Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Small ; : e2305940, 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-37803920

RESUMEN

Nanomedicine technologies are being developed for the prevention, diagnosis, and treatment of cardiovascular disease (CVD), which is the leading cause of death worldwide. Before delving into the nuances of cardiac nanomedicine, it is essential to comprehend the fundamental sex-specific differences in cardiovascular health. Traditionally, CVDs have been more prevalent in males, but it is increasingly evident that females also face significant risks, albeit with distinct characteristics. Females tend to develop CVDs at a later age, exhibit different clinical symptoms, and often experience worse outcomes compared to males. These differences indicate the need for sex-specific approaches in cardiac nanomedicine. This Perspective discusses the importance of considering sex in the safety and therapeutic efficacy of nanomedicine approaches for the prevention, diagnosis, and treatment of CVD.

2.
Small ; 19(36): e2301838, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37119440

RESUMEN

The protein corona forms spontaneously on nanoparticle surfaces when nanomaterials are introduced into any biological system/fluid. Reliable characterization of the protein corona is, therefore, a vital step in the development of safe and efficient diagnostic and therapeutic nanomedicine products. 2134 published manuscripts on the protein corona are reviewed and a down-selection of 470 papers spanning 2000-2021, comprising 1702 nanoparticle (NP) systems is analyzed. This analysis reveals: i) most corona studies have been conducted on metal and metal oxide nanoparticles; ii) despite their overwhelming presence in clinical practice, lipid-based NPs are underrepresented in protein corona research, iii) studies use new methods to improve reliability and reproducibility in protein corona research; iv) studies use more specific protein sources toward personalized medicine; and v) careful characterization of nanoparticles after corona formation is imperative to minimize the role of aggregation and protein contamination on corona outcomes. As nanoparticles used in biomedicine become increasingly prevalent and biochemically complex, the field of protein corona research will need to focus on developing analytical approaches and characterization techniques appropriate for each unique nanoparticle formulation. Achieving such characterization of the nano-bio interface of nanobiotechnologies will enable more seamless development and safe implementation of nanoparticles in medicine.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Corona de Proteínas , Corona de Proteínas/química , Reproducibilidad de los Resultados , Proteínas/química , Nanomedicina , Nanopartículas/química
3.
Circulation ; 144(3): 210-228, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-33951921

RESUMEN

BACKGROUND: Human induced pluripotent stem cells with normal (wild-type) or upregulated (overexpressed) levels of CCND2 (cyclin D2) expression were differentiated into cardiomyocytes (CCND2WTCMs or CCND2OECMs, respectively) and injected into infarcted pig hearts. METHODS: Acute myocardial infarction was induced by a 60-minute occlusion of the left anterior descending coronary artery. Immediately after reperfusion, CCND2WTCMs or CCND2OECMs (3×107 cells each) or an equivalent volume of the delivery vehicle was injected around the infarct border zone area. RESULTS: The number of the engrafted CCND2OECMs exceeded that of the engrafted CCND2WTCMs from 6- to 8-fold, rising from 1 week to 4 weeks after implantation. In contrast to the treatment with the CCND2WTCMs or the delivery vehicle, the administration of CCND2OECM was associated with significantly improved left ventricular function, as revealed by magnetic resonance imaging. This correlated with reduction of infarct size, fibrosis, ventricular hypertrophy, and cardiomyocyte apoptosis, and increase of vascular density and arterial density, as per histologic analysis of the treated hearts. Expression of cell proliferation markers (eg, Ki67, phosphorylated histone 3, and Aurora B kinase) was also significantly upregulated in the recipient cardiomyocytes from the CCND2OECM-treated than from the CCND2WTCM-treated pigs. The cell proliferation rate and the hypoxia tolerance measured in cultured human induced pluripotent stem cell cardiomyocytes were significantly greater after treatment with exosomes isolated from the CCND2OECMs (CCND2OEExos) than from the CCND2WTCMs (CCND2WTExos). As demonstrated by our study, CCND2OEExos can also promote the proliferation activity of postnatal rat and adult mouse cardiomyocytes. A bulk miRNA sequencing analysis of CCND2OEExos versus CCND2WTExos identified 206 and 91 miRNAs that were significantly upregulated and downregulated, respectively. Gene ontology enrichment analysis identified significant differences in the expression profiles of miRNAs from various functional categories and pathways, including miRNAs implicated in cell-cycle checkpoints (G2/M and G1/S transitions), or the mechanism of cytokinesis. CONCLUSIONS: We demonstrated that enhanced potency of CCND2OECMs promoted myocyte proliferation in both grafts and recipient tissue in a large mammal acute myocardial infarction model. These results suggest that CCND2OECMs transplantation may be a potential therapeutic strategy for the repair of infarcted hearts.


Asunto(s)
Diferenciación Celular/genética , Ciclina D2/genética , Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Trasplante de Células Madre , Animales , Biomarcadores , Técnicas de Cultivo de Célula , Proliferación Celular , Separación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/etiología , Miocitos Cardíacos/citología , Neovascularización Fisiológica/genética , Recuperación de la Función , Porcinos , Resultado del Tratamiento
4.
Immunology ; 166(4): 429-443, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35470422

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic has created unprecedented challenges worldwide. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19 and has a complex interaction with the immune system, including growing evidence of sex-specific differences in the immune response. Sex-disaggregated analyses of epidemiological data indicate that males experience more severe symptoms and suffer higher mortality from COVID-19 than females. Many behavioural risk factors and biological factors may contribute to the different immune response. This review examines the immune response to SARS-CoV-2 infection in the context of sex, with emphasis on potential biological mechanisms explaining differences in clinical outcomes. Understanding sex differences in the pathophysiology of SARS-CoV-2 infection will help promote the development of specific strategies to manage the disease.


Asunto(s)
COVID-19 , Femenino , Humanos , Inmunidad , Masculino , Pandemias , Factores de Riesgo , SARS-CoV-2 , Factores Sexuales
5.
Mol Pharm ; 18(8): 3171-3180, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34279974

RESUMEN

Current treatment of chronic wounds has been critically limited by various factors, including bacterial infection, biofilm formation, impaired angiogenesis, and prolonged inflammation. Addressing these challenges, we developed a multifunctional wound dressing-based three-pronged approach for accelerating wound healing. The multifunctional wound dressing, composed of nanofibers, functional nanoparticles, natural biopolymers, and selected protein and peptide, can target multiple endogenous repair mechanisms and represents a promising alternative to current wound healing products.


Asunto(s)
Anexina A1/administración & dosificación , Antiinflamatorios/administración & dosificación , Vendajes , Diabetes Mellitus Experimental/complicaciones , Proteínas Relacionadas con la Folistatina/administración & dosificación , Péptidos/administración & dosificación , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Herida Quirúrgica/complicaciones , Herida Quirúrgica/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Infección de Heridas/complicaciones , Infección de Heridas/tratamiento farmacológico , Células 3T3 , Animales , Materiales Biocompatibles/administración & dosificación , Biopolímeros/química , Supervivencia Celular/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Células HaCaT , Humanos , Nanopartículas Magnéticas de Óxido de Hierro/química , Masculino , Ensayo de Materiales/métodos , Ratones , Nanofibras/química , Ratas , Ratas Wistar , Infecciones Estafilocócicas/microbiología , Resultado del Tratamiento , Infección de Heridas/microbiología
6.
Chem Rev ; 119(21): 11352-11390, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31490059

RESUMEN

The adult myocardium has a limited regenerative capacity following heart injury, and the lost cells are primarily replaced by fibrotic scar tissue. Suboptimal efficiency of current clinical therapies to resurrect the infarcted heart results in injured heart enlargement and remodeling to maintain its physiological functions. These remodeling processes ultimately leads to ischemic cardiomyopathy and heart failure (HF). Recent therapeutic approaches (e.g., regenerative and nanomedicine) have shown promise to prevent HF postmyocardial infarction in animal models. However, these preclinical, clinical, and technological advancements have yet to yield substantial enhancements in the survival rate and quality of life of patients with severe ischemic injuries. This could be attributed largely to the considerable gap in knowledge between clinicians and nanobioengineers. Development of highly effective cardiac regenerative therapies requires connecting and coordinating multiple fields, including cardiology, cellular and molecular biology, biochemistry and chemistry, and mechanical and materials sciences, among others. This review is particularly intended to bridge the knowledge gap between cardiologists and regenerative nanomedicine experts. Establishing this multidisciplinary knowledge base may help pave the way for developing novel, safer, and more effective approaches that will enable the medical community to reduce morbidity and mortality in HF patients.


Asunto(s)
Insuficiencia Cardíaca/terapia , Nanomedicina/métodos , Medicina Regenerativa/métodos , Animales , Insuficiencia Cardíaca/prevención & control , Humanos
7.
Nature ; 525(7570): 479-85, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26375005

RESUMEN

The elucidation of factors that activate the regeneration of the adult mammalian heart is of major scientific and therapeutic importance. Here we found that epicardial cells contain a potent cardiogenic activity identified as follistatin-like 1 (Fstl1). Epicardial Fstl1 declines following myocardial infarction and is replaced by myocardial expression. Myocardial Fstl1 does not promote regeneration, either basally or upon transgenic overexpression. Application of the human Fstl1 protein (FSTL1) via an epicardial patch stimulates cell cycle entry and division of pre-existing cardiomyocytes, improving cardiac function and survival in mouse and swine models of myocardial infarction. The data suggest that the loss of epicardial FSTL1 is a maladaptive response to injury, and that its restoration would be an effective way to reverse myocardial death and remodelling following myocardial infarction in humans.


Asunto(s)
Proteínas Relacionadas con la Folistatina/metabolismo , Miocardio/metabolismo , Pericardio/crecimiento & desarrollo , Pericardio/metabolismo , Regeneración , Animales , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Proteínas Relacionadas con la Folistatina/genética , Humanos , Masculino , Ratones , Mioblastos Cardíacos/citología , Mioblastos Cardíacos/efectos de los fármacos , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Pericardio/citología , Pericardio/efectos de los fármacos , Ratas , Regeneración/efectos de los fármacos , Transducción de Señal , Porcinos , Transgenes/genética
8.
Curr Cardiol Rep ; 21(9): 105, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31367922

RESUMEN

PURPOSE OF REVIEW: Tissue engineering has expanded into a highly versatile manufacturing landscape that holds great promise for advancing cardiovascular regenerative medicine. In this review, we provide a summary of the current state-of-the-art bioengineering technologies used to create functional cardiac tissues for a variety of applications in vitro and in vivo. RECENT FINDINGS: Studies over the past few years have made a strong case that tissue engineering is one of the major driving forces behind the accelerating fields of patient-specific regenerative medicine, precision medicine, compound screening, and disease modeling. To date, a variety of approaches have been used to bioengineer functional cardiac constructs, including biomaterial-based, cell-based, and hybrid (using cells and biomaterials) approaches. While some major progress has been made using cellular approaches, with multiple ongoing clinical trials, cell-free cardiac tissue engineering approaches have also accomplished multiple breakthroughs, although drawbacks remain. This review summarizes the most promising methods that have been employed to generate cardiovascular tissue constructs for basic science or clinical applications. Further, we outline the strengths and challenges that are inherent to this field as a whole and for each highlighted technology.


Asunto(s)
Corazón/fisiología , Miocardio/citología , Ingeniería de Tejidos/métodos , Materiales Biocompatibles/administración & dosificación , Bioimpresión , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Humanos , Miocitos Cardíacos/fisiología , Impresión Tridimensional , Medicina Regenerativa/métodos , Medicina Regenerativa/tendencias , Ingeniería de Tejidos/tendencias , Andamios del Tejido , Investigación Biomédica Traslacional
9.
Chem Soc Rev ; 46(14): 4218-4244, 2017 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-28585944

RESUMEN

Nanoscale materials are increasingly found in consumer goods, electronics, and pharmaceuticals. While these particles interact with the body in myriad ways, their beneficial and/or deleterious effects ultimately arise from interactions at the cellular and subcellular level. Nanoparticles (NPs) can modulate cell fate, induce or prevent mutations, initiate cell-cell communication, and modulate cell structure in a manner dictated largely by phenomena at the nano-bio interface. Recent advances in chemical synthesis have yielded new nanoscale materials with precisely defined biochemical features, and emerging analytical techniques have shed light on nuanced and context-dependent nano-bio interactions within cells. In this review, we provide an objective and comprehensive account of our current understanding of the cellular uptake of NPs and the underlying parameters controlling the nano-cellular interactions, along with the available analytical techniques to follow and track these processes.


Asunto(s)
Nanopartículas/metabolismo , Fenómenos Bioquímicos , Transporte Biológico , Comunicación Celular , Humanos , Nanopartículas/química
10.
Adv Funct Mater ; 25(28): 4379-4389, 2015 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-27516731

RESUMEN

Biomaterials are extensively used to restore damaged tissues, in the forms of implants (e.g. tissue engineered scaffolds) or biomedical devices (e.g. pacemakers). Once in contact with the physiological environment, nanostructured biomaterials undergo modifications as a result of endogenous proteins binding to their surface. The formation of this macromolecular coating complex, known as 'protein corona', onto the surface of nanoparticles and its effect on cell-particle interactions are currently under intense investigation. In striking contrast, protein corona constructs within nanostructured porous tissue engineering scaffolds remain poorly characterized. As organismal systems are highly dynamic, it is conceivable that the formation of distinct protein corona on implanted scaffolds might itself modulate cell-extracellular matrix interactions. Here, we report that corona complexes formed onto the fibrils of engineered collagen scaffolds display specific, distinct, and reproducible compositions that are a signature of the tissue microenvironment as well as being indicative of the subject's health condition. Protein corona formed on collagen matrices modulated cellular secretome in a context-specific manner ex-vivo, demonstrating their role in regulating scaffold-cellular interactions. Together, these findings underscore the importance of custom-designing personalized nanostructured biomaterials, according to the biological milieu and disease state. We propose the use of protein corona as in situ biosensor of temporal and local biomarkers.

11.
Nanotechnology ; 26(6): 060501, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25611345

RESUMEN

Cell-based therapies are a recently established path for treating a wide range of human disease. Tissue engineering of contractile heart muscle for replacement therapy is among the most exciting and important of these efforts. However, current in vitro techniques of cultivating functional mature cardiac grafts have only been moderately successful due to the poor capability of traditional two-dimensional cell culture systems to recapitulate necessary in vivo conditions. In this issue, Kiefer et al introduce a laser-patterned nanostructured substrate (Al/Al2O3 nanowires) for efficient maintenance of oriented human cardiomyocytes, with great potential to open new roads to mass-production of contractile myocardial grafts for cardiovascular tissue engineering.


Asunto(s)
Miocitos Cardíacos/fisiología , Nanocables/uso terapéutico , Ingeniería de Tejidos/métodos , Humanos
14.
Acta Biomater ; 173: 109-122, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37925122

RESUMEN

Myocardial infarction (MI) is accompanied by the formation of a fibrotic scar in the left ventricle (LV) and initiates significant alterations in the architecture and constituents of the LV free wall (LVFW). Previous studies have shown that LV adaptation is highly individual, indicating that the identification of remodeling mechanisms post-MI demands a fully subject-specific approach that can integrate a host of structural alterations at the fiber-level to changes in bulk biomechanical adaptation at the tissue-level. We present an image-driven micromechanical approach to characterize remodeling, assimilating new biaxial mechanical data, histological studies, and digital image correlation data within an in-silico framework to elucidate the fiber-level remodeling mechanisms that drive tissue-level adaptation for each subject. We found that a progressively diffused collagen fiber structure combined with similarly disorganized myofiber architecture in the healthy region leads to the loss of LVFW anisotropy post-MI, offering an important tissue-level hallmark for LV maladaptation. In contrast, our results suggest that reductions in collagen undulation are an adaptive mechanism competing against LVFW thinning. Additionally, we show that the inclusion of subject-specific geometry when modeling myocardial tissue is essential for accurate prediction of tissue kinematics. Our approach serves as an essential step toward identifying fiber-level remodeling indices that govern the transition of MI to systolic heart failure. These indices complement the traditional, organ-level measures of LV anatomy and function that often fall short of early prognostication of heart failure in MI. In addition, our approach offers an integrated methodology to advance the design of personalized interventions, such as hydrogel injection, to reinforce and suppress native adaptive and maladaptive mechanisms, respectively, to prevent the transition of MI to heart failure. STATEMENT OF SIGNIFICANCE: Biomechanical and architectural adaptation of the LVFW remains a central, yet overlooked, remodeling process post-MI. Our study indicates the biomechanical adaptation of the LVFW post-MI is highly individual and driven by altered fiber network architecture and collective changes in collagen fiber content, undulation, and stiffness. Our findings demonstrate the possibility of using cardiac strains to infer such fiber-level remodeling events through in-silico modeling, paving the way for in-vivo characterization of multiscale biomechanical indices in humans. Such indices will complement the traditional, organ-level measures of LV anatomy and function that often fall short of early prognostication of heart failure in MI.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Humanos , Remodelación Ventricular , Miocardio/patología , Infarto del Miocardio/patología , Insuficiencia Cardíaca/patología , Colágeno
15.
Front Cardiovasc Med ; 11: 1432784, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39026997

RESUMEN

Introduction: Primary pulmonary vein stenosis (PVS) is a rare congenital heart disease that proves to be a clinical challenge due to the rapidly progressive disease course and high rates of treatment complications. PVS intervention is frequently faced with in-stent restenosis and persistent disease progression despite initial venous recanalization with balloon angioplasty or stenting. Alterations in wall shear stress (WSS) have been previously associated with neointimal hyperplasia and venous stenosis underlying PVS progression. Thus, the development of patient-specific three-dimensional (3D) in vitro models is needed to further investigate the biomechanical outcomes of endovascular and surgical interventions. Methods: In this study, deidentified computed tomography images from three patients were segmented to generate perfusable phantom models of pulmonary veins before and after catheterization. These 3D reconstructions were 3D printed using a clear resin ink and used in a benchtop experimental setup. Computational fluid dynamic (CFD) analysis was performed on models in silico utilizing Doppler echocardiography data to represent the in vivo flow conditions at the inlets. Particle image velocimetry was conducted using the benchtop perfusion setup to analyze WSS and velocity profiles and the results were compared with those predicted by the CFD model. Results: Our findings indicated areas of undesirable alterations in WSS before and after catheterization, in comparison with the published baseline levels in the healthy in vivo tissues that may lead to regional disease progression. Discussion: The established patient-specific 3D in vitro models and the developed in vitro-in silico platform demonstrate great promise to refine interventional approaches and mitigate complications in treating patients with primary PVS.

16.
Adv Sci (Weinh) ; 11(26): e2400476, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38696618

RESUMEN

Vascular cell overgrowth and lumen size reduction in pulmonary vein stenosis (PVS) can result in elevated PV pressure, pulmonary hypertension, cardiac failure, and death. Administration of chemotherapies such as rapamycin have shown promise by inhibiting the vascular cell proliferation; yet clinical success is limited due to complications such as restenosis and off-target effects. The lack of in vitro models to recapitulate the complex pathophysiology of PVS has hindered the identification of disease mechanisms and therapies. This study integrated 3D bioprinting, functional nanoparticles, and perfusion bioreactors to develop a novel in vitro model of PVS. Bioprinted bifurcated PV constructs are seeded with endothelial cells (ECs) and perfused, demonstrating the formation of a uniform and viable endothelium. Computational modeling identified the bifurcation point at high risk of EC overgrowth. Application of an external magnetic field enabled targeting of the rapamycin-loaded superparamagnetic iron oxide nanoparticles at the bifurcation site, leading to a significant reduction in EC proliferation with no adverse side effects. These results establish a 3D bioprinted in vitro model to study PV homeostasis and diseases, offering the potential for increased throughput, tunability, and patient specificity, to test new or more effective therapies for PVS and other vascular diseases.


Asunto(s)
Bioimpresión , Impresión Tridimensional , Venas Pulmonares , Sirolimus , Sirolimus/farmacología , Sirolimus/administración & dosificación , Bioimpresión/métodos , Humanos , Constricción Patológica , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Nanopartículas de Magnetita , Técnicas In Vitro , Sistemas de Liberación de Medicamentos/métodos , Proliferación Celular/efectos de los fármacos
17.
Adv Healthc Mater ; : e2401603, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38815975

RESUMEN

The ability to promote three-dimensional (3D) self-organization of induced pluripotent stem cells into complex tissue structures called organoids presents new opportunities for the field of developmental biology. Brain organoids have been used to investigate principles of neurodevelopment and neuropsychiatric disorders and serve as a drug screening and discovery platform. However, brain organoid cultures are currently limited by a lacking ability to precisely control their extracellular environment. Here, this work employs 3D bioprinting to generate a high-throughput, tunable, and reproducible scaffold for controlling organoid development and patterning. Additionally, this approach supports the coculture of organoids and vascular cells in a custom architecture containing interconnected endothelialized channels. Printing fidelity and mechanical assessments confirm that fabricated scaffolds closely match intended design features and exhibit stiffness values reflective of the developing human brain. Using organoid growth, viability, cytoarchitecture, proliferation, and transcriptomic benchmarks, this work finds that organoids cultured within the bioprinted scaffold long-term are healthy and have expected neuroectodermal differentiation. Lastly, this work confirms that the endothelial cells (ECs) in printed channel structures can migrate toward and infiltrate into the embedded organoids. This work demonstrates a tunable 3D culturing platform that can be used to create more complex and accurate models of human brain development and underlying diseases.

18.
Acta Biomater ; 162: 240-253, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36963596

RESUMEN

The myocardium possesses an intricately designed microarchitecture to produce an optimal cardiac contraction. The contractile behavior of the heart is generated at the sarcomere level and travels across several length scales to manifest as the systolic function at the organ level. While passive myocardial behavior has been studied extensively, the translation of active tension produced at the fiber level to the organ-level function is not well understood. Alterations in cardiac systolic function are often key sequelae in structural heart diseases, such as myocardial infarction and systolic heart failure; thus, characterization of the contractile behavior of the heart across multiple length scales is essential to improve our understanding of mechanisms collectively leading to depressed systolic function. In this study, we present a methodology to characterize the active behavior of left ventricle free wall (LVFW) myocardial tissues in mice. Combined with active tests in papillary muscle fibers and conventional in vivo contractility measurement at the organ level in an animal-specific manner, we establish a multiscale active characterization of the heart from fiber to organ. In addition, we quantified myocardial architecture from histology to shed light on the directionality of the contractility at the tissue level. The LVFW tissue activation-relaxation behavior under isometric conditions was qualitatively similar to that of the papillary muscle fiber bundle. However, the maximum stress developed in the LVFW tissue was an order of magnitude lower than that developed by a fiber bundle, and the time taken for active forces to plateau was 2-3 orders of magnitude longer. Although the LVFW tissue exhibited a slightly stiffer passive response in the circumferential direction, the tissues produced significantly larger active stresses in the longitudinal direction during active testing. Also, contrary to passive viscoelastic stress relaxation, active stresses relaxed faster in the direction with larger peak stresses. The multiscale experimental pipeline presented in this work is expected to provide crucial insight into the contractile adaptation mechanisms of the heart with impaired systolic function. STATEMENT OF SIGNIFICANCE: Heart failure cause significant alterations to the contractile-relaxation behavior of the yocardium. Multiscale characterization of the contractile behavior of the myocardium is essential to advance our understanding of how contractility translates from fiber to organ and to identify the multiscale mechanisms leading to impaired cardiac function. While passive myocardial behavior has been studied extensively, the investigation of tissue-level contractile behavior remains critically scarce in the literature. To the best of our knowledge, our study here is the first to investigate the contractile behavior of the left ventricle at multiple length scales in small animals. Our results indicate that the active myocardial wall is a function of transmural depth and relaxes faster in the direction with larger peak stresses.


Asunto(s)
Ventrículos Cardíacos , Corazón , Ratones , Animales , Corazón/fisiología , Miocardio , Contracción Miocárdica , Sístole
19.
Adv Biol (Weinh) ; 7(7): e2300124, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37132122

RESUMEN

Adhesive tissue engineering scaffolds (ATESs) have emerged as an innovative alternative means, replacing sutures and bioglues, to secure the implants onto target tissues. Relying on their intrinsic tissue adhesion characteristics, ATES systems enable minimally invasive delivery of various scaffolds. This study investigates development of the first class of 3D bioprinted ATES constructs using functionalized hydrogel bioinks. Two ATES delivery strategies, in situ printing onto the adherend versus printing and then transferring to the target surface, are tested using two bioprinting methods, embedded versus air printing. Dopamine-modified methacrylated hyaluronic acid (HAMA-Dopa) and gelatin methacrylate (GelMA) are used as the main bioink components, enabling fabrication of scaffolds with enhanced adhesion and crosslinking properties. Results demonstrate that dopamine modification improved adhesive properties of the HAMA-Dopa/GelMA constructs under various loading conditions, while maintaining their structural fidelity, stability, mechanical properties, and biocompatibility. While directly printing onto the adherend yields superior adhesive strength, embedded printing followed by transfer to the target tissue demonstrates greater potential for translational applications. Together, these results demonstrate the potential of bioprinted ATESs as off-the-shelf medical devices for diverse biomedical applications.


Asunto(s)
Bioimpresión , Adhesivos Tisulares , Ingeniería de Tejidos/métodos , Hidrogeles/química , Adhesivos , Bioimpresión/métodos , Dopamina , Gelatina/química , Metacrilatos/química , Impresión Tridimensional
20.
bioRxiv ; 2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37333261

RESUMEN

The oral microbiome is a complex community that matures with dental development while oral health is also a recognized risk factor for systemic disease. Despite the oral cavity having a substantial microbial burden, healing of superficial oral wounds occurs quickly and with little scarring. By contrast, creation of an oro-nasal fistula (ONF), often occurring after surgery to correct a cleft palate, is a significant wound healing challenge that is further complicated by a connection of the oral and nasal microbiome. In this study, we characterized the changes in the oral microbiome of mice following a freshly inflicted wound in the oral palate that results in an open and unhealed ONF. Creation of an ONF in mice significantly lowered oral microbiome alpha diversity, with concurrent blooms of Enterococcus faecalis, Staphylococcus lentus, and Staphylococcus xylosus in the oral cavity. Treatment of mice with oral antibiotics one week prior to ONF infliction resulted in a reduction in the alpha diversity, prevented E. faecalis and S. lentus, and S. xylosus blooms, but did not impact ONF healing. Strikingly, delivery of the beneficial microbe Lactococcus lactis subsp. cremoris (LLC) to the wound bed of the freshly inflicted ONF via a PEG-MAL hydrogel vehicle resulted in rapid healing of the ONF. Healing of the ONF was associated with the maintenance of relatively high microbiome alpha diversity, and limited the abundance of E. faecalis and S. lentus, and S. xylosus in the oral cavity. These data demonstrate that a freshly inflicted ONF in the murine palate is associated with a dysbiotic oral microbiome state that may prevent ONF healing, and a bloom of opportunistic pathogens. The data also demonstrate that delivery of a specific beneficial microbe, LLC, to the ONF can boost wound healing, can restore and/or preserve oral microbiome diversity, and inhibit blooms of opportunistic pathogens.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA