Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Int J Mol Sci ; 24(4)2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36834844

ABSTRACT

Annexin A1 (ANXA1) is an endogenous protein, which plays a central function in the modulation of inflammation. While the functions of ANXA1 and its exogenous peptidomimetics, N-Acetyl 2-26 ANXA1-derived peptide (ANXA1Ac2-26), in the modulation of immunological responses of neutrophils and monocytes have been investigated in detail, their effects on the modulation of platelet reactivity, haemostasis, thrombosis, and platelet-mediated inflammation remain largely unknown. Here, we demonstrate that the deletion of Anxa1 in mice upregulates the expression of its receptor, formyl peptide receptor 2/3 (Fpr2/3, orthologue of human FPR2/ALX). As a result, the addition of ANXA1Ac2-26 to platelets exerts an activatory role in platelets, as characterised by its ability to increase the levels of fibrinogen binding and the exposure of P-selectin on the surface. Moreover, ANXA1Ac2-26 increased the development of platelet-leukocyte aggregates in whole blood. The experiments carried out using a pharmacological inhibitor (WRW4) for FPR2/ALX, and platelets isolated from Fpr2/3-deficient mice ascertained that the actions of ANXA1Ac2-26 are largely mediated through Fpr2/3 in platelets. Together, this study demonstrates that in addition to its ability to modulate inflammatory responses via leukocytes, ANXA1 modulates platelet function, which may influence thrombosis, haemostasis, and platelet-mediated inflammation under various pathophysiological settings.


Subject(s)
Annexin A1 , Animals , Humans , Mice , Annexin A1/metabolism , Blood Platelets/metabolism , Inflammation/metabolism , Neutrophils/metabolism , Peptides/pharmacology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism
2.
Pulm Pharmacol Ther ; 61: 101896, 2020 04.
Article in English | MEDLINE | ID: mdl-31988027

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is related to smoking and anti-inflammatory therapy is indicated. Among the mediators with anti-inflammatory properties, we highlight piperlongumine (PL), an alkaloid/amide of Piper longum. Here we evaluated the PL administration on an experimental model of respiratory inflammation resulting from exposure to cigarette smoke. Male Balb/c mice were exposed to burning of 10 commercial cigarettes, 2x/day, for five weeks on specific equipment. PL efficacy was evaluated in control, exposed to smoke without treatment and PL treated (2.0 mg/kg, 3x/week) groups. Animals were weighed and plethysmographic analyses performed at the end of the exposure protocol. Inflammatory cells were evaluated in the bronchoalveolar lavage (BAL) and hemoglobin and glucose in the blood. Lung fragments were processed for histopathological studies and AnxA1, COX-2, NF-kB and neutrophil elastase expressions. Plethysmography revealed that PL maintained pulmonary frequency, volume and ventilation parameters similar to controls, with respiratory volume reduction compared to untreated animals. Final weight was reduced in both exposed groups. PL decreased hemoglobin concentration, attenuated the reduction of glucose levels and reduced influx of lymphocytes, neutrophils and macrophages in BAL. Histopathologically occured infiltration of inflammatory cells, increase of the interalveolar septa and intra-alveolar spaces in untreated animals. But, PL administration recovered lung tissues and, immunohistochemically, promoted increased expression of AnxA1 and reduction of COX-2, NF-kB and neutrophil elastase. Together the results indicate that PL attenuates systemic and pulmonary inflammatory changes, partially by modulating the expression the endogenous AnxA1, and may represent a promising therapy in preventing the inflammation induced by cigarette smoke.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dioxolanes/pharmacology , Inflammation/chemically induced , Lung Injury/drug therapy , Pulmonary Disease, Chronic Obstructive/drug therapy , Tobacco Smoking/adverse effects , Animals , Annexin A1/metabolism , Cyclooxygenase 2/metabolism , Lung Injury/metabolism , Lung Injury/pathology , Lung Injury/physiopathology , Lymphocytes/metabolism , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Neutrophils , Tobacco Smoking/metabolism , Tobacco Smoking/pathology , Tobacco Smoking/physiopathology
3.
J Neuroinflammation ; 16(1): 32, 2019 Feb 12.
Article in English | MEDLINE | ID: mdl-30755225

ABSTRACT

BACKGROUND: The inflammatory process has been described as a crucial mechanism in the pathophysiology of temporal lobe epilepsy. The anti-inflammatory protein annexin A1 (ANXA1) represents an interesting target in the regulation of neuroinflammation through the inhibition of leukocyte transmigration and the release of proinflammatory mediators. In this study, the role of the ANXA1-derived peptide Ac2-26 in an experimental model of status epilepticus (SE) was evaluated. METHODS: Male Wistar rats were divided into Naive, Sham, SE and SE+Ac2-26 groups, and SE was induced by intrahippocampal injection of pilocarpine. In Sham animals, saline was applied into the hippocampus, and Naive rats were only handled. Three doses of Ac2-26 (1 mg/kg) were administered intraperitoneally (i.p.) after 2, 8 and 14 h of SE induction. Finally, 24 h after the experiment-onset, rats were euthanized for analyses of neuronal lesion and inflammation. RESULTS: Pilocarpine induced generalised SE in all animals, causing neuronal damage, and systemic treatment with Ac2-26 decreased neuronal degeneration and albumin levels in the hippocampus. Also, both SE groups showed an intense influx of microglia, which was corroborated by high levels of ionised calcium binding adaptor molecule 1(Iba-1) and monocyte chemoattractant protein-1 (MCP-1) in the hippocampus. Ac2-26 reduced the astrocyte marker (glial fibrillary acidic protein; GFAP) levels, as well as interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and growth-regulated alpha protein (GRO/KC). These effects of the peptide were associated with the modulation of the levels of formyl peptide receptor 2, a G-protein-coupled receptor that binds to Ac2-26, and the phosphorylated extracellular signal-regulated kinase (ERK) in the hippocampal neurons. CONCLUSIONS: The data suggest a neuroprotective effect of Ac2-26 in the epileptogenic processes through downregulation of inflammatory mediators and neuronal loss.


Subject(s)
Annexin A1/therapeutic use , Cytokines/metabolism , Nerve Degeneration/drug therapy , Neuroprotective Agents/therapeutic use , Peptides/therapeutic use , Status Epilepticus/complications , Status Epilepticus/drug therapy , Animals , Annexin A1/metabolism , Anticonvulsants/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Diazepam/therapeutic use , Disease Models, Animal , Gliosis/etiology , Hippocampus/drug effects , Hippocampus/pathology , MAP Kinase Signaling System/drug effects , Male , Muscarinic Agonists/toxicity , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Tissue Proteins/metabolism , Neuroglia/pathology , Neurons/drug effects , Pilocarpine/toxicity , Rats , Rats, Wistar , Receptors, Lipoxin/metabolism , Status Epilepticus/chemically induced
4.
J Cell Physiol ; 233(9): 6591-6603, 2018 09.
Article in English | MEDLINE | ID: mdl-29115663

ABSTRACT

Annexin A1 (AnxA1) is a glucocorticoid-regulated anti-inflammatory protein secreted by phagocytes and other specialised cells. In the endocrine system, AnxA1 controls secretion of steroid hormones and it is abundantly expressed in the testis, ovaries, placenta and seminal fluid, yet its potential modulation of fertility has not been described. Here, we observed that AnxA1 knockout (KO) mice delivered a higher number of pups, with a higher percentage of female offsprings. This profile was not dependent on the male features, as sperm from KO male mice did not present functional alterations, and had an equal proportion of Y and X chromosomes, comparable to wild type (WT) male mice. Furthermore, mismatched matings of male WT mice with female KO yielded a higher percentage of female pups per litter, a phenomenon which was not observed when male KO mice mated with female WT animals. Indeed, AnxA1 KO female mice displayed several differences in parameters related to gestation including (i) an arrested estrous cycle at proestrus phase; (ii) increased sites of implantation; (iii) reduced pre- and post-implantation losses; (iv) exacerbated features of the inflammatory reaction in the uterine fluid during implantation phase; and (v) enhanced plasma progesterone in the beginning of pregnancy. In summary, herein we highlight that AnxA1 pathway as a novel determinant of fundamental non-redundant regulatory functions during early pregnancy.


Subject(s)
Annexin A1/metabolism , Embryo Implantation/physiology , Animals , Estrous Cycle/metabolism , Estrous Cycle/physiology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Animal , Pregnancy , Proestrus/metabolism , Proestrus/physiology , Sex Ratio , Uterus/metabolism , Uterus/physiology , X Chromosome/metabolism , X Chromosome/physiology , Y Chromosome/metabolism , Y Chromosome/physiology
5.
Microb Pathog ; 118: 277-284, 2018 May.
Article in English | MEDLINE | ID: mdl-29605649

ABSTRACT

Mast cells (MCs) have important immunoregulatory roles in skin inflammation. Annexin A1 (ANXA1) is an endogenous anti-inflammatory protein that can be expressed by mast cells, neutrophils, eosinophils, monocytes, epithelial and T cells. This study investigated MCs heterogeneity and ANXA1 expression in human dermatoses with special emphasis in leprosy. Sixty one skin biopsies from 2 groups were investigated: 40 newly diagnosed untreated leprosy patients (18 reaction-free, 11 type 1 reaction/T1R, 11 type 2 reaction/T2R); 21 patients with other dermatoses. Tryptase/try+ and chymase/chy + phenotypic markers and toluidine blue stained intact/degranulated MC counts/mm2 were evaluated. Try+/chy+ MCs and ANXA1 were identified by streptavidin-biotin-peroxidase immunostaining and density was reported. In leprosy, degranulated MCs outnumbered intact ones regardless of the leprosy form (from tuberculoid/TT to lepromatous/LL), leprosy reactions (reactional/reaction-free) and type of reaction (T1R/T2R). Compared to other dermatoses, leprosy skin lesions showed lower numbers of degranulated and intact MCs. Try+ MCs outnumbered chy+ in leprosy lesions (reaction-free/reactional, particularly in T2R), but not in other dermatoses. Compared to other dermatoses, ANXA1 expression, which is also expressed in mast cells, was higher in the epidermis of leprosy skin lesions, independently of reactional episode. In leprosy, higher MC degranulation and differential expression of try+/chy+ subsets independent of leprosy type and reaction suggest that the Mycobacterium leprae infection itself dictates the inflammatory MCs activation in skin lesions. Higher expression of ANXA1 in leprosy suggests its potential anti-inflammatory role to maintain homeostasis preventing tissue and nerve damage.


