Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Small ; 17(15): e2004258, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33094918

RESUMEN

Cardiotoxicity is one of the most serious side effects of cancer chemotherapy. Current approaches to monitoring of chemotherapy-induced cardiotoxicity (CIC) as well as model systems that develop in vivo or in vitro CIC platforms fail to notice early signs of CIC. Moreover, breast cancer (BC) patients with preexisting cardiac dysfunctions may lead to different incident levels of CIC. Here, a model is presented for investigating CIC where not only induced pluripotent stem cell (iPSC)-derived cardiac tissues are interacted with BC tissues on a dual-organ platform, but electrochemical immuno-aptasensors can also monitor cell-secreted multiple biomarkers. Fibrotic stages of iPSC-derived cardiac tissues are promoted with a supplement of transforming growth factor-ß 1 to assess the differential functionality in healthy and fibrotic cardiac tissues after treatment with doxorubicin (DOX). The production trend of biomarkers evaluated by using the immuno-aptasensors well-matches the outcomes from conventional enzyme-linked immunosorbent assay, demonstrating the accuracy of the authors' sensing platform with much higher sensitivity and lower detection limits for early monitoring of CIC and BC progression. Furthermore, the versatility of this platform is demonstrated by applying a nanoparticle-based DOX-delivery system. The proposed platform would potentially help allow early detection and prediction of CIC in individual patients in the future.


Asunto(s)
Neoplasias de la Mama , Cardiotoxicidad , Neoplasias de la Mama/tratamiento farmacológico , Cardiotoxicidad/diagnóstico , Cardiotoxicidad/etiología , Doxorrubicina/efectos adversos , Femenino , Corazón , Humanos , Dispositivos Laboratorio en un Chip , Miocitos Cardíacos
2.
Adv Healthc Mater ; 11(6): e2101678, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34971210

RESUMEN

In vitro cardiomyocyte (CM) maturation is an imperative step to replicate native heart tissue-like structures as cardiac tissue grafts or as drug screening platforms. CMs are known to interpret biophysical cues such as stiffness, topography, external mechanical stimulation or dynamic perfusion load through mechanotransduction and change their behavior, organization, and maturation. In this regard, a silk-based cardiac tissue (CT) coupled with a dynamic perfusion-based mechanical stimulation platform (DMM) for achieving maturation and functionality in vitro is tried to be delivered. Silk fibroin (SF) is used to fabricate lamellar scaffolds to provide native tissue-like anisotropic architecture and is found to be nonimmunogenic and biocompatible allowing cardiomyocyte attachment and growth in vitro. Further, the scaffolds display excellent mechanical properties by their ability to undergo cyclic compressions without any deformation when places in the DMM. Gradient compression strains (5% to 20%), mimicking the native physiological and pathological conditions, are applied to the cardiomyocyte culture seeded on lamellar silk scaffolds in the DMM. A strain-dependent difference in cardiomyocyte maturation, gene expression, sarcomere elongation, and extracellular matrix formation is observed. These silk-based CTs matured in the DMM can open up several avenues toward the development of host-specific grafts and in vitro models for drug screening.


Asunto(s)
Fibroínas , Materiales Biocompatibles , Fibroínas/química , Mecanotransducción Celular , Perfusión , Seda , Ingeniería de Tejidos , Andamios del Tejido/química
3.
Cells ; 10(12)2021 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-34943930

RESUMEN

Experimental models of the central nervous system (CNS) are imperative for developmental and pathophysiological studies of neurological diseases. Among these models, three-dimensional (3D) induced pluripotent stem cell (iPSC)-derived brain organoid models have been successful in mitigating some of the drawbacks of 2D models; however, they are plagued by high organoid-to-organoid variability, making it difficult to compare specific gene regulatory pathways across 3D organoids with those of the native brain. Single-cell RNA sequencing (scRNA-seq) transcriptome datasets have recently emerged as powerful tools to perform integrative analyses and compare variability across organoids. However, transcriptome studies focusing on late-stage neural functionality development have been underexplored. Here, we combine and analyze 8 brain organoid transcriptome databases to study the correlation between differentiation protocols and their resulting cellular functionality across various 3D organoid and exogenous brain models. We utilize dimensionality reduction methods including principal component analysis (PCA) and uniform manifold approximation projection (UMAP) to identify and visualize cellular diversity among 3D models and subsequently use gene set enrichment analysis (GSEA) and developmental trajectory inference to quantify neuronal behaviors such as axon guidance, synapse transmission and action potential. We showed high similarity in cellular composition, cellular differentiation pathways and expression of functional genes in human brain organoids during induction and differentiation phases, i.e., up to 3 months in culture. However, during the maturation phase, i.e., 6-month timepoint, we observed significant developmental deficits and depletion of neuronal and astrocytes functional genes as indicated by our GSEA results. Our results caution against use of organoids to model pathophysiology and drug response at this advanced time point and provide insights to tune in vitro iPSC differentiation protocols to achieve desired neuronal functionality and improve current protocols.


Asunto(s)
Encéfalo/metabolismo , Diferenciación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Organoides/metabolismo , Transcriptoma/genética , Encéfalo/embriología , Bases de Datos Genéticas , Regulación del Desarrollo de la Expresión Génica , Humanos , Neuronas/citología , Neuronas/metabolismo , Reproducción , Análisis de Secuencia de ARN , Transducción de Señal/genética , Análisis de la Célula Individual
4.
Sci Adv ; 7(33)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34380619

RESUMEN

Coronavirus disease 2019 (COVID-19) continues to burden society worldwide. Despite most patients having a mild course, severe presentations have limited treatment options. COVID-19 manifestations extend beyond the lungs and may affect the cardiovascular, nervous, and other organ systems. Current treatments are nonspecific and do not address potential long-term consequences such as pulmonary fibrosis, demyelination, and ischemic organ damage. Cell therapies offer great potential in treating severe COVID-19 presentations due to their customizability and regenerative function. This review summarizes COVID-19 pathogenesis, respective areas where cell therapies have potential, and the ongoing 89 cell therapy trials in COVID-19 as of 1 January 2021.

5.
Nat Biotechnol ; 39(4): 510-519, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33257861

RESUMEN

Human pluripotent stem cells (hPSCs) offer an unprecedented opportunity to model diverse cell types and tissues. To enable systematic exploration of the programming landscape mediated by transcription factors (TFs), we present the Human TFome, a comprehensive library containing 1,564 TF genes and 1,732 TF splice isoforms. By screening the library in three hPSC lines, we discovered 290 TFs, including 241 that were previously unreported, that induce differentiation in 4 days without alteration of external soluble or biomechanical cues. We used four of the hits to program hPSCs into neurons, fibroblasts, oligodendrocytes and vascular endothelial-like cells that have molecular and functional similarity to primary cells. Our cell-autonomous approach enabled parallel programming of hPSCs into multiple cell types simultaneously. We also demonstrated orthogonal programming by including oligodendrocyte-inducible hPSCs with unmodified hPSCs to generate cerebral organoids, which expedited in situ myelination. Large-scale combinatorial screening of the Human TFome will complement other strategies for cell engineering based on developmental biology and computational systems biology.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Oligodendroglía/citología , Células Madre Pluripotentes/citología , Factores de Transcripción/genética , Empalme Alternativo , Diferenciación Celular , Ingeniería Celular , Células Cultivadas , Técnicas de Cocultivo , Humanos , Oligodendroglía/metabolismo , Células Madre Pluripotentes/metabolismo , Biología de Sistemas
6.
Adv Sci (Weinh) ; 7(5): 1901878, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32154068

RESUMEN

Advances in biomanufacturing techniques have opened the doors to recapitulate human sensory organs such as the nose and ear in vitro with adequate levels of functionality. Such advancements have enabled simultaneous targeting of two challenges in engineered sensory organs, especially the nose: i) mechanically robust reconstruction of the nasal cartilage with high precision and ii) replication of the nose functionality: odor perception. Hybrid nasal organs can be equipped with remarkable capabilities such as augmented olfactory perception. Herein, a proof-of-concept for an odor-perceptive nose-like hybrid, which is composed of a mechanically robust cartilage-like construct and a biocompatible biosensing platform, is proposed. Specifically, 3D cartilage-like tissue constructs are created by multi-material 3D bioprinting using mechanically tunable chondrocyte-laden bioinks. In addition, by optimizing the composition of stiff and soft bioinks in macro-scale printed constructs, the competence of this system in providing improved viability and recapitulation of chondrocyte cell behavior in mechanically robust 3D constructs is demonstrated. Furthermore, the engineered cartilage-like tissue construct is integrated with an electrochemical biosensing system to bring functional olfactory sensations toward multiple specific airway disease biomarkers, explosives, and toxins under biocompatible conditions. Proposed hybrid constructs can lay the groundwork for functional bionic interfaces and humanoid cyborgs.

7.
Curr Pharm Des ; 24(45): 5471-5486, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30854951

RESUMEN

To reduce the required capital and time investment in the development of new pharmaceutical agents, there is an urgent need for preclinical drug testing models that are predictive of drug response in human tissues or organs. Despite tremendous advancements and rigorous multistage screening of drug candidates involving computational models, traditional cell culture platforms, animal models and most recently humanized animals, there is still a large deficit in our ability to predict drug response in patient groups and overall attrition rates from phase 1 through phase 4 of clinical studies remain well above 90%. Organ-on-a-chip (OOC) platforms have proven potential in providing tremendous flexibility and robustness in drug screening and development by employing engineering techniques and materials. More importantly, in recent years, there is a clear upward trend in studies that utilize human-induced pluripotent stem cell (hiPSC) to develop personalized tissue or organ models. Additionally, integrated multiple organs on the single chip with increasingly more sophisticated representation of absorption, distribution, metabolism, excretion and toxicity (ADMET) process are being utilized to better understand drug interaction mechanisms in the human body and thus showing great potential to better predict drug efficacy and safety. In this review, we summarize these advances, highlighting studies that took the next step to clinical trials and research areas with the utmost potential and discuss the role of the OOCs in the overall drug discovery process at a preclinical and clinical stage, as well as outline remaining challenges.


Asunto(s)
Descubrimiento de Drogas , Dispositivos Laboratorio en un Chip , Células Madre Pluripotentes/citología , Medicina de Precisión , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA