Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(5)2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38473763

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the gradual loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc), resulting in reduced dopamine levels in the striatum and eventual onset of motor symptoms. Linalool (3,7-dimethyl-1,6-octadien-3-ol) is a monoterpene in aromatic plants exhibiting antioxidant, antidepressant, and anti-anxiety properties. The objective of this study is to evaluate the neuroprotective impacts of linalool on dopaminergic SH-SY5Y cells, primary mesencephalic and cortical neurons treated with 1-methyl-4-phenylpyridinium ion (MPP+), as well as in PD-like mice induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Cell viability, α-tubulin staining, western blotting, immunohistochemistry and behavioral experiments were performed. In MPP+-treated SH-SY5Y cells, linalool increased cell viability, reduced neurite retraction, enhanced antioxidant defense by downregulation of apoptosis signaling (B-cell lymphoma 2 (Bcl-2), cleaved caspase-3 and poly ADP-ribose polymerase (PARP)) and phagocyte NADPH oxidase (gp91phox), as well as upregulation of neurotrophic signaling (brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF)) and nuclear factor-erythroid 2 related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. In MPP+-treated primary mesencephalic neurons, linalool enhanced the expressions of tyrosine hydroxylase (TH), Sirtuin 1 (SirT1), and parkin. In MPP+-treated primary cortical neurons, linalool upregulated protein expression of SirT1, γ-Aminobutyric acid type A-α1 (GABAA-α1), and γ-Aminobutyric acid type B (GABAB). In PD-like mice, linalool attenuated the loss of dopamine neurons in SNpc. Linalool improved the motor and nonmotor behavioral deficits and muscle strength of PD-like mice. These findings suggest that linalool potentially protects dopaminergic neurons and improves the impairment symptoms of PD.


Subject(s)
Acyclic Monoterpenes , Neuroblastoma , Neuroprotective Agents , Parkinson Disease , Humans , Mice , Animals , Parkinson Disease/metabolism , Dopaminergic Neurons/metabolism , Antioxidants/metabolism , Odorants , Sirtuin 1/metabolism , Neuroprotective Agents/pharmacology , Neuroblastoma/metabolism , 1-Methyl-4-phenylpyridinium , Muscle Strength , Models, Theoretical , gamma-Aminobutyric Acid/metabolism
2.
J Food Drug Anal ; 31(1): 116-136, 2023 03 15.
Article in English | MEDLINE | ID: mdl-37224559

ABSTRACT

Guilu Erxian Jiao (GEJ) is a commonly used nutritional supplement due to its rich content of amino acids. It is also a traditional herbal medicine for improving degenerative joint. This study aimed to investigate the effect and mechanism of GEJ water extract (GEJ-WE) on skeletal muscle in C2C12 myotubes and C57BL/6J mice. Analysis of GEJ-WE were performed by high-performance liquid chromatography fingerprinting with chemical standards. Protein expression, mRNA level, glycogen content, mitochondria activity and ATP level were evaluated by western blots, real-time PCR, PAS staining, MTT and ATP bioluminescence assay, respectively. Skeletal muscle strength was evaluated by grip strength. Skeletal muscle volume, mass and fiber types were evaluated by micro computed tomography, histological analysis and immunofluorescence staining, respectively. Motor function was evaluated by rotarod performance and locomotor activity. In C2C12 myotubes, GEJ-WE significantly enhanced myogenic differentiation and myotube growth, protein synthesis signaling IGF-1/IGF-1R/IRS-1/Akt, Glut4 translocation, glycogen content, mitochondrial biogenesis signaling PGC-1α/NRF1/TFAM, mitochondrial activity and ATP production. However, IGF-1R antagonist AG1024 and PI3K inhibitor wortmannin reduced GEJ-WE-induced protein expression of MyHC, p-Akt, p-mTOR and p-GSK-3ß, Glut4 translocation and glycogen content. In C57BL/6J mice, GEJ-WE not only upregulated protein synthesis and mitochondrial biogenesis signaling, but it also increased muscle volume, relative muscle weight, cross-sectional area of myofibers, glycogen content and transition of fast-to-slow type fibers of skeletal muscles. Moreover, GEJ-WE enhanced grip strength and motor activity of mice. In conclusion, the upregulation of protein synthesis, myogenic differentiation, glucose homeostasis, mitochondrial biogenesis and slow-twitch fibers contributes to the mechanisms of GEJ-WE on the enhancement of skeletal muscle mass and motor function.