Subject(s)
Annexin A1/biosynthesis , Annexin A1/immunology , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Leprosy/immunology , Leprosy/metabolism , Mast Cells/metabolism , Adult , Aged , Aged, 80 and over , Biopsy , Brazil , Chymases/metabolism , Epidermis/immunology , Epidermis/pathology , Female , Humans , Leprosy/pathology , Leprosy, Lepromatous/metabolism , Leprosy, Tuberculoid/metabolism , Male , Mast Cells/pathology , Middle Aged , Mycobacterium leprae/immunology , Mycobacterium leprae/pathogenicity , Skin/pathology , Skin Diseases/metabolism , Skin Diseases/pathology , Tryptases/metabolism , Young Adult
6.
FASEB J ; 29(5): 2161-71, 2015 May.
Article in English | MEDLINE | ID: mdl-25690650

ABSTRACT

Ischemia/reperfusion (I/R) injury following stroke can worsen patient outcome through excess inflammation. This study investigated the pharmacologic potential of targeting an endogenous anti-inflammatory circuit via formyl peptide receptor (FPR) 2/lipoxin receptor (ALX) (Fpr2/3 in mouse) in global cerebral I/R. Mice (C57BL/6 and Fpr2/3(-/-)) were subjected to bilateral common carotid artery occlusion, followed by reperfusion and treatment with FPR agonists: AnxA1Ac2-26 [Annexin A1 mimetic peptide (Ac-AMVSEFLKQAWFIENEEQEYVQTVK), 2.5 µg/kg] and 15-epimer-lipoxin A4 (15-epi-LXA4; FPR2/ALX specific, 12.5 and 100 ng/kg). Leukocyte-endothelial (L-E) interactions in the cerebral microvasculature were then quantified in vivo using intravital fluorescence microscopy. 15-epi-LXA4 administration at the start of reperfusion reduced L-E interactions after 40 min (which was sustained at 2 h with high-dose 15-epi-LXA4) to levels seen in sham-operated animals. AnxA1Ac2-26 treatment decreased leukocyte adhesion at 40 min and all L-E interactions at 2 h (up to 95%). Combined treatment with AnxA1Ac2-26 plus FPR antagonists t-Boc-FLFLF (250 ng/kg) or WRW4 (FPR2/ALX selective, 1.4 µg/kg) abrogated the effects of AnxA1Ac2-26 fully at 40 min. Antagonists were less effective at 2 h, which we demonstrate is likely because of their impact on early L-E interactions. Our findings indicate that FPR2/ALX activity elicits considerable control over vascular inflammatory responses during cerebral I/R and, therefore, provide evidence that targeting FPR2/ALX may be beneficial for patients who suffered from stroke.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Disease Models, Animal , Endothelium, Vascular/metabolism , Leukocytes/metabolism , Peptide Fragments/pharmacology , Receptors, Formyl Peptide/physiology , Stroke/physiopathology , Animals , Annexin A1/metabolism , Blotting, Western , Carotid Artery Diseases/pathology , Carotid Artery Diseases/prevention & control , Cell Adhesion , Cells, Cultured , Endothelium, Vascular/cytology , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Inflammation/pathology , Inflammation/prevention & control , Leukocytes/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Microvessels/metabolism , Microvessels/pathology , Neutrophils/cytology , Neutrophils/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control
7.
Hemoglobin ; 40(5): 356-358, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27535451

ABSTRACT

Phenotypic heterogeneity for sickle cell disease is associated to several genetic factors such as genotype for sickle cell disease, ß-globin gene cluster haplotypes and Hb F levels. The coinheritance of Hb S (HBB: c.20A > T) and Hb D-Punjab (HBB: c.364G > C) results in a double heterozygosity, which constitutes one of the genotypic causes of sickle cell disease. This study aimed to assess the phenotypic diversity of sickle cell disease presented by carriers of the Hb S/Hb D-Punjab genotype and the Bantu [- + - - - -] haplotype. We evaluated medical records from 12 patients with sickle cell disease whose Hb S/Hb D-Punjab genotype and Bantu haplotype were confirmed by molecular analysis. Hb S and Hb D-Punjab levels were quantified by chromatographic analysis. Mean concentrations of Hb S and Hb D-Punjab were 44.8 ± 2.3% and 43.3 ± 1.8%, respectively. Painful crises were present in eight (66.7%) patients evaluated, representing the most common clinical event. Acute chest syndrome (ACS) was the second most prevalent manifestation, occurring in two individuals (16.7%). Three patients were asymptomatic, while another two exhibited greater diversity of severe clinical manifestations. Medical records here analyzed reported a significant clinical diversity in sickle cell disease ranging from the absence of symptoms to wide phenotypic variety. The sickle cell disease genotype, Bantu haplotype and hemoglobin (Hb) levels did not influence the clinical diversity. Thus, we concluded that the phenotypic variation in sickle cell disease was present within a specific genotype for disease regardless of the ß-globin gene cluster haplotypes.


Subject(s)
Anemia, Sickle Cell/genetics , Hemoglobin, Sickle/analysis , Hemoglobins, Abnormal/analysis , Phenotype , Acute Chest Syndrome/etiology , Anemia, Sickle Cell/complications , Anemia, Sideroblastic , Genotype , Haplotypes , Heterozygote , Humans , Pain/etiology , beta-Globins/genetics
8.
Exp Eye Res ; 134: 24-32, 2015 May.
Article in English | MEDLINE | ID: mdl-25795053

ABSTRACT

Annexin A1 (ANXA1), a 37 kDa glucocorticoid-regulated protein, is a potent anti-inflammatory mediator effective in terminating acute inflammatory response, and its role in allergic settings has been poorly studied. The aim of this investigation was to evaluate the mechanism of action of ANXA1 in intraocular inflammation using a classical model of ovalbumin (OVA)-induced allergic conjunctivitis (AC). OVA-immunised Balb/c mice, wild-type (WT) and ANXA1-deficient (AnxA1(-/-)), were challenged with eye drops containing OVA on days 14-16 with a subset of WT animals pretreated intraperitoneally with the peptide Ac2-26 (N-terminal region of ANXA1) or dexamethasone (DEX). After 24 h of the last ocular challenge, WT mice treated with Ac2-26 and DEX had significantly reduced clinical signs of conjunctivitis (chemosis, conjunctival hyperaemia, lid oedema and tearing), plasma IgE levels, leukocyte (eosinophil and neutrophil) influx and mast cell degranulation in the conjunctiva compared to WT controls. These anti-inflammatory effects of DEX were associated with high endogenous levels of ANXA1 in the ocular tissues as detected by immunohistochemistry. Additionally, Ac2-26 administration was effective to reduce IL-2, IL-4, IL-10, IL-13, eotaxin and RANTES in the eye and lymph nodes compared to untreated WT animals. The lack of ANXA1 produced an exacerbated allergic response as detected by the density of the inflammatory cell influx to the conjunctiva and the cytokine/chemokine release. These different effects observed for Ac2-26 were correlated with diminished level of activated ERK at 24 h in the ocular tissues compared to untreated OVA group. Our findings demonstrate the protective effect of ANXA1 during the inflammatory allergic response suggesting this protein as a potential target for new ocular inflammation therapies.


Subject(s)
Annexin A1/therapeutic use , Conjunctivitis, Allergic/drug therapy , Disease Models, Animal , Peptides/therapeutic use , Animals , Blotting, Western , Conjunctivitis, Allergic/metabolism , Conjunctivitis, Allergic/pathology , Cytokines/metabolism , Dexamethasone/therapeutic use , Eosinophils/physiology , Glucocorticoids/therapeutic use , Immunoenzyme Techniques , Immunoglobulin E/blood , Lymph Nodes/metabolism , Male , Mast Cells/physiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/toxicity
9.
J Immunol ; 190(11): 5689-701, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23645879

ABSTRACT

Annexin A1 (AnxA1) is a protein that displays potent anti-inflammatory properties, but its expression in eye tissue and its role in ocular inflammatory diseases have not been well studied. We investigated the mechanism of action and potential uses of AnxA1 and its mimetic peptide (Ac2-26) in the endotoxin-induced uveitis (EIU) rodent model and in human ARPE-19 cells activated by LPS. In rats, analysis of untreated EIU after 24 and 48 h or EIU treated with topical applications or with a single s.c. injection of Ac2-26 revealed the anti-inflammatory actions of Ac2-26 on leukocyte infiltration and on the release of inflammatory mediators; the systemic administration of Boc2, a formylated peptide receptor (fpr) antagonist, abrogated the peptide's protective effects. Moreover, AnxA1(-/-) mice exhibited exacerbated EIU compared with wild-type animals. Immunohistochemical studies of ocular tissue showed a specific AnxA1 posttranslational modification in EIU and indicated that the fpr2 receptor mediated the anti-inflammatory actions of AnxA1. In vitro studies confirmed the roles of AnxA1 and fpr2 and the protective effects of Ac2-26 on the release of chemical mediators in ARPE-19 cells. Molecular analysis of NF-κB translocation and IL-6, IL-8, and cyclooxygenase-2 gene expression indicated that the protective effects of AnxA1 occur independently of the NF-κB signaling pathway and possibly in a posttranscriptional manner. Together, our data highlight the role of AnxA1 in ocular inflammation, especially uveitis, and suggest the use of AnxA1 or its mimetic peptide Ac2-26 as a therapeutic approach.


Subject(s)
Annexin A1/genetics , Anti-Inflammatory Agents/pharmacology , Peptides/pharmacology , Uveitis/genetics , Animals , Annexin A1/administration & dosage , Annexin A1/chemistry , Annexin A1/metabolism , Annexin A1/pharmacology , Anti-Inflammatory Agents/administration & dosage , Aqueous Humor/cytology , Aqueous Humor/immunology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Endotoxins/adverse effects , Gene Expression Regulation/drug effects , Lipopolysaccharides/immunology , Male , Mice , Mice, Knockout , Models, Biological , NF-kappa B/metabolism , Neutrophil Infiltration/immunology , Neutrophils/drug effects , Neutrophils/immunology , Oligopeptides/pharmacology , Peptides/administration & dosage , Phosphorylation , Protein Transport/drug effects , Rats , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Uveitis/chemically induced , Uveitis/immunology
10.
Heliyon ; 10(12): e32808, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38975186

ABSTRACT

For decades, animal models have been the standard approach in drug research and development, as they are required by regulations in the transition from preclinical to clinical trials. However, there is growing ethical and scientific concern regarding these trials, as 80 % of the therapeutic potential observed in pre-clinical studies are often unable to be replicated, despite demonstrating efficacy and safety. In response to this, Tissue Engineering has emerged as a promising alternative that enables the treatment of various diseases through the production of biological models for advanced biological assays or through the direct development of tissue repairs or replacements. One of the promising applications of Tissue Engineering is the development of three-dimensional (3D) models for in vitro tests, replacing the need for in vivo animal models. In this study, 3D skin equivalents (TSE) were produced and used as an in vitro model to test photobiostimulation using curcumin-loaded nanocapsules. Photodynamic biostimulation therapy uses photodynamic processes to generate small amounts of reactive oxygen species (ROS), which can activate important biological effects such as cell differentiation, modulation of inflammatory processes and contribution to cell regeneration. The PLGA nanocapsules (NC) used in the study were synthesized through a preformed polymer deposition method, exhibiting particle size <200 nm, Zeta potential >|30| and polydispersity index between 0.5 and 0.3. Atomic force microscopy analyzes confirmed that the particle size was <200 nm, with a spherical morphology and a predominantly smooth and uniform surface. The NC biocompatibility assay did not demonstrate cytotoxicity for the concentrations tested (2.5-25 µg mL-1).The in vitro release assay showed a slow and sustained release characteristic of the nanocapsules, and cellular uptake assays indicated a significant increase in cellular internalization of the curcumin-loaded nanostructure. Monolayer photobiostimulation studies revealed an increase in cell viability of the HDFn cell line (viability 134 %-228 %) for all LED fluences employed at λ = 450 nm (150, 300, and 450 mJ cm-2). Additionally, the scratch assays, monitoring in vitro scar injury, demonstrated more effective effects on cell proliferation with the fluence of 300 mJ cm-2. Staining of TSE with hematoxylin and eosin showed the presence of cells with different morphologies, confirming the presence of fibroblasts and keratinocytes. Immunohistochemistry using KI-67 revealed the presence of proliferating cells in TSE after irradiation with LED λ = 450 nm (150, 300, and 450 mJ cm-2).

11.
Biomed Pharmacother ; 165: 115230, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37531784

ABSTRACT

Diabetes mellitus (DM) is characterized by metabolic alterations that involve defects in the secretion and/or action of insulin, being responsible for several complications, such as impaired healing. Studies from our research group have shown that annexin A1 protein (AnxA1) is involved in the regulation of inflammation and cell proliferation. In light of these findings, we have developed a new technology and evaluated its effect on a wound healing in vivo model using type 1 diabetes (T1DM)-induced mice. We formulated a hydrogel containing AnxA12-26 using defined parameters such as organoleptic characteristics, pH, UV-vis spectroscopy and cytotoxicity assay. UV-vis spectroscopy confirmed the presence of the associated AnxA12-26 peptide in the three-dimensional hydrogel matrix, while the in vitro cytotoxicity assay showed excellent biocompatibility. Mice showed increased blood glucose levels, confirming the efficacy of streptozotocin (STZ) to induce T1DM. Treatment with AnxA12-26 hydrogel showed to improve diabetic wound healing, defined as complete re-epithelialization and tissue remodeling, with reduction of inflammatory infiltrate in diabetic animals. We envisage that the AnxA12-26 hydrogel, with its innovative composition and formulation be efficient on improving diabetic healing and contributing on the expansion of the therapeutic arsenal to treat diabetic wounds, at a viable cost.


Subject(s)
Annexin A1 , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Skin Diseases , Mice , Animals , Diabetes Mellitus, Type 1/drug therapy , Hydrogels/pharmacology , Hydrogels/chemistry , Annexin A1/pharmacology , Annexin A1/metabolism , Diabetes Mellitus, Experimental/metabolism , Wound Healing
12.
Ann Rheum Dis ; 71(11): 1872-80, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22562975

ABSTRACT

OBJECTIVES: To establish the role and effect of glucocorticoids and the endogenous annexin A1 (AnxA1) pathway in inflammatory arthritis. METHODS: Ankle joint mRNA and protein expression of AnxA1 and its receptors were analysed in naive and arthritic mice by real-time PCR and immunohistochemistry. Inflammatory arthritis was induced with the K/BxN arthritogenic serum in AnxA1(+/+) and AnxA1(-/-) mice; in some experiments, animals were treated with dexamethasone (Dex) or with human recombinant AnxA1 or a protease-resistant mutant (termed SuperAnxA1). Readouts were arthritic score, disease incidence, paw oedema and histopathology, together with pro-inflammatory gene expression. RESULTS: All elements of the AnxA1 pathway could be detected in naive joints, with augmentation during ongoing disease, due to the infiltration of immune cells. No difference in arthritis intensity of profile could be observed between AnxA1(+/+) and AnxA1(-/-) mice. Treatment of mice with Dex (10 µg intraperitoneally daily from day 2) afforded potent antiarthritic effects highly attenuated in the knockouts: macroscopic changes were mirrored by histopathological findings and pro-inflammatory gene (eg, Nos2) expression. Presence of proteinase 3 mRNA in the arthritic joints led the authors to test AnxA1 and the mutant SuperAnxA1 (1 µg intraperitoneally daily in both cases from day 2), with the latter one being able to accelerate the resolving phase of the disease. CONCLUSION: AnxA1 is an endogenous determinant for the therapeutic efficacy of Dex in inflammatory arthritis. Such an effect can be partially mimicked by application of SuperAnxA1 which may represent the starting point for novel antiarthritic therapeutic strategies.


Subject(s)
Annexin A1/physiology , Antirheumatic Agents/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Dexamethasone/therapeutic use , Glucocorticoids/therapeutic use , Animals , Annexin A1/chemistry , Annexin A1/pharmacology , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Disease Models, Animal , Drug Therapy, Combination , Edema/drug therapy , Edema/pathology , Gene Expression/drug effects , Mice , Mice, Knockout , Mutant Proteins/chemistry , Mutant Proteins/pharmacology , Recombinant Proteins/pharmacology
13.
Mol Vis ; 18: 1583-93, 2012.
Article in English | MEDLINE | ID: mdl-22740770

ABSTRACT

PURPOSE: The aim of this study was to evaluate the expression of the protein annexin A1 (ANXA1), a potent endogenous regulator of the inflammatory process, in ocular toxoplasmosis. METHODS: C57BL/6 female mice were infected using intravitreal injections of either 10(6) tachyzoites of Toxoplasma gondii (RH strain; T. gondii) or PBS only (control groups). After 24, 48, and 72 h, animals were sacrificed and their eyes were harvested for histopathological, immunohistochemical, and ultrastructural immunocytochemical analysis of ANXA1. Human retinal pigment epithelial (RPE) cells (ARPE-19) were infected in vitro with T. gondii and collected after 60, 120, 240 min, and 24 h. RESULTS: Compared with non-infected eyes, an intense inflammatory response was observed in the anterior (24 h after infection) and posterior segments (72 h after infection) of the infected eye, characterized by neutrophil infiltration and by the presence of tachyzoites and their consequent destruction along with disorganization of normal retina architecture and RPE vacuolization. T. gondii infection was associated with a significant increase of ANXA1 expression in the neutrophils at 24, 48, and 72 h, and in the RPE at 48 and 72 h. In vitro studies confirmed an upregulation of ANXA1 levels in RPE cells, after 60 and 120 min of infection with T. gondii. CONCLUSIONS: The positive modulation of endogenous ANXA1 in the inflammatory and RPE cells during T. gondii infection suggests that this protein may serve as a therapeutic target in ocular toxoplasmosis.


Subject(s)
Annexin A1/genetics , Anterior Eye Segment/immunology , Epithelial Cells/immunology , Posterior Eye Segment/immunology , Retinal Pigment Epithelium/immunology , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Ocular/veterinary , Animals , Annexin A1/metabolism , Anterior Eye Segment/parasitology , Anterior Eye Segment/pathology , Epithelial Cells/parasitology , Epithelial Cells/pathology , Female , Gene Expression Regulation/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Intravitreal Injections , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/immunology , Posterior Eye Segment/parasitology , Posterior Eye Segment/pathology , Retinal Pigment Epithelium/parasitology , Retinal Pigment Epithelium/pathology , Toxoplasma/immunology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/pathology , Toxoplasmosis, Ocular/immunology , Toxoplasmosis, Ocular/parasitology , Toxoplasmosis, Ocular/pathology
14.
Drug Discov Today ; 27(11): 103367, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36165812

ABSTRACT

Annexins (AnxAs) are Ca2+/phospholipid-binding proteins extensively studied and generally involved in several diseases. Although evidence exists regarding the distribuition of AnxAs in the visual system, their exact roles and the exact cell types of the eye where these proteins are expressed are not well-understood. AnxAs have pro-resolving roles in infectious, autoimmune, degenerative, fibrotic and angiogenic conditions, making them an important target in ocular tissue homeostasis. This review summarizes the current knowledge on the distribution and function of AnxA1-8 isoforms under normal and pathological conditions in the visual system, as well as perspectives for ophthalmologic treatments, including the potential use of the AnxA1 recombinant and/or its mimetic peptide Ac2-26.

15.
Toxicol Lett ; 363: 27-35, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35561849

ABSTRACT

Cisplatin is an antineoplastic agent widely used, and no effective treatments capable of preventing cisplatin-induced ototoxicity and neurotoxicity in humans have yet been identified. This study evaluated the effect of the anti-inflammatory annexin A1 (AnxA1)-derived peptide Ac2-26 in a cisplatin-induced ototoxicity model. Wistar rats received intraperitoneal injections of cisplatin (10 mg/kg/day) for 3 days to induce hearing loss, and Ac2-26 (1 mg/kg) was administered 15 min before cisplatin administration. Control animals received an equal volume of saline. Hearing thresholds were measured by distortion product otoacoustic emissions (DPOAE) before and after treatments. Pharmacological treatment with Ac2-26 protected against cisplatin-induced hearing loss, as evidenced by DPOAE results showing similar signal-noise ratios between the control and Ac2-26-treated groups. These otoprotective effects of Ac2-26 were associated with an increased number of ganglion neurons compared with the untreated cisplatin group. Additionally, Ac2-26 treatment produced reduced immunoreactivity on cleaved caspase 3 and phosphorylated ERK levels in the ganglion neurons, compared to the untreated group, supporting the neuroprotective effects of the Ac2-26. Our results suggest that Ac2-26 has a substantial otoprotective effect in this cisplatin-induced ototoxicity model mediated by neuroprotection and the regulation of the ERK pathway.


Subject(s)
Annexin A1 , Antineoplastic Agents , Hearing Loss , Ototoxicity , Animals , Annexin A1/pharmacology , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Hearing Loss/chemically induced , Hearing Loss/prevention & control , Otoacoustic Emissions, Spontaneous , Ototoxicity/prevention & control , Peptides/pharmacology , Rats , Rats, Wistar
16.
Inflammation ; 45(3): 1133-1145, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35031944

ABSTRACT

The pathogenesis of atopic dermatitis (AD) and psoriasis (Ps) overlaps, particularly the activation of the immune response and tissue damage. Here, we evaluated galectin (Gal)-1 and Gal-3 levels, which are beta-galactoside-binding proteins with immunomodulatory functions and examined their effects on human keratinocytes stimulated with either interleukin (IL)-4 or IL-17A. Skin biopsies from AD, Ps, and control patients were evaluated using histological and immunohistochemical analyses. Six studies containing publicly available transcriptome data were individually analyzed using the GEO2R tool to detect Gal-1 and Gal-3 mRNA levels. In vitro, IL-4- or IL-17A-stimulated keratinocytes were treated with or without Gal-1 or Gal-3 to evaluate cytokine release and migration. Our findings showed different patterns of expression for Gal-1 and Gal-3 in AD and Ps skins. Densitometric analysis in skin samples showed a marked increase in the protein Gal-1 levels in Ps epidermis and in both AD and Ps dermis compared to controls. Protein and mRNA Gal-3 levels were downregulated in AD and Ps lesional skin compared with the control samples. In vitro, both galectins addition abrogated the release of IL-8 and RANTES in IL-17-stimulated keratinocytes after 24 h, whereas IL-6 release was downregulated by Gal-3 and Gal-1 in IL-4- and IL-17-stimulated cells, respectively. Administration of both galectins also increased the rate of keratinocyte migration under IL-4 or IL-17 stimulation conditions compared with untreated cells. Altogether, the immunoregulatory and migration effects of Gal-1 and Gal-3 on keratinocytes under inflammatory microenvironment make them interesting targets for future therapies in cutaneous diseases.


Subject(s)
Dermatitis, Atopic , Psoriasis , Blood Proteins , Cells, Cultured , Galectin 1/metabolism , Galectin 1/pharmacology , Galectin 3/metabolism , Galectin 3/pharmacology , Galectins , Humans , Immunity , Interleukin-17/metabolism , Interleukin-4/metabolism , Interleukin-4/pharmacology , Keratinocytes/metabolism , Psoriasis/metabolism , RNA, Messenger/metabolism
17.
Life Sci ; 304: 120677, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35654117

ABSTRACT

AIMS: In this study we evaluated the effect of pharmacological treatment with AnxA1-derived peptide Ac2-26 in an experimental model of toxicity induced by cisplatin. MAIN METHODS: Male rats were divided into Sham (control), Cisplatin (received intraperitoneal injections of 10 mg/kg/day of cisplatin for 3 days) and Ac2-26 (received intraperitoneal injections of 1 mg/kg/day of peptide, 15 min before cisplatin) groups. KEY FINDINGS: After 6 h of the last dose of cisplatin, an acute inflammatory response was observed characterized by a marked increase in the number of neutrophils and GM-CSF, IL-ß, IL-6, IL-10 and TNF-α plasma levels. These findings were associated with increased AnxA1 protein levels in liver and kidneys, as well as positive AnxA1/Fpr2 circulating leukocytes. Treatment with Ac2-26 produced higher levels of GM-CSF, corroborating the high numbers of neutrophils, and the anti-inflammatory cytokine IL-4. Ac2-26 preserved the morphology of liver structures and increased Fpr1 expression, preventing the damage caused by cisplatin. In the kidneys, Ac2-26 caused downregulation of renal Fpr1 and Fpr2 levels and abrogated the increased levels of the CLU and KIM-1 biomarkers of kidney damage induced by cisplatin. However, no effect of peptide treatment was detected in cisplatin-induced kidney morphology injury. SIGNIFICANCE: Despite activation of the anti-inflammatory AnxA1/Fpr axis during cisplatin administration, treatment with Ac2-26 did not efficiently prevent its deleterious effects on the liver and kidneys.


Subject(s)
Annexin A1 , Animals , Annexin A1/chemistry , Annexin A1/metabolism , Annexin A1/pharmacology , Anti-Inflammatory Agents/pharmacology , Cisplatin/metabolism , Cisplatin/toxicity , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Kidney/metabolism , Liver/metabolism , Male , Peptides/chemistry , Rats
18.
BMC Immunol ; 12: 59, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-22011168

ABSTRACT

BACKGROUND: The balancing functions of pro/anti-inflammatory mediators of the complex innate responses have been investigated in a variety of experimental inflammatory settings. Annexin-A1 (AnxA1) is one mediator of endogenous anti-inflammation, affording regulation of leukocyte trafficking and activation in many contexts, yet its role in lung pathologies has been scarcely investigated, despite being highly expressed in lung cells. Here we have applied the bleomycin lung fibrosis model to AnxA1 null mice over a 21-day time-course, to monitor potential impact of this mediator on the control of the inflammatory and fibrotic phases. RESULTS: Analyses in wild-type mice revealed strict spatial and temporal regulation of the Anxa1 gene, e.g. up-regulation in epithelial cells and infiltrated granulocytes at day 7, followed by augmented protein levels in alveolar macrophages by day 21. Absence of AnxA1 caused increases in: i) the degree of inflammation at day 7; and ii) indexes of fibrosis (assessed by deposition of hydroxyproline in the lung) at day 7 and 21. These alterations in AnxA1 null mice were paralleled by augmented TGF-ß1, IFN-γ and TNF-α generation compared to wild-type mice. Finally, treatment of wild type animals with an AnxA1 peptido-mimetic, given prophylactically (from day 0 to 21) or therapeutically (from day 14 onward), ameliorated both signs of inflammation and fibrosis. CONCLUSION: Collectively these data reveal a pathophysiological relevance for endogenous AnxA1 in lung inflammation and, more importantly, fibrosis, and may open new insights for the pharmacological treatment of lung fibrosis.


Subject(s)
Epithelial Cells/metabolism , Lung/metabolism , Macrophages, Alveolar/metabolism , Pneumonia/metabolism , Pulmonary Fibrosis/metabolism , Animals , Annexin A1/genetics , Annexin A1/metabolism , Biomimetic Materials/administration & dosage , Biomimetic Materials/metabolism , Bleomycin/administration & dosage , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation/genetics , Hydroxyproline/metabolism , Inflammation Mediators/metabolism , Lung/drug effects , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Mice , Mice, Knockout , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Pneumonia/chemically induced , Pneumonia/drug therapy , Pneumonia/genetics , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/immunology , Time Factors
19.
Mol Vis ; 17: 1310-9, 2011.
Article in English | MEDLINE | ID: mdl-21633711

ABSTRACT

PURPOSE: To investigate the role of mast cells and annexin-A1 (Anxa1) in endotoxin-induced uveitis (EIU). METHODS: EIU was induced by injection of lipopolysaccharide (LPS) into the paws of rats, which were then sacrificed after 24 and 48 h. To assess EIU in the absence of mast cells, groups of animals were pretreated with compound 48/80 (c48/80) and sacrificed after 24 h after no treatment or EIU induction. The eyes were used for histological studies and the aqueous humor (AqH) pool was used for the analysis of transmigrated cells and Anxa1 levels. In inflammatory cells, Anxa1 expression was monitored by immunohistochemistry. RESULTS: After 24 h, rats with EIU exhibited degranulated mast cells, associated with elevated numbers of infiltrating leukocytes and the high expression of Anxa1 in the AqH and the neutrophils. After 48 h of EIU, the mast cells were intact, indicating granule re-synthesis, and there was a reduction of neutrophil transmigration and an increase in the number of mononuclear phagocytic cells in ocular tissues. Anxa1 expression was decreased in neutrophils but increased in mononuclear phagocytic cells. In the animals pretreated with c48/80 and subjected to EIU, mast cells responded to this secretagogue by degranulating and few transmigrated neutrophils were observed. CONCLUSTIONS: We report that mast cells are a potential source of pharmacological mediators that are strongly linked to the pathophysiology of EIU, and the endogenous protein Anxa1 is a mediator in the homeostasis of the inflammatory process with anti-migratory effects on leukocytes, which supports further studies of this protein as an innovative therapy for uveitis.


Subject(s)
Lipopolysaccharides , Mast Cells , Uveitis/chemically induced , Uveitis/physiopathology , Animals , Annexin A1/metabolism , Aqueous Humor/cytology , Aqueous Humor/metabolism , Blotting, Western , Cell Count , Cell Degranulation , Cell Movement/drug effects , Eye/pathology , Eye Proteins/metabolism , Homeostasis , Immunohistochemistry , Leukocytes/pathology , Male , Neutrophils/drug effects , Neutrophils/metabolism , Rats , Rats, Wistar , Uveitis/pathology , p-Methoxy-N-methylphenethylamine/pharmacology
20.
Dig Dis Sci ; 56(6): 1637-44, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21188523

ABSTRACT

BACKGROUND: This study evaluated the galectin-1 and -3 expression during N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)-induced gastric carcinogenesis in denervated rat stomachs using benzalkonium chloride. METHOD: Four experimental situations were evaluated: nondenervated and denervated stomachs without lesions and nondenervated and denervated stomachs with lesions. Sections of the pyloric region were stained with toluidine blue and incubated with mouse monoclonal anti-Gal-1 and rabbit polyclonal anti-Gal-3 for histopathological and immunohistochemical analysis, respectively. RESULT: MNNG caused the development of benign and malignant epithelial lesions, which were more pronounced in nondenervated stomachs with lesions and accompanied by inflammatory cell-enriched stroma. By immunostaining, the epithelial cells, blood vessels, muscle layer, and myenteric plexus were Gal-1 and -3 positive. Gal-3 was also detected in the gastric crypts, mucus secretion, and fibroblasts of pyloric fragments. Development of lesions in denervated stomachs was associated with a significant decrease in Gal-1 and -3 expression in epithelial cells, mast cells, and neutrophil cytoplasm, compared with that of nondenervated stomach lesions (P < 0.01; P < 0.001; P < 0.001, respectively). CONCLUSION: These results demonstrate that myenteric denervation downregulates endogenous Gal-1 and -3 expression, which might inhibit tumor development in this experimental model.


Subject(s)
Galectin 1/metabolism , Galectin 3/metabolism , Stomach Neoplasms/chemically induced , Stomach Neoplasms/metabolism , Adenocarcinoma/metabolism , Animals , Denervation , Down-Regulation , Enteric Nervous System , Galectin 1/genetics , Galectin 3/genetics , Gene Expression Regulation, Neoplastic , Male , Methylnitronitrosoguanidine/toxicity , Neoplasms, Experimental , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL