Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
J Orthop Res ; 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39282769

RESUMEN

This study aimed to investigate the biocompatibility and osseointegration of novel titanium (Ti) implants with a perforated part with high surface roughness (Ra >4 µm) and a smooth solid part (test group), as compared to smooth solid Ti implants (control group; Ra < 0.8 µm). Test and control implants were implanted in rabbit femurs. After 4 and 15 weeks, host tissue reaction and quality of tissue formed were evaluated with histopathology, while micro-CT scans were used to quantitatively assess bone-implant contact (BIC), surrounding bone formation, and bone ingrowth. After 4 and 15 weeks, minimal host reaction was found in the test group. Histopathological analysis showed new bone formation around the implants in both the test and control groups after 4 weeks. Furthermore, additional bone growth was often observed within the holes of the test implants. After 15 weeks, the test implants showed high bone ingrowth and the presence of mature bone in direct contact with the implant surface, whereas, bone ingrowth was poorer for the control group with 30% of the control implants, showing larger gaps at the bone-implant interface. Quantitative micro-CT analysis revealed comparable BIC and bone formation in both groups at 4 weeks, but higher BIC and more bone formation in the test group than in the control group after 15 weeks. No significant differences were observed in any of the analyses. In conclusion, partially perforated, high-roughness Ti implants showed excellent osseointegration and minimal host reaction, indicating their potential for orthopedic applications in bone repair and regeneration.

2.
Results Probl Cell Differ ; 73: 249-297, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39242383

RESUMEN

The landscape of exosome research has undergone a significant paradigm shift, with a departure from early conceptions of exosomes as vehicles for cellular waste disposal towards their recognition as integral components of cellular communication with therapeutic potential. This chapter presents an exhaustive elucidation of exosome biology, detailing the processes of exosome biogenesis, release, and uptake, and their pivotal roles in signal transduction, tissue repair, regeneration, and intercellular communication. Additionally, the chapter highlights recent innovations and anticipates future directions in exosome research, emphasizing their applicability in clinical settings. Exosomes have the unique ability to navigate through tissue spaces to enter the circulatory system, positioning them as key players in tissue repair. Their contributory role in various processes of tissue repair, although in the nascent stages of investigation, stands out as a promising area of research. These vesicles function as a complex signaling network for intracellular and organ-level communication, critical in both pathological and physiological contexts. The chapter further explores the tissue-specific functionality of exosomes and underscores the advancements in methodologies for their isolation and purification, which have been instrumental in expanding the scope of exosome research. The differential cargo profiles of exosomes, dependent on their cellular origin, position them as prospective diagnostic biomarkers for tissue damage and regenerative processes. Looking ahead, the trajectory of exosome research is anticipated to bring transformative changes to biomedical fields. This includes advancing diagnostic and prognostic techniques that utilize exosomes as non-invasive biomarkers for a plethora of diseases, such as cancer, neurodegenerative, and cardiovascular conditions. Additionally, engineering exosomes through alterations of their native content or surface properties presents a novel frontier, including the synthesis of artificial or hybrid variants with enhanced functional properties. Concurrently, the ethical and regulatory frameworks surrounding exosome research, particularly in clinical translation, will require thorough deliberation. In conclusion, the diverse aspects of exosome research are coalescing to redefine the frontiers of diagnostic and therapeutic methodologies, cementing its importance as a discipline of considerable consequence in the biomedical sciences.


Asunto(s)
Comunicación Celular , Exosomas , Exosomas/metabolismo , Humanos , Comunicación Celular/fisiología , Animales , Cicatrización de Heridas/fisiología , Transducción de Señal/fisiología
3.
Stem Cell Res Ther ; 15(1): 219, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39026298

RESUMEN

Epicardial epithelial-to-mesenchymal transition (EMT) plays a pivotal role in both heart development and injury response and involves dynamic cellular changes that are essential for cardiogenesis and myocardial repair. Specifically, epicardial EMT is a crucial process in which epicardial cells lose polarity, migrate into the myocardium, and differentiate into various cardiac cell types during development and repair. Importantly, following EMT, the epicardium becomes a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis and contribute to cardiac remodeling after injury. As such, EMT seems to represent a fundamental step in cardiac repair. Nevertheless, endogenous EMT alone is insufficient to stimulate adequate repair. Redirecting and amplifying epicardial EMT pathways offers promising avenues for the development of innovative therapeutic strategies and treatment approaches for heart disease. In this review, we present a synthesis of recent literature highlighting the significance of epicardial EMT reactivation in adult heart disease patients.


Asunto(s)
Transición Epitelial-Mesenquimal , Pericardio , Humanos , Pericardio/metabolismo , Pericardio/citología , Animales , Cardiopatías/metabolismo , Cardiopatías/patología , Cardiopatías/terapia , Miocardio/metabolismo , Miocardio/patología , Diferenciación Celular
4.
Bioengineering (Basel) ; 11(7)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39061739

RESUMEN

Flexor tendon lacerations are primarily treated by surgical repair. Limited intrinsic healing ability means the repair site can remain weak. Furthermore, adhesion formation may reduce range of motion post-operatively. Mesenchymal stromal cells (MSCs) have been trialled for repair and regeneration of multiple musculoskeletal structures. Our goal was to determine the efficacy of MSCs in enhancing the biomechanical properties of surgically repaired flexor tendons. A PRISMA systematic review was conducted using four databases (PubMed, Ovid, Web of Science, and CINAHL) to identify studies using MSCs to augment surgical repair of flexor tendon injuries in animals compared to surgical repair alone. Nine studies were included, which investigated either bone marrow- or adipose-derived MSCs. Results of biomechanical testing were extracted and meta-analyses were performed regarding the maximum load, friction and properties relating to viscoelastic behaviour. There was no significant difference in maximum load at final follow-up. However, friction, a surrogate measure of adhesions, was significantly reduced following the application of MSCs (p = 0.04). Other properties showed variable results and dissipation of the therapeutic benefits of MSCs over time. In conclusion, MSCs reduce adhesion formation following tendon injury. This may result from their immunomodulatory function, dampening the inflammatory response. However, this may come at the cost of favourable healing which will restore the tendon's viscoelastic properties. The short duration of some improvements may reflect MSCs' limited survival or poor retention. Further investigation is needed to clarify the effect of MSC therapy and optimise its duration of action.

5.
J Bone Miner Res ; 39(8): 1045-1060, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-38836494

RESUMEN

Beyond the sensation of pain, peripheral nerves have been shown to play crucial roles in tissue regeneration and repair. As a highly innervated organ, bone can recover from injury without scar formation, making it an interesting model in which to study the role of nerves in tissue regeneration. As a comparison, tendon is a musculoskeletal tissue that is hypo-innervated, with repair often resulting in scar formation. Here, we reviewed the significance of innervation in 3 stages of injury repair (inflammatory, reparative, and remodeling) in 2 commonly injured musculoskeletal tissues: bone and tendon. Based on this focused review, we conclude that peripheral innervation is essential for phases of proper bone and tendon repair, and that nerves may dynamically regulate the repair process through interactions with the injury microenvironment via a variety of neuropeptides or neurotransmitters. A deeper understanding of neuronal regulation of musculoskeletal repair, and the crosstalk between nerves and the musculoskeletal system, will enable the development of future therapies for tissue healing.


Accumulating evidence has shown that, across organs systems, peripheral nerves regulate the process of tissue repair and regeneration. This is particularly relevant in the context of musculoskeletal injuries such as those affecting the bone and tendon. The question then arises: what is the function of peripheral innervation in the repair of bone and tendon injuries? This review offers an in-depth look at the ways in which nerves regulate the healing of bone and tendon injuries at various stages of recovery. A deeper comprehension of the influence of nerves on the repair of these tissues could pave the way for the development of future therapeutic strategies for tissue healing.


Asunto(s)
Huesos , Traumatismos de los Tendones , Cicatrización de Heridas , Humanos , Traumatismos de los Tendones/fisiopatología , Traumatismos de los Tendones/patología , Animales , Huesos/patología , Neuronas/metabolismo , Neuronas/patología , Tendones/patología , Tendones/fisiopatología
6.
Int J Biol Macromol ; 275(Pt 1): 133458, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38945326

RESUMEN

The development of novel therapeutic approaches to facilitate endometrial repair and regeneration while preventing adhesion recurrence is a crucial research objective aimed at enhancing clinical outcomes for women with intrauterine adhesions (IUA). In this study, we introduced an injectable Alg-GMA/PTSB zwitterionic hydrogel, characterized by excellent biocompatibility, anti-protein adsorption properties, and biodegradability. In a rat model, the hydrogel significantly promoted the regeneration and angiogenesis of damaged endometrial tissue, leading to improved recovery of epithelial cells, glands, proliferation, and vascularization. Furthermore, it exhibited the ability to suppress cellular apoptosis and collagen deposition, thereby mitigating fibrosis. Additionally, the hydrogel restored the expression of estrogen/progesterone receptors and endometrial receptivity markers, contributing to enhanced embryo implantation and fertility. These findings underscore the potential of the hydrogel as a promising therapeutic strategy for addressing endometrial injury, reducing fibrosis, restoring fertility, and ultimately improving outcomes for women with IUA.


Asunto(s)
Alginatos , Endometrio , Fertilidad , Hidrogeles , Femenino , Hidrogeles/química , Hidrogeles/farmacología , Animales , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Alginatos/química , Alginatos/farmacología , Ratas , Fertilidad/efectos de los fármacos , Adherencias Tisulares/prevención & control , Adherencias Tisulares/tratamiento farmacológico , Ratas Sprague-Dawley , Inyecciones , Regeneración/efectos de los fármacos , Humanos
7.
Int J Biol Macromol ; 274(Pt 2): 133495, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38944089

RESUMEN

Cartilage repair and regeneration have become a global issue that millions of patients from all over the world need surgical intervention to repair the articular cartilage annually due to the limited self-healing capability of the cartilage tissues. Cartilage tissue engineering has gained significant attention in cartilage repair and regeneration by integration of the chondrocytes (or stem cells) and the artificial scaffolds. Recently, polysaccharide-protein based scaffolds have demonstrated unique and promising mechanical and biological properties as the artificial extracellular matrix of natural cartilage. In this review, we summarize the modification methods for polysaccharides and proteins. The preparation strategies for the polysaccharide-protein based hydrogel scaffolds are presented. We discuss the mechanical, physical and biological properties of the polysaccharide-protein based scaffolds. Potential clinical translation and challenges on the artificial scaffolds are also discussed.


Asunto(s)
Cartílago Articular , Polisacáridos , Regeneración , Ingeniería de Tejidos , Andamios del Tejido , Andamios del Tejido/química , Humanos , Polisacáridos/química , Regeneración/efectos de los fármacos , Ingeniería de Tejidos/métodos , Animales , Cartílago Articular/fisiología , Proteínas/química , Hidrogeles/química , Condrocitos/citología , Condrocitos/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Cartílago/fisiología , Cicatrización de Heridas/efectos de los fármacos
8.
Cell Regen ; 13(1): 12, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38861103

RESUMEN

Macrophages play crucial and versatile roles in regulating tissue repair and regeneration upon injury. However, due to their complex compositional heterogeneity and functional plasticity, deciphering the nature of different macrophage subpopulations and unraveling their dynamics and precise roles during the repair process have been challenging. With its distinct advantages, zebrafish (Danio rerio) has emerged as an invaluable model for studying macrophage development and functions, especially in tissue repair and regeneration, providing valuable insights into our understanding of macrophage biology in health and diseases. In this review, we present the current knowledge and challenges associated with the role of macrophages in tissue repair and regeneration, highlighting the significant contributions made by zebrafish studies. We discuss the unique advantages of the zebrafish model, including its genetic tools, imaging techniques, and regenerative capacities, which have greatly facilitated the investigation of macrophages in these processes. Additionally, we outline the potential of zebrafish research in addressing the remaining challenges and advancing our understanding of the intricate interplay between macrophages and tissue repair and regeneration.

9.
Phytomedicine ; 130: 155553, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38820664

RESUMEN

INTRODUCTION: Non-healing wounds resulting from trauma, surgery, and chronic diseases annually affect millions of individuals globally, with limited therapeutic strategies available due to the incomplete understanding of the molecular processes governing tissue repair and regeneration. Salvianolic acid B (Sal B) has shown promising bioactivities in promoting angiogenesis and inhibiting inflammation. However, its regulatory mechanisms in tissue regeneration remain unclear. PURPOSE: This study aims to investigate the effects of Sal B on wound healing and regeneration processes, along with its underlying molecular mechanisms, by employing zebrafish as a model organism. METHODS: In this study, we employed a multifaceted approach to evaluate the impact of Sal B on zebrafish tail fin regeneration. We utilized whole-fish immunofluorescence, TUNEL staining, mitochondrial membrane potential (MMP), and Acridine Orange (AO) probes to analyze the tissue repair and regenerative under Sal B treatment. Additionally, we utilized transgenic zebrafish strains to investigate the migration of inflammatory cells during different phases of fin regeneration. To validate the importance of Caveolin-1 (Cav1) in tissue regeneration, we delved into its functional role using molecular docking and Morpholino-based gene knockdown techniques. Additionally, we quantified Cav1 expression levels through the application of in situ hybridization. RESULTS: Our findings demonstrated that Sal B expedites zebrafish tail fin regeneration through a multifaceted mechanism involving the promotion of cell proliferation, suppression of apoptosis, and enhancement of MMP. Furthermore, Sal B was found to exert regulatory control over the dynamic aggregation and subsequent regression of immune cells during tissue regenerative processes. Importantly, we observed that the knockdown of Cav1 significantly compromised tissue regeneration, leading to an excessive infiltration of immune cells and increased levels of apoptosis. Moreover, the knockdown of Cav1 also affects blastema formation, a critical process influenced by Cav1 in tissue regeneration. CONCLUSION: The results of this study showed that Sal B facilitated tissue repair and regeneration through regulating of immune cell migration and Cav1-mediated fibroblast activation, promoting blastema formation and development. This study highlighted the potential pharmacological effects of Sal B in promoting tissue regeneration. These findings contributed to the advancement of regenerative medicine research and the development of novel therapeutic approaches for trauma.


Asunto(s)
Benzofuranos , Caveolina 1 , Cicatrización de Heridas , Pez Cebra , Animales , Aletas de Animales/efectos de los fármacos , Aletas de Animales/fisiología , Animales Modificados Genéticamente , Apoptosis/efectos de los fármacos , Benzofuranos/farmacología , Caveolina 1/metabolismo , Movimiento Celular/efectos de los fármacos , Depsidos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Simulación del Acoplamiento Molecular , Regeneración/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Proteínas de Pez Cebra/metabolismo
10.
Int J Biol Macromol ; 268(Pt 1): 131643, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38643918

RESUMEN

The rational design of hydrogel materials to modulate the immune microenvironment has emerged as a pivotal approach in expediting tissue repair and regeneration. Within the immune microenvironment, an array of immune cells exists, with macrophages gaining prominence in the field of tissue repair and regeneration due to their roles in cytokine regulation to promote regeneration, maintain tissue homeostasis, and facilitate repair. Macrophages can be categorized into two types: classically activated M1 (pro-inflammatory) and alternatively activated M2 (anti-inflammatory and pro-repair). By regulating the physical and chemical properties of hydrogels, the phenotypic transformation and cell behavior of macrophages can be effectively controlled, thereby promoting tissue regeneration and repair. A full understanding of the interaction between hydrogels and macrophages can provide new ideas and methods for future tissue engineering and clinical treatment. Therefore, this paper reviews the effects of hydrogel components, hardness, pore size, and surface morphology on cell behaviors such as macrophage proliferation, migration, and phenotypic polarization, and explores the application of hydrogels based on macrophage immune regulation in skin, bone, cartilage, and nerve tissue repair. Finally, the challenges and future prospects of macrophage-based immunomodulatory hydrogels are discussed.


Asunto(s)
Hidrogeles , Macrófagos , Regeneración , Cicatrización de Heridas , Hidrogeles/química , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Humanos , Animales , Regeneración/inmunología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/inmunología , Ingeniería de Tejidos , Inmunomodulación/efectos de los fármacos
11.
J Ethnopharmacol ; 329: 118141, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38570149

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The active ingredients of traditional Chinese medicine (TCM), such as naringin (NG), Eucommiol, isopsoralen, icariin, Astragalus polysaccharides, and chondroitin sulfate, contained in Drynariae Rhizoma, Eucommiae Cortex, Psoralea corylifolia, Herba Epimedii, Astragalus radix and deer antler, are considered promising candidates for enhancing the healing of osteoporotic defects due to their outstanding bone homeostasis regulating properties. They are commonly used to activate bone repair scaffolds. AIM OF THE REVIEW: Bone repair scaffolds are inadequate to meet the demands of osteoporotic defect healing due to the lack of regulation of bone homeostasis. Therefore, selecting bone scaffolds activated with TCM to improve the therapeutic effect of repairing osteoporotic bone defects. MATERIALS AND METHODS: To gather information on bone scaffold activated by traditional Chinese medicine, we conducted a thorough search of several scientific databases, including Google Scholar, Web of Science, Scifinder, Baidu Scholar, PubMed, and China National Knowledge Infrastructure (CNKI). RESULTS: This review discusses the mechanism of TCM active ingredients in regulating bone homeostasis, including stimulating bone formation and inhibiting bone resorption process and the healing mechanism of traditional bone repair scaffolds activated by them for osteoporotic defect healing. CONCLUSION: In general, the introduction of TCM active ingredients provides a novel therapeutic approach for modulating bone homeostasis and facilitating osteoporotic defect healing, and also offers a new strategy for design of other unconventional bone defect healing materials.


Asunto(s)
Regeneración Ósea , Medicamentos Herbarios Chinos , Homeostasis , Medicina Tradicional China , Osteoporosis , Andamios del Tejido , Osteoporosis/tratamiento farmacológico , Regeneración Ósea/efectos de los fármacos , Animales , Humanos , Medicina Tradicional China/métodos , Homeostasis/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Huesos/efectos de los fármacos , Huesos/metabolismo
12.
ACS Nano ; 18(16): 10667-10687, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38592060

RESUMEN

Cartilage injuries are escalating worldwide, particularly in aging society. Given its limited self-healing ability, the repair and regeneration of damaged articular cartilage remain formidable challenges. To address this issue, nanomaterials are leveraged to achieve desirable repair outcomes by enhancing mechanical properties, optimizing drug loading and bioavailability, enabling site-specific and targeted delivery, and orchestrating cell activities at the nanoscale. This review presents a comprehensive survey of recent research in nanomedicine for cartilage repair, with a primary focus on biomaterial design considerations and recent advances. The review commences with an introductory overview of the intricate cartilage microenvironment and further delves into key biomaterial design parameters crucial for treating cartilage damage, including microstructure, surface charge, and active targeting. The focal point of this review lies in recent advances in nano drug delivery systems and nanotechnology-enabled 3D matrices for cartilage repair. We discuss the compositions and properties of these nanomaterials and elucidate how these materials impact the regeneration of damaged cartilage. This review underscores the pivotal role of nanotechnology in improving the efficacy of biomaterials utilized for the treatment of cartilage damage.


Asunto(s)
Materiales Biocompatibles , Cartílago Articular , Nanomedicina , Humanos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Nanomedicina/métodos , Cartílago Articular/efectos de los fármacos , Animales , Sistemas de Liberación de Medicamentos , Ingeniería de Tejidos , Regeneración/efectos de los fármacos
13.
Mater Today Bio ; 25: 100978, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38434571

RESUMEN

A life-threatening illness that poses a serious threat to human health is myocardial infarction. It may result in a significant number of myocardial cells dying, dilated left ventricles, dysfunctional heart function, and ultimately cardiac failure. Based on the development of emerging biomaterials and the lack of clinical treatment methods and cardiac donors for myocardial infarction, hydrogels with good compatibility have been gradually applied to the treatment of myocardial infarction. Specifically, based on the three processes of pathophysiology of myocardial infarction, we summarized various types of hydrogels designed for myocardial tissue engineering in recent years, including natural hydrogels, intelligent hydrogels, growth factors, stem cells, and microRNA-loaded hydrogels. In addition, we also describe the heart patch and preparation techniques that promote the repair of MI heart function. Although most of these hydrogels are still in the preclinical research stage and lack of clinical trials, they have great potential for further application in the future. It is expected that this review will improve our knowledge of and offer fresh approaches to treating myocardial infarction.

14.
Regen Ther ; 25: 355-366, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38374989

RESUMEN

As a prominent research area in tissue repair and regeneration, mesenchymal stem cells (MSCs) have garnered substantial attention for their potential in the treatment of various diseases. It is now widely recognized that the therapeutic effects of MSCs primarily occur through paracrine mechanisms. Among these mechanisms, exosomes play a crucial role by exerting a series of regulatory effects on surrounding cells and tissues. While exosomes have shown promise in treating various diseases, they do have some limitations, such as limited secretion, poor targeting, and single functionality. However, MSC preconditioning can enhance the production of exosomes, lead to more stable functionality and improve therapeutic effects. Moreover, exosomes could also serve as carriers for specific drugs or genes, enabling more precise treatments of diseases. This review summarizes the most recent literatures on how preconditioning of MSCs influences the regenerative potential of their exosomes in tissue repair and provides new insights into the therapeutic application of exosomes derived from MSCs.

15.
Cell Regen ; 13(1): 4, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38349608

RESUMEN

The Golgi Apparatus (GA) is pivotal in vesicle sorting and protein modifications within cells. Traditionally, the GA has been described as a perinuclear organelle consisting of stacked cisternae forming a ribbon-like structure. Changes in the stacked structure or the canonical perinuclear localization of the GA have been referred to as "GA fragmentation", a term widely employed in the literature to describe changes in GA morphology and distribution. However, the precise meaning and function of GA fragmentation remain intricate. This review aims to demystify this enigmatic phenomenon, dissecting the diverse morphological changes observed and their potential contributions to cellular wound repair and regeneration. Through a comprehensive analysis of current research, we hope to pave the way for future advancements in GA research and their important role in physiological and pathological conditions.

16.
Biomater Adv ; 157: 213740, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38183690

RESUMEN

The fibrocartilaginous enthesis is a highly specialised tissue interface that ensures a smooth mechanical transfer between tendon or ligament and bone through a fibrocartilage area. This tissue is prone to injury and often does not heal, even after surgical intervention. Enthesis augmentation approaches are challenging due to the complexity of the tissue that is characterised by the coexistence of a range of cellular and extracellular components, architectural features and mechanical properties within only hundreds of micrometres. Herein, we discuss enthesis repair and regeneration strategies, with particular focus on elegant interfacial and functionalised scaffold-based designs.


Asunto(s)
Huesos , Tendones , Tendones/cirugía , Huesos/cirugía , Fibrocartílago/lesiones , Ligamentos
17.
Prog Biophys Mol Biol ; 187: 36-50, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38280492

RESUMEN

Tissue repair and regeneration is a vital biological process in organisms, which is influenced by various internal mechanisms and microenvironments. Pulsed electromagnetic fields (PEMFs) are becoming a potential medical technology due to its advantages of effectiveness and non-invasiveness. Numerous studies have demonstrated that PEMFs can stimulate stem cell proliferation and differentiation, regulate inflammatory reactions, accelerate wound healing, which is of great significance for tissue regeneration and repair, providing a solid basis for enlarging its clinical application. However, some important issues such as optimal parameter system and potential deep mechanisms remain to be resolved due to PEMFs window effect and biological complexity. Thus, it is of great importance to comprehensively summarizing and analyzing the literature related to the biological effects of PEMFs in tissue regeneration and repair. This review expounded the biological effects of PEMFs on stem cells, inflammation response, wound healing and musculoskeletal disorders in order to improve the application value of PEMFs in medicine. It is believed that with the continuous exploration of biological effects of PEMFs, it will be applied increasingly widely to tissue repair and other diseases.


Asunto(s)
Campos Electromagnéticos , Células Madre Mesenquimatosas , Diferenciación Celular , Cicatrización de Heridas , Células Madre
18.
Adv Healthc Mater ; 13(5): e2302664, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37902817

RESUMEN

Inorganic nanoparticulate biomaterials, such as calcium phosphate and bioglass particles, with chemical compositions similar to that of the inorganic component of natural bone, and hence having excellent biocompatibility and bioactivity, are widely used for the fabrication of synthetic bone graft substitutes. Growing evidence suggests that structurally anisotropic, or 1D inorganic micro-/nanobiomaterials are superior to inorganic nanoparticulate biomaterials in the context of mechanical reinforcement and construction of self-supporting 3D network structures. Therefore, in the past decades, efforts have been devoted to developing advanced synthetic scaffolds for bone regeneration using 1D micro-/nanobiomaterials as building blocks. These scaffolds feature extraordinary physical and biological properties, such as enhanced mechanical properties, super elasticity, multiscale hierarchical architecture, extracellular matrix-like fibrous microstructure, and desirable biocompatibility and bioactivity, etc. In this review, an overview of recent progress in the development of advanced scaffolds for bone regeneration is provided based on 1D inorganic micro-/nanobiomaterials with a focus on their structural design, mechanical properties, and bioactivity. The promising perspectives for future research directions are also highlighted.


Asunto(s)
Sustitutos de Huesos , Nanoestructuras , Andamios del Tejido/química , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/química , Regeneración Ósea , Sustitutos de Huesos/farmacología , Sustitutos de Huesos/química
19.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1021718

RESUMEN

BACKGROUND:In the construction of guided bone regeneration membrane with biological function,a single material cannot meet the clinical needs due to its insufficient function,so the composite of multiple materials has become a trend of tissue repair engineering. OBJECTIVE:To prepare silk fibroin/bioactive glass composite fiber membranes by electrospinning technology,and to characterize the physicochemical properties and biocompatibility in vitro. METHODS:The solution of electrospinning was prepared by dissolving 0.8 g silk fibroin protein in 10 mL hexafluoro-isopropanol alcohol,and the nanofiber membrane of silk fibroin protein was prepared by electrospinning technology(denoted as SF fiber membrane).0.1,0.3,0.5,and 0.8 g of bioactive glass were added to the electrospinning solution,and the silk fibroin/bioactive glass composite fiber membrane was prepared by electrospinning technology(recorded as SF/1BG,SF/3BG,SF/5BG,and SF/8BG fiber membrane in turn).The physicochemical properties and biocompatibility of five groups of fiber membranes were characterized. RESULTS AND CONCLUSION:(1)The scanning electron microscopy results showed that nanofibers of the prepared composite membrane were smooth,continuous and uniform and had no beaded structure.There was no obvious adhesion between the silk fibers,and they all showed random arrangement of disordered porous structures.The fiber diameter of the fiber membrane decreased after the addition of bioactive glass.Fourier infrared spectroscopy and X-ray diffraction detection results showed that the chemical structure of silk fibroin protein and bioactive glass in fiber membrane was stable.The water contact angles of SF,SF/1BG,SF/3BG,SF/5BG,and SF/8BG were 105.02°,72.58°,78.13°,79.35°,and 72.50°,respectively.(2)Bone marrow mesenchymal stem cells were inoculated on five groups of fiber membranes.CCK-8 assay results showed that SF/1BG,SF/3BG,and SF/5BG fiber membranes could promote the proliferation of bone marrow mesenchymal stem cells compared with SF and SF/8BG.Live cell/dead cell staining showed that the cell vitality on the surface of the five groups of fiber membranes was better,and the number and distribution of cells on the surface of SF/5BG fiber membrane were more uniform.Rhodamine phalloidin staining and scanning electron microscopy exhibited that compared with SF fiber membrane,the SF/5BG fiber membrane was more favorable to the adhesion of bone marrow mesenchymal stem cells.Bone marrow mesenchymal stem cells were inoculated on the fiber membrane of the five groups for osteogenic induction differentiation,and the alkaline phosphatase activity of the SF/3BG and SF/5BG groups was higher than that of the other three groups(P<0.05,P<0.01,P<0.001).Alizarin red staining showed that the formation of calcium nodules in fiber membrane increased after the addition of bioactive glass,and the formation of calcium nodules in the SF/5BG group was the most.(3)The results show that silk fibroin/bioactive glass composite fiber membrane has good biosafety and biocompatibility.

20.
Cureus ; 15(11): e49437, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38149156

RESUMEN

The relentless march of technological progress entails constant evolution and adaptation. A concerted effort is underway in medical research to unravel various diseases' cellular and molecular underpinnings. The traditional approaches to disease treatment often fall short of delivering entirely satisfactory outcomes, which has prompted a shifting spotlight on gene therapy as a versatile solution for many inherited and acquired disorders. Genes, intricate sequences of genetic code, are the complicated blueprints dictating the production of essential proteins within the human body. Remarkably, each individual's genetic makeup is uniquely distinct, with variations in these genetic sequences serving as the bedrock of our diversity. Gene therapy represents an innovative medical strategy that harnesses the power of genes themselves to function as therapeutic agents. It serves as a conduit through which defective genes are either substituted or mended with the introduction of remedial genetic material. This groundbreaking method can tackle various illnesses, from conditions originating from single-gene abnormalities to intricate disorders influenced by multiple genes. In dentistry and periodontics, gene therapy finds a promising array of applications. It contributes significantly to managing salivary gland disorders, autoimmune diseases, and the regeneration of damaged bone tissue, as well as addressing cancerous and precancerous conditions. Moreover, the possibilities extend into DNA vaccination and broader areas of oral health. The advent of gene therapy in dentistry represents a new era of significant progress, offering substantial advancements in the management of periodontal disease and the reconstruction of the dental alveolar apparatus. The aim of this narrative review is to provide a comprehensive overview of the landscape of gene therapy investigations in these disciplines, shedding light on its potential implications for oral health and treatment. With its potential to rectify the genetic underpinnings of various conditions, gene therapy offers a novel frontier in healthcare that continually shapes the landscape of medicine and holds the promise of more effective and personalised treatments.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA