Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Nat Commun ; 15(1): 3263, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38627393

RESUMEN

Gouty arthritis evokes joint pain and inflammation. Mechanisms driving gout pain and inflammation remain incompletely understood. Here we show that CXCL5 activates CXCR2 expressed on nociceptive sensory neurons to drive gout pain and inflammation. CXCL5 expression was increased in ankle joints of gout arthritis model mice, whereas CXCR2 showed expression in joint-innervating sensory neurons. CXCL5 activates CXCR2 expressed on nociceptive sensory neurons to trigger TRPA1 activation, resulting in hyperexcitability and pain. Neuronal CXCR2 coordinates with neutrophilic CXCR2 to contribute to CXCL5-induced neutrophil chemotaxis via triggering CGRP- and substance P-mediated vasodilation and plasma extravasation. Neuronal Cxcr2 deletion ameliorates joint pain, neutrophil infiltration and gait impairment in model mice. We confirmed CXCR2 expression in human dorsal root ganglion neurons and CXCL5 level upregulation in serum from male patients with gouty arthritis. Our study demonstrates CXCL5-neuronal CXCR2-TRPA1 axis contributes to gouty arthritis pain, neutrophil influx and inflammation that expands our knowledge of immunomodulation capability of nociceptive sensory neurons.


Asunto(s)
Artritis Gotosa , Animales , Humanos , Masculino , Ratones , Artralgia , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Inflamación , Nocicepción , Nociceptores/metabolismo , Dolor
2.
Micromachines (Basel) ; 15(4)2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38675248

RESUMEN

The rapid and sensitive detection of foodborne pathogens is crucial for ensuring food safety. Among virus testing methods, polymerase chain reaction (PCR) has served as the gold-standard technique in most food safety regulation organizations. However, to enhance the speed and efficiency of PCR, novel approaches are continually being explored. In this work, leveraging the photothermal effects and high thermal conductivity of gold nanoparticles, we have significantly improved the heating and cooling rates of thermal cycles, enabling ultra-fast PCR detection. Specifically, we present a pre-degassing multiplex digital PCR chip integrated with gold nanoparticles. We further developed a portable system with a light source for photothermal heating cycling, along with an optoelectronic sensor to analyze PCR amplification products after rapid thermal cycling. As proof of concept, the proposed chip and portable device was applied for the on-site detection of several types of foodborne pathogens, including Escherichia coli, Listeria monocytogenes, Staphylococcus aureus, and Salmonella. The whole system could distinguish those pathogens within 20 min, showing good potential for the rapid detection of multiple types of foodborne pathogens.

3.
J Agric Food Chem ; 72(9): 4689-4702, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38382537

RESUMEN

Plant metabolites from natural product extracts offer unique advantages against carcinogenesis in the development of drugs. The target-based virtual screening from food-derived compounds represents a promising approach for tumor therapy. In this study, we performed virtual screening to target the presumed inhibitor-binding pocket and identified a highly potent Kv10.1 inhibitor, liensinine (Lien), which can inhibit the channel in a dose-dependent way with an IC50 of 0.24 ± 0.07 µM. Combining molecular dynamics simulations with mutagenesis experiments, our data show that Lien interacts with Kv10.1 by binding with Y539, T543, D551, E553, and H601 in the C-linker domain of Kv10.1. In addition, the interaction of sequence alignment and 3D structural modeling revealed differences between the C-linker domain of the Kv10.1 channel and the Kv11.1 channel. Furthermore, antitumor experiments revealed that Lien suppresses the proliferation and migration of HCC both in vitro and in vivo. In summary, the food-derived compound, Lien, may serve as a lead compound for antihepatoma therapeutic drugs targeting Kv10.1.


Asunto(s)
Carcinoma Hepatocelular , Isoquinolinas , Neoplasias Hepáticas , Fenoles , Humanos , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Carcinogénesis/metabolismo
4.
Eur J Pharmacol ; 962: 176240, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38048981

RESUMEN

Ether-à-go-go (EAG) potassium channels play a crucial role in the regulation of neuronal excitability and cancer progression, rendering them potential drug targets for cancer therapy. However, the scarcity of information regarding the selection sites on hEAG1 has posed a challenge in the discovery of new hEAG1 inhibitors. In this study, we introduced a novel natural product, corydaline, which selectively inhibits the hEAG1 channel without sensitivity to other KCNH channels. The IC50 of corydaline for the hEAG1 channel was 11.3 ± 0.6 µM, whereas the IC50 for hEAG2 and hERG1 were 73.6 ± 9.9 µM and 111.4 ± 8.5 µM, respectively. Molecular dynamics simulations together with site-directed mutagenesis, have unveiled that the site corydaline forms interactions with Lys217, Phe273, Pro276, Trp295 and Arg366, situated within the intracellular transmembrane segments S1-S4 of the voltage-sensor domain, be considered a novel drug pocket for hEAG1. Additionally, the intergaration of sequence alignment and 3D structural modeling revealed differences between the voltage sensor domain of hEAG1 channel and other EAG channels, suggesting the feasibility of a VSD modulation approach that could potentially lead to the selective inhibition of hEAG1 channels. Furthermore, antitumor experiments demonstrated that corydaline can inhibit the proliferation and migration of hepatic carcinoma cells by targeting hEAG1. The identification of this novel druggable pocket offers the possibility for drug screening against diseases linked to abnormal hEAG1 channels.


Asunto(s)
Carcinoma , Canales de Potasio Éter-A-Go-Go , Humanos , Supervivencia Celular , Canales de Potasio Éter-A-Go-Go/metabolismo , Línea Celular
5.
Int J Biol Macromol ; 253(Pt 6): 127261, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37802433

RESUMEN

TMEM16A is highly expressed in a variety of tumor cells and is involved in the growth and metastasis of malignancies. It has been established that down-regulation of TMEM16A expression or functional activity can inhibit tumor cells growth. However, there is a lack of targeted inhibitors with high efficiency and low toxicity. Here, we identified a novel inhibitor daidzein from dozens of natural product molecules. Whole-cell patch clamp data indicated that daidzein inhibits TMEM16A channel in a dose-dependent manner, with IC50 of 1.39 ± 0.59 µM. Western blot result showed that daidzein can also reduce the expression of TMEM16A protein in LA795 cells. These results indicated that the inhibitory effects of daidzein exert on TMEM16A in two ways, both inhibiting TMEM16A current and decreasing its protein expression. In addition, the putative binding sites of daidzein on TMEM16A are G608, G628, and K839 through molecular docking. Moreover, daidzein concentration-dependently reduced cell viability and cell migration, causing G1/S cell cycle arrest in vitro. It was also confirmed that daidzein can effectively inhibit the growth of LA795 lung adenocarcinoma cells implanted nude mice in vivo. In conclusion, daidzein can be used as a lead compound for the development of therapeutic drugs for lung adenocarcinoma.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Ratones , Animales , Canales de Cloruro/metabolismo , Simulación del Acoplamiento Molecular , Ratones Desnudos , Adenocarcinoma del Pulmón/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico
6.
Small ; 19(46): e2304880, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37452439

RESUMEN

Porous noble metal nanoparticles have received particular attention recently for their unique optical, thermal, and catalytic functions in biomedicine. However, limited progress has been made to synthesize such porous metallic nanostructures with large mesopores (≥25 nm). Here, a green yet facile synthesis strategy using biocompatible liposomes as templates to mediate the formation of mesoporous metallic nanostructures in a controllable fashion is reported. Various monodispersed nanostructures with well-defined mesoporous shape and large mesopores (≈ 40 nm) are successfully synthesized from mono- (Au, Pd, and Pt), bi- (AuPd, AuPt, AuRh, PtRh, and PdPt), and tri-noble metals (AuPdRh, AuPtRh, and AuPdPt). Along with a successful demonstration of its effectiveness in synthesis of various mesoporous nanostructures, the possible mechanism of liposome-guided formation of such nanostructures via time sectioning of the synthesis process (monitoring time-resolved growth of mesoporous structures) and computational quantum molecular modeling (analyzing chemical interaction energy between metallic cations and liposomes at the enthalpy level) is also revealed. These mesoporous metallic nanostructures exhibit a strong photothermal effect in the near-infrared region, effective catalytic activities in hydrogen peroxide decomposition reaction, and high drug loading capacity. Thus, the liposome-templated method provides an inspiring and robust avenue to synthesize mesoporous noble metal-based nanostructures for versatile biomedical applications.


Asunto(s)
Liposomas , Nanoestructuras , Nanoestructuras/química , Metales/química
7.
Phys Chem Chem Phys ; 25(23): 16114-16125, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37278335

RESUMEN

Exploiting micelles of polyethylene glycol-dipalmitoylglycerophosphoethanolamine (PEG-DPPE) as a drug delivery approach is of great promise for improving therapeutic targeting and the half-lives of drugs. To optimize the micelle carriers, pending issues concerning the kinetics underlying the carrier-membrane interplay and the specific contributions of the micelle hydrophobic/hydrophilic components remain to be addressed. Relying on MARTINI coarse-grain (CG) molecular dynamics simulations, we explored the carrier-membrane fusion dynamics of PEG-DPPE micelles with different PEG repetitions in delivering doxorubicin (DOX). A bilayer model composed of 20% phosphatidylglycerol (POPG) and 80% phosphatidylcholine (POPC) was constructed to mimic anionic cancer cell membranes. The CG model of DOX was pioneeringly constructed herein, and it was found to distribute at the hydrophilic/hydrophobic interface of the PEGylated micelles, in agreement with experimental results. The free DOXs cause insignificant disorder of the membrane organization, whereas the PEG-DPPE micelles encapsulating DOX lead to a remarkable membrane invasion supported by the order parameter of the lipid acyl carbon tails and the membrane permeation free energy of DOX. The carrier-bilayer interaction shows a stepwise form attributed to the rearrangement of the zwitterionic/anionic lipids upon the absorption of the DOX-micelle complex on a membrane locality, which initiates the rapid release of DOX to the bilayer interior. Benefiting from the enhanced micelle-membrane interplay, the PEG1250-DPPE micelles result in severe bilayer breakage and deeper membrane insertion of DOX compared to the PEG2000-DPPE micelles. This study provides new theoretical insights into the mechanism of PEG-DPPE micelles in delivering drugs through membranes, which is of benefit for further optimization of PEGylated delivery systems.


Asunto(s)
Micelas , Polietilenglicoles , Polietilenglicoles/química , Línea Celular Tumoral , Doxorrubicina/química , Portadores de Fármacos/química
8.
J Chem Inf Model ; 63(14): 4423-4432, 2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37382878

RESUMEN

Transmissibility of SARS-CoV-2 initially relies on its trimeric Spike-RBDs to tether the ACE-2 on host cells, and enhanced self-association of ACE-2 engaged with Spike facilitates the viral infection. Two primary packing modes of Spike-ACE2 heteroproteins exist potentially due to discrepant amounts of RBDs loading on ACE-2, but the resultant self-association difference is inherently unclear. We used extensive coarse-grained dynamic simulations to characterize the self-association efficiency, the conformation relevance, and the molecular mechanism of ACE-2 with different RBD amounts. It was revealed that the ACE-2 hanging two/full RBDs (Mode-A) rapidly dimerized into the heteroprotein complex in a compact "linear" conformation, while the bare ACE-2 showed weakened self-association and a protein complex. The RBD-tethered ectodomains of ACE-2 presented a more upright conformation relative to the membrane, and the intermolecular ectodomains were predominantly packed by the neck domains, which was obligated to the rapid protein self-association in a compact pattern. Noted is the fact that the ACE-2 tethered by a single RBD (Mode-B) retained considerable self-association efficiency and clustering capability, which unravels the interrelation of ACE-2 colocalization and protein cross-linkage. The molecular perspectives in this study expound the self-association potency of ACE-2 with different RBD amounts and the viral activity implications, which can greatly enhance our comprehension of SARS-CoV-2 infection details.


Asunto(s)
COVID-19 , Humanos , Análisis por Conglomerados , Dimerización , Simulación de Dinámica Molecular , Unión Proteica , SARS-CoV-2
9.
Phys Chem Chem Phys ; 25(27): 18495, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37377001

RESUMEN

Correction for 'Delivery mechanism of doxorubicin by PEG-DPPE micelles on membrane invasion by dynamic simulations' by Lina Zhao et al., Phys. Chem. Chem. Phys., 2023, 25, 16114-16125, https://doi.org/10.1039/D2CP05946K.

10.
Biophys J ; 122(13): 2675-2685, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37218130

RESUMEN

Association of the cellular adhesive protein CD44 and the N-terminal (FERM) domain of cytoskeleton adaptors is critical for cell proliferation, migration, and signaling. Phosphorylation of the cytoplasmic domain (CTD) of CD44 acts as an important regulator of the protein association, but the structural transformation and dynamics mechanism remain enigmatic. In this study, extensive coarse-grained simulations were employed to explore the molecular details in the formation of CD44-FERM complex under S291 and S325 phosphorylation, a modification path known to exert reciprocal effects on the protein association. We find that phosphorylation of S291 inhibits complexation by causing the CTD of CD44 to adopt a more closed structure. In contrast, S325 phosphorylation liberates the CD44-CTD from the membrane surface and promotes the linkage with FERM. The phosphorylation-driven transformation is found to occur in a PIP2-dependent manner, with PIP2 effecting the relative stability of the closed and open conformation, and a replacement of PIP2 by POPS greatly abrogates this effect. The revealed interdependent regulation mechanism by phosphorylation and PIP2 in the association of CD44 and FERM further strengthens our understanding of the molecular basis of cellular signaling and migration.


Asunto(s)
Citoesqueleto , Proteínas , Transducción de Señal , Conformación Molecular , Unión Proteica
11.
J Biol Chem ; 299(6): 104780, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37142220

RESUMEN

The calcium-activated chloride channel TMEM16A is a potential drug target to treat hypertension, secretory diarrhea, and several cancers. However, all reported TMEM16A structures are either closed or desensitized, and direct inhibition of the open state by drug molecules lacks a reliable structural basis. Therefore, revealing the druggable pocket of TMEM16A exposed in the open state is important for understanding protein-ligand interactions and facilitating rational drug design. Here, we reconstructed the calcium-activated open conformation of TMEM16A using an enhanced sampling algorithm and segmental modeling. Furthermore, we identified an open-state druggable pocket and screened a potent TMEM16A inhibitor, etoposide, which is a derivative of a traditional herbal monomer. Molecular simulations and site-directed mutagenesis showed that etoposide binds to the open state of TMEM16A, thereby blocking the ion conductance pore of the channel. Finally, we demonstrated that etoposide can target TMEM16A to inhibit the proliferation of prostate cancer PC-3 cells. Together, these findings provide a deep understanding of the TMEM16A open state at an atomic level and identify pockets for the design of novel inhibitors with broad applications in chloride channel biology, biophysics, and medicinal chemistry.


Asunto(s)
Anoctamina-1 , Modelos Moleculares , Humanos , Masculino , Anoctamina-1/química , Anoctamina-1/metabolismo , Calcio/metabolismo , Etopósido/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Simulación por Computador
12.
Front Pharmacol ; 14: 1118584, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36937869

RESUMEN

Hand, foot, and mouth disease (HFMD) is a common childhood infectious disease caused by enterovirus (EV) infection. EV71 is one of the major pathogens causing hand, foot, and mouth disease and is more likely to cause exacerbation and death than other enteroviruses. Although a monovalent vaccine for EV71 has been developed, there are no clinically available anti-EV71 specific drugs. Here, we performed virtual screening and biological experiments based on the traditional Chinese medicine monomer library. We identified a traditional Chinese medicine monomer, Salvianolic acid A (SA), a polyphenolic compound isolated from Salvia miltiorrhiza. Salvianolic acid A inhibits EV71 virus infection in a concentration-dependent manner, and its antiviral activity is higher than that of other reported natural polyphenols and has a high biosafety. Furthermore, molecular dynamics simulations showed that salvianolic acid A can anchor to E71, a member of the enzyme catalytic triad, and cause H40 to move away from the catalytic center. Meanwhile, molecular mechanics generalized born surface area (MMGBSA) and steered molecular dynamics (SMD) results showed that the P1 group of SA was most easily unbound to the S1 pocket of 3Cpro, which provided theoretical support to further improve the affinity of salvianolic acid A with 3Cpro. These findings suggest that salvianolic acid A is a novel EV71 3Cpro inhibitor with excellent antiviral activity and is a promising candidate for clinical studies.

13.
Int J Biol Macromol ; 235: 123839, 2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-36842737

RESUMEN

TMEM16A, a Ca2+-activated chloride channel (CaCC), and its pharmacological inhibitors can inhibit the growth of lung adenocarcinoma cells. However,the poor efficacy, safety, and stability of TMEM16A inhibitors limit the development of these agents. Therefore, finding new therapeutic directions from already marketed drugs is a feasible strategy to obtain safe and effective therapeutic drugs. Here, we screened a library contain more than 2400 FDA, EMA, and NMPA-approved drugs through virtual screening. We identified a drug candidate, candesartan (CDST), which showed strong inhibitory effect on the TMEM16A in a concentration-dependent manner with an IC50 of 24.40 ± 3.21 µM. In addition, CDST inhibited proliferation, migration and induced apoptosis of LA795 cells targeting TMEM16A, and significantly inhibited lung adenocarcinoma tumor growth in vivo. The molecular mechanism of CDST inhibiting TMEM16A channel indicated it bound to R515/R535/E623/E624 in the drug pocket, thereby blocked the pore. In conclusion, we identified a novel TMEM16A channel inhibitor, CDST, which exhibited excellent inhibitory activity against lung adenocarcinoma. Considering that CDST has been used in clinical treatment of hypertension, it may play an important role in the combined treatment of hypertension and lung adenocarcinoma as a multi-target drug in the future.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Antihipertensivos/farmacología , Reposicionamiento de Medicamentos , Canales de Cloruro/metabolismo , Adenocarcinoma del Pulmón/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Calcio/metabolismo
14.
Biomed Mater ; 18(2)2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36758244

RESUMEN

The development of novel antibacterial nano-materials with synergistic biological effects has attracted extensive interest of the researchers. In the study, 0.5 mol% Ag and 0.5 mol% Cu co-doped K2Ti6O13(0.5 Ag-0.5 Cu-KTO) nanomaterial was successfully synthesized using two-step method of sol-gel and hydrothermal synthesis. The crystal structure of 0.5 Ag-0.5 Cu-KTO was the same as that of monoclinic K2Ti6O13. Ag ions and Cu ions were uniformly loaded on K2Ti6O13by replacing partial Ti ions, so that these antibacterial ions could be slowly released. High specific surface area of 0.5 Ag-0.5 Cu-KTO (337.6 m2g-1) provided more surface active sites for Ag-Cu doping and adsorption. More negative surface zeta potential (-32.83 mV in phosphate buffer solution and -21.45 mV in physiological saline solution, respectively) would be beneficial to prevent the aggregation of the nanowires in physiological environment. Under the same doping amount, compared to 1.0 mol% Cu doped K2Ti6O13, 0.5 Ag-0.5 Cu-KTO exhibited better antibacterial performance against gram-positive and gram-negative bacteria at only 100 µg ml-1dose concentration, near to 1.0 mol% Ag doped K2Ti6O13(1.0 Ag-KTO). And 0.5 Ag-0.5 Cu-KTO showed more excellent biocompatibility than 1.0 Ag-KTO, which was attribute to the introduction of Cu ions effectively decreasing the hemolytic and cytotoxic risks from Ag ions. As expected, the synthesized 0.5 Ag-0.5 Cu-KTO nanowires demonstrated excellent structural stability, high antibacterial activity, good hemocompatibility and cytocompatibility owing to the synergistic effects of Cu and Ag ions. 0.5 Ag-0.5 Cu-KTO nanowires will be a promising antimicrobial candidate for biomedical applications.


Asunto(s)
Antibacterianos , Nanocables , Antibacterianos/química , Titanio/química , Bacterias Gramnegativas , Bacterias Grampositivas , Iones
15.
Bioact Mater ; 20: 404-417, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35784636

RESUMEN

Human cancers typically express a high level of tumor-promoting mutant p53 protein (Mutp53) with a minimal level of tumor-suppressing wild-type p53 protein (WTp53). In this regard, inducing Mutp53 degradation while activating WTp53 is a viable strategy for precise anti-tumor therapy. Herein, a new carrier-free nanoprodrug (i.e., Mn-ZnO2 nanoparticles) was developed for concurrent delivery of dual Zn-Mn ions and reactive oxygen species (ROS) within tumor to regulate the p53 protein for high anti-tumor efficacy. In response to the mild tumor acidic environment, the released Zn2+ and H2O2 from Mn-ZnO2 NPs induced ubiquitination-mediated proteasomal degradation of Mutp53, while the liberative Mn2+ and increased ROS level activated the ATM-p53-Bax pathway to elevate WTp53 level. Both in vitro and in vivo results demonstrated that pH-responsive decomposition of Mn-ZnO2 NPs could effectively elevate the intracellular dual Zn-Mn ions and ROS level and subsequently generate the cytotoxic hydroxyl radical (•OH) through the Fenton-like reaction. With the integration of multiple functions (i.e., carrier-free ion and ROS delivery, tumor accumulation, p53 protein modulation, toxic •OH generation, and pH-activated MRI contrast) in a single nanosystem, Mn-ZnO2 NPs demonstrate its superiority as a promising nanotherapeutics for p53-mutated tumor therapy.

16.
Int J Biol Macromol ; 223(Pt A): 1145-1157, 2022 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-36400205

RESUMEN

Cancer chemotherapy drugs are widely criticized for their serious side effects and low cure rate. Therefore, adjuvant therapy as a combination with chemotherapy administration is being accepted by many patients. However, unclear drug targets and mechanisms limit the application of adjuvant treatment. In this study, we confirmed TMEM16A is a key drug target for lung adenocarcinoma, and narirutin is an effective anti-lung adenocarcinoma natural product. Virtual screening and fluorescence experiments confirmed that narirutin inhibits the molecular target TMEM16A, which is specific high-expression in lung adenocarcinoma. Molecular dynamics simulations and electrophysiological experiments revealed the precise molecular mechanism of narirutin regulating TMEM16A. The anticancer effect of narirutin and its synergistic effect on cisplatin were explored by cell proliferation, migration, and apoptosis assays. The signaling pathways regulated by narirutin were analyzed by western blot. Tumor xenograft mice experiments demonstrated the synergistic anticancer effect of narirutin and cisplatin, and the side effects of high concentrations of cisplatin were almost eliminated. Pharmacokinetic experiments showed the biological safety of narirutin is satisfactory in vivo. Based on the significant anticancer effect and high biosafety, naringin has great potential as a functional food in the adjuvant treatment of lung cancer.


Asunto(s)
Productos Biológicos , Neoplasias Pulmonares , Humanos , Ratones , Animales , Anoctamina-1/metabolismo , Anoctamina-1/farmacología , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Movimiento Celular , Neoplasias Pulmonares/patología , Proliferación Celular , Cisplatino/metabolismo , Línea Celular Tumoral
17.
Zool Res ; 43(5): 886-896, 2022 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-36052553

RESUMEN

Various peptide toxins in animal venom inhibit voltage-gated sodium ion channel Nav1.7, including Nav-targeting spider toxin (NaSpTx) Family I. Toxins in NaSpTx Family I share a similar structure, i.e., N-terminal, loops 1-4, and C-terminal. Here, we used Mu-theraphotoxin-Ca2a (Ca2a), a peptide isolated from Cyriopagopus albostriatus, as a template to investigate the general properties of toxins in NaSpTx Family I. The toxins interacted with the cell membrane prior to binding to Nav1.7 via similar hydrophobic residues. Residues in loop 1, loop 4, and the C-terminal primarily interacted with the S3-S4 linker of domain II, especially basic amino acids binding to E818. We also identified the critical role of loop 2 in Ca2a regarding its affinity to Nav1.7. Our results provide further evidence that NaSpTx Family I toxins share similar structures and mechanisms of binding to Nav1.7.


Asunto(s)
Venenos de Araña , Animales , Péptidos/química , Canales de Sodio , Venenos de Araña/química , Venenos de Araña/genética , Venenos de Araña/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico
18.
ACS Appl Mater Interfaces ; 14(28): 31715-31726, 2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35798541

RESUMEN

The gated state of anion channels is involved in the regulation of proliferation and migration of tumors. Specific regulators are urgently needed for efficacious cancer ablation. For this purpose, it is essential to understand the molecular mechanisms of interaction between the regulators and anion channels and apply this knowledge to regulate anion channels. Transmembrane 16A (TMEM16A) is the molecular basis of the calcium-activated chloride channels. It is an anion channel activated by Ca2+, and the inhibition of TMEM16A is associated with a decrease in tumorigenesis. Herein, we characterized a natural compound procyanidin (PC) as an efficacious and selective inhibitor of TMEM16A with an IC50 of 10.6 ± 0.6 µM. Our research revealed the precise sites (D383, R535, and E624) of electrostatic interactions between PC and TMEM16A. Near-infrared (NIR)-light-responsive photothermal conjugated polymer nanoparticles encapsulating PC (CPNs-PC) were established to remotely target and regulate the TMEM16A anion channel. Upon NIR irradiation, CPNs-PC downregulated the signaling pathway downstream of TMEM16A and arrested the cell cycle progression of cancer cells and improved the bioavailability of PC. The tumor inhibition ratio of CPNs-PC was superior to PC by 13.4%. Our findings enabled the development of a strategy to accurately and remotely regulate anion channels to promote tumor regression using NIR-light-responsive conjugated polymer nanoparticles containing specific inhibitors of TMEM16A.


Asunto(s)
Canales de Cloruro , Transducción de Señal , Aniones , Anoctamina-1/metabolismo , Calcio/metabolismo , Canales de Cloruro/metabolismo , Polímeros/metabolismo
19.
Biomed Pharmacother ; 153: 113392, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35834992

RESUMEN

Chemotherapy is one of the main methods for malignant lung cancer treatment. However, the side effects of chemotherapy drugs are serious and it is prone to drug resistance. Therefore, multi-drug combination chemotherapy is popular in lung cancer treatment. This study found that tracheloside (TCS) was a novel inhibitor of TMEM16A which was specific high expressed in lung cancer tissues. TCS concentration dependently inhibited TMEM16A with an IC50 of 3.09 ± 0.21 µM. It inhibited lung cancer cells proliferation, migration, and induced cells apoptosis targeting TMEM16A. In addition, molecular docking combined with site-directed mutagenesis confirmed that the binding sites of TCS to TMEM16A were S387, E623, E624. Subsequently, multi-target combined drug administration was conducted based on the different drug targets of TCS and doxorubicin (DOX). Both in vitro and in vivo experiments indicated that the combined administration of low concentration of TCS and DOX achieved satisfactory anticancer effect, and it offset the side effects caused by high concentration of DOX. Therefore, TCS is a safe and efficient anticancer lead compound which can enhance the effect of DOX.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , 4-Butirolactona/análogos & derivados , Adenocarcinoma del Pulmón/tratamiento farmacológico , Apoptosis , Línea Celular Tumoral , Doxorrubicina , Glucósidos , Humanos , Neoplasias Pulmonares/patología , Simulación del Acoplamiento Molecular
20.
Acta Biomater ; 148: 152-162, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35718101

RESUMEN

Hepatocellular carcinoma development is closely related to the changes in tissue mechanics induced by excess collagen deposition and crosslinking, which leads to liver fibrosis and malignant progression. The role of matrix stiffness has been widely assessed using various linearly elastic materials. However, the liver, like many soft tissues, also exhibits nonlinear elasticity by strain-stiffening, allowing cells to mechanically interact with their micromilieus which has attracted much attention in cellular processes recently. Here, we use a biomimetic hydrogel grafting of GRGDS peptide with tunable nonlinear mechanical properties, polyisocyanides (PIC), to investigate the influence of strain-stiffening on HepG2 liver cancer cell behavior by tuning PIC polymer length. Compared to short PIC polymer with lower critical stress, PIC hydrogels composed of long polymer with higher critical stress promote the motility and invasiveness of HepG2 cells, and induce more actin stress fibers and higher expression level of mechanotransducer YAP and its nuclear translocation. Strikingly, the expression of calcium-activated potassium channel KCa3.1, an important biomarker in hepatocellular carcinoma, is also affected by the mechanical property of PIC hydrogels. It was also shown that downregulating the KCa3.1 channel can be achieved by inhibiting the formation of actin fibers. Our findings imply that the strain-stiffening property of PIC hydrogels affects the expression of KCa3.1 potassium channel via mediating cytoskeletal stress fiber formation, and ultimately influences the liver carcinoma cell functional response. STATEMENT OF SIGNIFICANCE: The effect of nonlinear elasticity by strain-stiffening, is assessed in HepG2 liver cancer cell behavior by using a biomimetic hydrogel with tunable mechanical properties, polyisocyanides (PIC). PIC gels with higher critical stress promote the motility and invasiveness of HepG2 cells and induce upregulated expression levels of KCa3.1 potassium channel and YAP, but which can be suppressed by inhibiting the formation of actin fibers. Our findings imply that the strain-stiffening property of PIC gels influences the expression of KCa3.1 potassium channel via mediating cytoskeletal stress fiber formation and, ultimately affects the liver carcinoma cell functional response.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Actinas , Elasticidad , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Polímeros/química , Canales de Potasio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...