Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Opt Express ; 32(10): 16867-16878, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38858883

ABSTRACT

In this paper, a highly integrated terahertz (THz) biosensor is proposed and implemented, which pioneered the preparation of low-temperature gallium arsenide (LT-GaAs) thin film photoconductive antenna (PCA) on the sensor for direct generation and detection of THz waves, simplifying complex terahertz time-domain spectroscopy (THz-TDS) systems. A latch type metasurface is deposited in the detection region to produce a resonance absorption peak at 0.6 THz that is independent of polarisation. Microfluidics is utilised and automatic injection is incorporated to mitigate the experimental effects of hydrogen bond absorption of THz waves in aqueous-based environment. Additionally, cell damage is minimised by regulating the cell flow rate. The biosensor was utilised to detect the concentration of three distinct sizes of bacteria with successful results. The assay was executed as a proof of concept to detect two distinct types of breast cancer cells. Based on the experimental findings, it has been observed that the amplitude and blueshift of the resonance absorption peaks have the ability to identify and differentiate various cancer cell types. The findings of this study introduce a novel approach for developing microfluidic THz metasurface biosensors that possess exceptional levels of integration, sensitivity, and rapid label-free detection capabilities.


Subject(s)
Arsenicals , Biosensing Techniques , Gallium , Terahertz Spectroscopy , Gallium/chemistry , Arsenicals/chemistry , Biosensing Techniques/instrumentation , Terahertz Spectroscopy/instrumentation , Humans , Equipment Design , Microfluidics/instrumentation
2.
Rev Sci Instrum ; 93(4): 043004, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35489890

ABSTRACT

The bandwidth of very high gain (≥100 MV/A) transimpedance amplifiers is restricted to below 100 kHz, unless measures are employed to mitigate the effect of circuit parasitic capacitances. Current approaches involve significantly increased circuit complexity and component count. They may suffer unwanted noise pickup or destructive capacitive coupling to ground, the latter restricting the available bandwidth. We demonstrate that combining a positive feedback circuit with a low-pass filter network extends the bandwidth of a transimpedance amplifier out to the limit of gain peaking (>1 MHz) without increasing the noise signal. The circuit uses a single inverting amplifier and very large feedback-resistance to provide a canceling parasitic-capacitance positive feedback signal. This can negate both the negative feedback-resistor parasitic-capacitance and the input/output pin parasitic-capacitance of the transimpedance amplifier. The circuit solves the problem of destructive distributed-capacitive coupling to ground along the feedback resistor.

3.
J Agric Food Chem ; 70(9): 2889-2897, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35212537

ABSTRACT

Chronic stress induction in immunosuppression and splenocyte apoptosis is commonly associated with increased susceptibility to various diseases. Lycopene (LYC) is a member of the carotenoid family with immune restoration and anti-apoptotic function. However, little is known about the mechanisms underlying the protective roles of LYC against spleen injury induced by chronic stress. Herein, male Wistar rats were undergoing chronic restraint stress and/or administered LYC (10 mg/kg) for 21 days. The effective model establishment was validated by open-field tests and levels of corticosterone in serum. Histopathological staining observation displayed that LYC could reduce chronic stress-induced spleen structure damage. Furthermore, LYC treatment significantly reduced the number of apoptotic-positive splenocytes caused by chronic stress via the death receptor apoptotic pathway. We detected the interleukin 4 and interferon γ levels in serum and spleen to determine the ratio of Th1 and Th2 and found that LYC can alleviate the immunosuppression induced by chronic stress. Notably, western blot and real-time polymerase chain reaction indicated that LYC can reduce the expression of the Notch-pathway-related proteins and mRNA in rats exposed to chronic stress. Further study of the potential mechanisms by adding the Notch pathway inhibitor DAPT revealed that LYC alleviates the structure damage, apoptosis, and immunosuppression caused by chronic stress via the suppression of the Notch pathway. Overall, this study presents a strong rationale to target LYC as a treatment strategy to relieve chronic stress-induced spleen injury.


Subject(s)
Oxidative Stress , Spleen , Animals , Apoptosis , Immunosuppression Therapy , Lycopene/metabolism , Male , Rats , Rats, Wistar , Signal Transduction , Spleen/metabolism
4.
Appl Opt ; 60(29): 9036-9041, 2021 Oct 10.
Article in English | MEDLINE | ID: mdl-34623983

ABSTRACT

A terahertz (THz) photoconductive antenna is prepared using, to the best of our knowledge, a novel method, which has high yield and strong stability. It eliminates the stripping process of a thin-film THz antenna and effectively prevents the toxicity of the corrosion solution, the easy damage in the transfer process, and the weak bonding with the substrate. First, a 200 µm copper wire is bundled on a low-temperature GaAs epitaxial wafer, and then the electrode of the photoconductive antenna is fabricated using the vacuum evaporation method. Finally, the THz time-domain signal with a high signal-to-noise ratio and good repeatability is obtained using an 800 nm laser. Additionally, the influence of pump light and detection light power on THz signal intensity is studied when the total optical power is unchanged. Results show that when the total power of the laser is greater than a certain value, there is an optimal ratio between the pump power and the detection power, which can maximize the signal-to-noise ratio of the THz wave. This provides a basis for the effective application of a THz antenna and lays a foundation for improving the detection sensitivity of samples.

5.
Mol Neurobiol ; 58(11): 5533-5547, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34363182

ABSTRACT

Dexmedetomidine (DEX) has multiple biological effects. Here, we investigated the neuroprotective role and molecular mechanism of DEX against lipopolysaccharide (LPS)-induced hippocampal neuronal apoptosis. Sprague Dawley rats were intraperitoneally injected with LPS (10 mg/kg) and/or DEX (30 µg/kg). We found that DEX improved LPS-induced alterations of hippocampal microstructure (necrosis and neuronal loss in the CA1 and CA3 regions) and ultrastructure (mitochondrial damage). DEX also attenuated LPS-induced inflammation and hippocampal apoptosis by inhibiting the increase of interleukin-1ß, interleukin-6, interleukin-18, and tumor necrosis factor-α levels and downregulating the expression of mitochondrial apoptosis pathway-related proteins. Moreover, DEX prevented the LPS-induced activation of the c-Myc/chloride intracellular channel 4 (CLIC4) pathway. DEX inhibited the p38 MAPK pathway, but not JNK and ERK. To further clarify whether DEX alleviated LPS-induced neuronal apoptosis through the p38 MAPK/c-Myc/CLIC4 pathway, we treated PC12 cells with p38 MAPK inhibitor SB203582 (10 µM). DEX had the same effect as SB203582 in reducing the protein and mRNA expression of c-Myc and CLIC4. Furthermore, DEX and SB203582 diminished LPS-induced apoptosis, indicated by decreased Bax and Tom20 fluorescent double-stained cells, reduced annexin V-FITC/PI apoptosis rate, and reduced protein expression levels of Bax, cytochrome C, cleaved caspase-9, and cleaved caspase-3. Taken together, the findings indicate that DEX attenuates LPS-induced hippocampal neuronal apoptosis by regulating the p38 MAPK/c-Myc/CLIC4 signaling pathway. These findings provide new insights into the mechanism of Alzheimer's disease and depression and may help aid in drug development for these diseases.


Subject(s)
Apoptosis , Hippocampus , MAP Kinase Signaling System , Neurons , Animals , Male , Rats , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Chloride Channels/physiology , Cytokines/blood , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Hippocampus/drug effects , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/pathology , PC12 Cells , Proto-Oncogene Proteins c-myc/physiology , Random Allocation , Rats, Sprague-Dawley
6.
J Hazard Mater ; 411: 125179, 2021 06 05.
Article in English | MEDLINE | ID: mdl-33858114

ABSTRACT

Alumina nanoparticles (AlNPs) exposure causes hippocampal-dependent cognitive dysfunction. However, whether chronic stress exacerbates AlNPs-induced hippocampal lesion and its mechanism remains unclear. This study was aimed to investigate the combined effects and mechanisms of AlNPs and chronic stress on the hippocampal lesion. The behavioral tests demonstrated that combined exposure to AlNPs and chronic restraint stress (CRS) worsened both cognition and depression-like behavior than exposed to AlNPs and CRS alone. Microstructural and ultrastructural observations showed that combined exposure to AlNPs and CRS exacerbated hippocampal damage. Both AlNPs and CRS induced hippocampal neuronal ferroptosis, presenting as iron and glutamate metabolism disorder, GPX4 fluorescence of neurons decrease, LPO and ROS levels increase, and FJB-positive neurons increase. Meanwhile, combined exposure to AlNPs and CRS exacerbated hippocampal neuronal ferroptosis. Mechanism investigation revealed that combined exposure to AlNPs and CRS activated IFN-γ/ASK1/JNK signaling pathway. Furthermore, IFN-γ neutralizing antibody R4-6A2 effectively inhibited the activation of IFN-γ/ASK1/JNK signaling pathway, alleviated hippocampal neuronal ferroptosis and improved cognition ability. ASK1 inhibitor GS-4997 also improved hippocampal neuronal ferroptosis and cognitive dysfunction by inhibiting ASK1/JNK signaling pathway. Together, these results demonstrate that combined exposure to AlNPs and CRS exacerbates hippocampal neuronal ferroptosis via activating IFN-γ/ASK1/JNK signaling pathway.


Subject(s)
Ferroptosis , Nanoparticles , Aluminum Oxide , Animals , Apoptosis , Hippocampus , MAP Kinase Signaling System , Neurons , Rats
7.
Front Pharmacol ; 11: 790, 2020.
Article in English | MEDLINE | ID: mdl-32670056

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) is a severe complication of sepsis; however, no effective drugs have been found. Activation of the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome is a major pathogenic mechanism of AKI induced by lipopolysaccharide (LPS). Autophagy, a process of intracellular degradation related to renal homeostasis, effectively restricts inflammatory responses. Herein, we explored the potential protective mechanisms of dexmedetomidine (DEX), which has confirmed anti-inflammatory effects, on LPS-induced AKI. METHODS: AKI was induced in rats by injecting 10 mg/kg of LPS intraperitoneally (i.p.). Wistar rats received intraperitoneal injections of DEX (30 µg/kg) 30 min before an intraperitoneal injection of LPS. Atipamezole (ATI) (250 µg/kg) and 3-methyladenine (3-MA) (15 mg/kg) were intraperitoneally injected 30 min before the DEX injection. RESULTS: DEX significantly attenuated renal injury. Furthermore, DEX decreased activation of the NLRP3 inflammasome and expression of interleukins 1ß and 18. In addition, autophagy-related protein and gene analysis indicated that DEX could significantly enhance autophagy. Finally, we verified the pharmacological effects of DEX on the 5'-adenosine monophosphate-activated protein kinase (AMPK)/mechanistic target of rapamycin (mTOR) pathway. Atip and 3-MA significantly reversed the protective effects of DEX. CONCLUSIONS: Our results suggest that the protective effects of DEX were mediated by enhanced autophagy via the α2-adrenoreceptor/AMPK/mTOR pathway, which decreased activation of the NLRP3 inflammasome. Above all, we verified the renal protective effects of DEX and offer a new treatment strategy for AKI.

8.
Front Pharmacol ; 11: 128, 2020.
Article in English | MEDLINE | ID: mdl-32158395

ABSTRACT

BACKGROUND: Acute kidney injury (AKI) is often secondary to sepsis. Previous studies suggest that damaged mitochondria and the inhibition of autophagy results in AKI during sepsis, but dexmedetomidine (DEX) alleviates lipopolysaccharide (LPS)-induced AKI. However, it is uncertain whether the renoprotection of DEX is related to autophagy or the clearance of damaged mitochondria in sepsis-induced AKI. METHODS: In this study, AKI was induced in rats by injecting 10 mg/kg of LPS intraperitoneally (i.p.). The rats were also pretreated with DEX (30 µg/kg, i.p.) 30 min before the injection of LPS. The structure and function of kidneys harvested from the rats were evaluated, and the protein levels of autophagy-related proteins, oxidative stress levels, and apoptosis levels were measured. Further, atipamezole (Atip) and 3-Methyladenine (3-MA), which are inhibitors of DEX and autophagy, respectively, were administered before the injection of DEX to examine the protective mechanism of DEX. RESULTS: Pretreatment with DEX ameliorated kidney structure and function. DEX decreased the levels of blood urea nitrogen (BUN) and creatinine (Cre), urine kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), reactive oxygen species (ROS), and apoptosis proteins (such as cleaved caspase-9 and cleaved caspase-3). However, DEX upregulated the levels of autophagy and mitophagy proteins, such as Beclin-1, LC3 II and PINK1. These results suggest that DEX ameliorated LPS-induced AKI by reducing oxidative stress and apoptosis and enhancing autophagy. To promote autophagy, DEX inhibited the phosphorylation levels of PI3K, AKT, and mTOR. Furthermore, the administration of Atip and 3-MA inhibitors blocked the renoprotection effects of DEX. CONCLUSIONS: Here, we demonstrate a novel mechanism in which DEX protects against LPS-induced AKI. DEX enhances autophagy, which results in the removal of damaged mitochondria and reduces oxidative stress and apoptosis in LPS-induced AKI through the α2-AR and inhibition of the PI3K/AKT/mTOR pathway.

9.
Toxicology ; 429: 152312, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31693917

ABSTRACT

LPS-induced neuronal apoptosis leads to neurodegenerative diseases (NDs). However, the mechanisms underlying NDs pathogenesis remains unclear. The apoptotic response to activation of the c-Myc/chloride intracellular channel (CLIC4) pathway is directed through a mitochondrial pathway. In this study, we aimed to explore the c-Myc/CLIC4 pathway in the progression of NDs induced by lipopolysaccharide (LPS). In an in vivo experiment, the results of HE staining, transmission electron microscopic, immunofluorescence microscopy of cleaved caspase-3 and Bax and the increasing expression of apoptotic pathway related proteins in mitochondria showed that LPS (10 mg/kg) administration damaged mitochondrial and induced hippocampal neuron apoptosis. The Western blot and RT-PCR indicated that LPS induced the activation of c-Myc/CLIC4 pathway. Furthermore, in an in vitro experiment, PC12 cells were exposed to LPS to induce cell injuries to mimic the model of NDs. To further confirm the role of the c-Myc/CLIC4 pathway in LPS-induced neuronal apoptosis, the gene knockout of c-Myc and CLIC4 were performed by CRISPR/Cas9. The results of the flow cytometry assay and Annexin V-FITC/PI showed that knocking out c-Myc and CLIC4 significantly reduced cell apoptosis. The results of Western blot and dual immunofluorescence with Cyt c and TOM20 showed that knocking out c-Myc and CLIC4 significantly reduced the expression of mitochondrial apoptosis-related proteins. Our data confirmed that LPS-induced apoptosis is regulated by the activation of c-Myc/CLIC4 pathway. These results support further research mechanisms underlying neurodegenerative diseases and can provide effective pharmacodynamic targets for the clinical development of therapeutic drugs for neurodegenerative diseases.


Subject(s)
Apoptosis/physiology , Chloride Channels/metabolism , Neurodegenerative Diseases/physiopathology , Proto-Oncogene Proteins c-myc/metabolism , Animals , Chloride Channels/genetics , Disease Models, Animal , Disease Progression , Gene Knockout Techniques , Lipopolysaccharides/toxicity , Male , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , PC12 Cells , Proto-Oncogene Proteins c-myc/genetics , Rats , Rats, Sprague-Dawley
10.
Dalton Trans ; 40(24): 6490-3, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21594271

ABSTRACT

A pair of luminescent heterometallic lanthanide-transition-metal coordination polymers, namely, [PrLAg(3)(SCN)(6)·H(2)O](n) (1) and [PrLAg(3)(SCN)(6)](n) (2) [L = 2,6-di(pyrazol-1-yl)pyridine], have been obtained with different cooling rates under solvothermal conditions. The two structures are pseudo- supramolecular isomers constructed via the same [PrL(NCS)(6)](3-) subunit and different Ag-S clusters, presenting diverse two-dimensional and three dimensional frameworks, respectively. In both complexes, the tridentate chelate L, acting as an organic chromophore, along with the d(10)-block Ag-S clusters, are simultaneously immobilized, and effectively sensitize the Pr(III)-based luminescence.


Subject(s)
Fluorescent Dyes/chemistry , Polymers/chemistry , Praseodymium/chemistry , Silver Compounds/chemistry , Coordination Complexes/chemistry , Crystallography, X-Ray , Isomerism , Lanthanoid Series Elements/chemistry , Molecular Conformation , Transition Elements/chemistry
11.
Appl Opt ; 45(33): 8448-53, 2006 Nov 20.
Article in English | MEDLINE | ID: mdl-17086254

ABSTRACT

We demonstrate a wavelength-selective photodetector that combines a Fabry-Perot filtering cavity (FPC) with a taper absorption cavity (TAC). The taper cavity shows a nonresonant effect but exhibits an absorption enhancement effect, so that high speed, high quantum efficiency, wide tuning range, and an ultranarrow spectral linewidth can be achieved simultaneously. Device performance was theoretically investigated by including key factors such as taper angle, finite-size diffracting-beam input, and lateral walk-off in the taper cavity. The device was fabricated by bonding a GaAs-based FPC, which can be tuned via thermal-optic effect, with an InP-based TAC. An integrated device with a spectral linewidth of 0.6 nm (FWHM), a wavelength tuning range of 10.2 nm(1518.0-1528.2 nm), a 3 dB bandwidth of 12 GHz, and a quantum efficiency of approximately 70% was demonstrated, and the absorption layer thickness is only 0.3 microm.

SELECTION OF CITATIONS
SEARCH DETAIL
...