Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
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.
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
3.
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
4.
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
5.
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
7.
Front Immunol ; 13: 910021, 2022.
Article in English | MEDLINE | ID: mdl-36248833

ABSTRACT

Deficiency of adenosine deaminase 2 (DADA2) is an autosomal recessive disease associated with a highly variable clinical presentation, including vasculitis, immunodeficiency, and hematologic manifestations, potentially progressing over time. The present study describes the long-term evolution of the immuno-hematological features and therapeutic challenge of two identical adult twin sisters affected by DADA2. The absence of plasmatic adenosine deaminase 2 (ADA2) activity in both twins suggested the diagnosis of DADA2, then confirmed by genetic analysis. Exon sequencing revealed a missense (p.Leu188Pro) mutation on the paternal ADA2 allele. While, whole genome sequencing identified an unreported deletion (IVS6_IVS7del*) on the maternal allele predicted to produce a transcript missing exon 7. The patients experienced the disease onset during childhood with early strokes (Patient 1 at two years, Patient 2 at eight years of age), subsequently followed by other shared DADA2-associated features, including neutropenia, hypogammaglobulinemia, reduced switched memory B cells, inverted CD4:CD8 ratio, increased naïve T cells, reduced follicular regulatory T cells, the almost complete absence of NK cells, T-large granular cell leukemia, and osteoporosis. Disease evolution differed: clinical manifestations presented several years earlier and were more pronounced in Patient 1 than in Patient 2. Due to G-CSF refractory life-threatening neutropenia, Patient 1 successfully underwent an urgent hematopoietic stem cell transplantation (HSCT) from a 9/10 matched unrelated donor. Patient 2 experienced a similar, although delayed, disease evolution and is currently on anti-TNF therapy and anti-infectious prophylaxis. The unique cases confirmed that heterozygous patients with null ADA2 activity deserve deep investigation for possible structural variants on a single allele. Moreover, this report emphasizes the importance of timely recognizing DADA2 at the onset to allow adequate follow-up and detection of disease progression. Finally, the therapeutic management in these identical twins raises significant concerns as they share a similar phenotype, with a delayed but almost predictable disease evolution in one of them, who could benefit from a prompt definitive treatment like elective allogeneic HSCT. Additional data are required to assess whether the absence of enzymatic activity at diagnosis is associated with hematological involvement and is also predictive of bone marrow dysfunction, encouraging early HSCT to improve functional outcomes.


Subject(s)
Agammaglobulinemia , Neutropenia , Polyarteritis Nodosa , Adenosine Deaminase/genetics , Agammaglobulinemia/diagnosis , Agammaglobulinemia/genetics , Granulocyte Colony-Stimulating Factor , Humans , Intercellular Signaling Peptides and Proteins , Severe Combined Immunodeficiency , Tumor Necrosis Factor Inhibitors , Twins, Monozygotic/genetics
8.
Front Immunol ; 13: 898298, 2022.
Article in English | MEDLINE | ID: mdl-35844522

ABSTRACT

Gasdermins (GSDMs) are a class of pore-forming proteins related to pyroptosis, a programmed cell death pathway that is induced by a range of inflammatory stimuli. Small-scale GSDM activation and pore formation allow the passive release of cytokines, such as IL-1ß and IL-18, and alarmins, but, whenever numerous GSDM pores are assembled, osmotic lysis and cell death occur. Such GSDM-mediated pyroptosis promotes pathogen clearance and can help restore homeostasis, but recent studies have revealed that dysregulated pyroptosis is at the root of many inflammation-mediated disease conditions. Moreover, new homeostatic functions for gasdermins are beginning to be revealed. Here, we review the newly discovered mechanisms of GSDM activation and their prominent roles in host defense and human diseases associated with chronic inflammation. We also highlight the potential of targeting GSDMs as a new therapeutic approach to combat chronic inflammatory diseases and cancer and how we might overcome the current obstacles to realize this potential.


Subject(s)
Inflammasomes , Neoplasms , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Pyroptosis/physiology
10.
Blood Adv ; 5(16): 3174-3187, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34424322

ABSTRACT

Adenosine deaminase 2 deficiency (DADA2) is a rare inherited disorder that is caused by autosomal recessive mutations in the ADA2 gene. Clinical manifestations include early-onset lacunar strokes, vasculitis/vasculopathy, systemic inflammation, immunodeficiency, and hematologic defects. Anti-tumor necrosis factor therapy reduces strokes and systemic inflammation. Allogeneic hematopoietic stem/progenitor cell (HSPC) transplantation can ameliorate most disease manifestations, but patients are at risk for complications. Autologous HSPC gene therapy may be an alternative curative option for patients with DADA2. We designed a lentiviral vector encoding ADA2 (LV-ADA2) to genetically correct HSPCs. Lentiviral transduction allowed efficient delivery of the functional ADA2 enzyme into HSPCs from healthy donors. Supranormal ADA2 expression in human and mouse HSPCs did not affect their multipotency and engraftment potential in vivo. The LV-ADA2 induced stable ADA2 expression and corrected the enzymatic defect in HSPCs derived from DADA2 patients. Patients' HSPCs re-expressing ADA2 retained their potential to differentiate into erythroid and myeloid cells. Delivery of ADA2 enzymatic activity in patients' macrophages led to a complete rescue of the exaggerated inflammatory cytokine production. Our data indicate that HSPCs ectopically expressing ADA2 retain their multipotent differentiation ability, leading to functional correction of macrophage defects. Altogether, these findings support the implementation of HSPC gene therapy for DADA2.


Subject(s)
Adenosine Deaminase , Vasculitis , Adenosine Deaminase/genetics , Animals , Humans , Inflammation , Intercellular Signaling Peptides and Proteins , Macrophages , Mice
11.
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
12.
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
13.
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
14.
Front Immunol ; 10: 1566, 2019.
Article in English | MEDLINE | ID: mdl-31379813

ABSTRACT

The inflammasome is a multi-protein complex that mediates proteolytic cleavage and release of the pro-inflammatory cytokines IL-1ß and IL-18, and pyroptosis-a form of cell death induced by various pathogenic bacteria. Apoptosis-associated speck-like protein containing a CARD (ASC) has a pivotal role in inflammasome assembly and activation. While ASC function has been primarily implicated in innate immune cells, its contribution to lymphocyte biology is unclear. Here we report that ASC is constitutively expressed in naïve CD4+ T cells together with the inflammasome sensor NLRP3 and caspase-1. When adoptively transferred in immunocompromised Rag1-/- mice, Asc-/- CD4+ T cells exacerbate T-cell-mediated autoimmune colitis. Asc-/- CD4+ T cells exhibit a higher proliferative capacity in vitro than wild-type CD4+ T cells. The increased expansion of Asc-/- CD4+ T cells in vivo correlated with robust TCR-mediated activation, inflammatory activity, and higher metabolic profile toward a highly glycolytic phenotype. These findings identify ASC as a crucial intrinsic regulator of CD4+ T-cell expansion that serves to maintain intestinal homeostasis.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Homeostasis/immunology , Intestines/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Caspase 1/genetics , Caspase 1/immunology , Caspase 1/metabolism , Cells, Cultured , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Homeostasis/genetics , Inflammasomes/genetics , Inflammasomes/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
15.
Front Immunol ; 10: 1556, 2019.
Article in English | MEDLINE | ID: mdl-31333677

ABSTRACT

The inflammasome is an intracellular multi-protein complex that orchestrates the release of the pro-inflammatory cytokines IL-1ß and IL-18, and a form of cell death known as pyroptosis. Tyrosine phosphorylation of the inflammasome sensors NLRP3, AIM2, NLRC4, and the adaptor protein, apoptosis-associated speck-like protein (ASC) has previously been demonstrated to be essential in the regulation of the inflammasome. By using the pharmacological protein tyrosine phosphatase (PTPase) inhibitor, phenylarsine oxide (PAO), we have demonstrated that tyrosine dephosphorylation is an essential step for the activation of the NLRP3 and AIM2 inflammasomes in human and murine macrophages. We have also shown that PTPase activity is required for ASC nucleation leading to caspase-1 activation, IL-1ß, and IL-18 processing and release, and cell death. Furthermore, by site-directed mutagenesis of ASC tyrosine residues, we have identified the phosphorylation of tyrosine Y60 and Y137 of ASC as critical for inflammasome assembly and function. Therefore, we report that ASC tyrosine dephosphorylation and phosphorylation are crucial events for inflammasome activation.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , DNA-Binding Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphorylation/physiology , Tyrosine/metabolism , Animals , Caspase 1/metabolism , Cell Line , Cytokines/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Protein Tyrosine Phosphatases/metabolism , Th1 Cells
16.
Sci Rep ; 9(1): 4913, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894604

ABSTRACT

Interleukin-1ß (IL-1ß) is a major cytokine that initiates and enhances inflammatory responses. Excessive IL-1ß production is a characteristic of most chronic inflammatory diseases, including atherosclerosis, type 2 diabetes, and obesity, which affect a large proportion of the global population. The production of bioactive IL-1ß is mediated by a caspase-1-activating complex known as an 'inflammasome'. The NLRP3 inflammasome has been associated with several human inflammatory and autoimmune diseases and represents a potential therapeutic target for disrupting IL-1ß production. We used molecular modeling guided by molecular dynamics simulations to design α-helical stapled peptides targeting the pyrin domain of the adaptor protein ASC to interrupt the development of its filament, which is crucial for NLRP3 inflammasome formation. The peptides were effectively internalized by human monocytic cells and efficiently suppressed the release of the inflammasome-regulated cytokines IL-1ß and IL-18, following exogenous activation of the NLRP3 inflammasome. The peptides reduced ASC speck formation and caspase-1 processing thereby suppressing pro-IL-1ß processing and release of active IL-1ß. This is the first demonstration of the successful use of stapled peptides designed to target the adaptor protein ASC, and can be extended to other inflammatory pathways to disrupt excessive IL-1ß production.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , Cell-Penetrating Peptides/pharmacology , Inflammasomes/drug effects , Interleukin-1beta/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , Binding Sites , CARD Signaling Adaptor Proteins/antagonists & inhibitors , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/immunology , Cell-Penetrating Peptides/chemistry , Gene Expression Regulation , Humans , Hydrophobic and Hydrophilic Interactions , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Models, Molecular , NF-kappa B/genetics , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nigericin/pharmacology , Proof of Concept Study , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Signal Transduction , THP-1 Cells , Thermodynamics
17.
Front Immunol ; 9: 261, 2018.
Article in English | MEDLINE | ID: mdl-29515579

ABSTRACT

Calcineurin (Cn) is a protein phosphatase that regulates the activation of the nuclear factor of activated T-cells (NFAT) family of transcription factors, which are key regulators of T-cell development and function. Here, we generated a conditional Cnb1 mouse model in which Cnb1 was specifically deleted in CD4+ T cells (Cnb1CD4 mice) to delineate the role of the Cn-NFAT pathway in immune homeostasis of the intestine. The Cnb1CD4 mice developed severe, spontaneous colitis characterized at the molecular level by an increased T helper-1-cell response but an unaltered regulatory T-cell compartment. Antibiotic treatment ameliorated the intestinal inflammation observed in Cnb1CD4 mice, suggesting that the microbiota contributes to the onset of colitis. CD4+ T cells isolated from Cnb1CD4 mice produced high levels of IFNγ due to increased activation of the JAK2/STAT4 pathway induced by IL-12. Our data highlight that Cn signaling in CD4+ T cells is critical for intestinal immune homeostasis in part by inhibiting IL-12 responsiveness of CD4+ T cells.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Calcineurin/metabolism , Colitis/immunology , Inflammatory Bowel Diseases/immunology , Intestines/immunology , Animals , Calcineurin/genetics , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Gastrointestinal Microbiome/immunology , Homeostasis , Humans , Interferon-gamma/metabolism , Interleukin-12/metabolism , Janus Kinase 2/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT4 Transcription Factor/metabolism , Signal Transduction
18.
Nat Commun ; 9(1): 1102, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29549257

ABSTRACT

The intestinal immune system can respond to invading pathogens yet maintain immune tolerance to self-antigens and microbiota. Myeloid cells are central to these processes, but the signaling pathways that underlie tolerance versus inflammation are unclear. Here we show that mice lacking Calcineurin B in CD11chighMHCII+ cells (Cnb1 CD11c mice) spontaneously develop intestinal inflammation and are susceptible to induced colitis. In these mice, colitis is associated with expansion of T helper type 1 (Th1) and Th17 cell populations and a decrease in the number of FoxP3+ regulatory T (Treg) cells, and the pathology is linked to the inability of intestinal Cnb1-deficient CD11chighMHCII+ cells to express IL-2. Deleting IL-2 in CD11chighMHCII+ cells induces spontaneous colitis resembling human inflammatory bowel disease. Our findings identify that the calcineurin-NFAT-IL-2 pathway in myeloid cells is a critical regulator of intestinal homeostasis by influencing the balance of inflammatory and regulatory responses in the mouse intestine.


Subject(s)
CD11c Antigen/immunology , Calcineurin/immunology , Colitis/immunology , Interleukin-2/immunology , Intestines/immunology , Myeloid Cells/immunology , Animals , CD11c Antigen/genetics , Calcineurin/genetics , Colitis/genetics , Female , Genes, MHC Class II , Homeostasis , Humans , Interleukin-2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/immunology , Th17 Cells/immunology
19.
Sci Rep ; 8(1): 2847, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29434227

ABSTRACT

Diabetic retinopathy (DR) is a retinal microvascular disease characterized by inflammatory and angiogenic pathways. In this study, we evaluated NLRP3 inflammasome in a double transgenic mouse model, Akimba (Ins2 Akita xVEGF+/-), which demonstrates hyperglycemia, vascular hyperpermeability and neovascularization seen in the proliferative DR. Retinal structural integrity, vascular leakage and function were examined by fundus photography, fluorescein angiography, optical coherence tomography, retinal flat mounts, laser speckle flowgraphy (LSFG), and electroretinography in Akimba and its parental strains, Akita (Ins2 Akita ) and Kimba (trVEGF029) mice. Inflammatory mechanisms involving NLRP3 inflammasome were investigated using real time-PCR, immunohistochemistry, ELISA and western blots. We observed an increased vascular leakage, reduced retinal thickness, and function in Akimba retina. Also, Akimba retina depicts decreased relative flow volume measured by LSFG. Most importantly, high levels of IL-1ß along with increased NLRP3, ASC, and Caspase-1 at mRNA and protein levels were observed in Akimba retina. However, the in vivo functional role remains undefined. In conclusion, increased activation of macroglia (GFAP), microglia (Iba-1 and OX-42) and perivascular macrophages (F4/80 and CD14) together with pro-inflammatory (IL-1ß and IL-6) and pro-angiogenic markers (PECAM-1, ICAM-1, VEGF, Flt-1, and Flk-1), suggested a critical role for NLRP3 inflammasome in the Akimba mouse model depicting advanced stages of DR pathogenesis.


Subject(s)
Diabetic Retinopathy/diagnostic imaging , Insulin/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neovascularization, Pathologic/diagnostic imaging , Vascular Endothelial Growth Factor A/genetics , Animals , Cytokines/genetics , Cytokines/metabolism , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Disease Models, Animal , Electroretinography , Fluorescein Angiography , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Retina/diagnostic imaging , Retina/pathology , Tomography, Optical Coherence
20.
Sci Rep ; 7(1): 17429, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29234083

ABSTRACT

Acinetobacter baumannii (A. baumannii) is a significant cause of severe nosocomial pneumonia in immunocompromised individuals world-wide. With limited treatment options available, a better understanding of host immnity to A. baumannii infection is critical to devise alternative control strategies. Our previous study has identified that intracellular Nod1/Nod2 signaling pathway is required for the immune control of A. baumannii in airway epithelial cells in vitro. In the current study, using Nod2-/- mice and an in vivo sublethal model of pulmonary infection, we show that Nod2 contributes to the early lung defense against A. baumannii infection through reactive oxygen species (ROS)/reactive nitrogen species (RNS) production as Nod2-/- mice showed significantly reduced production of ROS/RNS in the lungs following A. baumannii infection. Consistent with the higher bacterial load, A. baumannii-induced neutrophil recruitment, cytokine/chemokine response and lung pathology was also exacerbated in Nod2-/- mice at early time points post-infection. Finally, we show that administration of Nod2 ligand muramyl dipeptide (MDP) prior to infection protected the wild- type mice from A. baumannii pulmonary challenge. Collectively, Nod2 is an important player in the early lung immunity against A. baumannii and modulating Nod2 pathway could be considered as a viable therapeutic strategy to control A. baumannii pulmonary infection.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Immunity, Innate/physiology , Lung/immunology , Nod2 Signaling Adaptor Protein/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Acinetobacter Infections/drug therapy , Acinetobacter Infections/pathology , Animals , Anti-Infective Agents/pharmacology , Female , Lung/drug effects , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/genetics , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...