Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.208
Filtrar
1.
J Biochem Mol Toxicol ; 38(7): e23761, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38952040

RESUMO

Non-small cell cancer (NSCLC) is the most common cancer in the world, but its effective therapeutic methods are limited. Tilianin and sufentanil alleviate various human tumors. This research aimed to clarify the functions and mechanisms of Tilianin and sufentanil in NSCLC. The functions of Tilianin and sufentanil on NSCLC cell viability, apoptosis, mitochondrial dysfunction, and immunity in vitro were examined using Cell Counting Kit-8 assay, flow cytometry, reactive oxygen species level analysis, CD8+ T cell percentage analysis, Western blot, and enzyme-linked immunosorbent assay, respectively. The molecular mechanism regulated by Tilianin and sufentanil in NSCLC was assessed using Western blot, and immunofluorescence assays. Meanwhile, the roles of Tilianin and sufentanil in NSCLC tumor growth, apoptosis, and immunity in vivo were determined by establishing a tumor xenograft mouse model, immunohistochemistry, and Western blot assays. When sufentanil concentration was proximity 2 nM, the inhibition rate of NSCLC cell viability was 50%. The IC50 for A549 cells was 2.36 nM, and the IC50 for H1299 cells was 2.18 nM. The IC50 of Tilianin for A549 cells was 38.7 µM, and the IC50 of Tilianin for H1299 cells was 44.6 µM. Functionally, 0.5 nM sufentanil and 10 µM Tilianin reduced NSCLC cell (A549 and H1299) viability in a dose-dependent manner. Also, 0.5 nM sufentanil and 10 µM Tilianin enhanced NSCLC cell apoptosis, yet this impact was strengthened after a combination of Tilianin and Sufentanil. Furthermore, 0.5 nM sufentanil and 10 µM Tilianin repressed NSCLC cell mitochondrial dysfunction and immunity, and these impacts were enhanced after a combination of Tilianin and Sufentanil. Mechanistically, 0.5 nM sufentanil and 10 µM Tilianin repressed the NF-κB pathway in NSCLC cells, while this repression was strengthened after a combination of Tilianin and Sufentanil. In vivo experimental data further clarified that 1 µg/kg sufentanil and 10 mg/kg Tilianin reduced NSCLC growth, immunity, and NF-κB pathway-related protein levels, yet these trends were enhanced after a combination of Tilianin and Sufentanil. Tilianin strengthened the antitumor effect of sufentanil in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Sufentanil , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Humanos , Sufentanil/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Animais , Camundongos , Apoptose/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Células A549 , Camundongos Nus , Sinergismo Farmacológico , Linhagem Celular Tumoral , Camundongos Endogâmicos BALB C , Antineoplásicos/farmacologia , Sulfatos de Condroitina/farmacologia , Venenos de Anfíbios
2.
ACS Appl Mater Interfaces ; 16(28): 35936-35948, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-38958205

RESUMO

Tissue-engineered heart valve (TEHV) has emerged as a prospective alternative to conventional valve prostheses. The decellularized heart valve (DHV) represents a promising TEHV scaffold that preserves the natural three-dimensional structure and retains essential biological activity. However, the limited mechanical strength, fast degradation, poor hemocompatibility, and lack of endothelialization of DHV restrict its clinical use, which is necessary for ensuring its long-term durability. Herein, we used oxidized chondroitin sulfate (ChS), one of the main components of the extracellular matrix with various biological activities, to cross-link DHV to overcome the above problems. In addition, the ChS-adipic dihydrazide was used to react with residual aldehyde groups, thus preventing potential calcification. The results indicated notable enhancements in mechanical properties and resilience against elastase and collagenase degradation in vitro as well as the ability to withstand extended periods of storage without compromising the structural integrity of valve scaffolds. Additionally, the newly cross-linked valves exhibited favorable hemocompatibility in vitro and in vivo, thereby demonstrating exceptional biocompatibility. Furthermore, the scaffolds exhibited traits of gradual degradation and resistance to calcification through a rat subcutaneous implantation model. In the rat abdominal aorta implantation model, the scaffolds demonstrated favorable endothelialization, commendable patency, and a diminished pro-inflammatory response. As a result, the newly constructed DHV scaffold offers a compelling alternative to traditional valve prostheses, which potentially advances the field of TEHV.


Assuntos
Sulfatos de Condroitina , Animais , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Ratos , Próteses Valvulares Cardíacas , Engenharia Tecidual , Valvas Cardíacas/efeitos dos fármacos , Valvas Cardíacas/química , Ratos Sprague-Dawley , Alicerces Teciduais/química , Teste de Materiais , Humanos , Reagentes de Ligações Cruzadas/química , Masculino , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Suínos
3.
Sci Rep ; 14(1): 16396, 2024 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-39013921

RESUMO

Most of the conditions involving cartilaginous tissues are irreversible and involve degenerative processes. The aim of the present study was to fabricate a biocompatible fibrous and film scaffolds using electrospinning and casting techniques to induce chondrogenic differentiation for possible application in cartilaginous tissue regeneration. Polycaprolactone (PCL) electrospun nanofibrous scaffolds and PCL film were fabricated and incorporated with multi-walled carbon nanotubes (MWCNTs). Thereafter, coating of chondroitin sulfate (CS) on the fibrous and film structures was applied to promote chondrogenic differentiation of human dental pulp stem cells (hDPSCs). First, the morphology, hydrophilicity and mechanical properties of the scaffolds were characterized by scanning electron microscopy (SEM), spectroscopic characterization, water contact angle measurements and tensile strength testing. Subsequently, the effects of the fabricated scaffolds on stimulating the proliferation of human dental pulp stem cells (hDPSCs) and inducing their chondrogenic differentiation were evaluated via electron microscopy, flow cytometry and RT‒PCR. The results of the study demonstrated that the different forms of the fabricated PCL-MWCNTs scaffolds analyzed demonstrated biocompatibility. The nanofilm structures demonstrated a higher rate of cellular proliferation, while the nanofibrous architecture of the scaffolds supported the cellular attachment and differentiation capacity of hDPSCs and was further enhanced with CS addition. In conclusion, the results of the present investigation highlighted the significance of this combination of parameters on the viability, proliferation and chondrogenic differentiation capacity of hDPSCs seeded on PCL-MWCNT scaffolds. This approach may be applied when designing PCL-based scaffolds for future cell-based therapeutic approaches developed for chondrogenic diseases.


Assuntos
Diferenciação Celular , Condrogênese , Sulfatos de Condroitina , Polpa Dentária , Nanofibras , Nanotubos de Carbono , Poliésteres , Células-Tronco , Alicerces Teciduais , Humanos , Polpa Dentária/citologia , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Poliésteres/química , Poliésteres/farmacologia , Nanofibras/química , Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Alicerces Teciduais/química , Nanotubos de Carbono/química , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Engenharia Tecidual/métodos
4.
Int J Nanomedicine ; 19: 5125-5138, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38855730

RESUMO

Purpose: Breast cancer is a prevalent malignancy among women worldwide, and malignancy is closely linked to the tumor microenvironment (TME). Here, we prepared mixed nano-sized formulations composed of pH-sensitive liposomes (Ber/Ru486@CLPs) and small-sized nano-micelles (Dox@CLGs). These liposomes and nano-micelles were modified by chondroitin sulfate (CS) to selectively target breast cancer cells. Methods: Ber/Ru486@CLPs and Dox@CLGs were prepared by thin-film dispersion and ethanol injection, respectively. To mimic actual TME, the in vitro "condition medium of fibroblasts + MCF-7" cell model and in vivo "4T1/NIH-3T3" co-implantation mice model were established to evaluate the anti-tumor effect of drugs. Results: The physicochemical properties showed that Dox@CLGs and Ber/Ru486@CLPs were 28 nm and 100 nm in particle size, respectively. In vitro experiments showed that the mixed formulations significantly improved drug uptake and inhibited cell proliferation and migration. The in vivo anti-tumor studies further confirmed the enhanced anti-tumor capabilities of Dox@CLGs + Ber/Ru486@CLPs, including smaller tumor volumes, weak collagen deposition, and low expression levels of α-SMA and CD31 proteins, leading to a superior anti-tumor effect. Conclusion: In brief, this combination therapy based on Dox@CLGs and Ber/Ru486@CLPs could effectively inhibit tumor development, which provides a promising approach for the treatment of breast cancer.


Assuntos
Neoplasias da Mama , Proliferação de Células , Doxorrubicina , Lipossomos , Microambiente Tumoral , Microambiente Tumoral/efeitos dos fármacos , Animais , Feminino , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Humanos , Camundongos , Lipossomos/química , Células MCF-7 , Doxorrubicina/farmacologia , Doxorrubicina/química , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Proliferação de Células/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Tamanho da Partícula , Sistemas de Liberação de Fármacos por Nanopartículas/química , Sistemas de Liberação de Medicamentos/métodos , Movimento Celular/efeitos dos fármacos , Nanopartículas/química
5.
Int J Biol Macromol ; 271(Pt 2): 132675, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38845259

RESUMO

Novel hydrogel-based multifunctional systems prepared utilizing photocrosslinking and freeze-drying processes (PhotoCross/Freeze-dried) dedicated for bone tissue regeneration are presented. Fabricated materials, composed of methacrylated gelatin, chitosan, and chondroitin sulfate, possess interesting features including bioactivity, biocompatibility, as well as antibacterial activity. Importantly, their degradation and swellability might be easily tuned by playing with the biopolymeric content in the photocrosllinked systems. To broaden the potential application and deliver the therapeutic features, mesoporous silica particles functionalized with methacrylate moieties decorated with hydroxyapatite and loaded with the antiosteoporotic drug, alendronate, (MSP-MA-HAp-ALN) were dispersed within the biopolymeric sol and photocrosslinked. It was demonstrated that the obtained composites are characterized by a significantly extended degradation time, ensuring optimal conditions for balancing hybrids removal with the deposition of fresh bone. We have shown that attachment of MSP-MA-HAp-ALN to the polymeric matrix minimizes the initial burst effect and provides a prolonged release of ALN (up to 22 days). Moreover, the biological evaluation in vitro suggested the capability of the resulted systems to promote bone remodeling. Developed materials might potentially serve as scaffolds that after implantation will fill up bone defects of various origin (osteoporosis, tumour resection, accidents) providing the favourable conditions for bone regeneration and supporting the infections' treatment.


Assuntos
Regeneração Óssea , Quitosana , Sulfatos de Condroitina , Gelatina , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Quitosana/química , Gelatina/química , Regeneração Óssea/efeitos dos fármacos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Alicerces Teciduais/química , Humanos , Reagentes de Ligações Cruzadas/química , Animais , Osso e Ossos/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/química , Hidrogéis/química , Hidrogéis/farmacologia
6.
J Food Sci ; 89(7): 4469-4479, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38837700

RESUMO

This study aimed to evaluate the anti-cervical cancer activity of chondroitin sulfate-functionalized selenium nanoparticles (SeCS) and to elucidate their action mechanism. Cytotoxic effect of SeCS on HeLa cells was assessed by MTT assay. Further molecular mechanism of SeCS was analyzed by flow cytometric assay and western blotting. The results showed that treatment with SeCS resulted in a dose- and time-dependent inhibition in the proliferation of HeLa cells. The data obtained from flow cytometry demonstrated that SeCS inhibited HeLa cell growth via the induction of S-phase arrest and cell apoptosis. Further mechanism analysis found that SeCS down-regulated expression levels of cyclin A and CDK2 and up-regulated p21 expression, which contributed to S arrest. Moreover, SeCS increased the level of Bax and decreased the expression of Bcl-2, resulting in the release of cytochrome C from mitochondria and activating caspase-3/8/9 for caspase-dependent apoptosis. Meanwhile, intracellular reactive oxygen species (ROS) levels were elevated after SeCS treatment, suggesting that ROS might be upstream of SeCS-induced S-phase arrest and cell apoptosis. These data show that SeCS has anti-tumor effects and possesses the potential to become a new therapeutic agent or adjuvant therapy for cancer patients. PRACTICAL APPLICATION: In our previous study, we used chondroitin sulfate to stabilize nano-selenium to obtain SeCS to improve the bioactivity and stability of nano-selenium. We found that it possessed an inhibitory effect on HeLa cells. However, the molecular mechanism remains unclear. This study elucidated the mechanism of SeCS damage to HeLa cells. SeCS has the potential to become a new therapeutic agent or adjuvant therapy for cancer patients.


Assuntos
Apoptose , Sulfatos de Condroitina , Nanopartículas , Espécies Reativas de Oxigênio , Selênio , Humanos , Células HeLa , Sulfatos de Condroitina/farmacologia , Sulfatos de Condroitina/química , Apoptose/efeitos dos fármacos , Selênio/farmacologia , Selênio/química , Nanopartículas/química , Espécies Reativas de Oxigênio/metabolismo , Proliferação de Células/efeitos dos fármacos , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Antineoplásicos/farmacologia
7.
Biomaterials ; 311: 122661, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38875883

RESUMO

Abdominal adhesion, a serious complication of abdominal surgery, often resists mitigation by current drug administration and physical barriers. To address this issue, we developed an injectable, antifouling hydrogel through the free-radical polymerization of methacrylate chondroitin sulfate (CS-GMA) and 2-methacryloyloxyethyl phosphorylcholine (MPC) monomers, dubbed the CGM hydrogel. We systematically analyzed its physicochemical properties, including rheological strength, biocompatibility, and antifouling capabilities. A rat abdominal cecum adhesion model was constructed to assess the effectiveness of CGM hydrogel in preventing postoperative adhesion and recurrent adhesion. In addition, multi-omics analyses identified the relationship between adhesion development and CCL2/CCR2 interaction. Notably, CGM hydrogel can thwart the recruitment and aggregation of fibroblasts and macrophages by inhibiting the CCL2/CCR2 interaction. Moreover, CGM hydrogel significantly dampens the activity of fibrosis-linked cytokines (TGF-ßR1) and recalibrates extracellular matrix deposition-related cytokines (t-PA and PAI-1, Col Ⅰ and MMP-9). Cumulatively, the dual action of CGM hydrogel-as a physical barrier and cytokine regulator-highlights its promising potential in clinical application for abdominal adhesion prevention.


Assuntos
Quimiocina CCL2 , Hidrogéis , Ratos Sprague-Dawley , Receptores CCR2 , Animais , Aderências Teciduais/prevenção & controle , Aderências Teciduais/metabolismo , Hidrogéis/química , Hidrogéis/farmacologia , Quimiocina CCL2/metabolismo , Ratos , Receptores CCR2/metabolismo , Fosforilcolina/análogos & derivados , Fosforilcolina/química , Fosforilcolina/farmacologia , Metacrilatos/química , Metacrilatos/farmacologia , Incrustação Biológica/prevenção & controle , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Camundongos , Abdome/cirurgia , Injeções , Masculino , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos
8.
Int J Biol Macromol ; 274(Pt 2): 133435, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38936580

RESUMO

Polyether-ether-ketone (PEEK) is clinically used as a bio-implant for the healing of skeletal defects. However, the osseointegration of clinical-sized bone grafts remains limited. In this study, surface-porous PEEK was created by using a sulfonation method and a metal-polysaccharide complex MgCS was introduced on the surface of sulfonated PEEK to form MgCS@SPEEK. The as-prepared MgCS@SPEEK was found to have a porous surface with good hydrophilicity and bioactivity. This was followed by an investigation into whether MgCS loaded onto sulfonated PEEK surfaces could promote osseointegration and angiogenesis. The in vitro results showed that MgCS@SPEEK had a positive effect on reducing the expression levels of inflammatory genes and promoting osteogenesis and angiogenesis-related genes expression levels. Furthermore, porous MgCS@SPEEK was implanted in critical-sized rat tibial defects for in vivo evaluation of osseointegration. The micro-computed tomography evaluation results revealed substantial bone formation at 4 and 8 weeks. Collectively, these findings indicate that MgCS@SPEEK could provide improved osseointegration and an attractive strategy for orthopaedic applications.


Assuntos
Benzofenonas , Sulfatos de Condroitina , Cetonas , Osseointegração , Osteogênese , Polietilenoglicóis , Polímeros , Animais , Polímeros/química , Ratos , Cetonas/química , Cetonas/farmacologia , Polietilenoglicóis/química , Osseointegração/efeitos dos fármacos , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Osteogênese/efeitos dos fármacos , Magnésio/farmacologia , Porosidade , Próteses e Implantes , Propriedades de Superfície , Ratos Sprague-Dawley , Masculino , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia
9.
Biomacromolecules ; 25(6): 3312-3324, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38728671

RESUMO

3D-printed hydrogel scaffolds biomimicking the extracellular matrix (ECM) are key in cartilage tissue engineering as they can enhance the chondrogenic differentiation of mesenchymal stem cells (MSCs) through the presence of active nanoparticles such as graphene oxide (GO). Here, biomimetic hydrogels were developed by cross-linking alginate, gelatin, and chondroitin sulfate biopolymers in the presence of GO as a bioactive filler, with excellent processability for developing bioactive 3D printed scaffolds and for the bioprinting process. A novel bioink based on our hydrogel with embedded human MSCs presented a cell survival rate near 100% after the 3D bioprinting process. The effects of processing and filler concentration on cell differentiation were further quantitatively evaluated. The nanocomposited hydrogels render high MSC proliferation and viability, exhibiting intrinsic chondroinductive capacity without any exogenous factor when used to print scaffolds or bioprint constructs. The bioactivity depended on the GO concentration, with the best performance at 0.1 mg mL-1. These results were explained by the rational combination of the three biopolymers, with GO nanoparticles having carboxylate and sulfate groups in their structures, therefore, biomimicking the highly negatively charged ECM of cartilage. The bioactivity of this biomaterial and its good processability for 3D printing scaffolds and 3D bioprinting techniques open up a new approach to developing novel biomimetic materials for cartilage repair.


Assuntos
Alginatos , Bioimpressão , Diferenciação Celular , Condrogênese , Sulfatos de Condroitina , Gelatina , Hidrogéis , Células-Tronco Mesenquimais , Nanocompostos , Impressão Tridimensional , Alicerces Teciduais , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Alginatos/química , Alginatos/farmacologia , Gelatina/química , Bioimpressão/métodos , Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Nanocompostos/química , Alicerces Teciduais/química , Hidrogéis/química , Hidrogéis/farmacologia , Engenharia Tecidual/métodos , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Grafite/química , Grafite/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas
10.
J Neural Eng ; 21(3)2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38806019

RESUMO

Objective.Severe traumatic brain injury (sTBI) induced neuronal loss and brain atrophy contribute significantly to long-term disabilities. Brain extracellular matrix (ECM) associated chondroitin sulfate (CS) glycosaminoglycans promote neural stem cell (NSC) maintenance, and CS hydrogel implants have demonstrated the ability to enhance neuroprotection, in preclinical sTBI studies. However, the ability of neuritogenic chimeric peptide (CP) functionalized CS hydrogels in promoting functional recovery, after controlled cortical impact (CCI) and suction ablation (SA) induced sTBI, has not been previously demonstrated. We hypothesized that neuritogenic (CS)CP hydrogels will promote neuritogenesis of human NSCs, and accelerate brain tissue repair and functional recovery in sTBI rats.Approach.We synthesized chondroitin 4-Osulfate (CS-A)CP, and 4,6-O-sulfate (CS-E)CP hydrogels, using strain promoted azide-alkyne cycloaddition (SPAAC), to promote cell adhesion and neuritogenesis of human NSCs,in vitro; and assessed the ability of (CS-A)CP hydrogels in promoting tissue and functional repair, in a novel CCI-SA sTBI model,in vivo. Main results.Results indicated that (CS-E)CP hydrogels significantly enhanced human NSC aggregation and migration via focal adhesion kinase complexes, when compared to NSCs in (CS-A)CP hydrogels,in vitro. In contrast, NSCs encapsulated in (CS-A)CP hydrogels differentiated into neurons bearing longer neurites and showed greater spontaneous activity, when compared to those in (CS-E)CP hydrogels. The intracavitary implantation of (CS-A)CP hydrogels, acutely after CCI-SA-sTBI, prevented neuronal and axonal loss, as determined by immunohistochemical analyses. (CS-A)CP hydrogel implanted animals also demonstrated the significantly accelerated recovery of 'reach-to-grasp' function when compared to sTBI controls, over a period of 5-weeks.Significance.These findings demonstrate the neuritogenic and neuroprotective attributes of (CS)CP 'click' hydrogels, and open new avenues for the development of multifunctional glycomaterials that are functionalized with biorthogonal handles for sTBI repair.


Assuntos
Lesões Encefálicas Traumáticas , Hidrogéis , Células-Tronco Neurais , Neuritos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Hidrogéis/administração & dosagem , Animais , Ratos , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Humanos , Células-Tronco Neurais/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Masculino , Sulfatos de Condroitina/administração & dosagem , Sulfatos de Condroitina/farmacologia , Glicosaminoglicanos/administração & dosagem , Células Cultivadas , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia
11.
Int J Biol Macromol ; 271(Pt 1): 132520, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38772463

RESUMO

Blocking the tumor nutrient supply through angiogenic inhibitors is an effective treatment approach for malignant tumors. However, using angiogenic inhibitors alone may not be enough to achieve a significant tumor response. Therefore, we recently designed a universal drug delivery system combining chemotherapy and anti-angiogenic therapy to target tumor cells while minimizing drug-related side effects. This system (termed as PCCE) is composed of biomaterial chondroitin sulfate (CS), the anti-angiogenic peptide ES2, and paclitaxel (PTX), which collectively enhance antitumor properties. Interestingly, the PCCE system is conferred exceptional cell membrane permeability due to inherent characteristics of CS, including CD44 receptor-mediated endocytosis. The PCCE could respond to the acidic and high glutathione conditions, thereby releasing PTX and ES2. PCCE could effectively inhibit the proliferation, migration, and invasion of tumor cells and cause apoptosis, while PCCE can affect the endothelial cells tube formation and exert anti-angiogenic function. Consistently, more potent in vivo antitumor efficacy and non-toxic sides were demonstrated in B16F10 xenograft mouse models. PCCE can achieve excellent antitumor activity via modulating angiogenic and apoptosis-related factors. In summary, we have successfully developed an intelligent and responsive CS-based nanocarrier known as PCCE for delivering various antitumor drugs, offering a promising strategy for treating malignant tumors.


Assuntos
Inibidores da Angiogênese , Sulfatos de Condroitina , Nanopartículas , Paclitaxel , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Paclitaxel/farmacologia , Paclitaxel/administração & dosagem , Paclitaxel/química , Paclitaxel/uso terapêutico , Animais , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/química , Inibidores da Angiogênese/uso terapêutico , Inibidores da Angiogênese/administração & dosagem , Humanos , Camundongos , Nanopartículas/química , Linhagem Celular Tumoral , Apoptose/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Proliferação de Células/efeitos dos fármacos , Portadores de Fármacos/química , Movimento Celular/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/administração & dosagem
12.
Carbohydr Res ; 541: 109163, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38805806

RESUMO

In this study, glycosaminoglycans (GAGs) were extracted from corb (Sciaena umbra) heads and thoroughly examined for their structure. Through cellulose acetate electrophoresis, the GAGs were identified as chondroitin sulfate (CS), with a recovery yield of 10.35 %. The CS exhibited notable characteristics including a high sulfate content (12.4 %) and an average molecular weight of 38.32 kDa. Further analysis via 1H NMR spectroscopy and SAX-HPLC revealed that the CS primarily consisted of alternating units predominantly composed of monosulfated disaccharides at positions 6 and 4 of GalNAc (52.6 % and 38.8 %, respectively). The ratio of sulfate groups between positions 4 and 6 of GalNAc (4/6 ratio) was approximately 0.74, resulting in an overall charge density of 0.98. Thermal properties of the CS were assessed using techniques such as differential scanning calorimetry and thermogravimetric analysis. Notably, the CS demonstrated concentration-dependent prolongation of activated partial thromboplastin time (aPTT) and thrombin time (TT) while showing no effect on platelet function. At 200 µg/mL, aPTT and TT coagulation times were 1.4 and 3.7 times faster than the control, respectively. These findings suggest that CS derived from corb heads holds promise as an anticoagulant agent for therapy, although further clinical investigations are necessary to validate its efficacy.


Assuntos
Anticoagulantes , Sulfatos de Condroitina , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Sulfatos de Condroitina/isolamento & purificação , Anticoagulantes/química , Anticoagulantes/farmacologia , Anticoagulantes/isolamento & purificação , Animais , Humanos , Coagulação Sanguínea/efeitos dos fármacos
13.
Int J Biol Macromol ; 269(Pt 2): 131952, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38692541

RESUMO

Thromboembolic diseases pose a serious risk to human health worldwide. Fucosylated chondroitin sulfate (FCS) is reported to have good anticoagulant activity with a low bleeding risk. Molecular weight plays a significant role in the anticoagulant activity of FCS, and FCS smaller than octasaccharide in size has no anticoagulant activity. Therefore, identifying the best candidate for developing novel anticoagulant FCS drugs is crucial. Herein, native FCS was isolated from sea cucumber Cucumaria frondosa (FCScf) and depolymerized into a series of lower molecular weights (FCScfs). A comprehensive assessment of the in vitro anticoagulant activity and in vivo bleeding risk of FCScfs with different molecule weights demonstrated that 10 kDa FCScf (FCScf-10 K) had a greater intrinsic anticoagulant activity than low molecular weight heparin (LMWH) without any bleeding risk. Using molecular modeling combined with experimental validation, we revealed that FCScf-10 K can specifically inhibit the formation of the Xase complex by binding the negatively charged sulfate group of FCScf-10 K to the positively charged side chain of arginine residues on the specific surface of factor IXa. Thus, these data demonstrate that the intermediate molecular weight FCScf-10 K is a promising candidate for the development of novel anticoagulant drugs.


Assuntos
Anticoagulantes , Sulfatos de Condroitina , Fator IXa , Peso Molecular , Animais , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Sulfatos de Condroitina/isolamento & purificação , Anticoagulantes/farmacologia , Anticoagulantes/química , Anticoagulantes/isolamento & purificação , Fator IXa/metabolismo , Fator IXa/antagonistas & inibidores , Fator IXa/química , Cucumaria/química , Pepinos-do-Mar/química , Coagulação Sanguínea/efeitos dos fármacos , Humanos , Modelos Moleculares
14.
J Mater Chem B ; 12(22): 5535-5550, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38747002

RESUMO

Invasive neural implants allow for high-resolution bidirectional communication with the nervous tissue and have demonstrated the ability to record neural activity, stimulate neurons, and sense neurochemical species with high spatial selectivity and resolution. However, upon implantation, they are exposed to a foreign body response which can disrupt the seamless integration of the device with the native tissue and lead to deterioration in device functionality for chronic implantation. Modifying the device surface by incorporating bioactive coatings has been a promising approach to camouflage the device and improve integration while maintaining device performance. In this work, we explored the novel application of a chondroitin sulfate (CS) based hydrophilic coating, with anti-fouling and neurite-growth promoting properties for neural recording electrodes. CS-coated samples exhibited significantly reduced protein-fouling in vitro which was maintained for up to 4-weeks. Cell culture studies revealed a significant increase in neurite attachment and outgrowth and a significant decrease in microglia attachment and activation for the CS group as compared to the control. After 1-week of in vivo implantation in the mouse cortex, the coated probes demonstrated significantly lower biofouling as compared to uncoated controls. Like the in vitro results, increased neuronal population (neuronal nuclei and neurofilament) and decreased microglial activation were observed. To assess the coating's effect on the recording performance of silicon microelectrodes, we implanted coated and uncoated electrodes in the mouse striatum for 1 week and performed impedance and recording measurements. We observed significantly lower impedance in the coated group, likely due to the increased wettability of the coated surface. The peak-to-peak amplitude and the noise floor levels were both lower in the CS group compared to the controls, which led to a comparable signal-to-noise ratio between the two groups. The overall single unit yield (% channels recording a single unit) was 74% for the CS and 67% for the control group on day 1. Taken together, this study demonstrates the effectiveness of the polysaccharide-based coating in reducing biofouling and improving biocompatibility for neural electrode devices.


Assuntos
Sulfatos de Condroitina , Materiais Revestidos Biocompatíveis , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Animais , Camundongos , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Propriedades de Superfície , Neurônios/efeitos dos fármacos , Incrustação Biológica/prevenção & controle , Eletrodos Implantados
15.
Mar Drugs ; 22(4)2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38667801

RESUMO

Fucosylated chondroitin sulfate is a unique glycosaminoglycan isolated from sea cucumbers, with excellent anticoagulant activity. The fucosyl branch in FCS is generally located at the 3-OH of D-glucuronic acid but, recently, a novel structure with α-L-fucose linked to the 6-OH of N-acetyl-galactosamine has been found. Here, using functionalized monosaccharide building blocks, we prepared novel FCS tetrasaccharides with fucosyl branches both at the 6-OH of GalNAc and 3-OH of GlcA. In the synthesis, the protective group strategy of selective O-sulfation, as well as stereoselective glycosylation, was established, which enabled the efficient synthesis of the specific tetrasaccharide compounds. This research enriches knowledge on the structural types of FCS oligosaccharides and facilitates the exploration of the structure-activity relationship in the future.


Assuntos
Sulfatos de Condroitina , Oligossacarídeos , Pepinos-do-Mar , Sulfatos de Condroitina/química , Sulfatos de Condroitina/síntese química , Sulfatos de Condroitina/farmacologia , Animais , Oligossacarídeos/síntese química , Oligossacarídeos/química , Pepinos-do-Mar/química , Glicosilação , Fucose/química , Anticoagulantes/farmacologia , Anticoagulantes/química , Anticoagulantes/síntese química , Relação Estrutura-Atividade , Acetilgalactosamina/química , Acetilgalactosamina/análogos & derivados
16.
Int J Biol Macromol ; 266(Pt 2): 131425, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38583830

RESUMO

Nano-MoS2 exhibit oxidoreductase-like activities, and has been shown to effectively eliminate excessive intracellular ROS and inhibit Aß aggregation, thus demonstrating promising potential for anti-Alzheimer's disease (anti-AD) intervention. However, the low water dispersibility and high toxicity of nano-MoS2 limits its further application. In this study, we developed a chondroitin sulphate (CS)-modified MoS2 nanoenzyme (CS@MoS2) by harnessing the excellent biocompatibility of CS and the exceptional activities of nano-MoS2 to explore its potential in anti-AD research. Promisingly, CS@MoS2 significantly inhibited Aß1-40 aggregation and prevented toxic injury in SH-SY5Y cells caused by Aß1-40. In addition, CS@MoS2 protected these cells from oxidative stress damage by regulating ROS production, as well as promoting the activities of SOD and GSH-Px. CS@MoS2 also modulated the intracellular Ca2+ imbalance and downregulated Tau hyperphosphorylation by activating GSK-3ß. CS@MoS2 suppressed p-NF-κB (p65) translocation to the nucleus by inhibiting MAPK phosphorylation, and modulated the expression of downstream anti- and proinflammatory cytokines. Owing to its multifunctional activities, CS@MoS2 effectively improved spatial learning, memory, and anxiety in D-gal/AlCl3-induced AD mice. Taken together, these results indicate that CS@MoS2 has significant potential for improving the therapeutic efficacy of the prevention and treatment of AD, while also presenting a novel framework for the application of nanoenzymes.


Assuntos
Doença de Alzheimer , Sulfatos de Condroitina , Dissulfetos , Molibdênio , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Animais , Camundongos , Humanos , Molibdênio/química , Molibdênio/farmacologia , Dissulfetos/química , Dissulfetos/farmacologia , Peptídeos beta-Amiloides/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Linhagem Celular Tumoral , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/química , Masculino , Modelos Animais de Doenças
17.
ACS Biomater Sci Eng ; 10(5): 3242-3254, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38632852

RESUMO

Osteoarthritis is characterized by enzymatic breakdown of the articular cartilage via the disruption of chondrocyte homeostasis, ultimately resulting in the destruction of the articular surface. Decades of research have highlighted the importance of inflammation in osteoarthritis progression, with inflammatory cytokines shifting resident chondrocytes into a pro-catabolic state. Inflammation can result in poor outcomes for cells implanted for cartilage regeneration. Therefore, a method to promote the growth of new cartilage and protect the implanted cells from the pro-inflammatory cytokines found in the joint space is required. In this study, we fabricate two gel types: polymer network hydrogels composed of chondroitin sulfate and hyaluronic acid, glycosaminoglycans (GAGs) known for their anti-inflammatory and prochondrogenic activity, and interpenetrating networks of GAGs and collagen I. Compared to a collagen-only hydrogel, which does not provide an anti-inflammatory stimulus, chondrocytes in GAG hydrogels result in reduced production of pro-inflammatory cytokines and enzymes as well as preservation of collagen II and aggrecan expression. Overall, GAG-based hydrogels have the potential to promote cartilage regeneration under pro-inflammatory conditions. Further, the data have implications for the use of GAGs to generally support tissue engineering in pro-inflammatory environments.


Assuntos
Condrócitos , Sulfatos de Condroitina , Ácido Hialurônico , Hidrogéis , Inflamação , Hidrogéis/química , Hidrogéis/farmacologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Sulfatos de Condroitina/farmacologia , Sulfatos de Condroitina/química , Inflamação/metabolismo , Inflamação/tratamento farmacológico , Inflamação/patologia , Animais , Cartilagem Articular/metabolismo , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Citocinas/metabolismo , Agrecanas/metabolismo , Engenharia Tecidual/métodos , Osteoartrite/patologia , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo
18.
Macromol Biosci ; 24(8): e2400051, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38663437

RESUMO

Bioactive scaffolds capable of simultaneously repairing osteochondral defects remain a big challenge due to the heterogeneity of bone and cartilage. Currently modular microgel-based bioassembly scaffolds are emerged as potential solution to this challenge. Here, microgels based on methacrylic anhydride (MA) and dopamine modified gelatin (GelMA-DA) are loaded with chondroitin sulfate (CS) (the obtained microgel named GC Ms) or bioactive glass (BG) (the obtained microgel named GB Ms), respectively. GC Ms and GB Ms show good biocompatibility with BMSCs, which suggested by the adhesion and proliferation of BMSCs on their surfaces. Specially, GC Ms promote chondrogenic differentiation of BMSCs, while GB Ms promote osteogenic differentiation. Furthermore, the injectable GC Ms and GB Ms are assembled integrally by bottom-up in situ cross-linking to obtain modular microgel-based bioassembly scaffold (GC-GB/HM), which show a distinct bilayer structure and good porous properties and swelling properties. Particularly, the results of in vivo and in vitro experiments show that GC-GB/HM can simultaneously regulate the expression levels of chondrogenic- and osteogenesis-related genes and proteins. Therefore, modular microgel-based assembly scaffold in this work with the ability to promote bidirectional differentiation of BMSCs and has great potential for application in the minimally invasive treatment of osteochondral tissue defects.


Assuntos
Diferenciação Celular , Condrogênese , Células-Tronco Mesenquimais , Microgéis , Osteogênese , Alicerces Teciduais , Osteogênese/efeitos dos fármacos , Alicerces Teciduais/química , Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Animais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Microgéis/química , Gelatina/química , Gelatina/farmacologia , Coelhos , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Proliferação de Células/efeitos dos fármacos , Engenharia Tecidual/métodos
19.
Int J Biol Macromol ; 266(Pt 2): 131051, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38556223

RESUMO

In situ-forming hydrogels that possess the ability to be injected in a less invasive manner and mimic the biochemical composition and microarchitecture of the native cartilage extracellular matrix are desired for cartilage tissue engineering. Besides, gelation time and stiffness of the hydrogel are two interdependent factors that affect cells' distribution and fate and hence need to be optimized. This study presented a bioinspired in situ-forming hydrogel composite of hyaluronic acid (HA), chondroitin sulfate (CS), and collagen short nanofiber (CSNF). HA and CS were functionalized with aldehyde and amine groups to form a gel through a Schiff-base reaction. CSNF was fabricated via electrospinning, followed by fragmentation by ultrasonics. Gelation time (11-360 s) and compressive modulus (1.4-16.2 kPa) were obtained by varying the concentrations of CS, HA, CSNFs, and CSNFs length. The biodegradability and biocompatibility of the hydrogels with varying gelation and stiffness were also assessed in vitro and in vivo. At three weeks, the assessment of hydrogels' chondrogenic differentiation also yields varying levels of chondrogenic differentiation. The subcutaneous implantation of the hydrogels in a mouse model indicated no severe inflammation. Results demonstrated that the injectable CS/HA@CSNF hydrogel was a promising hydrogel for tissue engineering and cartilage regeneration.


Assuntos
Sulfatos de Condroitina , Colágeno , Ácido Hialurônico , Hidrogéis , Nanocompostos , Nanofibras , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Nanofibras/química , Animais , Hidrogéis/química , Hidrogéis/farmacologia , Camundongos , Colágeno/química , Nanocompostos/química , Engenharia Tecidual/métodos , Cartilagem/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Alicerces Teciduais/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia
20.
Adv Healthc Mater ; 13(19): e2400207, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38529833

RESUMO

Magnesium phosphate bone cement (MPC) has gained widespread usage in orthopedic implantation due to its fast-setting and high initial strength benefits. However, the simultaneous attainment of drug-controlled release and osteogenic potential in MPC remains a significant challenge. Herein, a strategy to create a smart injectable cement system using nanocontainers and chondroitin sulfate is proposed. It employs nanocontainers containing alendronate-loaded mesoporous silica nanoparticles, which are surface-modified with polypyrrole to control drug release in response to near-infrared (NIR) stimulation. The alendronate-incorporated cement (ACMPC) exhibits improved compressive strength (70.6 ± 5.9 MPa), prolonged setting time (913 s), and exceptional injectability (96.5% of injection rate and 242 s of injection time). It also shows the capability to prevent degradation, thus preserving mechanical properties. Under NIR irradiation, the cement shows good antibacterial properties due to the combined impact of hyperthermia, reactive oxygen species, and alendronate. Furthermore, the ACMPC (NIR) group displays good biocompatibility and osteogenesis capabilities, which also lead to an increase in alkaline phosphatase activity, extracellular matrix mineralization, and the upregulation of osteogenic genes. This research has significant implications for developing multifunctional biomaterials and clinical applications.


Assuntos
Alendronato , Cimentos Ósseos , Osteogênese , Dióxido de Silício , Cimentos Ósseos/química , Cimentos Ósseos/farmacologia , Osteogênese/efeitos dos fármacos , Alendronato/química , Alendronato/farmacologia , Dióxido de Silício/química , Animais , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacologia , Preparações de Ação Retardada/farmacocinética , Nanopartículas/química , Antibacterianos/química , Antibacterianos/farmacologia , Antibacterianos/administração & dosagem , Polímeros/química , Magnésio/química , Magnésio/farmacologia , Força Compressiva , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Raios Infravermelhos , Camundongos , Humanos , Liberação Controlada de Fármacos , Fosfatos , Pirróis , Compostos de Magnésio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...