Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(36): e2201494119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037355

ABSTRACT

Pulmonary emphysema is associated with dysregulated innate immune responses that promote chronic pulmonary inflammation and alveolar apoptosis, culminating in lung destruction. However, the molecular regulators of innate immunity that promote emphysema are ill-defined. Here, we investigated whether innate immune inflammasome complexes, comprising the adaptor ASC, Caspase-1 and specific pattern recognition receptors (PRRs), promote the pathogenesis of emphysema. In the lungs of emphysematous patients, as well as spontaneous gp130F/F and cigarette smoke (CS)-induced mouse models of emphysema, the expression (messenger RNA and protein) and activation of ASC, Caspase-1, and the inflammasome-associated PRR and DNA sensor AIM2 were up-regulated. AIM2 up-regulation in emphysema coincided with the biased production of the mature downstream inflammasome effector cytokine IL-1ß but not IL-18. These observations were supported by the genetic blockade of ASC, AIM2, and the IL-1 receptor and therapy with AIM2 antagonistic suppressor oligonucleotides, which ameliorated emphysema in gp130F/F mice by preventing elevated alveolar cell apoptosis. The functional requirement for AIM2 in driving apoptosis in the lung epithelium was independent of its expression in hematopoietic-derived immune cells and the recruitment of infiltrating immune cells in the lung. Genetic and inhibitor-based blockade of AIM2 also protected CS-exposed mice from pulmonary alveolar cell apoptosis. Intriguingly, IL-6 trans-signaling via the soluble IL-6 receptor, facilitated by elevated levels of IL-6, acted upstream of the AIM2 inflammasome to augment AIM2 expression in emphysema. Collectively, we reveal cross-talk between the AIM2 inflammasome/IL-1ß and IL-6 trans-signaling axes for potential exploitation as a therapeutic strategy for emphysema.


Subject(s)
DNA-Binding Proteins , Immunity, Innate , Interleukin-1beta , Interleukin-6 , Pulmonary Emphysema , Animals , Apoptosis , Caspase 1/metabolism , Cytokine Receptor gp130/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Pulmonary Emphysema/immunology
2.
Gut ; 71(8): 1515-1531, 2022 08.
Article in English | MEDLINE | ID: mdl-34489308

ABSTRACT

OBJECTIVE: The absent in melanoma 2 (AIM2) cytosolic pattern recognition receptor and DNA sensor promotes the pathogenesis of autoimmune and chronic inflammatory diseases via caspase-1-containing inflammasome complexes. However, the role of AIM2 in cancer is ill-defined. DESIGN: The expression of AIM2 and its clinical significance was assessed in human gastric cancer (GC) patient cohorts. Genetic or therapeutic manipulation of AIM2 expression and activity was performed in the genetically engineered gp130 F/F spontaneous GC mouse model, as well as human GC cell line xenografts. The biological role and mechanism of action of AIM2 in gastric tumourigenesis, including its involvement in inflammasome activity and functional interaction with microtubule-associated end-binding protein 1 (EB1), was determined in vitro and in vivo. RESULTS: AIM2 expression is upregulated by interleukin-11 cytokine-mediated activation of the oncogenic latent transcription factor STAT3 in the tumour epithelium of GC mouse models and patients with GC. Genetic and therapeutic targeting of AIM2 in gp130 F/F mice suppressed tumourigenesis. Conversely, AIM2 overexpression augmented the tumour load of human GC cell line xenografts. The protumourigenic function of AIM2 was independent of inflammasome activity and inflammation. Rather, in vivo and in vitro AIM2 physically interacted with EB1 to promote epithelial cell migration and tumourigenesis. Furthermore, upregulated expression of AIM2 and EB1 in the tumour epithelium of patients with GC was independently associated with poor patient survival. CONCLUSION: AIM2 can play a driver role in epithelial carcinogenesis by linking cytokine-STAT3 signalling, innate immunity and epithelial cell migration, independent of inflammasome activation.


Subject(s)
Melanoma , Stomach Neoplasms , Animals , Carcinogenesis/genetics , Cell Movement/genetics , Cytokine Receptor gp130/metabolism , DNA , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Immunity, Innate/genetics , Inflammasomes/genetics , Inflammasomes/metabolism , Mice , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/pathology , Up-Regulation
3.
Eur J Immunol ; 51(7): 1686-1697, 2021 07.
Article in English | MEDLINE | ID: mdl-33860535

ABSTRACT

Cytosolic DNA receptor cyclic GMP-AMP (cGAMP) synthase (cGAS) has been shown to be critically involved in the detection of cytosolic, self- and non-self-DNA, initiating a type I IFN response through the adaptor protein Stimulator of Interferon Genes (STING) and interferon regulatory factor 3 (IRF3). Current studies propose that canonical binding of dsDNA by cGAS depends on DNA length, but not on base sequence. In contrast, activation of TLR9 is sequence dependent. It requires unmethylated CpG dinucleotides in microbial DNA, which is mimicked by synthetic oligodeoxynucleotides (ODN). Here, we provide evidence that d-type ODN (D-ODN), but not K-type ODN (K-ODN), bind to human cGAS and activate downstream signaling. Transfection of D-ODN into a TLR9-deficient, human monocytic cell line (THP-1) induced phosphorylation of IRF3 and secretion of IFN. This response was absent in cells with CRISPR/Cas9-mediated cGAS- or STING-deficiency. Utilizing a protein pulldown approach, we further demonstrate direct binding of D-ODN to cGAS. Induction of a type I IFN response by D-ODN was confirmed in human primary monocytes and monocyte-derived macrophages. These results are relevant to our understanding of self-nonself-discrimination by cGAS and to the pharmacologic effects of ODN, which currently are investigated in clinical studies.


Subject(s)
Cytosol/immunology , Interferon Type I/immunology , Membrane Proteins/immunology , Nucleotides, Cyclic/immunology , Oligodeoxyribonucleotides/immunology , Signal Transduction/immunology , Cells, Cultured , HEK293 Cells , Humans , Interferon Regulatory Factor-3/immunology , Macrophages/immunology , Monocytes/immunology , Phosphorylation/immunology , THP-1 Cells
4.
Am J Respir Crit Care Med ; 204(1): 53-63, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33760701

ABSTRACT

Rationale: Acute respiratory distress syndrome (ARDS) is a heterogeneous syndrome with a mortality of up to 40%. Precision medicine approaches targeting patients on the basis of their molecular phenotypes of ARDS might help to identify effective pharmacotherapies. The inflammasome-caspase-1 pathway contributes to the development of ARDS via IL-1ß and IL-18 production. Recent studies indicate that tetracycline can be used to treat inflammatory diseases mediated by IL-1ß and IL-18, although the molecular mechanism by which tetracycline inhibits inflammasome-caspase-1 signaling remains unknown. Objectives: To identify patients with ARDS characterized by IL-1ß and IL-18 expression and investigate the ability of tetracycline to inhibit inflammasome-caspase-1 signaling in ARDS. Methods: IL-1ß and IL-18 concentrations were quantified in BAL fluid from patients with ARDS. Tetracycline's effects on lung injury and inflammation were assessed in two mouse models of direct (pulmonary) acute lung injury, and its effects on IL-1ß and IL-18 production were assessed by alveolar leukocytes from patients with direct ARDS ex vivo. Murine macrophages were used to further characterize the effect of tetracycline on the inflammasome-caspase-1 pathway. Measurements and Main Results: BAL fluid concentrations of IL-1ß and IL-18 are significantly higher in patients with direct ARDS than those with indirect (nonpulmonary) ARDS. In experimental acute lung injury, tetracycline significantly diminished lung injury and pulmonary inflammation by selectively inhibiting caspase-1-dependent IL-1ß and IL-18 production, leading to improved survival. Tetracycline also reduced the production of IL-1ß and IL-18 by alveolar leukocytes from patients with direct ARDS. Conclusions: Tetracycline may be effective in the treatment of direct ARDS in patients with elevated caspase-1 activity. Clinical Trial registered with www.clinicaltrials.gov (NCT04079426).


Subject(s)
Acute Lung Injury/prevention & control , Caspase 1/metabolism , Inflammasomes/metabolism , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/genetics , Tetracycline/metabolism , Acute Lung Injury/etiology , Animals , Anti-Bacterial Agents/metabolism , Enzyme Inhibitors/metabolism , Humans , Immunomodulation , Interleukin-18/genetics , Interleukin-1beta/genetics , Mice , Models, Animal , Respiratory Distress Syndrome/physiopathology
5.
Int Immunol ; 32(1): 39-48, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31633763

ABSTRACT

Immune-mediated inflammation must be down-regulated to facilitate tissue remodeling during homeostatic restoration of an inflammatory response. Uncontrolled or over-exuberant immune activation can cause autoimmune diseases, as well as tissue destruction. A151, the archetypal example of a chemically synthesized suppressive oligodeoxynucleotide (ODN) based on repetitive telomere-derived TTAGGG sequences, was shown to successfully down-regulate a variety of immune responses. However, the degree, duration and breadth of A151-induced transcriptome alterations remain elusive. Here, we performed a comprehensive microarray analysis in combination with Ingenuity Pathway Analysis (IPA) using murine splenocytes to investigate the underlying mechanism of A151-dependent immune suppression. Our results revealed that A151 significantly down-regulates critical mammalian target of rapamycin (mTOR) activators (Pi3kcd, Pdpk1 and Rheb), elements downstream of mTOR signaling (Rps6ka1, Myc, Stat3 and Slc2a1), an important component of the mTORC2 protein complex (Rictor) and Mtor itself. The effects of A151 on mTOR signaling were dose- and time-dependent. Moreover, flow cytometry and immunoblotting analyses demonstrated that A151 is able to reverse mTOR phosphorylation comparably to the well-known mTOR inhibitor rapamycin. Furthermore, Seahorse metabolic assays showed an A151 ODN-induced decrease in both oxygen consumption and glycolysis implying that a metabolically inert state in macrophages could be triggered by A151 treatment. Overall, our findings suggested novel insights into the mechanism by which the immune system is metabolically modulated by A151 ODN.


Subject(s)
Immunosuppressive Agents/pharmacology , Oligodeoxyribonucleotides/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Amino Acid Motifs/drug effects , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/pharmacology , Phosphorylation/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
6.
Infect Immun ; 87(12)2019 12.
Article in English | MEDLINE | ID: mdl-31591164

ABSTRACT

Toll-like receptors (TLR) trigger the immune system to mount a rapid innate response capable of protecting the host from a wide variety of bacterial and viral pathogens. There is interest in harnessing TLR agonists to reduce the susceptibility of at-risk populations to infection. However, the widespread prophylactic use of TLR agonists has been compromised by the need to administer them by parenteral injection. An exception is the TLR7/8 agonist R848, which can boost gastrointestinal and systemic immunity when administered orally. This work examines the effect of R848 on host susceptibility to Listeria monocytogenes in a murine challenge model and describes the underlying mechanisms. Results show that prophylactic administration of R848 significantly reduces susceptibility to infection of BALB/c mice, an effect that lasts 1 week. Oral R848 directly stimulated B cells to produce cytokines and Ig. In the absence of B cells, R848-mediated protection was lost. These findings support the use of oral R848 to reduce the susceptibility of at-risk individuals to infection and identify the critical role of B cells in TLR7-mediated resistance to bacterial infection.


Subject(s)
B-Lymphocytes/immunology , Imidazoles/pharmacology , Listeria monocytogenes/immunology , Listeriosis/prevention & control , Membrane Glycoproteins/immunology , Toll-Like Receptor 7/immunology , Animals , Listeriosis/immunology , Mice , Mice, Inbred BALB C
7.
Eur J Immunol ; 48(4): 605-611, 2018 04.
Article in English | MEDLINE | ID: mdl-29215161

ABSTRACT

Type I interferon (IFN) is a critical mediator of autoimmune diseases such as systemic lupus erythematosus (SLE) and Aicardi-Goutières Syndrome (AGS). The recently discovered cyclic-GMP-AMP (cGAMP) synthase (cGAS) induces the production of type I IFN in response to cytosolic DNA and is potentially linked to SLE and AGS. Suppressive oligodeoxynucleotides (ODN) containing repetitive TTAGGG motifs present in mammalian telomeres have proven useful in the treatment of autoimmune diseases including SLE. In this study, we demonstrate that the suppressive ODN A151 effectively inhibits activation of cGAS in response to cytosolic DNA, thereby inhibiting type I IFN production by human monocytes. In addition, A151 abrogated cGAS activation in response to endogenous accumulation of DNA using TREX1-deficient monocytes. We demonstrate that A151 prevents cGAS activation in a manner that is competitive with DNA. This suppressive activity of A151 was dependent on both telomeric sequence and phosphorothioate backbone. To our knowledge this report presents the first cGAS inhibitor capable of blocking self-DNA. Collectively, these findings might lead to the development of new therapeutics against IFN-driven pathologies due to cGAS activation.


Subject(s)
Interferon Type I/biosynthesis , Monocytes/immunology , Nucleotide Motifs/genetics , Nucleotidyltransferases/antagonists & inhibitors , Oligodeoxyribonucleotides/genetics , Oligonucleotides/genetics , Cell Line , Cytosol , DNA/genetics , Exodeoxyribonucleases/genetics , HEK293 Cells , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Phosphoproteins/genetics , Telomere/genetics
8.
J Autoimmun ; 99: 24-32, 2019 05.
Article in English | MEDLINE | ID: mdl-30679006

ABSTRACT

Systematic Lupus Erythematosus (SLE) is an autoimmune syndrome of unclear etiology. While T and B cell abnormalities contribute to disease pathogenesis, recent work suggests that inflammatory M1-like macrophages also play a role. Previous work showed that the TLR2/1 agonist PAM3CSK4 (PAM3) could stimulate normal human monocytes to preferentially differentiate into immunosuppressive M2-like rather than inflammatory M1-like macrophages. This raised the possibility of PAM3 being used to normalize the M1:M2 ratio in SLE. Consistent with that possibility, monocytes from lupus patients differentiated into M2-like macrophages when treated with PAM3 in vitro. Furthermore, lupus-prone NZB x NZW F1 mice responded similarly to weekly PAM3 treatment. Normalization of the M2 macrophage frequency was associated with delayed disease progression, decreased autoantibody and inflammatory cytokine synthesis, reduced proteinuria and prolonged survival in NZB x NZW F1 mice. The ability of PAM3 to bias monocyte differentiation in favor of immunosuppressive macrophages may represent a novel approach to the therapy of SLE.


Subject(s)
Lipopeptides/pharmacology , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Cell Plasticity/drug effects , Cell Plasticity/immunology , Cytokines/metabolism , Endocytosis/immunology , Female , Immunophenotyping , Lupus Erythematosus, Systemic/complications , Lupus Nephritis/etiology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Macrophages/metabolism , Male , Mice , Monocytes/metabolism , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/metabolism
9.
J Immunol ; 198(2): 681-690, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27986906

ABSTRACT

Th cells sensitized against autoantigens acquire pathogenicity following two sequential events, namely activation by their target Ag and a process named "licensing." In this study, we analyzed these processes in a transgenic mouse system in which TCR-transgenic Th cells specific to hen egg lysozyme (HEL) are adoptively transferred to recipients and induce inflammation in eyes expressing HEL. Our data show that the notion that the lung is the organ where "licensing" for pathogenicity takes place is based on biased data collected with cells injected i.v., a route in which most transferred cells enter via the lung. Thus, we found that when donor cells were activated in vitro and injected intraperitoneally, or were activated in vivo, they migrated simultaneously to the lung, spleen, and other tested organs. In all, tested organs donor cells undergo "licensing" for pathogenicity, consisting of vigorous increase in number and changes in expression levels of inflammation-related genes, monitored by both flow cytometry and microarray analysis. After reaching peak numbers, around day 3, the "licensed" donor cells migrate to the circulation and initiate inflammation in the HEL-expressing recipient eyes. Importantly, the kinetics of increase in number and of changes in gene expression by the donor cells were similar in lung, spleen, and other tested organs of the recipient mice. Furthermore, the total numbers of donor cells in the spleen at their peaks were 10- to 100-fold larger in the spleen than in the lung, contradicting the notion that the lung is the organ where "licensing" takes place.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Autoantigens/immunology , Disease Models, Animal , Flow Cytometry , Lung/immunology , Mice , Mice, Transgenic , Muramidase/immunology , Spleen/immunology
10.
Eur J Immunol ; 46(3): 647-55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26613957

ABSTRACT

Synthetic oligonucleotides (ODNs) containing CpG motifs stimulate human plasmacytoid dendritic cells (pDCs) to produce type-1 interferons (IFNs) and proinflammatory cytokines. Previous studies demonstrated that interferon regulatory factors (IRFs) play a central role in mediating CpG-induced pDC activation. This work explores the inverse effects of IRF5 and IRF8 (also known as IFN consensus sequence-binding protein) on CpG-dependent gene expression in the human CAL-1 pDC cell line. This cell line shares many of the phenotypic and functional properties of freshly isolated human pDCs. Results from RNA interference and microarray studies indicate that IRF5 upregulates TLR9-driven gene expression whereas IRF8 downregulates the same genes. Several findings support the conclusion that IRF8 inhibits TLR9-dependent gene expression by directly blocking the activity of IRF5. First, the inhibitory activity of IRF8 is only observed when IRF5 is present. Second, proximity ligation analysis shows that IRF8 and IRF5 colocalize within the cytoplasm of resting human pDCs and cotranslocate to the nucleus after CpG stimulation. Taken together, these findings suggest that IRF5 and IRF8, two transcription factors with opposing functions, control TLR9 signaling in human pDCs.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factors/immunology , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , Cell Line , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/ultrastructure , Down-Regulation , Gene Expression Regulation , Humans , Microarray Analysis , Oligodeoxyribonucleotides/pharmacology , RNA Interference , Signal Transduction , Toll-Like Receptor 9/genetics
11.
Eur J Immunol ; 46(7): 1615-21, 2016 07.
Article in English | MEDLINE | ID: mdl-27125983

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are a major source of type I interferon (IFN) and are important for host defense by sensing microbial DNA via TLR9. pDCs also play a critical role in the pathogenesis of IFN-driven autoimmune diseases. Yet, this autoimmune reaction is caused by the recognition of self-DNA and has been linked to TLR9-independent pathways. Increasing evidence suggests that the cytosolic DNA receptor cyclic GMP-AMP (cGAMP) synthase (cGAS) is a critical component in the detection of pathogens and contributes to autoimmune diseases. It has been shown that binding of DNA to cGAS results in the synthesis of cGAMP and the subsequent activation of the stimulator of interferon genes (STING) adaptor to induce IFNs. Our results show that the cGAS-STING pathway is expressed and activated in human pDCs by cytosolic DNA leading to a robust type I IFN response. Direct activation of STING by cyclic dinucleotides including cGAMP also activated pDCs and knockdown of STING abolished this IFN response. These results suggest that pDCs sense cytosolic DNA and cyclic dinucleotides via the cGAS-STING pathway and that targeting this pathway could be of therapeutic interest.


Subject(s)
DNA/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon Type I/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction , Cells, Cultured , Cytosol/immunology , Cytosol/metabolism , Gene Expression , Humans , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Type I/genetics , Membrane Proteins/genetics , Nucleotidyltransferases/genetics , Toll-Like Receptor 9/metabolism
12.
J Autoimmun ; 78: 1-10, 2017 03.
Article in English | MEDLINE | ID: mdl-28082046

ABSTRACT

Systemic lupus is the prototypic human autoimmune disease. It is a kaleidoscope of autoreactivities, with clear indications of both a genetic and environmental basis. Indeed, it is a disease that can manifest in virtually every tissue and organ and can also be found spontaneously in a number of animal species, including dogs, cats and horses. Moreover, there are multiple murine models of lupus, the first of which, New Zealand Black (NZB) mice, were discovered in 1959. Despite an enormous effort from scientists in multiple disciplines, the etiology of lupus remains elusive and the introduction of new therapies has been disappointing. Fortunately, significant advances have occurred to help patients through the general principles of internal medicine, including antibiotics, dialysis, and of course use of steroids and immunosuppressive agents. However, the magic bullet has yet to be discovered. One of the major causes of morbidity in lupus remains lupus nephritis and there has been significant effort and encouragement in understanding the pathogenesis, renal histologic classification, and use of therapeutic protocols to induce and sustain remission of lupus nephritis. Indeed, the first use of evidence-based clinical trials in lupus was initiated by Dr. Alfred D. Steinberg at NIH in pioneering studies involving either oral or intravenous pulses of cyclophosphamide, azathioprine or corticosteroids alone and/or some combination. Cyclophosphamide intravenously proved to be superior and the use of cyclophosphamide in combination with methylprednisolone remained the standard protocol for the treatment of lupus nephritis for decades. Although alternative therapies have been introduced, including mycophenolate mofetil, the use of therapies first pioneered at NIH may still be considered standard of care in the appropriate indications. More targeted therapies are much desired. In this review we provide a comprehensive overview of lupus nephritis and the evolution of clinical treatments.


Subject(s)
Lupus Nephritis/therapy , Animals , Biopsy , Histological Techniques/history , Histological Techniques/methods , History, 20th Century , Humans , Lupus Nephritis/diagnosis , Lupus Nephritis/history
13.
J Immunol ; 194(9): 4215-21, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25825448

ABSTRACT

Tumors persist by occupying immunosuppressive microenvironments that inhibit the activity of tumoricidal T and NK cells. Monocytic myeloid-derived suppressor cells (mMDSC) are an important component of this immunosuppressive milieu. We find that the suppressive activity of mMDSC isolated from cancer patients can be reversed by treatment with TLR7/8 agonists, which induce human mMDSC to differentiate into tumoricidal M1-like macrophages. In contrast, agonists targeting TLR1/2 cause mMDSC to mature into immunosuppressive M2-like macrophages. These two populations of macrophage are phenotypically and functionally discrete and differ in gene expression profile. The ability of TLR7/8 agonists to reverse mMDSC-mediated immune suppression suggests that they might be useful adjuncts for tumor immunotherapy.


Subject(s)
Cell Differentiation/drug effects , Imidazoles/pharmacology , Immune Tolerance/drug effects , Monocytes/cytology , Monocytes/drug effects , Myeloid Cells/cytology , Myeloid Cells/drug effects , Toll-Like Receptors/agonists , Humans , Immune Tolerance/immunology , Monocytes/immunology , Myeloid Cells/immunology , Structure-Activity Relationship , Toll-Like Receptors/immunology
14.
Pharmacol Res ; 105: 216-25, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26779666

ABSTRACT

Synthetic oligodeoxynucleotides that can down-regulate cellular elements of the immune system have been developed and are being widely studied in preclinical models. These agents vary in sequence, mechanism of action, and cellular target(s) but share the ability to suppress a plethora of inflammatory responses. This work reviews the types of immunosuppressive oligodeoxynucleotide (Sup ODN) and compares their therapeutic activity against diseases characterized by pathologic levels of immune stimulation ranging from autoimmunity to septic shock to cancer (see graphical abstract). The mechanism(s) underlying the efficacy of Sup ODN and the influence size, sequence and nucleotide backbone on function are considered.


Subject(s)
Immunosuppressive Agents/chemistry , Immunosuppressive Agents/therapeutic use , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/therapeutic use , Animals , Base Sequence , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Immunosuppressive Agents/pharmacology , Interferon Regulatory Factors/immunology , Oligodeoxyribonucleotides/pharmacology , Toll-Like Receptor 9/immunology
15.
J Immunol ; 190(4): 1882-9, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23296706

ABSTRACT

The primary goal of cancer immunotherapy is to elicit an immune response capable of eliminating the tumor. One approach toward accomplishing that goal uses general (rather than tumor-specific) immunomodulatory agents to boost the number and activity of pre-existing CTLs. We find that the intratumoral injection of polyguanosine (poly-G) oligonucleotides (ODN) has such an effect, boosting antitumor immunity and promoting tumor regression. The antitumor activity of poly-G ODN was mediated through CD8 T cells in a TLR9-independent manner. Mechanistically, poly-G ODN directly induced the phosphorylation of Lck (an essential element of the T cell-signaling pathway), thereby enhancing the production of IL-2 and CD8 T cell proliferation. These findings establish poly-G ODN as a novel type of cancer immunotherapy.


Subject(s)
Antineoplastic Agents/metabolism , Guanosine/physiology , Interleukin-2/biosynthesis , Oligodeoxyribonucleotides/biosynthesis , Oligodeoxyribonucleotides/pharmacology , Up-Regulation/immunology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Animals , Cell Line, Tumor , CpG Islands/genetics , CpG Islands/immunology , Guanosine/biosynthesis , Guanosine/genetics , Humans , Interleukin-2/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Oligodeoxyribonucleotides/chemical synthesis , Phosphorylation/drug effects , Phosphorylation/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Tumor Cells, Cultured , Up-Regulation/genetics
16.
J Immunol ; 191(7): 3876-83, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23986531

ABSTRACT

Synthetic oligodeoxynucleotides (ODNs) comprised of the immunosuppressive motif TTAGGG block TLR9 signaling, prevent STAT1 and STAT4 phosphorylation and attenuate a variety of inflammatory responses in vivo. In this study, we demonstrate that such suppressive ODN abrogate activation of cytosolic nucleic acid-sensing pathways. Pretreatment of dendritic cells and macrophages with the suppressive ODN-A151 abrogated type I IFN, TNF-α, and ISG induction in response to cytosolic dsDNA. In addition, A151 abrogated caspase-1-dependent IL-1ß and IL-18 maturation in dendritic cells stimulated with dsDNA and murine CMV. Inhibition was dependent on A151's phosphorothioate backbone, whereas substitution of the guanosine residues for adenosine negatively affected potency. A151 mediates these effects by binding to AIM2 in a manner that is competitive with immune-stimulatory DNA and as a consequence prevents AIM2 inflammasome complex formation. Collectively, these findings reveal a new route by which suppressive ODNs modulate the immune system and unveil novel applications for suppressive ODNs in the treatment of infectious and autoimmune diseases.


Subject(s)
Inflammasomes/antagonists & inhibitors , Nuclear Proteins/metabolism , Nucleotide Motifs , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Cell Line , Cluster Analysis , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Cytosol/metabolism , DNA/metabolism , DNA-Binding Proteins , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Inflammasomes/chemistry , Inflammasomes/metabolism , Mice , Oligodeoxyribonucleotides/metabolism , Protein Binding , Protein Multimerization/drug effects , Signal Transduction/drug effects , Thionucleotides/chemistry
17.
J Immunol ; 191(1): 415-23, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23720812

ABSTRACT

The interaction between TLRs and their cognate ligands triggers both the innate and adaptive immune systems, and thus can play a pivotal role in the defense against pathogen invasion. This work investigates the differentiation of naive CD4 cells into Th1 or Th17 phenotypes in mice treated with different TLR ligands. We use a model system in which naive transgenic cells specific to hen egg lysozyme are adoptively transferred into recipients that express hen egg lysozyme in the lens of the eye. The transferred naive T cells induce ocular inflammation only in recipients treated with TLR ligands. Treatment with LPS preferentially stimulated IL-17 production, whereas CpG oligodeoxynucleotide and polyinosinic:polycytidylic acid primarily stimulated Th1 cells. Peptidoglycan stimulated the two Th subpopulations equally. The preferential induction of Th1 or Th17 by the four ligands was detected in the spleen (where a major portion of the adoptively transferred cells homed) and in the eyes, where activated Th cells initiate inflammation. Analysis of the cytokines present in recipient mice suggests that Th1 induction is elicited by IL-12 and/or IFN-α, whereas Th17 generation is preferentially mediated by IL-6. Importantly, we show in this article that treatment with LPS selectively promoted in the recipient mice the generation of IL-6-producing activated B cells. An inverse correlation was found between the level of regulatory T cells and severity of inflammation induced by the donor cells. Taken together, our data show that specific TLR ligands differentially activate the immune system as evidenced by the generation of distinct Th phenotypes from naive CD4 cells.


Subject(s)
Autoimmune Diseases/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Toll-Like Receptors/metabolism , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Chickens , Ligands , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/genetics , Peptidoglycan/metabolism , Poly I-C/metabolism , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/metabolism , Th17 Cells/pathology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Toll-Like Receptors/physiology
18.
J Immunol ; 191(2): 865-74, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23776172

ABSTRACT

Alarmins are a group of structurally diverse host defense antimicrobial peptides that are important immune activators. In this article, we present a novel role for two potent alarmins, human ß-defensin 2 and 3 (HBD2 and 3), in promoting IFN-α production by human plasmacytoid dendritic cells. We demonstrate that HBD2 and 3 activate pDCs by enhancing the intracellular uptake of CpG and self DNA and promote DNA-induced IFN-α production in a TLR9-dependent manner. Both CpG and host DNA form aggregates that resemble DNA nets when combined with HBD2 and 3. Isothermal titration calorimetry studies to elucidate the nature of HBD3/CpG complexes demonstrate involvement of enthalpy-driven interactions, in addition to hydrophobic interactions, with the formation of complexes at a molar ratio of 2:1 defensin/CpG. The i.v. administration of HBD3/CpG complexes induced proinflammatory cytokines like IL-12, IFN-γ, IL-6, IFN-α, and IL-10 in serum, associated with an increased recruitment of APCs in the spleen. Subcutaneous injections of these complexes showed enhanced infiltration of inflammatory cells at the injection site, indicating a potential pathophysiological role for alarmin/DNA complexes in contributing to inflammation. Intraperitoneal immunization of HBD3/CpG complexes with OVA enhanced both cellular and humoral responses to OVA, compared with OVA/HBD3 or OVA/CPG alone, indicative of a much more potent adjuvant effect of the HBD3/CpG complexes. Thus, the ability of defensins to enhance cellular uptake of nucleic acids can lead to improved vaccine formulations by promoting their uptake by various cells, resulting in an enhanced immune response.


Subject(s)
Dendritic Cells/metabolism , Inflammation/immunology , Interferon-alpha/biosynthesis , beta-Defensins/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/immunology , Biological Transport , Cells, Cultured , CpG Islands , DNA/metabolism , Dendritic Cells/immunology , Female , Humans , Hydrophobic and Hydrophilic Interactions , Interferon-alpha/blood , Interferon-gamma/blood , Interleukin-12/blood , Interleukin-6/blood , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Toll-Like Receptor 9/metabolism
19.
Carcinogenesis ; 35(5): 1078-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24403310

ABSTRACT

Silicosis is an inflammatory lung disease induced by the inhalation of silica-containing dust particles. There is conflicting data on whether patients with silicosis are more susceptible to lung cancer induced by cigarette smoke. To examine this issue experimentally, a model was developed in which one of the most abundant and potent carcinogens present in cigarette smoke [4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone (NNK)] was administered to mice at the peak of silica-induced pulmonary inflammation. Results show that the incidence of lung tumors in silicotic mice treated with NNK was significantly increased compared with mice exposed to silica or NNK alone. Synthetic oligonucleotides (ODN) containing repetitive TTAGGG motifs can block pathologic inflammation. We therefore examined whether treatment with these suppressive (Sup) ODN could block silica-induced pulmonary inflammation and thereby reduce susceptibility to lung cancer. Results show that Sup (but not control) ODN inhibit pulmonary fibrosis and other inflammatory manifestations of chronic silicosis. Of greater import, Sup ODN reduced lung tumor incidence and multiplicity in silicotic mice exposed to NNK. These findings establish an experimental model for examining the role of silicotic inflammation in cancer susceptibility and demonstrate that Sup ODN represent a novel therapy for chronic silicosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Disease Susceptibility , Lung Neoplasms/etiology , Lung Neoplasms/prevention & control , Oligodeoxyribonucleotides/pharmacology , Silicosis/complications , Animals , Anti-Inflammatory Agents/administration & dosage , Biomarkers , Disease Models, Animal , Female , Gene Expression , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lung Neoplasms/pathology , Mice , Nitrosamines/adverse effects , Oligodeoxyribonucleotides/administration & dosage , Pneumonia/complications , Pneumonia/drug therapy , Pneumonia/etiology , Pneumonia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Silicon Dioxide/adverse effects , Silicosis/pathology
20.
Eur J Immunol ; 43(7): 1896-906, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23616277

ABSTRACT

Synthetic oligonucleotides (ODN) expressing CpG motifs mimic the ability of bacterial DNA to trigger the innate immune system via TLR9. Plasmacytoid dendritic cells (pDCs) make a critical contribution to the ensuing immune response. This work examines the induction of antiviral (IFN-ß) and pro-inflammatory (IL-6) cytokines by CpG-stimulated human pDCs and the human CAL-1 pDC cell line. Results show that interferon regulatory factor-5 (IRF-5) and NF-κB p50 are key co-regulators of IFN-ß and IL-6 expression following TLR9-mediated activation of human pDCs. The nuclear accumulation of IRF-1 was also observed, but this was a late event that was dependant on type 1 IFN and unrelated to the initiation of gene expression. IRF-8 was identified as a novel negative regulator of gene activation in CpG-stimulated pDCs. As variants of IRF-5 and IRF-8 were recently found to correlate with susceptibility to certain autoimmune diseases, these findings are relevant to our understanding of the pharmacologic effects of "K" ODN and the role of TLR9 ligation under physiologic, pathologic, and therapeutic conditions.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factors/immunology , Interferon-beta/biosynthesis , Interleukin-6/biosynthesis , NF-kappa B p50 Subunit/immunology , Cell Line , Dendritic Cells/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation/immunology , Humans , Immunoblotting , Immunoprecipitation , Interferon Regulatory Factors/metabolism , Interferon-beta/immunology , Interleukin-6/immunology , NF-kappa B p50 Subunit/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 9/immunology
SELECTION OF CITATIONS
SEARCH DETAIL