Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 15.472
1.
Signal Transduct Target Ther ; 9(1): 142, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38825657

Radiotherapy combined with immune checkpoint blockade holds great promise for synergistic antitumor efficacy. Targeted radionuclide therapy delivers radiation directly to tumor sites. LNC1004 is a fibroblast activation protein (FAP)-targeting radiopharmaceutical, conjugated with the albumin binder Evans Blue, which has demonstrated enhanced tumor uptake and retention in previous preclinical and clinical studies. Herein, we demonstrate that 68Ga/177Lu-labeled LNC1004 exhibits increased uptake and prolonged retention in MC38/NIH3T3-FAP and CT26/NIH3T3-FAP tumor xenografts. Radionuclide therapy with 177Lu-LNC1004 induced a transient upregulation of PD-L1 expression in tumor cells. The combination of 177Lu-LNC1004 and anti-PD-L1 immunotherapy led to complete eradication of all tumors in MC38/NIH3T3-FAP tumor-bearing mice, with mice showing 100% tumor rejection upon rechallenge. Immunohistochemistry, single-cell RNA sequencing (scRNA-seq), and TCR sequencing revealed that combination therapy reprogrammed the tumor microenvironment in mice to foster antitumor immunity by suppressing malignant progression and increasing cell-to-cell communication, CD8+ T-cell activation and expansion, M1 macrophage counts, antitumor activity of neutrophils, and T-cell receptor diversity. A preliminary clinical study demonstrated that 177Lu-LNC1004 was well-tolerated and effective in patients with refractory cancers. Further, scRNA-seq of peripheral blood mononuclear cells underscored the importance of addressing immune evasion through immune checkpoint blockade treatment. This was emphasized by the observed increase in antigen processing and presentation juxtaposed with T cell inactivation. In conclusion, our data supported the efficacy of immunotherapy combined with 177Lu-LNC1004 for cancer patients with FAP-positive tumors.


Immune Checkpoint Inhibitors , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Endopeptidases/genetics , NIH 3T3 Cells , Radiopharmaceuticals/therapeutic use , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Xenograft Model Antitumor Assays , Immunotherapy , Gelatinases/genetics , Gelatinases/immunology , Lutetium/pharmacology , Cell Line, Tumor
2.
Asian Pac J Cancer Prev ; 25(5): 1737-1743, 2024 May 01.
Article En | MEDLINE | ID: mdl-38809646

OBJECTIVE: This study aimed to investigate the cytotoxicity effect of the ethyl acetate extract of Aaptos suberitoides on colorectal cancer cells (DLD-1) and murine fibroblast cells (NIH-3T3). METHODS: A. suberitoides was collected from Putus Island, Bunaken National Park, North Sulawesi, Indonesia, and was processed with maceration and ethyl acetate extraction. The sponge extract was characterized based on Thin Layer Chromatography (TLC) and then identified by using LCMS/MS analysis. DLD-1 and NIH-3T3 cells were treated with the ethyl acetate extract and then followed by 3- [4, 5-dimethylthiazol-2-yl] -2.5 diphenyl tetrazolium bromide (MTT) assay to assess their cytotoxicity effect. RESULTS: LCMS/MS analysis showed that the most abundant compounds in this extract were identified as aaptamine (1). Furthermore, this study revealed that the active ethyl acetate fraction of A. suberitoides has cytotoxic effects in colorectal cancer DLD-1 cells with an IC50 value of 9.597 µg/mL, higher than NIH-3T3 cells with an IC50 value of 12.23 µg/mL Thus, the active ethyl acetate fraction of A. suberitoides is considered more toxic to cancer cells than normal cells. CONCLUSION: This study provides the first evidence to support the role of the ethyl acetate extract of A. suberitoides sponge extracts to be developed as a colorectal anticancer agent.


Cell Proliferation , Colorectal Neoplasms , Porifera , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Porifera/chemistry , Mice , Humans , Indonesia , Cell Proliferation/drug effects , NIH 3T3 Cells , Antineoplastic Agents/pharmacology , Tumor Cells, Cultured , Apoptosis/drug effects , Naphthyridines
3.
Anticancer Res ; 44(6): 2393-2406, 2024 Jun.
Article En | MEDLINE | ID: mdl-38821585

BACKGROUND/AIM: Cholangiocarcinoma (CCA) is an aggressive tumor with limited treatment options especially in 2nd line or later treatments. Targeting fibroblast growth factor receptor (FGFR) 2 has recently emerged as a promising treatment option for patients with CCA harboring FGFR2-fusion. This study investigated the antitumor activities of tasurgratinib as an orally available FGFR1-3 inhibitor, in preclinical FGFR2-driven CCA models. MATERIALS AND METHODS: Antitumor activities of tasurgratinib were examined in vitro and in vivo using NIH/3T3 cells expressing FGFR2-fusion as FGFR2-driven CCA models, and in vivo using a CCA patient-derived xenograft model. The molecular mechanism of action of tasurgratinib was elucidated through co-crystal structure analysis with FGFR1, manual complex model analysis with FGFR2, and binding kinetics analysis with FGFR2. Furthermore, the cell-based inhibitory activities against acquired resistant FGFR2 mutations in patients with CCA treated with FGFR inhibitors were evaluated. RESULTS: Tasurgratinib showed antitumor activity in preclinical FGFR2-driven CCA models by inhibiting the FGFR signaling pathway in vitro and in vivo. Furthermore, cell-based target engagement assays indicated that tasurgratinib had potent inhibitory activities against FGFR2 mutations, such as N549H/K, which are the major acquired mutations in CCA. We also confirmed that tasurgratinib exhibited fast association and slow dissociation kinetics with FGFR2, binding to the ATP-binding site and the neighboring region, and adopting an Asp-Phe-Gly (DFG)-"in" conformation. CONCLUSION: These data demonstrate the therapeutic potential of tasurgratinib in FGFR2-driven CCA and provide molecular mechanistic insights into its unique inhibitory profile against secondary FGFR2 resistance mutations in patients with CCA treated with FGFR inhibitors.


Bile Duct Neoplasms , Cholangiocarcinoma , Receptor, Fibroblast Growth Factor, Type 2 , Xenograft Model Antitumor Assays , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Animals , Humans , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Mice , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Administration, Oral , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , NIH 3T3 Cells , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrimidines/administration & dosage , Cell Proliferation/drug effects , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors
4.
Biomater Adv ; 161: 213886, 2024 Jul.
Article En | MEDLINE | ID: mdl-38735200

Altering the route of uptake by the cells is an attractive strategy to overcome drug-receptor adaptation problems. Carbon nanoparticles (CNPs) with emission beyond tissue autofluorescence for imaging biological tissues were used to study the phenomenon of uptake by the cells. In this regard, red-emitting carbon nanoparticles (CNPs) were synthesized and incorporated onto lipid microbubbles (MBs). The CNPs showed red emissions in the range of 640 nm upon excitation with 480 nm wavelength of light. Atomic force microscopic and confocal microscopic images showed the successful loading of CNPs onto the MB. Carbon nanoparticle loaded microbubbles (CNP-MBs) were treated with NIH 3 T3 cells at different concentrations. Confocal microscopic imaging studies confirm the presence of CNPs inside the treated cells. Cytotoxicity studies revealed that the CNPs showed minimal toxicity towards cells after loading onto MBs. The CNPs are usually taken up by the cells through the clathrin-mediated (CME) pathway, but when loaded onto MBs, the mechanism of uptake of CNPs is altered, and the uptake by the cells was observed even in the presence of inhibitors for the CME pathway. Loading CNPs onto MBs resulted in the uptake of CNPs by the cell through micropinocytosis and sonophoresis in the presence of ultrasound. The in vivo uptake CNP-MBs were performed in Danio rerio (Zebrafish larvae). This study provides insights into altering the uptake pathway through reformulation by loading nanoparticles onto MBs.


Carbon , Microbubbles , Nanoparticles , Zebrafish , Animals , Carbon/chemistry , Mice , Nanoparticles/chemistry , NIH 3T3 Cells , Drug Delivery Systems/methods
5.
J Cell Biol ; 223(9)2024 Sep 02.
Article En | MEDLINE | ID: mdl-38787349

Cell processes require precise regulation of actin polymerization that is mediated by plus-end regulatory proteins. Detailed mechanisms that explain plus-end dynamics involve regulators with opposing roles, including factors that enhance assembly, e.g., the formin mDia1, and others that stop growth (capping protein, CP). We explore IQGAP1's roles in regulating actin filament plus-ends and the consequences of perturbing its activity in cells. We confirm that IQGAP1 pauses elongation and interacts with plus ends through two residues (C756 and C781). We directly visualize the dynamic interplay between IQGAP1 and mDia1, revealing that IQGAP1 displaces the formin to influence actin assembly. Using four-color TIRF, we show that IQGAP1's displacement activity extends to formin-CP "decision complexes," promoting end-binding protein turnover at plus-ends. Loss of IQGAP1 or its plus-end activities disrupts morphology and migration, emphasizing its essential role. These results reveal a new role for IQGAP1 in promoting protein turnover on filament ends and provide new insights into how plus-end actin assembly is regulated in cells.


Actin Capping Proteins , Actin Cytoskeleton , Formins , ras GTPase-Activating Proteins , Animals , Humans , Actin Capping Proteins/metabolism , Actin Capping Proteins/genetics , Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Movement , Formins/metabolism , HeLa Cells , Protein Binding , ras GTPase-Activating Proteins/metabolism , ras GTPase-Activating Proteins/genetics , Mice , NIH 3T3 Cells
6.
Int Immunopharmacol ; 134: 112230, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38744171

Accelerating wound healing, as well as preventing infection and scar formation are among the most important medical challenges. This study aims to examine the antimicrobial, immunomodulatory, and anticancer properties of Morus nigra. The antimicrobial activities of ripe and unripe M. nigra fruit (MNF) extracts were tested. HPLC was employed to measure the components in the extract. Oserin ointment was made with 8 % extract. To test the ointment, 48 Wistar rats were randomly assigned into eight groups. The ointment was used daily by treating the wounds. Tissue histology and wound healing were assessed over nine days. Comparative evaluation of wound healing was conducted by analyzing TGF-ß, TNF-α, and IL-1 mRNA levels. Finally, cytotoxic effects on AGS cancer and NIH-3 T3 fibroblast cells were examined. The ANOVA test and Prsim program were used for statistical analysis. Unripe MNF extract had good antimicrobial properties in standard and nosocomial strains. The most abundant compound in the extract was ascorbic acid (0.0441 mg/10 mg extract), followed by naringenin and gallic acid. In all groups treated with MNF extract ointment, a significant reduction in wound area was observed compared to other groups (p < 0.05). After six days of treatment, the microbial load was uncountable. In the microscopic studies of the wounds, a significant increase was observed in fibroblasts, angiogenesis, and in neutrophils in the first days as well as a decrease in the final days. The treatment caused a significant decline in the expression of IL-1 and TNF-α genes, as well as an increase in the expression of TGF-ß (p < 0.05). This extract had no significant cytotoxic effects on human fibroblast cells (p > 0.05). In general, it can be concluded that the unripe MNF extract ointment can be a suitable option for the treatment of infectious and non-infectious skin wounds.


Fruit , Morus , Ointments , Plant Extracts , Rats, Wistar , Wound Healing , Animals , Wound Healing/drug effects , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Fruit/chemistry , Mice , Humans , Rats , NIH 3T3 Cells , Morus/chemistry , Male , Skin/drug effects , Skin/pathology , Fibroblasts/drug effects , Anti-Infective Agents/pharmacology , Anti-Infective Agents/therapeutic use , Cell Line, Tumor
7.
Viruses ; 16(5)2024 05 11.
Article En | MEDLINE | ID: mdl-38793642

Mouse adenoviruses (MAdV) play important roles in studying host-adenovirus interaction. However, easy-to-use reverse genetics systems are still lacking for MAdV. An infectious plasmid pKRMAV1 was constructed by ligating genomic DNA of wild-type MAdV-1 with a PCR product containing a plasmid backbone through Gibson assembly. A fragment was excised from pKRMAV1 by restriction digestion and used to generate intermediate plasmid pKMAV1-ER, which contained E3, fiber, E4, and E1 regions of MAdV-1. CMV promoter-controlled GFP expression cassette was inserted downstream of the pIX gene in pKMAV1-ER and then transferred to pKRMAV1 to generate adenoviral plasmid pKMAV1-IXCG. Replacement of transgene could be conveniently carried out between dual BstZ17I sites in pKMAV1-IXCG by restriction-assembly, and a series of adenoviral plasmids were generated. Recombinant viruses were rescued after transfecting linearized adenoviral plasmids to mouse NIH/3T3 cells. MAdV-1 viruses carrying GFP or firefly luciferase genes were characterized in gene transduction, plaque-forming, and replication in vitro or in vivo by observing the expression of reporter genes. The results indicated that replication-competent vectors presented relevant properties of wild-type MAdV-1 very well. By constructing viruses bearing exogenous fragments with increasing size, it was found that MAdV-1 could tolerate an insertion up to 3.3 kb. Collectively, a replication-competent MAdV-1 vector system was established, which simplified procedures for the change of transgene or modification of E1, fiber, E3, or E4 genes.


Genetic Vectors , Plasmids , Virus Replication , Animals , Mice , Genetic Vectors/genetics , Plasmids/genetics , Adenoviridae/genetics , NIH 3T3 Cells , Cloning, Molecular , Genes, Reporter
8.
Molecules ; 29(10)2024 May 16.
Article En | MEDLINE | ID: mdl-38792206

Various conjugates with rhodamines were prepared by starting with betulinic acid (BA) and platanic acid (PA). The molecules homopiperazine and piperazine, which were identified in earlier research, served as linkers between the rhodamine and the triterpene. The pentacyclic triterpene's ring A was modified with two acetyloxy groups in order to possibly boost its cytotoxic activity. The SRB assays' cytotoxicity data showed that conjugates 13-22, derived from betulinic acid, had a significantly higher cytotoxicity. Of these hybrids, derivatives 19 (containing rhodamine B) and 22 (containing rhodamine 101) showed the best values with EC50 = 0.016 and 0.019 µM for A2780 ovarian carcinoma cells. Additionally, based on the ratio of EC50 values, these two compounds demonstrated the strongest selectivity between malignant A2780 cells and non-malignant NIH 3T3 fibroblasts. A375 melanoma cells were used in cell cycle investigations, which showed that the cells were halted in the G1/G0 phase. Annexin V/FITC/PI staining demonstrated that the tumor cells were affected by both necrosis and apoptosis.


Apoptosis , Rhodamines , Triterpenes , Triterpenes/chemistry , Triterpenes/pharmacology , Triterpenes/chemical synthesis , Humans , Rhodamines/chemistry , Mice , Animals , Cell Line, Tumor , NIH 3T3 Cells , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Betulinic Acid , Pentacyclic Triterpenes/pharmacology , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/chemical synthesis , Cell Cycle/drug effects , Cell Survival/drug effects , Cell Proliferation/drug effects , Lupanes
9.
Toxicol In Vitro ; 98: 105845, 2024 Jun.
Article En | MEDLINE | ID: mdl-38754600

Current clinical therapies for metastatic breast cancer (MBC) have limited therapeutic efficacy and induce significant systemic side effects, leading to poor patient compliance. To address this challenge, this investigation focuses on the design of LINC02535 + miR-30a-5p for treating breast cancer. In vitro cytotoxicity studies confirmed that LINC02535 + miR-30a-5p was more effective in 4 T1 cells, with reduced toxicity in NIH3T3 cells. Further verification of cellular morphology was achieved through various biochemical staining methods. Additionally, the antimetastatic attributes of LINC02535 + miR-30a-5p have been evaluated using both migration scratch and Transwell migration assays, which have collectively revealed excellent antimetastatic potential. The DNA fragmentation of the 4 T1 cells was assessed using a comet assay. LINC02535 + miR-30a-5p improved ROS levels and caused mitochondrial membrane potential alterations and DNA damage, which resulted in apoptosis. Therefore, we propose that LINC02535 + miR-30a-5p could be an alternative therapeutic strategy for breast cancer therapy.


Apoptosis , Breast Neoplasms , Cell Proliferation , MicroRNAs , RNA, Long Noncoding , MicroRNAs/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Apoptosis/drug effects , Animals , Mice , Humans , Cell Proliferation/drug effects , Female , Cell Line, Tumor , RNA, Long Noncoding/genetics , Cell Movement , NIH 3T3 Cells , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , DNA Damage
10.
Int J Biol Macromol ; 270(Pt 1): 131856, 2024 Jun.
Article En | MEDLINE | ID: mdl-38693000

Biomacromolecules derived from natural sources offer superior biocompatibility, biodegradability, and water-holding capacity, which make them promising scaffolds for tissue engineering. Psyllium seed has gained attention in biomedical applications recently due to its gel-forming ability, which is provided by its polysaccharide-rich content consisting mostly of arabinoxylan. This study focuses on the extraction and gelation of Psyllium seed hydrocolloid (PSH) in a single-step water-based protocol, and scaffold fabrication using freeze-drying method. After characterization of the scaffold, including morphological, mechanical, swelling, and protein adsorption analyses, 3D cell culture studies were done using NIH-3 T3 fibroblast cells on PSH scaffold, and cell viability was assessed using Live/Dead and Alamar Blue assays. Starting from day 1, high cell viability was obtained, and it reached 90 % at the end of 15-day culture period. Cellular morphology on PSH scaffold was monitored via SEM analysis; cellular aggregates then spheroid formation were observed throughout the study. Collagen Type-I and F-actin expressions were followed by immunostaining revealing a 9- and 10-fold increase during long-term culture. Overall, a single-step and non-toxic protocol was developed for extraction and gelation of PSH. Obtained results unveiled that PSH scaffold provided a favorable 3D microenvironment for cells, holding promise for further tissue engineering applications.


Colloids , Psyllium , Seeds , Tissue Engineering , Tissue Scaffolds , Xylans , Psyllium/chemistry , Xylans/chemistry , Xylans/pharmacology , Tissue Engineering/methods , Animals , Seeds/chemistry , Mice , Colloids/chemistry , Tissue Scaffolds/chemistry , NIH 3T3 Cells , Cell Survival/drug effects , Water/chemistry
11.
Biomed Phys Eng Express ; 10(3)2024 Apr 02.
Article En | MEDLINE | ID: mdl-38564254

The high demand for bone grafts has motivated the development of implants with excellent osteogenic activity, whereas the risk of implant-associated infection, particularly given the rise of antimicrobial resistance, has compelled the development of implants with innovative antimicrobial strategies in which a small amount of bactericidal agent can effectively kill a wide range of bacteria. To induce antibacterial property, the surface of Grade-5 bone plate titanium implants used in clinical applications was modified using direct current (DC) sputter coating followed by thermal annealing. The 15 nm silver film-coated implants were thermally annealed in the furnace for 15 min at 750 °C. The modified implant surface's antibacterial efficacy againstEscherichia coli(E. coli),Staphylococcus aureus(S. aureus),Salmonella typhi, andMethicillin-resistant staphylococcus aureusbacteria has been assessed using a colony-forming assay. On the modified implant surface, the growth ofE. coliandS. aureusbacteria is reduced by 99.72%, while highly drug-resistant bacteria are inhibited by 96.59%. The MTT assay was used to assess the cytotoxicity of the modified bone-implant surface against NIH3T3 mouse fibroblast cells. The modified bone-implant surface promoted fibroblast growth and demonstrated good cytocompatibility. Furthermore, the mechanical properties of the implant were not harmed by this novel surface modification method. This method is simple and provides new insight into surface modification of commercial metallic implants to have effective antibacterial properties against various classes of bacteria.


Alloys , Methicillin-Resistant Staphylococcus aureus , Silver , Animals , Mice , Titanium , Bone Plates , Escherichia coli , NIH 3T3 Cells , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology
12.
Biomed Mater ; 19(3)2024 Apr 26.
Article En | MEDLINE | ID: mdl-38574669

Recently,in vitromodels of intestinal mucosa have become important tools for drug screening and studying the physiology and pathology of the intestine. These models enable the examination of cellular behavior in diseased states or in reaction to alterations in the microenvironment, potentially serving as alternatives to animal models. One of the major challenges in constructing physiologically relevantin vitromodels of intestinal mucosa is the creation of three-dimensional microstructures that accurately mimic the integration of intestinal epithelium and vascularized stroma. Here, core-shell alginate (Alg) microspheres were generated to create the compartmentalized extracellular matrix microenvironment needed to simulate the epithelial and vascularized stromal compartments of the intestinal mucosa. We demonstrated that NIH-3T3 and human umbilical vein endothelial cells embedded in the core of the microspheres can proliferate and develop a vascular network, while human colorectal adenocarcinoma cells (Caco-2) can form an epithelial monolayer in the shell. Compared to Caco-2 monolayer encapsulated within the shell, the presence of the vascularized stroma enhances their proliferation and functionality. As such, our core-shell Alg microspheres provide a valuable method for generatingin vitromodels of vascularized intestinal mucosa with epithelial and vascularized stroma arranged in a spatially relevant manner and demonstrating near-physiological functionality.


Alginates , Cell Proliferation , Human Umbilical Vein Endothelial Cells , Intestinal Mucosa , Microspheres , Tissue Engineering , Alginates/chemistry , Humans , Intestinal Mucosa/metabolism , Animals , Mice , Caco-2 Cells , Tissue Engineering/methods , NIH 3T3 Cells , Extracellular Matrix/metabolism , Tissue Scaffolds/chemistry , Hexuronic Acids/chemistry
13.
ACS Appl Mater Interfaces ; 16(15): 18327-18343, 2024 Apr 17.
Article En | MEDLINE | ID: mdl-38588343

58S bioactive glass (BG) has effective biocompatibility and bioresorbable properties for bone tissue engineering; however, it has limitations regarding antibacterial, antioxidant, and mechanical properties. Therefore, we have developed BGAC biocomposites by reinforcing 58S BG with silver and ceria nanoparticles, which showed effective bactericidal properties by forming inhibited zones of 2.13 mm (against Escherichia coli) and 1.96 mm (against Staphylococcus aureus; evidenced by disc diffusion assay) and an increment in the antioxidant properties by 39.9%. Moreover, the elastic modulus, hardness, and fracture toughness were observed to be increased by ∼84.7% (∼51.9 GPa), ∼54.5% (∼3.4 GPa), and ∼160% (∼1.3 MPam1/2), whereas the specific wear rate was decreased by ∼55.2% (∼1.9 × 10-11 m3/Nm). X-ray diffraction, high-resolution transmission electron microscopy, and field emission scanning electron microscopy confirmed the fabrication of biocomposites and the uniform distribution of the nanomaterials in the BG matrix. The addition of silver nanoparticles in the 58S BG matrix (in BGA) increased mechanical properties by composite strengthening and bactericidal properties by damaging the cytoplasmic membrane of bacterial cells. The addition of nanoceria in 58S BG (BGC) increased the antioxidant properties by 44.5% (as evidenced by the 2,2-diphenyl-1-picrylhydrazyl assay). The resazurin reduction assay and MTT assay confirmed the effective cytocompatibility for BGAC biocomposites against mouse embryonic fibroblast cells (NIH3T3) and mouse bone marrow stromal cells. Overall, BGAC resulted in mechanical properties comparable to those of cancellous bone, and its effective antibacterial and cytocompatibility properties make it a good candidate for bone healing.


Cerium , Metal Nanoparticles , Silver , Animals , Mice , Antioxidants , NIH 3T3 Cells , Fibroblasts , Anti-Bacterial Agents/pharmacology , Glass
14.
Int J Biol Macromol ; 267(Pt 2): 131389, 2024 May.
Article En | MEDLINE | ID: mdl-38582461

This work developed Acer tegmentosum extract-mediated silver nanoparticles (AgNPs) loaded chitosan (CS)/alginic acid (AL) scaffolds (CS/AL-AgNPs) to enhance the healing of E. coli-infected wounds. The SEM-EDS and XRD results revealed the successful formation of the CS/AL-AgNPs. FTIR analysis evidenced that the anionic group of AL (-COO-) and cationic amine groups of CS (-NH3+) were ionically crosslinked to form scaffold (CS/AL). The CS/AL-AgNPs exhibited significant antimicrobial activity against both Gram-positive (G+) and Gram-negative (G-) bacterial pathogens, while being non-toxic to red blood cells (RBCs), the hen's egg chorioallantoic membrane (HET-CAM), and a non-cancerous cell line (NIH3T3). Treatment with CS/AL-AgNPs significantly accelerated the healing of E. coli-infected wounds by regulating the collagen deposition and blood parameters as evidenced by in vivo experiments. Overall, these findings suggest that CS/AL-AgNPs are promising for the treatment of infected wounds.


Acer , Alginates , Anti-Bacterial Agents , Chitosan , Escherichia coli , Metal Nanoparticles , Plant Extracts , Silver , Wound Healing , Chitosan/chemistry , Chitosan/pharmacology , Metal Nanoparticles/chemistry , Silver/chemistry , Silver/pharmacology , Animals , Wound Healing/drug effects , Escherichia coli/drug effects , Mice , Acer/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , NIH 3T3 Cells , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Alginates/chemistry , Alginates/pharmacology , Escherichia coli Infections/drug therapy , Tissue Scaffolds/chemistry
15.
Int J Biol Macromol ; 267(Pt 2): 131375, 2024 May.
Article En | MEDLINE | ID: mdl-38604424

Compostable zein-polycaprolactone (PZ) electrospun nanofiber integrated with different concentrations of Aster yomena extract loaded halloysite nanotubes (A. yomena-HNT) as bioactive nanofibrous food packaging is reported. SEM micrographs reveal heterogeneous nanofibers. A. yomena extract used in the study showed weak antioxidant activity with AAI and TEAC values of 0.229 and 0.346. In vitro, release profile over 7 days of A. yomena indicates a controlled, sustained, and prolonged release. The prepared nanofibers were effective against both gram-positive and gram-negative bacteria. The prepared composite nanofibers were rendered biocompatible and nontoxic when subjected to WST-1 and LDH assay after incubating with NIH 3T3 mouse fibroblast cell line. PZ-15 nanofiber packaging showed the best postharvest quality preservation in Black mulberry fruits after 4 days of storage at 25 °C and 85 % Rh. Moreover, the in vitro decomposition test reveals that the fabricated nanofibers decompose in the soil and do not pose as a threat to the environment.


Clay , Food Packaging , Nanofibers , Nanotubes , Plant Extracts , Polyesters , Zein , Nanofibers/chemistry , Polyesters/chemistry , Zein/chemistry , Mice , Animals , Clay/chemistry , Food Packaging/methods , Nanotubes/chemistry , NIH 3T3 Cells , Plant Extracts/chemistry , Plant Extracts/pharmacology , Antioxidants/pharmacology , Antioxidants/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry
16.
Int J Biol Macromol ; 267(Pt 1): 131328, 2024 May.
Article En | MEDLINE | ID: mdl-38574901

The management of invasive fungal infections in humans poses significant challenges due to the intricate nature of the treatment, which is both arduous and costly, necessitating routine diagnostic procedures. Consequently, this investigation aimed to formulate a chitosan-based nanoemulsion (CS NEMs) incorporating the antifungal agent undecanoic acid (UDA), characterizing these NEMs and assessing their antifungal efficacy against both filamentous and non-filamentous fungal pathogens. The CS-based UDA NEMs were synthesized by introducing the surfactant Triton X-100 and the stabilizer glycerol. Nanoparticle tracking analysis (NTA) and SEM demonstrated the CS-UDA NEMs with an average size of 145 nm and 164.5 ± 24 nm, respectively. The successful formation of CS-UDA NEMs was verified through FTIR and XRD. CS-UDA NEMs exhibited exceptional inhibition against Aspergillus flavus, Aspergillus fumigatus, Aspergillus niger, and Candida albicans with MFC of 500, 500, 250 and 250 µg/mL, respectively. Additionally, CS-UDA NEMs displayed comparatively lower antioxidant activity as determined by DPPH and ABTS radical scavenging assays. Importantly, CS-UDA NEMs demonstrated no cytotoxic effects on NIH3T3 cells even at higher concentration (1000 µg/mL), as confirmed by cell viability and fluorescent staining assays. In conclusion, this study suggests that the developed CS-UDA NEMs hold promise as potent antifungal agents with diverse potential applications.


Antifungal Agents , Chitosan , Emulsions , Fatty Acids , Chitosan/chemistry , Chitosan/pharmacology , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Emulsions/chemistry , Fatty Acids/chemistry , Mice , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Microbial Sensitivity Tests , Nanoparticles/chemistry , NIH 3T3 Cells , Antioxidants/pharmacology , Antioxidants/chemistry , Candida albicans/drug effects
17.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article En | MEDLINE | ID: mdl-38673904

Chagas disease is one of the world's neglected tropical diseases, caused by the human pathogenic protozoan parasite Trypanosoma cruzi. There is currently a lack of effective and tolerable clinically available therapeutics to treat this life-threatening illness and the discovery of modern alternative options is an urgent matter. T. cruzi glucokinase (TcGlcK) is a potential drug target because its product, d-glucose-6-phosphate, serves as a key metabolite in the pentose phosphate pathway, glycolysis, and gluconeogenesis. In 2019, we identified a novel cluster of TcGlcK inhibitors that also exhibited anti-T. cruzi efficacy called the 3-nitro-2-phenyl-2H-chromene analogues. This was achieved by performing a target-based high-throughput screening (HTS) campaign of 13,040 compounds. The selection criteria were based on first determining which compounds strongly inhibited TcGlcK in a primary screen, followed by establishing on-target confirmed hits from a confirmatory assay. Compounds that exhibited notable in vitro trypanocidal activity over the T. cruzi infective form (trypomastigotes and intracellular amastigotes) co-cultured in NIH-3T3 mammalian host cells, as well as having revealed low NIH-3T3 cytotoxicity, were further considered. Compounds GLK2-003 and GLK2-004 were determined to inhibit TcGlcK quite well with IC50 values of 6.1 µM and 4.8 µM, respectively. Illuminated by these findings, we herein screened a small compound library consisting of thirteen commercially available 3-nitro-2-phenyl-2H-chromene analogues, two of which were GLK2-003 and GLK2-004 (compounds 1 and 9, respectively). Twelve of these compounds had a one-point change from the chemical structure of GLK2-003. The analogues were run through a similar primary screening and confirmatory assay protocol to our previous HTS campaign. Subsequently, three in vitro biological assays were performed where compounds were screened against (a) T. cruzi (Tulahuen strain) infective form co-cultured within NIH-3T3 cells, (b) T. brucei brucei (427 strain) bloodstream form, and (c) NIH-3T3 host cells alone. We report on the TcGlcK inhibitor constant determinations, mode of enzyme inhibition, in vitro antitrypanosomal IC50 determinations, and an assessment of structure-activity relationships. Our results reveal that the 3-nitro-2-phenyl-2H-chromene scaffold holds promise and can be further optimized for both Chagas disease and human African trypanosomiasis early-stage drug discovery research.


Benzopyrans , Glucokinase , Trypanocidal Agents , Trypanosoma cruzi , Animals , Humans , Mice , Benzopyrans/pharmacology , Benzopyrans/chemistry , Chagas Disease/drug therapy , Chagas Disease/parasitology , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Glucokinase/metabolism , Glucokinase/antagonists & inhibitors , High-Throughput Screening Assays , Molecular Docking Simulation , NIH 3T3 Cells , Structure-Activity Relationship , Trypanocidal Agents/pharmacology , Trypanocidal Agents/chemistry , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/enzymology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
18.
Bioorg Chem ; 147: 107374, 2024 Jun.
Article En | MEDLINE | ID: mdl-38636433

The incidence of idiopathic pulmonary fibrosis (IPF) has been steadily increasing each year, posing significant challenges in its treatment. In this study, we conducted the design and synthesis of 23 new inhibitors that specifically target the TGF-ß1/Smad3 pathway. Initially, we employed a cell model of TGF-ß-induced pulmonary fibrosis, using cell survival rate and HYP expression as indicators to identify the potent ingredient 5aa, which demonstrated significant anti-pulmonary fibrosis activity. Subsequently, we induced mice with bleomycin (BLM) to establish an experimental animal model of pulmonary fibrosis, and evaluated the pharmacodynamics of 5aa in vivo against pulmonary fibrosis. The alterations in HYP and collagen levels in BLM-induced pulmonary fibrosis mice were analyzed using ELISA and immunohistochemistry techniques. The results indicated that compound 5aa effectively suppressed the fibrotic response induced by TGF-ß1, inhibited the expression of the fibrotic marker α-SMA, and hindered the EMT process in NIH3T3 cells. Additionally, oral administration of 5aa demonstrated significant therapeutic effects in a mouse model of IPF, comparable to the established drug Nintedanib. Moreover, compound 5aa exhibited higher bioavailability in vivo compared to Nintedanib. These collective outcomes suggest that 5aa holds promise as a potential inhibitor of TGF-ß1/Smad3 signaling for the treatment of IPF.


Idiopathic Pulmonary Fibrosis , Signal Transduction , Smad3 Protein , Transforming Growth Factor beta1 , Animals , Smad3 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/chemically induced , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Mice , Signal Transduction/drug effects , Molecular Structure , Humans , Bleomycin , Structure-Activity Relationship , Mice, Inbred C57BL , NIH 3T3 Cells , Dose-Response Relationship, Drug , Male
19.
Cells ; 13(8)2024 Apr 17.
Article En | MEDLINE | ID: mdl-38667312

The assessment of nanoparticle cytotoxicity is challenging due to the lack of customized and standardized guidelines for nanoparticle testing. Nanoparticles, with their unique properties, can interfere with biochemical test methods, so multiple tests are required to fully assess their cellular effects. For a more reliable and comprehensive assessment, it is therefore imperative to include methods in nanoparticle testing routines that are not affected by particles and allow for the efficient integration of additional molecular techniques into the workflow. Digital holographic microscopy (DHM), an interferometric variant of quantitative phase imaging (QPI), has been demonstrated as a promising method for the label-free assessment of the cytotoxic potential of nanoparticles. Due to minimal interactions with the sample, DHM allows for further downstream analyses. In this study, we investigated the capabilities of DHM in a multimodal approach to assess cytotoxicity by directly comparing DHM-detected effects on the same cell population with two downstream biochemical assays. Therefore, the dry mass increase in RAW 264.7 macrophages and NIH-3T3 fibroblast populations measured by quantitative DHM phase contrast after incubation with poly(alkyl cyanoacrylate) nanoparticles for 24 h was compared to the cytotoxic control digitonin, and cell culture medium control. Viability was then determined using a metabolic activity assay (WST-8). Moreover, to determine cell death, supernatants were analyzed for the release of the enzyme lactate dehydrogenase (LDH assay). In a comparative analysis, in which the average half-maximal effective concentration (EC50) of the nanocarriers on the cells was determined, DHM was more sensitive to the effect of the nanoparticles on the used cell lines compared to the biochemical assays.


Nanoparticles , Animals , Mice , NIH 3T3 Cells , Nanoparticles/toxicity , Nanoparticles/chemistry , RAW 264.7 Cells , Cell Survival/drug effects , Holography/methods , Quantitative Phase Imaging
20.
Int J Pharm ; 654: 123994, 2024 Apr 10.
Article En | MEDLINE | ID: mdl-38484859

Small interfering RNA (siRNA) holds great potential to treat many difficult-to-treat diseases, but its delivery remains the central challenge. This study aimed at investigating the suitability of polymer-lipid hybrid nanomedicines (HNMeds) as novel siRNA delivery platforms for locoregional therapy of glioblastoma. Two HNMed formulations were developed from poly(lactic-co-glycolic acid) polymer and a cationic lipid: 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP) or 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol (DC-Chol). After characterization of the HNMeds, a model siRNA was complexed onto their surface to form HNMed/siRNA complexes. The physicochemical properties and siRNA binding ability of complexes were assessed over a range of nitrogen-to-phosphate (N/P) ratios to optimize the formulations. At the optimal N/P ratio of 10, complexes effectively bound siRNA and improved its protection from enzymatic degradation. Using the NIH3T3 mouse fibroblast cell line, DOTAP-based HNMeds were shown to possess higher cytocompatibility in vitro over the DC-Chol-based ones. As proof-of-concept, uptake and bioefficacy of formulations were also assessed in vitro on U87MG human glioblastoma cell line expressing luciferase gene. Complexes were able to deliver anti-luciferase siRNA and induce a remarkable suppression of gene expression. Noteworthy, the effect of DOTAP-based formulation was not only about three-times higher than DC-Chol-based one, but also comparable to lipofectamine model transfection reagent. These findings set the basis to exploit this nanosystem for silencing relevant GB-related genes in further in vitro and in vivo studies.


Fatty Acids, Monounsaturated , Glioblastoma , Liposomes , Quaternary Ammonium Compounds , Mice , Animals , Humans , Liposomes/chemistry , Polymers/chemistry , RNA, Small Interfering , Glioblastoma/genetics , Glioblastoma/therapy , NIH 3T3 Cells , Nanomedicine , Lipids/chemistry
...