Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.790
Filtrar
1.
Biomaterials ; 313: 122769, 2025 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-39208698

RESUMO

Minimally invasive transcatheter interventional therapy utilizing cardiac occluders represents the primary approach for addressing congenital heart defects and left atrial appendage (LAA) thrombosis. However, incomplete endothelialization and delayed tissue healing after occluder implantation collectively compromise clinical efficacy. In this study, we have customized a recombinant humanized collagen type I (rhCol I) and developed an rhCol I-based extracellular matrix (ECM)-mimetic coating. The innovative coating integrates metal-phenolic networks with anticoagulation and anti-inflammatory functions as a weak cross-linker, combining them with specifically engineered rhCol I that exhibits high cell adhesion activity and elicits a low inflammatory response. The amalgamation, driven by multiple forces, effectively serves to functionalize implantable materials, thereby responding positively to the microenvironment following occluder implantation. Experimental findings substantiate the coating's ability to sustain a prolonged anticoagulant effect, enhance the functionality of endothelial cells and cardiomyocyte, and modulate inflammatory responses by polarizing inflammatory cells into an anti-inflammatory phenotype. Notably, occluder implantation in a canine model confirms that the coating expedites reendothelialization process and promotes tissue healing. Collectively, this tailored ECM-mimetic coating presents a promising surface modification strategy for improving the clinical efficacy of cardiac occluders.


Assuntos
Materiais Revestidos Biocompatíveis , Matriz Extracelular , Cicatrização , Animais , Matriz Extracelular/metabolismo , Cães , Humanos , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Cicatrização/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Células Endoteliais da Veia Umbilical Humana , Reepitelização/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos
2.
Mol Biol Rep ; 51(1): 1002, 2024 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-39305332

RESUMO

BACKGROUND: Recent 23Na-MRI reports show higher salt deposition in malignant breast tissue than in surrounding normal tissue. The effect of high salt on cancer progression remains controversial. Here, we investigated the direct effect of high salt on breast cancer progression in vitro. METHODS: Here, the impact of high salt on apoptosis, proliferation, cell cycle, adhesion, and migration of MDA-MB-231 and MCF-7 cells was studied using MTT, scratch, and clonogenic assays, as well as RT-PCR and flow cytometry. Gene expression was analyzed using Real-Time PCR and western blotting. The effect of high salt on global transcriptomics changes in MDA MB-231 cells was studied using RNA-sequencing analysis. RESULTS: Flow cytometry with Annexin V and CFSE revealed that high salt-induced dose-dependent apoptosis and inhibited proliferation. High salt-induced cell cycle arrest at the G1/S phase of the cell cycle. p-MDM2 is known to suppress p53, which plays a crucial role in regulating apoptosis and cell cycle arrest under cellular stress conditions. High salt treatment led to decreased p-MDM2 and increased p53 expression, suggesting that high salt induces apoptosis through p53 stabilization. decreased p-MDM2 and increased p53 expression. High salt also reduced migration and adhesion of cells in a dose-dependent manner suggesting its inhibitory effect on metastatic properties as evident from wound healing assay. RNA sequencing analysis revealed overexpression of tumor suppressor genes and genes associated with anti-tumor activity (PCDHGA11, EIF3CL, RAVER1, TNFSF15, RANBP3L) and under-expression of genes involved in cancer-promoting activity (MT1X, CLDN14, CSF-2). CONCLUSION: Our results unequivocally demonstrate the anti-tumor efficacy of high salt against breast cancer cells, suggesting its potential as a therapeutic strategy in cancer treatment.


Assuntos
Apoptose , Neoplasias da Mama , Movimento Celular , Proliferação de Células , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células MCF-7 , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Antineoplásicos/farmacologia , Cloreto de Sódio/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos
3.
J Mater Sci Mater Med ; 35(1): 58, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39348082

RESUMO

Tissue engineering scaffolds are three-dimensional structures that provide an appropriate environment for cellular attachment, proliferation, and differentiation. Depending on their specific purpose, these scaffolds must possess distinct features, including appropriate mechanical properties, porosity, desired degradation rate, and cell compatibility. This investigation aimed to fabricate a new nanocomposite scaffold using a 3D printing technique composed of poly(ε-caprolactone) (PCL)/Gelatin (GEL)/ordered mesoporous calcium-magnesium silicate (om-CMS) particles. Different weight ratios of om-CMS were added and optimized, and a series of scaffolds were constructed for comparison purposes, including PCL 50%/Gel 50%, PCL 50%/Gel 45%/om-CMS%5, and PCL 50%/Gel 40%/om-CMS%10. The optimized weight ratio of om-CMS was 10% without leaving behind negative effects on the filaments' structure. The scaffolds' physical and chemical properties were assessed using various techniques, and their degradation rate, bioactivity potential, cell viability, attachment, and ALP activity were evaluated in vitro. The results demonstrated that the PCL 50%/Gel 40%/om-CMS10% scaffold had promising potential for further studies in bone tissue regeneration.


Assuntos
Regeneração Óssea , Gelatina , Nanocompostos , Poliésteres , Impressão Tridimensional , Silicatos , Engenharia Tecidual , Alicerces Teciduais , Gelatina/química , Alicerces Teciduais/química , Poliésteres/química , Nanocompostos/química , Regeneração Óssea/efeitos dos fármacos , Engenharia Tecidual/métodos , Silicatos/química , Porosidade , Sobrevivência Celular/efeitos dos fármacos , Silicatos de Magnésio/química , Teste de Materiais , Humanos , Proliferação de Células/efeitos dos fármacos , Compostos de Cálcio/química , Magnésio/química , Materiais Biocompatíveis/química , Osso e Ossos , Animais , Adesão Celular/efeitos dos fármacos , Cálcio/química
4.
Colloids Surf B Biointerfaces ; 244: 114186, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39226849

RESUMO

Developing biomaterials with high osteogenic properties is crucial for achieving rapid bone repair and regeneration. This study focuses on the application of nanocrystal hydroxyapatite (nHAp) as a drug carrier to load Fu Yuan Huo Xue Decoction (FYHXD), a traditional Chinese medicine derived from Angelica sinensis, aiming to achieve improved efficacy in treating bone diseases such as osteoporosis. Through a facile physical adsorption approach, the FTIR result emerges new characteristic absorption peaks in the range of 1200-950 cm-1, proving the successful absorption of FYHXD onto the nHAp with a loading efficiency of 39.76 %. The modified nHAp exhibits a similar shape to the bone-derived hydroxyapatite nanocrystals, and their diameter increases slightly after modification. The drug release assay implies the rapid release of FYHXD in the first 10 h, followed by a continuously slow release within 70 h. The developed nHAp effectively enhances the adhesion, spreading, and proliferation of MC3T3-E1 cells in vitro, and significantly promotes their osteogenic differentiation, as indicated by increased alkaline phosphatase activity. Overall, the biocomposites hold great promise as active ingredients for integration into bone-associated biomaterials, offering the potential to stimulate spontaneous osteogenesis without requiring exogenous osteogenic factors.


Assuntos
Diferenciação Celular , Medicamentos de Ervas Chinesas , Durapatita , Nanopartículas , Osteogênese , Durapatita/química , Osteogênese/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Camundongos , Animais , Nanopartículas/química , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacologia , Proliferação de Células/efeitos dos fármacos , Medicina Tradicional Chinesa , Liberação Controlada de Fármacos , Fosfatase Alcalina/metabolismo , Tamanho da Partícula , Linhagem Celular , Adesão Celular/efeitos dos fármacos , Portadores de Fármacos/química
6.
Biomolecules ; 14(9)2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39334945

RESUMO

Various strategies have been employed to improve the reliability of 2D, 3D, and co-culture in vitro models of nonalcoholic fatty liver disease, including using extracellular matrix proteins such as collagen I to promote cell adhesion. While studies have demonstrated the significant benefits of culturing cells on collagen I, its effects on the HepG2 cell line after exposure to palmitate (PA) have not been investigated. Therefore, this study aimed to assess the effects of PA-induced lipotoxicity in HepG2 cultured in the absence or presence of collagen I. HepG2 cultured in the absence or presence of collagen I was exposed to PA, followed by analyses that assessed cell proliferation, viability, adhesion, cell death, mitochondrial respiration, reactive oxygen species production, gene and protein expression, and triacylglycerol accumulation. Culturing HepG2 on collagen I was associated with increased cell proliferation, adhesion, and expression of integrin receptors, and improved cellular spreading compared to culturing them in the absence of collagen I. However, PA-induced lipotoxicity was greater in collagen I-cultured HepG2 than in those cultured in the absence of collagen I and was associated with increased α2ß1 receptors. In summary, the present study demonstrated for the first time that collagen I-cultured HepG2 exhibited exacerbated cell death following exposure to PA through integrin-mediated death. The findings from this study may serve as a caution to those using 2D models or 3D scaffold-based models of HepG2 in the presence of collagen I.


Assuntos
Adesão Celular , Proliferação de Células , Sobrevivência Celular , Colágeno Tipo I , Humanos , Células Hep G2 , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Proliferação de Células/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Palmitatos/toxicidade , Palmitatos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Morte Celular/efeitos dos fármacos , Integrina alfa2beta1/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Integrinas/metabolismo , Integrinas/genética
7.
Int J Mol Sci ; 25(18)2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39337295

RESUMO

To repair damaged mesothelium tissue, which lines internal organs and cavities, a tissue engineering approach with mesothelial cells seeded to a functional nanostructured scaffold is a promising approach. Therefore, this study explored the uses of electrospun nanofiber membrane scaffolds (NMSs) as scaffolds for mesothelial cell culture and transplantation. We fabricated a composite NMS through electrospinning by blending polycaprolactone (PCL) with gelatin. The addition of gelatin enhanced the membrane's hydrophilicity while maintaining its mechanical strength and promoted cell attachment. The in vitro study demonstrated enhanced adhesion of mesothelial cells to the scaffold with improved morphology and increased phenotypic expression of key marker proteins calretinin and E-cadherin in PCL/gelatin compared to pure PCL NMSs. In vivo studies in rats revealed that only cell-seeded PCL/gelatin NMS constructs fostered mesothelial healing. Implantation of these constructs leads to the regeneration of new mesothelium tissue. The neo-mesothelium is similar to native mesothelium from hematoxylin and eosin (H&E) and immunohistochemical staining. Taken together, the PCL/gelatin NMSs can be a promising scaffold for mesothelial cell attachment, proliferation, and differentiation, and the cell/scaffold construct can be used in therapeutic applications to reconstruct a mesothelium layer.


Assuntos
Gelatina , Nanofibras , Poliésteres , Engenharia Tecidual , Alicerces Teciduais , Nanofibras/química , Gelatina/química , Alicerces Teciduais/química , Poliésteres/química , Animais , Ratos , Epitélio/efeitos dos fármacos , Engenharia Tecidual/métodos , Proliferação de Células/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/citologia , Células Cultivadas , Humanos
8.
Int J Mol Sci ; 25(18)2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39337558

RESUMO

Meningiomas are predominantly benign tumors, but there are also malignant forms that are associated with a poor prognosis. Like almost all tumors, meningiomas metabolize glucose as part of aerobic glycolysis (Warburg effect) for energy supply, so there are attempts to influence the prognosis of tumor diseases using a glucose-reduced diet. This altered metabolism leads to so called hallmarks of cancer, such as glycation and glycosylation. In this study, we investigated the influence of low (3 mM), normal (5.5 mM) and high glucose (15 mM) on a malignant meningioma cell line (IOMM-Lee, WHO grade 3). In addition, the influence of methylglyoxal, a by-product of glycolysis and a precursor for glycation, was investigated. Impedance-based methods (ECIS and RTCA) were used to study migration and invasion, and immunoblotting was used to analyze the expression of proteins relevant to these processes, such as focal adhesion kinase (FAK), merlin or integrin ß1. We were able to show that low glucose reduced the invasive potential of the cells, which was associated with a reduced amount of sialic acid. Under high glucose, barrier function was impaired and adhesion decreased, which correlated with a decreased expression of FAK.


Assuntos
Movimento Celular , Glucose , Neoplasias Meníngeas , Meningioma , Humanos , Meningioma/metabolismo , Meningioma/patologia , Movimento Celular/efeitos dos fármacos , Glucose/metabolismo , Glucose/farmacologia , Linhagem Celular Tumoral , Glicosilação , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/patologia , Invasividade Neoplásica , Aldeído Pirúvico/metabolismo , Aldeído Pirúvico/farmacologia , Adesão Celular/efeitos dos fármacos
9.
Molecules ; 29(18)2024 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-39339476

RESUMO

Poly(N-isopropylacrylamide) (PNIPAM) offers a promising platform for non-invasive and gentle cell detachment. However, conventional PNIPAM-based substrates often suffer from limitations including limited stability and reduced reusability, which hinder their widespread adoption in biomedical applications. In this study, PNIPAM copolymer films were formed on the surfaces of glass slides or silicon wafers using a two-step film-forming method involving coating and grafting. Subsequently, a comprehensive analysis of the films' surface wettability, topography, and thickness was conducted using a variety of techniques, including contact angle analysis, atomic force microscopy (AFM), and ellipsometric measurements. Bone marrow mesenchymal stem cells (BMMSCs) were then seeded onto PNIPAM copolymer films prepared from different copolymer solution concentrations, ranging from 0.2 to 10 mg·mL-1, to select the optimal culture substrate that allowed for good cell growth at 37 °C and effective cell detachment through temperature reduction. Furthermore, the stability and reusability of the optimal copolymer films were assessed. Finally, AFM and X-ray photoelectron spectroscopy (XPS) were employed to examine the surface morphology and elemental composition of the copolymer films after two rounds of BMMSC adhesion and detachment. The findings revealed that the surface properties and overall characteristics of PNIPAM copolymer films varied significantly with the solution concentration. Based on the selection criteria, the copolymer films derived from 1 mg·mL-1 solution were identified as the optimal culture substrates for BMMSCs. After two rounds of cellular adhesion and detachment, some proteins remained on the film surfaces, acting as a foundation for subsequent cellular re-adhesion and growth, thereby implicitly corroborating the practicability and reusability of the copolymer films. This study not only introduces a stable and efficient platform for stem cell culture and harvesting but also represents a significant advance in the fabrication of smart materials tailored for biomedical applications.


Assuntos
Resinas Acrílicas , Adesão Celular , Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Resinas Acrílicas/química , Adesão Celular/efeitos dos fármacos , Técnicas de Cultura de Células/métodos , Propriedades de Superfície , Proliferação de Células/efeitos dos fármacos , Temperatura , Animais , Microscopia de Força Atômica , Células Cultivadas , Células da Medula Óssea/citologia
10.
Int J Biol Sci ; 20(11): 4114-4127, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39247831

RESUMO

Osteosarcoma is the most prevalent form of primary malignant bone tumor, primarily affecting children and adolescents. The nerve growth factors (NGF) referred to as neurotrophins have been associated with cancer-induced bone pain; however, the role of NGF in osteosarcoma has yet to be elucidated. In osteosarcoma samples from the Genomic Data Commons data portal, we detected higher levels of NGF and M2 macrophage markers, but not M1 macrophage markers. In cellular experiments, NGF-stimulated osteosarcoma conditional medium was shown to facilitate macrophage polarization from the M0 to the M2 phenotype. NGF also enhanced VCAM-1-dependent monocyte adhesion within the osteosarcoma microenvironment by down-regulating miR-513c-5p levels through the FAK and c-Src cascades. In in vivo xenograft models, the overexpression of NGF was shown to enhance tumor growth, while the oral administration of the TrK inhibitor larotrectinib markedly antagonized NGF-promoted M2 macrophage expression and tumor progression. These results suggest that larotrectinib could potentially be used as a therapeutic agent aimed at mitigating NGF-mediated osteosarcoma progression.


Assuntos
Monócitos , Fator de Crescimento Neural , Osteossarcoma , Microambiente Tumoral , Molécula 1 de Adesão de Célula Vascular , Osteossarcoma/metabolismo , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Humanos , Fator de Crescimento Neural/metabolismo , Animais , Microambiente Tumoral/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/metabolismo , Camundongos , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Macrófagos/metabolismo , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Camundongos Nus
11.
Int J Mol Sci ; 25(17)2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39273096

RESUMO

In recent years, with the advent of a super-aged society, lifelong dental care has gained increasing emphasis, and implant therapy for patients with an edentulous jaw has become a significant option. However, for implant therapy to be suitable for elderly patients with reduced regenerative and immunological capabilities, higher osteoconductive and antimicrobial properties are required on the implant surfaces. Silicon nitride, a non-oxide ceramic known for its excellent mechanical properties and biocompatibility, has demonstrated high potential for inducing hard tissue differentiation and exhibiting antibacterial properties. In this study, silicon nitride was deposited on pure titanium metal surfaces and evaluated for its biocompatibility and antibacterial properties. The findings indicate that silicon nitride improves the hydrophilicity of the material surface, enhancing the initial adhesion of rat bone marrow cells and promoting hard tissue differentiation. Additionally, the antibacterial properties were assessed using Staphylococcus aureus, revealing that the silicon nitride-coated surfaces exhibited significant antibacterial activity. Importantly, no cytotoxicity was observed, suggesting that silicon nitride-coated titanium could serve as a novel implant material.


Assuntos
Antibacterianos , Materiais Revestidos Biocompatíveis , Compostos de Silício , Staphylococcus aureus , Propriedades de Superfície , Titânio , Titânio/química , Titânio/farmacologia , Animais , Antibacterianos/farmacologia , Antibacterianos/química , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Ratos , Staphylococcus aureus/efeitos dos fármacos , Compostos de Silício/química , Compostos de Silício/farmacologia , Teste de Materiais , Adesão Celular/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos
12.
ACS Appl Mater Interfaces ; 16(36): 47178-47191, 2024 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-39222394

RESUMO

Guided bone regeneration (GBR) technology has been demonstrated to be an effective method for reconstructing bone defects. A membrane is used to cover the bone defect to stop soft tissue from growing into it. The biosurface design of the barrier membrane is key to the technology. In this work, an asymmetric functional gradient Janus membrane was designed to address the bidirectional environment of the bone and soft tissue during bone reconstruction. The Janus membrane was simply and efficiently prepared by the multilayer self-assembly technique, and it was divided into the polycaprolactone isolation layer (PCL layer, GBR-A) and the nanohydroxyapatite/polycaprolactone/polyethylene glycol osteogenic layer (HAn/PCL/PEG layer, GBR-B). The morphology, composition, roughness, hydrophilicity, biocompatibility, cell attachment, and osteogenic mineralization ability of the double surfaces of the Janus membrane were systematically evaluated. The GBR-A layer was smooth, dense, and hydrophobic, which could inhibit cell adhesion and resist soft tissue invasion. The GBR-B layer was rough, porous, hydrophilic, and bioactive, promoting cell adhesion, proliferation, matrix mineralization, and expression of alkaline phosphatase and RUNX2. In vitro and in vivo results showed that the membrane could bind tightly to bone, maintain long-term space stability, and significantly promote new bone formation. Moreover, the membrane could fix the bone filling material in the defect for a better healing effect. This work presents a straightforward and viable methodology for the fabrication of GBR membranes with Janus-based bioactive surfaces. This work may provide insights for the design of biomaterial surfaces and treatment of bone defects.


Assuntos
Regeneração Óssea , Osteogênese , Poliésteres , Regeneração Óssea/efeitos dos fármacos , Animais , Poliésteres/química , Poliésteres/farmacologia , Osteogênese/efeitos dos fármacos , Durapatita/química , Durapatita/farmacologia , Polietilenoglicóis/química , Membranas Artificiais , Adesão Celular/efeitos dos fármacos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Proliferação de Células/efeitos dos fármacos , Humanos , Regeneração Tecidual Guiada/métodos , Coelhos , Camundongos
13.
Biointerphases ; 19(5)2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39235276

RESUMO

In order to properly satisfy biomedical constraints for cardiovascular applications, additively manufactured NiTi scaffolds required further process and metallurgical engineering. Additively manufactured NiTi materials for cardiovascular use will have to undergo surface finishing in order to minimize negative surface interactions within the artery. In this study, we sought to understand biocompatibility from chemically etched additively manufactured NiTi scaffolds by laser powder bed fusion (LPBF). Although two distinct oxide films were created in the surface etching process (labeled CP-A and CP-B), no qualitative changes in microroughness were seen between the two conditions. CP-A possessed significantly less Ni at the surface (0.19 at. %) than the CP-B group (3.30 at. %), via x-ray photoelectron spectroscopy, alongside a concomitant shift in the O1 s peak presentation alluding to a greater formation of a Ni based oxide in the CP-B group. Our live dead staining revealed significant toxicity and reduced cellular attachment for the CP-B group, in addition to inducing more cell lysis (20.9 ± 5.1%), which was significantly increased when compared to CP-A (P < 0.01). Future practices of manufacturing NiTi scaffolds using LPBF should focus on producing surface films that are not only smooth, but free of cytotoxic Ni based oxides.


Assuntos
Materiais Biocompatíveis , Níquel , Propriedades de Superfície , Titânio , Titânio/química , Titânio/farmacologia , Níquel/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Humanos , Óxidos/química , Óxidos/farmacologia , Teste de Materiais , Sobrevivência Celular/efeitos dos fármacos , Espectroscopia Fotoeletrônica , Adesão Celular/efeitos dos fármacos , Alicerces Teciduais/química
14.
Biomater Adv ; 165: 214001, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39216317

RESUMO

Artificial periosteum is deemed a novel strategy for inducing endogenous bone regeneration, but ideal periosteum substitutes achieved by orchestrating a biomimetic microenvironment for bone regeneration remain a significant challenge. Here, we design and fabricate a hybridized nanofiber-based artificial periosteum with boosted osteoinduction properties. Via a "molecular cage" biomineralization strategy, nano-hydroxyapatite (nano-HAp) with a controllable size (∼22 nm) and excellent dispersion serves as unique nano-additives for water-soluble polyvinyl-alcohol (PVA)-based artificial periosteum. The PVA/HAp composite is electrospun into nanofibers to replicate the extracellular-matrix-inspired nanostructure for inducing cell adhesion, proliferation, and fate manipulation. A simple post-crosslinking treatment is subsequently applied to further booster its mechanical strength (6.6 MPa) and swelling stability. The optimized sample of C-PVA/HAp (10 wt% nano-HAp) artificial periosteum features excellent biocompatibility and remarkable in vitro mineralization. Cell experiments demonstrate that its effectively boasted cell modulation for enhanced osteogenesis without the aid of growth factors, showing a possible activation of the ERK/MAPK signaling pathway. This work provides an effective strategy for designing novel HAp nano-additives and expands the possibility of biomimetic fabrication for more advanced nanofiber-based artificial periosteum.


Assuntos
Durapatita , Nanofibras , Osteogênese , Periósteo , Álcool de Polivinil , Nanofibras/química , Álcool de Polivinil/química , Durapatita/química , Durapatita/farmacologia , Osteogênese/efeitos dos fármacos , Humanos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Regeneração Óssea/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Animais , Materiais Biocompatíveis/química , Adesão Celular/efeitos dos fármacos , Substitutos Ósseos/química
15.
ACS Appl Bio Mater ; 7(9): 6089-6100, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39183644

RESUMO

Extracellular matrix (ECM) is essential for tissue development, providing structural support and a microenvironment that is necessary for cells. As tissue engineering advances, there is a growing demand for ECM mimics. Polycaprolactone (PCL) is a commonly used synthetic polymer for ECM mimic materials. However, its biologically inactive surface limits its direct application in tissue engineering. Our study aimed to improve the biocompatibility of PCL by incorporating hemoglobin nanofibrils (HbFs) into PCL using an electrospinning technique. HbFs were formed from bovine hemoglobin (Hb) extracted from industrial byproducts and designed to offer PCL an improved cell adhesion property. The fabricated HbFs@PCL electrospun scaffold exhibits improved fibroblast adherence, proliferation, and deeper fibroblast infiltration into the scaffold compared with the pure PCL scaffold, indicating its potential to be an ECM mimic. This study represents the pioneering utilization of Hb-sourced nanofibrils in the electrospun PCL scaffolds for tissue engineering applications.


Assuntos
Materiais Biocompatíveis , Matriz Extracelular , Hemoglobinas , Teste de Materiais , Nanofibras , Poliésteres , Engenharia Tecidual , Hemoglobinas/química , Animais , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Bovinos , Nanofibras/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Poliésteres/química , Proliferação de Células/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Alicerces Teciduais/química , Tamanho da Partícula , Camundongos , Fibroblastos
16.
Bioconjug Chem ; 35(9): 1318-1323, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39213494

RESUMO

We present a novel approach to the formation of cell aggregates by employing click chemistry with water-soluble zwitterionic dibenzo cyclooctadiyne (WS-CODY) and azide-modified hyaluronic acid (HA-N3) as a linker to facilitate rapid and stable cell aggregation. By optimizing the concentrations of HA-N3 and WS-CODY, we achieved efficient cross-linking between azide-modified cell surfaces and HA-N3, generating cell aggregates within 10 min, and the resulting aggregates remained stable for up to 5 days, with cell viability maintained at approximately 80%. Systematic experiments revealed that a stoichiometric balance between HA-N3 and WS-CODY is important for effective cross-linking, highlighting the roles of both cell-surface azide modification and HA in the aggregate formation. We also investigated the genetic basis of altered cell behavior within these aggregates. Transcriptome analysis (RNA-seq) of aggregates postcultivation revealed a marked fluctuation of genes associated with 'cell migration' and 'cell adhesion', including notable changes in the expression of HYAL1, ICAM-1, CEACAM5 and RHOB. These findings suggest that HA-N3-mediated cell aggregation can induce intrinsic cellular responses that not only facilitate cell aggregate formation but also modulate cell-matrix interactions. We term this phenomenon 'chemo-resilience', The simplicity and efficacy of this click chemistry-based approach suggest it may have broad applicability for forming cell aggregates and modulating cell-matrix interactions in tissue engineering and regenerative medicine.


Assuntos
Azidas , Agregação Celular , Movimento Celular , Química Click , Reagentes de Ligações Cruzadas , Ácido Hialurônico , Ácido Hialurônico/química , Azidas/química , Humanos , Movimento Celular/efeitos dos fármacos , Reagentes de Ligações Cruzadas/química , Agregação Celular/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos
17.
J Mater Chem B ; 12(36): 8911-8918, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39145600

RESUMO

Polymethyl methacrylate (PMMA) based biomaterials have been widely utilized in clinics. However, currently, PMMA catalyzed by benzoyl peroxide (BPO) exhibits disquieting disadvantages including an exothermic polymerization reaction and a lack of bioactivity. Here, we first designed three industrial-scale synthesis methods for high-purity butoxydibutylborane (BODBB), achieving purity levels greater than 95% (maximum: 97.6%) and ensuring excellent fire safety. By utilizing BODBB as a catalyst, the highest polymerization temperature of PMMA bone cement (PMMA-BODBB) reached only 36.05 °C, ensuring that no thermal damage occurred after implantation. Compared to PMMA catalyzed by BPO and partially oxidized tributylborane (TBBO, catalyst of Super Bond C&B), PMMA-BODBB exhibited superior cell adhesion, proliferation, and osteogenesis, attributed to the reduced release of free radicals and toxic monomer, and moderate bioactive boron release. After injection into a 5 mm defect in the rat cranial bone, PMMA-BODBB demonstrated the highest level of osteointegration. This work not only presents an industrial-scale synthesis of high-purity BODBB, but also offers an innovative PMMA biomaterial system with intrinsic biocompatibility and osseointegration, paving the way for the next generation of PMMA-based biomaterials with broader applications.


Assuntos
Materiais Biocompatíveis , Cimentos Ósseos , Osseointegração , Polimerização , Polimetil Metacrilato , Polimetil Metacrilato/química , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/síntese química , Catálise , Ratos , Cimentos Ósseos/química , Cimentos Ósseos/síntese química , Osseointegração/efeitos dos fármacos , Boranos/química , Boranos/síntese química , Proliferação de Células/efeitos dos fármacos , Teste de Materiais , Ratos Sprague-Dawley , Camundongos , Adesão Celular/efeitos dos fármacos
18.
J Cancer Res Clin Oncol ; 150(8): 402, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39198302

RESUMO

PURPOSE: Uterine serous carcinoma (USC) is a highly aggressive and frequently recurring subtype of endometrial cancer with limited treatment options for advanced or recurrent stages. Sulindac, a classic non-steroidal anti-inflammatory drug, has demonstrated anti-tumor activity in several pre-clinical tumor models. This study aims to evaluate the effect of sulindac on cell proliferation and invasion in USC cells. METHODS: Human USC cell lines ARK-1 and SPEC2 were treated with different concentrations of sulindac. Cell proliferation was assessed using MTT and colony formation assays. ELISA assays measured cellular stress, cleaved caspase 3 activity, antioxidant ability, and adhesion. Cell cycle arrest was evaluated by Cellometer. The invasive capability was detected by wound healing assay. Western blotting was used to analyze the changes in protein expression induced by sulindac. RESULTS: Exposure to sulindac decreased cellular viability in a dose-dependent manner in ARK-1 and SPEC2 cells. Sulindac effectively inhibited cell cycle progression, increased cellular stress, caused apoptosis, and reduced cell adhesion and invasion in USC cells. Additionally, sulindac decreased the expression of COX-2 and blocked phosphorylation of NF-κB induced by TNF-α. CONCLUSION: Sulindac is a potential therapeutic agent for USC that deserves further exploration in pre-clinical studies and potentially future clinical trials.


Assuntos
Apoptose , Proliferação de Células , Cistadenocarcinoma Seroso , Sulindaco , Neoplasias Uterinas , Humanos , Feminino , Sulindaco/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Uterinas/tratamento farmacológico , Neoplasias Uterinas/patologia , Linhagem Celular Tumoral , Cistadenocarcinoma Seroso/tratamento farmacológico , Cistadenocarcinoma Seroso/patologia , Cistadenocarcinoma Seroso/metabolismo , Apoptose/efeitos dos fármacos , Invasividade Neoplásica , Adesão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos
19.
Int J Immunopathol Pharmacol ; 38: 3946320241276894, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39135409

RESUMO

Background: Pentagalloyl glucose (PGG) is a polyphenol with vasoprotective properties. Targeted delivery of PGG reversed aortic aneurysm growth in several rodent models associated with decreased number of macrophages and transforming growth factor-ß (TGF-ß) expression. Thus, we sought to determine cellular mechanisms by which PGG reduces macrophage-induced aortic pathogenicity and its relationship to TGF-ß. Methods: Using THP-1 cells, primary human aortic cells, and explanted rat aortas, we assessed the anti-inflammatory effect of PGG. Expression of pro/anti-inflammatory macrophage markers was analyzed. Adhesion of monocytes as well as oxidative stress status, viability, and TGF-ß expression after primary aortic cell exposure to macrophage-conditioned medium with and without PGG were assessed. The release of TGF-ß was also examined in elastase-treated cultured rat aortas. Results: PGG pre-treatment of human aortic cell monolayers reduced the adhesion of THP-1 monocytes. PGG enhanced the expression of anti-inflammatory markers in THP-1-derived macrophages, and increased mitochondrial reactive oxygen species as well as mitochondrial polarization. Conditioned medium from THP-1-derived macrophages induced reactive oxygen species, cell death, and TGF-ß release from human aortic cells, which was suppressed by PGG. In explanted rat aortas, PGG reduced elastase mediated TGF-ß release. Conclusions: Combining anti-inflammatory, cytotoxic, and oxidative effects, PGG has high cardiovascular therapeutic potential. We confirmed previous in vivo observations whereby PGG suppressed TGF-ß response associated with disease resolution.


Assuntos
Anti-Inflamatórios , Aorta , Taninos Hidrolisáveis , Macrófagos , Fator de Crescimento Transformador beta , Taninos Hidrolisáveis/farmacologia , Humanos , Animais , Fator de Crescimento Transformador beta/metabolismo , Células THP-1 , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Anti-Inflamatórios/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Masculino , Adesão Celular/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos
20.
J Appl Oral Sci ; 32: e20240034, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39140581

RESUMO

OBJECTIVE: Bisphosphonates are prescribed to treat excessive bone resorption in patients with osteoporosis. However, its use is associated with potential adverse effects such as medication-related osteonecrosis of the jaw, prompting the introduction of the drug holiday concept in patients prior to dentoalveolar surgery. Furthermore, bisphosphonate discontinuation has been studied in vivo, in humans, and in animal models. However, it is not known whether this approach could affect bone cells in vitro. Therefore, the objective of this study was to investigate the potential effects of bisphosphonate discontinuation on pre-osteoblast and osteoblast activities in vitro. METHODOLOGY: Pre-osteoblasts (MC3T3) and osteoblasts were treated with bisphosphonate (alendronate) at concentrations of 1, 5, and 10 µM. Alendronate was then withdrawn at different time points. The negative control consisted of untreated cells (0 µM), while the positive control consisted of cells incubated with alendronate throughout the experiment. Cell viability, cell adhesion, cell cytoskeleton, mineralization, and gene expressions were investigated. RESULTS: Pre-osteoblasts and osteoblasts showed a decrease in cell viability after treatment with 5-10 µM alendronate for 4 days or longer. Two days of alendronate discontinuation significantly increased cell viability compared with the positive control. However, these levels did not reach those of the negative control. Bone nodule formation was reduced by alendronate. Discontinuation of alendronate regained bone nodule formation. Longer periods of discontinuation were more effective in restoring nodule formation than shorter periods. Addition of alendronate resulted in an increase in the percentage of dead cells, which, in turn, decreased when alendronate was discontinued. Alendronate affected the cell cytoskeleton by disassembling actin stress fibers. Cell adhesion and cell morphological parameters were also affected by alendronate. Discontinuation of alendronate restored cell adhesion and these parameters. Overall, the highest improvement after alendronate discontinuation was seen at 10 µM. However, alendronate treatment and discontinuation did not affect osteoblast gene expression. CONCLUSION: Discontinuation of alendronate helps to reverse the negative effects of the drug on cell viability, cell adhesion, and mineralization by restoring the cell cytoskeleton. Our data suggest the benefits of drug holiday and/or intermittent strategies for alendronate administration at the cellular level.


Assuntos
Alendronato , Conservadores da Densidade Óssea , Calcificação Fisiológica , Adesão Celular , Sobrevivência Celular , Citoesqueleto , Osteoblastos , Osteoblastos/efeitos dos fármacos , Alendronato/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Conservadores da Densidade Óssea/farmacologia , Citoesqueleto/efeitos dos fármacos , Animais , Adesão Celular/efeitos dos fármacos , Fatores de Tempo , Calcificação Fisiológica/efeitos dos fármacos , Camundongos , Expressão Gênica/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real , Análise de Variância
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA