Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Omega ; 8(12): 10968-10979, 2023 Mar 28.
Article in English | MEDLINE | ID: mdl-37008095

ABSTRACT

The synthesis of fungicides in eco-friendly and cost-effective ways is significantly essential for agriculture. Plant pathogenic fungi cause many ecological and economic issues worldwide, which must be treated with effective fungicides. Here, this study proposes the biosynthesis of fungicides, which combines copper and Cu2O nanoparticles (Cu/Cu2O) synthesized using durian shell (DS) extract as a reducing agent in aqueous media. Sugar and polyphenol compounds contained in DS, as the main phytochemicals acting in the reduction procedure, were extracted under different temperatures and duration conditions to obtain the highest yields. We confirmed the extraction process performed at 70 °C for 60 min to be the most effective in extracting sugar (6.1 g/L) and polyphenols (22.7 mg/L). We determined the suitable conditions for Cu/Cu2O synthesis using a DS extract as a reducing agent for a synthesis time of 90 min, a volume ratio of DR extract/Cu2+ of 15:35, an initial pH solution of 10, a synthesis temperature of 70 °C, and a CuSO4 concentration of 10 mM. The characterization results of as-prepared Cu/Cu2O NP showed a highly crystalline structure of Cu2O and Cu with sizes estimated in the range of 40-25 nm and 25-30 nm, respectively. Through in vitro experiments, the antifungal efficacy of Cu/Cu2O against Corynespora cassiicola and Neoscytalidium dimidiatum was investigated by the inhibition zone. The green-synthesized Cu/Cu2O nanocomposites, which are potential antifungals against plant pathogens, exhibited excellent antifungal efficacy against both Corynespora cassiicola (MIC = 0.25 g/L, the diameter of the inhibition zone was 22.00 ± 0.52 mm) and Neoscytalidium dimidiatum (MIC = 0.0625 g/L, the diameter of the inhibition zone was 18.00 ± 0.58 mm). Cu/Cu2O nanocomosites prepared in this study could be a valuable suggestion for the control of plant pathogenic fungi affecting crop species globally.

2.
PLoS One ; 6(8): e23660, 2011.
Article in English | MEDLINE | ID: mdl-21858193

ABSTRACT

Inflammation and oxidative stress have been implicated in the pathophysiology of Parkinson's disease (PD) and inhibition of microglial activation attenuates degeneration of dopaminergic (DA) neurons in animal models of PD. Loss-of-function mutations in the parkin gene, which encodes an E3 ubiquitin ligase, cause autosomal recessive parkinsonism. While most studies on Parkin have focused on its function in neurons, here we demonstrate that Parkin mRNA and protein is detectable in brain-resident microglia and peripheral macrophages. Using pharmacologic and genetic approaches, we found that Parkin levels are regulated by inflammatory signaling. Specifically, exposure to LPS or Tumor Necrosis Factor (TNF) induced a transient and dose-dependent decrease in Parkin mRNA and protein in microglia, macrophages and neuronal cells blockable by inhibitors of Nuclear Factor-Kappa B (NF-κB) signaling and not observed in MyD88-null cells. Moreover, using luciferase reporter assays, we identified an NF-κB response element in the mouse parkin promoter responsible for mediating the transcriptional repression, which was abrogated when the consensus sequence was mutated. Functionally, activated macrophages from Parkin-null mice displayed increased levels of TNF, IL-1ß, and iNOS mRNA compared to wild type macrophages but no difference in levels of Nrf2, HO-1, or NQO1. One implication of our findings is that chronic inflammatory conditions may reduce Parkin levels and phenocopy parkin loss-of-function mutations, thereby increasing the vulnerability for degeneration of the nigrostriatal pathway and development of PD.


Subject(s)
Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases/genetics , Animals , Binding Sites/drug effects , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Immunoblotting , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Models, Biological , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Neurons/drug effects , Neurons/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
3.
J Neuroinflammation ; 5: 14, 2008 May 12.
Article in English | MEDLINE | ID: mdl-18474101

