Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(22): e2219216120, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37216514

RESUMO

The assembly of the ß-amyloid peptide (Aß) to form oligomers and fibrils is closely associated with the pathogenesis and progression of Alzheimer's disease. Aß is a shape-shifting peptide capable of adopting many conformations and folds within the multitude of oligomers and fibrils the peptide forms. These properties have precluded detailed structural elucidation and biological characterization of homogeneous, well-defined Aß oligomers. In this paper, we compare the structural, biophysical, and biological characteristics of two different covalently stabilized isomorphic trimers derived from the central and C-terminal regions Aß. X-ray crystallography reveals the structures of the trimers and shows that each trimer forms a ball-shaped dodecamer. Solution-phase and cell-based studies demonstrate that the two trimers exhibit markedly different assembly and biological properties. One trimer forms small soluble oligomers that enter cells through endocytosis and activate capase-3/7-mediated apoptosis, while the other trimer forms large insoluble aggregates that accumulate on the outer plasma membrane and elicit cellular toxicity through an apoptosis-independent mechanism. The two trimers also exhibit different effects on the aggregation, toxicity, and cellular interaction of full-length Aß, with one trimer showing a greater propensity to interact with Aß than the other. The studies described in this paper indicate that the two trimers share structural, biophysical, and biological characteristics with oligomers of full-length Aß. The varying structural, assembly, and biological characteristics of the two trimers provide a working model for how different Aß trimers can assemble and lead to different biological effects, which may help shed light on the differences among Aß oligomers.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Peptídeos beta-Amiloides/metabolismo , Conformação Proteica , Cristalografia por Raios X , Membrana Celular/metabolismo , Fragmentos de Peptídeos/química
2.
J Appl Toxicol ; 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38711185

RESUMO

Dental resin systems, used for artificial replacement of teeth and their surrounding structures, have gained popularity due to the Food and Drug Administration's (FDA) recommendation to reduce dental amalgam use in high-risk populations and medical circumstances. Bisphenol A (BPA), an endocrine-disrupting chemical, is an essential monomer within dental resin in the form of various analogues and derivatives. Leaching of monomers from resins results in toxicity, affecting hormone metabolism and causing long-term health risks. Understanding cellular-level toxicity profiles of bisphenol derivatives is crucial for conducting toxicity studies in in vivo models. This review provides insights into the unique expression patterns of BPA and its analogues among different cell types and their underlying toxicity mechanisms. Lack of a consistent cell line for toxic effects necessitates exploring various cell lines. Among the individual monomers, BisGMA was found to be the most toxic; however, BisDMA and BADGE generates BPA endogenously and found to elicit severe adverse reactions. In correlating in vitro data with in vivo findings, further research is necessary to classify the elutes as human carcinogens or xenoestrogens. Though the basic mechanisms underlying toxicity were believed to be the production of intracellular reactive oxygen species and a corresponding decline in glutathione levels, several underlying mechanisms were identified to stimulate cellular responses at low concentrations. The review calls for further research to assess the synergistic interactions of co-monomers and other components in dental resins. The review emphasizes the clinical relevance of these findings, highlighting the necessity for safer dental materials and underscoring the potential health risks associated with current dental resin systems.

3.
Plant Cell Physiol ; 63(12): 1927-1942, 2023 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-35997763

RESUMO

Plants activate a myriad of signaling cascades to tailor adaptive responses under environmental stresses, such as salinity. While the roles of exogenous karrikins (KARs) in salt stress mitigation are well comprehended, genetic evidence of KAR signaling during salinity responses in plants remains unresolved. Here, we explore the functions of the possible KAR receptor KARRIKIN-INSENSITIVE2 (KAI2) in Arabidopsis thaliana tolerance to salt stress by investigating comparative responses of wild-type (WT) and kai2-mutant plants under a gradient of NaCl. Defects in KAI2 functions resulted in delayed and inhibited cotyledon opening in kai2 seeds compared with WT seeds, suggesting that KAI2 played an important role in enhancing seed germination under salinity. Salt-stressed kai2 plants displayed more phenotypic aberrations, biomass reduction, water loss and oxidative damage than WT plants. kai2 shoots accumulated significantly more Na+ and thus had a lower K+/Na+ ratio, than WT, indicating severe ion toxicity in salt-stressed kai2 plants. Accordingly, kai2 plants displayed a lower expression of genes associated with Na+ homeostasis, such as SALT OVERLY SENSITIVE (SOS) 1, SOS2, HIGH-AFFINITY POTASSIUM TRANSPORTER 1;1 (HKT1;1) and CATION-HYDROGEN EXCHANGER 1 (NHX1) than WT plants. WT plants maintained a better glutathione level, glutathione-related redox status and antioxidant enzyme activities relative to kai2 plants, implying KAI2's function in oxidative stress mitigation in response to salinity. kai2 shoots had lower expression levels of genes involved in the biosynthesis of strigolactones (SLs), salicylic acid and jasmonic acid and the signaling of abscisic acid and SLs than those of WT plants, indicating interactive functions of KAI2 signaling with other hormone signaling in modulating plant responses to salinity. Collectively, these results underpin the likely roles of KAI2 in the alleviation of salinity effects in plants by regulating several physiological and biochemical mechanisms involved in ionic and osmotic balance, oxidative stress tolerance and hormonal crosstalk.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Tolerância ao Sal/genética , Proteínas de Transporte/metabolismo , Glutationa/metabolismo , Regulação da Expressão Gênica de Plantas
4.
Environ Res ; 236(Pt 1): 116776, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37517486

RESUMO

A steadily increasing production volume of nanoparticles reflects their numerous industrial and domestic applications. These economic successes come with the potential adverse effects on natural systems that are associated with their presence in the environment. Biological activities and effects of nanoparticles are affected by their entry method together with their specificities like their size, shape, charge, area, and chemical composition. Particles can be classified as safe or dangerous depending on their specific properties. As both aquatic and terrestrial systems suffer from organic and inorganic contamination, nanoparticles remain a sink for these contaminants. Researching the sources, synthesis, fate, and toxicity of nanoparticles has advanced significantly during the last ten years. We summarise nanoparticle pathways throughout the ecosystem and their interactions with beneficial microorganisms in this research. The prevalence of nanoparticles in the ecosystem causes beneficial microorganisms to become hazardous to their cells, which prevents the synthesis of bioactive molecules from undergoing molecular modifications and diminishes the microbe population. Recently, observed concentrations in the field could support predictions of ambient concentrations based on modeling methodologies. The aim is to illustrate the beneficial and negative effects that nanoparticles have on aqueous and terrestrial ecosystems, as well as the methods utilized to reduce their toxicity.

5.
Environ Res ; 230: 115046, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-36525994

RESUMO

Exposure to asbestos and asbestos-like minerals has been related to the development of severe lung diseases, including cancer and malignant mesothelioma (MM). A high incidence of non-occupational MM was observed in New Caledonia (France) in people living in proximity of serpentinite outcrops, containing chrysotile and fibrous antigorite. Antigorite is a magnesium silicate, which shares with chrysotile asbestos the chemical formula. To achieve information on antigorite toxicity, we investigated the physico-minero-chemical features relevant for toxicity and cellular effects elicited on murine macrophages (MH-S) and alveolar epithelial cells (A549) of three fibrous antigorites (f-Atg) collected in a Caledonian nickel lateritic ore and subjected to supergene alteration. Field Atg were milled to obtain samples suitable for toxicological studies with a similar particle size distribution. UICC chrysotile (Ctl) and a non-fibrous antigorite (nf-Atg) were used as reference minerals. A high variability in toxicity was observed depending on shape, chemical alteration, and surface reactivity. The antigorites shared with Ctl a similar surface area (16.3, 12.1, 20.3, 13.4, and 15.6 m2/g for f-Atg1, 2, 3, nf-Atg, and Ctl). f-Atg showed different level of pedogenetic weathering (Ni depletion f-Atg1 ≪ f-Atg2 and 3) and contained about 50% of elongated mineral particles, some of which exhibited high aspect ratios (AR > 10 µm, 20%, 26%, 31% for f-Atg1, 2, and 3, respectively). The minerals differed in bio-accessible iron at pH 4.5 (f-Atg1 ≪ f-Atg3, < f-Atg2, nf-Atg < Ctl), and surface reactivity (ROS release in solution, f-Atg1 ≪ f-Atg2, 3, nf-Atg, and Ctl). f-Atg2 and f-Atg3 induced oxidative stress and pro-inflammatory responses, while the less altered, poorly reactive sample (f-Atg1) induced negligible effects, as well nf-Atg. The slow dissolution kinetics observed in simulated body fluids may signal a high biopersistence. Overall, our work revealed a significative cellular toxicity of f-Atg that correlates with fibrous habit and surface reactivity.


Assuntos
Asbestos Serpentinas , Amianto , Humanos , Camundongos , Animais , Asbestos Serpentinas/toxicidade , Nova Caledônia , Amianto/toxicidade , Minerais/toxicidade , Silicatos
6.
J Appl Toxicol ; 43(11): 1576-1593, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36806101

RESUMO

Microplastics (MPs) are plastic particles of a diameter of less than 5 mm and a major carrier of pollution. In accordance with its diameter range, MPs can be divided into microplastics (100-5 mm) and nanoplastics (<100 nm). In recent years, in addition to the impact of MPs on the environment, the ways in which MPs affect the body has also attracted continuous attention. However, relevant studies on the cytotoxicity of MPs are not comprehensive. Based on the current research, this paper summarizes four main cytotoxic mechanisms of MPs, inducing oxidative stress, damaging cell membrane organelles, inducing immune response, and genotoxicity. Generally, MPs cause cytotoxicity such as oxidative stress, damage to cell membranes and organelles, activation of immune responses, and genotoxicity through mechanical damage or induction of cells to produce reactive oxygen species. Understanding these toxic mechanisms is helpful for the evaluation and prevention of human toxicity of MPs. This paper also analyzes the limitations of current research and prospects for future research into cellular MPs, with the aim of providing a scientific basis and reference for further research into the toxic mechanism of MPs.

7.
Curr Issues Mol Biol ; 44(3): 1294-1315, 2022 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-35723310

RESUMO

Hepatic cells are sensitive to internal and external signals. Ethanol is one of the oldest and most widely used drugs in the world. The focus on the mechanistic engine of the alcohol-induced injury has been in the liver, which is responsible for the pathways of alcohol metabolism. Ethanol undergoes a phase I type of reaction, mainly catalyzed by the cytoplasmic enzyme, alcohol dehydrogenase (ADH), and by the microsomal ethanol-oxidizing system (MEOS). Reactive oxygen species (ROS) generated by cytochrome (CYP) 2E1 activity and MEOS contribute to ethanol-induced toxicity. We aimed to: (1) Describe the cellular, pathophysiological and clinical effects of alcohol misuse on the liver; (2) Select the biomarkers and analytical methods utilized by the clinical laboratory to assess alcohol exposure; (3) Provide therapeutic ideas to prevent/reduce alcohol-induced liver injury; (4) Provide up-to-date knowledge regarding the Corona virus and its affect on the liver; (5) Link rare diseases with alcohol consumption. The current review contributes to risk identification of patients with alcoholic, as well as non-alcoholic, liver disease and metabolic syndrome. Additional prevalence of ethnic, genetic, and viral vulnerabilities are presented.

8.
Int J Mol Sci ; 23(20)2022 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-36293416

RESUMO

Curing salts composed of mixtures of nitrates and nitrites are preservatives widely used in processed meats. Despite many desirable technological effects, their use in meat products has been linked to methemoglobinemia and the formation of nitrosamines. Therefore, an increasing "anti-nitrite feeling" has grown among meat consumers, who search for clean label products. In this view, the use of natural compounds as alternatives represents a challenge for the meat industry. Processing (including formulation and fermentation) induces chemical or physical changes of food matrix that can modify the bioaccessibility of nutrients and the formation of peptides, impacting on the real nutritional value of food. In this study we investigated the effect of nitrate/nitrite replacement with a combination of polyphenols, ascorbate, and nitrate-reducing microbial starter cultures on the bioaccessibility of fatty acids, the hydrolysis of proteins and the release of bioactive peptides after in vitro digestion. Moreover, digested salami formulations were investigated for their impacts on cell proliferation and genotoxicity in the human intestinal cellular model (HT-29 cell line). The results indicated that a replacement of synthetic nitrates/nitrites with natural additives can represent a promising strategy to develop innovative "clean label" salamis without negatively affecting their nutritional value.


Assuntos
Produtos da Carne , Nitrosaminas , Humanos , Nitratos/metabolismo , Sais , Nitritos/metabolismo , Carne/análise , Nutrientes , Ácidos Graxos
9.
Biochem Biophys Res Commun ; 529(1): 23-27, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32560814

RESUMO

BACKGROUND: Our previous reports demonstrated the prospects of a new series of imidazoles as a source of alternative anti-parasite treatments, thus warranting further studies that include toxicity profiling. OBJECTIVE: In this study, we evaluated three imidazoles: bis-imidazole (compound 1), phenyl-substituted 1H-imidazole (compound 2), and thiopene-imidazole (compound 3) for cellular toxicity and possible mechanisms. METHODS: The three (3) compounds were assessed for in vitro cytotoxic action. Additionally, we probed likely mechanistic actions of these imidazoles. Findings showed dose-dependent cellular toxicity by these imidazoles. RESULTS: In the presence of antioxidant (Trolox), cytotoxicity was improved for compounds 2 and 3 but not for compound 1. Meantime, compound 7 promoted reactive oxygen species (ROS) production, which was abated in the presence of a standard antioxidant (Trolox). Additionally, the three (3) imidazoles impaired mitochondrial membrane potential (MMP). While MMP was not restored after treatment removal, the addition of antioxidant (Trolox) improved MMP for compounds 2 and 3 treatment. Additionally, compound 1 elevated expression of hypoxia-inducing factor 1-alpha (HIF-1α). This may not be unconnected with the capacity of compound 1 to cause oxidative stress. CONCLUSION: We show evidence that supports the cytotoxic action of imidazoles involves likely impairment to redox balance and mitochondrial membrane potential. The findings help our understanding of the mechanistic action of these imidazoles in living cells, and altogether may boost their prospects as new and alternative anti-protozoans.


Assuntos
Antiparasitários/toxicidade , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imidazóis/toxicidade , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Antiparasitários/química , Antiparasitários/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Imidazóis/química , Imidazóis/farmacologia , Oxirredução/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade
10.
Respir Res ; 21(1): 305, 2020 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-33213456

RESUMO

BACKGROUND: Exposure to electronic-cigarette (e-cig) aerosols induces potentially fatal e-cig or vaping-associated lung injury (EVALI). The cellular and molecular mechanisms underlying these effects, however, are unknown. We used an air-liquid interface (ALI) in vitro model to determine the influence of two design characteristics of third-generation tank-style e-cig devices-resistance and voltage-on (1) e-cig aerosol composition and (2) cellular toxicity. METHODS: Human bronchial epithelial cells (H292) were exposed to either butter-flavored or cinnamon-flavored e-cig aerosols at the ALI in a Vitrocell exposure system connected to a third-generation e-cig device. Exposures were conducted following a standard vaping topography profile for 2 h per day, for 1 or 3 consecutive days. 24 h after ALI exposures cellular and molecular outcomes were assessed. RESULTS: We found that butter-flavored e-cig aerosol produced under 'sub-ohm' conditions (< 0.5 Ω) contains high levels of carbonyls (7-15 µg/puff), including formaldehyde, acetaldehyde and acrolein. E-cig aerosol produced under regular vaping conditions (resistance > 1 Ω and voltage > 4.5 V), contains lower carbonyl levels (< 2 µg/puff). We also found that the levels of carbonyls produced in the cinnamon-flavored e-cig aerosols were much lower than that of the butter-flavored aerosols. H292 cells exposed to butter-flavored or cinnamon-flavored e-cig aerosol at the ALI under 'sub-ohm' conditions for 1 or 3 days displayed significant cytotoxicity, decreased tight junction integrity, increased reactive oxygen species production, and dysregulated gene expression related to biotransformation, inflammation and oxidative stress (OS). Additionally, the cinnamon-flavored e-cig aerosol induced pro-oxidant effects as evidenced by increases in 8-hydroxy-2-deoxyguanosine protein levels. Moreover, we confirmed the involvement of OS as a toxicity process for cinnamon-flavored e-cig aerosol by pre-treating the cells with N-acetyl cysteine (NAC), an antioxidant that prevented the cells from the OS-mediated damage induced by the e-cig aerosol. CONCLUSION: The production of high levels of carbonyls may be flavor specific. Overall, inhaling e-cig aerosols produced under 'sub-ohm' conditions is detrimental to lung epithelial cells, potentially via mechanisms associated with OS. This information could help policymakers take the necessary steps to prevent the manufacturing of sub-ohm atomizers for e-cig devices.


Assuntos
Brônquios/efeitos dos fármacos , Citotoxinas/toxicidade , Sistemas Eletrônicos de Liberação de Nicotina , Aromatizantes/toxicidade , Mucosa Respiratória/efeitos dos fármacos , Vaping/efeitos adversos , Aerossóis , Antioxidantes/farmacologia , Brônquios/citologia , Brônquios/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Expressão Gênica , Humanos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Mucosa Respiratória/metabolismo
11.
Bioorg Med Chem Lett ; 29(14): 1831-1835, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31097376

RESUMO

In this investigation, a series of 1-phenyl-3-(5-(pyrimidin-4-ylthio)-1,3,4- thiadiazol-2-yl)urea receptor tyrosine kinase inhibitors were synthesized by a simple and efficient structure-based design. Structure-activity relationship (SAR) analysis of these compounds based on cellular assays led to the discovery of a number of compounds that showed potent activity against human chronic myeloid leukemia (CML) cell line K562, but very weak or no cellular toxicity through monitoring the growth kinetics of K562 cell during a period of 72 h using the real-time live-cell imaging. Among these compounds, 1-(5-((6-((3-morpholinopropyl) amino)pyrimidin-4-yl)thio)-1,3,4-thiadiazol-2-yl)-3-(4-(trifluoromethyl)phenyl)urea (7) exhibited the least cellular toxicity and better biological activity in cellular assays (K562, IC50: 0.038 µM). Compound 7 also displayed very good induced-apoptosis effect for human CML cell line K562 and exerted its effect via a significantly reduced protein phosphorylation of PI3K/Akt signal pathway by Human phospho-kinase array analysis. In vitro results indicate that 1-phenyl-3-(5-(pyrimidin-4-ylthio)-1,3,4- thiadiazol-2-yl)urea derivatives are lead molecules for further development as treatment of chronic myeloid leukemia and cancer.


Assuntos
Antineoplásicos/uso terapêutico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Antineoplásicos/farmacologia , Desenho de Fármacos , Humanos , Transdução de Sinais , Relação Estrutura-Atividade
12.
Arch Toxicol ; 92(7): 2259-2271, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29556720

RESUMO

Increasing incidence of multidrug-resistant bacteria presents an imminent risk to global health. Polymyxins are 'last-resort' antibiotics against Gram-negative 'superbugs'; however, nephrotoxicity remains a key impediment in their clinical use. Molecular mechanisms underlying this nephrotoxicity remain poorly defined. Here, we examined the pathways which led to polymyxin B induced cell death in vitro and in vivo. Human proximal tubular cells were treated with polymyxin B (12.5-100 µM) for up to 24 h and showed a significant increase in micronuclei frequency, as well as abnormal mitotic events (over 40% in treated cells, p < 0.05). Time-course studies were performed using a mouse nephrotoxicity model (cumulative 72 mg/kg). Kidneys were collected over 48 h and investigated for histopathology and DNA damage. Notable increases in γH2AX foci (indicative of double-stranded breaks) were observed in both cell culture (up to ~ 44% cells with 5+ foci at 24 h, p < 0.05) and mice treated with polymyxin B (up to ~ 25%, p < 0.05). Consistent with these results, in vitro assays showed high binding affinity of polymyxin B to DNA. Together, our results indicate that polymyxin B nephrotoxicity is associated with DNA damage, leading to chromosome missegregation and genome instability. This novel mechanistic information may lead to new strategies to overcome the nephrotoxicity of this important last-line class of antibiotics.


Assuntos
Antibacterianos/toxicidade , Dano ao DNA , Reparo do DNA , Rim/efeitos dos fármacos , Polimixina B/toxicidade , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Feminino , Instabilidade Genômica/efeitos dos fármacos , Humanos , Rim/patologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Camundongos , Necrose
13.
Pestic Biochem Physiol ; 149: 8-19, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30033020

RESUMO

In vitro experiments were performed to ascertain the impact of kitazin, hexaconazole, metalaxyl and carbendazim on growth behaviour, enzymatic profile, ultrastructure, cell permeability and bioactive molecules of phosphate-solubilizing bacterium. Strain BC8 isolated from Brassica oleracea rhizosphere was characterized and identified as Bacillus subtilis by 16S rDNA sequencing (Accession no. MG028650) technique. Strain BC8 was unambiguously chosen due to its high tolerance capability to various fungicides and substantial production of plant growth regulators. The biomarker enzymatic assays (lipid peroxidation, lactate dehydrogenase) and oxidative stress (catalase) induced by fungicides exhibited significant (p < 0.05) toxicity of fungicides toward strain BC8. Fungicides caused the cellular/ultrastructural damage and reduced the viability of strain BC8 as clearly revealed under scanning (SEM), high resolution transmission (HR-TEM) and confocal laser scanning (CLSM) microscopy. As the concentration of fungicides increased, a gradual drop in the plant growth promoting traits of B. subtilis strain BC8 was observed. Kitazin at 2400 µg mL-1, hexaconazole at 1500 µg mL-1, metalaxyl at 1200 µg mL-1 and carbendazim at 1200 µg mL-1decreased the IAA production by 35 (48.3 µg mL-1), 27 (51.5 µg mL-1), 39 (43.6 µg mL-1) and 47% (37.3 µg mL-1), respectively, over control (71.3 µg mL-1), while, α-ketobutyrate was declined by 51 (29.6), 56 (26.2), 61 (22.8) and 68 (19)%, respectively, over untreated control (59.9 mg protein-1 h-1). Also, with increase in the concentration of fungicides there was a significant decrease in plant nutrient (P); the maximum being (19.6 µg mL-1) observed at 1500 µg mL-1 hexaconazole with consequent drop in pH (from pH 6.4 to 4.2). The current findings clearly suggest that despite injury, B. subtilis maintained secreting active biomolecules and this property makes this organism truly indispensable for enhancing crop production under fungicide stressed conditions.


Assuntos
Bacillus subtilis/efeitos dos fármacos , Enzimas/metabolismo , Fungicidas Industriais/farmacologia , Reguladores de Crescimento de Plantas/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/crescimento & desenvolvimento , Bacillus subtilis/ultraestrutura , Brassica/microbiologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular , DNA Ribossômico/genética , Testes de Sensibilidade Microbiana , Microscopia Confocal , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Estresse Oxidativo , RNA Bacteriano/genética , RNA Ribossômico 16S/genética
14.
Cytotherapy ; 19(12): 1426-1437, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29037943

RESUMO

BACKGROUND AIMS: Light chain (AL) amyloidosis is a protein misfolding disease characterized by extracellular deposition of immunoglobulin light chains (LC) as amyloid fibrils. Patients with LC amyloid involvement of the heart have the worst morbidity and mortality. Current treatments target the plasma cells to reduce further production of amyloid proteins. There is dire need to understand the mechanisms of cardiac tissue damage from amyloid to develop novel therapies. We recently reported that LC soluble and fibrillar species cause apoptosis and inhibit cell growth in human cardiomyocytes. Mesenchymal stromal cells (MSCs) can promote wound healing and tissue remodeling. The objective of this study was to evaluate MSCs to protect cardiomyocytes affected by AL amyloid fibrils. METHODS: We used live cell imaging and proteomics to analyze the effect of MSCs in the growth arrest caused by AL amyloid fibrils. RESULTS: We evaluated the growth of human cardiomyocytes (RFP-AC16 cells) in the presence of cytotoxic LC amyloid fibrils. MSCs reversed the cell growth arrest caused by LC fibrils. We also demonstrated that this effect requires cell contact and may be mediated through paracrine factors modulating cell adhesion and extracellular matrix remodeling. To our knowledge, this is the first report of MSC protection of human cardiomyocytes in amyloid disease. CONCLUSIONS: This important proof of concept study will inform future rational development of MSC therapy in cardiac LC amyloid.


Assuntos
Amiloide/toxicidade , Amiloidose de Cadeia Leve de Imunoglobulina/patologia , Células-Tronco Mesenquimais/citologia , Miócitos Cardíacos/patologia , Amiloide/metabolismo , Apoptose , Células Cultivadas , Técnicas de Cocultura , Humanos , Cadeias Leves de Imunoglobulina/metabolismo , Amiloidose de Cadeia Leve de Imunoglobulina/terapia , Células-Tronco Mesenquimais/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo
15.
Environ Toxicol ; 32(6): 1836-1843, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28206703

RESUMO

Etoposide, a topoisomerase II inhibitor, has been widely used as a clinical anticancer drug to treat diverse cancer patients. Since not only rapidly dividing cancer cells but also the cells of normal human tissues and every living organism in environmental ecosystems have topoisomerases, it is crucial to study the toxicity of etoposide in other organisms in addition to cancer cells. In this study, we evaluated the toxicity of etoposide in both a soil nematode, Caenorhabditis elegans, and 3T3-L1 normal murine cells. Etoposide significantly retarded the growth, egg laying, and hatching in C. elegans. Etoposide also affected the reproductive gonad tissue, decreased the number of germ cells and induced abnormally enlarged nuclei in C. elegans. In addition, etoposide inhibited 3T3-L1 cell proliferation, with IC50 values of 37.8 ± 7.3 and 9.8 ± 1.8 µM after 24 and 48 hours of treatment, respectively, via the induction of cell cycle arrest at the G2/M phase and apoptotic cell death. Etoposide also induced nuclear enlargement in 3T3-L1 normal murine cells. The reproductive toxicity and abnormal nuclear morphological changes seemed to correlate with the adverse effects of etoposide. We suggest that these experimental platforms, i.e., the toxicological evaluation of both nematodes and 3T3-L1 cells, may be useful to study the mechanisms underlying the side effects of chemicals, including topoisomerase inhibitors.


Assuntos
Antineoplásicos/toxicidade , Caenorhabditis elegans/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Etoposídeo/toxicidade , Inibidores da Topoisomerase II/toxicidade , Células 3T3-L1 , Animais , Apoptose/efeitos dos fármacos , Técnicas de Cultura de Células , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Núcleo Celular/patologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Camundongos , Microscopia de Fluorescência , Reprodução/efeitos dos fármacos
16.
Int J Mol Sci ; 17(10)2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27775595

RESUMO

Homocysteine (Hcy) is a sulfur-containing non-proteinogenic amino acid derived in methionine metabolism. The increased level of Hcy in plasma, hyperhomocysteinemia, is considered to be an independent risk factor for cardio and cerebrovascular diseases. However, it is still not clear if Hcy is a marker or a causative agent of diseases. More and more research data suggest that Hcy is an important indicator for overall health status. This review represents the current understanding of molecular mechanism of Hcy metabolism and its link to hyperhomocysteinemia-related pathologies in humans. The aberrant Hcy metabolism could lead to the redox imbalance and oxidative stress resulting in elevated protein, nucleic acid and carbohydrate oxidation and lipoperoxidation, products known to be involved in cytotoxicity. Additionally, we examine the role of Hcy in thiolation of proteins, which results in their molecular and functional modifications. We also highlight the relationship between the imbalance in Hcy metabolism and pathogenesis of diseases, such as cardiovascular diseases, neurological and psychiatric disorders, chronic kidney disease, bone tissue damages, gastrointestinal disorders, cancer, and congenital defects.


Assuntos
Nível de Saúde , Homocisteína/metabolismo , Hiper-Homocisteinemia/metabolismo , Estresse Oxidativo/fisiologia , Doenças Ósseas/patologia , Doenças Cardiovasculares/patologia , Transtornos Cerebrovasculares/patologia , Gastroenteropatias/patologia , Homocisteína/sangue , Humanos , Nefropatias/patologia , Neoplasias/patologia , Oxirredução , Fatores de Risco
17.
Environ Toxicol ; 29(8): 908-15, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23047665

RESUMO

Increases in asthma worldwide have been associated epidemiologically with expanding urban air pollution. The mechanistic relationship between airway hyper-responsiveness, inflammation, and ambient airborne triggers remains ambiguous. Acrolein, a ubiquitous aldehyde pollutant, is a product of incomplete combustion reactions. Acrolein is abundant in cigarette smoke, effluent from industrial smokestacks, diesel exhaust, and even hot oil cooking vapors. Acrolein is a potent airway irritant and can induce airway hyper-responsiveness and inflammation in the lungs of animal models. In the present study, we utilized the mast cell analog, RBL-2H3, to interrogate the responses of cells relevant to airway inflammation and allergic responses as a model for the induction of asthma-like conditions upon exposure to acrolein. We hypothesized that acrolein would induce oxidative stress and degranulation in airway mast cells. Our results indicate that acrolein at 1 ppm initiated degranulation and promoted the generation of reactive oxygen species (ROS). Introduction of antioxidants to the system significantly reduced both ROS generation and degranulation. At higher levels of exposure (above 100 ppm), RBL-2H3 cells displayed signs of severe toxicity. This experimental data indicates acrolein can induce an allergic inflammation in mast cell lines, and the initiation of degranulation was moderated by the application of antioxidants.


Assuntos
Acroleína/toxicidade , Poluentes Atmosféricos/toxicidade , Degranulação Celular/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Citocinas/metabolismo , Mastócitos/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo
18.
Chemosphere ; 357: 142090, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38648983

RESUMO

The growing utilization of rare earth elements (REEs) in industrial and technological applications has captured global interest, leading to the development of high-performance technologies in medical diagnosis, agriculture, and other electronic industries. This accelerated utilization has also raised human exposure levels, resulting in both favourable and unfavourable impacts. However, the effects of REEs are dependent on their concentration and molecular species. Therefore, scientific interest has increased in investigating the molecular interactions of REEs with biomolecules. In this current review, particular attention was paid to the molecular mechanism of interactions of Lanthanum (La), Cerium (Ce), and Gadolinium (Gd) with biomolecules, and the biological consequences were broadly interpreted. The review involved gathering and evaluating a vast scientific collection which primarily focused on the impact associated with REEs, ranging from earlier reports to recent discoveries, including studies in human and animal models. Thus, understanding the molecular interactions of each element with biomolecules will be highly beneficial in elucidating the consequences of REEs accumulation in the living organisms.


Assuntos
Lantânio , Metais Terras Raras , Metais Terras Raras/química , Humanos , Lantânio/química , Animais , Cério/química , Gadolínio/química , Substâncias Macromoleculares/química
19.
Toxics ; 12(6)2024 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-38922068

RESUMO

Cadmium (Cd), a prevalent environmental contaminant, exerts widespread toxic effects on human health through various biochemical and molecular mechanisms. This review encapsulates the primary pathways through which Cd inflicts damage, including oxidative stress induction, disruption of Ca2+ signaling, interference with cellular signaling pathways, and epigenetic modifications. By detailing the absorption, distribution, metabolism, and excretion (ADME) of Cd, alongside its interactions with cellular components such as mitochondria and DNA, this paper highlights the extensive damage caused by Cd2+ at the cellular and tissue levels. The role of Cd in inducing oxidative stress-a pivotal mechanism behind its toxicity-is discussed with emphasis on how it disrupts the balance between oxidants and antioxidants, leading to cellular damage and apoptosis. Additionally, the review covers Cd's impact on signaling pathways like Mitogen-Activated Protein Kinase (MAPK), Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB), and Tumor Protein 53 (p53) pathways, illustrating how its interference with these pathways contributes to pathological conditions and carcinogenesis. The epigenetic effects of Cd, including DNA methylation and histone modifications, are also explored to explain its long-term impact on gene expression and disease manifestation. This comprehensive analysis not only elucidates the mechanisms of Cd toxicity but also underscores the critical need for enhanced strategies to mitigate its public health implications.

20.
Toxicol Lett ; 398: 28-37, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38851367

RESUMO

This work investigated the influence of surface chirality on cellular internalization, cytotoxicity, and tissue distribution of silver nanoparticles (AgNPs). D-cysteine and L-cysteine are chiral forms of the amino acid cysteine. These enantiomers exhibit distinct spatial arrangements, with D-cysteine having a different configuration from L-cysteine. This structural dissimilarity can lead to variations in how these forms interact with biological systems, potentially impacting their cytotoxic responses. Four distinct types of AgNPs were synthesized, each possessing a unique surface coating: pristine AgNPs (pAgNPs), L-cysteine coated AgNPs (AgNPs@L-Cys), D-cysteine coated AgNPs (AgNPs@D-Cys), and racemic AgNPs coated with both L-Cys and D-Cys (AgNPs@L/D-Cys). We found chiral-dependent cytotoxicity of AgNPs on J774A.1 cells. Specifically, AgNPs@L-Cys exhibited the highest toxicity, and AgNPs@D-Cys exhibited the lowest toxicity. Meanwhile, the cellular uptake of the AgNPs correlated nicely with their cytotoxicity, with AgNPs@L-Cys being internalized to the greatest extent while AgNPs@D-Cys displays the least internalization. Scavenger receptors and clathrin predominantly mediate the cellular internalization of these AgNPs. Strikingly, the dissimilar cellular internalization and cytotoxicity of AgNPs with different chirality were eliminated upon protein corona coverage. Notably, following intravenous injection in mice, these four types of AgNPs showed similar patterns among various organs due to the inevitable protein adsorption in the bloodstream. These findings underscored the pivotal role of surface chirality in governing the biological interactions and toxicity of AgNPs.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa