Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Allergy ; 78(8): 2168-2180, 2023 08.
Article in English | MEDLINE | ID: mdl-36951658

ABSTRACT

BACKGROUND: Exacerbations of asthma are thought to be strongly dependent on reactivation of allergen-induced lung tissue-resident and circulatory memory CD4 T cells. Strategies that broadly inhibit multiple T cell populations might then be useful to limit asthma. Accordingly, we tested whether targeting CD3 during exposure to inhaled allergen could prevent the accumulation of lung-localized effector memory CD4 T cells and block exacerbations of asthmatic inflammation. METHODS: House dust mite-sensitized and repetitively challenged BL/6 mice were transiently treated therapeutically with F(ab')2 anti-CD3ε and memory T cell responses and lung inflammation were assessed. PBMCs from HDM-allergic donors were examined for the effect of anti-CD3 on expansion of allergen-reactive T cells. RESULTS: Allergen-sensitized mice undergoing exacerbations of asthma were protected from lung inflammation by transient therapeutic treatment with F(ab')2 anti-CD3. Regardless of whether sensitized mice underwent a secondary or tertiary recall response to inhaled allergen, anti-CD3 inhibited all phenotypes of effector memory CD4 T cells in the lung tissue and lung vasculature by 80%-90%, including those derived from tissue-resident and circulatory memory T cells. This did not depend on Treg cells suggesting it was primarily a blocking effect on memory T cell signaling. Correspondingly, anti-CD3 also strongly inhibited proliferation of human allergen-reactive memory CD4 T cells from allergic individuals. In contrast, the number of surviving tissue-resident memory CD4 T cells that were maintained in the lungs at later times was not robustly reduced by anti-CD3. CONCLUSION: Anti-CD3 F(ab')2 administration at the time of allergen exposure represents a viable strategy for limiting the immediate activity of allergen-responding memory T cells and asthma exacerbations.


Subject(s)
Asthma , Hypersensitivity , Pneumonia , Animals , Mice , Humans , Memory T Cells , CD4-Positive T-Lymphocytes , Th2 Cells , Asthma/prevention & control , Allergens/adverse effects , Pyroglyphidae , Disease Models, Animal
2.
J Allergy Clin Immunol ; 151(4): 976-990.e5, 2023 04.
Article in English | MEDLINE | ID: mdl-36473503

ABSTRACT

BACKGROUND: Dysregulation of airway smooth muscle cells (ASM) is central to the severity of asthma. Which molecules dominantly control ASM in asthma is unclear. High levels of the cytokine LIGHT (aka TNFSF14) have been linked to asthma severity and lower baseline predicted FEV1 percentage, implying that signals through its receptors might directly control ASM dysfunction. OBJECTIVE: Our study sought to determine whether signaling via lymphotoxin beta receptor (LTßR) or herpesvirus entry mediator from LIGHT dominantly drives ASM hyperreactivity induced by allergen. METHODS: Conditional knockout mice deficient for LTßR or herpesvirus entry mediator in smooth muscle cells were used to determine their role in ASM deregulation and airway hyperresponsiveness (AHR) in vivo. Human ASM were used to study signals induced by LTßR. RESULTS: LTßR was strongly expressed in ASM from normal and asthmatic subjects compared to several other receptors implicated in smooth muscle deregulation. Correspondingly, conditional deletion of LTßR only in smooth muscle cells in smMHCCreLTßRfl/fl mice minimized changes in their numbers and mass as well as AHR induced by house dust mite allergen in a model of severe asthma. Intratracheal LIGHT administration independently induced ASM hypertrophy and AHR in vivo dependent on direct LTßR signals to ASM. LIGHT promoted contractility, hypertrophy, and hyperplasia of human ASM in vitro. Distinguishing LTßR from the receptors for IL-13, TNF, and IL-17, which have also been implicated in smooth muscle dysregulation, LIGHT promoted NF-κB-inducing kinase-dependent noncanonical nuclear factor kappa-light-chain enhancer of activated B cells in ASM in vitro, leading to sustained accumulation of F-actin, phosphorylation of myosin light chain kinase, and contractile activity. CONCLUSIONS: LTßR signals directly and dominantly drive airway smooth muscle hyperresponsiveness relevant for pathogenesis of airway remodeling in severe asthma.


Subject(s)
Asthma , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Mice , Animals , Lymphotoxin beta Receptor/genetics , Asthma/pathology , Muscle, Smooth , Myocytes, Smooth Muscle/pathology , Mice, Knockout , Allergens , Lung/pathology
3.
Front Immunol ; 13: 951361, 2022.
Article in English | MEDLINE | ID: mdl-35936001

ABSTRACT

Tissue-resident memory CD4 T cells (Trm) are thought to be a major contributor to asthma relapse, but the role of circulatory T cells in asthma exacerbations or to maintaining the population of lung Trm cells is not fully understood. Here, we used a house dust mite allergen-based murine model of asthma relapse, and monitored the development of lung effector/Trm phenotype CD44hiCD62LloCD69+ CD4 T cells. To determine the contribution of circulatory cells, mice were treated with FTY720, to block lymphocyte egress from lymph nodes. Inhibiting the primary migration of circulatory cells to the lungs mitigated the accumulation and expansion of allergen-driven Trm phenotype cells, but subsequent allergen challenges still resulted in strong lung inflammation and Trm cell accumulation. This was blocked if FTY720 was also given at the time of allergen re-exposure, showing that new circulatory cells contributed to this lung memory/effector T cell pool at times well after the initial sensitization. However, once lung-localized Trm cells developed at high frequency, circulatory cells were not required to maintain this population following allergen re-encounter, even though circulatory cells still were major contributors to the overall asthmatic lung inflammatory response. Our results suggest that strategies that target the response of circulatory memory T cells and Trm cells together might be required to strongly inhibit T cell reactivity to airborne allergens and to limit exacerbations of asthma and their reoccurrence, but the contribution of circulatory T cells might vary in long-term asthmatics possessing a large stable Trm cell population in the lungs.


Subject(s)
Asthma , Pneumonia , Allergens , Animals , Asthma/pathology , CD4-Positive T-Lymphocytes , Fingolimod Hydrochloride/pharmacology , Immunologic Memory , Lung , Mice , Pneumonia/pathology , Recurrence
4.
J Allergy Clin Immunol ; 147(6): 2316-2329, 2021 06.
Article in English | MEDLINE | ID: mdl-33160971

ABSTRACT

BACKGROUND: The selective reduction of memory TH2 cell responses could be key to affording tolerance and protection from the recurrence of damaging allergic pathology. OBJECTIVE: We asked whether TNF family costimulatory molecules cooperated to promote accumulation and reactivity of effector memory CD4 T cells to inhaled complex allergen, and whether their neutralization could promote airway tolerance to subsequent reexposure to allergen. METHODS: Mice were sensitized intraperitoneally or intranasally with house dust mite and challenged with intranasal allergen after memory had developed. We assessed whether single or combined blockade of OX40L/CD252 and CD30L/CD153 inhibited memory T cells from driving acute asthmatic lung inflammation and protected mice following exposure to allergen at a later time. RESULTS: OX40- or CD30-deficient animals showed strong or partial protection against allergic airway inflammation; however, neutralizing either molecule alone during the secondary response to allergen had little effect on the frequency of effector memory CD4 T cells formed and acute lung inflammation. In contrast, a significant reduction in eosinophilic inflammation was observed when OX40L and CD30L were simultaneously neutralized, with dual blockade inhibiting effector memory TH2 cell expansion in the lungs, whereas formation of peripherally induced regulatory T cells remained intact. Moreover, dual blockade during the secondary response resulted in a tolerogenic state such that mice did not develop a normal tertiary memory TH2 cell and lung inflammatory response when challenged weeks later with allergen. CONCLUSION: Memory T-cell responses to complex allergens are controlled by several TNF costimulatory interactions, and their combination targeting might represent a strategy to reduce the severity of inflammatory reactions following reexposure to allergen.


Subject(s)
Allergens/immunology , CD30 Ligand/antagonists & inhibitors , Immunologic Memory , OX40 Ligand/antagonists & inhibitors , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Asthma/etiology , Asthma/metabolism , Asthma/pathology , Biomarkers , Disease Models, Animal , Disease Susceptibility/immunology , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Mice , Pneumonia/etiology , Pneumonia/metabolism , Pneumonia/pathology
5.
Inflamm Res ; 70(1): 109-126, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33136175

ABSTRACT

OBJECTIVE: Cysteinyl leukotrienes (CysLTs), a group of inflammatory lipid mediators, are found elevated in obese-asthmatic patients. Leukotriene D4 (LTD4), a representative CysLT, is implicated in promoting lung inflammation and remodelling in allergic asthma, but its role in non-allergic asthma, especially in obese-asthmatic patients, is not known. Here, using primary human small airway epithelial cells (SAECs) we have investigated the mechanism of LTD4-induced inflammation and remodelling and assessed high proneness of obese mice to develop asthma upon challenge with allergen ovalbumin (OVA). METHODS: Primary human small airway epithelial cells (SAECs) were stimulated with different concentrations of LTD4 for different time intervals and various inflammatory markers were measured through cytokine array, membrane-based ELISA and Western blotting. An air-liquid interface (ALI) model of SAECs was used to study the effects of LTD4-induced remodelling in SAECs using Western blotting, H&E staining and PAS staining. Further, OVA-based murine model was used to examine the propensity of high-fat diet (HFD)-fed obese mice to develop asthma symptoms by studying the infiltration of inflammatory cells (assessed by bronchioalveolar lavage (BAL) cytology) and airway remodelling (assessed by histopathology) upon allergen exposure. RESULTS: The human primary small airway epithelial cells (SAECs) treated with LTD4 showed significant alterations in the levels of inflammatory markers such as GM-CSF, TNF-α, IL-1ß, EGF and eotaxin in dose- and time-dependent manner. Further, LTD4 enhanced the activation of inflammasomes as evidenced by increased levels of NALP3, cleaved caspase-1 and IL-1ß. LTD4 also enhanced inflammation by increasing the expression of COX-2 in SAECs. The airway remodelling markers Vimentin and Muc5AC were found elevated in ALI culture of SAECs when stimulated with LTD4, as it also increased TGF-ß levels and activation of Smad2/3 phosphorylation in SAECs. Last, sensitization and challenge of HFD-fed obese mice with OVA showed increased infiltration of inflammatory cells in BAL and enhanced levels of remodeling phenotypes like loss of cilia, mucus cell metaplasia and collagen deposition in mice lung tissues. CONCLUSION: The results suggest that LTD4 could induce inflammatory response in human airway epithelial cell by activating NALP3 inflammasome. LTD4 could further promote airway epithelial cells' remodelling through TGF-ß/smad2/3-mediated pathway. Our in vivo results suggested that obesity predisposed the OVA challenged mice to develop lung inflammation and remodelling akin to asthma-like phenotypes during obesity.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Epithelial Cells/immunology , Inflammation/immunology , Leukotriene D4/immunology , Obesity/immunology , Allergens/immunology , Animals , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Cytokines/immunology , Humans , Inflammasomes/immunology , Inflammation/pathology , Leukocyte Count , Male , Mice, Inbred BALB C , Mucin 5AC/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Obesity/pathology , Ovalbumin/immunology , Smad2 Protein/immunology , Smad3 Protein/immunology , Vimentin/immunology
6.
J Immunol ; 205(9): 2414-2422, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32958689

ABSTRACT

Lung fibrosis and tissue remodeling are features of chronic diseases such as severe asthma, idiopathic pulmonary fibrosis, and systemic sclerosis. However, fibrosis-targeted therapies are currently limited. We demonstrate in mouse models of allergen- and bleomycin-driven airway inflammation that neutralization of the TNF family cytokine TL1A through Ab blocking or genetic deletion of its receptor DR3 restricted increases in peribronchial smooth muscle mass and accumulation of lung collagen, primary features of remodeling. TL1A was found as a soluble molecule in the airways and expressed on the surface of alveolar macrophages, dendritic cells, innate lymphoid type 2 cells, and subpopulations of lung structural cells. DR3 was found on CD4 T cells, innate lymphoid type 2 cells, macrophages, fibroblasts, and some epithelial cells. Suggesting in part a direct activity on lung structural cells, administration of recombinant TL1A into the naive mouse airways drove remodeling in the absence of other inflammatory stimuli, innate lymphoid cells, and adaptive immunity. Correspondingly, human lung fibroblasts and bronchial epithelial cells were found to express DR3 and responded to TL1A by proliferating and/or producing fibrotic molecules such as collagen and periostin. Reagents that disrupt the interaction of TL1A with DR3 then have the potential to prevent deregulated tissue cell activity in lung diseases that involve fibrosis and remodeling.


Subject(s)
Airway Remodeling/immunology , Idiopathic Pulmonary Fibrosis/immunology , Lung/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Adaptive Immunity/immunology , Animals , Asthma/immunology , Bleomycin/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Dendritic Cells/immunology , Epithelial Cells/immunology , Female , Humans , Immunity, Innate/immunology , Inflammation/immunology , Lymphocytes/immunology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL
7.
Biochem Pharmacol ; 179: 113925, 2020 09.
Article in English | MEDLINE | ID: mdl-32217103

ABSTRACT

Lipids and their mediators are known to play a pro-inflammatory role in several human diseases including asthma. The influence of leukotrienes and prostaglandins through arachidonate metabolism in asthma pathophysiology is well established and hence, prompted the way for therapeutic strategies targeting lipid metabolites. In addition, various types of fatty acids have been reported to play a diverse role in asthma. For instance, CD4+ T-lymphocytes differentiation towards T-effector (Teff) or T-regulatory (Tregs) cells seems to be controlled reciprocally by fatty acid metabolic pathways. Further, the dysregulated lipid status in obesity complicates the asthma manifestations suggesting the role of lipid metabolites particularly ω-6 fatty acids in the process. On the other hand, clinical and pre-clinical studies suggests the role of short chain fatty acids in curbing asthma through upregulation of T-regulatory cells or clearance of inflammatory cells through promoting apoptosis. Accordingly, the present review compiles various studies for comprehensive analysis of different types of lipid based metabolites in asthma manifestation. Finally, we have proposed certain strategies which may enhance the usefulness of lipid mediators for balanced immune response during asthma.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/diet therapy , Asthma/drug therapy , Asthma/metabolism , Anti-Asthmatic Agents/adverse effects , Anti-Asthmatic Agents/therapeutic use , Asthma/physiopathology , Fatty Acids/metabolism , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/pharmacology , Glucocorticoids/pharmacology , Humans , Lipid Metabolism , Lipids/physiology , Lymphocytes/metabolism , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Obesity/metabolism , Obesity/physiopathology
8.
Inflammation ; 43(1): 358-369, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31720988

ABSTRACT

Curcumin, extracted from the roots of Curcuma longa, has been used as an anti-inflammatory agent since the time of Ayurveda. The present work was designed to evaluate the potential of curcumin in amelioration of ovalbumin (OVA) induced AD in mice. Female BALB/c mice were subjected to skin OVA-patch application for a period of 1 week followed by resting period of 2 weeks, and the same protocol was repeated thrice. Curcumin was administered daily at dose of 20 mg/kg (i.p.) for 7 consecutive days during last sensitization phase. The phytochemical ameliorated the OVA-induced skin pathology as evident by normalization of epidermal thickness and suppressed infiltration of inflammatory cells in dermal region. The expression of Th2 promoting cytokines (TSLP/IL-33) and Th2 cytokines (IL-4/IL-5/IL-13/IL-31) was suppressed markedly along with reduced STAT-6 phosphorylation and GATA-3 expression. Curcumin administration also restored the redox balance and phosphorylation status of P65-NF-κB. Additionally, the epicutaneously sensitized mice challenged with aerosolized OVA developed asthmatic features which were effectively thwarted back upon curcumin treatment as reflected by data on total/differential cells in BALF and mRNA expression of Th2 cytokines in lungs. Overall, our findings demonstrate that curcumin treatment blunts the development of AD as well as associated atopic march in experimental mice.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Curcumin/pharmacology , Dermatitis, Atopic/prevention & control , Skin/drug effects , Animals , Asthma/chemically induced , Asthma/immunology , Asthma/metabolism , Asthma/prevention & control , Cytokines/metabolism , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/immunology , Dermatitis, Atopic/metabolism , Disease Models, Animal , Disease Progression , Female , GATA3 Transcription Factor/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Mice, Inbred BALB C , Ovalbumin , Phosphorylation , STAT6 Transcription Factor/metabolism , Skin/immunology , Skin/metabolism , Skin/pathology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Transcription Factor RelA/metabolism
9.
IUBMB Life ; 71(7): 1003-1013, 2019 07.
Article in English | MEDLINE | ID: mdl-30964965

ABSTRACT

Despite the reported role of poly(ADP-ribose) polymerase (PARP) in asthma inflammation, its contribution during remodeling is not clearly known. The main aim of the current investigation was to examine the potential of olaparib, a pharmacological inhibitor of PARP against airway remodeling using an ovalbumin (OVA)-based murine model of chronic asthma. The results demonstrated that post-challenge olaparib treatment (5 mg/kg i.p., 30 min after OVA exposure) for six weeks (3 days/week) attenuates inflammation, mucus production, and collagen deposition in lungs. Additionally, olaparib blunted the protein expression of STAT-6 and GATA-3 considerably along with a modest reduction in p65-NF-κB phosphorylation. Furthermore, olaparib normalized the OVA-induced redox imbalance as reflected by data on reactive oxygen species, malondialdehyde, protein carbonyls, and reduced glutathione/oxidized glutathione ratio. Interestingly, the protection offered by olaparib was further linked with the altered level of NLRP3 inflammasome-mediated IL-1ß release and consequent expression of its downstream targets matrix metalloproteinase-9 and transforming growth factor beta. Suppressed collagen deposition in the lungs correlates well with the reduced expression of vimentin upon olaparib treatment. Finally, olaparib restored the expression of histone deacetylase 2, a steroid-responsive element in asthma. Overall, results suggest that olaparib prevents OVA-induced airway inflammation as well as remodeling via modulating inflammasome signaling in mice. © 2019 IUBMB Life, 1-11, 2019.


Subject(s)
Airway Remodeling/immunology , Asthma/complications , Inflammasomes/drug effects , Phthalazines/pharmacology , Piperazines/pharmacology , Pneumonia/prevention & control , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Airway Remodeling/drug effects , Animals , Apoptosis , Asthma/chemically induced , Asthma/pathology , Cell Proliferation , Chronic Disease , Male , Mice , Mice, Inbred BALB C , Ovalbumin/toxicity , Pneumonia/etiology , Pneumonia/metabolism , Tumor Cells, Cultured
10.
Int Immunopharmacol ; 60: 121-131, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29729496

ABSTRACT

Persistent inflammation and remodeling of airways are the major hallmarks of asthma. Though airway inflammation diminishes in ovalbumin (OVA)-based mouse model of chronic asthma owing to immune-tolerance linked with repeated allergen exposure, which limits the application of the disease model. Accordingly, the present study was designed to develop a murine model of chronic asthma which presents persistent airway inflammation coupled with remodeling traits. Herein, OVA-sensitized BALB/c mice were challenged with increasing (modified protocol) or constant concentration (conventional protocol) of the allergen for 6 weeks; 3 times/week. The results, indeed, revealed that mice subjected to modified protocol demonstrate an improved response to the allergen as reflected by the significant increase in inflammatory cells particularly, eosinophils in bronchoalveolar lavage fluid compared to conventional protocol. Moreover, the expression of Th2 cytokines and their responsible transcription factors (GATA-3 and STAT-6) was markedly enhanced in lungs. The increase in inflammation was further accompanied by a marked increase in mucus production, collagen deposition, and the expression of allied factors (Muc5ac, Col1α1, and α-SMA). Interestingly, pre-treatment of dexamethasone, a corticosteroid (0.5 mg/kg b.wt., i.p.), suppressed the allergen-induced airway inflammation and mucus production without altering collagen deposition. Failure of dexamethasone seems to be related to their ineffectiveness to modulate the expression of TGF-ß, MMP-9, COL1α1, and α-SMA. Overall, our results strongly suggest that mice underwent modified chronic protocol bears more resemblance with asthmatics as it imitates persistent airway inflammation allied with steroid-refractory remodeling traits; hence, may be useful for the evaluation of new/alternative drugs in steroid-refractory asthmatic conditions.


Subject(s)
Allergens/immunology , Asthma/immunology , Ovalbumin/immunology , Acute Disease , Animals , Bronchoalveolar Lavage Fluid/cytology , Chronic Disease , Cytokines/genetics , Disease Models, Animal , Immune Tolerance , Leukocyte Count , Lung/immunology , Male , Mice, Inbred BALB C , RNA, Messenger/metabolism
11.
Front Immunol ; 8: 1172, 2017.
Article in English | MEDLINE | ID: mdl-28974953

ABSTRACT

Asthma, acute lung injury (ALI), and chronic obstructive pulmonary disease (COPD) are lung inflammatory disorders with a common outcome, that is, difficulty in breathing. Corticosteroids, a class of potent anti-inflammatory drugs, have shown less success in the treatment/management of these disorders, particularly ALI and COPD; thus, alternative therapies are needed. Poly(ADP-ribose)polymerases (PARPs) are the post-translational modifying enzymes with a primary role in DNA repair. During the last two decades, several studies have reported the critical role played by PARPs in a good of inflammatory disorders. In the current review, the studies that address the role of PARPs in asthma, ALI, and COPD have been discussed. Among the different members of the family, PARP-1 emerges as a key player in the orchestration of lung inflammation in asthma and ALI. In addition, PARP activation seems to be associated with the progression of COPD. Furthermore, PARP-14 seems to play a crucial role in asthma. STAT-6 and GATA-3 are reported to be central players in PARP-1-mediated eosinophilic inflammation in asthma. Interestingly, oxidative stress-PARP-1-NF-κB axis appears to be tightly linked with inflammatory response in all three-lung diseases despite their distinct pathophysiologies. The present review sheds light on PARP-1-regulated factors, which may be common or differential players in asthma/ALI/COPD and put forward our prospective for future studies.

SELECTION OF CITATIONS
SEARCH DETAIL
...