Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 14: 886601, 2023.
Article in English | MEDLINE | ID: mdl-36960058

ABSTRACT

Introduction: Pulmonary fibrosis is a destructive, progressive disease that dramatically reduces life quality of patients, ultimately leading to death. Therapeutic regimens for pulmonary fibrosis have shown limited benefits, hence justifying the efforts to evaluate the outcome of alternative treatments. Methods: Using a mouse model of bleomycin (BLM)-induced lung fibrosis, in the current work we asked whether treatment with pro-resolution molecules, such as pro-resolving lipid mediators (SPMs) could ameliorate pulmonary fibrosis. To this end, we injected aspirin-triggered resolvin D1 (7S,8R,17R-trihydroxy-4Z,9E,11E,13Z,15E19Z-docosahexaenoic acid; ATRvD1; i.v.) 7 and 10 days after BLM (intratracheal) challenge and samples were two weeks later. Results and discussion: Assessment of outcome in the lung tissues revealed that ATRvD1 partially restored lung architecture, reduced leukocyte infiltration, and inhibited formation of interstitial edema. In addition, lung tissues from BLM-induced mice treated with ATRvD1 displayed reduced levels of TNF-α, MCP-1, IL-1-ß, and TGF-ß. Of further interest, ATRvD1 decreased lung tissue expression of MMP-9, without affecting TIMP-1. Highlighting the beneficial effects of ATRvD1, we found reduced deposition of collagen and fibronectin in the lung tissues. Congruent with the anti-fibrotic effects that ATRvD1 exerted in lung tissues, α-SMA expression was decreased, suggesting that myofibroblast differentiation was inhibited by ATRvD1. Turning to culture systems, we next showed that ATRvD1 impaired TGF-ß-induced fibroblast differentiation into myofibroblast. After showing that ATRvD1 hampered extracellular vesicles (EVs) release in the supernatants from TGF-ß-stimulated cultures of mouse macrophages, we verified that ATRvD1 also inhibited the release of EVs in the bronco-alveolar lavage (BAL) fluid of BLM-induced mice. Motivated by studies showing that BLM-induced lung fibrosis is linked to angiogenesis, we asked whether ATRvD1 could blunt BLM-induced angiogenesis in the hamster cheek pouch model (HCP). Indeed, our intravital microscopy studies confirmed that ATRvD1 abrogates BLM-induced angiogenesis. Collectively, our findings suggest that treatment of pulmonary fibrosis patients with ATRvD1 deserves to be explored as a therapeutic option in the clinical setting.


Subject(s)
Pulmonary Fibrosis , Humans , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Aspirin/pharmacology , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Lung/pathology , Bleomycin/pharmacology , Transforming Growth Factor beta/metabolism
2.
Int J Mol Sci ; 22(21)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34769064

ABSTRACT

Novel strategies for the prevention and treatment of sepsis-associated acute kidney injury and its long-term outcomes have been required and remain a challenge in critical care medicine. Therapeutic strategies using lipid mediators, such as aspirin-triggered resolvin D1 (ATRvD1), can contribute to the resolution of acute and chronic inflammation. In this study, we examined the potential effect of ATRvD1 on long-term kidney dysfunction after severe sepsis. Fifteen days after cecal ligation and puncture (CLP), sepsis-surviving BALB/c mice were subjected to a tubulointerstitial injury through intraperitoneal injections of bovine serum albumin (BSA) for 7 days, called the subclinical acute kidney injury (subAKI) animal model. ATRvD1 treatment was performed right before BSA injections. On day 22 after CLP, the urinary protein/creatinine ratio (UPC), histologic parameters, fibrosis, cellular infiltration, apoptosis, inflammatory markers levels, and mRNA expression were determined. ATRvD1 treatment mitigated tubulointerstitial injury by reducing proteinuria excretion, the UPC ratio, the glomerular cell number, and extracellular matrix deposition. Pro-fibrotic markers, such as transforming growth factor ß (TGFß), type 3 collagen, and metalloproteinase (MMP)-3 and -9 were reduced after ATRvD1 administration. Post-septic mice treated with ATRvD1 were protected from the recruitment of IBA1+ cells. The interleukin-1ß (IL-1ß) levels were increased in the subAKI animal model, being attenuated by ATRvD1. Tumor necrosis factor-α (TNF-α), IL-10, and IL-4 mRNA expression were increased in the kidney of BSA-challenged post-septic mice, and it was also reduced after ATRvD1. These results suggest that ATRvD1 protects the kidney against a second insult such as BSA-induced tubulointerstitial injury and fibrosis by suppressing inflammatory and pro-fibrotic mediators in renal dysfunction after sepsis.


Subject(s)
Acute Kidney Injury/drug therapy , Aspirin/pharmacology , Docosahexaenoic Acids/pharmacology , Kidney Glomerulus/drug effects , Sepsis/drug therapy , Acute Kidney Injury/chemically induced , Albumins/pharmacology , Animals , Biomarkers/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Inflammation/drug therapy , Inflammation/metabolism , Kidney Function Tests/methods , Kidney Glomerulus/metabolism , Male , Mice , Mice, Inbred BALB C , Proteinuria/chemically induced , Proteinuria/drug therapy , Proteinuria/metabolism , RNA, Messenger/metabolism , Sepsis/metabolism
3.
PLoS One ; 10(7): e0133227, 2015.
Article in English | MEDLINE | ID: mdl-26197455

ABSTRACT

Sepsis is a deadly disease characterized by an overwhelming release of inflammatory mediators and the activation of different types of cells. This altered state of cell activation, termed leukocyte reprogramming, contributes to patient outcome. However, the understanding of the process underlying sepsis and the role of regulatory T cells (Tregs) in sepsis remains to be elucidated. In this study, we investigated the role of CCR4, the CCL17/CCL22 chemokine receptor, in the innate and acquired immune responses during severe sepsis and the role of Tregs in effecting the outcome. In contrast with wild-type (WT) mice subjected to cecal ligation and puncture (CLP) sepsis, CCR4-deficient (CCR4-/-) septic mice presented an increased survival rate, significant neutrophil migration toward the infection site, a low bacterial count in the peritoneum, and reduced lung inflammation and serum cytokine levels. Thus, a better early host response may favor an adequate long-term response. Consequently, the CCR4-/- septic mice were not susceptible to secondary fungal infection, in contrast with the WT septic mice. Furthermore, Tregs cells from the CCR4-/- septic mice showed reduced suppressive effects on neutrophil migration (both in vivo and in vitro), lymphocyte proliferation and ROS production from activated neutrophils, in contrast with what was observed for Tregs from the WT septic mice. These data show that CCR4 is involved in immunosuppression after severe sepsis and suggest that CCR4+ Tregs negatively modulate the short and long-term immune responses.


Subject(s)
Receptors, CCR4/immunology , Sepsis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Female , Gene Deletion , Interleukin-10/immunology , Male , Mice , Mice, Inbred C57BL , Neutrophil Activation , Reactive Oxygen Species/immunology , Receptors, CCR4/genetics , Sepsis/genetics , Sepsis/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/immunology
4.
J Invest Dermatol ; 134(5): 1436-1445, 2014 May.
Article in English | MEDLINE | ID: mdl-24226420

ABSTRACT

Lipid mediators derived from 5-lipoxygenase (5-LO) metabolism can activate both pro- and anti-inflammatory pathways, but their role in wound healing remains largely unexplored. In this study we show that 5-LO knockout (5-LO(-/-)) mice exhibited faster wound healing than wild-type (WT) animals, and exhibited upregulation of heme oxygenase-1 (HO-1). Furthermore, HO-1 inhibition in 5-LO(-/-) mice abolished the beneficial effect observed. Despite the fact that 5-LO(-/-) mice exhibited faster healing, in in vitro assays both migration and proliferation of human dermal fibroblasts (HDFs) were inhibited by the 5-LO pharmacologic inhibitor AA861. No changes were observed in the expression of fibronectin, transforming growth factor (I and III), and α-smooth muscle actin (α-SMA). Interestingly, AA861 treatment significantly decreased ROS formation by stimulated fibroblasts. Similar to 5-LO(-/-) mice, induction of HO-1, but not superoxide dismutase-2 (SOD-2), was also observed in response to 5-LO (AA861) or 5-LO activating protein (MK886) inhibitors. HO-1 induction was independent of nuclear factor (erythroid derived-2) like2 (Nrf-2), cyclooxygenase 2 (COX-2) products, or lipoxin action. Taken together, our results show that 5-LO disruption improves wound healing and alters fibroblast function by an antioxidant mechanism based on HO-1 induction. Overexpression of HO-1 in wounds may facilitate early wound resolution.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Dermatitis/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Wound Healing/physiology , Adult , Animals , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/immunology , Dermatitis/genetics , Dermatitis/immunology , Dermis/cytology , Dermis/immunology , Dermis/metabolism , Disease Models, Animal , Fibroblasts/immunology , Fibroblasts/metabolism , Heme Oxygenase-1/immunology , Humans , Male , Membrane Proteins/immunology , Mice , Mice, 129 Strain , Mice, Knockout , Oxidative Stress/physiology , Primary Cell Culture , Reactive Oxygen Species/metabolism
5.
Am J Respir Cell Mol Biol ; 49(6): 1029-37, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23848293

ABSTRACT

No successful therapies are available for pulmonary fibrosis, indicating the need for new treatments. Lipoxins and their 15-epimers, aspirin-triggered lipoxins (ATL), present potent antiinflammatory and proresolution effects (Martins et al., J Immunol 2009;182:5374-5381). We show that ATLa, an ATL synthetic analog, therapeutically reversed a well-established pulmonary fibrotic process induced by bleomycin (BLM) in mice. We investigated the mechanisms involved in its effect and found that systemic treatment with ATLa 1 week after BLM instillation considerably reversed the inflammatory response, total collagen and collagen type 1 deposition, vascular endothelial growth factor, and transforming growth factor (TGF)-ß expression in the lung and restored surfactant protein C expression levels. ATLa also inhibited BLM-induced apoptosis and cellular accumulation in bronchoalveolar lavage fluid and in the lung parenchyma as evaluated by light microscopy and flow cytometry (Ly6G(+), F4/80(+), CD11c(+), CD4(+), and B220(+) cells) assays. Moreover, ATLa inhibited the lung production of IL-1ß, IL-17, TNF-α, and TGF-ß induced by BLM-challenged mice. ATLa restored the balance of inducible nitric oxide synthase-positive and arginase-positive cells in the lungs, suggesting a prevalence of M2 versus M1 macrophages. Together, these effects improved pulmonary mechanics because ATLa treatment brought to normal levels lung resistance and elastance, which were clearly altered at 7 days after BLM challenge. Our findings support ATLa as a promising therapeutic agent to treat lung fibrosis.


Subject(s)
Lipoxins/therapeutic use , Pulmonary Fibrosis/drug therapy , Animals , Arginase/metabolism , Aspirin/therapeutic use , Biomarkers/metabolism , Bleomycin/toxicity , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cytokines/biosynthesis , Disease Models, Animal , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Peptides/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , Pulmonary Surfactant-Associated Protein C , Respiratory Mechanics/drug effects , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...