Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.771.682
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
Biomaterials ; 312: 122733, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39106819

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) demonstrates unique characteristics in anticancer therapies as it selectively induces apoptosis in cancer cells. However, most cancer cells are TRAIL-resistant. Odanacatib (ODN), a cathepsin K inhibitor, is considered a novel sensitizer for cancer treatment. Combination therapy between TRAIL and sensitizers is considered a potent platform that improves TRAIL-based anticancer therapies beyond TRAIL monotherapy. Herein, we developed ODN loaded poly(lactic-co-glycolic) nanoparticles conjugated to GST-TRAIL (TRAIL-ODN-PLGA-NPs) to target and treat TRAIL-resistant cancer. TRAIL-ODN-PLGA-NPs demonstrated a significant increase in cellular uptake via death receptors (DR5 and DR4) on surface of cancer cells. TRAIL-ODN-PLGA-NPs exposure destroyed more TRAIL-resistant cells compared to a single treatment with free drugs. The released ODN decreased the Raptor protein, thereby increasing damage to mitochondria by elevating reactive oxygen species (ROS) generation. Additionally, Bim protein stabilization improved TRAIL-resistant cell sensitization to TRAIL-induced apoptosis. The in vivo biodistribution study revealed that TRAIL-ODN-PLGA-NPs demonstrated high location and retention in tumor sites via the intravenous route. Furthermore, TRAIL-ODN-PLGA-NPs significantly inhibited xenograft tumor models of TRAIL-resistant Caki-1 and TRAIL-sensitive MDA-MB-231 cells.The inhibition was associated with apoptosis activation, Raptor protein stabilizing Bim protein downregulation, Bax accumulation, and mitochondrial ROS generation elevation. Additionally, TRAIL-ODN-PLGA-NPs affected the tumor microenvironment by increasing tumor necrosis factor-α and reducing interleukin-6. In conclusion, we evealed that our formulation demonstrated synergistic effects against TRAIL compared with the combination of free drug in vitro and in vivo models. Therefore, TRAIL-ODN-PLGA-NPs may be a novel candidate for TRAIL-induced apoptosis in cancer treatment.


Assuntos
Antineoplásicos , Compostos de Bifenilo , Resistencia a Medicamentos Antineoplásicos , Nanopartículas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ligante Indutor de Apoptose Relacionado a TNF , Animais , Feminino , Humanos , Camundongos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Compostos de Bifenilo/uso terapêutico , Compostos de Bifenilo/farmacologia , Compostos de Bifenilo/química , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Espécies Reativas de Oxigênio/metabolismo , Distribuição Tecidual , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
2.
Biomaterials ; 312: 122707, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39121729

RESUMO

Polypyrimidine tract-binding protein 1 (PTBP1) regulates numerous alternative splicing events during tumor progression and neurogenesis. Previously, PTBP1 downregulation was reported to convert astrocytes into functional neurons; however, how PTBP1 regulates astrocytic physiology remains unclear. In this study, we revealed that PTBP1 modulated glutamate uptake via ATP1a2, a member of Na+/K+-ATPases, and glutamate transporters in astrocytes. Ptbp1 knockdown altered mitochondrial function and energy metabolism, which involved PTBP1 regulating mitochondrial redox homeostasis via the succinate dehydrogenase (SDH)/Nrf2 pathway. The malfunction of glutamate transporters following Ptbp1 knockdown resulted in enhanced excitatory synaptic transmission in the cortex. Notably, we developed a biomimetic cationic triblock polypeptide system, i.e., polyethylene glycol44-polylysine30-polyleucine10 (PEG44-PLL30-PLLeu10) with astrocytic membrane coating to deliver Ptbp1 siRNA in vitro and in vivo, which approach allowed Ptbp1 siRNA to efficiently cross the blood-brain barrier and target astrocytes in the brain. Collectively, our findings suggest a framework whereby PTBP1 serves as a modulator in glutamate transport machinery, and indicate that biomimetic methodology is a promising route for in vivo siRNA delivery.


Assuntos
Astrócitos , Ácido Glutâmico , Ribonucleoproteínas Nucleares Heterogêneas , Homeostase , Fator 2 Relacionado a NF-E2 , Proteína de Ligação a Regiões Ricas em Polipirimidinas , RNA Interferente Pequeno , Animais , Astrócitos/metabolismo , Ácido Glutâmico/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Fator 2 Relacionado a NF-E2/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Camundongos , Transdução de Sinais , Membrana Celular/metabolismo , Camundongos Endogâmicos C57BL , Masculino , Humanos , Mitocôndrias/metabolismo
3.
Protein Expr Purif ; 225: 106595, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39197671

RESUMO

We previously reported a chromatography system for purifying immunoglobulin M (IgM) using N,N,N',N'-ethylenediaminetetrakis(methylenephosphonic acid)-modified zirconia particles that selectively absorb immunoglobulins. Here, we report a simple procedure for preparing biotinylated IgM from hybridoma culture medium using this zirconia-based chromatography system. The culture medium of an IgM-producing hybridoma cell line was used as the starting sample solution, and the IgM in the medium was concentrated and partially purified by zirconia chromatography. Next, 9-(biotinamido)-4,7-dioxanonanoic acid N-succinimidyl ester was added to react with the proteins in the sample. Subsequently, only the biotinylated IgM was isolated by Capto Core 400 polishing column chromatography. The entire process was easy to perform, could be completed within 2 h, and provided highly pure biotin-labeled IgM. This procedure is expected to be applicable to the labeling of IgM with various compounds and drugs.


Assuntos
Biotinilação , Meios de Cultura , Hibridomas , Imunoglobulina M , Imunoglobulina M/química , Imunoglobulina M/isolamento & purificação , Animais , Meios de Cultura/química , Camundongos , Zircônio/química , Biotina/química
4.
Food Chem ; 462: 140920, 2025 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-39208732

RESUMO

The use of direct injection ion mobility mass spectrometry (DI-IM-MS) to detect and identify betacyanin pigments in A. hortensis 'rubra' extracts was explored for the first time, with results compared to conventional LC-MS/MS analysis. The anti-inflammatory activities of leaf and seed extracts, alongside purified amaranthin and celosianin pigments, were investigated using a model of lipopolysaccharide (LPS)-activated murine macrophages. Extracts and purified pigments significantly inhibited the production of prostaglandin E2 and NO by up to 90% and 70%, respectively, and reduced the expression of Il6, Il1b, Nos2, and Cox2. Leaf and seed extracts also decreased secretion of Il6 and Il1b cytokines and reduced protein levels of Nos2 and Cox2. Furthermore, extracts and purified pigments demonstrated potent dose-dependent radical scavenging activity in a cellular antioxidant activity assay (CAA) without any cytotoxic effects. Our research highlights the promising biological potential of edible, climate-resilient A. hortensis 'rubra' as a valuable source of bioactive compounds.


Assuntos
Lipopolissacarídeos , Macrófagos , Estresse Oxidativo , Extratos Vegetais , Camundongos , Animais , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Células RAW 264.7 , Estresse Oxidativo/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Lipopolissacarídeos/farmacologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Anti-Inflamatórios/isolamento & purificação , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Ciclo-Oxigenase 2/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Espectrometria de Massas em Tandem
5.
Biomaterials ; 313: 122753, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39217793

RESUMO

Non-viral nanoparticles (NPs) have seen heightened interest as a delivery method for a variety of clinically relevant nucleic acid cargoes in recent years. While much of the focus has been on lipid NPs, non-lipid NPs, including polymeric NPs, have the possibility of improved efficacy, safety, and targeting, especially to non-liver organs following systemic administration. A safe and effective systemic approach for intracellular delivery to the lungs could overcome limitations to intratracheal/intranasal delivery of NPs and improve clinical benefit for a range of diseases including cystic fibrosis. Here, engineered biodegradable poly (beta-amino ester) (PBAE) NPs are shown to facilitate efficient delivery of mRNA to primary human airway epithelial cells from both healthy donors and individuals with cystic fibrosis. Optimized NP formulations made with differentially endcapped PBAEs and systemically administered in vivo lead to high expression of mRNA within the lungs in BALB/c and C57 B/L mice without requiring a complex targeting ligand. High levels of mRNA-based gene editing were achieved in an Ai9 mouse model across bronchial, epithelial, and endothelial cell populations. No toxicity was observed either acutely or over time, including after multiple systemic administrations of the NPs. The non-lipid biodegradable PBAE NPs demonstrate high levels of transfection in both primary human airway epithelial cells and in vivo editing of lung cell types that are targets for numerous life-limiting diseases particularly single gene disorders such as cystic fibrosis and surfactant deficiencies.


Assuntos
Pulmão , Camundongos Endogâmicos C57BL , Nanopartículas , Polímeros , RNA Mensageiro , Animais , Pulmão/metabolismo , Humanos , Nanopartículas/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Polímeros/química , Camundongos Endogâmicos BALB C , Camundongos , Fibrose Cística , Feminino , Ligantes , Células Epiteliais/metabolismo
6.
Biomaterials ; 313: 122777, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39222545

RESUMO

Telomere length plays a crucial role in cellular aging and the risk of diseases. Unlike normal cells, cancer cells can extend their own survival by maintaining telomere stability through telomere maintenance mechanism. Therefore, regulating the lengths of telomeres have emerged as a promising approach for anti-cancer treatment. In this study, we introduce a nanoscale octopus-like structure designed to induce physical entangling of telomere, thereby efficiently triggering telomere dysfunction. The nanoscale octopus, composed of eight-armed PEG (8-arm-PEG), are functionalized with cell penetrating peptide (TAT) to facilitate nuclear entry and are covalently bound to N-Methyl Mesoporphyrin IX (NMM) to target G-quadruplexes (G4s) present in telomeres. The multi-armed configuration of the nanoscale octopus enables targeted binding to multiple G4s, physically disrupting and entangling numerous telomeres, thereby triggering telomere dysfunction. Both in vitro and in vivo experiments indicate that the nanoscale octopus significantly inhibits cancer cell proliferation, induces apoptosis through telomere entanglement, and ultimately suppresses tumor growth. This research offers a novel perspective for the development of innovative anti-cancer interventions and provides potential therapeutic options for targeting telomeres.


Assuntos
Apoptose , Telômero , Telômero/metabolismo , Apoptose/efeitos dos fármacos , Humanos , Animais , Linhagem Celular Tumoral , Camundongos , Quadruplex G/efeitos dos fármacos , Camundongos Nus , Polietilenoglicóis/química , Proliferação de Células/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Neoplasias/patologia , Neoplasias/tratamento farmacológico , Feminino , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/farmacologia , Nanoestruturas/química
7.
Biomaterials ; 312: 122714, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39079462

RESUMO

Osteosarcoma, a malignant bone tumor often characterized by high hedgehog signaling activity, residual tumor cells, and substantial bone defects, poses significant challenges to both treatment response and postsurgical recovery. Here, we developed a nanocomposite hydrogel for the sustained co-delivery of bioactive magnesium ions, anti-PD-L1 antibody (αPD-L1), and hedgehog pathway antagonist vismodegib, to eradicate residual tumor cells while promoting bone regeneration post-surgery. In a mouse model of tibia osteosarcoma, this hydrogel-mediated combination therapy led to remarkable tumor growth inhibition and hence increased animal survival by enhancing the activity of tumor-suppressed CD8+ T cells. Meanwhile, the implanted hydrogel improved the microenvironment of osteogenesis through long-term sustained release of Mg2+, facilitating bone defect repair by upregulating the expression of osteogenic genes. After 21 days, the expression levels of ALP, COL1, RUNX2, and BGLAP in the Vis-αPD-L1-Gel group were approximately 4.1, 5.1, 5.5, and 3.4 times higher than those of the control, respectively. We believe that this hydrogel-based combination therapy offers a potentially valuable strategy for treating osteosarcoma and addressing the tumor-related complex bone diseases.


Assuntos
Neoplasias Ósseas , Hidrogéis , Imunoterapia , Nanocompostos , Osteossarcoma , Osteossarcoma/patologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/terapia , Animais , Hidrogéis/química , Nanocompostos/química , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Neoplasias Ósseas/terapia , Camundongos , Imunoterapia/métodos , Linhagem Celular Tumoral , Regeneração Óssea/efeitos dos fármacos , Humanos , Osteogênese/efeitos dos fármacos , Antígeno B7-H1/metabolismo , Camundongos Endogâmicos BALB C , Magnésio/química
8.
Biomaterials ; 312: 122720, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39084098

RESUMO

Mesenchymal stem cells (MSCs) are expected to be useful therapeutics in osteoarthritis (OA), the most common joint disorder characterized by cartilage degradation. However, evidence is limited with regard to cartilage repair in clinical trials because of the uncontrolled differentiation and weak cartilage-targeting ability of MSCs after injection. To overcome these drawbacks, here we synthesized CuO@MSN nanoparticles (NPs) to deliver Sox9 plasmid DNA (favoring chondrogenesis) and recombinant protein Bmp7 (inhibiting hypertrophy). After taking up CuO@MSN/Sox9/Bmp7 (CSB NPs), the expressions of chondrogenic markers were enhanced while hypertrophic markers were decreased in response to these CSB-engineered MSCs. Moreover, a cartilage-targeted peptide (designated as peptide W) was conjugated onto the surface of MSCs via a click chemistry reaction, thereby prolonging the residence time of MSCs in both the knee joint cavity of mice and human-derived cartilage. In a surgery-induced OA mouse model, the NP and peptide dual-modified W-CSB-MSCs showed an enhancing therapeutic effect on cartilage repair in knee joints compared with other engineered MSCs after intra-articular injection. Most importantly, W-CSB-MSCs accelerated cartilage regeneration in damaged cartilage explants derived from OA patients. Thus, this new peptide and NPs dual engineering strategy shows potential for clinical applications to boost cartilage repair in OA using MSC therapy.


Assuntos
Diferenciação Celular , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Nanopartículas , Osteoartrite , Peptídeos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Osteoartrite/terapia , Osteoartrite/patologia , Nanopartículas/química , Humanos , Diferenciação Celular/efeitos dos fármacos , Peptídeos/química , Transplante de Células-Tronco Mesenquimais/métodos , Condrogênese/efeitos dos fármacos , Camundongos , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Cartilagem Articular/patologia , Cartilagem Articular/efeitos dos fármacos , Proteína Morfogenética Óssea 7/química , Proteína Morfogenética Óssea 7/farmacologia , Engenharia Tecidual/métodos , Regeneração/efeitos dos fármacos
9.
Biomaterials ; 312: 122711, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39088911

RESUMO

The unsuitable deformation stimulus, harsh urine environment, and lack of a regenerative microenvironment (RME) prevent scaffold-based urethral repair and ultimately lead to irreversible urethral scarring. The researchers clarify the optimal elastic modulus of the urethral scaffolds for urethral repair and design a multilayered PVA hydrogel scaffold for urethral scar-free healing. The inner layer of the scaffold has self-healing properties, which ensures that the wound effectively resists harsh urine erosion, even when subjected to sutures. In addition, the scaffold's outer layer has an extracellular matrix-like structure that synergizes with adipose-derived stem cells to create a favorable RME. In vivo experiments confirm successful urethral scar-free healing using the PVA multilayered hydrogel scaffold. Further mechanistic study shows that the PVA multilayer hydrogel effectively resists the urine-induced inflammatory response and accelerates the transition of urethral wound healing to the proliferative phase by regulating macrophage polarization, thus providing favorable conditions for urethral scar-free healing. This study provides mechanical criteria for the fabrication of urethral tissue-engineered scaffolds, as well as important insights into their design.


Assuntos
Módulo de Elasticidade , Hidrogéis , Alicerces Teciduais , Uretra , Cicatrização , Alicerces Teciduais/química , Animais , Hidrogéis/química , Engenharia Tecidual/métodos , Camundongos , Regeneração , Cicatriz/patologia , Masculino , Microambiente Celular , Ratos Sprague-Dawley , Células-Tronco/citologia
10.
J Ethnopharmacol ; 336: 118741, 2025 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-39197801

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Amyotrophic lateral sclerosis (ALS) is a fetal neuromuscular disorder characterized by the gradual deterioration of motor neurons. Semen Strychni pulveratum (SSP), a processed version of Semen Strychni (SS) powder, is widely used to treat ALS in China. Vomicine is one of the most primary components of SS. However, their pharmacological effects and mechanisms for ALS remain elusive. AIM OF THE STUDY: This study aimed to evaluate the neuroprotective and anti-neuroinflammatory effects of SSP and vomicine, as well as to explore their protective roles in ALS and the underlying mechanisms. MATERIALS AND METHODS: In vivo, 8-week-old hSOD1-WT mice and hSOD1-G93A mice were orally administered different concentrations of SSP (SSP-L = 5.46 mg/ml, SSP-M = 10.92 mg/ml or SSP-H = 16.38 mg/ml) once every other day for 8 weeks. A series of experiments, including body weight measurement, footprint tests, Hematoxylin & Eosin staining, and Nissl staining, were performed to evaluate the preventive effect of SSP. Immunofluorescence staining, western blotting, and RT-qPCR were subsequently performed to evaluate activation of the cGAS-STING-TBK1 pathway in the spinal cord. In vitro, hSOD1G93A NSC-34 cells were treated with vomicine to further explore the pharmacological mechanism of vomicine in the treatment of ALS via the cGAS-STING-TBK1 pathway. RESULTS: SSP improved motor function, body weight loss, gastrocnemius muscle atrophy, and motor neuron loss in the spine and cortex of hSOD1-G93A mice. Furthermore, the cGAS-STING-TBK1 pathway was activated in the spinal cord of hSOD1-G93A mice, with activation predominantly observed in neurons and microglia. However, the levels of cGAS, STING, and pTBK1 proteins and cGAS, IRF3, IL-6, and IL-1ß mRNA were reversed following intervention with SSP. Vomicine not only downregulated the levels of cGAS, TBK1, IL-6 and IFN-ß mRNA, but also the levels of cGAS and STING protein in hSOD1G93A NSC-34 cells. CONCLUSION: This study demonstrated that SSP and vomicine exert neuroprotective and anti-neuroinflammatory effects in the treatment of ALS. SSP and vomicine may reduce neuroinflammation by regulating the cGAS-STING-TBK1 pathway, and could thereby play a role in ALS treatment.


Assuntos
Esclerose Lateral Amiotrófica , Proteínas de Membrana , Fármacos Neuroprotetores , Nucleotidiltransferases , Proteínas Serina-Treonina Quinases , Animais , Proteínas Serina-Treonina Quinases/metabolismo , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/metabolismo , Camundongos , Proteínas de Membrana/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Nucleotidiltransferases/metabolismo , Masculino , Transdução de Sinais/efeitos dos fármacos , Camundongos Transgênicos , Doenças Neuroinflamatórias/tratamento farmacológico , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Modelos Animais de Doenças
11.
Biomaterials ; 313: 122773, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39217794

RESUMO

The development of artificial Antigen Presenting Cells (aAPCs) has led to improvements in adoptive T cell therapy (ACT), an immunotherapy, for cancer treatment. aAPCs help to streamline the consistent production and expansion of T cells, thus reducing the time and costs associated with ACT. However, several issues still exist with ACT, such as insufficient T cell potency, which diminishes the translational potential for ACT. While aAPCs have been used primarily to increase production efficiency of T cells for ACT, the intrinsic properties of a biomaterial-based aAPC may affect T cell phenotype and function. In CD8+ T cells, reactive oxygen species (ROS) and oxidative stress accumulation can activate Forkhead box protein O1 (FOXO1) to transcribe antioxidants which reduce ROS and improve memory formation. Alginate, a biocompatible and antioxidant rich biomaterial, is promising for incorporation into an aAPC formulation to modulate T cell phenotype. To investigate its utility, a novel alginate-based aAPC platform was developed that preferentially expanded CD8+ T cells with memory related features. Alginate-based aAPCs allowed for greater control of CD8+ T cell qualities, including, significantly improved in vivo persistence and augmented in vivo anti-tumor T cell responses.


Assuntos
Alginatos , Células Apresentadoras de Antígenos , Linfócitos T CD8-Positivos , Memória Imunológica , Imunoterapia Adotiva , Alginatos/química , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Animais , Imunoterapia Adotiva/métodos , Células Apresentadoras de Antígenos/imunologia , Memória Imunológica/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Humanos , Proliferação de Células/efeitos dos fármacos
12.
J Colloid Interface Sci ; 678(Pt A): 20-29, 2025 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-39178688

RESUMO

Bacterial biofilms present a profound challenge to global public health, often resulting in persistent and recurrent infections that resist treatment. Chemodynamic therapy (CDT), leveraging the conversion of hydrogen peroxide (H2O2) to highly reactive hydroxyl radicals (•OH), has shown potential as an antibacterial approach. Nonetheless, CDT struggles to eliminate biofilms due to limited endogenous H2O2 and the protective extracellular polymeric substances (EPS) within biofilms. This study introduces a multifunctional nanoplatform designed to self-supply H2O2 and generate nitric oxide (NO) to overcome these hurdles. The nanoplatform comprises calcium peroxide (CaO2) for sustained H2O2 production, a copper-based metal-organic framework (HKUST-1) encapsulating CaO2, and l-arginine (l-Arg) as a natural NO donor. When exposed to the acidic microenvironment within biofilms, the HKUST-1 layer decomposes, releasing Cu2+ ions and l-Arg, and exposing the CaO2 core to initiate a cascade of reactions producing reactive species such as H2O2, •OH, and superoxide anions (•O2-). Subsequently, H2O2 catalyzes l-Arg to produce NO, which disperses the biofilm and reacts with •O2- to form peroxynitrite, synergistically eradicating bacteria with •OH. In vitro assays demonstrated the nanoplatform's remarkable antibiofilm efficacy against both Gram-positive Methicillin-resistant Staphylococcus aureus and Gram-negative Pseudomonas aeruginosa, significantly reducing bacterial viability and EPS content. In vivo mouse model experiments validated the nanoplatform's effectiveness in eliminating biofilms and promoting infected wound healing without adverse effects. This study represents a breakthrough in overcoming traditional CDT limitations by integrating self-supplied H2O2 with NO's biofilm-disrupting capabilities, offering a promising therapeutic strategy for biofilm-associated infection.


Assuntos
Antibacterianos , Biofilmes , Peróxido de Hidrogênio , Óxido Nítrico , Biofilmes/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Peróxido de Hidrogênio/química , Animais , Óxido Nítrico/metabolismo , Óxido Nítrico/química , Óxido Nítrico/farmacologia , Camundongos , Antibacterianos/farmacologia , Antibacterianos/química , Pseudomonas aeruginosa/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Nanopartículas/química , Arginina/química , Arginina/farmacologia , Cobre/química , Cobre/farmacologia , Tamanho da Partícula , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Propriedades de Superfície , Peróxidos
13.
J Colloid Interface Sci ; 678(Pt A): 42-52, 2025 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-39180847

RESUMO

Photodynamic therapy (PDT) and catalytic therapy were promising treatment modes, but tumor hypoxia and poor catalytic activity severely limited their efficacies. Herein, using a porphyrin metal-organic framework (PCN-224) as nanocarrier, a platinum/palladium (Pt/Pd) dual-modified PCN-224 nanoprobe (PCN-224-Pt@Pd) with strong peroxidase (POD)/catalase (CAT)-like activities was developed, achieving photothermal-promoted PDT/catalytic therapy. Compared with single ultrasmall Pt modifying, CAT-like activity of Pt/Pd dual-modifying increased oxygen concentration from 6.24 to 9.35 mg/L, which improved singlet oxygen (1O2) yield from 63.8 % to 82.9 %. Moreover, POD-like activity of Pt/Pd dual-modifying significantly accelerated hydroxyl radicals (·OH) generation. Importantly, PCN-224-Pt@Pd possessed near-infrared II (NIR-II) photothermal effect with a high efficiency (55.6 %), which further promoted ·OH production. Under combined therapy of PCN-224-Pt@Pd, the cell survival rate greatly reduced to 5.8 %, and the tumors were cured, suggesting NIR-II photothermal-enhanced PDT/catalytic therapy.


Assuntos
Raios Infravermelhos , Estruturas Metalorgânicas , Paládio , Fotoquimioterapia , Fármacos Fotossensibilizantes , Platina , Porfirinas , Paládio/química , Paládio/farmacologia , Platina/química , Platina/farmacologia , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Catálise , Porfirinas/química , Porfirinas/farmacologia , Humanos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Camundongos , Antineoplásicos/química , Antineoplásicos/farmacologia , Tamanho da Partícula , Propriedades de Superfície , Ensaios de Seleção de Medicamentos Antitumorais , Proliferação de Células/efeitos dos fármacos
14.
J Colloid Interface Sci ; 678(Pt A): 108-118, 2025 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-39182385

RESUMO

Chemotherapy is the main clinical treatment for ovarian cancer, but still faces challenges of low drug targeting efficiency and insufficient drug permeability. Drug-loaded nanoparticle collectives, which are actuated by magnetic field, could be targeted to a designated location and achieve targeted drug delivery. In this work, we report a strategy that utilizes magnetic mesoporous silica nanoparticles loaded with cis-diaminodichloroplatinum (Fe3O4@SiO2-CDDP) for targeted delivery of chemotherapeutic drugs and enhances penetration into deep tumors. The Fe3O4@SiO2-CDDP collectives actively moved to the target tumor site, and this movement was regulated by a magnetic actuation system. Under the action of a torque-force hybrid magnetic field (TFMF), Fe3O4@SiO2-CDDP could further penetrate into the interior of tumors and achieve pH-responsive drug release in the tumor environment. The feasibility of this strategy was verified in three-dimensional cell spheres in vitro and in a tumor-bearing mouse model in vivo. This magnetically actuated nanoparticle collectives enhanced drug penetration strategy provides a new paradigm for targeted drug delivery and potentiated tumor therapy.


Assuntos
Antineoplásicos , Cisplatino , Neoplasias Ovarianas , Dióxido de Silício , Cisplatino/farmacologia , Cisplatino/química , Cisplatino/farmacocinética , Cisplatino/administração & dosagem , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Feminino , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Humanos , Camundongos , Dióxido de Silício/química , Nanopartículas de Magnetita/química , Tamanho da Partícula , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Sistemas de Liberação de Medicamentos , Propriedades de Superfície , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Campos Magnéticos , Nanopartículas/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Camundongos Endogâmicos BALB C
15.
J Colloid Interface Sci ; 678(Pt A): 260-271, 2025 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-39197369

RESUMO

The demand for breast mesh with antitumor properties is critical in post-mastectomy breast reconstruction to prevent local tumor recurrence. Molybdenum-based oxide (MoOx) exhibits enzyme-like activities by catalyzing endogenous hydrogen peroxide to produce reactive oxygen species for inducing tumor cell apoptosis. However, its catalytic activity is limited by insufficient active sites. Herein, a defect engineering strategy is proposed to create redox nanozymes with multiple enzymatic activities by incorporating Fe into MoOx (Fe-MoOv). Fe-MoOv is subsequently integrated into polycaprolactone (PCL) to fabricate breast meshes for establishing an enzyme-catalyzed antitumor platform. The doping of Fe into MoOx formed numerous defect sites, including oxygen vacancies (OV) and Fe substitution sites, synergistically boosting the binding capacity and catalytic activity of Fe-MoOv. Density functional theory calculations demonstrated that the outstanding peroxidase-like catalytic activity of Fe-MoOv resulted from the synergy between OV and Fe sites. Additionally, OV contributes to the localized surface plasmon resonance effect, enhancing the photothermal capability of the PCL/Fe-MoOv mesh. Upon near-infrared laser exposure, the catalytic activity of the PCL/Fe-MoOv mesh is further improved, leading to increased generation of reactive oxygen species and enhanced antitumor efficacy, achieving 86.7% tumor cell mortality, a 264% enhancement compared to the PCL/MoOx mesh.


Assuntos
Antineoplásicos , Molibdênio , Óxidos , Molibdênio/química , Humanos , Catálise , Antineoplásicos/química , Antineoplásicos/farmacologia , Óxidos/química , Óxidos/farmacologia , Ferro/química , Ferro/metabolismo , Neoplasias da Mama/patologia , Espécies Reativas de Oxigênio/metabolismo , Feminino , Ensaios de Seleção de Medicamentos Antitumorais , Camundongos , Poliésteres/química , Animais , Proliferação de Células/efeitos dos fármacos , Propriedades de Superfície , Tamanho da Partícula , Telas Cirúrgicas , Teoria da Densidade Funcional , Apoptose/efeitos dos fármacos
16.
J Biomech Eng ; 147(1)2025 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-39297758

RESUMO

Collagen XI is ubiquitous in tissues such as joint cartilage, cancellous bone, muscles, and tendons and is an important contributor during a crucial part in fibrillogenesis. The COL11A1 gene encodes one of three alpha chains of collagen XI. The present study elucidates the role of collagen XI in the establishment of mechanical properties of tendons and ligaments. We investigated the mechanical response of three tendons and one ligament tissues from wild type and a targeted mouse model null for collagen XI: Achilles tendon (ACH), the flexor digitorum longus tendon (FDL), the supraspinatus tendon (SST), and the anterior cruciate ligament (ACL). Area was substantially lower in Col11a1ΔTen/ΔTen ACH, FDL, and SST. Maximum load and maximum stress were significantly lower in Col11a1ΔTen/ΔTen ACH and FDL. Stiffness was lower in Col11a1ΔTen/ΔTen ACH, FDL, and SST. Modulus was reduced in Col11a1ΔTen/ΔTen FDL and SST (both insertion site and midsubstance). Collagen fiber distributions were more aligned under load in both wild type group and Col11a1ΔTen/ΔTen groups. Results also revealed that the effect of collagen XI knockout on collagen fiber realignment is tendon-dependent and location-dependent (insertion versus midsubstance). In summary, this study clearly shows that the regulatory role of collagen XI on tendon and ligament is tissue specific and that joint hypermobility in type II Stickler's Syndrome may in part be due to suboptimal mechanical response of the soft tissues surrounding joints.


Assuntos
Colágeno Tipo XI , Ligamentos , Tendões , Animais , Camundongos , Fenômenos Biomecânicos , Tendões/metabolismo , Tendões/fisiologia , Ligamentos/metabolismo , Ligamentos/fisiologia , Colágeno Tipo XI/metabolismo , Colágeno Tipo XI/genética , Fenômenos Mecânicos , Estresse Mecânico
17.
Mol Med Rep ; 31(1)2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39422036

RESUMO

The nuclear receptor subfamily 4 group A member 1 (NR4A1) gene plays a crucial role in both osteoporosis and adipogenesis. The present study investigated the mechanisms by which NR4A1 influences osteoblastogenesis and adipogenesis in human bone marrow­derived mesenchymal stem cells (BMD­MSCs). NR4A1 was overexpressed or knocked down in mouse MC3T3­E1 osteoblast cells and 3T3­L1 adipocyte cells, as well as in PCS­500­012, a BMD­MSC line. The alkaline phosphatase (ALP) assay and Alizarin Red S staining were performed using MC3T3­E1 and BMD­MSCs to assess ALP activity and mineralization, while Oil Red O staining was used to assess the lipid content in 3T3­L1 cells and BMD­MSCs. Total RNA was isolated from control, NR4A1­overexpressing and NR4A1 small interfering RNA (siRNA; siNR4A1)­treated BMD­MSCs. RNA sequencing (RNA­seq) was performed to identify differentially expressed genes, followed by ingenuity pathway analysis (IPA) to determine the role of NR4A1 in osteoblastogenesis and adipogenesis. NR4A1 or Nr4a1 knockdown tended to increase ALP activity and significantly increased calcification in BMD­MSCs (P<0.005) and MC3T3­E1 cells (P<0.005), respectively. By contrast, NR4A1 or Nr4a1 overexpression significantly decreased ALP activity and calcification. NR4A1 or Nr4a1 knockdown and overexpression significantly decreased and increased adipogenesis, respectively, in BMD­MSCs (P<0.005 and <0.05, respectively) and 3T3­L1 cells (P<0.005 in both). Treatments of BMD­MSCs with an NR4A1 antagonist, 1,1­bis(3'­indolyl)­1­(p­hydroxyphenyl) methane and siNR4A1 showed similar results. RNA­seq and IPA in control, NR4A1 knockdown and NR4A1 overexpressing cells indicated that Notch signaling mediated the effects of NR4A1 in osteoblastogenesis and adipogenesis. Expression of mastermind­like transcriptional coactivator 3 was reduced in the Notch signaling pathway in cells treated with siNR4A1. In conclusion, NR4A1 suppressed osteoblastogenesis and promotes adipogenesis in human BMD­MSCs. The present study also suggested that NR4A1 plays a role in the progression of osteoporosis and adipogenesis by modulating the Notch signaling cascade.


Assuntos
Adipogenia , Diferenciação Celular , Células-Tronco Mesenquimais , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Osteoblastos , Osteogênese , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Humanos , Adipogenia/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Camundongos , Osteoblastos/metabolismo , Osteoblastos/citologia , Animais , Osteogênese/genética , Células 3T3-L1 , Transdução de Sinais
18.
Methods Mol Biol ; 2857: 159-167, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39348064

RESUMO

Microglia and astrocytes are the main components of the central nervous system (CNS). Upon activation, microglia is able to phagocyte cell debris, pathogens, and toxins; astrocytes support neuronal functions, blood-brain barrier (BBB) homeostasis, and neurotransmitter uptake and metabolism. Furthermore, both cell types can produce cytokines and chemokines. Aging impacts microglia and astrocytes by promoting the production of pro-inflammatory cytokines, impairing microglial phagocytosis and motility and astrocyte glutamate uptake. During neurodegenerative and neuroinflammatory diseases, the aging process may be accelerated contributing to the alteration of CNS glial cells functions. Multiple sclerosis (MS) is an autoimmune, demyelinating disease in which immunosenescence can promote the conversion from relapsing-remitting form to progressive disease. The murine model of experimental autoimmune encephalomyelitis (EAE) allows to investigate MS pathogenesis. Furthermore, EAE can be developed as acute or progressive, mimicking different forms of human MS. Microglia and astrocytes report morphological and functional changes during neuroinflammation that can be investigated in different ways. We here present a protocol for the study of glial cell activation in the spinal cord tissue of EAE mice.


Assuntos
Astrócitos , Encefalomielite Autoimune Experimental , Gliose , Microglia , Medula Espinal , Animais , Microglia/metabolismo , Microglia/patologia , Camundongos , Medula Espinal/patologia , Medula Espinal/metabolismo , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/metabolismo , Astrócitos/metabolismo , Astrócitos/patologia , Gliose/patologia , Gliose/metabolismo , Imunofluorescência/métodos , Modelos Animais de Doenças , Esclerose Múltipla/patologia , Esclerose Múltipla/metabolismo
19.
Methods Mol Biol ; 2857: 169-180, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39348065

RESUMO

Acute skeletal muscle injury initiates a process of necrosis, debris clearance, and ultimately tissue regeneration via myogenesis. While skeletal muscle stem cells (MuSCs) are responsible for populating the proliferative myogenic progenitor pool to fuel muscle repair, recruited and resident immune cells have a central role in the regulation of muscle regeneration via the execution of phagocytosis and release of soluble factors that act directly on MuSCs to regulate myogenic differentiation. Therefore, the timing of MuSC proliferation and differentiation is closely linked to the populations and behaviors of immune cells present within skeletal muscle. This has important implications for aging and muscle repair, as systemic changes in immune system function contribute to a decline in muscle regenerative capacity. Here, we present adapted protocols for the isolation of mononuclear cells from skeletal muscles for the quantification of immune cell populations using flow cytometry. We also describe a cardiotoxin skeletal muscle injury protocol and detail the expected outcomes including immune cell infiltration to the injured sites and formation of new myocytes. As immune cell function is substantially influenced by aging, we extend these approaches and outcomes to aged mice.


Assuntos
Envelhecimento , Modelos Animais de Doenças , Músculo Esquelético , Regeneração , Animais , Camundongos , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Envelhecimento/fisiologia , Desenvolvimento Muscular , Citometria de Fluxo/métodos , Diferenciação Celular , Proliferação de Células
20.
Biomaterials ; 313: 122757, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39178558

RESUMO

Recent progress in stem cell therapy has demonstrated the therapeutic potential of intravenous stem cell infusions for treating the life-threatening lung disease of pulmonary fibrosis (PF). However, it is confronted with limitations, such as a lack of control over cellular function and rapid clearance by the host after implantation. In this study, we developed an innovative PF therapy through tracheal administration of microfluidic-templated stem cell-laden microcapsules, which effectively reversed the progression of inflammation and fibrotic injury. Our findings highlight that hydrogel microencapsulation can enhance the persistence of donor mesenchymal stem cells (MSCs) in the host while driving MSCs to substantially augment their therapeutic functions, including immunoregulation and matrix metalloproteinase (MMP)-mediated extracellular matrix (ECM) remodeling. We revealed that microencapsulation activates the MAPK signaling pathway in MSCs to increase MMP expression, thereby degrading overexpressed collagen accumulated in fibrotic lungs. Our research demonstrates the potential of hydrogel microcapsules to enhance the therapeutic efficacy of MSCs through cell-material interactions, presenting a promising yet straightforward strategy for designing advanced stem cell therapies for fibrotic diseases.


Assuntos
Matriz Extracelular , Fatores Imunológicos , Fibrose Pulmonar , Células-Tronco , Cápsulas/química , Fatores Imunológicos/química , Fatores Imunológicos/farmacologia , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/terapia , Células Cultivadas , Humanos , Matriz Extracelular/química , Microfluídica , Sobrevivência Celular/efeitos dos fármacos , Hidrogéis/química , Masculino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Metaloproteinases da Matriz/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA