Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.207
Filter
1.
ACS Appl Mater Interfaces ; 16(28): 35936-35948, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38958205

ABSTRACT

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.


Subject(s)
Chondroitin Sulfates , Animals , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Rats , Heart Valve Prosthesis , Tissue Engineering , Heart Valves/drug effects , Heart Valves/chemistry , Rats, Sprague-Dawley , Tissue Scaffolds/chemistry , Materials Testing , Humans , Cross-Linking Reagents/chemistry , Male , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Swine
2.
J Biochem Mol Toxicol ; 38(7): e23761, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38952040

ABSTRACT

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.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Sufentanil , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Humans , Sufentanil/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , Mice , Apoptosis/drug effects , Xenograft Model Antitumor Assays , A549 Cells , Mice, Nude , Drug Synergism , Cell Line, Tumor , Mice, Inbred BALB C , Antineoplastic Agents/pharmacology , Chondroitin Sulfates/pharmacology , Amphibian Venoms
3.
Sci Rep ; 14(1): 16396, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39013921

ABSTRACT

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.


Subject(s)
Cell Differentiation , Chondrogenesis , Chondroitin Sulfates , Dental Pulp , Nanofibers , Nanotubes, Carbon , Polyesters , Stem Cells , Tissue Scaffolds , Humans , Dental Pulp/cytology , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Polyesters/chemistry , Polyesters/pharmacology , Nanofibers/chemistry , Cell Differentiation/drug effects , Chondrogenesis/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Tissue Scaffolds/chemistry , Nanotubes, Carbon/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Tissue Engineering/methods
4.
Int J Biol Macromol ; 271(Pt 2): 132675, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38845259

ABSTRACT

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.


Subject(s)
Bone Regeneration , Chitosan , Chondroitin Sulfates , Gelatin , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Chitosan/chemistry , Gelatin/chemistry , Bone Regeneration/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Scaffolds/chemistry , Humans , Cross-Linking Reagents/chemistry , Animals , Bone and Bones/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology
5.
J Food Sci ; 89(7): 4469-4479, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38837700

ABSTRACT

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.


Subject(s)
Apoptosis , Chondroitin Sulfates , Nanoparticles , Reactive Oxygen Species , Selenium , Humans , HeLa Cells , Chondroitin Sulfates/pharmacology , Chondroitin Sulfates/chemistry , Apoptosis/drug effects , Selenium/pharmacology , Selenium/chemistry , Nanoparticles/chemistry , Reactive Oxygen Species/metabolism , Cell Proliferation/drug effects , S Phase Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/drug effects , Antineoplastic Agents/pharmacology
6.
Int J Nanomedicine ; 19: 5125-5138, 2024.
Article in English | MEDLINE | ID: mdl-38855730

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Cell Proliferation , Doxorubicin , Liposomes , Tumor Microenvironment , Tumor Microenvironment/drug effects , Animals , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Humans , Mice , Liposomes/chemistry , MCF-7 Cells , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Cell Proliferation/drug effects , Mice, Inbred BALB C , NIH 3T3 Cells , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Particle Size , Nanoparticle Drug Delivery System/chemistry , Drug Delivery Systems/methods , Cell Movement/drug effects , Nanoparticles/chemistry
7.
Carbohydr Res ; 541: 109163, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38805806

ABSTRACT

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.


Subject(s)
Anticoagulants , Chondroitin Sulfates , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Chondroitin Sulfates/isolation & purification , Anticoagulants/chemistry , Anticoagulants/pharmacology , Anticoagulants/isolation & purification , Animals , Humans , Blood Coagulation/drug effects
8.
Int J Biol Macromol ; 269(Pt 2): 131952, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692541

ABSTRACT

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.


Subject(s)
Anticoagulants , Chondroitin Sulfates , Factor IXa , Molecular Weight , Animals , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Chondroitin Sulfates/isolation & purification , Anticoagulants/pharmacology , Anticoagulants/chemistry , Anticoagulants/isolation & purification , Factor IXa/metabolism , Factor IXa/antagonists & inhibitors , Factor IXa/chemistry , Cucumaria/chemistry , Sea Cucumbers/chemistry , Blood Coagulation/drug effects , Humans , Models, Molecular
9.
J Neural Eng ; 21(3)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38806019

ABSTRACT

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.


Subject(s)
Brain Injuries, Traumatic , Hydrogels , Neural Stem Cells , Neurites , Rats, Sprague-Dawley , Recovery of Function , Hydrogels/administration & dosage , Animals , Rats , Recovery of Function/drug effects , Recovery of Function/physiology , Humans , Neural Stem Cells/drug effects , Neurites/drug effects , Neurites/physiology , Male , Chondroitin Sulfates/administration & dosage , Chondroitin Sulfates/pharmacology , Glycosaminoglycans/administration & dosage , Cells, Cultured , Neurogenesis/drug effects , Neurogenesis/physiology
10.
Int J Biol Macromol ; 271(Pt 1): 132520, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38772463

ABSTRACT

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.


Subject(s)
Angiogenesis Inhibitors , Chondroitin Sulfates , Nanoparticles , Paclitaxel , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Paclitaxel/chemistry , Paclitaxel/therapeutic use , Animals , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Humans , Mice , Nanoparticles/chemistry , Cell Line, Tumor , Apoptosis/drug effects , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Drug Carriers/chemistry , Cell Movement/drug effects , Neovascularization, Pathologic/drug therapy , Human Umbilical Vein Endothelial Cells/drug effects , Drug Delivery Systems , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage
11.
J Mater Chem B ; 12(22): 5535-5550, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38747002

ABSTRACT

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.


Subject(s)
Chondroitin Sulfates , Coated Materials, Biocompatible , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Animals , Mice , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Surface Properties , Neurons/drug effects , Biofouling/prevention & control , Electrodes, Implanted
12.
Biomacromolecules ; 25(6): 3312-3324, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38728671

ABSTRACT

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.


Subject(s)
Alginates , Bioprinting , Cell Differentiation , Chondrogenesis , Chondroitin Sulfates , Gelatin , Hydrogels , Mesenchymal Stem Cells , Nanocomposites , Printing, Three-Dimensional , Tissue Scaffolds , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Alginates/chemistry , Alginates/pharmacology , Gelatin/chemistry , Bioprinting/methods , Cell Differentiation/drug effects , Chondrogenesis/drug effects , Nanocomposites/chemistry , Tissue Scaffolds/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Engineering/methods , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Graphite/chemistry , Graphite/pharmacology , Cell Proliferation/drug effects , Cells, Cultured
13.
ACS Biomater Sci Eng ; 10(5): 3242-3254, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38632852

ABSTRACT

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.


Subject(s)
Chondrocytes , Chondroitin Sulfates , Hyaluronic Acid , Hydrogels , Inflammation , Hydrogels/chemistry , Hydrogels/pharmacology , Chondrocytes/drug effects , Chondrocytes/metabolism , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Chondroitin Sulfates/pharmacology , Chondroitin Sulfates/chemistry , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cytokines/metabolism , Aggrecans/metabolism , Tissue Engineering/methods , Osteoarthritis/pathology , Osteoarthritis/drug therapy , Osteoarthritis/metabolism
14.
Mar Drugs ; 22(4)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667801

ABSTRACT

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.


Subject(s)
Chondroitin Sulfates , Oligosaccharides , Sea Cucumbers , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/chemical synthesis , Chondroitin Sulfates/pharmacology , Animals , Oligosaccharides/chemical synthesis , Oligosaccharides/chemistry , Sea Cucumbers/chemistry , Glycosylation , Fucose/chemistry , Anticoagulants/pharmacology , Anticoagulants/chemistry , Anticoagulants/chemical synthesis , Structure-Activity Relationship , Acetylgalactosamine/chemistry , Acetylgalactosamine/analogs & derivatives
15.
Int J Biol Macromol ; 266(Pt 2): 131425, 2024 May.
Article in English | MEDLINE | ID: mdl-38583830

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Chondroitin Sulfates , Disulfides , Molybdenum , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Animals , Mice , Humans , Molybdenum/chemistry , Molybdenum/pharmacology , Disulfides/chemistry , Disulfides/pharmacology , Amyloid beta-Peptides/metabolism , Reactive Oxygen Species/metabolism , Oxidative Stress/drug effects , Cell Line, Tumor , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Male , Disease Models, Animal
16.
Int J Biol Macromol ; 266(Pt 2): 131051, 2024 May.
Article in English | MEDLINE | ID: mdl-38556223

ABSTRACT

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.


Subject(s)
Chondroitin Sulfates , Collagen , Hyaluronic Acid , Hydrogels , Nanocomposites , Nanofibers , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Nanofibers/chemistry , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Mice , Collagen/chemistry , Nanocomposites/chemistry , Tissue Engineering/methods , Cartilage/drug effects , Chondrogenesis/drug effects , Cell Differentiation/drug effects , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
17.
J Mater Chem B ; 12(14): 3417-3435, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38525920

ABSTRACT

Due to the increasing aging population and the advancements in transcatheter aortic valve replacement (TAVR), the use of bioprosthetic heart valves (BHVs) in patients diagnosed with valvular disease has increased substantially. Commercially available glutaraldehyde (GA) cross-linked biological valves suffer from reduced durability due to a combination of factors, including the high cell toxicity of GA, subacute thrombus, inflammation and calcification. In this study, oxidized chondroitin sulfate (OCS), a natural polysaccharide derivative, was used to replace GA to cross-link decellularized bovine pericardium (DBP), carrying out the first crosslinking of DBP to obtain OCS-BP. Subsequently, the zwitterion radical copolymerization system was introduced in situ to perform double cross-linking to obtain double crosslinked BHVs with biomimetic modification (P(APM/MPC)-OCS-BP). P(APM/MPC)-OCS-BP presented enhanced mechanical properties, collagen stability and enzymatic degradation resistance due to double crosslinking. The ex vivo AV-shunt assay and coagulation factors test suggested that P(APM/MPC)-OCS-BP exhibited excellent anticoagulant and antithrombotic properties due to the introduction of P(APM/MPC). P(APM/MPC)-OCS-BP also showed good HUVEC-cytocompatibility due to the substantial reduction of its residual aldehyde group. The subcutaneous implantation also demonstrated that P(APM/MPC)-OCS-BP showed a weak inflammatory response due to the anti-inflammatory effect of OCS. Finally, in vivo and in vitro results revealed that P(APM/MPC)-OCS-BP exhibited an excellent anti-calcification property. In a word, this simple cooperative crosslinking strategy provides a novel solution to obtain BHVs with good mechanical properties, and HUVEC-cytocompatibility, anti-coagulation, anti-inflammatory and anti-calcification properties. It might be a promising alternative to GA-fixed BP and exhibited good prospects in clinical applications.


Subject(s)
Calcinosis , Heart Valve Prosthesis , Humans , Animals , Cattle , Aged , Chondroitin Sulfates/pharmacology , Cross-Linking Reagents/pharmacology , Heart Valves , Glutaral , Anti-Inflammatory Agents/pharmacology , Pericardium
18.
Carbohydr Polym ; 334: 121972, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38553198

ABSTRACT

Chronic wounds with bacterial infection present formidable clinical challenges. In this study, a versatile hydrogel dressing with antibacterial and angiogenic activity composite of silk fibroin (SF), chondroitin sulfate (CS), and graphene oxide quantum dots (GOQDs) is fabricated. GOQDs@SF/CS (GSC) hydrogel is rapidly formed through the enzyme catalytic action of horseradish peroxidase. With the incorporation of GOQDs both gelation speed and mechanical properties have been enhanced, and the photothermal characteristics of GOQDs in GSC hydrogel enabled bacterial killing through photothermal treatment (PTT) at ∼51 °C. In vitro studies show that the GSC hydrogels demonstrate excellent antibacterial performance and induce type H vessel differentiation of endothelial cells via the activated ERK1/2 signaling pathway and upregulated SLIT3 expression. In vivo results show that the hydrogel significantly promotes type H vessels formation, which is related to the collagen deposition, epithelialization and, ultimately, accelerates the regeneration of infected skin defects. Collectively, this multifunctional GSC hydrogel, with dual action of antibacterial efficacy and angiogenesis promotion, emerges as an innovative skin dressing with the potential for advancing in infected wound healing.


Subject(s)
Fibroins , Graphite , Quantum Dots , Fibroins/pharmacology , Chondroitin Sulfates/pharmacology , Hydrogels/pharmacology , Endothelial Cells , Wound Healing , Anti-Bacterial Agents/pharmacology
19.
Cryobiology ; 115: 104857, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38350589

ABSTRACT

The cryopreservation of endothelial cell monolayers is an important step that bridges the cryopreservation of cells in suspension to that of tissues. Previous studies have identified clear distinctions in freezing mechanisms between cells in suspension and in monolayers, as well as developed novel protocols for monolayer cryopreservation. Recently, our group has shown that human umbilical vein endothelial cell (HUVEC) and porcine corneal endothelial cell (PCEC) monolayers grown on Rinzl plastic substrate can be cryopreserved in 5% dimethyl sulfoxide, 6% hydroxyethyl starch, and 2% chondroitin sulfate, following a slow-cooling protocol (-1 °C/min) with rapid plunge into liquid nitrogen from -40 °C. However, membrane integrity assessments were done immediately post thaw, which may result in an overestimation of cell viability due to possible delayed injury responses. Here, we show that for the optimal protocol condition of plunge at the -40 °C interrupt temperature, HUVEC and PCEC monolayers exhibited no significant immediate post-thaw injuries nor delayed injury responses during the 24-h post-thaw overnight culture period. HUVEC monolayers experienced no significant impact to their natural growth rate during the post-thaw culture, while PCEC monolayers experienced significantly higher growth than the unfrozen controls. The difference in the low-temperature responses between HUVEC and PCEC monolayers was further shown under high temperature plunge conditions. At these suboptimal plunge temperatures, HUVEC monolayers exhibited moderate immediate membrane injury but a pronounced delayed injury response during the 24-h post-thaw culture, while PCEC monolayers showed significant immediate membrane injury but no additional delayed injury response during the same period. Therefore, we provide further validation of our group's previously designed endothelial monolayer cryopreservation protocol for HUVEC and PCEC monolayers, and we identify several cell-type-specific responses to the freezing process.


Subject(s)
Cell Survival , Cryopreservation , Cryoprotective Agents , Dimethyl Sulfoxide , Human Umbilical Vein Endothelial Cells , Cryopreservation/methods , Humans , Animals , Cell Survival/drug effects , Cryoprotective Agents/pharmacology , Swine , Dimethyl Sulfoxide/pharmacology , Chondroitin Sulfates/pharmacology , Endothelial Cells/cytology , Hydroxyethyl Starch Derivatives/pharmacology , Cells, Cultured , Endothelium, Corneal/cytology , Endothelium, Corneal/injuries
20.
Signal Transduct Target Ther ; 9(1): 39, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38355690

ABSTRACT

Immunostaining in lungs of patients who died with COVID-19 infection showed increased intensity and distribution of chondroitin sulfate and decline in N-acetylgalactostamine-4-sulfatase (Arylsulfatase B; ARSB). To explain these findings, human small airway epithelial cells were exposed to the SARS-CoV-2 spike protein receptor binding domain (SPRBD) and transcriptional mechanisms were investigated. Phospho-p38 MAPK and phospho-SMAD3 increased following exposure to the SPRBD, and their inhibition suppressed the promoter activation of the carbohydrate sulfotransferases CHST15 and CHST11, which contributed to chondroitin sulfate biosynthesis. Decline in ARSB was mediated by phospho-38 MAPK-induced N-terminal Rb phosphorylation and an associated increase in Rb-E2F1 binding and decline in E2F1 binding to the ARSB promoter. The increases in chondroitin sulfotransferases were inhibited when treated with phospho-p38-MAPK inhibitors, SMAD3 (SIS3) inhibitors, as well as antihistamine desloratadine and antibiotic monensin. In the mouse model of carrageenan-induced systemic inflammation, increases in phospho-p38 MAPK and expression of CHST15 and CHST11 and declines in DNA-E2F binding and ARSB expression occurred in the lung, similar to the observed effects in this SPRBD model of COVID-19 infection. Since accumulation of chondroitin sulfates is associated with fibrotic lung conditions and diffuse alveolar damage, increased attention to p38-MAPK inhibition may be beneficial in ameliorating Covid-19 infections.


Subject(s)
COVID-19 , N-Acetylgalactosamine-4-Sulfatase , Mice , Animals , Humans , N-Acetylgalactosamine-4-Sulfatase/metabolism , Chondroitin Sulfates/pharmacology , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Spike Glycoprotein, Coronavirus , Carbohydrate Sulfotransferases , Angiotensin-Converting Enzyme 2 , p38 Mitogen-Activated Protein Kinases/genetics , SARS-CoV-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL