Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Nanomedicine ; 56: 102726, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38052371

ABSTRACT

The pressing demand for innovative approaches to create delivery systems with heightened drug loading and prolonged circulation has spurred numerous efforts, yielding some successes but accompanied by constraints. Our study proposes employing dendritic lipopeptide with precisely balanced opposing charges to extend blood residency for biomimetic nanoplatforms. Neutrally mixed-charged zwitterionic nanoparticles (NNPs) achieved a notable 19 % simvastatin loading content and kept stable even after one-month storage at 4 °C. These nanoplatforms demonstrated low cytotoxicity in NIH-3T3 and L02 cells and negligible hemolysis (<5 %). NNPs inhibited protein adhesion (>95 %) from positively and negatively charged sources through surface hydration. In comparison to positively charged CNPs, NNPs demonstrated an 86 % decrease in phagocytic rate by BMDMs, highlighting their efficacy. Importantly, NNPs showed prolonged circulation compared to CNPs and free simvastatin. These findings highlight the potential of this biomimetic nanoplatform for future therapeutic applications with enhanced drug loading and circulation traits.


Subject(s)
Biomimetics , Nanoparticles , Pharmaceutical Preparations , Simvastatin/pharmacology , Nanoparticles/chemistry , Drug Delivery Systems
2.
Int J Biol Macromol ; 253(Pt 5): 127146, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37778581

ABSTRACT

There is a great demand for the fabrication of soft electronics using hydrogels due to their biomimetic structures and good flexibility. However, conventional hydrogels have poor mechanical properties, which restricts their applications as stretchable sensors. Herein, a facile one-step strategy is proposed to fabricate tough and conductive hydrogels by making use of the graftability of carboxymethyl chitosan without extra conductive matter and crosslinking agent. The obtained polyacrylamide/carboxymethyl chitosan composite hydrogels possess outstanding transmittance and excellent mechanical performances, with tensile breaking stress of 630 kPa, breaking strain of 4560 %, toughness of 8490 kJ/m3. These hydrogels have low modulus of 5-20 kPa, fast recoverability after unloading, high conductivity of ∼0.85 S/m without the addition of other conductive substances and good biocompatibility. The ionic conductivity of the gels originates from the counterions of carboxymethyl chitosan, affording the hydrogels as resistive-type sensors. The resultant hydrogel sensors demonstrate a broad strain window (0.12-1500 %), excellent linear response, high sensitivity with the gauge factor reaching 11.72, and great durability, capable of monitoring diverse human motions. This work provides a new strategy to develop stretchable conductive hydrogels with promising applications in the fields of artificial intelligence and flexible electronics.


Subject(s)
Chitosan , Humans , Chitosan/chemistry , Artificial Intelligence , Hydrogels/chemistry , Electric Conductivity
3.
Adv Mater ; 35(51): e2305529, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37549042

ABSTRACT

Drug treatment is required for both resectable and unresectable cancers to strive for any meaningful improvement in patient outcomes. However, the clinical benefit of receiving conventional systemic administrations is often less than satisfactory. Drug delivery systems are preferable substitutes but still fail to meet diverse clinical demands due to the difficulty in programming drug release profiles. Herein, a microfabrication concept, termed "Hierarchical Multiple Polymers Immobilization" (HMPI), is introduced and biodegradable-polymer-based hierarchical microdevices (HMDs) that can pre-program any desired controlled release profiles are engineered. Based on the first-line medication of pancreatic and breast cancer, controlled release of single gemcitabine and the doxorubicin/paclitaxel combination in situ for multiple courses is implemented, respectively. Preclinical models of postsurgical pancreatic, postsurgical breast, and unresectable breast cancer are established, and the designed HMDs are demonstrated as well-tolerable and effective treatments for inhibiting tumor growth, recurrence, and metastasis. The proposed HMPI strategy allows the creation of tailorable and high-resolution hierarchical microstructures for pre-programming controlled release according to clinical medication schedules, which may provide promising alternative treatments for postsurgical and unresectable tumor control.


Subject(s)
Breast Neoplasms , Drug Delivery Systems , Humans , Female , Delayed-Action Preparations , Gemcitabine , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Paclitaxel/therapeutic use
4.
Acta Biomater ; 169: 243-255, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37572980

ABSTRACT

Despite 3D bioprinting having emerged as an advanced method for fabricating complex in vitro models, developing suitable bioinks that fulfill the opposing requirements for the biofabrication window still remains challenging. Although naturally derived hydrogels can better mimic the extracellular matrix (ECM) of numerous tissues, their weak mechanical properties usually result in architecturally simple shapes and patchy functions of in vitro models. Here, this limitation is addressed by a peptide-dendrimer-reinforced bioink (HC-PDN) which contained the peptide-dendrimer branched PEG with end-grafted norbornene (PDN) and the cysteamine-modified HA (HC). The extensive introduction of ethylene end-groups facilitates the grafting of sufficient moieties and enhances thiol-ene-induced crosslinking, making HC-PDN exhibits improved mechanical and rheological properties, as well as a significant reduction in reactive oxygen species (ROS) accumulation than that of methacrylated hyaluronic acid (HAMA). In addition, HC-PDN can be applied for the bioprinting of numerous complex structures with superior shape fidelity and soft matrix microenvironment. A heterogeneous and biomimetic hepatic tissue is concretely constructed in this work. The HepG2-C3As, LX-2s, and EA.hy.926s utilized with HC-PDN and assisted GelMA bioinks closely resemble the parenchymal and non-parenchymal counterparts of the native liver. The bioprinted models show the endothelium barrier function, hepatic functions, as well as increased activity of drug-metabolizing enzymes, which are essential functions of liver tissue in vivo. All these properties make HC-PDN a promising bioink to open numerous opportunities for in vitro model biofabrication. STATEMENT OF SIGNIFICANCE: In this manuscript, we introduced a peptide dendrimer system, which belongs to the family of hyperbranched 3D nanosized macromolecules that exhibit high molecular structure regularity and various biological advantages. Specifically, norbornene-modified peptide dendrimer was grafted onto PEG, and hyaluronic acid (HA) was selected as a base material for bioink formulation because it is a component of the ECM. Peptide dendrimers confer the following advantages to bioinks: (a) Geometric symmetry can facilitate construction of bioinks with homogeneous networks; (b) abundant surface functional groups allow for abundant crosslinking points; (c) the biological origin can promote biocompatibility. This study shows conceptualization to application of a peptide-dendrimer bioink to extend the Biofabrication Window of natural bioinks and will expand use of 3D bioprinting of in vitro models.


Subject(s)
Bioprinting , Dendrimers , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Bioprinting/methods , Biomimetics , Hyaluronic Acid , Printing, Three-Dimensional , Hydrogels/chemistry , Peptides , Norbornanes
5.
Biomater Sci ; 11(19): 6384-6402, 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37552036

ABSTRACT

Many soft tissues of the human body possess hierarchically anisotropic structures, exhibiting orientation-specific mechanical properties and biological functionality. Hydrogels have been proposed as promising scaffold materials for tissue engineering applications due to their water-rich composition, excellent biocompatibility, and tunable physico-chemical properties. However, conventional hydrogels with homogeneous structures often exhibit isotropic properties that differ from those of biological tissues, limiting their further application. Recently, magnetically anisotropic hydrogels containing long-range ordered magneto-structures have attracted widespread interest owing to their highly controllable assembly strategy, rapid magnetic responsiveness and remote spatiotemporal regulation. In this review, we summarize the latest progress of magnetically anisotropic hydrogels for tissue engineering. The fabrication strategy of magnetically anisotropic hydrogels from magnetic nanofillers with different dimensions is systemically introduced. Then, the effects of magnetically anisotropic cues on the physicochemical properties of hydrogels and the cellular biological behavior are discussed. And the applications of magnetically anisotropic hydrogels in the engineering of different tissues are emphasized. Finally, the current challenges and the future perspectives for magnetically anisotropic hydrogels are presented.


Subject(s)
Hydrogels , Tissue Engineering , Humans , Tissue Engineering/methods , Hydrogels/chemistry
6.
Carbohydr Polym ; 317: 121103, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37364964

ABSTRACT

Injectable and stable hydrogels have great promise for clinical applications. Fine-tuning the injectability and the stability of the hydrogels at different stages has been challenging due to the limited number of coupling reactions. A distinct "reversible to irreversible" concept using a thiazolidine-based bioorthogonal reaction between 1,2-aminothiols and aldehydes in physiological conditions to surmount the dilemma between injectability and stability is presented for the first time. Upon mixing aqueous solutions of aldehyde-functionalized hyaluronic acid (SA-HA) and cysteine-capped ethylenediamine (DI-Cys), SA-HA/DI-Cys hydrogels formed through reversible hemithioacetal crosslinking within 2 min. The reversible kinetic intermediate facilitated thiol-triggered gel-to-sol transition, shear-thinning and injectability of the SA-HA/DI-Cys hydrogel but then converted to the irreversible thermodynamic network after injection, thereby permitting the resulting gel with improved stability. As compared to the Schiff base hydrogels, the hydrogels generated from this simple, yet effective concept awarded improved protection to the embedded mesenchymal stem cells and fibroblast during injection, retained the cells homogeneously within the gel, and allowed them further proliferation in vitro and in vivo. There is potential for the proposed approach of "reversible to irreversible" based on thiazolidine chemistry to be applied as a general coupling technique for developing injectable and stable hydrogels for biomedical applications.


Subject(s)
Hydrogels , Mesenchymal Stem Cells , Hyaluronic Acid , Thiazolidines , Sulfhydryl Compounds , Cysteine
7.
Int J Biol Macromol ; 243: 124971, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37236562

ABSTRACT

Mesenchymal stem cells (MSCs) have gained increasing attention in various biomedical applications. However, conventional therapeutic approaches, such as direct intravenous injection, are associated with low cell survival due to the shear force during injection and the oxidative stress microenvironments in the lesion area. Herein, a photo-crosslinkable antioxidant hydrogel based on tyramine- and dopamine-modified hyaluronic acid (HA-Tyr/HA-DA) was developed. Meanwhile, human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) were encapsulated in HA-Tyr/HA-DA hydrogel using a microfluidic system to create size-controllable microgels (hUC-MSCs@microgels). The HA-Tyr/HA-DA hydrogel was demonstrated to have good rheology, biocompatibility, and antioxidant properties for cell microencapsulation. The hUC-MSCs encapsulated in microgels showed a high viability and a significantly improved the survival rate under oxidative stress conditions. Therefore, the presented work provides a promising platform for MSCs microencapsulation, which may further improve the stem cell-based biomedical applications.


Subject(s)
Mesenchymal Stem Cells , Microgels , Humans , Reactive Oxygen Species , Hyaluronic Acid , Antioxidants , Hydrogels
8.
ACS Appl Mater Interfaces ; 15(20): 24034-24046, 2023 May 24.
Article in English | MEDLINE | ID: mdl-37159919

ABSTRACT

The emergence and innovation of three-dimensional (3D) bioprinting provide new development opportunities for tissue engineering and regenerative medicine. However, how to obtain bioinks with both biomimicry and manufacturability remains a great issue in 3D bioprinting. Developing intelligent responsive biomaterials is conducive to break through the current dilemma. Herein, a stepwise multi-cross-linking strategy concerning thermosensitive thiolated Pluronic F127 (PF127-SH) and hyaluronic acid methacrylate (HAMA) is proposed to achieve temperature-controlled 3D embedded bioprinting, specifically pre-cross-linking (Michael addition reaction) at low temperatures (4-20 °C) and subsequently self-assembly (hydrophobic interaction) in a high-temperature (30-37 °C) suspension bath as well as final photo-cross-linking (mainly thiol-ene "click" reaction). The unique stepwise cross-linking mechanism promises the thermosensitive bioink appropriate viscosity at different printing stages, making it possible to print complex structures with excellent shape fidelity and simultaneously maintain the biological activity of cells. In vitro studies reveal that 3D-printed hydrogels are beneficial for enhancing cell viability. Further, in vivo experiments demonstrate that cell-laden printed hydrogels significantly promote wound healing and re-epithelialization by modulating inflammation and accelerating collagen deposition and angiogenesis. Therefore, the proposed stepwise multi-cross-linking strategy is expected to accelerate the development of novel bioinks and promote the clinical applications of 3D bioprinting.


Subject(s)
Bioprinting , Tissue Scaffolds , Tissue Scaffolds/chemistry , Bioprinting/methods , Printing, Three-Dimensional , Tissue Engineering , Hydrogels/chemistry , Wound Healing
9.
Int J Biol Macromol ; 240: 124364, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37044319

ABSTRACT

Three-dimensional (3D) bioprinting is a promising technique to construct heterogeneous architectures that mimic cell microenvironment. However, the current bioinks for 3D bioprinting usually show some limitations, such as low printing accuracy, unsatisfactory mechanical properties and compromised cytocompatibility. Herein, a novel bioink comprising hydroxyphenyl propionic acid-conjugated gelatin and tyramine-modified alginate is developed for printing 3D constructs. The bioink takes advantage of an ionic/covalent intertwined network that combines covalent bonds formed by photo-mediated redox reaction and ionic bonds formed by chelate effect. Benefiting from the thermosensitivity of gelatin and the double-crosslinking mechanism, the developed bioink shows controllable rheological behaviors, enhanced mechanical behavior, improved printing accuracy and structure stability. Moreover, the printed cell-laden hydrogels exhibit a homogeneous cell distribution and considerable cell survival because the pre-crosslinking of the bioink prevents cellular sedimentation and the visible light crosslinking mechanism preserves cell viability. Further in vivo studies demonstrate that resulting cell-laden hydrogels are beneficial for the reduction of inflammation response and the promotion of collagen deposition and angiogenesis, thereby improving the quality of skin wound healing. This convenient and effective strategy is of great significance for accelerating the development of multifunctional bioinks and broadening the biomedical applications of 3D bioprinting.


Subject(s)
Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Gelatin/chemistry , Alginates/pharmacology , Alginates/chemistry , Printing, Three-Dimensional , Hydrogels/pharmacology , Hydrogels/chemistry , Wound Healing
10.
Biomacromolecules ; 24(2): 921-928, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36644840

ABSTRACT

Poly(trimethylene carbonate) (PTMC), as one of the representatives of biodegradable aliphatic polycarbonates, has been found to degrade in vivo via surface erosion. This unique degradation behavior and the resulting nonacidic products make it more competitive with aliphatic polyesters (e.g., polylactide) in clinical practice. However, this surface degradation mechanism is complicated and not fully understood to date despite the findings that several reactive oxygen species and enzymes can specifically degrade PTMC in vitro. Herein, the biodegradation mechanism of PTMC was investigated by using possible degradation factors, distinct cell lines, and the inhibitors of these factors. The results demonstrate that PTMC undergoes a specific macrophage-mediated erosion. Macrophages tend to fuse into giant cells and elicit a typical inflammatory response by releasing proinflammatory cytokines. In addition, macrophages are suggested to primarily secrete enzymes (lipase specifically) to erode the PTMC bulk extracellularly as inhibiting their activity effectively prevented this eroding process. The clarification of the biodegradation mechanism in this work suggests that the degradation of PTMC highly depends on the foreign body response. Thus, it reminds the researchers to consider the effect of the microenvironment on the degradation and drug release of PTMC-based implantation devices and localized drug delivery systems.


Subject(s)
Lipase , Polymers , Polymers/pharmacology , Dioxanes , Macrophages/metabolism
11.
J Adv Res ; 54: 89-104, 2023 12.
Article in English | MEDLINE | ID: mdl-36632888

ABSTRACT

INTRODUCTION: The bone ingrowth depth in the porous scaffolds is greatly affected by the structural design, notably the pore size, pore geometry, and the pore distribution. To enhance the bone regeneration capability of scaffolds, the bionic design can be regarded as a potential solution. OBJECTIVES: We proposed a Haversian system-like gradient structure based on the triply periodic minimal surface architectures with pore size varying from the edge to the center. And its effects in promoting bone regeneration were evaluated in the study. METHODS: The gradient scaffold was designed using the triply periodic minimal surface architectures. The mechanical properties were analyzed by the finite element simulation and confirmed using the universal machine. The fluid characteristics were calculated by the computational fluid dynamics analysis. The bone regeneration process was simulated using a in silico computational model containing the main biological, physical, and chemical variation during the bone growth process. Finally, the in vitro and in vivo studies were carried out to verify the actual osteogenic effect. RESULTS: Compared to the uniform scaffold, the biomimetic gradient scaffold demonstrated better performance in stress conduction and reduced stress shielding effects. The fluid features were appropriate for cell migration and flow diffusion, and the permeability was in the same order of magnitude with the natural bone. The bone ingrowth simulation exhibited improved angiogenesis and bone regeneration. Higher expression of the osteogenesis-related genes, higher alkaline phosphatase activity, and increased mineralization could be observed on the gradient scaffold in the in vitro study. The 12-week in vivo study proved that the gradient scaffold had deeper bone inserting depth and a more stable bone-scaffold interface. CONCLUSION: The Haversian system-like gradient structure can effectively promote the bone regeneration. This structural design can be used as a new solution for the clinical application of prosthesis design.


Subject(s)
Haversian System , Tissue Scaffolds , Tissue Scaffolds/chemistry , Porosity , Osteogenesis , Bone Regeneration
12.
ACS Biomater Sci Eng ; 9(1): 520-530, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36459430

ABSTRACT

Smart materials have great potential in many biomedical applications, in which biodegradable shape memory polymers (SMPs) can be used as surgical sutures, implants, and stents. Poly(dl-lactide-co-trimethylene carbonate) (PDLLTC) represents one of the promising SMPs and is widely used in biomedical applications. However, the relationship between its shape memory property and chemical structure has not been fully studied and needs further elaboration. In this work, PDLLTC copolymers in different compositions have been synthesized, and their shape memory properties have been investigated. It has been found that the shape memory property is related to the chemical composition and polymeric chain segments. The copolymer with a DLLA/TMC ratio of 75:25 (PDLLTC7525) has been demonstrated with great shape fixation and recovery ratio at human body temperature. Furthermore, PDLLTC7525-based self-morphing small-diameter vascular scaffolds adhered with inner electrospun aligned gelatin/hyaluronic acid (Gel/HA) nanofibers have been constructed, as a merit of its shape memory property. The scaffolds have been demonstrated to facilitate the proliferation and adhesion of endothelial cells on the inner layer. Therefore, PDLLTC with tailorable shape memory properties represents a promising candidate for the development of SMPs, as well as for small-diameter vascular scaffolds construction.


Subject(s)
Endothelial Cells , Polymers , Humans , Polymers/chemistry , Dioxanes/chemistry
13.
Carbohydr Polym ; 296: 119953, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36087997

ABSTRACT

Hydrogel tissue adhesives that currently available are often fabricated by mixing two or more polymeric components. Single-component hydrogels afford injectability, strong and reversible adhesion remain a formidable challenge. This research describes the creation of the first single-component hyaluronic acid hydrogel adhesive-based on phenylboronic acid-diol ester linkages. Phenylboronic acid can not only serve as a cross-linker to form hydrogel, but also act as an adhesion site for glycosyl compounds found in biological cell membranes. The rheological and compressive tests for the hydrogel show that it has excellent self-healing properties, good injectability and strong compressive strength. Adhesion tests demonstrated that the hydrogel has significantly greater adhesion strength than commercial fibrin glue. These findings suggest that the rational design of hydrogel precursors facilitates the formation of single-component networks and multiple functionalities. In vivo studies further proved the hydrogel was an ideal bio-adhesive with biocompatibility, absorbed wound exudate and hemostasis, and accelerated wound closure.


Subject(s)
Hyaluronic Acid , Hydrogels , Adhesives , Esters , Hemostasis , Hydrogels/chemistry , Wound Healing
14.
J Clin Lab Anal ; 36(11): e24705, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36125894

ABSTRACT

BACKGROUND: Congenital coagulation factor V (FV) deficiency is a very rare hemorrhagic disease with an incidence of approximately one in a million. The common clinical manifestations of FV deficiency include ecchymosis and mucosal bleeding. Life-threatening intracranial bleeding is rare. It has been reported in several cases. However, the molecular basis has been established in only a few cases. METHODS: We reported a 2-month-old girl with congenital FV deficiency and intracranial hemorrhage. Coagulation screening combined with clinical manifestations was performed to diagnose congenital FV deficiency. Genetic testing was performed to identify the pathogenic genes. A literature review was included to emphasize the clinical manifestation, diagnosis, and treatment for congenital FV deficiency with intracranial bleeding. RESULTS: The coagulation tests revealed a significantly prolonged prothrombin time (PT) of 51 s and an activated partial thromboplastin time (APTT) of 73.7 s. The patient had a plasma FV activity of 0.9%. Genetic testing showed compound heterozygous mutations of the patient's FV gene. A literature review showed that patients with homozygous or compound heterozygous variants of the FV gene were often associated with a severe bleeding phenotype. CONCLUSION: Our study provides a direction for the rapid and accurate diagnosis and treatment for FV deficiency to avoid life-threatening bleeding. Infants with spontaneous cranial hematoma and intracranial hemorrhage should be investigated for underlying hemostatic defects. Congenital coagulation factor deficiency should be considered. Once congenital FV deficiency is diagnosed, fresh frozen plasma (FFP) should be given on a regular basis. Liver transplantation may be performed in severe cases.


Subject(s)
Activated Protein C Resistance , Factor V Deficiency , Humans , Factor V/genetics , Factor V Deficiency/complications , Factor V Deficiency/genetics , Factor V Deficiency/congenital , Intracranial Hemorrhages/genetics
15.
Front Bioeng Biotechnol ; 10: 964080, 2022.
Article in English | MEDLINE | ID: mdl-35910020

ABSTRACT

Shape memory polymers (SMPs) have a wide range of potential applications in many fields. In particular, electrically driven SMPs have attracted increasing attention due to their unique electrical deformation behaviors. Carbon nanotubes (CNTs) are often used as SMP conductive fillers because of their excellent electrical conductivities. However, raw CNTs do not disperse into the polymer matrix well. This strictly limits their use. In this study, to improve their dispersion performance characteristics in the polymer matrix, hydroxylated multi-walled carbon nanotubes (MWCNT-OHs) were functionalized with octadecyl isocyanate (i-MWCNTs). Polyurethane with shape memory properties (SMPU) was synthesized using polycaprolactone diol (PCL-diol), hexamethylene diisocyanate (HDI), and 1,4-butanediol (BDO) at a 1:5:4 ratio. Then, electroactive shape memory composites were developed by blending SMPU with i-MWCNTs to produce SMPU/i-MWCNTs. The functionalized i-MWCNTs exhibited better dispersibility characteristics in organic solvents and SMPU composites than the MWCNT-OHs. The addition of i-MWCNTs reduced the crystallinity of SMPU without affecting the original chemical structure. In addition, the hydrogen bond index and melting temperature of the SMPU soft segment decreased significantly, and the thermal decomposition temperatures of the composites increased. The SMPU/i-MWCNT composites exhibited conductivity when the i-MWCNT content was 0.5 wt%. This conductivity increased with the i-MWCNT content. In addition, when the i-MWCNT content exceeded 1 wt%, the composite temperature could increase beyond 60°C within 140 s and the temporary structure could be restored to its initial state within 120 s using a voltage of 30 eV. Therefore, the functionalized CNTs exhibit excellent potential for use in the development of electroactive shape memory composites, which may be used in flexible electronics and other fields.

16.
Biomater Sci ; 10(16): 4479-4491, 2022 Aug 09.
Article in English | MEDLINE | ID: mdl-35792832

ABSTRACT

Although significant breakthroughs have been achieved in constructing complex tissue/organ models in vitro, the progress of 3D bioprinting has long been subjected to trade-offs between the printability and biocompatibility of bioinks. Methacrylated hyaluronic acid (HAMA) has been widely adopted for 3D bioprinting due to its limited immunogenicity and simple synthetic method. However, the challenges arising from HAMA are its limited mechanical properties, low printing resolution, inhomogeneity networks, and the accumulation of reactive oxygen species (ROS) during polymerization. Here, these limitations are addressed by developing a thiol-norbornene photoclick polysaccharide-based bioink (HC-HN), which is prepared through the modification of hyaluronic acid with norbornene functional groups (Nor) and cysteamine hydrochloride (Cys). Compared with traditional HAMA, the HC-HN bioink via a step-growth polymerization mechanism can realize increased viscoelastic properties, reduced ROS accumulation, and superior shape fidelity in a range of complex structures by 3D embedded bioprinting. To further confirm its potential in 3D bioprinting applications, the HC-HN bioink is employed to print a liver model in vitro, which shows higher albumin secretion and urea production. Furthermore, a markedly increased sensitivity to drug-induced hepatotoxicity is observed in the bioprinted liver model compared to the 2D culture. Therefore, the proposed photoclick HC-HN bioink expands the palette of available polysaccharide-based bioinks and greatly extends the biofabrication window to broaden the application opportunities of 3D bioprinting.


Subject(s)
Bioprinting , Bioprinting/methods , Hyaluronic Acid , Hydrogels/chemistry , Norbornanes , Printing, Three-Dimensional , Reactive Oxygen Species , Tissue Engineering/methods , Tissue Scaffolds/chemistry
17.
Adv Healthc Mater ; 11(15): e2200874, 2022 08.
Article in English | MEDLINE | ID: mdl-35657075

ABSTRACT

Although tissue adhesives have potential advantages over traditional sutures, existing ones suffer from several limitations: slow adhesion kinetic, low mechanical strength, and poor interfacial bonding with wet biological tissues. Herein, a cooperative mussel/slug double-bioinspired hydrogel adhesive (DBHA) composed of a robust adhesive interface and a stretchable dissipative matrix is developed. The DBHA is formed by a cationic polysaccharide (chitosan), an anionic polysaccharide (carboxymethyl cellulose), and a barbell-like dendritic lysine grafted with catechol groups (G3KPCA). Compared to various commercial bio-glues and traditional adhesives, the DBHA has significantly stronger tissue adhesion and enhanced toughness both ex vivo and in vivo. Meanwhile, the DBHA exhibits fast, strong, tough, and durable adhesion to diverse ex vivo tissue surfaces with blood. The adhesion energy between the adhesive and porcine skin can reach 200-900 J m-2 . Additionally, in vivo studies prove that DBHA has good hemostasis of rabbit artery trauma and achieves better wound healing of tissue incision than commercial bio-glues. This study provides a novel strategy for fabricating fast and strong wet adhesives, which can be used in many applications, such as soft robots, tissue adhesives and hemostats.


Subject(s)
Dendrimers , Tissue Adhesives , Adhesives/pharmacology , Animals , Hydrogels , Peptides , Rabbits , Swine , Tissue Adhesions , Tissue Adhesives/pharmacology
18.
Drug Deliv ; 29(1): 1595-1607, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35612309

ABSTRACT

At present, the 10-year survival rate of patients with pancreatic cancer is still less than 4%, mainly due to the high cancer recurrence rate caused by incomplete surgery and lack of effective postoperative adjuvant treatment. Systemic chemotherapy remains the only choice for patients after surgery; however, it is accompanied by off-target effects and server systemic toxicity. Herein, we proposed a biodegradable microdevice for local sustained drug delivery and postoperative pancreatic cancer treatment as an alternative and safe option. Biodegradable poly(l-lactic-co-glycolic acid) (P(L)LGA) was developed as the matrix material, gemcitabine hydrochloride (GEM·HCl) was chosen as the therapeutic drug and polyethylene glycol (PEG) was employed as the drug release-controlled regulator. Through adjusting the amount and molecular weight of PEG, the controllable degradation of matrix and the sustained release of GEM·HCl were obtained, thus overcoming the unstable drug release properties of traditional microdevices. The drug release mechanism of microdevice and the regulating action of PEG were studied in detail. More importantly, in the treatment of the postoperative recurrence model of subcutaneous pancreatic tumor in mice, the microdevice showed effective inhibition of postoperative in situ recurrences of pancreatic tumors with excellent biosafety and minimum systemic toxicity. The microdevice developed in this study provides an option for postoperative adjuvant pancreatic treatment, and greatly broadens the application prospects of traditional chemotherapy drugs.


Subject(s)
Neoplasm Recurrence, Local , Pancreatic Neoplasms , Animals , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Drug Delivery Systems , Humans , Lactic Acid , Mice , Neoplasm Recurrence, Local/drug therapy , Pancreatic Neoplasms/drug therapy , Polyethylene Glycols/therapeutic use , Gemcitabine , Pancreatic Neoplasms
19.
ACS Appl Mater Interfaces ; 14(19): 22426-22442, 2022 May 18.
Article in English | MEDLINE | ID: mdl-35533377

ABSTRACT

The metal gallium has enormous promise in fighting infections by disrupting bacterial iron metabolism via a "Trojan horse" trick. It is well worth trying to study the potential of gallium-mediated hydrogel for treating infected wounds. Herein, on the basis of a conventional gelation strategy of sodium alginate combined with metal ions, Ga3+ has been innovatively given a dual role in a dual-cross-linked hydrogel. It acts nor only as a cross-linking agent to form a hydrogel material but also as a therapeutic agent to slow-release and continuously treat infected wounds. Further photo-cross-linking is introduced to improve the mechanical properties of the hydrogel. Thus, a new gallium ionic- and photo-dual-cross-linked alginate hydrogel, with broad-spectrum antimicrobial activity and strengthened mechanical performance, for the treatment of infected wounds is reported. The morphology, degradability, swelling behavior, rheological properties, and gallium release kinetics together indicated the homogeneous and the strengthened mechanical performance of this hydrogel but did not impede the release of gallium ions. Interestingly, in vitro and in vivo results also demonstrated its favorable biocompatibility, reduced bacterial growth, and accelerated infected wound healing, making the gallium-incorporated hydrogel an ideal antimicrobial dressing.


Subject(s)
Gallium , Wound Infection , Alginates/pharmacology , Anti-Bacterial Agents/pharmacology , Gallium/pharmacology , Humans , Hydrogels/pharmacology , Ions , Wound Healing
20.
J Mater Chem B ; 10(37): 7148-7160, 2022 09 28.
Article in English | MEDLINE | ID: mdl-35475512

ABSTRACT

Due to their inherent and tunable biomechanical and biochemical performances, bioactive hydrogels based on polysaccharides and peptides have shown attractive potential for wound management. In this review, the recent progress of bioactive hydrogels prepared by polysaccharides and peptides for soft tissue wound management is overviewed. Meanwhile, we focus on the elaboration of the relationship between chemical structures and inherent bioactive functions of polysaccharides and peptides, as well as the strategies that are taken for achieving multiple wound repairing effects including hemostasis, adhesion, wound contraction and closure, anti-bacteria, anti-oxidation, immunomodulation, molecule delivery, etc. Some innovative and important works are well introduced as well. In the end, current study limitations, clinical unmet needs, and future directions are discussed.


Subject(s)
Hydrogels , Polysaccharides , Hydrogels/chemistry , Hydrogels/pharmacology , Peptides/pharmacology , Polysaccharides/chemistry , Polysaccharides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...