Subject(s)
Organelle Biogenesis , Phosphatidylinositol 3-Kinases , Animals , Mice , Mice, Inbred C57BL , Glycogen Synthase Kinase 3 beta , Proto-Oncogene Proteins c-akt , X-Ray Microtomography , Muscle, Skeletal , Homeostasis , Glucose , Adenosine Triphosphate
3.
Int J Mol Sci ; 23(21)2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36361718

ABSTRACT

Inflammation is a major cause of skeletal muscle atrophy in various diseases. 2-Hydroxy-4'-methoxychalcone (AN07) is a chalcone-based peroxisome-proliferator-activated receptor gamma (PPARγ) agonist with various effects, such as antiatherosclerosis, anti-inflammation, antioxidative stress, and neuroprotection. In this study, we examined the effects of AN07 on protein homeostasis pathway and mitochondrial function in inflammation-associated myotube atrophy induced by lipopolysaccharides (LPS). We found that AN07 significantly attenuated NF-κB activation, inflammatory factors (TNF-α, IL-1ß, COX-2, and PGE2), Nox4 expression, and reactive oxygen species levels in LPS-treated C2C12 myotubes. Moreover, AN07 increased SOD2 expression and improved mitochondrial function, including mitochondrial membrane potential and mitochondrial oxygen consumption rate. We also demonstrated that AN07 attenuated LPS-induced reduction of myotube diameter, MyHC expression, and IGF-1/IGF-1R/p-Akt-mediated protein synthesis signaling. Additionally, AN07 downregulated LPS-induced autophagy-lysosomal protein degradation molecules (LC3-II/LC3-I and degraded p62) and ubiquitin-proteasome protein degradation molecules (n-FoxO1a/MuRF1/atrogin-1). However, the regulatory effects of AN07 on protein synthesis and degradation signaling were inhibited by the IGF-1R inhibitor AG1024 and the PI3K inhibitor wortmannin. In addition, the PPARγ antagonist GW9662 attenuated the effects of AN07 against LPS-induced inflammation, oxidation, and protein catabolism. In conclusion, our findings suggest that AN07 possesses protective effects on inflammation-induced myotube atrophy and mitochondrial dysfunction.


Subject(s)
Chalcone , Chalcones , Humans , Lipopolysaccharides/adverse effects , Phosphatidylinositol 3-Kinases/metabolism , PPAR gamma/metabolism , Chalcones/pharmacology , Chalcones/metabolism , Chalcone/metabolism , Muscular Atrophy/chemically induced , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscle Fibers, Skeletal/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism
4.
Biomolecules ; 12(5)2022 04 30.
Article in English | MEDLINE | ID: mdl-35625589

ABSTRACT

Hyperbaric oxygen therapy (HBOT) has been suggested as a potential adjunctive therapy for Parkinson's disease (PD). PD is a neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The aim of this study was to investigate the protective mechanisms of HBOT on neurons and motor function in a 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD and 1-methyl-4-phenylpyridinium (MPP+)-mediated neurotoxicity in SH-SY5Y cells on the potential protective capability. In vivo: male C57BL/6 mice were randomly divided into three groups: control, MPTP group and MPTP+HBOT group. The MPTP-treated mice were intraperitoneally received MPTP (20 mg/kg) four times at 2 h intervals within a day. The day after MPTP treatment, MPTP+HBOT mice were exposed to hyperbaric oxygen at 2.5 atmosphere absolute (ATA) with 100% oxygen for 1 h once daily for 7 consecutive days. In vitro: retinoic acid (RA)-differentiated SH-SY5Y cells were treated with MPP+ for 1 h followed by hyperbaric oxygen at 2.5 ATA with 100% oxygen for 1 h. The results showed that MPTP induced a significant loss in tyrosine hydroxylase (TH)-positive neurons in the SNpc of mice. HBOT treatment significantly increased the number of TH-positive neurons, with enhanced neurotrophic factor BDNF, decreased apoptotic signaling and attenuated inflammatory mediators in the midbrain of MPTP-treated mice. In addition, MPTP treatment decreased the locomotor activity and grip strength of mice, and these effects were shown to improve after HBOT treatment. Furthermore, MPTP decreased mitochondrial biogenesis signaling (SIRT-1, PGC-1α and TFAM), as well as mitochondrial marker VDAC expression, while HBOT treatment was shown to upregulate protein expression. In cell experiments, MPP+ reduced neurite length, while HBOT treatment attenuated neurite retraction. Conclusions: the effects of HBOT in MPTP-treated mice might come from promoting mitochondrial biogenesis, decreasing apoptotic signaling and attenuating inflammatory mediators in the midbrain, suggesting its potential benefits in PD treatment.


Subject(s)
Hyperbaric Oxygenation , MPTP Poisoning , Neurodegenerative Diseases , Parkinson Disease , Sirtuins , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Dopaminergic Neurons/metabolism , Inflammation Mediators/metabolism , MPTP Poisoning/metabolism , MPTP Poisoning/therapy , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Organelle Biogenesis , Oxygen/metabolism , Parkinson Disease/metabolism , Parkinson Disease/therapy , Sirtuins/metabolism
5.
Respir Res ; 22(1): 159, 2021 May 22.
Article in English | MEDLINE | ID: mdl-34022899

ABSTRACT

BACKGROUND: Patients in intensive care units (ICUs) often received broad-spectrum antibiotic treatment and Acinetobacter baumannii (A.b.) and Pseudomonas aeruginosa (P.a.) were the most common pathogens causing ventilator-associated pneumonia (VAP). This study aimed to examine the effects and mechanism of mechanical ventilation (MV) on A.b.-induced lung injury and the involvement of alveolar macrophages (AMs). METHODS: C57BL/6 wild-type (WT) and c-Jun N-terminal kinase knockout (JNK1-/-) mice received MV for 3 h at 2 days after nasal instillation of A.b., P.a. (1 × 106 colony-forming unit, CFU), or normal saline. RESULTS: Intranasal instillation of 106 CFU A.b. in C57BL/6 mice induced a significant increase in total cells and protein levels in the bronchoalveolar lavage fluid (BALF) and neutrophil infiltration in the lungs. MV after A.b. instillation increases neutrophil infiltration, interleukin (IL)-6 and vascular cell adhesion molecule (VCAM) mRNA expression in the lungs and total cells, IL-6 levels, and nitrite levels in the BALF. The killing activity of AMs against A.b. was lower than against P.a. The diminished killing activity was parallel with decreased tumor necrosis factor-α production by AMs compared with A.b. Inducible nitric oxide synthase inhibitor, S-methylisothiourea, decreased the total cell number in BALF on mice receiving A.b. instillation and ventilation. Moreover, MV decreased the A.b. and P.a. killing activity of AMs. MV after A.b. instillation induced less total cells in the BALF and nitrite production in the serum of JNK1-/- mice than those of WT mice. CONCLUSION: A.b. is potent in inducing neutrophil infiltration in the lungs and total protein in the BALF. MV enhances A.b.-induced lung injury through an increase in the expression of VCAM and IL-6 levels in the BALF and a decrease in the bacteria-killing activity of AMs. A lower inflammation level in JNK1-/- mice indicates that A.b.-induced VAP causes lung injury through JNK signaling pathway in the lungs.


Subject(s)
Acinetobacter Infections/enzymology , Acinetobacter baumannii/pathogenicity , Lung/enzymology , Mitogen-Activated Protein Kinase 8/metabolism , Pneumonia, Ventilator-Associated/enzymology , Respiration, Artificial/adverse effects , Ventilator-Induced Lung Injury/enzymology , Acinetobacter Infections/microbiology , Acinetobacter Infections/pathology , Animals , Cells, Cultured , Disease Models, Animal , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/microbiology , Lung/pathology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/microbiology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Neutrophil Infiltration , Nitric Oxide Synthase Type II/metabolism , Pneumonia, Ventilator-Associated/microbiology , Pneumonia, Ventilator-Associated/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Ventilator-Induced Lung Injury/microbiology , Ventilator-Induced Lung Injury/pathology
6.
Br J Pharmacol ; 178(15): 2998-3016, 2021 08.
Article in English | MEDLINE | ID: mdl-33788266

ABSTRACT

BACKGROUND AND PURPOSE: Increasing evidence suggests systemic inflammation-caused skeletal muscle atrophy as a major clinical feature of cachexia. Triptolide obtained from Tripterygium wilfordii Hook F possesses potent anti-inflammatory and immunosuppressive effects. The present study aims to evaluate the protective effects and molecular mechanisms of triptolide on inflammation-induced skeletal muscle atrophy. EXPERIMENTAL APPROACH: The effects of triptolide on skeletal muscle atrophy were investigated in LPS-treated C2C12 myotubes and C57BL/6 mice. Protein expressions and mRNA levels were analysed by western blot and qPCR, respectively. Skeletal muscle mass, volume and strength were measured by histological analysis, micro-CT and grip strength, respectively. Locomotor activity was measured using the open field test. KEY RESULTS: Triptolide (10-100 fM) up-regulated protein synthesis signals (IGF-1/p-IGF-1R/IRS-1/p-Akt/p-mTOR) and down-regulated protein degradation signal atrogin-1 in C2C12 myotubes. In LPS (100 ng·ml-1 )-treated C2C12 myotubes, triptolide up-regulated MyHC, IGF-1, p-IGF-1R, IRS-1 and p-Akt. Triptolide also down-regulated ubiquitin-proteasome molecules (n-FoxO3a/atrogin-1/MuRF1), proteasome activity, autophagy-lysosomal molecules (LC3-II/LC3-I and Bnip3) and inflammatory mediators (NF-κB, Cox-2, NLRP3, IL-1ß and TNF-α). However, AG1024, an IGF-1R inhibitor, suppressed triptolide-mediated effects on MyHC, myotube diameter, MuRF1 and p62 in LPS-treated C2C12 myotubes. In LPS (1 mg·kg-1 , i.p.)-challenged mice, triptolide (5 and 20 µg·kg-1 ·day-1 , i.p.) decreased plasma TNF-α levels and it increased skeletal muscle volume, cross-sectional area of myofibers, weights of the gastrocnemius and tibialis anterior muscles, forelimb grip strength and locomotion. CONCLUSIONS AND IMPLICATIONS: These findings reveal that triptolide prevented LPS-induced inflammation and skeletal muscle atrophy and have implications for the discovery of novel agents for preventing muscle wasting.


Subject(s)
NF-kappa B , Tumor Necrosis Factor-alpha , Animals , Diterpenes , Epoxy Compounds , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal , Muscle, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/drug therapy , Muscular Atrophy/prevention & control , Phenanthrenes
7.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525680

ABSTRACT

Kynurenic acid (KYNA, 4-oxoquinoline-2-carboxylic acid), an intermediate of the tryptophan metabolism, has been recognized to exert different neuroactive actions; however, the need of how it or its aminoalkylated amide derivative N-(2-(dimethylamino)ethyl)-3-(morpholinomethyl)-4-oxo-1,4-dihydroquinoline-2-carboxamide (KYNA-A4) exerts any effects on ion currents in excitable cells remains largely unmet. In this study, the investigations of how KYNA and other structurally similar KYNA derivatives have any adjustments on different ionic currents in pituitary GH3 cells and hippocampal mHippoE-14 neurons were performed by patch-clamp technique. KYNA or KYNA-A4 increased the amplitude of M-type K+ current (IK(M)) and concomitantly enhanced the activation time course of the current. The EC50 value required for KYNA- or KYNA-A4 -stimulated IK(M) was yielded to be 18.1 or 6.4 µM, respectively. The presence of KYNA or KYNA-A4 shifted the relationship of normalized IK(M)-conductance versus membrane potential to more depolarized potential with no change in the gating charge of the current. The voltage-dependent hysteretic area of IK(M) elicited by long-lasting triangular ramp pulse was observed in GH3 cells and that was increased during exposure to KYNA or KYNA-A4. In cell-attached current recordings, addition of KYNA raised the open probability of M-type K+ channels, along with increased mean open time of the channel. Cell exposure to KYNA or KYNA-A4 mildly inhibited delayed-rectifying K+ current; however, neither erg-mediated K+ current, hyperpolarization-activated cation current, nor voltage-gated Na+ current in GH3 cells was changed by KYNA or KYNA-A4. Under whole-cell, current-clamp recordings, exposure to KYNA or KYNA-A4 diminished the frequency of spontaneous action potentials; moreover, their reduction in firing frequency was attenuated by linopirdine, yet not by iberiotoxin or apamin. In hippocampal mHippoE-14 neurons, the addition of KYNA also increased the IK(M) amplitude effectively. Taken together, the actions presented herein would be one of the noticeable mechanisms through which they modulate functional activities of excitable cells occurring in vivo.


Subject(s)
Hippocampus/physiology , KCNQ Potassium Channels/drug effects , Kynurenic Acid/pharmacology , Animals , Apamin/pharmacology , Cell Line , Hippocampus/drug effects , Hippocampus/metabolism , Indoles/pharmacology , Kynurenic Acid/chemistry , Membrane Potentials/drug effects , Mice , Patch-Clamp Techniques , Peptides/pharmacology , Pyridines/pharmacology , Rats
8.
Nutr Neurosci ; 24(1): 71-81, 2021 Jan.
Article in English | MEDLINE | ID: mdl-30900959

ABSTRACT

Objectives: Recent studies revealed the neuroprotective effects of naringenin (NGEN), a common dietary bioflavonoid contained in citrus fruits. However, there are limited data on its protection against methylglyoxal (MG), the most potent precursor of advanced glycation end-products. The present study was to investigate the protection of NGEN on MG-induced neurotoxicity and the involvement of insulin-like growth factor 1 receptor (IGF-1R) signaling. Methods: NSC34 motor neuron-like cells was used. Cell viability was measured by MTT assay. Protein expressions were analyzed by western blots. Morphological changes of neurites were observed by an inverted microscope. Reactive oxygen species (ROS) production and apoptotic cell numbers were measured by flow cytometer. Glutathione (GSH) level and superoxide dismutase (SOD) activity were measured by ELISA. Results: >NGEN attenuated ROS production and increased GSH level, SOD activity and nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear expression in MG-treated NSC34 cells. NGEN also increased neurite length and enhanced IGF-1R and p-Akt in MG-treated NSC34 cells. Furthermore, NGEN attenuated MG-induced apoptotic death accompanied with down-regulation of cleaved-poly (ADP-ribose) polymerase (PARP) and up-regulation of B-cell lymphoma-2 (Bcl-2). However, AG1024, an IGF-1R antagonist, attenuated the anti-oxidative and anti-apoptotic effects of NGEN in MG-treated cells. Discussion: The present results demonstrated that NGEN decreased neuronal apoptosis and improved antioxidant defense in MG-treated NSC34 cells. Moreover, IGF-1R-mediated antioxidant defense plays an important role in this protective mechanism. These findings suggest the potential benefits of NGEN on the prevention of MG-induced or diabetes/hyperglycemia-related neurotoxicity. In vivo studies are needed for further confirmation on NGEN-mediated neuroprotection.


Subject(s)
Antioxidants/administration & dosage , Apoptosis/drug effects , Flavanones/administration & dosage , Motor Neurons/drug effects , Neurites/drug effects , Neuroprotective Agents/administration & dosage , Pyruvaldehyde/toxicity , Receptor, IGF Type 1/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Mice , Motor Neurons/metabolism , Oxidative Stress/drug effects
9.
Phytomedicine ; 53: 96-106, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30668418

ABSTRACT

BACKGROUND: Liuwei dihuang (LWDH), a widely used traditional Chinese herbal medicine, has been noticed for its potential on the improvement of diabetic complications including diabetic nephropathy and diabetic encephalopathy. However, whether LWDH can improve the effects of diabetic skeletal muscle atrophy has not yet been reported. PURPOSE: The present study aimed to investigate the protective effects and mechanisms of the water extract of Liuwei dihuang (LWDH-WE) on skeletal muscle in cellular and animal models of diabetic muscle atrophy. STUDY DESIGN: The muscle protective effects of LWDH-WE on diabetic muscle atrophy and weakness were examined in methylglyoxal (MG)-treated C2C12 myotubes and streptozotocin (STZ)-treated C57BL/6 mice, respectively. METHODS: C2C12 myoblasts were differentiated by differentiation medium to form myotube structures. C2C12 myotubes were pre-treated with LWDH-WE 1 h before MG treatment. Diabetic mice were induced by single intraperitoneal injection of STZ (150 mg/kg) in C57BL/6 mice. Cell viability was determined by MTT and LDH assays. Protein expressions were detected by western blots. Morphological changes of cells were observed by an inverted microscope. Mitochondria membrane potential and reactive oxygen species (ROS) production were measured by flow cytometry. Muscle strength was evaluated by measuring grip strength of mice. RESULTS: In C2C12 myotubes, LWDH-WE attenuated MG-induced cellular death and oxidative damage accompanied with improving mitochondrial membrane potential, inhibiting NADPH oxidase (Nox) activation, and ROS production. Moreover, LWDH-WE could attenuate MG-induced atrophy of C2C12 myotubes accompanied with regulating protein synthesis (IGF-1R, Akt, mTOR) and protein degradation (FoxO3a, atrogin-1, MuRF-1) signals. In STZ-induced diabetic mice, LWDH-WE improved body weight and skeletal muscle mass of mice. LWDH-WE also enhanced muscle strength of STZ-induced diabetic mice. Furthermore, LWDH-WE enhanced the improvement of insulin on gastrocnemius muscle mass and grip strength in STZ-treated mice. CONCLUSION: LWDH-WE possesses skeletal muscle protection via reducing oxidative damage and regulating protein synthesis and degradation pathways in MG-induced atrophy of C2C12 myotubes. We also reveal the novel protection of LWDH-WE against STZ-induced reduction of muscle mass and muscle strength in mice.


Subject(s)
Diabetes Mellitus, Experimental/complications , Drugs, Chinese Herbal/pharmacology , Muscle, Skeletal/drug effects , Muscular Atrophy/drug therapy , Animals , Cell Line , Cell Survival/drug effects , Drugs, Chinese Herbal/chemistry , Insulin/pharmacology , Male , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Reactive Oxygen Species/metabolism , Streptozocin , Water/chemistry
10.
Phytother Res ; 33(3): 690-701, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30556245

ABSTRACT

Loganin, a major iridoid glycoside obtained from fruits of Cornus officinalis, possesses anti-inflammatory, antitumor, antidiabetic, and osteoporosis prevention effects. Loganin has been linked to neuroprotection in several models of neurodegeneration, including Parkinson's disease (PD). However, mechanisms underlying the neuroprotective effects of loganin are still mostly unknown. Here, we demonstrated the protective effects of loganin against PD mimetic toxin 1-methyl-4-phenylpyridinium (MPP+ ) and the important roles of insulin-like growth factor 1 receptor (IGF-1R) and glucagon-like peptide 1 receptor (GLP-1R) in the neuroprotective mechanisms of loganin. In primary mesencephalic neuronal cultures treated with or without MPP+ , loganin up-regulated expressions of neurotrophic signals including IGF-1R, GLP-1R, p-Akt, BDNF, and tyrosine hydroxylase. Loganin protected against MPP+ -induced apoptosis by up-regulating antiapoptotic protein and down-regulating proapoptotic protein. Moreover, loganin attenuated MPP+ -induced neurite damage via up-regulation of GAP43 and down-regulation of membrane-RhoA/ROCK2/p-LIMK/p-cofilin. Loganin also attenuated MPP+ -induced reactive oxygen species (ROS) production. However, both AG1024, an IGF-1R antagonist, and exendin 9-39, a GLP-1R antagonist, attenuated the protective effects of loganin on MPP+ -induced cytotoxicity, apoptosis, neurite length decrease, and ROS production. Our results suggest that loganin attenuates MPP+ -induced apoptotic death, neurite damage, and oxidative stress through enhancement of neurotrophic signaling, activation of IGF-1R/GLP-1R, and inhibition of RhoA/ROCK pathway, providing the evidence that loganin possesses novel neuroprotective effects.


Subject(s)
Glucagon-Like Peptide-1 Receptor/physiology , Iridoids/pharmacology , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Receptor, IGF Type 1/physiology , Signal Transduction/drug effects , rho-Associated Kinases/antagonists & inhibitors , rhoA GTP-Binding Protein/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Cells, Cultured , Humans , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
11.
J Transl Med ; 16(1): 225, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30103798

ABSTRACT

BACKGROUND: Widespread use of antibiotics in the intensive care unit is a potential cause of the emergence of hospital-acquired pneumonia. This study determined whether Lactobacillus salivarius feeding could reverse antibiotic-induced lung defense impairment in a ventilator model. METHODS: C57BL/6 wild-type (WT) mice received mechanical ventilation for 3 h after intramuscular antibiotic treatment for 6 days. Treatment with dead Lactobacillus salivarius and fructo-oligosaccharides (FOS) feeding were used to stimulate antibacterial protein expression in the intestine. Reactive oxygen species (ROS) in the intestinal mucosa was detected using 2'7'-dichlorofluorescein diacetate. The peroxynitrite production of alveolar macrophages (AMs) was measured using dihydrorhodamine 123 oxidation assay. N-acetylcysteine (NAC), an ROS scavenger, was orally administered to mice receiving antibiotics with FOS feeding. RESULTS: Antibiotic treatment decreased Pseudomonas aeruginosa (PA) phagocytic activity and activity of AMs and protein expression of regenerating islet-derived protein 3ß (Reg3ß) as well as Toll-like receptor 4 (TLR4) in the intestinal mucosa in the ventilator model. Antibiotic treatment also decreased ROS production in the intestinal mucosa, peroxynitrite production of AMs, and RELMß expression as well as NF-κB DNA binding activity of the intestinal mucosa in WT mice but not in MyD88-/- mice. Treatment with dead L. salivarius or FOS feeding increased ROS production, bacterial killing activity, and protein expression of Reg3ß as well as TLR4 in the intestinal mucosa and reversed the inhibitory effects of antibiotics on PA phagocytic activity of AMs. CONCLUSION: Taken together with the finding that ablation of FOS-induced intestinal ROS using NAC decreased peroxynitrite production as well as PA phagocytic activity of AMs and protein expression of CRP-ductin, IL-17, Reg3ß, and RELMß in the intestinal mucosa, we conclude that commensal microflora plays a key role in stimulating lung immunity. Intestinal ROS plays a role as a predictive indicator and modulator of pulmonary defense mechanisms. Antibiotic treatment reduces lung defense against PA infection through the decrease in intestinal Reg3ß and TLR4 expression. Treatment with dead L. salivarius or FOS feeding reverses the antibiotic-induced lung defense impairment through the intestinal ROS/MyD88 pathways.


Subject(s)
Anti-Bacterial Agents/adverse effects , Ligilactobacillus salivarius/physiology , Lung/immunology , Ventilators, Mechanical , Acetylcysteine/pharmacology , Animals , DNA/metabolism , Hormones, Ectopic/metabolism , Intercellular Signaling Peptides and Proteins , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Lung/microbiology , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Neutrophil Infiltration/drug effects , Pancreatitis-Associated Proteins/metabolism , Peroxynitrous Acid/metabolism , Phagocytosis/drug effects , Pneumonia/complications , Protein Binding/drug effects , Pseudomonas aeruginosa/drug effects , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/metabolism
12.
Exp Toxicol Pathol ; 65(6): 887-96, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23352990

ABSTRACT

Zinc oxide nanoparticles (ZnONP) have great potential for medical applications. However, ZnONP is reported to induce acute lung inflammation, which limits its application in humans. We designed in vivo and in vitro studies to clarify ZnONP inflammation and its associated molecular signals. ZnONP with a single dose of 80 µg/30 µl was instilled into the tracheas of mice sacrificed at days 2, 7, 14, and 28 after instillation. Bronchoalveolar lavage fluid showed increased neutrophils and macrophages after treatment. Lung pathology showed a mixed inflammatory infiltrate of neutrophils, lymphocytes, and macrophages primarily in the bronchioles and peribronchiolar areas. Proinflammatory gene expression of TNF-α, IL-6, CXCL1, and MCP-1 was increased at day 2 and decreased after 7 days. The lung pathology resolved at day 28, without fibrosis. It remains unclear whether this acute lung inflammation was caused by ZnONP themselves or Zn(2+) iron released from the nanoparticles. In vitro studies confirming the results of in vivo studies showed increased expression of proinflammatory genes in both MLE12 cells (mouse lung epithelial cells) and RAW264.7 cells (mouse macrophages) with either ZnONP or Zn(NO3)2 treatment; notably, increased levels of proinflammatory genes were obviously higher in cells treated with ZnONP than in cells treated with Zn(NO3)2 at the same molarity dose. TNF-α and MCP-1 were induced only in MLE12 cells. MyD88, an adaptor protein for most Toll-like receptors (TLR) signaling pathways, initiated the ZnONP or Zn(NO3)2-induced lung inflammation. Silencing MyD88 expression with siRNA significantly reduced ZnONP or Zn(NO3)2-induced proinflammatory gene expression in MLE12 and RAW264.7 cells. Single-dose exposure to ZnONP produced the short-term lung inflammation via a MyD88-dependent TLR pathway. These data suggest that although both ZnONP and zinc ion might participate in the inflammatory reactions, ZnONP more effectively induced MyD88-dependent proinflammatory cytokines than zinc ion in lung epithelial cells.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Nanoparticles/toxicity , Pneumonia/chemically induced , Zinc Oxide/toxicity , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Cell Survival/drug effects , Cytokines/immunology , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred ICR , Myeloid Differentiation Factor 88/genetics , Particle Size , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/pathology , RNA, Small Interfering/genetics , Respiratory Function Tests
13.
Kaohsiung J Med Sci ; 28(7 Suppl): S53-62, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22871604

ABSTRACT

The safety of quantum dots (QDs) 705 was evaluated in this study. Mice were treated with QD705 (intravenous) at a single dose of (40 pmol) for 4, 12, 16, and 24 weeks. Effects of QD705 on kidneys were examined. While there was a lack of histopathology, reduction in renal functions was detected at 16 weeks. Electron microscopic examination revealed alterations in proximal convoluted tubule (PCT) cell mitochondria at even much earlier time, including disorientation and reduction of mitochondrial number (early change), mitochondrial swelling, and later compensatory mitochondrial hypertrophy (enlargement mitochondria: giant mitochondria with hyperplastic inner cristae) as well as mitochondrial hyperplasia (increase in mitochondrial biogenesis and numbers) were observed. Such changes probably represent compensatory attempts of the mitochondria for functional loss or reduction of mitochondria in QD705 treated animals. Moreover, degeneration of mitochondria (myelin-figure and cytoplasmic membranous body formation) and degradation of cytoplasmic materials (isolated cytoplasmic pockets of degenerated materials and focal cytoplasmic degradation) also occurred in later time points (16-24 weeks). Such mitochondrial changes were not identical with those induced by pure cadmium. Taken together, we suggest that mitochondria appeared to be the target of QD705 toxicity and specific mitochondrial markers may be useful parameters for toxicity assessments of QDs or other metal-based nanomaterials.


Subject(s)
Fluorescent Dyes/toxicity , Mitochondria/drug effects , Quantum Dots , Animals , Blood Urea Nitrogen , Cadmium/pharmacology , Cadmium/toxicity , Creatinine/blood , Epithelial Cells/drug effects , Epithelial Cells/pathology , Fluorescent Dyes/pharmacology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Male , Mice , Mice, Inbred ICR , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Size/drug effects , Selenium/pharmacology , Selenium/toxicity , Tellurium/pharmacology , Tellurium/toxicity
14.
Nanotechnology ; 23(8): 085102, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22293282

ABSTRACT

Although zinc oxide nanoparticles (ZnONPs) have been applied in nanotechnology, their kinetics and tissue distribution in vivo are unknown. Here we compared the kinetics and tissue distribution of 10 nm (65)ZnONPs, 71 nm (65)ZnONPs and (65)Zn(NO(3))(2) in mice after intravenous injection. The areas under the curves and the half-lives in the second compartment of (65)Zn(NO(3))(2) were greater than those of (65)ZnONPs; the kinetic parameters were similar for both (65)ZnONPs. However, the tissue distributions for the three forms were different. ZnONPs preferentially accumulated in the liver and spleen at 24 h. At day 28, (65)Zn concentration was highest in bone and the proportion of recovered (65)Zn radioactivity was highest in the carcass; these had the same ranking, 10 nm (65)ZnONPs > 71 nm (65)ZnONPs>  (65)Zn(NO(3))(2). Although more than 80% of the 10 nm (65)ZnONPs had been excreted by day 28, greater amounts of the 10 nm (65)ZnONPs than the 71 nm (65)ZnONPs or (65)Zn(NO(3))(2) had accumulated in other organs (brain, lung, heart and kidneys). Zn ions seem to have a longer half-life in the plasma, but ZnONPs show greater tissue accumulation. Although the size of the ZnONPs had no obvious effect on the kinetics, nevertheless the smaller ZnONPs tended to accumulate preferentially in some organs.


Subject(s)
Nanoparticles/chemistry , Nitrates/pharmacokinetics , Zinc Compounds/pharmacokinetics , Zinc Oxide/pharmacokinetics , Animals , Kinetics , Male , Materials Testing , Metabolic Clearance Rate , Mice , Mice, Inbred ICR , Nanoparticles/radiation effects , Nanoparticles/ultrastructure , Neutrons , Nitrates/chemistry , Nitrates/radiation effects , Particle Size , Tissue Distribution , Zinc Compounds/chemistry , Zinc Compounds/radiation effects , Zinc Oxide/chemistry , Zinc Oxide/radiation effects
15.
Nanotoxicology ; 5(4): 650-63, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21142715

ABSTRACT

The objective of this study was to investigate whether quantum dot 705 (QD705) disrupts the cellular antioxidant systems leading to hepatotoxicity in mice. Mice were intravenously injected with QD705 and then sacrificed at week 12 or 16. Homeostasis of antioxidant-related metals, antioxidant activities, induction of oxidative stress, and toxicity in the liver were investigated. Although no histopathological change was observed, a time- and dose-dependent increase in metallothionein expression and reduction in liver function was noticed. Increased copper, zinc, and selenium levels and enhancements of the trace metal-corresponding transporters were noted at week 12. At week 16, a decline of selenium from its elevated level at week 12 was observed, which was accompanied by changes in glutathione peroxidase activity as well as in redox status. A significant reduction in superoxide dismutase activity was observed at 16 weeks. Furthermore, a corresponding elevation of heme oxygenase-1 expression, 8-oxo-7,8-dihydro-2'-deoxyguanosine, interleukin-6 and tumor necrosis factor-alpha suggested the presence of oxidative stress, oxidative DNA damage and inflammation.


Subject(s)
Cadmium/chemistry , Chemical and Drug Induced Liver Injury/etiology , Quantum Dots , Selenium/chemistry , Tellurium/chemistry , Animals , Cadmium/toxicity , Cation Transport Proteins/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cytokines/metabolism , Gene Expression/drug effects , Immunohistochemistry , Liver/chemistry , Liver/metabolism , Liver/pathology , Male , Metallothionein/metabolism , Mice , Mice, Inbred ICR , Oxidative Stress/drug effects , Selenium/toxicity , Superoxide Dismutase/metabolism , Tellurium/toxicity
16.
Nanotoxicology ; 5(1): 91-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21155710

ABSTRACT

Cadmium (Cd) is a component in quantum dot 705 (QD705). Whether QD705 behaves similar to Cd in vivo is of great concern. We compared the distributional kinetics of cadmium chloride (CdCl(2)) and QD705 in mice after intravenous injection. QD705 showed a longer plasma and body retention than CdCl(2) and could be detected in the brain during early exposure. While both the liver and spleen demonstrated a constant Cd concentration for 28 days after QD705 injection, it is likely that this represents intact QD705 stored in mononuclear phagocytes. The kidneys showed a time-dependent accumulation of Cd in the QD705-exposed animals. By day 28, Cd in the kidneys from QD705 was 3-fold that of CdCl(2). QD705 and CdCl(2) have very different kinetics in distribution and metabolism. The long body retention of QD705 in the kidneys may mean that QD705 has even more renal toxicity than CdCl(2).


Subject(s)
Cadmium Chloride/pharmacokinetics , Hazardous Substances/toxicity , Metal Nanoparticles/toxicity , Quantum Dots , Animals , Cadmium Chloride/toxicity , Diagnostic Imaging/methods , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Metallothionein/metabolism , Mice , Mice, Inbred ICR , Tissue Distribution
17.
Nanotechnology ; 20(21): 215101, 2009 May 27.
Article in English | MEDLINE | ID: mdl-19423922

ABSTRACT

QD705 is a cadmium/selenium/tellurium (Cd/Se/Te)-based quantum dot with good potential for biomedical applications. Although the biological fate of QD705 is established, its chemical fate in the biological system is still unknown. Since the chemical nature of Cd in QD705 (either stays as bounded Cd or becomes free Cd) is closely related to the toxicity of this nanocrystal, information on its chemical fate is critically needed. In this study we investigated the chemical fate of QD705 in the kidneys of mice. We used the molar ratio of Cd and Te (increased Cd/Te ratio signifies increased Cd release from QD705) and the induction of tissue metallothionein (MT) as markers for elevated free Cd in tissues. Our study indicated that 100% of QD705 (measured as Cd) was still retained in the body 16 weeks after exposure, with significant time redistribution to the kidneys. Furthermore, there were an elevation in both the molar Cd/Te ratio and MT-1 expression in the kidneys, suggesting that free Cd was released from QD705. Thus QD705 is not as stable or biologically inert as many may have once believed. Our study demonstrated that free Cd indeed can be released from QD705 in the kidneys and increases the risk of renal toxicity.


Subject(s)
Cadmium Compounds/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney/drug effects , Kidney/pathology , Selenium Compounds/toxicity , Tellurium/toxicity , Animals , Cadmium Compounds/pharmacokinetics , Dose-Response Relationship, Drug , Kidney/metabolism , Kidney Diseases/metabolism , Male , Materials Testing , Mice , Mice, Inbred ICR , Quantum Dots , Selenium Compounds/pharmacokinetics , Tellurium/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...