Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4720, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38830847

RESUMEN

Bioadhesive materials and patches are promising alternatives to surgical sutures and staples. However, many existing bioadhesives do not meet the functional requirements of current surgical procedures and interventions. Here, we present a translational patch material that exhibits instant adhesion to tissues (2.5-fold stronger than Tisseel, an FDA-approved fibrin glue), ultra-stretchability (stretching to >300% its original length without losing elasticity), compatibility with rapid photo-projection (<2 min fabrication time/patch), and ability to deliver therapeutics. Using our established procedures for the in silico design and optimization of anisotropic-auxetic patches, we created next-generation patches for instant attachment to tissues while conforming to a broad range of organ mechanics ex vivo and in vivo. Patches coated with extracellular vesicles derived from mesenchymal stem cells demonstrate robust wound healing capability in vivo without inducing a foreign body response and without the need for patch removal that can cause pain and bleeding. We further demonstrate a single material-based, void-filling auxetic patch designed for the treatment of lung puncture wounds.


Asunto(s)
Adhesivos Tisulares , Cicatrización de Heridas , Animales , Humanos , Elasticidad , Células Madre Mesenquimatosas/citología , Ratones , Adhesivo de Tejido de Fibrina , Masculino , Materiales Biocompatibles/química
2.
Nat Commun ; 15(1): 3784, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710716

RESUMEN

Probiotic and engineered microbe-based therapeutics are an emerging class of pharmaceutical agents. They represent a promising strategy for treating various chronic and inflammatory conditions by interacting with the host immune system and/or delivering therapeutic molecules. Here, we engineered a targeted probiotic yeast platform wherein Saccharomyces boulardii is designed to bind to abundant extracellular matrix proteins found within inflammatory lesions of the gastrointestinal tract through tunable antibody surface display. This approach enabled an additional 24-48 h of probiotic gut residence time compared to controls and 100-fold increased probiotic concentrations within the colon in preclinical models of ulcerative colitis in female mice. As a result, pharmacodynamic parameters including colon length, colonic cytokine expression profiles, and histological inflammation scores were robustly improved and restored back to healthy levels. Overall, these studies highlight the potential for targeted microbial therapeutics as a potential oral dosage form for the treatment of inflammatory bowel diseases.


Asunto(s)
Colitis Ulcerosa , Colon , Modelos Animales de Enfermedad , Matriz Extracelular , Probióticos , Saccharomyces boulardii , Animales , Probióticos/administración & dosificación , Femenino , Ratones , Matriz Extracelular/metabolismo , Colitis Ulcerosa/terapia , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Colon/microbiología , Colon/metabolismo , Colon/patología , Ratones Endogámicos C57BL , Colitis/terapia , Colitis/microbiología , Colitis/patología , Citocinas/metabolismo , Humanos
3.
Biomater Sci ; 11(15): 5262-5273, 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37341642

RESUMEN

Live biotherapeutic products (LBPs) are an emerging class of therapeutics comprised of engineered living organisms such as bacteria or yeast. Bioprinting with living materials has now become possible using modern three-dimensional (3D) printing strategies. While there has been significant progress in bioprinting cells, bioprinting LBPs, specifically yeast, remains in its infancy and has not been optimized. Yeasts are a promising platform to develop into protein biofactories because they (1) grow rapidly, (2) are easy to engineer and manufacture, and (3) are inexpensive to produce. Here we developed an optimized method for loading yeast into hydrogel patches using digital light processing (DLP) 3D printing. We assessed the effects of patch geometry, bioink composition, and yeast concentration on yeast viability, patch stability, and protein release, and in doing so developed a patch formulation capable of supporting yeast growth and sustained protein release for at least ten days.


Asunto(s)
Bioimpresión , Andamios del Tejido , Ingeniería de Tejidos/métodos , Saccharomyces cerevisiae , Bioimpresión/métodos , Impresión Tridimensional , Hidrogeles , Proteínas
4.
Trends Biotechnol ; 40(3): 354-369, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34481657

RESUMEN

Genetically engineered microbes that secrete therapeutics, sense and respond to external environments, and/or target specific sites in the gut fall under an emergent class of therapeutics, called live biotherapeutic products (LBPs). As live organisms that require symbiotic host interactions, LBPs offer unique therapeutic opportunities, but also face distinct challenges in the gut microenvironment. In this review, we describe recent approaches (often demonstrated using traditional probiotic microorganisms) to discover LBP chassis and genetic parts utilizing omics-based methods and highlight LBP delivery strategies, with a focus on addressing physiological challenges that LBPs encounter after oral administration. Finally, we share our perspective on the opportunity to apply an integrated approach, wherein discovery and delivery strategies are utilized synergistically, towards tailoring and optimizing LBP efficacy.


Asunto(s)
Probióticos , Administración Oral , Ingeniería Genética , Probióticos/uso terapéutico
5.
Bioeng Transl Med ; 6(2): e10214, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34027097

RESUMEN

Cell therapies have emerged as a promising therapeutic modality with the potential to treat and even cure a diverse array of diseases. Cell therapies offer unique clinical and therapeutic advantages over conventional small molecules and the growing number of biologics. Particularly, living cells can simultaneously and dynamically perform complex biological functions in ways that conventional drugs cannot; cell therapies have expanded the spectrum of available therapeutic options to include key cellular functions and processes. As such, cell therapies are currently one of the most investigated therapeutic modalities in both preclinical and clinical settings, with many products having been approved and many more under active clinical investigation. Here, we highlight the diversity and key advantages of cell therapies and discuss their current clinical advances. In particular, we review 28 globally approved cell therapy products and their clinical use. We also analyze >1700 current active clinical trials of cell therapies, with an emphasis on discussing their therapeutic applications. Finally, we critically discuss the major biological, manufacturing, and regulatory challenges associated with the clinical translation of cell therapies.

6.
AAPS J ; 23(4): 76, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34009532

RESUMEN

A new modality in microbe-mediated drug delivery has recently emerged wherein genetically engineered microbes are used to locally deliver recombinant therapeutic proteins to the gastrointestinal tract. These engineered microbes are often referred to as live biotherapeutic products (LBPs). Despite advanced genetic engineering and recombinant protein expression approaches, little is known on how to control the spatiotemporal dynamics of LBPs and their secreted therapeutics within the gastrointestinal tract. To date, the fundamental pharmacokinetic analyses for microbe-mediated drug delivery systems have not been described. Here, we explore the pharmacokinetics of an engineered, model protein-secreting Saccharomyces cerevisiae, which serves as an ideal organism for the oral delivery of complex, post-translationally modified proteins. We establish three methods to modulate the pharmacokinetics of an engineered, recombinant protein-secreting fungi system: (i) altering oral dose of engineered fungi, (ii) co-administering antibiotics, and (iii) altering recombinant protein secretion titer. Our findings establish the fundamental pharmacokinetics which will be essential in controlling downstream therapeutic response for this new delivery modality.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Tracto Gastrointestinal/metabolismo , Proteínas Recombinantes/farmacocinética , Saccharomyces cerevisiae/genética , Administración Oral , Animales , Femenino , Absorción Gastrointestinal , Ingeniería Genética , Humanos , Ratones , Modelos Animales , Proteínas Recombinantes/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA