Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Allergol Int ; 71(4): 512-519, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35718709

ABSTRACT

BACKGROUND: The mechanism of allergic reactions to COVID-19 mRNA vaccines has not been clarified. Polyethylene glycol (PEG) is a potential antigen in the components of vaccines. However, there is little evidence that allergy after COVID-19 mRNA vaccination is related to PEG. Furthermore, the role of polysorbate (PS) as an antigen has also not been clarified. The objective of this study was to investigate whether PEG and PS allergies are reasonable causes of allergic symptoms after vaccination by detecting PEG-specific and PS-specific antibodies. METHODS: Fourteen patients who developed immediate allergic reactions to BNT162b2 (Pfizer-BioNTech) or mRNA-1273 (Moderna) vaccines and nineteen healthy controls who did not present allergic symptoms were recruited. Serum PEG-specific immunoglobulin E (IgE) and immunoglobulin G (IgG) and PS-specific IgE and IgG were measured by enzyme-linked immunosorbent assay. Skin tests using PEG-2000 and PS-80 were applied to five patients and three controls. RESULTS: Serum levels of PEG-specific IgE and IgG in patients with immediate allergic reactions to the COVID-19 mRNA vaccine were higher than those in the control group. Serum levels of PS-specific IgE in patients with allergy to the vaccine were higher than those in patients of the control group. Intradermal tests using PEG verified the results for PEG-specific IgE and IgG. CONCLUSIONS: The results suggest that PEG is one of the antigens in the allergy to COVID-19 mRNA vaccines. Cross-reactivity between PEG and PS might be crucial for allergy to the vaccines. PEG-specific IgE and IgG may be useful in diagnosing allergy to COVID-19 mRNA vaccines.


Subject(s)
BNT162 Vaccine/adverse effects , COVID-19 , Hypersensitivity , COVID-19/diagnosis , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , Humans , Hypersensitivity/diagnosis , Hypersensitivity, Immediate , Immunoglobulin E/blood , Immunoglobulin G/blood , Polyethylene Glycols , Polysorbates , RNA, Messenger , Vaccines, Synthetic , mRNA Vaccines
2.
J Med Virol ; 93(7): 4549-4552, 2021 07.
Article in English | MEDLINE | ID: mdl-33739483

ABSTRACT

BACKGROUND: The gold standard for coronavirus disease (COVID-19) diagnosis has been the detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA by nucleic acid amplification testing (NAAT). On the other hand, serological testing for COVID-19 may offer advantages in detecting possibly overlooked infections by NAAT. METHODS: To evaluate seroconversion of NAAT-negative pneumonia patients, immunoglobulin M (IgM) and IgG targeting the spike protein of SARS-CoV-2 were semiquantified by an immunofluorescence assay. Seroconversion was confirmed by another serological method, targeting the nucleocapsid protein. RESULTS: Eight suspected but unconfirmed COVID-19 pneumonia patients (median age, 39 years; range, 21-55) were included. The median period between symptom onset and NAAT sample collection was 6 days (2-27 days). None of them had tested positive for SARS-CoV-2 by NAAT. In contrast, all eight patients revealed seropositivity with the two serological methods, indicating actual seroconversion against SARS-CoV-2. The median period between onset and blood sampling was 26.5 days (7-51 days). CONCLUSION: Eight patients with COVID-19 pneumonia, initially tested negative for SARS-CoV-2 by NAAT, were finally confirmed of the diagnosis by serological testing. To cover the whole spectrum of this heterogenous infectious disease, serology testing should be implemented to the multitiered diagnostic algorithm for COVID-19.


Subject(s)
Antibodies, Viral/blood , COVID-19 Serological Testing/methods , COVID-19/diagnosis , Immunoglobulin G/blood , Immunoglobulin M/blood , Adult , Coronavirus Nucleocapsid Proteins/immunology , Female , Humans , Male , Middle Aged , Phosphoproteins/immunology , SARS-CoV-2/immunology , Seroconversion , Spike Glycoprotein, Coronavirus/immunology , Young Adult
3.
Mod Rheumatol ; 31(6): 1087-1093, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33491519

ABSTRACT

OBJECTIVES: Rheumatoid arthritis (RA) in patients undergoing immunosuppressive therapy (IS) is sometimes involved with other iatrogenic immunodeficiency-associated lymphoproliferative disorders (LPD). We aimed to clarify the effects of LPD treatment on RA and the current status of RA treatment options after LPD onset and subsequent IS withdrawal. METHODS: We retrospectively analyzed data of patients who had RA with LPD and examined the relationship between LPD course and RA treatment as well as that between RA relapse and LPD treatment. RESULTS: LPD patients were categorized into two groups: patients who regressed spontaneously (n = 19) and those who needed chemotherapy (n = 12). The chemotherapy group had significantly less RA relapse than the spontaneous regression group (p = .041). RA almost relapsed early in the spontaneous regression group and needed treatment for RA. Chemotherapy with rituximab prevented long-term RA relapse, and RA did not relapse for long even after rituximab monotherapy. The total dose of rituximab in monotherapy correlated with the time to RA relapse. Six patients with RA relapse received biologics and had no LPD relapse for more than 1 year. CONCLUSIONS: Rituximab in chemotherapy for LPD may help prevent RA relapse with LPD. Large-scale studies are required in the future for verification.


Subject(s)
Arthritis, Rheumatoid , Lymphoproliferative Disorders , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/drug therapy , Humans , Iatrogenic Disease , Lymphoproliferative Disorders/drug therapy , Lymphoproliferative Disorders/etiology , Methotrexate , Recurrence , Retrospective Studies
4.
Eur Respir J ; 56(3)2020 09.
Article in English | MEDLINE | ID: mdl-32366484

ABSTRACT

It is currently not understood whether cigarette smoke exposure facilitates sensitisation to self-antigens and whether ensuing auto-reactive T cells drive chronic obstructive pulmonary disease (COPD)-associated pathologies.To address this question, mice were exposed to cigarette smoke for 2 weeks. Following a 2-week period of rest, mice were challenged intratracheally with elastin for 3 days or 1 month. Rag1-/- , Mmp12-/- , and Il17a-/- mice and neutralising antibodies against active elastin fragments were used for mechanistic investigations. Human GVAPGVGVAPGV/HLA-A*02:01 tetramer was synthesised to assess the presence of elastin-specific T cells in patients with COPD.We observed that 2 weeks of cigarette smoke exposure induced an elastin-specific T cell response that led to neutrophilic airway inflammation and mucus hyperproduction following elastin recall challenge. Repeated elastin challenge for 1 month resulted in airway remodelling, lung function decline and airspace enlargement. Elastin-specific T cell recall responses were dose dependent and memory lasted for over 6 months. Adoptive T cell transfer and studies in T cells deficient Rag1-/- mice conclusively implicated T cells in these processes. Mechanistically, cigarette smoke exposure-induced elastin-specific T cell responses were matrix metalloproteinase (MMP)12-dependent, while the ensuing immune inflammatory processes were interleukin 17A-driven. Anti-elastin antibodies and T cells specific for elastin peptides were increased in patients with COPD.These data demonstrate that MMP12-generated elastin fragments serve as a self-antigen and drive the cigarette smoke-induced autoimmune processes in mice that result in a bronchitis-like phenotype and airspace enlargement. The study provides proof of concept of cigarette smoke-induced autoimmune processes and may serve as a novel mouse model of COPD.


Subject(s)
Elastin , Pulmonary Disease, Chronic Obstructive , Animals , Autoimmunity , Disease Models, Animal , Humans , Lung , Mice , Mice, Inbred C57BL , Smoke/adverse effects , Smoking/adverse effects
5.
J Immunol ; 190(12): 5927-38, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23650617

ABSTRACT

We reported that Pla2g5-null mice lacking group V secretory phospholipase A2 (gV-sPLA2) showed reduced eosinophilic pulmonary inflammation and Th2 cytokine generation when challenged with an extract from house dust mite Dermatophagoides farinae, compared with wild-type (WT) controls. Adoptive transfer studies suggested that gV-sPLA2 in dendritic cells was necessary for sensitization of Pla2g5-null mice, but was not sufficient to induce the effector phase of pulmonary inflammation. In this study, we demonstrate that gV-sPLA2 is inducibly expressed in mouse and human macrophages (M) activated by IL-4 and is required for the acquisition of M effector functions that facilitate the effector phase of pulmonary inflammation. We demonstrate that gV-sPLA2 expression in M is sufficient for the development of pulmonary inflammation, even when inflammation is induced by intrapulmonary administration of IL-4. The concentrations of CCL22/CCL17 and effector T cell recruitment are severely impaired in Pla2g5-null mice. Intratracheal transfers of enriched CD68(+) cells isolated from the lungs of D. farinae-challenged WT donor mice induce eosinophilia, chemokine production, and recruitment of T cells into the lungs of Pla2g5-null recipients previously sensitized by WT D. farinae-loaded dendritic cells. Our studies identified a unique function of gV-sPLA2 in activation of M and in their capacity to recruit T cells to amplify the effector phase of pulmonary inflammation.


Subject(s)
Group V Phospholipases A2/immunology , Hypersensitivity/immunology , Macrophage Activation/immunology , Pneumonia/immunology , Animals , Fluorescent Antibody Technique , Group V Phospholipases A2/metabolism , Humans , Hypersensitivity/metabolism , Immunohistochemistry , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
6.
J Immunol ; 191(6): 2879-89, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23935198

ABSTRACT

Bisphosphonates (BPs) have been widely used to treat osteoporosis. They act by inhibiting farnesyl diphosphate synthase in the mevalonate pathway. This resembles the action of statins, whose immune-modulating effect has recently been highlighted. In contrast, the effect of BPs on immune responses has not been elucidated well. In this study, we examined the effect of alendronate (ALN), a nitrogen-containing BP, on allergic airway inflammation in a mouse model. BALB/c mice were sensitized twice with OVA and challenged three times with nebulized OVA to induce eosinophilic airway inflammation. ALN was administered by an intragastric tube before each inhalation. ALN strongly suppressed airway eosinophilia and Th2, as well as Th17 cytokine production in the lung. ALN also attenuated eotaxin-2 production in the lung. Immunohistochemistry demonstrated that the major cell source of eotaxin-2 was peribronchial/perivascular macrophages, and flow cytometrical studies confirmed that ALN decreased eotaxin-2 expression in these macrophages. Furthermore, ALN attenuated eotaxin-2 production from mouse pleural macrophages and human monocyte/macrophage-like THP-1 cells in vitro. These results suggest that ALN suppressed Ag-induced airway responses in the mouse model. The suppression of eotaxin-2 production from macrophages appears to be one of ALN's immunomodulatory effects, whereas the mechanism by which ALN suppressed Th2 and Th17 responses could not be fully elucidated in this study. Although a clinical study should be conducted, ALN could be a novel therapeutic option for asthma.


Subject(s)
Alendronate/pharmacology , Bone Density Conservation Agents/pharmacology , Eosinophils/immunology , Macrophages/drug effects , Pneumonia/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Animals , Chemokine CCL24/biosynthesis , Chemokine CCL24/immunology , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Eosinophils/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Pneumonia/metabolism , Real-Time Polymerase Chain Reaction , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/metabolism
7.
Biochem Biophys Res Commun ; 443(1): 266-71, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24309112

ABSTRACT

Perillyl alcohol (POH) is an isoprenoid which inhibits farnesyl transferase and geranylgeranyl transferase, key enzymes that induce conformational and functional changes in small G proteins to conduct signal production for cell proliferation. Thus, it has been tried for the treatment of cancers. However, although it affects the proliferation of immunocytes, its influence on immune responses has been examined in only a few studies. Notably, its effect on antigen-induced immune responses has not been studied. In this study, we examined whether POH suppresses Ag-induced immune responses with a mouse model of allergic airway inflammation. POH treatment of sensitized mice suppressed proliferation and cytokine production in Ag-stimulated spleen cells or CD4(+) T cells. Further, sensitized mice received aerosolized OVA to induce allergic airway inflammation, and some mice received POH treatment. POH significantly suppressed indicators of allergic airway inflammation such as airway eosinophilia. Cytokine production in thoracic lymph nodes was also significantly suppressed. These results demonstrate that POH suppresses antigen-induced immune responses in the lung. Considering that it exists naturally, POH could be a novel preventive or therapeutic option for immunologic lung disorders such as asthma with minimal side effects.


Subject(s)
Hypersensitivity/drug therapy , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Lung/drug effects , Monoterpenes/therapeutic use , Pulmonary Eosinophilia/drug therapy , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Disease Models, Animal , Hypersensitivity/immunology , Hypersensitivity/prevention & control , Immunosuppressive Agents/adverse effects , Inflammation/drug therapy , Inflammation/immunology , Inflammation/prevention & control , Lung/immunology , Male , Mice , Mice, Inbred BALB C , Monoterpenes/adverse effects , Ovalbumin/immunology , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/prevention & control
8.
J Immunol ; 186(10): 5975-82, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21471450

ABSTRACT

Allergic airway inflammation is generally considered a Th2-type immune response. Recent studies, however, demonstrated that Th17-type immune responses also play important roles in this process, especially in the pathogenesis of neutrophilic airway inflammation, a hallmark of severe asthma. We previously reported that dendritic cells release dopamine to naive CD4(+) T cells in Ag-specific cell-cell interaction, in turn inducing Th17 differentiation through dopamine D1-like receptor (D1-like-R). D1-like-R antagonist attenuates Th17-mediated diseases such as experimental autoimmune encephalomyelitis and autoimmune diabetes. However, the effect of antagonizing D1-like-R on Th17-mediated airway inflammation has yet to be studied. In this study, we examined whether D1-like-R antagonist suppresses OVA-induced neutrophilic airway inflammation in OVA TCR-transgenic DO11.10 mice and then elucidated the mechanism of action. DO11.10 mice were nebulized with OVA or PBS, and some mice received D1-like-R antagonist orally before OVA nebulization. D1-like-R antagonist significantly suppressed OVA-induced neutrophilic airway inflammation in DO11.10 mice. It also inhibited the production of IL-17 and infiltration of Th17 cells in the lung. Further, D1-like-R antagonist suppressed the production of IL-23 by lung CD11c(+) APCs. In contrast, D1-like-R antagonist did not increase Foxp3(+) regulatory T cells in the lung. D1-like-R antagonist neither suppressed nonspecific LPS-induced neutrophilic airway inflammation nor OVA-induced eosinophilic airway inflammation. These results indicate that D1-like-R antagonist could suppress Th17-mediated neutrophilic airway inflammation, raising the possibility that antagonizing D1-like-R serves as a promising new strategy for treating neutrophil-dominant severe asthma.


Subject(s)
Benzazepines/pharmacology , Neutrophils/immunology , Receptors, Dopamine D1/antagonists & inhibitors , Respiratory Hypersensitivity/immunology , Th17 Cells/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Dopamine/immunology , Dopamine/metabolism , Female , Forkhead Transcription Factors/metabolism , Inflammation/immunology , Interleukin-23/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology , Receptors, Dopamine D1/metabolism , Respiratory Hypersensitivity/metabolism , T-Lymphocytes, Regulatory/immunology
9.
J Immunol ; 187(10): 5077-89, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21998459

ABSTRACT

Allergic inflammation in the airway is generally considered a Th2-type immune response. However, Th17-type immune responses also play important roles in this process, especially in the pathogenesis of severe asthma. IL-22 is a Th17-type cytokine and thus might play roles in the development of allergic airway inflammation. There is increasing evidence that IL-22 can act as a proinflammatory or anti-inflammatory cytokine depending on the inflammatory context. However, its role in Ag-induced immune responses is not well understood. This study examined whether IL-22 could suppress allergic airway inflammation and its mechanism of action. BALB/c mice were sensitized and challenged with OVA-Ag to induce airway inflammation. An IL-22-producing plasmid vector was delivered before the systemic sensitization or immediately before the airway challenge. Delivery of the IL-22 gene before sensitization, but not immediately before challenge, suppressed eosinophilic airway inflammation. IL-22 gene delivery suppressed Ag-induced proliferation and overall cytokine production in CD4(+) T cells, indicating that it could suppress Ag-induced T cell priming. Antagonism of IL-22 by IL-22-binding protein abolished IL-22-induced immune suppression, suggesting that IL-22 protein itself played an essential role. IL-22 gene delivery neither increased regulatory T cells nor suppressed dendritic cell functions. The suppression by IL-22 was abolished by deletion of the IL-10 gene or neutralization of the IL-10 protein. Finally, IL-22 gene delivery increased IL-10 production in draining lymph nodes. These findings suggested that IL-22 could have an immunosuppressive effect during the early stage of an immune response. Furthermore, IL-10 plays an important role in the immune suppression by IL-22.


Subject(s)
Down-Regulation/immunology , Eosinophilia/immunology , Eosinophilia/pathology , Immunosuppressive Agents/metabolism , Interleukin-10/physiology , Interleukins/biosynthesis , Up-Regulation/immunology , Animals , Down-Regulation/genetics , Eosinophilia/genetics , Gene Transfer Techniques , Humans , Immunosuppressive Agents/administration & dosage , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/metabolism , Interleukins/administration & dosage , Interleukins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Up-Regulation/genetics , Interleukin-22
10.
Sci Rep ; 12(1): 9147, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35650227

ABSTRACT

Recently, immune response to coronavirus disease (COVID-19) has attracted attention where an association between higher antibody titer and worsening disease severity has been reported. However, our experiences with severe COVID-19 patients with low antibody titers led to hypothesizing that suppressed humoral immune response may be associated with poorer prognosis in severe COVID19. In this study, antibody titers in severe COVID19 patients were measured at 7, 10, 12, and 14 days after onset. Patients were divided into survivors and non-survivors. SARS-CoV-2 IgM in survivors and non-survivors were 0.06 AU and 0.02 AU (P = 0.048) at 10 days, 0.1 AU and 0.03 AU (P = 0.02) at 12 days, and 0.17 AU and 0.06 AU (P = 0.02) at 14 days. IgG in survivors and non-survivors were 0.01 AU and 0.01 AU (P = 0.04) at 7 days, 0.42 AU and 0.01 AU (P = 0.04) at 12 days, and 0.42 AU and 0.01 AU (P = 0.02) at 14 days. Multivariate analysis showed better survival among patients with IgM positivity at 12 days (P = 0.04), IgG positivity at 12 days (P = 0.04), IgM positivity at 14 days (P = 0.008), and IgG positivity at 14 days (P = 0.005). In severe COVID-19, low antibody titers on days 12 and 14 after onset were associated with poorer prognosis.


Subject(s)
COVID-19 , Antibodies, Viral , Humans , Immunoglobulin G , Immunoglobulin M , SARS-CoV-2
11.
Respir Investig ; 60(6): 750-761, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35934631

ABSTRACT

BACKGROUND: The role of programmed cell death, especially pyroptosis and apoptosis, in unfavorable immune responses in COVID-19 remains to be elucidated. METHODS: We conducted a cross-sectional analysis to investigate the association between the serum gasdermin D (GSDMD) levels, a pyroptotic marker, and caspase-cleaved cytokeratin 18 fragment (M30), an apoptotic marker, and the clinical status and abnormal chest computed tomography (CT) findings in patients with COVID-19. RESULTS: In this study, 46 patients diagnosed with COVID-19 were divided into the following three groups according to the disease severity: mild to moderate group (n = 10), severe group (n = 14), and critical group (n = 22). The serum GSDMD levels were higher in the critical group than in the mild to moderate group (P = 0.016). In contrast, serum M30 levels were lower in the critical group than in the severe group (P = 0.048). Patients who required mechanical ventilation or died had higher serum GSDMD levels than those who did not (P = 0.007). Area of consolidation only and of ground glass opacity plus consolidation positively correlated with serum GSDMD levels (r = 0.56, P < 0.001 and r = 0.53, P < 0.001, respectively). CONCLUSION: Higher serum GSDMD levels are associated with critical respiratory status and the consolidation area on chest CT in patients with COVID-19, suggesting that excessive activation of pyroptosis may affect the clinical manifestations in patients with COVID-19.


Subject(s)
COVID-19 , Humans , Phosphate-Binding Proteins/metabolism , COVID-19/diagnostic imaging , Cross-Sectional Studies , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Tomography , Tomography, X-Ray Computed
12.
PLoS One ; 17(11): e0276738, 2022.
Article in English | MEDLINE | ID: mdl-36327268

ABSTRACT

Presently, coronavirus disease-19 (COVID-19) is spreading worldwide without an effective treatment method. For COVID-19, which is often asymptomatic, it is essential to adopt a method that does not cause aggravation, as well as a method to prevent infection. Whether aggravation can be predicted by analyzing the extent of lung damage on chest computed tomography (CT) scans was examined. The extent of lung damage on pre-intubation chest CT scans of 277 patients with COVID-19 was assessed. It was observed that aggravation occurred when the CT scan showed extensive damage associated with ground-glass opacification and/or consolidation (p < 0.0001). The extent of lung damage was similar across the upper, middle, and lower fields. Furthermore, upon comparing the extent of lung damage based on the number of days after onset, a significant difference was found between the severe pneumonia group (SPG) with intubation or those who died and non-severe pneumonia group (NSPG) ≥3 days after onset, with aggravation observed when ≥14.5% of the lungs exhibited damage at 3-5 days (sensitivity: 88.2%, specificity: 72.4%) and when ≥20.1% of the lungs exhibited damage at 6-8 days (sensitivity: 88.2%, specificity: 69.4%). Patients with aggravation suddenly developed hypoxemia after 7 days from the onset; however, chest CT scans obtained in the paucisymptomatic phase without hypoxemia indicated that subsequent aggravation could be predicted based on the degree of lung damage. Furthermore, in subjects aged ≥65 years, a significant difference between the SPG and NSPG was observed in the extent of lung damage early beginning from 3 days after onset, and it was found that the degree of lung damage could serve as a predictor of aggravation. Therefore, to predict and improve prognosis through rapid and appropriate management, evaluating patients with factors indicating poor prognosis using chest CT is essential.


Subject(s)
COVID-19 , Humans , COVID-19/diagnostic imaging , SARS-CoV-2 , Tomography, X-Ray Computed/methods , Lung/diagnostic imaging , Hypoxia , Retrospective Studies
13.
Am J Pathol ; 177(4): 1665-73, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20709801

ABSTRACT

Acute kidney injury (AKI) is a serious problem in critically ill patients of intensive care units. It has been reported previously that AKI can induce acute lung injury (ALI), as well as cause injuries to other remote organs, including the lungs. Patients with AKI complicated by ALI show remarkably high mortality. ALI is characterized by neutrophil infiltration into the lung. Neutrophil elastase (NE) is a key enzyme for tissue injury caused by activated neutrophils, such as occurs in ALI. Therefore, this study investigated the role of NE in AKI-induced ALI using a specific NE inhibitor, sivelestat sodium hydrate (ONO-5046), in a mouse bilateral nephrectomy model. Bilateral nephrectomy showed not only a remarkable increase in blood urea nitrogen levels, but also demonstrated neutrophil infiltration into the lung, increased pulmonary inflammatory cytokine expression [interleukin-6, neutrophil chemokine keratinocyte-derived chemokine, and tumor necrosis factor-α], and protein leakage with early increases in both systemic and pulmonary NE activity. ONO-5046 treatment reduced NE activity and improved these pulmonary inflammatory responses. Additionally, ONO-5046-treated animals had longer survival times. These data demonstrate that increasing NE activity induces pulmonary inflammatory damage in a bilateral nephrectomy model. Blockade of NE activity will be a useful therapeutic strategy for ALI complications in AKI patients.


Subject(s)
Acute Lung Injury/enzymology , Acute Lung Injury/etiology , Leukocyte Elastase/physiology , Nephrectomy/adverse effects , Acute Lung Injury/drug therapy , Animals , Blotting, Western , Chemokines/genetics , Chemokines/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophil Activation/drug effects , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serine Proteinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Survival Rate , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
J Immunol ; 183(1): 209-20, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542432

ABSTRACT

Allergic inflammation in the airway is generally considered a Th2-type immune response. However, recent studies demonstrated that Th1- and Th17-type immune responses also play important roles in this process. IFN-gamma is a Th1-type cytokine that generally counteracts the Th2 response. Although previous studies suggest that exogenous IFN-gamma suppresses allergic airway inflammation, the mechanism of suppression has not been fully clarified. In this study, we elucidated whether IFN-gamma suppresses Ag-induced immune responses including the production of Th1- and Th17-type cytokines in the lung, and examined its mechanism of action. BALB/c mice were sensitized and challenged with OVA-Ag to induce airway inflammation. An IFN-gamma-producing plasmid vector was delivered before systemic Ag sensitization. IFN-gamma suppressed indicators of Th2-type immune responses such as airway eosinophilia, IL-5 and IL-13 production in the lung, and bronchial mucus production. Moreover, IFN-gamma also suppressed the production of IL-17 and IFN-gamma itself. The suppression was not mediated by inducing regulatory T cells or by inducing apoptosis in immunocytes. Instead, IFN-gamma suppressed the Ag-presenting capacity and cytokine production of splenic dendritic cells and thus subsequently suppressed OVA-induced activation of CD4(+) T cells. Furthermore, IFN-gamma also attenuated allergic airway inflammation when delivered during the OVA challenge. Various functions of lung CD11c(+) APCs and their migration to regional lymph nodes were also suppressed. These results suggest that the Th1 cytokine IFN-gamma has broad immune regulatory potential through suppressing APC functions. They also suggest that delivery of IFN-gamma could be an effective strategy for regulating Ag-induced immune responses in the lung.


Subject(s)
Antigens/administration & dosage , Antigens/immunology , Cytokines/antagonists & inhibitors , Interferon-gamma/physiology , Lung/immunology , Th1 Cells/immunology , Animals , Antigen-Presenting Cells/metabolism , CD11c Antigen/biosynthesis , CD11c Antigen/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/physiology , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Lung/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Plasmids/administration & dosage , Plasmids/immunology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/prevention & control , Th1 Cells/metabolism
15.
Int Arch Allergy Immunol ; 149 Suppl 1: 25-30, 2009.
Article in English | MEDLINE | ID: mdl-19494501

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is now considered a chronic inflammatory disease. Although dendritic cells (DCs) are thought to play a key role in immune responses, studies investigating the role of DCs in COPD are quite limited. METHODS: Porcine pancreas elastase was intratracheally administered to C57BL/6J mice on day 0. On days 2, 7 and 14, emphysema formation was evaluated by pressure-volume relationships and microscopic findings, including measurement of the mean linear intercept. Lung DCs were isolated on day 2, and their ability to stimulate allogeneic T cells and to produce cytokines was examined. RESULTS: Pathologic emphysematous change was observed on day 2 and a significant increase in lung volume was observed on day 14. Lung DC function, such as the induction of T-cell proliferation and IL-10 production, was upregulated. CONCLUSIONS: Upregulation of DC function was observed in elastase-induced emphysema. Further investigation on the contribution to emphysema formation may provide a useful target for future therapy.


Subject(s)
Dendritic Cells/immunology , Emphysema/immunology , Up-Regulation , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Proliferation , Disease Models, Animal , Interleukin-10/biosynthesis , Male , Mice , Mice, Inbred C57BL , Pancreatic Elastase/immunology , T-Lymphocytes/cytology
16.
Int Arch Allergy Immunol ; 149 Suppl 1: 14-20, 2009.
Article in English | MEDLINE | ID: mdl-19494499

ABSTRACT

Hepatocyte growth factor (HGF) has an important role in many biological events such as angiogenesis and cell proliferation, as well as anti-fibrotic and anti-apoptotic effects. In addition, we found that HGF suppresses antigen-induced immune responses in the airway by suppressing dendritic cell functions, using a HGF-producing plasmid vector. In the present study, we examined whether delivery of the HGF protein in the lung attenuates allergic airway inflammation in a mouse model. Generally, HGF is rapidly cleared from organs. So, to achieve the efficient delivery of HGF, we prepared a slow-releasing form by mounting recombinant human (rh) HGF protein in biodegradable gelatin hydrogels. BALB/c mice were immunized and then challenged with ovalbumin (OVA) to induce eosinophilic airway inflammation. Intratracheal delivery of a very small amount of gelatin-coupled rhHGF (0.3 microg) just once before the inhalation of OVA significantly suppressed eosinophilic airway inflammation. In addition, cytokine production in thoracic lymph nodes and the antigen-presenting capacity of lung CD11c+ cells were reduced. In contrast, delivery of 1.0 microg of rhHGF did not exhibit any significantly suppressive effect. These results suggest that the controlled release of rhHGF protein can suppress antigen-induced allergic immune responses in the lung. Therefore, HGF could be a novel therapeutic option for asthma.


Subject(s)
Asthma/drug therapy , Hepatocyte Growth Factor/administration & dosage , Animals , Delayed-Action Preparations , Gelatin/administration & dosage , Gels/administration & dosage , Humans , Inflammation/drug therapy , Male , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage
17.
JCI Insight ; 52019 07 09.
Article in English | MEDLINE | ID: mdl-31287803

ABSTRACT

The prevalence of obesity is rising worldwide and obese patients comprise a specific population in the intensive care unit. Acute respiratory distress syndrome (ARDS) incidence is increased in obese patients. Exposure of rodents to hyperoxia mimics many of the features of ARDS. In this report, we demonstrate that high fat diet induced obesity increases the severity of hyperoxic acute lung injury in mice in part by altering fatty acid synthase (FASN) levels in the lung. Obese mice exposed to hyperoxia had significantly reduced survival and increased lung damage. Transcriptomic analysis of lung homogenates identified Fasn as one of the most significantly altered mitochondrial associated genes in mice receiving 60% compared to 10% fat diet. FASN protein levels in the lung of high fat diet mice were lower by immunoblotting and immunohistochemistry. Depletion of FASN in type II alveolar epithelial cells resulted in altered mitochondrial bioenergetics and more severe lung injury with hyperoxic exposure, even upon the administration of a 60% fat diet. This is the first study to show that a high fat diet leads to altered FASN expression in the lung and that both a high fat diet and reduced FASN expression in alveolar epithelial cells promote lung injury.


Subject(s)
Alveolar Epithelial Cells/pathology , Fatty Acid Synthase, Type I/metabolism , Hyperoxia/complications , Obesity/metabolism , Respiratory Distress Syndrome/pathology , Adaptor Proteins, Signal Transducing/genetics , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Down-Regulation , Energy Metabolism , Gene Expression Profiling , Humans , Hyperoxia/metabolism , Male , Mice , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Obesity/etiology , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/metabolism
18.
Nat Commun ; 10(1): 3390, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358769

ABSTRACT

Accumulating evidence illustrates a fundamental role for mitochondria in lung alveolar type 2 epithelial cell (AEC2) dysfunction in the pathogenesis of idiopathic pulmonary fibrosis. However, the role of mitochondrial fusion in AEC2 function and lung fibrosis development remains unknown. Here we report that the absence of the mitochondrial fusion proteins mitofusin1 (MFN1) and mitofusin2 (MFN2) in murine AEC2 cells leads to morbidity and mortality associated with spontaneous lung fibrosis. We uncover a crucial role for MFN1 and MFN2 in the production of surfactant lipids with MFN1 and MFN2 regulating the synthesis of phospholipids and cholesterol in AEC2 cells. Loss of MFN1, MFN2 or inhibiting lipid synthesis via fatty acid synthase deficiency in AEC2 cells exacerbates bleomycin-induced lung fibrosis. We propose a tenet that mitochondrial fusion and lipid metabolism are tightly linked to regulate AEC2 cell injury and subsequent fibrotic remodeling in the lung.


Subject(s)
GTP Phosphohydrolases/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lipid Metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Cells, Cultured , Cholesterol/metabolism , GTP Phosphohydrolases/genetics , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Mice, Knockout , Mice, Transgenic , Mitochondrial Dynamics/drug effects , Mitochondrial Dynamics/genetics , Phospholipids/biosynthesis
19.
JCI Insight ; 3(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29415885

ABSTRACT

Renal fibrosis is a common pathogenic response to injury in chronic kidney disease (CKD). The receptor-interacting protein kinase-3 (RIPK3), a regulator of necroptosis, has been implicated in disease pathogenesis. In mice subjected to unilateral ureteral obstruction-induced (UUO-induced) or adenine diet-induced (AD-induced) renal fibrosis, models of progressive kidney fibrosis, we demonstrate increased kidney expression of RIPK3. Mice genetically deficient in RIPK3 displayed decreased kidney fibrosis and improved kidney function relative to WT mice when challenged with UUO or AD. In contrast, mice genetically deficient in mixed-lineage kinase domain-like protein (MLKL), a downstream RIPK3 target, were not protected from UUO-induced kidney fibrosis. We demonstrate a pathway by which RIPK3 promotes fibrogenesis through the AKT-dependent activation of ATP citrate lyase (ACL). Genetic or chemical inhibition of RIPK3 suppressed the phosphorylation of AKT and ACL in response to TGF-ß1 in fibroblasts. Inhibition of AKT or ACL suppressed TGF-ß1-dependent extracellular matrix production and myofibroblast differentiation in fibroblasts. Pharmacological inhibition of ACL suppressed UUO-induced kidney fibrosis. RIPK3 expression was highly regulated in human CKD kidney. In conclusion, we identify a pathway by which RIPK3 promotes kidney fibrosis independently of MLKL-dependent necroptosis as a promising therapeutic target in CKD.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Kidney/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Renal Insufficiency, Chronic/pathology , ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , ATP Citrate (pro-S)-Lyase/genetics , Adenine/administration & dosage , Adenine/toxicity , Adult , Aged , Aged, 80 and over , Animals , Cell Differentiation/drug effects , Disease Models, Animal , Female , Fibrosis , Gene Knockdown Techniques , Humans , Kidney/drug effects , Male , Mice , Middle Aged , Myofibroblasts , NIH 3T3 Cells , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Renal Insufficiency, Chronic/etiology
20.
JCI Insight ; 3(9)2018 05 03.
Article in English | MEDLINE | ID: mdl-29720570

ABSTRACT

In patients requiring ventilator support, mechanical ventilation (MV) may induce acute lung injury (ventilator-induced lung injury [VILI]). VILI is associated with substantial morbidity and mortality in mechanically ventilated patients with and without acute respiratory distress syndrome. At the cellular level, VILI induces necrotic cell death. However, the contribution of necroptosis, a programmed form of necrotic cell death regulated by receptor-interacting protein-3 kinase (RIPK3) and mixed-lineage kinase domain-like pseudokinase (MLKL), to the development of VILI remains unexplored. Here, we show that plasma levels of RIPK3, but not MLKL, were higher in patients with MV (i.e., those prone to VILI) than in patients without MV (i.e., those less likely to have VILI) in two large intensive care unit cohorts. In mice, RIPK3 deficiency, but not MLKL deficiency, ameliorated VILI. In both humans and mice, VILI was associated with impaired fatty acid oxidation (FAO), but in mice this association was not observed under conditions of RIPK3 deficiency. These findings suggest that FAO-dependent RIPK3 mediates pathogenesis of acute lung injury.


Subject(s)
Protein Kinases/blood , Protein Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/blood , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Respiration, Artificial/adverse effects , Ventilator-Induced Lung Injury/blood , Aged , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cell Death/genetics , Cohort Studies , Fatty Acids/metabolism , Female , Humans , Intensive Care Units , Male , Mice , Middle Aged , Oxidation-Reduction , Protein Kinases/genetics , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ventilator-Induced Lung Injury/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL