Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Appl Opt ; 63(10): 2710-2718, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38568556

ABSTRACT

Aimed at the regional open-path detection of benzene (C 6 H 6) in the atmosphere, a power-modulated integrated path differential absorption (PM-IPDA) lidar is introduced and demonstrated. Two tunable interband cascade lasers (ICLs) with about 3.2 µm wavelength are utilized to generate the required PM optical signal. These two operation central wavelengths (CWs) of the PM-IPDA lidar are, respectively, 3236.6 and 3187.1 nm, which can mitigate the influence of significant gases such as H 2 O, C H 4, and HCl on the detection performance. In this work, the fast Fourier transform algorithm is used to retrieve the measured values with the time resolution of 0.1 s corresponding to 104 sampling bins at the sampling rate of 100 kSps/s. The modulated frequency of the PM-IPDA lidar is selected as 10 kHz by laboratory experiments. The slow fluctuation characteristic of the benzene absorption spectrum within the vicinity region of 3.2 µm reduces the impact of small wavelength fluctuations on the performance of PM-IPDA lidar, although a scheme modulated only the driving current causes wavelength fluctuations of ∼±0.2n m. These laboratory experiments also indicate the PM-IPDA lidar can reduce the error resulting from 1/f noise. Open-path observation experiments show that the detection limit is about 0.60m g⋅m -3 and that the PM-IPDA lidar can be used for the regional open-path real-time detection of benzene.

2.
J Thorac Dis ; 15(10): 5414-5427, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37969261

ABSTRACT

Background: Atrial fibrosis caused by long-term atrial fibrillation influences the outcomes of clinical treatment. An improved understanding of the mechanisms underlying atrial fibrillation may reveal new therapeutic targets. This study was conducted to analyze the changes in protein levels in the atrial tissue of a rat model of atrial fibrillation based on proteome sequencing. Methods: Sprague-Dawley rats were used to develop a model of atrial fibrillation induced by chronic intermittent hypoxia (CIH). Histopathological changes were detected using hematoxylin and eosin staining and Masson's staining, and immunohistochemistry and western blotting for the levels of fibrosis biomarkers. Atrial fibrosis tissue samples were also evaluated by proteome sequencing. Differentially expressed proteins (DEPs) between the CIH and control groups were evaluated in functional assay. The expression levels of several key proteins were validated using western blotting. Results: CIH resulted in atrial fibrosis and induced atrial fibrillation. We identified 145 DEPs between the CIH and control groups. These included Myh7, Myl2, Myl3, and Atpla3, which are involved in signaling pathways related to hypertrophic cardiomyopathy, glycerolipid metabolism, and cardiac muscle contraction. Western blotting revealed the upregulation of Myh7, Myl2, and Myl3 and the downregulation of Atpla3 in the CIH group compared with the control group. These results were consistent with the sequencing results. Conclusions: Myh7, Myl2, Myl3, and Atpla3 may play key roles in the progression of atrial fibrillation through their involvement in cardiovascular-disease-related signaling pathways.

3.
Ann Med ; 55(1): 2234934, 2023 12.
Article in English | MEDLINE | ID: mdl-37452706

ABSTRACT

BACKGROUND: This study aimed to identify the appropriate signature veins for the right adrenal gland using a 3D model fused with adrenal venography images and to verify their accuracy through the selectivity index (SI) >2. METHODS: We analyzed the right adrenal venography images of 41 patients who underwent adrenal venous sampling (AVS). These images were merged with a 3D structure of the adrenal gland to identify the signature veins of the right adrenal gland. We then used the signature veins observed during adrenal venography to determine the optimal position of the catheter tip during AVS for 53 other patients. Finally, we verified the accuracy of this method according to the SI. RESULTS: We successfully fused the 3D models of 41 cases with adrenal venography images. We identified the trunk branch type as the major venous morphology in the right anterior oblique at degrees of 30 (38 cases, 92.7%). In addition, the central vein, brush vein, uvula vein, and capsular vein were identified as signature veins for the right AVS. The accuracy of AVS was 100% in the other 53 patients, as verified by an SI >2. CONCLUSIONS: Our study identified the right adrenal signature veins, including the previously overlooked uvula vein, which can be used to determine the position of the catheter tip and improve the success rate of AVS.


The majority of the venography types observed in patients in the right anterior oblique at 30 degrees during adrenal venography were trunk branch types, while irregular or hollow triangle types were infrequent.The signature veins identified during right adrenal venous sampling were the central vein, brush vein, uvula vein, and capsular vein.The right adrenal signature veins, particularly the uvula vein, which has not been given much attention in the past, can serve as a reference to verify the position of the catheter tip and enhance the success rate of adrenal venous sampling.


Subject(s)
Hyperaldosteronism , Humans , Hyperaldosteronism/diagnosis , Catheterization/methods , Adrenal Glands/diagnostic imaging , Adrenal Glands/blood supply , Phlebography/methods , Retrospective Studies , Aldosterone
4.
Pharmacol Res ; 192: 106791, 2023 06.
Article in English | MEDLINE | ID: mdl-37156450

ABSTRACT

Endothelial dysfunction is a key proponent of pathophysiological process of traumatic brain injury (TBI). We previously demonstrated that extracellular vesicles (EVs) released from injured brains led to endothelial barrier disruption and vascular leakage. However, the molecular mechanisms of this EV-induced endothelial dysfunction (endotheliopathy) remain unclear. Here, we enriched plasma EVs from TBI patients (TEVs), and detected high mobility group box 1 (HMGB1) exposure to 50.33 ± 10.17% of TEVs and the number of HMGB1+TEVs correlated with injury severity. We then investigated for the first time the impact of TEVs on endothelial function using adoptive transfer models. We found that TEVs induced dysfunction of cultured human umbilical vein endothelial cells and mediated endothelial dysfunction in both normal and TBI mice, which were propagated through the HMGB1-activated receptor for advanced glycation end products (RAGE)/Cathepsin B signaling, and the resultant NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation and canonical caspase-1/gasdermin D (GSDMD)-dependent pyroptosis. Finally, von Willebrand factor (VWF) was detected on the surface of 77.01 ± 7.51% of HMGB1+TEVs. The TEV-mediated endotheliopathy was reversed by a polyclonal VWF antibody, indicating that VWF might serve a coupling factor that tethered TEVs to ECs, thus facilitating HMGB1-induced endotheliopathy. These results suggest that circulating EVs isolated from patients with TBI alone are sufficient to induce endothelial dysfunction and contribute to secondary brain injury that are dependent on immunologically active HMGB1 exposed on their surface. This finding provided new insight for the development of potential therapeutic targets and diagnostic biomarkers for TBI.


Subject(s)
Brain Injuries, Traumatic , Extracellular Vesicles , HMGB1 Protein , Vascular Diseases , Humans , Mice , Animals , von Willebrand Factor , Brain Injuries, Traumatic/complications , Human Umbilical Vein Endothelial Cells
5.
Math Biosci Eng ; 20(3): 5413-5429, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36896551

ABSTRACT

BACKGROUND: Cardiac fibrosis has gradually gained significance in the field of cardiovascular disease; however, its specific pathogenesis remains unclear. This study aims to establish the regulatory networks based on whole-transcriptome RNA sequencing analyses and reveal the underlying mechanisms of cardiac fibrosis. METHODS: An experimental model of myocardial fibrosis was induced using the chronic intermittent hypoxia (CIH) method. Expression profiles of long non-coding RNA (lncRNA), microRNA (miRNA), and messenger RNA (mRNA) were acquired from right atrial tissue samples of rats. Differentially expressed RNAs (DERs) were identified, and functional enrichment analysis was performed. Moreover, a protein-protein interaction (PPI) network and competitive endogenous RNA (ceRNA) regulatory network that are related to cardiac fibrosis were constructed, and the relevant regulatory factors and functional pathways were identified. Finally, the crucial regulators were validated using qRT-PCR. RESULTS: DERs, including 268 lncRNAs, 20 miRNAs, and 436 mRNAs, were screened. Further, 18 relevant biological processes, such as "chromosome segregation, " and 6 KEGG signaling pathways, such as "cell cycle, " were significantly enriched. The regulatory relationship of miRNA-mRNA-KEGG pathways showed eight overlapping disease pathways, including "pathways in cancer." In addition, crucial regulatory factors, such as Arnt2, WNT2B, GNG7, LOC100909750, Cyp1a1, E2F1, BIRC5, and LPAR4, were identified and verified to be closely related to cardiac fibrosis. CONCLUSION: This study identified the crucial regulators and related functional pathways in cardiac fibrosis by integrating the whole transcriptome analysis in rats, which might provide novel insights into the pathogenesis of cardiac fibrosis.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Rats , Animals , Gene Regulatory Networks , Gene Expression Profiling , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Fibrosis
6.
Infect Dis Ther ; 11(5): 1999-2015, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36058990

ABSTRACT

INTRODUCTION: AOD01 is a novel, fully human immunoglobulin (Ig) G1 neutralizing monoclonal antibody that was developed as a therapeutic against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2). This first-in-human study assessed safety, tolerability, pharmacokinetics (PK), and pharmacodynamics of AOD01 in healthy volunteers. METHODS: Intravenous doses of AOD01 were evaluated in escalating cohorts [four single-dose cohorts (2, 5, 10, and 20 mg/kg) and one two-dose cohort (two doses of 20 mg/kg, 24 h apart)]. RESULTS: Twenty-three subjects were randomized to receive AOD01 or a placebo in blinded fashion. A total of 34 treatment-emergent adverse events (TEAEs) were reported; all were mild in severity. Related events (headache and diarrhea) were reported in one subject each. No event of infusion reactions, serious adverse event (SAE), or discontinuation due to AE were reported. The changes in laboratory parameters, vital signs, and electrocardiograms were minimal. Dose-related exposure was seen from doses 2 to 20 mg/kg as confirmed by Cmax and AUC0-tlast. The median Tmax was 1.5-3 h. Clearance was dose independent. Study results revealed long half-lives (163-465 h). Antidrug antibodies (ADA) to AOD01 were not detected among subjects, except in one subject of the two-dose cohort on day 92. Sustained ex vivo neutralization of SARS-CoV-2 was recorded until day 29 with single doses from 2 to 20 mg/kg and until day 43 with two doses of 20 mg/kg. CONCLUSIONS: AOD01 was safe and well tolerated, demonstrated dose-related PK, non-immunogenic status, and sustained ex vivo neutralization of SARS-CoV-2 after single intravenous dose ranging from 2 to 20 mg/kg and two doses of 20 mg/kg and show good potential for treatment of SARS-CoV-2 infection. (Health Sciences Authority identifier number CTA2000119).

7.
J Interv Card Electrophysiol ; 63(3): 723-736, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35124782

ABSTRACT

PURPOSE: Dysregulation of long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) plays important roles in atrial fibrillation (AF). This study aimed to identify crucial lncRNAs, miRNAs, and mRNAs in AF based on whole transcriptome sequencing. METHODS: Thirty Sprague Dawley rats were randomly stratified into control and chronic intermittent hypoxia (CIH) groups (n = 15 in each). Hematoxylin-eosin staining, Masson staining, immunohistochemical assay, and western blotting were used to evaluate this model. Thereafter, atrial tissues were sent for whole transcriptome sequencing. Finally, fibrosis-related competing endogenous RNA (ceRNA) regulatory networks were built, and the relative levels of lncRNAs, miRNAs, and mRNAs were validated by real-time quantitative polymerase chain reaction (RT-qPCR) or western blotting. RESULTS: A CIH-induced atrial fibrosis rat model was successfully constructed. After sequencing, 268 differentially expressed lncRNAs (DELs), 20 differentially expressed miRNAs (DEMs), and 436 differentially expressed genes (DEGs) were identified. Functional analyses showed that these DEGs were associated with several processes and pathways, including "cell division," "IL-17 signaling pathway," "NOD-like receptor signaling pathway," and "cell adhesion molecules." Fibrosis-related ceRNA networks were then built, comprising five lncRNAs, seven miRNAs, and 19 DEGs. RT-qPCR and western blotting results showed that the patterns of lncRNAs (NONRATT016396.2, NONRATT001596.2, NONRATT011456.2), miRNAs (miR-10b-3p, miR-29b-3p), and mRNAs (Gng7 and Wnt2b) were consistent with sequencing analyses. CONCLUSIONS: The DELs, DEMs, and DEGs identified in this study may participate in atrial fibrosis processes, and the occurrence and progression of AF.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Animals , Fibrosis , Gene Regulatory Networks , Humans , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Exome Sequencing
8.
J Immunother Cancer ; 10(2)2022 02.
Article in English | MEDLINE | ID: mdl-35131861

ABSTRACT

BACKGROUND: Despite significant progress in cancer immunotherapy in recent years, resistance to existing immune checkpoint therapies (ICT) is common. V-domain Ig suppressor of T cell activation (VISTA), a predominantly myeloid immune checkpoint regulator, represents a promising therapeutic target due to its role in suppressing proinflammatory antitumor responses in myeloid-enriched tumor microenvironments. However, uncertainty around the cognate VISTA ligand has made the development of effective anti-VISTA antibodies challenging. The expression of VISTA on normal immune cell subtypes argues for a neutralizing non-depleting antibody, however, previous reported anti-VISTA antibodies use IgG1 Fc isotypes that deplete VISTA+ cells by antibody dependent cellular cytotoxicity/complement dependent cytotoxicity and these antibodies have shown fast serum clearance and immune toxicities. METHOD: Here we used a rational antibody discovery approach to develop the first Fc-independent anti-VISTA antibody, HMBD-002, that binds a computationally predicted functional epitope within the C-C-loop, distinct from other known anti-VISTA antibodies. This epitope is species-conserved allowing robust in vitro and in vivo testing of HMBD-002 in human and murine models of immune activation and cancer including humanized mouse models. RESULTS: We demonstrate here that blockade by HMBD-002 inhibits VISTA binding to potential partners, including V-Set and Immunoglobulin domain containing 3, to reduce myeloid-derived suppression of T cell activity and prevent neutrophil migration. Analysis of immune cell milieu suggests that HMBD-002 treatment stimulates a proinflammatory phenotype characterized by a Th1/Th17 response, recapitulating a phenotype previously noted in VISTA knockout models. This mechanism of action is further supported by immune-competent syngenic and humanized mouse models of colorectal, breast and lung cancer where neutralizing VISTA, without depleting VISTA expressing cells, significantly inhibited tumor growth while decreasing infiltration of suppressive myeloid cells and increasing T cell activity. Finally, we did not observe either the fast serum clearance or immune toxicities that have been reported for IgG1 antibodies. CONCLUSION: In conclusion, we have shown that VISTA-induced immune suppression can be reversed by blockade of the functional C-C' loop region of VISTA with a first-in-class rationally targeted and non-depleting IgG4 isotype anti-VISTA antibody, HMBD-002. This antibody represents a highly promising novel therapy in the VISTA-suppressed ICT non-responder population.


Subject(s)
Immunosuppression Therapy/methods , Lymphocyte Activation/immunology , Neoplasms/immunology , Receptors, Fc/metabolism , Animals , Disease Models, Animal , Humans , Mice , Tumor Microenvironment
9.
Food Funct ; 12(20): 9719-9738, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34664590

ABSTRACT

Obesity is a global chronic disease epidemic that is attributed to the abnormal accumulation of lipids in adipose tissue. Astaxanthin (AST) from Haematococcus pluvialis, a natural carotenoid, exhibits antioxidant, anti-lipogenic, anti-diabetic and other potent effects. Herein, we evaluated the effect of AST to illuminate its efficacy and mechanisms in high-fat diet-fed mice. AST supplementation not only significantly decreased body weight and lipid droplet accumulation in the liver but also modulated liver function and serum lipid levels. Lipidomic analysis revealed that 13 lipids might be potential biomarkers responsible for the effects of AST in lipid reduction, such as total free fatty acids (FFAs), triacylglycerols (TGs) and cholesterol esters (CEs). The gut microbiota sequencing results indicated that AST alleviated HFD-induced gut microbiota dysbiosis by optimizing the ratio of Firmicutes to Bacteroides and inhibiting the abundance of obesity-related pathogenic microbiota while promoting the abundance of probiotics related to glucose and lipid metabolism. In addition, qRT-PCR demonstrated that AST could regulate the gene expressions of the AMPK/SREBP1c pathway by downregulating lipogenesis correlated-genes and upregulating the lipid oxidant related-gene. The present study revealed the new function of AST in regulating lipid metabolism, which provided a theoretical basis for the development of high-quality AST functional food and the application of diet active substances in obesity, as demonstrated in mice.


Subject(s)
Chlorophyta , Lipid Metabolism/drug effects , Animals , Diet, High-Fat , Dysbiosis/prevention & control , Gastrointestinal Microbiome/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/prevention & control , Xanthophylls/pharmacology
10.
PLoS One ; 16(6): e0253487, 2021.
Article in English | MEDLINE | ID: mdl-34161386

ABSTRACT

Although SARS-CoV-2-neutralizing antibodies are promising therapeutics against COVID-19, little is known about their mechanism(s) of action or effective dosing windows. We report the generation and development of SC31, a potent SARS-CoV-2 neutralizing antibody, isolated from a convalescent patient. Antibody-mediated neutralization occurs via an epitope within the receptor-binding domain of the SARS-CoV-2 Spike protein. SC31 exhibited potent anti-SARS-CoV-2 activities in multiple animal models. In SARS-CoV-2 infected K18-human ACE2 transgenic mice, treatment with SC31 greatly reduced viral loads and attenuated pro-inflammatory responses linked to the severity of COVID-19. Importantly, a comparison of the efficacies of SC31 and its Fc-null LALA variant revealed that the optimal therapeutic efficacy of SC31 requires Fc-mediated effector functions that promote IFNγ-driven anti-viral immune responses, in addition to its neutralization ability. A dose-dependent efficacy of SC31 was observed down to 5mg/kg when administered before viral-induced lung inflammatory responses. In addition, antibody-dependent enhancement was not observed even when infected mice were treated with SC31 at sub-therapeutic doses. In SARS-CoV-2-infected hamsters, SC31 treatment significantly prevented weight loss, reduced viral loads, and attenuated the histopathology of the lungs. In rhesus macaques, the therapeutic potential of SC31 was evidenced through the reduction of viral loads in both upper and lower respiratory tracts to undetectable levels. Together, the results of our preclinical studies demonstrated the therapeutic efficacy of SC31 in three different models and its potential as a COVID-19 therapeutic candidate.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , COVID-19/therapy , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2/genetics , Animals , Antibodies, Neutralizing/metabolism , COVID-19/immunology , COVID-19/virology , Chemokines/blood , Chemokines/genetics , Chlorocebus aethiops , Convalescence , Cricetinae , Cytokines/blood , Cytokines/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Macaca mulatta , Male , Mice, Transgenic , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells , Viral Load
11.
Eur J Pharm Biopharm ; 147: 87-101, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31899369

ABSTRACT

In recent decades, targeted drug delivery systems (TDDS) have been widely used as an ideal method of improving therapeutic effects and reducing systemic side effects of chemotherapeutic agents. Historically, a handful of methods have been developed to further improve the targeting ability of delivery systems. Thus, in this study, two methods, taking advantage of tumor characteristics, were used for the creation of a multi-targeted delivery system. The first was the fabrication of pH-sensitive micelles, lending the ability to increase drug release by exploiting the acidic tumor environment. The second method was through utilization of the surface-exposed phosphatidylserine (PS) of tumors, which is normally found in the inner leaflet in healthy cells. Using PS as a target site, PS binding peptide (PSBP-6) was conjugated to pH-sensitive mixed micelles, (consisting of poly (ethylene glycol)-b-poly (D, L-lactide) (PEG-PDLLA) and poly (ethylene glycol)-b-poly (L-histidine) (PEG-PHIS)). After successful preparation of micelles, paclitaxel (PTX), a common chemotherapeutic agent, was selected to measure drug loading capacity and encapsulation efficiency, showing 7.9% and 83.5%, respectively. The in vitro release of PTX from mixed micelles at pH 5.0, 6.5, and 7.4 was 78.1, 56.8, and 51.4%, respectively, indicating acid-triggered drug release. The PSBP-6-modified, mixed micelles exhibited significantly enhanced in vitro cytotoxicity and demonstrated more efficient cellular uptake compared to unmodified mixed micelles in the HeLa cell line. Moreover, pharmacokinetic, in vivo biodistribution, and fluorescence imaging studies showed that PSBP-6-PEG-PDLLA/PEG-PHIS mixed micelles provide prolonged time in blood circulation and enhanced tumor accumulation. These results suggest that the use of PS as a novel targeting site is advantageous, and that these new multi-targeted mixed micelles show great potential for realization of broad prospects in the targeted treatment of tumors for chemotherapeutic delivery.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Drug Delivery Systems , Paclitaxel/administration & dosage , Phosphatidylserines/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Cell Line, Tumor , Drug Liberation , Female , HeLa Cells , Humans , Hydrogen-Ion Concentration , Mice , Micelles , Paclitaxel/pharmacokinetics , Peptides/chemistry , Polymers/chemistry , Rats , Rats, Sprague-Dawley , Tissue Distribution , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
Mol Cancer Ther ; 19(2): 490-501, 2020 02.
Article in English | MEDLINE | ID: mdl-31911530

ABSTRACT

In recent years, HER3 has increasingly been implicated in the progression of a variety of tumor types and in acquired resistance to EGFR and HER2 therapies. Whereas EGFR and HER2 primarily signal through the MAPK pathway, HER3, as a heterodimer with EGFR or HER2, potently activates the PI3K pathway. Despite its critical role, previous attempts to target HER3 with neutralizing antibodies have shown disappointing efficacy in the clinic, most likely due to suboptimal and indirect mechanisms of action that fail to completely block heterodimerization; for example, tumors can escape inhibition of ligand binding by upregulating ligand-independent mechanisms of HER3 activation. We therefore developed 10D1F, a picomolar affinity, highly specific anti-HER3 neutralizing antibody that binds the HER3 heterodimerization interface, a region that was hitherto challenging to raise antibodies against. We demonstrate that 10D1F potently inhibits both EGFR:HER3 and HER2:HER3 heterodimerization to durably suppress activation of the PI3K pathway in a broad panel of tumor models. Even as a monotherapy, 10D1F shows superior inhibition of tumor growth in the same cell lines both in vitro and in mouse xenograft experiments, when compared with other classes of anti-HER3 antibodies. This includes models demonstrating ligand-independent activation of heterodimerization as well as constitutively activating mutations in the MAPK pathway. Possessing favorable pharmacokinetic and toxicologic profiles, 10D1F uniquely represents a new class of anti-HER3 neutralizing antibodies with a novel mechanism of action that offers significant potential for broad clinical benefit.10D1F is a novel anti-HER3 antibody that uniquely binds the receptor dimerization interface to block ligand-dependent and independent heterodimerization with EGFR/HER2 and thus more potently inhibits tumor growth than existing anti-HER3 antibodies.


Subject(s)
Immunoglobulin G/pharmacology , Neoplasms/therapy , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/immunology , Animals , Epitopes/immunology , Female , Humans , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
13.
Curr Drug Metab ; 20(10): 815-834, 2019.
Article in English | MEDLINE | ID: mdl-31580248

ABSTRACT

BACKGROUND: Target-homing drug delivery systems are now gaining significant attention for use as novel therapeutic approaches in antitumor targeting for cancer therapy. Numerous targeted drug delivery systems have been designed to improve the targeting effects because these systems can display a range of favorable properties, thus, providing suitable characteristics for clinical applicability of anticancer drugs, such as increasing the solubility, and improving the drug distribution at target sites. The majority of these targeting systems are designed with respect to differences between cancerous and normal tissues, for instance, the low pH of tumor tissues or overexpressed receptors on tumor cell membranes. Due to the growing number of targeting possibilities, it is important to know the tumor-specific recognition strategies for designing novel, targeted, drug delivery systems. Herein, we identify and summarize literature pertaining to various recognition sites for optimizing the design of targeted drug delivery systems to augment current chemotherapeutic approaches. OBJECTIVE: This review focuses on the identification of the recognition sites for developing targeted drug delivery systems for use in cancer therapeutics. METHODS: We have reviewed and compiled cancer-specific recognition sites and their abnormal characteristics within tumor tissues (low pH, high glutathione, targetable receptors, etc.), tumor cells (receptor overexpression or tumor cell membrane changes) and tumor cell organelles (nuclear and endoplasmic reticular dysregulation) utilizing existing scientific literature. Moreover, we have highlighted the design of some targeted drug delivery systems that can be used as homing tools for these recognition sites. RESULTS AND CONCLUSION: Targeted drug delivery systems are a promising therapeutic approach for tumor chemotherapy. Additional research focused on finding novel recognition sites, and subsequent development of targeting moieties for use with drug delivery systems will aid in the evaluation and clinical application of new and improved chemotherapeutics.


Subject(s)
Drug Delivery Systems , Molecular Targeted Therapy , Neoplasms/drug therapy , Animals , Humans , Neoplasms/metabolism , Organelles
14.
Opt Express ; 27(16): 22855-22867, 2019 Aug 05.
Article in English | MEDLINE | ID: mdl-31510570

ABSTRACT

We present a scheme for the electromagnetically induced transparency (EIT)-like nonlinear ground-state cooling in a double-cavity optomechanical system in which an optical cavity mode is coupled parametrically to the square of the position of a mechanical oscillator, an additional auxiliary cavity is coupled to the optomechanical cavity. The optimum cooling conditions is derived, based on which the heating process can be well suppressed and the mechanical resonator can be cooled with an optimal effect to near its ground state through EIT-like cooling mechanism even in unresolved sideband regime. It is demonstrated by numerical simulations that not only the average phonon number of steady state is lower than that of single-cavity optomechanical system, but also the cooling rate is greatly faster than that of the linear optomechanical coupling due to the two-phonon cooling process in the quadratic coupling. Also, the ground-state cooling is achievable even with a relatively weak quadratic coupling strengthby tunning the coupling between two cavities to reach the optimum cooling conditions, thus provides an solution for overcoming the limitations of weak quadratic coupling rate in experiments. The proposed approach provides a platform for quantum manipulation of macroscopic mechanical devices beyond the resolved sideband limit and linear coupling regime.

15.
FEBS Lett ; 592(1): 112-121, 2018 01.
Article in English | MEDLINE | ID: mdl-29237230

ABSTRACT

Focal adhesion (FA) proteins, kindlin-2 and integrin-linked kinase (ILK), regulate cell adhesion and migration. ILK interacts with and promotes kindlin-2 targeting to FAs. Leu353 and Leu357 in kindlin-2 have been reported to be important for the interaction between kindlin-2 and ILK. However, the binding interface between kindlin-2 and ILK remains unclear. Using molecular modeling and molecular dynamics simulations, we show that Asp344, Asp352, and Thr356 in kindlin-2 and Arg243 and Arg334 in ILK kinase domain (KD) are important in kindlin-2/ILK complex formation. Mutations that disrupt these interactions abrogate kindlin-2 and ILK colocalization in HeLa cells. The interactions are direct based on data from pull-down assays using purified recombinant kindlin-2 F2-pleckstrin homology and ILK KDs. These data provide additional insights into the binding interface between kindlin-2 and ILK.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Movement/genetics , Cell Movement/physiology , HeLa Cells , Humans , Membrane Proteins/genetics , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Neoplasm Proteins/genetics , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity
16.
Biochem Biophys Rep ; 7: 214-217, 2016 Sep.
Article in English | MEDLINE | ID: mdl-28955909

ABSTRACT

We showed that the αLß2 integrin with the non-functional mutation G150D cannot be induced with Mg/EGTA to express the mAb KIM127 epitope, which reports the leg-extended conformation. We extended the study to the αIIbß3, an integrin without an αI domain. The equivalent mutation, i.e. G161D, also resulted in an expressible, but non-adhesive αIIbß3 integrin. An NMR study of synthetic peptides spanning the α1-α1' helix of the ß3 I domain shows that both wild-type and mutant peptides are α-helical. However, whereas in the wild-type peptide this helix is continuous, the mutant presents a discontinuity, or kink, precisely at the site of mutation G161D. Our results suggest that the mutation may lock integrin heterodimers in a bent conformation that prevents integrin activation via conformational extension.

17.
Biochem Biophys Res Commun ; 458(2): 251-5, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25640842

ABSTRACT

We have shown that Mg/EGTA (5 mM Mg(2+) and 1.5 mM EGTA) could effectively promote the adhesion of integrin αLß2 to its ligand ICAM-1 but could not promote that of the αMß2 to denatured BSA. In order to determine the structural differences between αL and αM that specifically contribute to Mg/EGTA sensitivity, a series of αL/αM chimeras were constructed. Our results showed that αLß2 with αM calf-1 domain completely lost the response to Mg/EGTA activation. In the reverse experiment, αMß2 would require the presence of both the αL calf-1 and calf-2 domain to initiate the Mg/EGTA sensitivity.


Subject(s)
Cell Adhesion/physiology , Egtazic Acid/chemistry , Egtazic Acid/metabolism , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Magnesium/chemistry , Magnesium/metabolism , Binding Sites , HEK293 Cells , Humans , Protein Binding , Structure-Activity Relationship
18.
Blood Cells Mol Dis ; 54(2): 177-82, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25514840

ABSTRACT

Leukocyte adhesion deficiency 1 (LAD-1) is caused by defects in the ß2 integrin subunit. We studied 18 missense mutations, 14 of which fail to support the surface expression of the ß2 integrins. Integrins with the ß2-G150D mutation fail to bind ligands, possibly due to the failure of the α1 segment of the ßI domain to assume an α-helical structure. Integrins with the ß2-G716A mutation are not maintained in their resting states, and the patient has the severe phenotype of LAD-1. The ß2-S453N and ß2-P648L mutants support the expression of integrins and adhesion functions. They should be re-classified as polymorphic variants.


Subject(s)
CD18 Antigens/chemistry , Mutation, Missense , Protein Subunits/chemistry , Amino Acid Sequence , Amino Acid Substitution , CD18 Antigens/genetics , CD18 Antigens/metabolism , Cell Adhesion , Gene Expression , HEK293 Cells , Humans , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/pathology , Leukocytes/metabolism , Leukocytes/pathology , Models, Molecular , Molecular Sequence Data , Plasmids/chemistry , Plasmids/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Transfection
19.
Drug Dev Res ; 75(8): 489-96, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25452110

ABSTRACT

Preclinical Research The abnormal migration and proliferation of vascular smooth muscle cells (VSMCs) plays a pivotal role in the development of neointimal hyperplasia after vascular injury. Nobiletin, a citrus bioflavonoid, exhibits anti-inflammatory and anti-oxidative activities. The present study evalutaed whether nobiletin could inhibit platelet-derived growth factor (PDGF)-BB- stimulated VSMC proliferation and migration and decrease neointimal hyperplasia in a rat carotid artery injury model. Cultured VSMCs from rat thoracic aortas were treated with nobiletin before being stimulated with 20 ng/ml PDGF-BB, and rats were subjected to carotid artery injury. Nobiletin inhibited PDGF-BB-induced VSMC proliferation and migration, attenuated reactive oxygen species (ROS) production and reduced phosphorylation of ERK1/2 and the expression of nuclear NF-κB p65 in PDGF-BB-stimulated VSMCs. Nobiletin decreased the intima area and the ratio of neointima to media in balloon-injured rat carotid arteries. Serum levels of TNF-α and IL-6 in nobiletin-treated rats were decreased. These results indicated that nobiletin could be a potential protective agent for the prevention and treatment of restenosis after angioplasty.


Subject(s)
Antioxidants/administration & dosage , Aorta, Thoracic/injuries , Carotid Artery Injuries/drug therapy , Flavones/administration & dosage , Muscle, Smooth, Vascular/drug effects , Neointima/prevention & control , Animals , Antioxidants/pharmacology , Aorta, Thoracic/metabolism , Carotid Artery Injuries/etiology , Carotid Artery Injuries/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Flavones/pharmacology , Gene Expression Regulation/drug effects , Male , Muscle, Smooth, Vascular/cytology , Neointima/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...