Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(14)2022 Jul 11.
Article in English | MEDLINE | ID: mdl-35886983

ABSTRACT

Melanoma is a highly metastatic and rapidly progressing cancer, a leading cause of mortality among skin cancers. The melanoma microenvironment, formed from the activity of malignant cells on the extracellular matrix and the recruitment of immune cells, plays an active role in the development of drug resistance and tumor recurrence, which are clinical challenges in cancer treatment. These tumoral metabolic processes are affected by proteins, including Galectin-3 (Gal-3), which is extensively involved in cancer development. Previously, we characterized a partially methylated mannogalactan (MG-Pe) with antimelanoma activities. In vivo models of melanoma were used to observe MG-Pe effects in survival, spontaneous, and experimental metastases and in tissue oxidative stress. Analytical assays for the molecular interaction of MG-Pe and Gal-3 were performed using a quartz crystal microbalance, atomic force microscopy, and contact angle tensiometer. MG-Pe exhibits an additive effect when administered together with the chemotherapeutic agent dacarbazine, leading to increased survival of treated mice, metastases reduction, and the modulation of oxidative stress. MG-Pe binds to galectin-3. Furthermore, MG-Pe antitumor effects were substantially reduced in Gal-3/KO mice. Our results showed that the novel Gal-3 ligand, MG-Pe, has both antitumor and antimetastatic effects, alone or in combination with chemotherapy.


Subject(s)
Antineoplastic Agents , Galectin 3 , Melanoma , Skin Neoplasms , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Dacarbazine/metabolism , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Galectin 3/metabolism , Galectin 3/pharmacology , Galectin 3/therapeutic use , Ligands , Melanoma/drug therapy , Melanoma/metabolism , Mice , Neoplasm Recurrence, Local , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
2.
Int J Biol Macromol ; 185: 551-561, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34216657

ABSTRACT

Advanced melanoma patients that are not included in common genetic classificatory groups lack effective and safe therapeutic options. Chemotherapy and immunotherapy show unsatisfactory results and devastating adverse effects for these called triple wild-type patients. New approaches exploring the intrinsic antitumor properties of gold nanoparticles might reverse this scenario as a safer and more effective alternative. Therefore, we investigated the efficacy and safety of a composite made of gum arabic-functionalized gold nanorods (GA-AuNRs) against triple wild-type melanoma. The natural polymer gum arabic successfully stabilized the nanorods in the biological environment and was essential to improve their biocompatibility. In vivo results obtained from treating triple wild-type melanoma-bearing mice showed that GA-AuNRs remarkably reduced primary tumor growth by 45%. Furthermore, GA-AuNRs induced tumor histological features associated with better prognosis while also reducing superficial lung metastasis depth and the incidence of intrapulmonary metastasis. GA-AuNRs' efficacy comes from their capacity to reduce melanoma cells ability to invade the extracellular matrix and grow into colonies, in addition to a likely immunomodulatory effect induced by gum arabic. Additionally, a broad safety investigation found no evidence of adverse effects after GA-AuNRs treatment. Therefore, this study unprecedentedly reports GA-AuNRs as a potential nanomedicine for advanced triple wild-type melanomas.


Subject(s)
Gold/administration & dosage , Gum Arabic/chemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Melanoma/drug therapy , Animals , BALB 3T3 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Extracellular Matrix/metabolism , Gold/chemistry , Gold/pharmacology , Humans , Lung Neoplasms/metabolism , Melanoma/metabolism , Metal Nanoparticles , Mice , Treatment Outcome , Xenograft Model Antitumor Assays
3.
Microbiol Res ; 248: 126768, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33873141

ABSTRACT

The increase in the number of deaths from infections caused by multidrug-resistant bacteria and cancer diseases highlights the need for new molecules with biological activity. Actinobacteria represent a potential source of new compounds, as these microorganisms have already produced a great diversity of clinically employed antibiotics. Endophytes from unexplored biomes, such as the Pantanal (the largest wetland in the world), can be a source of new molecules. Hymenachne amplexicaulis is among the unexplored native plants of the Pantanal in terms of its endophytic community. This plant is considered a weed in other countries due to its ability to adapt and compete with native plants, and there is evidence to suggest that the endophytic community of H. amplexicaulis plays an important role in this competitiveness. To explore its therapeutic potential, the present study isolated, identified (using partial sequence of the 16S rDNA) and bioprospected H. amplexicaulis endophytic actinobacteria. Ten isolates belonging to the genera Streptomyces, Microbispora, Leifsonia, and Verrucosispora were obtained from root fragments. The susceptibility profile of the isolates to the different classes of antibiotics was evaluated, with 80 % of the isolates showing resistance to the antibiotics Nalidixic Acid, Ampicillin, Chloramphenicol, Oxacillin, and Rifampicin. To assess antibacterial and antitumor activities, methanolic extracts were obtained by fermentation in SG culture medium at 36 °C at 180 rpm for 10 days. The extract produced from the S. albidoflavus CMRP4854 isolate was the only one to show activity against the Gram-negative bacterium Acinetobacter baumanii. Due to the great clinical importance of this pathogen and the difficulty in obtaining active compounds against it, the CMRP4854 isolate should be further investigated for the identification of active compounds and mode of action. We also emphasize the results obtained by the extract of the isolates Streptomyces albidoflavus CMRP4852 and Verrucosispora sp. CMRP4860 that presented antibacterial effect against Methicilin-resistant Staphylococcus aureus (MRSA) (MIC: 1.5 µg/mL and 13 µg/mL, respectively) and Vancomycin-resistant Enterococcus (VRE) (MIC: 40 µg/mL for both extracts). Extracts (200 µg/mL) of these two endophytes also showed selective cytotoxicity action against murine B16-F10 melanoma cells. However, the CMRP4852 extract also affected the density of normal cells. Due to these results, the crude extract of isolate CMRP4860 Verrucosispora sp., which was the only one that presented cytotoxicity and reduced cell density only in tumor cells, was selected for subsequent analysis involving scale-up fermentation of the CMRP4860 resulting in 9 fractions that were tested against both bacteria and tumor cells, with particular fractions showing promise and meriting further investigation. Taken together, the results of this study not only show for the first time that the endophytic community of H. amplexicaulis actinobacteria can produce secondary metabolites that potentially possess important antibacterial and cytotoxic properties, but also reinforce the pressing need to conserve biomes such as the Brazilian Pantanal.


Subject(s)
Actinobacteria/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Endophytes/chemistry , Poaceae/microbiology , Actinobacteria/classification , Actinobacteria/isolation & purification , Actinobacteria/metabolism , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Brazil , Cell Line, Tumor , Cell Survival/drug effects , Endophytes/classification , Endophytes/isolation & purification , Endophytes/metabolism , Enterococcus/drug effects , Enterococcus/growth & development , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice , Wetlands
4.
J Clin Med ; 9(8)2020 Aug 18.
Article in English | MEDLINE | ID: mdl-32824699

ABSTRACT

Natural killer (NK) cells are innate lymphocytes responsible for the elimination of infected or transformed cells. The activation or inhibition of NK cells is determined by the balance of target cell ligand recognition by stimulatory and inhibitory receptors on their surface. Previous reports have suggested that the glycosaminoglycan heparin is a ligand for the natural cytotoxicity receptors NKp30, NKp44 (human), and NKp46 (both human and mouse). However, the effects of heparin on NK cell homeostasis and function remain unclear. Here, we show that heparin does not enhance NK cell proliferation or killing through NK cell activation. Alternatively, in mice models, heparin promoted NK cell survival in vitro and controlled B16-F10 melanoma metastasis development in vivo. In human NK cells, heparin promisingly increased interferon (IFN)-γ production in synergy with IL-12, although the mechanism remains elusive. Our data showed that heparin is not able to increase NK cell cytotoxicity.

5.
J Pharm Sci ; 109(7): 2294-2301, 2020 07.
Article in English | MEDLINE | ID: mdl-32311369

ABSTRACT

This work describes the development of polysaccharide-coated liposomes to modulate the delivery of epidermal growth factor (EGF), with the aim to produce different EGF release profiles depending on the milieu of infected wounds. For this purpose, cationic liposomes were coated with one layer of sodium alginate (ALG) followed by one layer of chitosan (CHI) using the layer-by-layer (LbL) technique. The coated liposomes exhibited apparent hydrodynamic diameters of 278 ± 36 and 216 ± 96 nm for Lip-ALG and Lip-ALG-CHI, respectively. Thus, it appears that adding the CHI layer compacted the Lip-ALG one. The incorporation efficiency of EGF was a maximum of 55% for liposomes with a polymeric coating. In vitro release experiments showed that Lip-ALG-CHI exhibits a higher release rate constant under acidic pH conditions, resembling those of infected tissue. Using an ex vivo model of EGF release in porcine ear skin, these liposomes were found to accumulate in the epidermis. Thus, coated liposomes could represent a local EGF delivery mechanism to promote healing.


