Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(23)2021 Nov 28.
Article in English | MEDLINE | ID: mdl-34884674

ABSTRACT

Acid sphingomyelinase deficiency (ASMD) is a lysosomal storage disease caused by deficient activity of acid sphingomyelinase (ASM) enzyme, leading to the accumulation of varying degrees of sphingomyelin. Lipid storage leads to foam cell infiltration in tissues, and clinical features including hepatosplenomegaly, pulmonary insufficiency and in some cases central nervous system involvement. ASM enzyme replacement therapy is currently in clinical trial being the first treatment addressing the underlying pathology of the disease. Therefore, presently, it is critical to better comprehend ASMD to improve its diagnose and monitoring. Lung disease, including recurrent pulmonary infections, are common in ASMD patients. Along with lung disease, several immune system alterations have been described both in patients and in ASMD animal models, thus highlighting the role of ASM enzyme in the immune system. In this review, we summarized the pivotal roles of ASM in several immune system cells namely on macrophages, Natural Killer (NK) cells, NKT cells, B cells and T cells. In addition, an overview of diagnose, monitoring and treatment of ASMD is provided highlighting the new enzyme replacement therapy available.


Subject(s)
Lysosomal Storage Diseases/immunology , Sphingomyelin Phosphodiesterase/deficiency , Animals , Enzyme Replacement Therapy , Humans , Lung Diseases/enzymology , Lysosomal Storage Diseases/diagnosis , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/immunology
2.
FEBS Lett ; 595(12): 1622-1638, 2021 06.
Article in English | MEDLINE | ID: mdl-33960414

ABSTRACT

Ixodes scapularis ticks feed on humans and other vertebrate hosts and transmit several pathogens of public health concern. Tick saliva is a complex mixture of bioactive proteins, lipids and immunomodulators, such as I. scapularis sphingomyelinase (IsSMase)-like protein, an ortholog of dermonecrotoxin SMase D found in the venom of Loxosceles spp. of spiders. IsSMase modulates the host immune response towards Th2, which suppresses Th1-mediated cytokines to facilitate pathogen transmission. Arboviruses utilize exosomes for their transmission from tick to the vertebrate host, and exosomes derived from tick saliva/salivary glands suppress C-X-C motif chemokine ligand 12 and interleukin-8 immune response(s) in human skin to delay wound healing and repair processes. IsSMase affects also viral replication and exosome biogenesis, thereby inhibiting tick-to-vertebrate host transmission of pathogenic exosomes. In this review, we elaborate on exosomes and their biogenesis as potential candidates for developing novel control measure(s) to combat tick-borne diseases. Such targets could help with the development of an efficient anti-tick vaccine for preventing the transmission of tick-borne pathogens.


Subject(s)
Arbovirus Infections , Arboviruses/immunology , Arthropod Proteins/immunology , Immunologic Factors/immunology , Ixodes , Sphingomyelin Phosphodiesterase/immunology , Animals , Arbovirus Infections/immunology , Arbovirus Infections/prevention & control , Arbovirus Infections/transmission , Humans , Ixodes/immunology , Ixodes/virology , Salivary Glands/immunology , Salivary Glands/virology , Th1 Cells/immunology , Th2 Cells/immunology
3.
J Clin Invest ; 131(10)2021 05 17.
Article in English | MEDLINE | ID: mdl-33830943

ABSTRACT

A complete carcinogen, ultraviolet B (UVB) radiation (290-320 nm), is the major cause of skin cancer. UVB-induced systemic immunosuppression that contributes to photocarcinogenesis is due to the glycerophosphocholine-derived lipid mediator platelet-activating factor (PAF). A major question in photobiology is how UVB radiation, which only absorbs appreciably in the epidermal layers of skin, can generate systemic effects. UVB exposure and PAF receptor (PAFR) activation in keratinocytes induce the release of large numbers of microvesicle particles (MVPs; extracellular vesicles ranging from 100 to 1000 nm in size). MVPs released from skin keratinocytes in vitro in response to UVB (UVB-MVPs) are dependent on the keratinocyte PAFR. Here, we used both pharmacologic and genetic approaches in cells and mice to show that both the PAFR and enzyme acid sphingomyelinase (aSMase) were necessary for UVB-MVP generation. Our discovery that the calcium-sensing receptor is a keratinocyte-selective MVP marker allowed us to determine that UVB-MVPs leaving the keratinocyte can be found systemically in mice and humans following UVB exposure. Moreover, we found that UVB-MVPs contained bioactive contents including PAFR agonists that allowed them to serve as effectors for UVB downstream effects, in particular UVB-mediated systemic immunosuppression.


Subject(s)
Cell-Derived Microparticles/immunology , Immune Tolerance/radiation effects , Keratinocytes/immunology , Ultraviolet Rays , Animals , Cell Line , Cell-Derived Microparticles/genetics , Female , Humans , Mice , Mice, Knockout , Platelet Activating Factor/genetics , Platelet Activating Factor/immunology , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/immunology
4.
Am J Respir Cell Mol Biol ; 64(5): 629-640, 2021 05.
Article in English | MEDLINE | ID: mdl-33662226

ABSTRACT

Deficiency of ASM (acid sphingomyelinase) causes the lysosomal storage Niemann-Pick disease (NPD). Patients with NPD type B may develop progressive interstitial lung disease with frequent respiratory infections. Although several investigations using the ASM-deficient (ASMKO) mouse NPD model revealed inflammation and foamy macrophages, there is little insight into the pathogenesis of NPD-associated lung disease. Using ASMKO mice, we report that ASM deficiency is associated with a complex inflammatory phenotype characterized by marked accumulation of monocyte-derived CD11b+ macrophages and expansion of airspace/alveolar CD11c+ CD11b- macrophages, both with increased size, granularity, and foaminess. Both the alternative and classical pathways were activated, with decreased in situ phagocytosis of opsonized (Fc-coated) targets, preserved clearance of apoptotic cells (efferocytosis), secretion of Th2 cytokines, increased CD11c+/CD11b+ cells, and more than a twofold increase in lung and plasma proinflammatory cytokines. Macrophages, neutrophils, eosinophils, and noninflammatory lung cells of ASMKO lungs also exhibited marked accumulation of chitinase-like protein Ym1/2, which formed large eosinophilic polygonal Charcot-Leyden-like crystals. In addition to providing insight into novel features of lung inflammation that may be associated with NPD, our report provides a novel connection between ASM and the development of crystal-associated lung inflammation with alterations in macrophage biology.


Subject(s)
Glycoproteins/immunology , Lysophospholipase/immunology , Macrophages, Alveolar/immunology , Macrophages/immunology , Niemann-Pick Disease, Type A/immunology , Niemann-Pick Disease, Type B/immunology , Pneumonia/immunology , Sphingomyelin Phosphodiesterase/immunology , Animals , CD11 Antigens/genetics , CD11 Antigens/immunology , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Size , Chitinases/genetics , Chitinases/immunology , Disease Models, Animal , Eosinophils/immunology , Eosinophils/pathology , Female , Gene Expression , Glycoproteins/genetics , Humans , Lectins/genetics , Lectins/immunology , Lung/immunology , Lung/pathology , Lysophospholipase/genetics , Macrophages/pathology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Niemann-Pick Disease, Type A/enzymology , Niemann-Pick Disease, Type A/genetics , Niemann-Pick Disease, Type A/pathology , Niemann-Pick Disease, Type B/enzymology , Niemann-Pick Disease, Type B/genetics , Niemann-Pick Disease, Type B/pathology , Phagocytosis , Pneumonia/enzymology , Pneumonia/genetics , Pneumonia/pathology , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Th1-Th2 Balance/genetics , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/immunology
5.
Cell Physiol Biochem ; 52(2): 280-301, 2019.
Article in English | MEDLINE | ID: mdl-30816675

ABSTRACT

Acid sphingomyelinase hydrolyzes sphingomyelin to ceramide and phosphorylcholine. Ceramide molecules spontaneously interact with each other and generate ceramide-enriched membrane domains. These ceramide-enriched domains further fuse, forming large ceramideenriched platforms that participate in the organization of receptors and in the amplification of signaling molecules. Recent studies have suggested several bacteria and bacterial toxins that stimulate the activation and the translocation of acid sphingomyelinase, which leads to the release of ceramide. The acid sphingomyelinase/ceramide system also regulates the internalization of bacteria into the host cell, the subsequent cytokine release, inflammatory response, and initiation of host cell apoptosis. In addition, ceramide has been implicated in the fusion of phagosomes and lysosomes upon bacterial infection. Thus, this system modulates the reorganization of cell membrane receptors and intracellular signaling molecules during bacteria-host interactions. The acid sphingomyelinase and ceramide system may thus serve as a novel therapeutic target for treating infections.


Subject(s)
Bacterial Infections/immunology , Bacterial Toxins/immunology , Ceramides/immunology , Signal Transduction/immunology , Sphingomyelin Phosphodiesterase/immunology , Animals , Bacterial Infections/pathology , Enzyme Activation/immunology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Lysosomes/immunology , Lysosomes/microbiology , Phagosomes/immunology , Phagosomes/microbiology
6.
Front Immunol ; 9: 2670, 2018.
Article in English | MEDLINE | ID: mdl-30515160

ABSTRACT

Betaine is a critical nutrient for mammal health, and has been found to alleviate inflammation by lowering interleukin (IL)-1ß secretion; however, the underlying mechanisms by which betaine inhibits IL-1ß secretion remain to be uncovered. In this review, we summarize the current understanding about the mechanisms of betaine in IL-1ß production and release. For IL-1ß production, betaine affects canonical and non-canonical inflammasome-mediated processing of IL-1ß through signaling pathways, such as NF-κB, NLRP3 and caspase-8/11. For IL-1ß release, betaine inhibits IL-1ß release through blocking the exocytosis of IL-1ß-containing secretory lysosomes, reducing the shedding of IL-1ß-containing plasma membrane microvesicles, suppressing the exocytosis of IL-1ß-containing exosomes, and attenuating the passive efflux of IL-1ß across hyperpermeable plasma membrane during pyroptotic cell death, which are associated with ERK1/2/PLA2 and caspase-8/A-SMase signaling pathways. Collectively, this review highlights the anti-inflammatory property of betaine by inhibiting the production and release of IL-1ß, and indicates the potential application of betaine supplementation as an adjuvant therapy in various inflammatory diseases associating with IL-1ß secretion.


Subject(s)
Betaine/pharmacology , Exocytosis/drug effects , MAP Kinase Signaling System/drug effects , Animals , Caspase 8/immunology , Exocytosis/immunology , Humans , Interleukin-1beta/immunology , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/immunology , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Sphingomyelin Phosphodiesterase/immunology
7.
Cell Physiol Biochem ; 51(4): 1815-1829, 2018.
Article in English | MEDLINE | ID: mdl-30504725

ABSTRACT

BACKGROUND/AIMS: Mycobacteria-induced diseases, especially tuberculosis, cause more than 1 million deaths each year, which is higher than any other single bacterial pathogen. Neutral sphingomyelinase 2 (Nsm2) has been implied in many physiological processes and diseases, but the role of Nsm2 in pathogen-host interactions and mycobacterial infections has barely been studied. METHODS: We investigated the role of the Nsm2/ceramide system in systemic infection of mice and murine macrophages with Mycobacterium bovis Bacillus Calmette-Guérin (BCG) as a model for mycobacterial infection. For in vitro assays we isolated bone marrow-derived macrophages from Wildtype mice or Nsm2-heterozygous and investigated the role of Nsm2 for macrophage migration/clustering as well as the involvement of p38 mitogen-activated protein kinases (p38K), c-Jun N-terminal kinase (JNK), ß1-integrin and Rac1 activity by Western blot and microscopic studies. For in vivo assays we injected mice intravenously with BCG and analyzed infected tissues for the role of Nsm2-mediated activation of ß1-integrin in granuloma formation and bacterial burden. RESULTS: Our results reveal that BCG infection of macrophages results in rapid stimulation of Nsm2. Genetic and pharmacological studies demonstrate that Nsm2 stimulates a signaling cascade via p38K and JNK to an activation of surface ß1-integrin and Rac1 that leads to the formation of granuloma-like macrophages clusters in vitro and granuloma in vivo. Heterozygosity of Nsm2 in macrophages or antibody-mediated neutralization of active b1-integrin reduced macrophage clusters in vitro and granuloma formation in vivo. Most importantly, Nsm2 heterozygosity or treatment with neutralizing antibodies against ß1-integrin protected mice from systemic BCG infections and chronic infections of the liver and spleen. CONCLUSION: The findings indicate that the Nsm2/ ceramide system plays an important role in systemic infection of mice with mycobacteria by regulating a signaling cascade via p38K, JNK, b1-integrin and Rac1.


Subject(s)
Integrin beta1/immunology , Mycobacterium bovis/immunology , Signal Transduction , Sphingomyelin Phosphodiesterase/immunology , Tuberculosis/veterinary , Animals , Ceramides/immunology , Granuloma/immunology , Granuloma/microbiology , Granuloma/pathology , Granuloma/veterinary , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology
8.
Front Immunol ; 9: 1186, 2018.
Article in English | MEDLINE | ID: mdl-29896202

ABSTRACT

Tumor microenvironment is fundamental for cancer progression and chemoresistance. Among stromal cells tumor-associated macrophages (TAMs) represent the largest population of infiltrating inflammatory cells in malignant tumors, promoting their growth, invasion, and immune evasion. M2-polarized TAMs are endowed with the nitric oxide (NO)-generating enzyme inducible nitric oxide synthase (iNOS). NO has divergent effects on tumors, since it can either stimulate tumor cells growth or promote their death depending on the source of it; likewise the role of iNOS in cancer differs depending on the cell type. The role of NO generated by TAMs has not been investigated. Using different tumor models in vitro and in vivo we found that NO generated by iNOS of M2-polarized TAMs is able to protect tumor cells from apoptosis induced by the chemotherapeutic agent cisplatin (CDDP). Here, we demonstrate that the protective effect of NO depends on the inhibition of acid sphingomyelinase (A-SMase), which is activated by CDDP in a pathway involving the death receptor CD95. Mechanistic insights indicate that NO actions occur via generation of cyclic GMP and activation of protein kinase G (PKG), inducing phosphorylation of syntaxin 4 (synt4), a SNARE protein responsible for A-SMase trafficking and activation. Noteworthy, phosphorylation of synt4 at serine 78 by PKG is responsible for the proteasome-dependent degradation of synt4, which limits the CDDP-induced exposure of A-SMase to the plasma membrane of tumor cells. This inhibits the cytotoxic mechanism of CDDP reducing A-SMase-triggered apoptosis. This is the first demonstration that endogenous NO system is a key mechanism through which TAMs protect tumor cells from chemotherapeutic drug-induced apoptosis. The identification of the pathway responsible for A-SMase activity downregulation in tumors leading to chemoresistance warrants further investigations as a means to identify new anti-cancer molecules capable of specifically inhibiting synt4 degradation.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/immunology , Glioma/immunology , Macrophages/immunology , Neoplasm Proteins/immunology , Nitric Oxide/immunology , Qa-SNARE Proteins/immunology , Sphingomyelin Phosphodiesterase/immunology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , Humans , Macrophages/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Nitric Oxide/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Qa-SNARE Proteins/genetics , Sphingomyelin Phosphodiesterase/genetics
9.
Front Immunol ; 9: 815, 2018.
Article in English | MEDLINE | ID: mdl-29720981

ABSTRACT

By promoting ceramide release at the cytosolic membrane leaflet, the neutral sphingomyelinase 2 (NSM) is capable of organizing receptor and signalosome segregation. Its role in T cell receptor (TCR) signaling remained so far unknown. We now show that TCR-driven NSM activation is dispensable for TCR clustering and initial phosphorylation, but of crucial importance for further signal amplification. In particular, at low doses of TCR stimulatory antibodies, NSM is required for Ca2+ mobilization and T cell proliferation. NSM-deficient T cells lack sustained CD3ζ and ZAP-70 phosphorylation and are unable to polarize and stabilize their microtubular system. We identified PKCζ as the key NSM downstream effector in this second wave of TCR signaling supporting dynamics of microtubule-organizing center (MTOC). Ceramide supplementation rescued PKCζ membrane recruitment and MTOC translocation in NSM-deficient cells. These findings identify the NSM as essential in TCR signaling when dynamic cytoskeletal reorganization promotes continued lateral and vertical supply of TCR signaling components: CD3ζ, Zap70, and PKCζ, and functional immune synapses are organized and stabilized via MTOC polarization.


Subject(s)
Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/physiology , Sphingomyelin Phosphodiesterase/immunology , T-Lymphocytes/immunology , CD3 Complex/immunology , Cell Differentiation , Ceramides/pharmacology , Humans , Jurkat Cells , Lymphocyte Activation , Microtubule-Organizing Center/immunology , Phosphorylation , Signal Transduction , T-Lymphocytes/drug effects , ZAP-70 Protein-Tyrosine Kinase/metabolism
10.
Front Immunol ; 9: 204, 2018.
Article in English | MEDLINE | ID: mdl-29467770

ABSTRACT

Microvesicles (MVs) are membrane particles of 200-500 nm released by all cell types constitutively. MVs of myeloid origin are found increased in the cerebrospinal fluid (CSF) of patients suffering from neuroinflammatory disorders, although the factors triggering their production have never been defined. Here, we report that both pro- and anti-inflammatory cytokines, specifically interferon-γ and interleukin-4, are equally able to stimulate the production of MVs from microglia cells and monocytes. Additionally, we found this process to be independent from the best characterized molecular pathway so far described for membrane shedding, which is centered on the purinergic receptor P2X7, whose activation by high concentrations of extracellular ATP (exATP) results in membrane blebbing operated by the secreted enzyme acid sphingomyelinase (ASMase). Moreover, a potent inhibitor of ASMase, injected in a mouse model of multiple sclerosis, failed to reduce the number of MVs in their CSF. This suggests that cytokines, rather than exATP, may exert a long-term control of MV production in the context of chronic inflammation, where both pro- and anti-inflammatory factors play coordinated roles.


Subject(s)
Cell-Derived Microparticles/immunology , Cytokines/immunology , Multiple Sclerosis/immunology , Myeloid Cells/immunology , Signal Transduction/immunology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Line , Cell-Derived Microparticles/drug effects , Cell-Derived Microparticles/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Imipramine/pharmacology , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Myeloid Cells/metabolism , Receptors, Purinergic P2X7/immunology , Receptors, Purinergic P2X7/metabolism , Retinoids/pharmacology , Signal Transduction/drug effects , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/immunology , Sphingomyelin Phosphodiesterase/metabolism
11.
Eur J Med Chem ; 153: 73-104, 2018 Jun 10.
Article in English | MEDLINE | ID: mdl-29031494

ABSTRACT

Ceramide generation is involved in signal transduction of cellular stress response, in particular during stress-induced apoptosis in response to stimuli such as minimally modified Low-density lipoproteins, TNFalpha and exogenous C6-ceramide. In this paper we describe 48 diverse synthetic products and evaluate their lysosomotropic and acid sphingomyelinase inhibiting activities in macrophages. A stimuli-induced increase of C16-ceramide in macrophages can be almost completely suppressed by representative compound NB 06 providing an effective protection of macrophages against apoptosis. Compounds like NB 06 thus offer highly interesting fields of application besides prevention of apoptosis of macrophages in atherosclerotic plaques in vessel walls. Most importantly, they can be used for blocking pH-dependent lysosomal processes and enzymes in general as well as for analyzing lysosomal dependent cellular signaling. Modulation of gene expression of several prominent inflammatory messengers IL1B, IL6, IL23A, CCL4 and CCL20 further indicate potentially beneficial effects in the field of (systemic) infections involving bacterial endotoxins like LPS or infections with influenza A virus.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Lysosomes/drug effects , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Cell Line , Cells, Cultured , Ceramides/immunology , Humans , Inflammation/drug therapy , Inflammation/immunology , Lipopolysaccharides/immunology , Lysosomes/immunology , Macrophages/drug effects , Macrophages/immunology , Signal Transduction/drug effects , Sphingomyelin Phosphodiesterase/immunology
12.
Biochem Biophys Res Commun ; 482(4): 645-650, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27865842

ABSTRACT

Niemann-Pick disease (NPD) type B is a rare autosomal recessive disease characterized by variable levels of impairment in sphingomyelin phosphodiesterase 1 (SMPD1) activity. Lung involvement is the most important prognostic factor in NPD-B, with recurrent respiratory infections starting in infancy being the major cause of morbidity and mortality. We hypothesized that decreased SMPD1 activity impaired airway epithelium host defense response. SMPD1 activity was reduced using inducible shRNA. Surprisingly, decreasing SMPD1 activity by 50%, resulted in increased neutrophil recruitment, both at baseline and in response to bacterial stimulation. This correlated with elevated levels of cytokine mRNA shown to contribute to neutrophil recruitment in unstimulated (e.g. IL-8 and GRO-α) and infected cells (e.g. IL-8, GRO-α, GM-CSF and CCL20). Instead of preventing the host defence responses, decreased SMPD1 activity results in an inflammatory response even in the absence of infection. Moreover, decreasing SMPD1 activity resulted in a pro-oxidative shift. Accordingly, expression of an inactive mutant, SMPD1[L225P] but not the WT enzyme increased activation of the antioxidant transcription factor NRF2. Therefore, decreasing SMPD1 activity by 50% in airway epithelial cells, the equivalent of the loss of one allele, results in the accumulation of oxidants that activates NRF2 and a concomitant increased cytokine production as well as neutrophil recruitment. This can result in a chronic inflammatory state that impairs host defence similar to scenarios observe in other chronic inflammatory lung disease such as Chronic Obstructive Pulmonary Disease or Cystic Fibrosis.


Subject(s)
Cytokines/immunology , NF-E2-Related Factor 2/immunology , Neutrophil Infiltration , Niemann-Pick Disease, Type B/immunology , Respiratory Mucosa/immunology , Sphingomyelin Phosphodiesterase/immunology , Bronchi/cytology , Bronchi/immunology , Bronchi/pathology , Cell Line , Humans , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Reactive Oxygen Species/immunology , Respiratory Mucosa/cytology , Respiratory Mucosa/pathology
13.
Cell Physiol Biochem ; 39(3): 985-95, 2016.
Article in English | MEDLINE | ID: mdl-27512981

ABSTRACT

BACKGROUND/AIMS: Regulatory T cell (Treg) is required for the maintenance of tolerance to various tissue antigens and to protect the host from autoimmune disorders. However, Treg may, indirectly, support cancer progression and bacterial infections. Therefore, a balance of Treg function is pivotal for adequate immune responses. Acid sphingomyelinase (ASM) is a rate limiting enzyme involved in the production of ceramide by breaking down sphingomyelin. Previous studies in T-cells have suggested that ASM is involved in CD28 signalling, T lymphocyte granule secretion, degranulation, and vesicle shedding similar to the formation of phosphatidylserine-exposing microparticles from glial cells. However, whether ASM affects the development of Treg has not yet been described. METHODS: Splenocytes, isolated Naive T lymphocytes and cultured T cells were characterized for various immune T cell markers by flow cytometery. Cell proliferation was measured by Carboxyfluorescein succinimidyl ester (CFSE) dye, cell cycle analysis by Propidium Iodide (PI), mRNA transcripts by q-RT PCR and protein expression by Western Blotting respectively. RESULTS: ASM deficient mice have higher number of Treg compared with littermate control mice. In vitro induction of ASM deficient T cells in the presence of TGF-ß and IL-2 lead to a significantly higher number of Foxp3+ induced Treg (iTreg) compared with control T-cells. Further, ASM deficient iTreg has less AKT (serine 473) phosphorylation and Rictor levels compared with control iTreg. Ceramide C6 led to significant reduction of iTreg in both ASM deficient and WT mice. The reduction in iTreg leads to induction of IL-1ß, IL-6 and IL-17 but not IFN-γ mRNA levels. CONCLUSION: ASM is a negative regulator of natural and iTreg.


Subject(s)
Cell Differentiation/immunology , RNA, Messenger/genetics , Sphingomyelin Phosphodiesterase/genetics , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Differentiation/drug effects , Cell Proliferation , Cell Separation , Ceramides/immunology , Ceramides/metabolism , Female , Fluoresceins , Fluorescent Dyes , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression , Gene Expression Regulation , Immunity, Innate , Interleukin-2/pharmacology , Interleukins/genetics , Interleukins/immunology , Male , Mice , Mice, Knockout , Phosphorylation , Primary Cell Culture , Propidium , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , RNA, Messenger/immunology , Rapamycin-Insensitive Companion of mTOR Protein , Signal Transduction , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/immunology , Spleen/drug effects , Spleen/pathology , Succinimides , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/pharmacology
14.
Immunol Res ; 64(4): 869-86, 2016 08.
Article in English | MEDLINE | ID: mdl-26798039

ABSTRACT

Protein kinase C theta (PKCθ) is a novel, calcium-independent member of the PKC family of kinases that was identified as a central player in T cell signaling and proliferation. Upon T cell activation by antigen-presenting cells, PKCθ gets phosphorylated and activated prior to its translocation to the immunological synapse where it couples with downstream effectors. PKCθ may be regulated by ceramide, a crucial sphingolipid that is known to promote differentiation, growth arrest, and apoptosis. To further investigate the mechanism, we stimulated human Jurkat T cells with either PMA or anti-CD3/anti-CD28 antibodies following induction of ceramide accumulation by adding exogenous ceramide, bacterial sphingomyelinase, or Fas ligation. Our results suggest that ceramide regulates the PKCθ pathway through preventing its critical threonine 538 (Thr538) phosphorylation and subsequent activation, thereby inhibiting the kinase's translocation to lipid rafts. Moreover, this inhibition is not likely to be a generic effect of ceramide on membrane reorganization. Other lipids, namely dihydroceramide, palmitate, and sphingosine, did not produce similar effects on PKCθ. Addition of the phosphatase inhibitors okadaic acid and calyculin A reversed the inhibition exerted by ceramide, and this suggests involvement of a ceramide-activated protein phosphatase. Such previously undescribed mechanism of regulation of PKCθ raises the possibility that ceramide, or one of its derivatives, and may prove valuable in novel therapeutic approaches for disorders involving autoimmunity or excessive inflammation-where PKCθ plays a critical role.


Subject(s)
Ceramides/metabolism , Membrane Microdomains/metabolism , Protein Kinase C-theta/metabolism , T-Lymphocytes/physiology , Cell Proliferation , Humans , Jurkat Cells , Lymphocyte Activation , Marine Toxins , Okadaic Acid/pharmacology , Oxazoles/pharmacology , Phosphorylation/drug effects , Protein Transport , Signal Transduction , Sphingomyelin Phosphodiesterase/immunology , Tetradecanoylphorbol Acetate/immunology , fas Receptor/metabolism
15.
J Immunol ; 196(1): 72-9, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26597010

ABSTRACT

Multiple sclerosis (MS) is a chronic demyelinating disorder of the CNS characterized by immune cell infiltration across the brain vasculature into the brain, a process not yet fully understood. We previously demonstrated that the sphingolipid metabolism is altered in MS lesions. In particular, acid sphingomyelinase (ASM), a critical enzyme in the production of the bioactive lipid ceramide, is involved in the pathogenesis of MS; however, its role in the brain vasculature remains unknown. Transmigration of T lymphocytes is highly dependent on adhesion molecules in the vasculature such as intercellular adhesion molecule-1 (ICAM-1). In this article, we hypothesize that ASM controls T cell migration by regulating ICAM-1 function. To study the role of endothelial ASM in transmigration, we generated brain endothelial cells lacking ASM activity using a lentiviral shRNA approach. Interestingly, although ICAM-1 expression was increased in cells lacking ASM activity, we measured a significant decrease in T lymphocyte adhesion and consequently transmigration both in static and under flow conditions. As an underlying mechanism, we revealed that upon lack of endothelial ASM activity, the phosphorylation of ezrin was perturbed as well as the interaction between filamin and ICAM-1 upon ICAM-1 clustering. Functionally this resulted in reduced microvilli formation and impaired transendothelial migration of T cells. In conclusion, in this article, we show that ASM coordinates ICAM-1 function in brain endothelial cells by regulating its interaction with filamin and phosphorylation of ezrin. The understanding of these underlying mechanisms of T lymphocyte transmigration is of great value to develop new strategies against MS lesion formation.


Subject(s)
Brain/metabolism , Intercellular Adhesion Molecule-1/metabolism , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes/immunology , Transendothelial and Transepithelial Migration/immunology , Adult , Aged , Aged, 80 and over , Brain/cytology , Brain/immunology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Line , Ceramides/metabolism , Cytoskeletal Proteins/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Filamins/metabolism , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/immunology , Male , Middle Aged , Multiple Sclerosis/immunology , Phosphorylation/genetics , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/immunology
16.
Cell Death Dis ; 6: e1828, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26203857

ABSTRACT

Acid sphingomyelinase (ASM), a lipid hydrolase enzyme, has the potential to modulate various cellular activation responses via the generation of ceramide and by interaction with cellular receptors. We have hypothesized that ASM modulates CD4(+) T-cell receptor activation and impacts immune responses. We first observed interactions of ASM with the intracellular domains of both CD3 and CD28. ASM further mediates T-cell proliferation after anti-CD3/CD28 antibody stimulation and alters CD4(+) T-cell activation signals by generating ceramide. We noted that various pharmacological inhibitors of ASM or knockdown of ASM using small hairpin RNA inhibit CD3/CD28-mediated CD4(+) T-cell proliferation and activation. Furthermore, such blockade of ASM bioactivity by biochemical inhibitors and/or molecular-targeted knockdown of ASM broadly abrogate T-helper cell responses. In conclusion, we detail immune, pivotal roles of ASM in adaptive immune T-cell responses, and propose that these pathways might provide novel targets for the therapy of autoimmune and inflammatory diseases.


Subject(s)
Adaptive Immunity/genetics , Lymphocyte Activation/drug effects , Sphingomyelin Phosphodiesterase/immunology , T-Lymphocytes, Helper-Inducer/immunology , Amitriptyline/pharmacology , Antibodies/pharmacology , CD28 Antigens/agonists , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Carnitine/pharmacology , Cell Proliferation/drug effects , Ceramides/biosynthesis , Ceramides/immunology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Humans , Imipramine/pharmacology , Primary Cell Culture , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/genetics , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects
17.
Vet Immunol Immunopathol ; 162(3-4): 96-107, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25454469

ABSTRACT

Staphylococcus aureus is the most frequently isolated pathogen from bovine intramammary infections worldwide. Commercially available vaccines for mastitis control are composed either of S. aureus lysates or inactivated whole-cells formulated with traditional adjuvants. We recently showed the ability of a S. aureus CP5 lysate vaccine adjuvanted with Iscom Matrix to generate a longer lasting specific antibody response in blood and milk, with improved opsonic capacity, compared with a S. aureus CP5 whole-cell formulation. The aim of the present study was to obtain an experimental immunogen composed of lysed cells of a CP5 S. aureus strain supplemented with recombinant clumping factor A, fibronectin binding protein A and ß-toxin formulated with Iscom Matrix, characterize the immune response generated when immunizing pregnant heifers and assess the functional role of antibodies raised against this immunogen in experimental models. Both a lysate vaccine and a lysate+recombinant antigens vaccine elicited antibodies that promoted neutrophil phagocytosis and inhibited internalization into mammary epithelial cells, in vitro. Incorporation of defined antigenic molecules to the lysate formulation elicited a strong specific humoral immune response against both lysate and recombinant antigens and was associated with higher expression of regulatory and pro-inflammatory cytokines. In addition, antibodies were efficient for blocking S. aureus binding to bovine fibrinogen and fibronectin, and neutralizing ß-toxin effect in vitro, placing these antigens as candidates to be included in a formulation directed to prevent staphylococcal bovine mastitis.


Subject(s)
Immunization/veterinary , Mastitis, Bovine/microbiology , Staphylococcal Infections/veterinary , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Vaccines, Synthetic/immunology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cattle , Cell Line , Coagulase/genetics , Coagulase/immunology , Cytokines/blood , Female , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , ISCOMs/pharmacology , Immunization/methods , Mastitis, Bovine/immunology , Mastitis, Bovine/prevention & control , Milk/microbiology , Pregnancy , Random Allocation , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/administration & dosage , Statistics, Nonparametric , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/standards
18.
J Immunol ; 193(7): 3366-77, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25172498

ABSTRACT

CD39 (ENTPD1) is expressed by subsets of pathogenic human CD4(+) T cells, such as Th17 cells. These Th17 cells are considered important in intestinal inflammation, such as seen in Crohn's disease (CD). Recently, CD161 (NKR-P1A) was shown to be a phenotypic marker of human Th17 cells. In this study, we report that coexpression of CD161 and CD39 not only identifies these cells but also promotes Th17 generation. We note that human CD4(+)CD39(+)CD161(+) T cells can be induced under stimulatory conditions that promote Th17 in vitro. Furthermore, CD4(+)CD39(+)CD161(+) cells purified from blood and intestinal tissues, from both healthy controls and patients with CD, are of the Th17 phenotype and exhibit proinflammatory functions. CD39 is coexpressed with CD161, and this association augments acid sphingomyelinase (ASM) activity upon stimulation of CD4(+) T cells. These pathways regulate mammalian target of rapamycin and STAT3 signaling to drive the Th17 phenotype. Inhibition of ASM activity by pharmacological blockers or knockdown of ASM abrogates STAT3 signaling, thereby limiting IL-17 production in CD4(+) T cells obtained from both controls and patients with active CD. Increased levels of CD39(+)CD161(+) CD4(+) T cells in blood or lamina propria are noted in patients with CD, and levels directly correlate with clinical disease activity. Hence, coexpression of CD39 and CD161 by CD4(+) T cells might serve as a biomarker to monitor Th17 responsiveness. Collectively, CD39 and CD161 modulate human Th17 responses in CD through alterations in purinergic nucleotide-mediated responses and ASM catalytic bioactivity, respectively.


Subject(s)
Antigens, CD/immunology , Apyrase/immunology , Crohn Disease/immunology , Mucous Membrane/immunology , NK Cell Lectin-Like Receptor Subfamily B/immunology , Th17 Cells/immunology , Adult , Aged , Biomarkers , Crohn Disease/pathology , Female , Humans , Inflammation/immunology , Inflammation/pathology , Interleukin-17/immunology , Male , Middle Aged , Mucous Membrane/pathology , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Sphingomyelin Phosphodiesterase/immunology , Th17 Cells/pathology
19.
Vaccine ; 31(25): 2749-55, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23664158

ABSTRACT

A chimeric protein was constructed expressing three epitopes of LiD1, a dermonecrotic toxin from the venom of Loxosceles intermedia spider. This species is responsible for a large number of accidents involving spiders in Brazil. We demonstrated that the chimeric protein (rCpLi) generated is atoxic and that antibodies previously developed in rabbits against synthetic epitopes reactive with rCpLi in ELISA and immunoblot assays. The antibody response in rabbits against the rCpLi was evaluated by ELISA and we have detected an antibody response in all immunized animals. Overlapping peptides covering the amino acid sequence of the rCpLi were synthesized on a cellulose membrane, and their recognition by rabbit anti-rCpLi serum assessed. Three different antigenic regions were identified. The percentage of inhibition of the dermonecrotic, hemorrhagic and edematogenic activities caused by the recombinant protein LiD1r in naïve rabbits was assessed by pre-incubation with anti-rCpLi antibodies. Anti-rCpLi induced good dermonecrotic and hemorrhagic protection. The levels of protection were similar to the antiboides anti-LiD1r. In summary, we have developed a polyepitope recombinant chimeric protein capable of inducing multiple responses of neutralizing antibodies in a rabbit model. This engineered protein may be a promising candidate for therapeutic serum development or vaccination.


Subject(s)
Recombinant Fusion Proteins/chemistry , Spider Venoms/chemistry , Animals , Antibodies, Neutralizing/immunology , Antivenins/immunology , Edema/immunology , Edema/prevention & control , Epitopes, B-Lymphocyte/genetics , Hemorrhage/immunology , Hemorrhage/prevention & control , Necrosis/immunology , Necrosis/prevention & control , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Skin/immunology , Skin/pathology , Sphingomyelin Phosphodiesterase/chemistry , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/immunology , Spider Venoms/genetics , Spider Venoms/immunology , Spiders
20.
J Allergy Clin Immunol ; 131(2): 421-7.e1-2, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23246020

ABSTRACT

BACKGROUND: The skin of patients with atopic dermatitis (AD) has defects in keratinocyte differentiation, particularly in expression of the epidermal barrier protein filaggrin. AD skin lesions are often exacerbated by Staphylococcus aureus-mediated secretion of the virulence factor α-toxin. It is unknown whether lack of keratinocyte differentiation predisposes to enhanced lethality from staphylococcal toxins. OBJECTIVE: We investigated whether keratinocyte differentiation and filaggrin expression protect against cell death induced by staphylococcal α-toxin. METHODS: Filaggrin-deficient primary keratinocytes were generated through small interfering RNA gene knockdown. RNA expression was determined by using real-time PCR. Cell death was determined by using the lactate dehydrogenase assay. Keratinocyte cell survival in filaggrin-deficient (ft/ft) mouse skin biopsies was determined based on Keratin 5 staining. α-Toxin heptamer formation and acid sphingomyelinase expression were determined by means of immunoblotting. RESULTS: We found that filaggrin expression, occurring as the result of keratinocyte differentiation, significantly inhibits staphylococcal α-toxin-mediated pathogenicity. Furthermore, filaggrin plays a crucial role in protecting cells by mediating the secretion of sphingomyelinase, an enzyme that reduces the number of α-toxin binding sites on the keratinocyte surface. Finally, we determined that sphingomyelinase enzymatic activity directly prevents α-toxin binding and protects keratinocytes against α-toxin-induced cytotoxicity. CONCLUSIONS: The current study introduces the novel concept that S aureus α-toxin preferentially targets and destroys filaggrin-deficient keratinocytes. It also provides a mechanism to explain the increased propensity for S aureus-mediated exacerbation of AD skin disease.


Subject(s)
Bacterial Toxins/toxicity , Hemolysin Proteins/toxicity , Intermediate Filament Proteins/biosynthesis , Keratinocytes/drug effects , Keratinocytes/immunology , Sphingomyelin Phosphodiesterase/immunology , Sphingomyelin Phosphodiesterase/metabolism , Animals , Bacterial Toxins/immunology , Cell Death/drug effects , Cell Death/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/pathology , Filaggrin Proteins , Hemolysin Proteins/immunology , Humans , Intermediate Filament Proteins/deficiency , Intermediate Filament Proteins/immunology , Keratinocytes/cytology , Keratinocytes/enzymology , Mice , Mice, Inbred BALB C , Skin/cytology , Skin/immunology , Skin/metabolism , Skin/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...