Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
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
2.
Brain ; 141(6): 1609-1621, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29741608

ABSTRACT

Dermatomyositis is an acquired auto-immune disease characterized by skin lesions and muscle-specific pathological features such as perifascicular muscle fibre atrophy and vasculopathy. Dermatomyositis patients display an upregulation of type I interferon-inducible genes in muscle fibres, endothelial cells, skin and peripheral blood. However, the effect of type I interferon on muscle tissue has not yet been determined. Our aim was to study the pathogenicity of type I interferon in vitro and to evaluate the efficacy of the type I interferon pathway blockade for therapeutic purposes. The activation of type I interferon in differentiating myoblasts abolished myotube formation with reduced myogenin expression while in differentiated myotubes, we observed a reduction in surface area and an upregulation of atrophy-associated genes. In vitro endothelial cells exposure to type I interferon disrupted vascular network organization. All the pathogenic effects observed in vitro were abolished by ruxolitinib. Finally, four refractory dermatomyositis patients were treated with ruxolitinib and improvement ensued in skin lesions, muscle weakness and a reduced serum type I interferon levels and interferon-inducbile genes scores. We propose JAK inhibition as a mechanism-based treatment for dermatomyositis, a finding that is relevant for the design of future clinical trials targeting dermatomyositis.


Subject(s)
Dermatomyositis , Interferon Type I/toxicity , Janus Kinase Inhibitors/therapeutic use , Muscle, Skeletal/drug effects , Pyrazoles/therapeutic use , Signal Transduction/drug effects , Aged , Aged, 80 and over , Cell Line, Transformed , Dermatomyositis/chemically induced , Dermatomyositis/drug therapy , Dermatomyositis/pathology , Endothelial Cells/drug effects , Female , Humans , Male , Middle Aged , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Neovascularization, Pathologic/chemically induced , Nitriles , Pyrimidines , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Up-Regulation/drug effects
3.
Mediators Inflamm ; 2017: 2086840, 2017.
Article in English | MEDLINE | ID: mdl-28894350

ABSTRACT

TLRs recognize a broad spectrum of microorganism molecules, triggering a variety of cellular responses. Among them, phagocytosis is a critical process for host defense. Leukotrienes (LTs), lipid mediators produced from 5-lipoxygenase (5-LO) enzyme, increase FcγR-mediated phagocytosis. Here, we evaluated the participation of TLR2, TLR3, TLR4, and TLR9 in FcγR-mediated phagocytosis and whether this process is modulated by LTs. Rat alveolar macrophages (AMs), murine bone marrow-derived macrophages (BMDMs), and peritoneal macrophages (PMs) treated with TLR2, TLR3, and TLR4 agonists, but not TLR9, enhanced IgG-opsonized sheep red blood cell (IgG-sRBC) phagocytosis. Pretreatment of AMs or BMDMs with drugs that block LT synthesis impaired the phagocytosis promoted by TLR ligands, and TLR potentiation was also abrogated in PMs and BMDMs from 5-LO-/- mice. LTB4 production induced by IgG engagement was amplified by TLR ligands, while cys-LTs were amplified by activation of TLR2 and TLR4, but not by TLR3. We also noted higher ERK1/2 phosphorylation in IgG-RBC-challenged cells when preincubated with TLR agonists. Furthermore, ERK1/2 inhibition by PD98059 reduced the phagocytic activity evoked by TLR agonists. Together, these data indicate that TLR2, TLR3, and TLR4 ligands, but not TLR9, amplify IgG-mediated phagocytosis by a mechanism which requires LT production and ERK-1/2 pathway activation.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Animals , Arachidonate 5-Lipoxygenase/genetics , Flavonoids/pharmacology , Immunoblotting , Leukotrienes/metabolism , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Knockout , Phagocytosis/drug effects , Phagocytosis/genetics , Phagocytosis/physiology , Phosphorylation/drug effects , Phosphorylation/genetics , Rats , Rats, Wistar , Sheep , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
4.
Am J Respir Cell Mol Biol ; 50(1): 87-95, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23947598

ABSTRACT

Postsepsis lung injury is a common clinical problem associated with significant morbidity and mortality. Leukotrienes (LTs) are important lipid mediators of infection and inflammation derived from the 5-lipoxygenase (5-LO) metabolism of arachidonate with the potential to contribute to lung damage after sepsis. To test the hypothesis that LTs are mediators of lung injury after sepsis, we assessed lung structure, inflammatory mediators, and mechanical changes after cecal ligation and puncture surgery in wild-type (WT) and 5-LO knockout (5-LO(-/-)) mice and in WT mice treated with a pharmacologic LT synthesis inhibitor (MK886) and LT receptor antagonists (CP105,696 and montelukast). Sixteen hours after surgery, WT animals exhibited severe lung injury (by histological analysis), substantial mechanical impairment (i.e., an increase in static lung elastance), an increase in neutrophil infiltration, and high levels of LTB4, cysteinyl-LTs (cys-LTs), prostaglandin E2, IL-1ß, IL-6, IL-10, IL-17, KC (CXCL1), and monocyte chemotactic protein-1 (CCL2) in lung tissue and plasma. 5-LO(-/-) mice and WT mice treated with a pharmacologic 5-LO inhibitor were significantly protected from lung inflammation and injury. Selective antagonists for BLT1 or cys-LT1, the high-affinity receptors for LTB4 and cys-LTs, respectively, were insufficient to provide protection when used alone. These results point to an important role for 5-LO products in sepsis-induced lung injury and suggest that the use of 5-LO inhibitors may be of therapeutic benefit clinically.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Lung Injury/metabolism , Sepsis/metabolism , Signal Transduction/physiology , Animals , Cecum/drug effects , Cecum/metabolism , Cytokines/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Leukotriene Antagonists/pharmacology , Leukotriene B4/metabolism , Lung Injury/drug therapy , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Receptors, Leukotriene/metabolism , Receptors, Leukotriene B4/metabolism , Sepsis/drug therapy , Signal Transduction/drug effects
5.
Int Wound J ; 11(2): 190-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-22905783

ABSTRACT

Silver is used worldwide in dressings for wound management. Silver has demonstrated great efficacy against a broad range of microorganisms, but there is very little data about the systemic absorption and toxicity of silver in vivo. In this study, the antimicrobial effect of the silver-coated dressing (SilverCoat(®)) was evaluated in vitro against the most common microorganisms found in wounds, including Pseudomonas aeruginosa, Candida albicans, Staphylococcus aureus, Methicillin-resistant Staphylococcus aureus and Klebsiella pneumoniae. We also performed an excisional skin lesion assay in mice to evaluate wound healing after 14 days of treatment with a silver-coated dressing, and we measured the amount of silver in the blood, the kidneys and the liver after treatment. Our data demonstrated that the nylon threads coated with metallic silver have a satisfactory antimicrobial effect in vitro, and the prolonged use of these threads did not lead to systemic silver absorption, did not induce toxicity in the kidneys and the liver and were not detrimental to the normal wound-healing process.


Subject(s)
Bandages , Polyesters/pharmacology , Polyethylenes/pharmacology , Silver/administration & dosage , Wound Healing , Animals , Argyria/epidemiology , Cell Survival , Kidney/drug effects , Liver/drug effects , Male , Malondialdehyde/analysis , Mice , Polyesters/administration & dosage , Polyethylenes/administration & dosage , Wound Healing/drug effects , Wound Healing/physiology , Wound Infection/prevention & control , Wounds and Injuries/microbiology
6.
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
7.
Crit Care Med ; 41(4): 1056-68, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23385098

ABSTRACT

OBJECTIVE: It is well known that sepsis causes damage in different organs, including kidneys. However, few studies have been conducted on the magnitude of the long-term effects of sepsis on the surviving population, in particular, in relation to kidney disease. In this study, we examined the impact of long-term effects of sepsis on a second kidney insult. DESIGN: Prospective experimental study. SETTING: University research laboratory. INTERVENTIONS: Wild-type mice were subjected to the cecal ligation and puncture sepsis model. Control animals underwent identical laparotomy but without ligation and cecum puncture. On days 0, 7, and 14 after surgery, the ratio between urinary protein and creatinine was measured. Fifteen days after surgery, surviving mice were subjected to a second kidney insult through intraperitoneal injections of bovine serum albumin for 7 days. On day 22 after surgery, urinary protein and creatinine, γ-glutamyl transpeptidase, lactate dehydrogenase, histologic parameters, macrophage infiltration, apoptotic cell, renal and plasmatic cytokines were determined. MEASUREMENTS AND MAIN RESULTS: On days 7 and 14 after surgery, the urinary protein and creatinine observed in the septic animal group were higher than those observed in the control group. On day 22 after surgery, sepsis-surviving animals that were subjected to a second kidney insult showed more severe tubular injury compared with controls. This process seems to involve an immunosuppressive state because the concentrations of some renal cytokines, such as tumor necrosis factor-α, interleukin 6, interferon-γ and chemokine ligand 2, were decreased and leukocyte numbers were increased. CONCLUSIONS: These results suggest that sepsis induces long-term effects in kidney structure aggravating tubule damage in a second kidney insult.


Subject(s)
Acute Kidney Injury/pathology , Acute Kidney Injury/urine , Shock, Septic/pathology , Shock, Septic/urine , Acute Kidney Injury/diagnosis , Animals , Biomarkers/urine , Cecum , Disease Models, Animal , Mice , Prospective Studies , Punctures , Random Allocation , Reference Values
8.
J Immunol ; 186(11): 6562-7, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21536805

ABSTRACT

High concentrations of free heme found during hemolytic events or cell damage leads to inflammation, characterized by neutrophil recruitment and production of reactive oxygen species, through mechanisms not yet elucidated. In this study, we provide evidence that heme-induced neutrophilic inflammation depends on endogenous activity of the macrophage-derived lipid mediator leukotriene B(4) (LTB(4)). In vivo, heme-induced neutrophil recruitment into the peritoneal cavity of mice was attenuated by pretreatment with 5-lipoxygenase (5-LO) inhibitors and leukotriene B(4) receptor 1 (BLT1) receptor antagonists as well as in 5-LO knockout (5-LO(-/-)) mice. Heme administration in vivo increased peritoneal levels of LTB(4) prior to and during neutrophil recruitment. Evidence that LTB(4) was synthesized by resident macrophages, but not mast cells, included the following: 1) immuno-localization of heme-induced LTB(4) was compartmentalized exclusively within lipid bodies of resident macrophages; 2) an increase in the macrophage population enhanced heme-induced neutrophil migration; 3) depletion of resident mast cells did not affect heme-induced LTB(4) production or neutrophil influx; 4) increased levels of LTB(4) were found in heme-stimulated peritoneal cavities displaying increased macrophage numbers; and 5) in vitro, heme was able to activate directly macrophages to synthesize LTB(4). Our findings uncover a crucial role of LTB(4) in neutrophil migration induced by heme and suggest that beneficial therapeutic outcomes could be achieved by targeting the 5-LO pathway in the treatment of inflammation associated with hemolytic processes.


Subject(s)
Cell Movement/drug effects , Heme/pharmacology , Leukotriene B4/metabolism , Neutrophils/drug effects , Animals , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Cells, Cultured , Female , Macrophages/drug effects , Macrophages/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Neutrophils/metabolism , Receptors, Leukotriene B4/metabolism , Thioglycolates/pharmacology , p-Methoxy-N-methylphenethylamine/pharmacology
9.
Trop Med Infect Dis ; 8(7)2023 Jun 25.
Article in English | MEDLINE | ID: mdl-37505632

ABSTRACT

Neutrophils are multifaceted cells that, upon activation, release meshes of chromatin associated with different proteins, known as neutrophil extracellular traps (NETs). Leishmania amazonensis promastigotes and amastigotes induce NET release, and we have identified the signaling pathways involved in NET extrusion activated by promastigotes. Amastigotes maintain the infection in vertebrate hosts, and we have shown the association of NETs with amastigotes in human biopsies of cutaneous leishmaniasis. However, the interaction of amastigotes and neutrophils remains poorly understood. Our study aimed to characterize the pathways involved in the formation of NETs induced by axenic amastigotes from L. infantum, the causal agent of visceral leishmaniasis. Human neutrophils pretreated with signaling pathway inhibitors were incubated with amastigotes, and NET release was quantified in the culture supernatant. Amastigote viability was checked after incubation with NETs. We found that the release of NETs by neutrophils stimulated with these amastigotes requires the participation of elastase and peptidyl arginine deaminase and the involvement of PI3K, ROS, and calcium. Moreover, amastigotes are not susceptible to NET-mediated killing. Altogether, these findings improve our comprehension of the signaling pathways implicated in the interaction between amastigotes and human neutrophils.

10.
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
11.
J Immunol ; 182(9): 5374-81, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380784

ABSTRACT

Despite an increase in the knowledge of mechanisms and mediators involved in pulmonary fibrosis, there are no successful therapeutics available. Lipoxins (LX) and their 15-epimers, aspirin-triggered LX (ATL), are endogenously produced eicosanoids with potent anti-inflammatory and proresolution effects. To date, few studies have been performed regarding their effect on pulmonary fibrosis. In the present study, using C57BL/6 mice, we report that bleomycin (BLM)-induced lung fibrosis was prevented by the concomitant treatment with an ATL synthetic analog, ATLa, which reduced inflammation and matrix deposition. ATLa inhibited BLM-induced leukocyte accumulation and alveolar collapse as evaluated by histology and morphometrical analysis. Moreover, Sirius red staining and lung hydroxyproline content showed an increased collagen deposition in mice receiving BLM alone that was decreased upon treatment with the analog. These effects resulted in benefits to pulmonary mechanics, as ATLa brought to normal levels both lung resistance and compliance. Furthermore, the analog improved mouse survival, suggesting an important role for the LX pathway in the control of disease establishment and progression. One possible mechanism by which ATLa restrained fibrosis was suggested by the finding that BLM-induced myofibroblast accumulation/differentiation in the lung parenchyma was also reduced by both simultaneous and posttreatment with the analog (alpha-actin immunohistochemistry). Interestingly, ATLa posttreatment (4 days after BLM) showed similar inhibitory effects on inflammation and matrix deposition, besides the TGF-beta level reduction in the lung, reinforcing an antifibrotic effect. In conclusion, our findings show that LX and ATL can be considered as promising therapeutic approaches to lung fibrotic diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antifibrinolytic Agents/therapeutic use , Aspirin/pharmacology , Bleomycin/toxicity , Lipoxins/therapeutic use , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/prevention & control , Animals , Bleomycin/antagonists & inhibitors , Immunity, Innate/drug effects , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/mortality , Pulmonary Fibrosis/physiopathology , Respiratory Function Tests , Survival Analysis
12.
PLoS Negl Trop Dis ; 15(9): e0009760, 2021 09.
Article in English | MEDLINE | ID: mdl-34492013

ABSTRACT

In insects the reserve proteins are stored in the oocytes into endocytic-originated vesicles named yolk organelles. VPS38/UVRAG and ATG14 are the variant regulatory subunits of two class-III ATG6/Beclin1 PI3K complexes that regulate the recruitment of the endocytic (complex II) and autophagic (complex I) machineries. In a previous work from our group, we found that the silencing of ATG6/Beclin1 resulted in the formation of yolk-deficient oocytes due to defects in the endocytosis of the yolk proteins. Because ATG6/Beclin1 is present in the two above-described PI3K complexes, we could not identify the contributions of each complex to the yolk defective phenotypes. To address this, here we investigated the role of the variant subunits VPS38/UVRAG (complex II, endocytosis) and ATG14 (complex I, autophagy) in the biogenesis of the yolk organelles in the insect vector of Chagas Disease Rhodnius prolixus. Interestingly, the silencing of both genes phenocopied the silencing of ATG6/Beclin1, generating 1) accumulation of yolk proteins in the hemolymph; 2) white, smaller, and yolk-deficient oocytes; 3) abnormal yolk organelles in the oocyte cortex; and 4) unviable F1 embryos. However, we found that the similar phenotypes were the result of a specific cross-silencing effect among the PI3K subunits where the silencing of VPS38/UVRAG and ATG6/Beclin1 resulted in the specific silencing of each other, whereas the silencing of ATG14 triggered the silencing of all three PI3K components. Because the silencing of VPS38/UVRAG and ATG6/Beclin1 reproduced the yolk-deficiency phenotypes without the cross silencing of ATG14, we concluded that the VPS38/UVRAG PI3K complex II was the major contributor to the previously observed phenotypes in silenced insects. Altogether, we found that class-III ATG6/Beclin1 PI3K complex II (VPS38/UVRAG) is essential for the yolk endocytosis and that the subunits of both complexes are under an unknown transcriptional regulatory system.


Subject(s)
Beclin-1/metabolism , Insect Proteins/metabolism , Oocytes/physiology , Organelles/physiology , Phosphatidylinositol 3-Kinases/metabolism , Rhodnius/physiology , Animals , Beclin-1/genetics , Chagas Disease/transmission , Egg Yolk/physiology , Gene Expression Regulation , Gene Silencing , Insect Proteins/genetics , Insect Vectors/physiology , Oocytes/cytology , Phosphatidylinositol 3-Kinases/genetics
13.
J Gen Virol ; 91(Pt 1): 259-64, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19793908

ABSTRACT

Feline leukemia virus (FeLV), a common, naturally occurring gammaretrovirus in domestic cats, is associated with degenerative diseases of the haematopoietic system, immunodeficiency and neoplasia. FeLV infection causes an important suppression of neutrophil function, leading to opportunistic infections. Recently, a new microbicidal mechanism named NETosis was described in human, bovine and fish neutrophils, as well as in chicken heterophils. The purpose of the present study was to characterize NETosis in feline neutrophils, as well as to evaluate neutrophil function in FeLV naturally infected symptomatic and asymptomatic cats through the phagocytosis process, release of neutrophil extracellular traps (NETs) and myeloperoxidase (MPO) activity. The results showed that feline neutrophils stimulated with protozoa parasites released structures comprising DNA and histones, which were characterized as NETs by immunofluorescence. Quantification of NETs after neutrophil stimulation showed a significant increase in NET release by neutrophils from FeLV(-) and FeLV(+) asymptomatic cats compared with FeLV(+) symptomatic cats. Moreover, the number of released NETs and MPO activity in unstimulated neutrophils of FeLV(+) symptomatic cats were higher than those in unstimulated neutrophils from FeLV(-) and FeLV(+) asymptomatic cats. This study reports, for the first time, NET release by feline neutrophils, along with the fact that NET induction may be modulated by a viral infection. The results indicate that the NET mechanism appears to be overactivated in FeLV(+) cats and that this feature could be considered a marker of disease progression in FeLV infection.


Subject(s)
Cat Diseases/immunology , Cat Diseases/virology , Leukemia Virus, Feline/immunology , Neutrophils/immunology , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Cats , DNA/metabolism , Histones/metabolism , Peroxidase/metabolism , Phagocytosis , Retroviridae Infections/immunology , Retroviridae Infections/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
14.
Front Physiol ; 11: 573347, 2020.
Article in English | MEDLINE | ID: mdl-33071827

ABSTRACT

Growing evidence demonstrates a continuous interaction between the immune system and the skeletal muscle in inflammatory diseases of different pathogenetic origins, in dystrophic conditions such as Duchenne Muscular Dystrophy as well as during normal muscle regeneration. Although one component of the innate immunity, the macrophage, has been extensively studied both in disease conditions and during cell or gene therapy strategies aiming at restoring muscular functions, much less is known about dendritic cells and their primary immunological targets, the T lymphocytes. This review will focus on the dendritic cells and T lymphocytes (including effector and regulatory T-cells), emphasizing the potential cross talk between these cell types and their influence on the structure and function of skeletal muscle.

15.
Int Immunol ; 20(1): 129-39, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18056919

ABSTRACT

In the present study, we show that the intra-thoracic injection of ovalbumin (OVA, 12.5 microg per cavity) into C57BL/10 mice induced a significant increase in gammadelta T lymphocyte numbers in the pleural cavity, blood and thoracic lymph node of challenged mice. Such increase was significant within 12 h, peaked within 48 h and returned to basal counts within 120 h. Levels of CC chemokine ligand (CCL)-2/monocyte chemotactic protein-1, CCL5/regulated upon activation, normal T cell expressed and secreted, CCL3/macrophage inflammatory protein-1 alpha and CCL25/thymus-expressed chemokine were above control values in pleural washes recovered 24 h after OVA challenge (OPW) and were likely produced by pleural macrophages and mesothelial cells. Antigenic challenge also induced an up-regulation in CC chemokine receptor (CCR)-2, CCR5 and CCR9 on gammadelta T cells from pleural cavities, blood and lymph nodes, suggesting that cells found in mice pleural cavity migrate from secondary lymphoid organs into the inflammatory site via blood stream. The in vitro neutralization of CCL2 (but not of CCL3, CCL5 or CCL25) abrogated OPW-induced gammadelta T lymphocyte transmigration. Confirming such results, the in vivo administration of alpha-CCL2 mAb inhibited gammadelta T lymphocyte accumulation in the pleural cavity of challenged mice, whereas the blockade of CCL3, CCL5 or CCL25 showed no effect on gammadelta T cell mobilization. In addition, OVA challenge failed to induce gammadelta T lymphocyte accumulation in the pleural cavity of C57BL/6 CCR2 knockout mice, which also showed decreased numbers of these cells in blood and lymph nodes when compared with wild-type mice. Overall, such results demonstrate that CCR2/CCL2 pathway is crucial for gammadelta T lymphocyte mobilization during the allergic response.


Subject(s)
Chemokine CCL2/metabolism , Hypersensitivity/immunology , Pleurisy/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, CCR2/metabolism , T-Lymphocytes , Animals , Cells, Cultured , Chemokine CCL2/genetics , Chemotaxis, Leukocyte/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Ovalbumin/immunology , Pleura/cytology , Pleura/immunology , Receptors, CCR2/genetics , T-Lymphocytes/immunology , T-Lymphocytes/physiology
16.
J Invest Dermatol ; 139(5): 1161-1170, 2019 05.
Article in English | MEDLINE | ID: mdl-30465800

ABSTRACT

Wound healing is a well-coordinated process that involves inflammatory mediators and cellular responses; however, if any disturbances are present during this process, tissue repair is impaired. Chronic wounds are one of the serious long-term complications associated with diabetes mellitus. The chemokine receptor CCR4 and its respective ligands, CCL17 and CCL22, are involved in regulatory T cell recruitment and activation in inflamed skin; however, the role of regulatory T cells in wounds is still not clear. Our aim was to investigate the role of CCR4 and regulatory T cells in cutaneous wound healing in diabetic mice. Alloxan-induced diabetic wild- type mice (diabetic) developed wounds that were difficult to heal, differently from CCR4-/- diabetic mice (CCR4-/- diabetic), and also from anti-CCL17/22 or anti-CD25-injected diabetic mice that presented with accelerated wound healing and fewer regulatory T cells in the wound bed. Consequently, CCR4-/- diabetic mice also presented with alteration on T cells population in the wound and draining lymph nodes; on day 14, these mice also displayed an increase of collagen fiber deposition. Still, cytokine levels were decreased in the wounds of CCR4-/- diabetic mice on day 2. Our data suggest that the receptor CCR4 and regulatory T cells negatively affect wound healing in diabetic mice.


Subject(s)
Chemokine CCL17/antagonists & inhibitors , Chemokine CCL22/antagonists & inhibitors , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Receptors, CCR4/metabolism , Wound Healing/drug effects , Alloxan/pharmacology , Analysis of Variance , Animals , Biopsy, Needle , Chemokine CCL17/pharmacology , Chemokine CCL22/pharmacology , Chemokines/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/drug therapy , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction/methods , Wound Healing/physiology
17.
Cell Death Dis ; 9(5): 551, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29748534

ABSTRACT

Idiopathic Inflammatory Myopathies (IIMs) are a heterogeneous group of autoimmune diseases affecting skeletal muscle tissue homeostasis. They are characterized by muscle weakness and inflammatory infiltration with tissue damage. Amongst the cells in the muscle inflammatory infiltration, dendritic cells (DCs) are potent antigen-presenting and key components in autoimmunity exhibiting an increased activation in inflamed tissues. Since, the IIMs are characterized by the focal necrosis/regeneration and muscle atrophy, we hypothesized that DCs may play a role in these processes. Due to the absence of a reliable in vivo model for IIMs, we first performed co-culture experiments with immature DCs (iDC) or LPS-activated DCs (actDC) and proliferating myoblasts or differentiating myotubes. We demonstrated that both iDC or actDCs tightly interact with myoblasts and myotubes, increased myoblast proliferation and migration, but inhibited myotube differentiation. We also observed that actDCs increased HLA-ABC, HLA-DR, VLA-5, and VLA-6 expression and induced cytokine secretion on myoblasts. In an in vivo regeneration model, the co-injection of human myoblasts and DCs enhanced human myoblast migration, whereas the absolute number of human myofibres was unchanged. In conclusion, we suggest that in the early stages of myositis, DCs may play a crucial role in inducing muscle-damage through cell-cell contact and inflammatory cytokine secretion, leading to muscle regeneration impairment.


Subject(s)
Cell Differentiation , Cell Proliferation , Dendritic Cells/metabolism , Myoblasts, Skeletal/metabolism , Adult , Antigens, Differentiation/biosynthesis , Dendritic Cells/cytology , Female , Humans , Infant, Newborn , Lipopolysaccharides/pharmacology , Male , Middle Aged , Myoblasts, Skeletal/cytology
18.
Mol Neurobiol ; 55(1): 435-444, 2018 01.
Article in English | MEDLINE | ID: mdl-27966074

ABSTRACT

Sepsis survivors frequently develop late cognitive impairment. Because little is known on the mechanisms of post-septic memory deficits, there are no current effective approaches to prevent or treat such symptoms. Here, we subjected mice to severe sepsis induced by cecal ligation and puncture (CLP) and evaluated the sepsis-surviving animals in the open field, novel object recognition (NOR), and step-down inhibitory avoidance (IA) task at different times after surgery. Post-septic mice (30 days post-surgery) failed in the NOR and IA tests but exhibited normal performance when re-evaluated 45 days after surgery. Cognitive impairment in post-septic mice was accompanied by reduced hippocampal levels of proteins involved in synaptic plasticity, including synaptophysin, cAMP response element-binding protein (CREB), CREB phosphorylated at serine residue 133 (CREBpSer133), and GluA1 phosphorylated at serine residue 845 (GluA1pSer845). Expression of tumor necrosis factor α (TNF-α) was increased and brain insulin signaling was disrupted, as indicated by increased hippocampal IRS-1 phosphorylation at serine 636 (IRS-1pSer636) and decreased phosphorylation of IRS-1 at tyrosine 465 (IRS-1pTyr465), in the hippocampus 30 days after CLP. Phosphorylation of Akt at serine 473 (AktpSer473) and of GSK3 at serine 9 (GSK3ßpSer9) were also decreased in hippocampi of post-septic animals, further indicating that brain insulin signaling is disrupted by sepsis. We then treated post-septic mice with liraglutide, a GLP-1 receptor agonist with insulinotropic activity, or TDZD-8, a GSK3ß inhibitor, which rescued NOR memory. In conclusion, these results establish that hippocampal inflammation and disrupted insulin signaling are induced by sepsis and are linked to late memory impairment in sepsis survivors.


Subject(s)
Brain/metabolism , Cognitive Dysfunction/metabolism , Insulin/metabolism , Sepsis/metabolism , Signal Transduction/physiology , Animals , Brain/pathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Exploratory Behavior/physiology , Male , Mice , Sepsis/complications , Sepsis/pathology
19.
Front Immunol ; 9: 1979, 2018.
Article in English | MEDLINE | ID: mdl-30258438

ABSTRACT

In chronic schistosomiasis, liver fibrosis is linked to portal hypertension, which is a condition associated with high mortality and morbidity. High mobility group box 1 (HMGB1) was originally described as a nuclear protein that functions as a structural co-factor in transcriptional regulation. However, HMGB1 can also be secreted into the extracellular milieu under appropriate signal stimulation. Extracellular HMGB1 acts as a multifunctional cytokine that contributes to infection, injury, inflammation, and immune responses by binding to specific cell-surface receptors. HMGB1 is involved in fibrotic diseases. From a clinical perspective, HMGB1 inhibition may represent a promising therapeutic approach for treating tissue fibrosis. In this study, we demonstrate elevated levels of HMGB1 in the sera in experimental mice or in patients with schistosomiasis. Using immunohistochemistry, we demonstrated that HMGB1 trafficking in the hepatocytes of mice suffering from acute schistosomiasis was inhibited by Glycyrrhizin, a well-known HMGB1 direct inhibitor, as well as by DIC, a novel and potential anti-HMGB1 compound. HMGB1 inhibition led to significant downregulation of IL-6, IL4, IL-5, IL-13, IL-17A, which are involved in the exacerbation of the immune response and liver fibrogenesis. Importantly, infected mice that were treated with DIC or GZR to inhibit HMGB1 pro-inflammatory activity showed a significant increase in survival and a reduction of over 50% in the area of liver fibrosis. Taken together, our findings indicate that HMGB1 is a key mediator of schistosomotic granuloma formation and liver fibrosis and may represent an outstanding target for the treatment of schistosomiasis.


Subject(s)
Granuloma , HMGB1 Protein/immunology , Liver Cirrhosis , Liver , Schistosoma mansoni/immunology , Schistosomiasis mansoni , Animals , Cytokines/immunology , Female , Granuloma/immunology , Granuloma/parasitology , Granuloma/pathology , Humans , Liver/immunology , Liver/parasitology , Liver/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/parasitology , Liver Cirrhosis/pathology , Male , Mice, Inbred BALB C , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/pathology
SELECTION OF CITATIONS
SEARCH DETAIL