Subject(s)
Chitosan , Liposomes , Alginates , Animals , Epidermal Growth Factor , Skin , Swine
6.
Front Immunol ; 11: 73, 2020.
Article in English | MEDLINE | ID: mdl-32063906

ABSTRACT

The tumor microenvironment (TME) is composed of multiple infiltrating host cells (e.g., endothelial cells, fibroblasts, lymphocytes, and myeloid cells), extracellular matrix, and various secreted or cell membrane-presented molecules. Group 1 innate lymphoid cells (ILCs), which includes natural killer (NK) cells and ILC1, contribute to protecting the host against cancer and infection. Both subsets are able to quickly produce cytokines such as interferon gamma (IFN-γ), chemokines, and other growth factors in response to activating signals. However, the TME provides many molecules that can prevent the potential effector function of these cells, thereby protecting the tumor. For example, TME-derived tumor growth factor (TGF)-ß and associated members of the superfamily downregulate NK cell cytotoxicity, cytokine secretion, metabolism, proliferation, and induce effector NK cells to upregulate ILC1-like characteristics. In concert, a family of carbohydrate-binding proteins called galectins, which can be produced by different cells composing the TME, can downregulate NK cell function. Matrix metalloproteinase (MMP) and a disintegrin and metalloproteinase (ADAM) are also enzymes that can remodel the extracellular matrix and shred receptors from the tumor cell surface, impairing the activation of NK cells and leading to less effective effector functions. Gaining a better understanding of the characteristics of the TME and its associated factors, such as infiltrating cells and extracellular matrix, could lead to tailoring of new personalized immunotherapy approaches. This review provides an overview of our current knowledge on the impact of the TME and extracellular matrix-associated components on differentiation, impairment, and function of NK cells.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Tumor Microenvironment/immunology , Cell Differentiation , Chemokines/metabolism , Cytokines/metabolism , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Galectins , Glycosaminoglycans , Humans , Immunotherapy , Interferon-gamma/metabolism , Proteoglycans
7.
Carbohydr Polym ; 225: 115203, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31521290

ABSTRACT

A fucomannogalactan (FMG-Hm), with a molecular weight of 17.1 kDa, obtained from fruiting bodies of Hypsizygus marmoreus exhibited promising in vitro antimelanoma effects. FMG-Hm was not cytotoxic, nor did it alter the cell morphology and proliferation, but was able to inhibit colony-forming ability and cell migration in B16-F10 murine melanoma cells. An analysis of the monosaccharide composition indicated that FMG-Hm was composed of fucose, mannose, and galactose in a ratio of 1.00:1.08:3.17. The FMG-Hm was structurally characterized based on methylation analysis, partial acid hydrolysis, and NMR experiments. The results indicated that FMG-Hm contained a α-(1→6)-linked galactopyranosyl main chain, partially substituted at O-2 by non-reducing ends of α-L-fucopyranose and ß-D-mannopyranose. The predicted structure of the heteropolysaccharide was established as.


Subject(s)
Agaricales/metabolism , Galactans/chemistry , Galactans/isolation & purification , Galactans/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Cell Line, Tumor , Cell Movement/drug effects , Fruiting Bodies, Fungal/metabolism , Molecular Structure , Molecular Weight
8.
Int J Oncol ; 55(3): 721-732, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31364728

ABSTRACT

Metastasis is responsible for the majority of deaths among patients with malignant melanoma. Despite recent advances, the majority of current and modern therapies are ineffective and/or financially unfeasible. Thus, in this study, we investigated two low­cost highly­diluted natural complexes (HDNCs) that have been shown to be effective against malignant melanoma in a murine model in vivo. The aim of this study was to determine the mechanisms through which these HDNCs directly affect melanoma cells, either alone or in an artificial tumor microenvironment, suppressing the metastatic phenotype, thus explaining previous in vivo effects. For this purpose, HDNC in vitro treatments of B16­F10 melanoma cells, alone or in co­culture with Balb/3T3 fibroblasts, were carried out. Molecular biology techniques and standard functional assays were used to assess the changes in molecule expression and in cell behaviors related to the metastatic phenotype. Melanoma progression features were found to be regulated by HDNCs. Molecules related to cell adhesion (N­cadherin, ß1­integrin and CD44), and migration, extracellular matrix remodeling and angiogenesis were modulated. The cell migratory, invasive and clonogenic capacities were reduced by the HDNCs. No loss of cell proliferation or viability were observed. On the whole, the findings of this study indicate that HDNCs directly reprogram, molecularly and functionally, melanoma cells in vitro, modulating their metastatic phenotype. Such findings are likely to be responsible for the attenuation of tumor growth and lung colonization previously observed in vivo.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Hyaluronan Receptors/metabolism , Integrin beta1/metabolism , Melanoma/metabolism , Plant Extracts/pharmacology , Skin Neoplasms/metabolism , Animals , BALB 3T3 Cells , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic , Humans , Melanoma/drug therapy , Mice , Neoplasm Metastasis , Plant Extracts/chemistry , Plants/chemistry , Skin Neoplasms/drug therapy , Melanoma, Cutaneous Malignant
9.
Sleep Sci ; 12(3): 196-202, 2019.
Article in English | MEDLINE | ID: mdl-31890096

ABSTRACT

Excitotoxicity has been related to play a crucial role in Parkinson's disease (PD) pathogenesis. Pedunculopontine tegmental nucleus (PPT) represents one of the major sources of glutamatergic afferences to nigrostriatal pathway and putative reciprocal connectivity between these structures may exert a potential influence on rapid eye movement (REM) sleep control. Also, PPT could be overactive in PD, it seems that dopaminergic neurons are under abnormally high levels of glutamate and consequently might be more vulnerable to neurodegeneration. We decided to investigate the neuroprotective effect of riluzole administration, a N-methyl-D-aspartate (NMDA) receptor antagonist, in rats submitted simultaneously to nigrostrial rotenone and 24h of REM sleep deprivation (REMSD). Our findings showed that blocking NMDA glutamatergic receptors in the SNpc, after REMSD challenge, protected the dopaminergic neurons from rotenone lesion. Concerning rotenone-induced hypolocomotion, riluzole reversed this impairment in the control groups. Also, REMSD prevented the occurrence of rotenone-induced motor impairment as a result of dopaminergic supersensitivity. In addition, higher Fluoro Jade C (FJC) staining within the SNpc was associated with decreased cognitive performance observed in rotenone groups. Such effect was counteracted by riluzole suggesting the occurrence of an antiapoptotic effect. Moreover, riluzole did not rescue cognitive impairment impinged by rotenone, REMSD or their combination. These data indicated that reductions of excitotoxicity, by riluzole, partially protected dopamine neurons from neuronal death and appeared to be effective in relieve specific rotenone-induce motor disabilities.

10.
Int J Biol Macromol ; 121: 239-248, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30267823

ABSTRACT

World fisheries and aquaculture production totaled 167 million tons in 2014. This high fish production generates a lot of waste that could be used as raw material for extraction of substances of pharmacological interest. In this work, we extract and characterize glycosaminoglycans (GAGs) present in the viscera of Nile tilapia (Oreochromis niloticus) and Pacu (Piaractus mesopotamicus), which are among the most vastly produced fishes in inland aquaculture in Brazil. Moreover, the anticoagulant activity of the GAGs fractions was evaluated. GAGs were obtained from total defatted viscera, after proteolysis, precipitation with ethanol, anion exchange chromatography and treatment with chondroitinase. Chondroitin sulfate (CS), dermatan sulfate (DS) and heparan sulfate (HS) were identified by agarose gel electrophoresis and NMR analyses. CS, DS and HS were identified in equivalent fractions obtained from both fishes, and all GAGs fractions showed anticoagulant activity.


Subject(s)
Anticoagulants/isolation & purification , Anticoagulants/pharmacology , Characiformes/anatomy & histology , Cichlids/anatomy & histology , Glycosaminoglycans/isolation & purification , Glycosaminoglycans/pharmacology , Viscera/chemistry , Animals
11.
Front Microbiol ; 9: 2211, 2018.
Article in English | MEDLINE | ID: mdl-30356683

ABSTRACT

The fungal genus Fonsecaea comprises etiological agents of human chromoblastomycosis, a chronic implantation skin disease. The current hypothesis is that patients acquire the infection through an injury from plant material. The present study aimed to evaluate a model of infection in plant and animal hosts to understand the parameters of trans-kingdom pathogenicity. Clinical strains of causative agents of chromoblastomycosis (Fonsecaea pedrosoi and Fonsecaea monophora) were compared with a strain of Fonsecaea erecta isolated from a living plant. The clinical strains of F. monophora and F. pedrosoi remained concentrated near the epidermis, whereas F. erecta colonized deeper plant tissues, resembling an endophytic behavior. In an invertebrate infection model with larvae of a beetle, Tenebrio molitor, F. erecta exhibited the lowest survival rates. However, F. pedrosoi produced dark, spherical to ovoidal cells that resembled muriform cells, the invasive form of human chromoblastomycosis confirming the role of muriform cells as a pathogenic adaptation in animal tissues. An immunologic assay in BALB/c mice demonstrated the high virulence of saprobic species in animal models was subsequently controlled via host higher immune response.

12.
Carbohydr Polym ; 190: 50-56, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29628259

ABSTRACT

An aqueous extract containing polysaccharides was obtained from the giant mushroom Macrocybe titans, and it was purified by amylase treatment, freeze-thawing process and dialysis. The purified fraction (ESP) was analyzed by HPSEC and GC-MS which showed a homogenous polysaccharide with Mw 14.2 × 103 g/mol composed by galactose and fucose. NMR and methylation analysis of ESP confirmed the presence of a fucogalactan with a (1 → 6)-linked α-d-Galp main chain partially substituted at O-2 by non reducing end units of α-l-Fucp residues in the side chain. Its biological activity was evaluated against murine melanoma cells B16-F10. The fucogalactan did not alter the viability, proliferative capacity and morphology of cells. However, this polysaccharide was able to reduce the cell migration in vitro at 40% (100 µg/mL) and 33% (250 µg/mL). The results obtained showed that Macrocybe titans fucogalactan is a promising agent capable of altering melanoma cell migration without decrease the cell viability.


Subject(s)
Agaricales/chemistry , Cell Movement/drug effects , Galactans/pharmacology , Melanoma, Experimental/pathology , Melanoma/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Galactans/chemistry
13.
Article in English | MEDLINE | ID: mdl-28750863

ABSTRACT

Hemoplasmas were detected in two apparently healthy captive South American coatis (Nasua nasua) from southern Brazil during an investigation for vector-borne pathogens. Blood was subjected to packed cell volume (PCV) determination, a commercial real-time PCR panel for the detection of Anaplasma spp., Babesia spp., Bartonella spp., Hepatozoon spp., Leishmania spp., Mycoplasma haemofelis, 'Candidatus Mycoplasma turicensis', 'Candidatus Mycoplasma haemominutum', Neorickettsia risticii, Rickettsia rickettsii and Leptospira spp., and a pan-hemoplasma conventional PCR assay. PCV was normal, but both coatis tested positive for hemoplasmas and negative for all the remaining pathogens tested. Using different techniques for microscopy (light, confocal or SEM), structures compatible with hemoplasmas were identified. Sequencing of the 16S rRNA gene identified an organism resembling Mycoplasma haemofelis and another hemotropic Mycoplasma sp., with a sequence identity of 96.8% to a Mycoplasma sp. previously detected in capybaras.


Subject(s)
Mycoplasma Infections/veterinary , Mycoplasma/isolation & purification , Procyonidae/microbiology , Animals , Animals, Domestic , Animals, Wild/microbiology , Brazil/epidemiology , DNA, Bacterial/genetics , Female , Male , Microscopy , Microscopy, Confocal , Microscopy, Electron, Scanning/methods , Mycoplasma/genetics , Mycoplasma/ultrastructure , Mycoplasma Infections/epidemiology , Mycoplasma Infections/microbiology , Mycoplasma Infections/transmission , RNA, Ribosomal, 16S/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
14.
Fish Shellfish Immunol ; 40(1): 296-303, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25038277

ABSTRACT

The non-steroidal anti-inflammatory drugs are emerging contaminants in aquatic ecosystems. This study aimed to evaluate toxic effects of some representative drugs of this pharmaceutical group on primary culture of monocytic lineage of Hoplias malabaricus anterior kidney. The effects of diclofenac, acetaminophen and ibuprofen in cell viability, lipopolysaccharide (LPS)-induced NO production and genotoxicity were evaluated. Cytometry analysis CD11b(+) cells showed 71.5% of stem cells, 19.5% of macrophages and 9% of monocytes. Cell viability was lower in the ficoll compared to percoll separation. LPS-induced NO production by these cells was blocked after treatment with dexamethasone and NG-Methyl-L-Arginine (L-NMMA). Exposure of the cells to diclofenac (0.2-200 ng/mL), acetaminophen (0.025-250 ng/mL) ibuprofen (10-1000 ng/mL) reduced basal NO production and inhibited LPS-induced NO production at all concentrations after 24 h of exposure. Genotoxicity occurred at the highest concentration of diclofenac and at the intermediary concentration of acetaminophen. Genotoxicity was also observed by ibuprofen. In summary, the pharmaceuticals influenced NO production and caused DNA damage in monocytic cells suggesting that these drugs can induce immunosuppression and genotoxicity in fish.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Characidae/metabolism , Animals , Cell Culture Techniques , Cell Survival/drug effects , Cells, Cultured , Diclofenac/pharmacology , Ibuprofen/pharmacology , Lipopolysaccharides/pharmacology , Mutagenicity Tests , Nitric Oxide/metabolism
15.
Chem Biol Interact ; 205(1): 29-37, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23756126

ABSTRACT

Galactofuranoside derivatives were synthesised by the classic Fischer glycosydation method, and their immune modulation properties were studied in vitro and in vivo. NMR spectroscopic and ESI-MS analyses confirmed the purity and authenticity of all derivatives. Their phagocyte capacities were tested in resident macrophages. Methyl ß-galactofuranoside (GFB-Me) and n-octyl ß-galactofuranoside (GFB-O) had an immune stimulant effect at 25µmolml(-1) with an enhancement of 35.12%±0.06 SD and 17.49%±0.11 SD, respectively, but Methyl α-galactofuranoside (GFA-Me) and n-octyl α-galactofuranoside (GFA-O) gave a low immune response. Methyl α-galactofuranoside 5,6-O-isopropylidene (GFA-IP) and Methyl ß-galactofuranoside 5,6-O-isopropylidene (GFB-IP) had negative values relative to the control group of minus 4.96%±0.10 SD and -40.72%±0.07 SD, respectively. Furthermore, GFB-Me and GFB-Me-IP were evaluated in vivo on the lethality induced by cecal ligation and puncture. Cytokine levels and iNOS expression were determined and correlated to mortality data. The results showed that the free HO-5 and HO-6 and the ß-configuration are essential for the induction of phagocytic activity by the galactofuranosyl units. The methyl ß-galactofuranosides also enhanced lethality during sepsis, increasing the levels of pro-inflammatory cytokines and iNOS expression.


Subject(s)
Glycosides/pharmacology , Immunologic Factors/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Sepsis/immunology , Animals , Cytokines/biosynthesis , Glycosides/chemistry , Glycosides/toxicity , Hydrogen Peroxide/metabolism , Ileum/drug effects , Ileum/immunology , Ileum/metabolism , Immunologic Factors/chemistry , Immunologic Factors/toxicity , Magnetic Resonance Spectroscopy , Male , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Phagocytosis/drug effects , Sepsis/etiology , Sepsis/metabolism , Superoxides/metabolism
16.
Mol Pharm ; 9(9): 2686-97, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22873645

ABSTRACT

In recent years, the antitumoral activity of antimicrobial peptides (AMPs) has been the goal of many research studies. Among AMPs, gomesin (Gm) displays antitumor activity by unknown mechanisms. Herein, we studied the cytotoxicity of Gm in the Chinese hamster ovary (CHO) cell line. Furthermore, we investigated the temporal ordering of organelle changes and the dynamics of Ca(2+) signaling during Gm-induced cell death. The results indicated that Gm binds to the plasma membrane and rapidly translocates into the cytoplasm. Moreover, 20 µM Gm increases the cytosolic Ca(2+) and induces membrane permeabilization after 30 min of treatment. Direct Ca(2+) measurements in CHO cells transfected with the genetically encoded D1-cameleon to the endoplasmic reticulum (ER) revealed that Gm induces ER Ca(2+) depletion, which in turn resulted in oscillatory mitochondrial Ca(2+) signal, as measured in cells expressing the genetically encoded probe to the mitochondrial matrix (mit)Pericam. This leads to mitochondria disruption, loss of mitochondrial membrane potential and increased reactive oxygen species prior to membrane permeabilization. Gm-induced membrane permeabilization by a Ca(2+)-dependent pathway involving Gm translocation into the cell, ER Ca(2+) depletion and disruption, mitochondrial Ca(2+) overload and oxidative stress.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Calcium/metabolism , Cell Death/drug effects , Cell Membrane Permeability/drug effects , Animals , CHO Cells , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cytoplasm/drug effects , Cytoplasm/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
17.
Toxicon ; 60(3): 396-405, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22538195

ABSTRACT

Brown spider (Loxosceles sp.) venom affects the endothelium of vessels and triggers disruptive activity in the subendothelial matrix. The vascular disorders observed after venom exposure include leukocyte and platelet activation, disseminated intravascular coagulation, an increase in vessel permeability and hemorrhage into the dermis. In this study, we report additional evidence regarding the mechanism of endothelial cell cytotoxicity induced by Loxosceles intermedia venom. Exposure to venom led to endothelial cell detachment in a time-dependent manner. Loss of cell anchorage and cell-cell adhesion following venom exposure was accompanied by changes in the distribution of the α5ß1 integrin and VE-cadherin. An ultrastructural analysis of cells treated with venom revealed morphological alterations characteristic of apoptosis. Moreover, after venom exposure, the ratio between Bax and Bcl-2 proteins was disturbed in favor of Bax. In addition, late apoptosis was only observed in cells detached by the action of venom. Accordingly, there was no increase in apoptosis when cells were exposed to L. intermedia venom in suspension, suggesting that the loss of cell anchorage provides the signal to initiate apoptosis. Thus, L. intermedia venom likely triggers endothelial cell death indirectly through an apoptotic mechanism known as anoikis.


Subject(s)
Anoikis/drug effects , Endothelium, Vascular/drug effects , Spider Venoms/pharmacology , Spiders/metabolism , Animals , Antigens, CD/metabolism , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/ultrastructure , Apoptosis Regulatory Proteins/metabolism , Brazil , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Shape/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Integrin alpha5beta1/metabolism , Kinetics , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Rabbits
18.
Matrix Biol ; 31(5): 308-16, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22504459

ABSTRACT

Heparin and its derivatives are known to regulate a variety of pathophysiological events related to vascular biology. In the present manuscript we examine a variety of heparinomimetics biochemically (electrophoretic behavior and enzymatic degradation) and pharmacologically (in vitro anticoagulant activity and in vivo hemorrhagic and antithrombotic tests) as well as their interactions with cells from the vessel wall using a time resolved fluorometric method and confocal microscopy. Data were determined for unfractionated heparin (UFH), enoxaparin, synthetic heparin pentasaccharide, C3 heparin derived oligosaccharides and phosphosulfomannan (PI-88). While being structurally distinct from UFH, all compounds exhibited anticoagulant, antithrombotic and hemorrhagic activities. In addition, besides the pentasaccharide, they all stimulated the synthesis of an antithrombotic heparan sulfate present at the cell surface and secreted by endothelial cells. Also, like UFH, they interacted with both endothelial and smooth muscle cells and dislodged UFH from its binding sites in a dose dependent manner but, with distinct saturable curves showing that the binding of polymeric structures to extracellular matrix (ECM) proteins does not depend on a glycosaminoglycan backbone. The data also suggest a common pathway, which does not depend on the presence of the conventionally accepted antithrombin binding pentasaccharide, for ECM dependent activity of the heparinomimetic stimulated synthesis of antithrombotic heparan sulfate. Notably, although of similar molecular weight as well as polymeric backbone, the synthetic heparin pentasaccharide showed significant hemorrhagic action and negligible antithrombotic activity in a venous thrombosis model, contrasting with C3, that displayed negligible hemorrhagic effect and potent antithrombotic action. These results provide evidence that structurally unrelated polymers can elicit similar hemostatic activities and show that polymeric sequence is not always crucial for certain activities. The results also suggest that non-GAG structures may provide an alternative route for the pharmaceutical control of hemostasis.


Subject(s)
Extracellular Matrix/drug effects , Hemostasis , Heparin/analogs & derivatives , Heparin/pharmacology , Animals , Anticoagulants/pharmacology , Binding Sites , Dose-Response Relationship, Drug , Endothelial Cells/chemistry , Endothelial Cells/drug effects , Extracellular Matrix/chemistry , Fibrinolytic Agents/pharmacology , Heparin Lyase/chemistry , Molecular Weight , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/drug effects , Oligosaccharides/pharmacology , Protein Binding , Proteolysis , Rabbits , Rats , Substrate Specificity
19.
J Cell Physiol ; 227(6): 2740-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21898414

ABSTRACT

Endothelial cells (ECs) are a source of physiologically important molecules that are synthesized and released to the blood and/or to the subendothelial extracellular matrix such as a heparan sulfate proteoglycan (HSPG) with antithrombotic properties. Previously, we have shown that heparin stimulates the synthesis and modifies the sulfation pattern of this HSPG. Here the molecular mechanisms involved in the up-regulation of HSPG synthesis by heparin in endothelial cells were decoded. The cells were stimulated with heparin and the expression of HSPG and intracellular pathways were evaluated by a combination of methods involving confocal microscopy, flow cytometry, Western blotting analyses, and [(35) S]-sulfate metabolically labeling of the cells. We observed that the up-regulation of HSPG synthesis evoked by heparin is dependent on the interaction of heparin with integrin since RGD peptide abolishes the effect. The activation of integrin leads to tyrosine-phosphorylation of focal adhesion-associated proteins such as FAK, Src, and paxillin. In addition, heparin induces ERK1/2 phosphorylation and inhibitors of Ras and MEK decreased heparin-dependent HSPG synthesis. Moreover, heparin also induced intracellular Ca(2+) release, PLCγ1 (phospholipase Cγ1) and CaMKII (calcium calmodulin kinase II) activation, as well as an increase in nitric oxide (NO) production. Finally, an intracellular Ca(2+) chelator, Ca(2+) signaling inhibitors, and an endothelial NO synthase inhibitor were all able to abolish the effect in heparan sulfate synthesis. In conclusion, the heparin-induced up-regulation of HSPG expression is associated with the phosphorylation of focal adhesion proteins and Ras/Raf/MEK/ERK MAP and Ca(2+) /NO pathways.


Subject(s)
Endothelial Cells/drug effects , Gene Expression Regulation/physiology , Heparin/metabolism , Heparitin Sulfate/metabolism , Integrins/metabolism , Animals , Anticoagulants/metabolism , Anticoagulants/pharmacology , Blotting, Western , Calcium/metabolism , Calcium Signaling , Cell Adhesion , Fibronectins/chemistry , Fibronectins/metabolism , Flow Cytometry , Heparin/pharmacology , Heparitin Sulfate/genetics , Microscopy, Confocal , Nitric Oxide , Phosphorylation , Protein Binding , Rabbits , Up-Regulation
20.
Hypertension ; 56(4): 713-21, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20805441

ABSTRACT

Previous reports have shown that heparin may promote human hypotension and vascular relaxation by elevation of NO levels through unclear mechanisms. We hypothesized that endothelial muscarinic M(3) receptor activation mediates the heparin-induced vasodilation of rat aortic rings. The experiments were carried out using unfractionated heparin extracted from bovine intestinal mucosa, which elicited an endothelium and NO-dependent relaxation of aortic segments with maximal potency and efficacy (EC(50): 100±10 µmol/L; E(max): 41±3%). Atropine and 1,1-dimethyl-4-diphenylacetoxypiperidinium iodide inhibitors reduced the heparin-dependent relaxation, indicating that M(3) muscarinic receptor is involved in this phenomenon. However, no direct binding of heparin to muscarinic receptors was observed. More importantly, studies performed using the arginine-glycine-aspartic acid peptide and 1-(1,1-dimethylethyl)-3-(1-naphthalenyl)-1H-pyrazolo[3,4-day]pyrimidin-4-amine, an Src family inhibitor, reduced by 51% and 73% the heparin-dependent relaxation, respectively, suggesting the coupling of heparin and M(3) receptor through extracellular matrix molecules and integrin. Furthermore, unfractionated heparin induced activation of focal adhesion protein kinase, Src, and paxillin. Finally, fluorescence resonance energy transfer approach confirmed the interaction of the M(3) receptor to integrin. Taken together, these data demonstrate the participation of M(3) receptor and integrin in heparin-dependent relaxation of vascular smooth muscle. These results provide new insights into the molecular mechanism and potential pharmacological action of heparin in vascular physiology.


Subject(s)
Aorta, Thoracic/drug effects , Heparin/pharmacology , Integrins/metabolism , Receptor, Muscarinic M3/metabolism , Vasodilation/drug effects , Acetylcholine/pharmacology , Animals , Anticoagulants/pharmacology , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiology , Atropine/pharmacology , Blotting, Western , Cattle , Cell Line , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Fluorescence Resonance Energy Transfer , In Vitro Techniques , Male , Nitric Oxide/biosynthesis , Oligopeptides/pharmacology , Paxillin/metabolism , Protein Binding , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rats , Rats, Wistar , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...