ABSTRACT

BACKGROUND: Recent animal and human studies implicate chronic activation of microglia in the progressive loss of CNS neurons. The inflammatory mechanisms that have neurotoxic effects and contribute to neurodegeneration need to be elucidated and specifically targeted without interfering with the neuroprotective effects of glial activities. Synthetic triterpenoid analogs of oleanolic acid, such as methyl-2-cyano-3,12-dioxooleana-1,9-dien-28-oate (CDDO-Me, RTA 402) have potent anti-proliferative and differentiating effects on tumor cells, and anti-inflammatory activities on activated macrophages. We hypothesized that CDDO-Me may be able to suppress neurotoxic microglial activities while enhancing those that promote neuronal survival. Therefore, the aims of our study were to identify specific microglial activities modulated by CDDO-Me in vitro, and to determine the extent to which this modulation affords neuroprotection against inflammatory stimuli. METHODS: We tested the synthetic triterpenoid methyl-2-cyano-3,12-dioxooleana-1,9-dien-28-oate (CDDO-Me, RTA 402) in various in vitro assays using the murine BV2 microglia cell line, mouse primary microglia, or mouse primary peritoneal macrophages to investigate its effects on proliferation, inflammatory gene expression, cytokine secretion, and phagocytosis. The antioxidant and neuroprotective effects of CDDO-Me were also investigated in primary neuron/glia cultures from rat basal forebrain or ventral midbrain. RESULTS: We found that at low nanomolar concentrations, treatment of rat primary mesencephalon neuron/glia cultures with CDDO-Me resulted in attenuated LPS-, TNF- or fibrillar amyloid beta 1-42 (A beta 1-42) peptide-induced increases in reactive microglia and inflammatory gene expression without an overall effect on cell viability. In functional assays CDDO-Me blocked death in the dopaminergic neuron-like cell line MN9D induced by conditioned media (CM) of LPS-stimulated BV2 microglia, but did not block cell death induced by addition of TNF to MN9D cells, suggesting that dopaminergic neuroprotection by CDDO-Me involved inhibition of microglial-derived cytokine production and not direct inhibition of TNF-dependent pro-apoptotic pathways. Multiplexed immunoassays of CM from LPS-stimulated microglia confirmed that CDDO-Me-treated BV2 cells produced decreased levels of specific subsets of cytokines, in particular TNF. Lastly, CDDO-Me enhanced phagocytic activity of BV2 cells in a stimulus-specific manner but inhibited generation of reactive oxygen species (ROS) in mixed neuron/glia basal forebrain cultures and dopaminergic cells. CONCLUSION: The neuroimmune modulatory properties of CDDO-Me indicate that this potent antioxidant and anti-inflammatory compound may have therapeutic potential to modify the course of neurodegenerative diseases characterized by chronic neuroinflammation and amyloid deposition. The extent to which synthetic triterpenoids afford therapeutic benefit in animal models of Parkinson's and Alzheimer's disease deserves further investigation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Dopamine/metabolism , Microglia/drug effects , Neuroprotective Agents/pharmacology , Oleanolic Acid/analogs & derivatives , Tumor Necrosis Factor-alpha/biosynthesis , Amyloid beta-Peptides/metabolism , Animals , Cell Division/drug effects , Cells, Cultured/drug effects , Cytokines/metabolism , Drug Evaluation, Preclinical , Gene Expression Profiling , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Mesencephalon/cytology , Mice , Neurons/drug effects , Oleanolic Acid/pharmacology , Peptide Fragments/metabolism , Phagocytosis/drug effects , Prosencephalon/cytology , Rats , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/genetics
4.
Front Biosci ; 13: 709-17, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17981581

ABSTRACT

The inflammatory response in the brain associated with most chronic neurodegenerative diseases is termed neuroinflammation. Neuropathological and neuroradiological studies indicate that in certain neurodegenerative disorders neuroinflammation may be detectable years before significant loss of neurons occurs. In this review, we discuss the evidence from human studies and experimental models that implicate neuroinflammatory processes in the progressive neurodegeneration of the nigrostriatal pathway, the hallmark of Parkinson's Disease (PD). We discuss the neurotoxic role of microglia-derived inflammatory mediators which are suspected to hasten the death of nigral dopaminergic neurons, in particular the pro-inflammatory cytokine Tumor Necrosis Factor (TNF) and its downstream signaling pathways. We also entertain the possibility that chronic microglia activation links proteinopathies to neurodegeneration. The rationale for current and future use of anti-inflammatory approaches to protect vulnerable neuronal populations in PD is also reviewed.


Subject(s)
Inflammation , Parkinson Disease/immunology , Parkinson Disease/therapy , Animals , Anti-Inflammatory Agents/pharmacology , Antiparkinson Agents/therapeutic use , Brain/pathology , Disease Models, Animal , Humans , Models, Biological , Nerve Degeneration/drug therapy , Neurons/metabolism , Oxidative Stress , Parkinson Disease/epidemiology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...