Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Colloid Interface Sci ; 672: 724-735, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38870763

ABSTRACT

The integration of functional nanomaterials with tissue engineering scaffolds has emerged as a promising solution for simultaneously treating malignant bone tumors and repairing resected bone defects. However, achieving a uniform bioactive interface on 3D-printing polymer scaffolds with minimized microstructural heterogeneity remains a challenge. In this study, we report a facile metal-coordination self-assembly strategy for the surface engineering of 3D-printed polycaprolactone (PCL) scaffolds with nanostructured two-dimensional conjugated metal-organic frameworks (cMOFs) consisting of Cu ions and 2,3,6,7,10,11-hexahydroxytriphenylene (HHTP). A tunable thickness of Cu-HHTP cMOF on PCL scaffolds was achieved via the alternative deposition of metal ions and HHTP. The resulting composite PCL@Cu-HHTP scaffolds not only demonstrated potent photothermal conversion capability for efficient OS ablation but also promoted the bone repair process by virtue of their cell-friendly hydrophilic interfaces. Therefore, the cMOF-engineered dual-functional 3D-printing scaffolds show promising potential for treating bone tumors by offering sequential anti-tumor effects and bone regeneration capabilities. This work also presents a new avenue for the interface engineering of bioactive scaffolds to meet multifaceted demands in osteosarcoma-related bone defects.


Subject(s)
Bone Neoplasms , Bone Regeneration , Osteosarcoma , Polyesters , Printing, Three-Dimensional , Tissue Scaffolds , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Osteosarcoma/therapy , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/therapy , Polyesters/chemistry , Humans , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Metal-Organic Frameworks/chemical synthesis , Surface Properties , Copper/chemistry , Copper/pharmacology , Hyperthermia, Induced , Tissue Engineering , Particle Size , Catalysis , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice , Cell Survival/drug effects , Nanostructures/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects
2.
Acta Neurochir (Wien) ; 166(1): 210, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38735896

ABSTRACT

PURPOSE: To evaluate the safety and efficacy of stereoelectroencephalography (SEEG)-guided radiofrequency thermocoagulation (RFTC) for drug-resistant focal epilepsy and investigate the relationship between post-RFTC remission duration and delayed excision surgery effectiveness. METHODS: We conducted a retrospective analysis of 43 patients with drug-resistant focal epilepsy who underwent RFTC via SEEG electrodes. After excluding three, the remaining 40 were classified into subgroups based on procedures and outcomes. Twenty-four patients (60%) underwent a secondary excision surgery. We determined the predictive value of RFTC outcome upon subsequent surgical outcome by categorizing the delayed secondary surgery outcome as success (Engel I/II) versus failure (Engel III/IV). Demographic information, epilepsy characteristics, and the duration of seizure freedom after RFTC were assessed. RESULTS: Among 40 patients, 20% achieved Engel class I with RFTC alone, while 24 underwent delayed secondary excision surgery. Overall, 41.7% attained Engel class I, with a 66.7% success rate combining RFTC with delayed surgery. Seizure freedom duration was significantly longer in the success group (mean 4.9 months, SD = 2.7) versus the failure group (mean 1.9 months, SD = 1.1; P = 0.007). A higher proportion of RFTC-only and delayed surgical success group patients had preoperative lesional findings (p = 0.01), correlating with a longer time to seizure recurrence (p < 0.05). Transient postoperative complications occurred in 10%, resolving within a year. CONCLUSION: This study demonstrates that SEEG-guided RFTC is a safe and potential treatment option for patients with drug-resistant focal epilepsy. A prolonged duration of seizure freedom following RFTC may serve as a predictive marker for the success of subsequent excision surgery.


Subject(s)
Drug Resistant Epilepsy , Electrocoagulation , Electroencephalography , Epilepsies, Partial , Humans , Male , Female , Adult , Electrocoagulation/methods , Electroencephalography/methods , Retrospective Studies , Drug Resistant Epilepsy/surgery , Treatment Outcome , Epilepsies, Partial/surgery , Epilepsies, Partial/physiopathology , Young Adult , Middle Aged , Adolescent , Prognosis , Stereotaxic Techniques , Child
3.
Adv Healthc Mater ; : e2400431, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38768997

ABSTRACT

The repair and regeneration of cartilage has always been a hot topic in medical research. Cartilage organoids (CORGs) are special cartilage tissue created using tissue engineering techniques outside the body. These engineered organoids tissues provide models that simulate the complex biological functions of cartilage, opening new possibilities for cartilage regenerative medicine and treatment strategies. However, it is crucial to establish suitable matrix scaffolds for the cultivation of CORGs. In recent years, utilizing hydrogel to culture stem cells and induce their differentiation into chondrocytes has emerged as a promising method for the in vitro construction of CORGs. In this review, the methods for establishing CORGs are summarized and an overview of the advantages and limitations of using matrigel in the cultivation of such organoids is provided. Furthermore, the importance of cartilage tissue ECM and alternative hydrogel substitutes for Matrigel, such as alginate, peptides, silk fibroin, and DNA derivatives is discussed, and the pros and cons of using these hydrogels for the cultivation of CORGs are outlined. Finally, the challenges and future directions in hydrogel research for CORGs are discussed. It is hoped that this article provides valuable references for the design and development of hydrogels for CORGs.

4.
J Am Chem Soc ; 146(18): 12547-12555, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38656766

ABSTRACT

Three-dimensional (3D) crystalline organic frameworks with complex topologies, high surface area, and low densities afford a variety of application prospects. However, the design and construction of these frameworks have been largely limited to systems containing polyhedron-shaped building blocks or those relying on component interpenetration. Here, we report the synthesis of a 3D crystalline organic framework based on molecular mortise-and-tenon jointing. This new material takes advantage of tetra(4-pyridylphenyl)ethylene and chlorinated bis(benzodioxaborole)benzene as building blocks and is driven by dative B-N bonds. A single-crystal X-ray diffraction analysis of the framework reveals the presence of two-dimensional (2D) layers with helical channels that are formed presumably during the boron-nitrogen coordination process. The protrusion of dichlorobenzene units from the upper and lower surfaces of the 2D layers facilitates the key mortise-and-tenon connections. These connections enable the interlocking of adjacent layers and the stabilization of an overall 3D framework. The resulting framework is endowed with high porosity and attractive mechanical properties, rendering it potentially suitable for the removal of impurities from acetylene.

5.
J Mater Chem B ; 12(17): 4148-4161, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38591180

ABSTRACT

Cyaonoside A (CyA), derived from the natural Chinese medicine, Cyathula officinalis Kuan, which was for a long time used to treat knee injuries and relieve joint pain in traditional Chinese medicine, showed an unclear mechanism for protecting cartilage. In addition, CyA was poorly hydrosoluble and incapable of being injected directly into the joint cavity, which limited its clinical application. This study reveals that CyA resisted IL-1ß-mediated chondrogenic inflammation and apoptosis. Next, transcriptome sequencing is used to explore the potential mechanisms underlying CyA regulation of MSC chondrogenic differentiation. Based on these findings, CyA-loaded composite hydrogel microspheres (HLC) were developed and they possessed satisfactory loading efficiency, a suitable degradation rate and good biocompatibility. HLC increased chondrogenic anabolic gene (Acan, COL2A, and SOX9) expression, while downregulating the expression of the catabolic marker MMP13 in vitro. In the osteoarthritis mouse model, HLC demonstrated promising therapeutic capabilities by protecting the integrity of articular cartilage. In conclusion, this study provides insights into the regulatory mechanisms of CyA for chondrocytes and proposes a composite hydrogel microsphere-based advanced therapeutic strategy for osteoarthritis.


Subject(s)
Chondrocytes , Hydrogels , Microspheres , Osteoarthritis , Chondrocytes/drug effects , Chondrocytes/metabolism , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Mice , Inflammation/drug therapy , Mice, Inbred C57BL , Male , Particle Size , Cells, Cultured
6.
Angew Chem Int Ed Engl ; 63(23): e202405761, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38587998

ABSTRACT

Vitrimers offer a unique combination of mechanical performance, reprocessability, and recyclability that makes them highly promising for a wide range of applications. However, achieving dynamic behavior in vitrimeric materials at their intended usage temperatures, thus combining reprocessability with adaptivity through associative dynamic covalent bonds, represents an attractive but formidable objective. Herein, we couple boron-nitrogen (B-N) dative bonds and B-O covalent bonds to generate a new class of vitrimers, boron-nitrogen vitrimers (BNVs), to endow them with dynamic features at usage temperatures. Compared with boron-ester vitrimers (BEVs) without B-N dative bonds, the BNVs with B-N dative bonds showcase enhanced mechanical performance. The excellent mechanical properties come from the synergistic effect of the dative B-N supramolecular polymer and covalent boron-ester networks. Moreover, benefiting from the associative exchange of B-O dynamic covalent bonds above their topological freezing temperature (Tv), the resultant BNVs also possess the processability. This study leveraged the structural characteristics of a boron-based vitrimer to achieve material reinforcement and toughness enhancement, simultaneously providing novel design concepts for the construction of new vitrimeric materials.

7.
Adv Mater ; : e2309875, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38642033

ABSTRACT

Addressing large bone defects remains a significant challenge owing to the inherent limitations in self-healing capabilities, resulting in prolonged recovery and suboptimal regeneration. Although current clinical solutions are available, they have notable shortcomings, necessitating more efficacious approaches to bone regeneration. Organoids derived from stem cells show great potential in this field; however, the development of bone organoids has been hindered by specific demands, including the need for robust mechanical support provided by scaffolds and hybrid extracellular matrices (ECM). In this context, bioprinting technologies have emerged as powerful means of replicating the complex architecture of bone tissue. The research focused on the fabrication of a highly intricate bone ECM analog using a novel bioink composed of gelatin methacrylate/alginate methacrylate/hydroxyapatite (GelMA/AlgMA/HAP). Bioprinted scaffolds facilitate the long-term cultivation and progressive maturation of extensive bioprinted bone organoids, foster multicellular differentiation, and offer valuable insights into the initial stages of bone formation. The intrinsic self-mineralizing quality of the bioink closely emulates the properties of natural bone, empowering organoids with enhanced bone repair for both in vitro and in vivo applications. This trailblazing investigation propels the field of bone tissue engineering and holds significant promise for its translation into practical applications.

8.
Mater Today Bio ; 25: 101014, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38464497

ABSTRACT

Traditional hydrogel design and optimization methods usually rely on repeated experiments, which is time-consuming and expensive, resulting in a slow-moving of advanced hydrogel development. With the rapid development of artificial intelligence (AI) technology and increasing material data, AI-energized design and optimization of hydrogels for biomedical applications has emerged as a revolutionary breakthrough in materials science. This review begins by outlining the history of AI and the potential advantages of using AI in the design and optimization of hydrogels, such as prediction and optimization of properties, multi-attribute optimization, high-throughput screening, automated material discovery, optimizing experimental design, and etc. Then, we focus on the various applications of hydrogels supported by AI technology in biomedicine, including drug delivery, bio-inks for advanced manufacturing, tissue repair, and biosensors, so as to provide a clear and comprehensive understanding of researchers in this field. Finally, we discuss the future directions and prospects, and provide a new perspective for the research and development of novel hydrogel materials for biomedical applications.

9.
Bioact Mater ; 35: 1-16, 2024 May.
Article in English | MEDLINE | ID: mdl-38298451

ABSTRACT

Segmental bone defects, stemming from trauma, infection, and tumors, pose formidable clinical challenges. Traditional bone repair materials, such as autologous and allogeneic bone grafts, grapple with limitations including source scarcity and immune rejection risks. The advent of nucleic acid nanotechnology, particularly the use of DNA hydrogels in tissue engineering, presents a promising solution, attributed to their biocompatibility, biodegradability, and programmability. However, these hydrogels, typically hindered by high gelation temperatures (∼46 °C) and high construction costs, limit cell encapsulation and broader application. Our research introduces a novel polymer-modified DNA hydrogel, developed using nucleic acid nanotechnology, which gels at a more biocompatible temperature of 37 °C and is cost-effective. This hydrogel then incorporates tetrahedral Framework Nucleic Acid (tFNA) to enhance osteogenic mineralization. Furthermore, considering the modifiability of tFNA, we modified its chains with Aptamer02 (Apt02), an aptamer known to foster angiogenesis. This dual approach significantly accelerates osteogenic differentiation in bone marrow stromal cells (BMSCs) and angiogenesis in human umbilical vein endothelial cells (HUVECs), with cell sequencing confirming their targeting efficacy, respectively. In vivo experiments in rats with critical-size cranial bone defects demonstrate their effectiveness in enhancing new bone formation. This innovation not only offers a viable solution for repairing segmental bone defects but also opens avenues for future advancements in bone organoids construction, marking a significant advancement in tissue engineering and regenerative medicine.

10.
World Neurosurg ; 184: e408-e416, 2024 04.
Article in English | MEDLINE | ID: mdl-38309654

ABSTRACT

OBJECTIVE: To analyze the relationship between trajectory-skull angle and stereoelectroencephalography electrode implantation accuracy in drug-resistant epilepsy patients, aiming to guide clinical electrode placement and enhance surgical precision and safety. METHODS: We conducted a retrospective analysis of medical records and surgical characteristics of 32 consecutive patients diagnosed with drug-resistant epilepsy, who underwent stereoelectroencephalography procedures at our center from June 2020 to June 2023. To evaluate the accuracy of electrode implantation, we utilized preoperative and postoperative computed tomography scans fused with SinoPlan software-planned trajectories. Entry radial error and target vector error were assessed as measurements of electrode implantation accuracy. RESULTS: After adjusting for confounders, we found a significant positive correlation between trajectory-skull angle and entry radial error (ß = 0.02, 95% CI: 0.01-0.03, P < 0.001). Likewise, a significant positive correlation existed between trajectory-skull angle and target vector error in all three models (ß = 0.03, 95% CI: 0.01-0.04, P < 0.001). Additionally, a U-shaped relationship between trajectory-skull angle and target vector error was identified using smooth curve fitting. This U-shaped pattern persisted in both frame-based and robot-guided stereotactic techniques. According to the two-piecewise linear regression model, the inflection points were 9° in the frame-based group and 16° in the robot-guided group. CONCLUSIONS: This study establishes a significant positive linear correlation between trajectory-skull angle and entry radial error, along with a distinctive U-shaped pattern in the relationship between trajectory-skull angle and target vector error. Our findings suggest that trajectory-skull angles of 9° (frame-based) and 16° (robot-guided) may optimize the accuracy of target vector error.


Subject(s)
Drug Resistant Epilepsy , Electroencephalography , Humans , Retrospective Studies , Electroencephalography/methods , Electrodes, Implanted , Stereotaxic Techniques , Drug Resistant Epilepsy/diagnostic imaging , Drug Resistant Epilepsy/surgery , Skull
11.
Angew Chem Int Ed Engl ; 63(13): e202317947, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38298087

ABSTRACT

Although our knowledge and understanding of adsorptions in natural and artificial systems has increased dramatically during the past century, adsorption associated with nonporous polymers remains something of a mystery, hampering applications. Here we demonstrate a model system for adaptisorption of nonporous polymers, wherein dative B-N bonds and host-guest binding units act as the kinetic and thermodynamic components, respectively. The coupling of these two components enables nonporous polymer crystals to adsorb molecules from solution and undergo recrystallization as thermodynamically favored crystals. Adaptisorption of nonporous polymer crystals not only extends the types of adsorption in which the sorbate molecules are integrated in a precise and orderly manner in the sorbent systems, but also provides a facile and accurate approach to the construction of polymeric materials with precise architectures and integrated functions.

12.
Acta Pharm Sin B ; 14(2): 579-601, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38322344

ABSTRACT

Lipid nanovehicles are currently the most advanced vehicles used for RNA delivery, as demonstrated by the approval of patisiran for amyloidosis therapy in 2018. To illuminate the unique superiority of lipid nanovehicles in RNA delivery, in this review, we first introduce various RNA therapeutics, describe systemic delivery barriers, and explain the lipid components and methods used for lipid nanovehicle preparation. Then, we emphasize crucial advances in lipid nanovehicle design for overcoming barriers to systemic RNA delivery. Finally, the current status and challenges of lipid nanovehicle-based RNA therapeutics in clinical applications are also discussed. Our objective is to provide a comprehensive overview showing how to utilize lipid nanovehicles to overcome multiple barriers to systemic RNA delivery, inspiring the development of more high-performance RNA lipid nanovesicles in the future.

13.
Bioact Mater ; 35: 429-444, 2024 May.
Article in English | MEDLINE | ID: mdl-38390528

ABSTRACT

Osteoarthritis (OA), a common degenerative disease, is characterized by high disability and imposes substantial economic impacts on individuals and society. Current clinical treatments remain inadequate for effectively managing OA. Organoids, miniature 3D tissue structures from directed differentiation of stem or progenitor cells, mimic native organ structures and functions. They are useful for drug testing and serve as active grafts for organ repair. However, organoid construction requires extracellular matrix-like 3D scaffolds for cellular growth. Hydrogel microspheres, with tunable physical and chemical properties, show promise in cartilage tissue engineering by replicating the natural microenvironment. Building on prior work on SF-DNA dual-network hydrogels for cartilage regeneration, we developed a novel RGD-SF-DNA hydrogel microsphere (RSD-MS) via a microfluidic system by integrating photopolymerization with self-assembly techniques and then modified with Pep-RGDfKA. The RSD-MSs exhibited uniform size, porous surface, and optimal swelling and degradation properties. In vitro studies demonstrated that RSD-MSs enhanced bone marrow mesenchymal stem cells (BMSCs) proliferation, adhesion, and chondrogenic differentiation. Transcriptomic analysis showed RSD-MSs induced chondrogenesis mainly through integrin-mediated adhesion pathways and glycosaminoglycan biosynthesis. Moreover, in vivo studies showed that seeding BMSCs onto RSD-MSs to create cartilage organoid precursors (COPs) significantly enhanced cartilage regeneration. In conclusion, RSD-MS was an ideal candidate for the construction and long-term cultivation of cartilage organoids, offering an innovative strategy and material choice for cartilage regeneration and tissue engineering.

14.
Mater Today Bio ; 24: 100948, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38269053

ABSTRACT

Articular cartilage injury is a frequent worldwide disease, while effective treatment is urgently needed. Due to lack of blood vessels and nerves, the ability of cartilage to self-repair is limited. Despite the availability of various clinical treatments, unfavorable prognoses and complications remain prevalent. However, the advent of tissue engineering and regenerative medicine has generated considerable interests in using biomaterials for articular cartilage repair. Nevertheless, there remains a notable scarcity of comprehensive reviews that provide an in-depth exploration of the various strategies and applications. Herein, we present an overview of the primary biomaterials and bioactive substances from the tissue engineering perspective to repair articular cartilage. The strategies include regeneration, substitution, and immunization. We comprehensively delineate the influence of mechanically supportive scaffolds on cellular behavior, shedding light on emerging scaffold technologies, including stimuli-responsive smart scaffolds, 3D-printed scaffolds, and cartilage bionic scaffolds. Biologically active substances, including bioactive factors, stem cells, extracellular vesicles (EVs), and cartilage organoids, are elucidated for their roles in regulating the activity of chondrocytes. Furthermore, the composite bioactive scaffolds produced industrially to put into clinical use, are also explicitly presented. This review offers innovative solutions for treating articular cartilage ailments and emphasizes the potential of biomaterials for articular cartilage repair in clinical translation.

15.
Small ; 20(24): e2306389, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38168513

ABSTRACT

In view of the increased levels of reactive oxygen species (ROS) that disturb the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), the repair of diabetic bone defects remains a great challenge. Herein, a factor-free hydrogel is reported with ROS scavenging and responsive degradation properties for enhanced diabetic bone healing. These hydrogels contain ROS-cleavable thioketal (TK) linkers and ultraviolet (UV)-responsive norbornene (NB) groups conjugated with 8-arm PEG macromers, which are formed via UV crosslinking-mediated gelation. Upon reacting with high levels of ROS in the bone defect microenvironment, ROS-cleavable TK linkers are destroyed, allowing the responsive degradation of hydrogels, which promotes the migration of BMSCs. Moreover, ROS levels are reduced through hydrogel-mediated ROS scavenging to reverse BMSC differentiation from adipogenic to osteogenic phenotype. As such, a favorable microenvironment is created after simultaneous ROS scavenging and hydrogel degradation, leading to the effective repair of bone defects in diabetic mouse models, even without the addition of growth factors. Thus, this study presents a responsive hydrogel platform that regulates ROS scavenging and stromal degradation in bone engineering.


Subject(s)
Cell Differentiation , Hydrogels , Mesenchymal Stem Cells , Osteogenesis , Reactive Oxygen Species , Animals , Reactive Oxygen Species/metabolism , Hydrogels/chemistry , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Mice , Cell Differentiation/drug effects , Diabetes Mellitus, Experimental , Free Radical Scavengers/pharmacology , Free Radical Scavengers/chemistry , Wound Healing/drug effects , Bone and Bones , Male
16.
Small ; 20(8): e2302506, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37814373

ABSTRACT

Osteoarthritis (OA) is a chronic disease that causes pain and disability in adults, affecting ≈300 million people worldwide. It is caused by damage to cartilage, including cellular inflammation and destruction of the extracellular matrix (ECM), leading to limited self-repairing ability due to the lack of blood vessels and nerves in the cartilage tissue. Organoid technology has emerged as a promising approach for cartilage repair, but constructing joint organoids with their complex structures and special mechanisms is still challenging. To overcome these boundaries, 3D bioprinting technology allows for the precise design of physiologically relevant joint organoids, including shape, structure, mechanical properties, cellular arrangement, and biological cues to mimic natural joint tissue. In this review, the authors will introduce the biological structure of joint tissues, summarize key procedures in 3D bioprinting for cartilage repair, and propose strategies for constructing joint organoids using 3D bioprinting. The authors also discuss the challenges of using joint organoids' approaches and perspectives on their future applications, opening opportunities to model joint tissues and response to joint disease treatment.


Subject(s)
Bioprinting , Tissue Engineering , Humans , Tissue Engineering/methods , Bioprinting/methods , Printing, Three-Dimensional , Organoids , Extracellular Matrix/chemistry , Tissue Scaffolds/chemistry
17.
J Am Chem Soc ; 146(1): 1109-1121, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38141046

ABSTRACT

The energy dissipative features of hydrogen bonds under conditions of mechanical strain have provided an ongoing incentive to explore hydrogen bonding units for the purpose of controlling and customizing the mechanical properties of polymeric materials. However, there remains a need for hydrogen bond units that (1) possess directionality, (2) provide selectivity, (3) dissipate energy effectively, and (4) can be incorporated readily into polymeric materials to regulate their mechanical properties. Here, we report mechanically interlocked hydrogen bond units that incorporate multiple hydrogen bonds within a [2]catenane structure. The conformational flexibility and associated spatial folding characteristics of the [2]catenane units allow for molecular scale motion under external stress, while the interlocked structure serves as a pivot that maintains the directionality and selectivity of the resultant hydrogen bonding units. When incorporated into polymers, these interlocked hydrogen bond motifs serve to strengthen and toughen the resulting materials. This study not only presents a novel hydrogen bond unit for creating polymeric materials with improved mechanical properties but also underscores the unique opportunities that mechanically interlocked hydrogen bond structures may provide across a diverse range of applications.

18.
Chemistry ; 29(71): e202303071, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-37843981

ABSTRACT

Fluorescent supramolecular polymers have garnered significant attention due to their successful integration of supramolecular polymers and fluorescence, offering vast potential for applications in sensing, imaging, optoelectronics, and photonics. In this study, we present a novel supramolecular polymer based on P5-OH, derived from mono-substituted pillararene macrocycles. Notably, these formed supramolecular polymeric aggregates exhibit a prominent blue emission, representing a rare instance of fluorescent polymers devoid of conventional chromophores. Furthermore, through the modification of alkyl chain ending groups attached to pillar[5]arenes, slight shifts in the emission peak could be observed. This research expands the scope of functional supramolecular polymeric systems utilizing pillararenes, providing valuable insights for the design of innovative luminescent materials and optical devices.

19.
Pharmaceutics ; 15(7)2023 Jun 25.
Article in English | MEDLINE | ID: mdl-37514001

ABSTRACT

Osteoarthritis (OA) is a degenerative disease of the joints characterized by cartilage damage and severe pain. Despite various pharmacological and surgical interventions, current therapies fail to halt OA progression, leading to high morbidity and an economic burden. Thus, there is an urgent need for alternative therapeutic approaches that can effectively address the underlying pathophysiology of OA. Extracellular Vesicles (EVs) derived from mesenchymal stromal cells (MSCs) represent a new paradigm in OA treatment. MSC-EVs are small membranous particles released by MSCs during culture, both in vitro and in vivo. They possess regenerative properties and can attenuate inflammation, thereby promoting cartilage healing. Importantly, MSC-EVs have several advantages over MSCs as cell-based therapies, including lower risks of immune reactions and ethical issues. Researchers have recently explored different strategies, such as modifying EVs to enhance their delivery, targeting efficiency, and security, with promising results. This article reviews how MSC-EVs can help treat OA and how they might work. It also briefly discusses the benefits and challenges of using MSC-EVs and talks about the possibility of allogeneic and autologous MSC-EVs for medical use.

20.
Sci Adv ; 9(27): eadi1169, 2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37406124

ABSTRACT

The integration of mechanically interlocked molecules (MIMs) into purely organic crystalline materials is expected to produce materials with properties that are not accessible using more classic approaches. To date, this integration has proved elusive. We present a dative boron-nitrogen bond-driven self-assembly strategy that allows for the preparation of polyrotaxane crystals. The polyrotaxane nature of the crystalline material was confirmed by both single-crystal x-ray diffraction analysis and cryogenic high-resolution low-dose transmission electron microscopy. Enhanced softness and greater elasticity are seen for the polyrotaxane crystals than for nonrotaxane polymer controls. This finding is rationalized in terms of the synergetic microscopic motion of the rotaxane subunits. The present work thus highlights the benefits of integrating MIMs into crystalline materials.

SELECTION OF CITATIONS
SEARCH DETAIL
...