharmacology
13.
J Immunol ; 188(4): 1789-98, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22250091

ABSTRACT

A simultaneous engagement of different pathogen recognition receptors provides a tailor-made adaptive immunity for an efficient defense against distinct pathogens. For example, cross-talk of TLR and C-type lectin signaling effectively shapes distinct gene expression patterns by integrating the signals at the level of NF-κB. In this study, we extend this principle to a strong synergism between the dectin-1 agonist curdlan and an inflammatory growth factor, GM-CSF. Both together act in synergy in inducing a strong inflammatory signature that converts immature dendritic cells (DCs) to potent effector DCs. A variety of cytokines (IL-1ß, IL-6, TNF-α, IL-2, and IL-12p70), costimulatory molecules (CD80, CD86, CD40, and CD70), chemokines (CXCL1, CXCL2, CXCL3, CCL12, CCL17), as well as receptors and molecules involved in fugal recognition and immunity such as Mincle, dectin-1, dectin-2, and pentraxin 3 are strongly upregulated in DC treated simultaneously with curdlan and GM-CSF. The synergistic effect of both stimuli resulted in strong IκBα phosphorylation, its rapid degradation, and enhanced nuclear translocation of all NF-κB subunits. We further identified MAPK ERK as one possible integration site of both signals, because its phosphorylation was clearly augmented when curdlan was coapplied with GM-CSF. Our data demonstrate that the immunomodulatory activity of curdlan requires an additional signal provided by GM-CSF to successfully initiate a robust ß-glucan-specific cytokine and chemokine response. The integration of both signals clearly prime and tailor a more effective innate and adaptive response against invading microbes and fungi.


Subject(s)
Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunologic Factors , beta-Glucans/immunology , Animals , Antigens, CD/biosynthesis , Cell Differentiation , Chemokines/biosynthesis , Cytokines/biosynthesis , Drug Synergism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , I-kappa B Proteins/metabolism , Lectins, C-Type/agonists , Lectins, C-Type/biosynthesis , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation , Polysaccharides, Bacterial/immunology , Signal Transduction , beta-Glucans/pharmacology
14.
Hum Gene Ther ; 35(7-8): 219-231, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38323580

ABSTRACT

Inflammatory diseases are conditions characterized by abnormal and often excessive immune responses, leading to tissue and organ inflammation. The complexity of these disorders arises from the intricate interplay of genetic factors and immune responses, which challenges conventional therapeutic approaches. However, the field of genetic manipulation has sparked unprecedented optimism in addressing these complex disorders. This review aims to comprehensively explore the application of gene therapy and gene editing in the context of inflammatory diseases, offering solutions that range from correcting genetic defects to precise immune modulation. These therapies have exhibited remarkable potential in ameliorating symptoms, improving quality of life, and even achieving disease remission. As we delve into recent breakthroughs and therapeutic applications, we illustrate how these advancements offer novel and transformative solutions for conditions that have traditionally eluded conventional treatments. By examining successful case studies and preclinical research, we emphasize the favorable results and substantial transformative impacts that gene-based interventions have demonstrated in patients and animal models of inflammatory diseases such as chronic granulomatous disease, cryopyrin-associated syndromes, and adenosine deaminase 2 deficiency, as well as those of multifactorial origins such as arthropathies (osteoarthritis, rheumatoid arthritis) and inflammatory bowel disease. In conclusion, gene therapy and gene editing offer transformative opportunities to address the underlying causes of inflammatory diseases, ushering in a new era of precision medicine and providing hope for personalized, targeted treatments.


Subject(s)
Gene Editing , Severe Combined Immunodeficiency , Animals , Humans , Gene Editing/methods , Quality of Life , Genetic Therapy/methods , Genetic Engineering , CRISPR-Cas Systems
15.
Commun Biol ; 7(1): 615, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777862

ABSTRACT

Deficiency of adenosine deaminase 2 (DADA2) is an inborn error of immunity caused by loss-of-function mutations in the adenosine deaminase 2 (ADA2) gene. Clinical manifestations of DADA2 include vasculopathy and immuno-hematological abnormalities, culminating in bone marrow failure. A major gap exists in our knowledge of the regulatory functions of ADA2 during inflammation and hematopoiesis, mainly due to the absence of an ADA2 orthologue in rodents. Exploring these mechanisms is essential for understanding disease pathology and developing new treatments. Zebrafish possess two ADA2 orthologues, cecr1a and cecr1b, with the latter showing functional conservation with human ADA2. We establish a cecr1b-loss-of-function zebrafish model that recapitulates the immuno-hematological and vascular manifestations observed in humans. Loss of Cecr1b disrupts hematopoietic stem cell specification, resulting in defective hematopoiesis. This defect is caused by induced inflammation in the vascular endothelium. Blocking inflammation, pharmacological modulation of the A2r pathway, or the administration of the recombinant human ADA2 corrects these defects, providing insights into the mechanistic link between ADA2 deficiency, inflammation and immuno-hematological abnormalities. Our findings open up potential therapeutic avenues for DADA2 patients.


Subject(s)
Adenosine Deaminase , Hematopoiesis , Hematopoietic Stem Cells , Inflammation , Zebrafish , Animals , Zebrafish/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Adenosine Deaminase/deficiency , Hematopoietic Stem Cells/metabolism , Inflammation/genetics , Inflammation/metabolism , Hematopoiesis/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Humans , Signal Transduction , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism
16.
Trends Immunol ; 31(1): 18-23, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19836307

ABSTRACT

Dendritic cells (DC) are professional phagocytes possessing a unique ability to sense perturbations in the tissue microenvironment and promote adaptive immune responses, whilst maintaining immunological tolerance. Mouse myeloid DC progenitors with the ability to migrate through the blood and replenish the DC pool have been identified in bone marrow but the ontogeny of human DC is poorly understood. Access to lymphoid tissues for human DC isolation is severely limited and researchers have resorted to the use of in vitro derivation systems in attempts to understand DC development, which may result in misleading conclusions. The identification of a human DC progenitor in blood would greatly enhance the understanding of DC homeostasis and their role in pathogenesis.


Subject(s)
Blood Cells/cytology , Dendritic Cells/cytology , Stem Cells/cytology , Animals , Humans , Mice , Monocytes/cytology
17.
J Immunol ; 187(1): 258-65, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21622857

ABSTRACT

Ag recognition is achieved through the communication across intercellular contacts between T cells and APCs such as dendritic cells (DC). Despite remarkable progress in delineating detailed molecular components at the intercellular contacts, little is known about the functional roles of physical cross-junctional adhesion between T and DC in shaping T cell responses. In addition, the mechanisms underlying sensitivity and specificity of Ag discrimination by T cells at intercellular contacts remain to be elucidated. In this study, we use single-cell force spectroscopy to probe the mechanical interactions between DC and T cells in response to stimulation with a panel of altered peptide ligands. The results show that intercellular interactions of DC-T cell conjugates exhibited different ranges of interaction forces in peptide-dependent manners that match the ability of the peptides to activate T cells. Elevated calcium mobilization and IL-2 secretion by T cells were only promoted in response to antigenic peptides that induce strong interaction forces, suggesting that mechanically stable DC-T cell contacts are crucial for driving T cell activation. Strong interactions were not solely dependent on cell-surface molecules such as TCRs and the adhesion molecule LFA-1, but were also controlled by cytoskeletal dynamics and the integrity of membrane lipid rafts. These data provide novel mechanical insights into the effect of Ag affinity on intercellular contacts that align with T cell responsiveness.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Amino Acid Sequence , Animals , Calcium/metabolism , Cell Communication/genetics , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Cholesterol/deficiency , Cholesterol/metabolism , Cytoskeleton/immunology , Cytoskeleton/pathology , Histocompatibility Antigens Class II/immunology , Interleukin-2/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Atomic Force , Molecular Sequence Data , Ovalbumin/immunology , Peptide Fragments/immunology , Peptides/metabolism , Peptides/physiology
18.
J Immunol ; 187(11): 5842-50, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22058415

ABSTRACT

Uric acid is released from damaged cells and serves as a danger signal that alerts the immune system to potential threats, even in the absence of microbial infection. Uric acid modulation of innate immune responses has been extensively studied, but the impact of this damage-associated molecular pattern on adaptive responses remains largely unknown. In this study, we report that, in the presence of NF-κB signaling, uric acid crystals were capable of stimulating dendritic cells to promote the release of cytokines associated with Th17 polarization. Accordingly, naive CD4(+) T cells cocultured with uric acid-treated dendritic cells differentiated toward the Th17 lineage. Th17 differentiation required the inflammasome-dependent cytokines IL-1α/ß and IL-18 in both in vitro and in vivo models, and the inflammasome adaptor protein ASC and caspase-1 were essential for Th17 responses. Collectively, our findings indicate a novel role for the danger signal uric acid, in cooperation with NF-κB activation, in driving proinflammatory Th17 differentiation. Our data indicate that sterile inflammation shapes adaptive immunity, in addition to influencing early innate responses.


Subject(s)
Cell Differentiation/immunology , Inflammasomes/immunology , Interleukin-18/biosynthesis , Interleukin-1/biosynthesis , Th17 Cells/cytology , Uric Acid/immunology , Adaptive Immunity/immunology , Adjuvants, Immunologic , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Hemocyanins/immunology , Hemocyanins/pharmacology , Interleukin-1/immunology , Interleukin-18/immunology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , NF-kappa B/metabolism , Real-Time Polymerase Chain Reaction , Th17 Cells/immunology
19.
Cells ; 12(13)2023 07 03.
Article in English | MEDLINE | ID: mdl-37443800

ABSTRACT

Inflammasome complexes and their integral receptor proteins have essential roles in regulating the innate immune response and inflammation at the post-translational level. Yet despite their protective role, aberrant activation of inflammasome proteins and gain of function mutations in inflammasome component genes seem to contribute to the development and progression of human autoimmune and autoinflammatory diseases. In the past decade, our understanding of inflammasome biology and activation mechanisms has greatly progressed. We therefore provide an up-to-date overview of the various inflammasomes and their known mechanisms of action. In addition, we highlight the involvement of various inflammasomes and their pathogenic mechanisms in common autoinflammatory, autoimmune and neurodegenerative diseases, including atherosclerosis, rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease, Alzheimer's disease, Parkinson's disease, and multiple sclerosis. We conclude by speculating on the future avenues of research needed to better understand the roles of inflammasomes in health and disease.


Subject(s)
Arthritis, Rheumatoid , Autoimmune Diseases , Humans , Inflammasomes/metabolism , Immunity, Innate , Inflammation
20.
Sci Transl Med ; 15(698): eade3856, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37256935

ABSTRACT

Dysregulation of the interleukin-1 (IL-1) pathway leads to immune diseases that can result in chronic tissue and organ inflammation. Although IL-1 blockade has shown promise in ameliorating these symptoms and improving patients' quality of life, there is an urgent need for more effective, long-lasting treatments. We developed a lentivirus (LV)-mediated gene transfer strategy using transplanted autologous hematopoietic stem/progenitor cells (HSPCs) as a source of IL-1 receptor antagonist (IL-1RA) for systemic delivery to tissues and organs. Transplantation of mouse and human HSPCs transduced with an IL-1RA-encoding LV ensured stable IL-1RA production while maintaining the clonogenic and differentiation capacities of HSPCs in vivo. We examined the efficacy of cell-mediated IL-1RA delivery in three models of IL-1-dependent inflammation, for which treatment hindered neutrophil recruitment in an inducible model of gout, prevented systemic and multi-tissue inflammation in a genetic model of cryopyrin-associated periodic syndromes, and reduced disease severity in an experimental autoimmune encephalomyelitis model of multiple sclerosis. Our findings demonstrate HSPC-mediated IL-1RA delivery as a potential therapeutic modality that can be exploited to suppress tissue and organ inflammation in diverse immune-related diseases involving IL-1-driven inflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Interleukin 1 Receptor Antagonist Protein , Animals , Humans , Encephalomyelitis, Autoimmune, Experimental/therapy , Inflammation/therapy , Interleukin-1 , Lentivirus , Quality of Life , Mice
SELECTION OF CITATIONS
SEARCH DETAIL