Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
ACS Appl Mater Interfaces ; 16(10): 12332-12338, 2024 Mar 13.
Article En | MEDLINE | ID: mdl-38426453

Organic dye-based agents with near-infrared (NIR)-II absorption have great potential for cancer theranostics because of the deeper tissue penetration and good biocompatibility. However, proper design is required to develop NIR-II-absorbing dyes with good optical properties. We proposed to construct chalcogen atom-modulated croconaine for NIR-II light-triggered photothermal theranostics. By introducing different chalcogen atoms (O, S, Se, or Te) into the structure of croconaine, the light absorption of croconaine can be precisely regulated from the NIR-I to the NIR-II range due to the heavy-atom effect. Especially, Te-substituted croconaine (CRTe) and its nanoformulations exhibit superior NIR-II responsiveness, a high photothermal conversion efficiency (70.6%), and good photostability. With their favorable tumor accumulation, CRTe-NPs from tumor regions can be visualized by NIR-II optoacoustic systems with high resolution and high contrast; meanwhile, their superior photothermal performance also contributes to efficient cell killing and tumor elimination upon 1064 nm laser irradiation. Therefore, this work provides an efficient strategy for the molecular design of NIR-II organic photothermal agents.


Chalcogens , Nanoparticles , Neoplasms , Humans , Theranostic Nanomedicine , Neoplasms/drug therapy , Coloring Agents/chemistry , Chalcogens/pharmacology , Nanoparticles/chemistry , Phototherapy , Cell Line, Tumor
2.
Protein J ; 42(6): 753-765, 2023 Dec.
Article En | MEDLINE | ID: mdl-37690089

Excessive production of transforming growth factor ß1 (TGF-ß1) in activated hepatic stellate cells (aHSCs) promotes liver fibrosis by activating the TGF-ß1/Smad signaling pathway. Thus, specifically inhibiting the pro-fibrotic activity of TGF-ß1 in aHSCs is an ideal strategy for treating liver fibrosis. Overexpression of platelet-derived growth factor ß receptor (PDGFßR) has been demonstrated on the surface of aHSCs relative to normal cells in liver fibrosis. Interferon-gamma peptidomimetic (mIFNγ) and truncated TGF-ß receptor type II (tTßRII) inhibit the TGF-ß1/Smad signaling pathway by different mechanisms. In this study, we designed a chimeric protein by the conjugation of (1) mIFNγ and tTßRII coupled via plasma protease-cleavable linker sequences (FNPKTP) to (2) PDGFßR-recognizing peptide (BiPPB), namely BiPPB-mIFNγ-tTßRII. This novel protein BiPPB-mIFNγ-tTßRII was effectively prepared using Escherichia coli expression system. The active components BiPPB-mIFNγ and tTßRII were slowly released from BiPPB-mIFNγ-tTßRII by hydrolysis using the plasma protease thrombin in vitro. Moreover, BiPPB-mIFNγ-tTßRII highly targeted to fibrotic liver tissues, markedly ameliorated liver morphology and fibrotic responses in chronic liver fibrosis mice by both inhibiting the phosphorylation of Smad2/3 and inducing the expression of Smad7. Meanwhile, BiPPB-mIFNγ-tTßRII markedly reduced the deposition of collagen fibrils and expression of fibrosis-related proteins in acute liver fibrosis mice. Furthermore, BiPPB-mIFNγ-tTßRII showed a good safety performance in both liver fibrosis mice. Taken together, BiPPB-mIFNγ-tTßRII improved the in vivo anti-liver fibrotic activity due to its high fibrotic liver-targeting potential and the dual inhibition of the TGF-ß1/Smad signaling pathway, which may be a potential candidate for targeting therapy on liver fibrosis.

3.
Endokrynol Pol ; 74(3): 234-242, 2023.
Article En | MEDLINE | ID: mdl-37335067

INTRODUCTION: Familial hypercholesterolaemia (FH) is a common hereditary genetic disorder, characterized by elevated circulating low-density lipoprotein cholesterol (LDL-C) and lipoprotein (a) [Lp(a)] concentrations, leading to atherosclerotic cardiovascular disease (ASCVD). Two types of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors- alirocumab and evolocumab- are efficient drugs in the treatment of FH, which can effectively reduce Lp(a) levels. MATERIAL AND METHODS: Embase, MEDLINE, and PubMed up to November 2022 were searched for randomized clinical trials (RCTs) evaluating the effect of alirocumab/evolocumab and placebo treatment on plasma Lp(a) levels in FH. Statistics were analysed by Review Manager (RevMan 5.3) and Stata 15.1. RESULTS: Eleven RCTs involved a total of 2408 participants. Alirocumab/evolocumab showed a significant efficacy in reducing Lp(a) [weighted mean difference (WMD): -20.10%, 95% confidence interval (CI): -25.59% to -14.61%] compared with placebo. In the drug type subgroup analyses, although the efficacy of evolocumab was slightly low (WMD: -19.98%, 95% CI: -25.23% to -14.73%), there was no difference with alirocumab (WMD: -20.54%, 95% CI: -30.07% to -11.02%). In the treatment duration subgroup analyses, the efficacy of the 12-week duration group (WMD: -17.61%, 95% CI: -23.84% to -11.38%) was lower than in the group of ≥ 24 weeks' duration (WMD: -22.81%, 95% CI: -31.56% to -14.07%). In the participants' characteristics subgroup analyses, the results showed that no differential effect of alirocumab/evolocumab therapy on plasma Lp(a) concentrations was observed (heterozygous FH [HeFH] WMD: -20.07%, 95% CI: -26.07% to -14.08%; homozygous FH [HoFH] WMD: -20.04%, 95% CI: -36.31% to -3.77%). Evaluation of all-cause adverse events (AEs) between alirocumab/evolocumab groups and placebo groups [relative risk (RR): 1.05, 95% CI: 0.98-1.12] implied no obvious difference between the 2 groups. CONCLUSIONS: Anti-PCSK9 drugs (alirocumab and evolocumab) may be effective as therapy for reducing serum Lp(a) levels in FH, and no differences were observed in treatment durations, participant characteristics, and other aspects of the 2 types of PCSk9 inhibitors. However, further experimental studies and RCTs are warranted to clarify the mechanism of PSCK9 inhibitors to lowering Lp(a) concentrations in FH.


Anticholesteremic Agents , Hyperlipoproteinemia Type II , Humans , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Lipoprotein(a)/therapeutic use , Randomized Controlled Trials as Topic , Hyperlipoproteinemia Type II/drug therapy , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use
4.
Exp Ther Med ; 25(5): 221, 2023 May.
Article En | MEDLINE | ID: mdl-37123202

Lactate dehydrogenase (LDH) and aspartate aminotransferase (AST) are important indicators of cardiovascular, muscle and liver lesions, and can be used as prognostic indicators for infectious diseases, such as coronavirus disease 2019 (COVID-19). The present systematic review and meta-analysis assessed the prognostic value of LDH and AST levels for COVID-19 severity. Ovid-Medline, PubMed, Embase and The Cochrane Library were used to search for articles, according to the inclusion and exclusion criteria, until July 2022. The meta-analysis was performed using Revman5.3 and Stata15.1. Standardized mean difference (SMD) and 95% confidence intervals (CIs) of LDH and AST concentrations were analyzed using a random-effects model. Heterogeneity was investigated using meta-regression and subgroup methods. A total of 4,342 patients with COVID-19 in 23 articles were included in the present study. LDH (SMD=1.21; 95% CI: 0.98, 1.44) and AST (SMD=0.68; 95% CI: 0.54, 0.81) were significantly higher in patients with severe COVID-19 compared with in those with non-severe COVID-19. Serum LDH and AST levels in critically ill patients with COVID-19 were increased, suggesting a correlation between the levels of LDH and AST and the severity of COVID-19. These findings may help to develop a risk-stratified approach to the care of patients with this disease.

5.
Appl Microbiol Biotechnol ; 107(1): 447-457, 2023 Jan.
Article En | MEDLINE | ID: mdl-36460807

The lack of electron donors prevents the effective degradation of azo dyes by bacteria, which severely limits the practical application of conventional biological treatment. Herein, we innovatively designed a bio-photoelectric reduction degradation system composed of CdS and Shewanella decolorationis, which could effectively degrade amaranth in anaerobic conditions driven by light when electron donors were unavailable. Compared with bare S. decolorationis and S. decolorationis (heat-killed)-CdS biohybrid, S. decolorationis-CdS biohybrid had 39.36-fold and 3.82-fold higher first-order kinetic constants, respectively. The morphology, particle size, elemental composition, crystalline type, photovoltaic properties, and band structure of the nanoparticles synthesized by S. decolorationis were carefully examined and analyzed. Light-driven biodegradation experiments showed that amaranth was degraded by the synergy of CdS and S. decolorationis. Reductive degradation of amaranth by electrons was demonstrated by electron and hole trapping. The effect of potential coexisting contaminants, which might serve as hole scavengers, on the degradation of amaranth was evaluated. Membrane protein inhibition experiments also suggested that NADH dehydrogenase, menaquinone, and cytochrome P450 played an important role in electron transfer between CdS and Shewanella decolorationis. The cyclic conversion of NAD+/NADH was probably the most critical rate-limiting step. Electrochemical measurements suggested that faster electron transfer might facilitate the degradation of amaranth. Our findings might contribute to the degradation of azo dyes in wastewater lacking electron donors and deepen our recognition of the microbe-material interface. KEY POINTS: • A BPRDS was constructed with Shewanella decolorationis and CdS. • Amaranth was effectively degraded by BPRDS in anaerobic conditions driven by light. • NDH, MQ, and CYP450 were involved in electron transfer.


Azo Compounds , Shewanella , Azo Compounds/metabolism , Wastewater , Electrons , Coloring Agents/metabolism , Oxidation-Reduction , Shewanella/metabolism , Amaranth Dye/metabolism , Amaranth Dye/pharmacology
6.
Exp Ther Med ; 19(2): 1439-1447, 2020 Feb.
Article En | MEDLINE | ID: mdl-32010320

Long non-coding RNAs (lncRNAs) have been indicated to serve critical roles in cancer development and progression. Long intergenic non-protein coding RNA 70 (LINC00707) was recently reported to be an oncogene involved in the tumorigenesis of several types of human cancer. However, the clinical role, biological functions and molecular mechanism of LINC00707 in colorectal cancer (CRC) remain unclear. The aim of the present study was to investigate the biological effects and mechanism of LINC00707 in CRC. Reverse transcription-quantitative PCR was used to detect the expression levels of LINC00707 in 65 CRC tissue samples and CRC cell lines (HCT116, HT29 and SW480). Cell Counting Kit-8 and colony formation assays were performed to investigate the effects of LINC00707 on CRC cell proliferation. A dual-luciferase reporter assay was conducted to investigate the mechanisms of LINC00707 in CRC. The upregulation of LINC00707 expression was significantly associated with tumor size, stage and poor survival in patients with CRC. LINC00707 also acted as an independent prognostic factor for CRC. Functional analyses revealed that the knockdown of LINC00707 could inhibit CRC cell proliferation. Furthermore, bioinformatics analysis demonstrated that microRNA (miR)-485-5p could directly bind to LINC00707, which was confirmed by a dual-luciferase reporter assay. In conclusion, the upregulation of LINC00707 is associated with a shorter survival time in patients with CRC. Knockdown of LINC00707 may inhibit the proliferation of CRC cells by binding with miR-485-5p.

7.
Autophagy ; 10(10): 1801-13, 2014 Oct 01.
Article En | MEDLINE | ID: mdl-25126734

Transient cerebral ischemia leads to endoplasmic reticulum (ER) stress. However, the contributions of ER stress to cerebral ischemia are not clear. To address this issue, the ER stress activators tunicamycin (TM) and thapsigargin (TG) were administered to transient middle cerebral artery occluded (tMCAO) mice and oxygen-glucose deprivation-reperfusion (OGD-Rep.)-treated neurons. Both TM and TG showed significant protection against ischemia-induced brain injury, as revealed by reduced brain infarct volume and increased glucose uptake rate in ischemic tissue. In OGD-Rep.-treated neurons, 4-PBA, the ER stress releasing mechanism, counteracted the neuronal protection of TM and TG, which also supports a protective role of ER stress in transient brain ischemia. Knocking down the ER stress sensor Eif2s1, which is further activated by TM and TG, reduced the OGD-Rep.-induced neuronal cell death. In addition, both TM and TG prevented PARK2 loss, promoted its recruitment to mitochondria, and activated mitophagy during reperfusion after ischemia. The neuroprotection of TM and TG was reversed by autophagy inhibition (3-methyladenine and Atg7 knockdown) as well as Park2 silencing. The neuroprotection was also diminished in Park2(+/-) mice. Moreover, Eif2s1 and downstream Atf4 silencing reduced PARK2 expression, impaired mitophagy induction, and counteracted the neuroprotection. Taken together, the present investigation demonstrates that the ER stress induced by TM and TG protects against the transient ischemic brain injury. The PARK2-mediated mitophagy may be underlying the protection of ER stress. These findings may provide a new strategy to rescue ischemic brains by inducing mitophagy through ER stress activation.


Brain Ischemia/drug therapy , Endoplasmic Reticulum Stress , Mitophagy , Neuroprotective Agents/therapeutic use , Thapsigargin/therapeutic use , Tunicamycin/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Brain Ischemia/pathology , Endoplasmic Reticulum Stress/drug effects , Glucose/deficiency , Mice , Mitophagy/drug effects , Neuroprotective Agents/pharmacology , Oxygen/metabolism , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Thapsigargin/pharmacology , Tunicamycin/pharmacology
8.
Neurosci Lett ; 422(1): 34-8, 2007 Jul 05.
Article En | MEDLINE | ID: mdl-17590512

The histidine-containing dipeptide, carnosine (beta-alanyl-L-histidine), is present in high concentrations in mammalian brain of mammals. There are many theories about its biological functions, such as anti-inflammatory agent, free radical scavenger, and protein glycosylation inhibitor, however, the role of carnosine in morphine addiction is less understood. Therefore, the objectives of this study were to determine the effects of carnosine on the development of morphine-induced conditioned place preference (CPP) and investigate its possible mechanism of action in Sprague-Dawley rats. Intraperitioneal (i.p.) injection of carnosine (200, 500, 1000 mg/kg) significantly inhibited the development of morphine-induced CPP in a dose-dependent manner. Although carnosine had no appreciable effect on the levels of histamine in the ventral tegmental area (VTA), nucleus accumbens (NAc) and prefrontal cortex (PFC), it significantly decreased glutamate level in the VTA, dopamine levels in the NAc and PFC, and DOPAC level in the NAc of morphine-treated rats. These results indicate that carnosine inhibits morphine-induced CPP in rats, and its action may be due to modulation of dopaminergic and glutaminergic activity. The study suggests that carnosine has potential as a new anti-addictive drug.


Analgesics, Opioid/antagonists & inhibitors , Analgesics, Opioid/pharmacology , Carnosine/pharmacology , Conditioning, Operant/drug effects , Morphine/antagonists & inhibitors , Morphine/pharmacology , Animals , Brain Chemistry/drug effects , Dopamine/metabolism , Dose-Response Relationship, Drug , Histamine/metabolism , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
9.
Biochem Pharmacol ; 73(5): 709-17, 2007 Mar 01.
Article En | MEDLINE | ID: mdl-17169331

Since the histidine-containing dipeptide carnosine (beta-alanyl-L-histidine) is believed to have many physiological functions in the brain, we investigated the neuroprotective effects of carnosine and its mechanisms of action in an in vitro model of neurotoxicity induced by N-methyl-d-aspartate (NMDA) in differentiated PC12 cells. Pretreatment with carnosine increased the viability and decreased the number of apoptotic and necrotic cells measured by MTT and Hoechst 33342 and propidium iodide (PI) double staining assays. Carnosine also can inhibit the glutamate release and increase HDC activity and the intracellular and extracellular contents of carnosine, histidine and histamine detected by high-performance liquid chromatography (HPLC). The protection by carnosine was reversed by alpha- fluoromethylhistidine, a selective and irreversible inhibitor of histidine decarboxylase (HDC). Pyrilamine and thioperamide, selective central histamine H(1) and H(3) antagonists also significantly reversed the protection of carnosine. Further, the inhibition of glutamate release by carnosine was reversed by thioperamide. Therefore, the protective mechanism of carnosine may not only involve the carnosine-histidine-histamine pathway, but also H(1)/H(3) receptors and the effective inhibition of glutamate release. This study indicates that carnosine may be an endogenous protective factor and calls for its further study as a new antiexcitotoxic agent.


Carnosine/metabolism , Carnosine/pharmacology , Histamine/metabolism , Histidine/metabolism , N-Methylaspartate/toxicity , Neurons/drug effects , Animals , Cell Differentiation , Glutamic Acid/metabolism , Methylhistidines , PC12 Cells , Piperidines/pharmacology , Rats , Receptors, Histamine H1/metabolism , Receptors, Histamine H2 , Receptors, Histamine H3/metabolism , gamma-Aminobutyric Acid/metabolism
...