Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Pharm ; 21(9): 4664-4672, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39133897

RESUMEN

The blood-brain barrier (BBB) poses a significant challenge for drug delivery and is linked to various neurovascular disorders. In vitro BBB models provide a tool to investigate drug permeation across the BBB and the barrier's response to external injury events. Yet, existing models lack fidelity in replicating the BBB's complexity, hindering a comprehensive understanding of its functions. This study introduces a three-dimensional (3D) model using polyethylene glycol (PEG) hydrogels modified with biomimetic peptides that represent recognition sequences of key proteins in the brain. Hydrogels were functionalized with recognition sequences for laminin (IKVAV) and fibronectin peptides (RGD) and chemically cross-linked with matrix metalloprotease-sensitive peptides (MMPs) to mimic the extracellular matrix of the BBB. Astrocytes and endothelial cells were seeded within and on the surface of the hydrogels, respectively. The barrier integrity was assessed through different tests including transendothelial electrical resistance (TEER), the permeability of sodium fluorescence (Na-F), the permeability of Evan's blue bound to albumin (EBA), and the expression of zonula occluden-1 (ZO-1) in seeded endothelial cells. Hydrogels with a combination of RGD and IKVAV peptides displayed superior performance, exhibiting significantly higher TEER values (55.33 ± 1.47 Ω·cm2) at day 5 compared to other 2D controls including HAECs-monoculture and HAECs-cocultured with NHAs seeded on well inserts and 3D controls including RGD hydrogel and RGD-IKVAV monoculture with HAECs and RGD hydrogel cocultured with HAECs and NHAs. The designed 3D system resulted in the lowest Evan's blue permeability at 120 min (0.215 ± 0.055 µg/mL) compared to controls. ZO-1 expression was significantly higher and formed a relatively larger network in the functionalized hydrogel cocultured with astrocytes and endothelial cells compared to the controls. Thus, the designed 3D model effectively recapitulates the main BBB structure and function in vitro and is expected to contribute to a deeper understanding of pathological CNS angiogenesis and the development of effective CNS medications.


Asunto(s)
Astrocitos , Barrera Hematoencefálica , Técnicas de Cocultivo , Células Endoteliales , Hidrogeles , Péptidos , Polietilenglicoles , Barrera Hematoencefálica/metabolismo , Astrocitos/metabolismo , Polietilenglicoles/química , Células Endoteliales/metabolismo , Técnicas de Cocultivo/métodos , Hidrogeles/química , Péptidos/química , Humanos , Oligopéptidos/química , Fibronectinas/química , Fibronectinas/metabolismo , Laminina/química , Animales , Biomimética/métodos , Materiales Biomiméticos/química , Células Cultivadas
2.
Biofabrication ; 16(4)2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39121863

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer, a leading cause of cancer-related deaths globally. Initial lesions of PDAC develop within the exocrine pancreas' functional units, with tumor progression driven by interactions between PDAC and stromal cells. Effective therapies require anatomically and functionally relevantin vitrohuman models of the pancreatic cancer microenvironment. We employed tomographic volumetric bioprinting, a novel biofabrication method, to create human fibroblast-laden constructs mimicking the tubuloacinar structures of the exocrine pancreas. Human pancreatic ductal epithelial (HPDE) cells overexpressing the KRAS oncogene (HPDE-KRAS) were seeded in the multiacinar cavity to replicate pathological tissue. HPDE cell growth and organization within the structure were assessed, demonstrating the formation of a thin epithelium covering the acini inner surfaces. Immunofluorescence assays showed significantly higher alpha smooth muscle actin (α-SMA) vs. F-actin expression in fibroblasts co-cultured with cancerous versus wild-type HPDE cells. Additionally,α-SMA expression increased over time and was higher in fibroblasts closer to HPDE cells. Elevated interleukin (IL)-6 levels were quantified in supernatants from co-cultures of stromal and HPDE-KRAS cells. These findings align with inflamed tumor-associated myofibroblast behavior, serving as relevant biomarkers to monitor early disease progression and target drug efficacy. To our knowledge, this is the first demonstration of a 3D bioprinted model of exocrine pancreas that recapitulates its true 3-dimensional microanatomy and shows tumor triggered inflammation.


Asunto(s)
Bioimpresión , Fibroblastos , Páncreas Exocrino , Humanos , Páncreas Exocrino/metabolismo , Fibroblastos/metabolismo , Fibroblastos/citología , Impresión Tridimensional , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Tomografía , Actinas/metabolismo , Interleucina-6/metabolismo , Ingeniería de Tejidos , Técnicas de Cocultivo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética
3.
Trends Biotechnol ; 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39025680

RESUMEN

Understanding the highly complex tumor-immune landscape is an important goal for developing novel immune therapies for solid cancers. To this end, 3D cancer-immune models have emerged as patient-relevant in vitro tools for modeling the tumor-immune landscape and the cellular interactions within it. In this review, we provide an overview of the components and applications of 3D cancer-immune models and discuss their evolution from 2015 to 2023. Specifically, we observe trends in primary cell-sourced, T cell-based complex models used for therapy evaluation and biological discovery. Finally, we describe the challenges of implementing 3D cancer-immune models and the opportunities for maximizing their potential for deciphering the complex tumor-immune microenvironment and identifying novel, clinically relevant drug targets.

4.
In Vitro Cell Dev Biol Anim ; 60(8): 832-841, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38888654

RESUMEN

Since March 2013, animal testing for toxicity evaluation of cosmetic ingredients is banned in Europe. This directive applies to all personal care ingredients including oral ingredients. Gingival in vitro 3D models are commercially available. However, it is essential to develop "in house model" to modulate several parameters to study oral diseases, determine the toxicity of ingredients, test biocompatibility, and evaluate different formulations of cosmetic ingredients. Our expertise in tissue engineering allowed us to reconstruct human oral tissues from normal human gingival cells (fibroblasts and keratinocytes). Indeed, isolation from surgical leftover was performed to culture these gingival cells. These cells keep their endogenous capacity to proliferate allowing reconstruction of equivalent tissue close to in vivo tissue. Reconstruction of gingival epithelium, chorion equivalent, and the combination of these two tissues (full thickness) using primary gingival cells displayed all characteristics of an in vivo gingival model.


Asunto(s)
Fibroblastos , Encía , Ingeniería de Tejidos , Humanos , Encía/citología , Ingeniería de Tejidos/métodos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Modelos Biológicos , Células Cultivadas , Proliferación Celular/efectos de los fármacos , Técnicas de Cultivo Tridimensional de Células/métodos
5.
Biomater Biosyst ; 14: 100095, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38912165

RESUMEN

This study evaluates the cytocompatibility of cerium-doped mesoporous bioactive glasses (Ce-MBGs) loaded with polyphenols (Ce-MBGs-Poly) for possible application in bone tissue engineering after tumour resection. We tested MBGs powders and pellets on 2D and 3D in vitro models using human bone marrow-derived mesenchymal stem cells (hMSCs), osteosarcoma cells (U2OS), and endothelial cells (EA.hy926). Promisingly, at a low concentration in culture medium, Poly-loaded MBGs powders containing 1.2 mol% of cerium inhibited U2OS metabolic activity, preserved hMSCs viability, and had no adverse effects on EA.hy926 migration. Moreover, the study discussed the possible interaction between cerium and Poly, influencing anti-cancer effects. In summary, this research provides insights into the complex interactions between Ce-MBGs, Poly, and various cell types in distinct 2D and 3D in vitro models, highlighting the potential of loaded Ce-MBGs for post-resection bone tissue engineering with a balance between pro-regenerative and anti-tumorigenic activities.

6.
Int J Mol Sci ; 25(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892151

RESUMEN

Astaxanthin (AST) is a natural compound derived from shellfish, microorganisms, and algae, with several healthy properties. For this reason, it is widely used in the diet of humans and animals, such as pigs, broilers, and fish, where its addition is related to its pigmenting properties. Moreover, AST's ability to reduce free radicals and protect cells from oxidative damage finds application during the weaning period, when piglets are exposed to several stressors. To better elucidate the mechanisms involved, here we generate ad hoc pig and rainbow trout in vitro platforms able to mimic the intestinal mucosa. The morphology is validated through histological and molecular analysis, while functional properties of the newly generated intestinal barriers, both in porcine and rainbow trout models, are demonstrated by measuring trans-epithelial electrical resistance and analyzing permeability with fluorescein isothiocyanate-dextran. Exposure to AST induced a significant upregulation of antioxidative stress markers and a reduction in the transcription of inflammation-related interleukins. Altogether, the present findings demonstrate AST's ability to interact with the molecular pathways controlling oxidative stress and inflammation both in the porcine and rainbow trout species and suggest AST's positive role in prevention and health.


Asunto(s)
Mucosa Intestinal , Oncorhynchus mykiss , Estrés Oxidativo , Xantófilas , Animales , Xantófilas/farmacología , Oncorhynchus mykiss/metabolismo , Porcinos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Antioxidantes/farmacología , Intestinos/efectos de los fármacos , Modelos Biológicos , Permeabilidad/efectos de los fármacos
7.
ACS Biomater Sci Eng ; 10(5): 2616-2635, 2024 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-38664996

RESUMEN

Primary brain tumor is one of the most fatal diseases. The most malignant type among them, glioblastoma (GBM), has low survival rates. Standard treatments reduce the life quality of patients due to serious side effects. Tumor aggressiveness and the unique structure of the brain render the removal of tumors and the development of new therapies challenging. To elucidate the characteristics of brain tumors and examine their response to drugs, realistic systems that mimic the tumor environment and cellular crosstalk are desperately needed. In the past decade, 3D GBM models have been presented as excellent platforms as they allowed the investigation of the phenotypes of GBM and testing innovative therapeutic strategies. In that scope, 3D bioprinting technology offers utilities such as fabricating realistic 3D bioprinted structures in a layer-by-layer manner and precisely controlled deposition of materials and cells, and they can be integrated with other technologies like the microfluidics approach. This Review covers studies that investigated 3D bioprinted brain tumor models, especially GBM using 3D bioprinting techniques and essential parameters that affect the result and quality of the study like frequently used cells, the type and physical characteristics of hydrogel, bioprinting conditions, cross-linking methods, and characterization techniques.


Asunto(s)
Bioimpresión , Neoplasias Encefálicas , Glioblastoma , Impresión Tridimensional , Humanos , Glioblastoma/patología , Bioimpresión/métodos , Neoplasias Encefálicas/patología , Animales , Encéfalo/patología , Ingeniería de Tejidos/métodos
8.
Tissue Eng Part C Methods ; 30(9): 368-382, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38587434

RESUMEN

Dental implants have been clinically used for almost five decades with high success rates. In vitro research models used in implant dentistry are limited to two-dimensional experiments, which are reproducible and well adapted to evaluate a single parameter but do not reproduce the complexity of clinical settings. On the contrary, the in vivo research models using animals offer similar histological and anatomical features to humans, and tissue healing can be close to a clinical situation, but those models are usually accompanied with ethical concerns, and their outcomes could not be extrapolated to humans because of interspecies variabilities. This makes the development of novel in vitro models that recapitulate physiological events occurring during dental implant placement of particular interest for current research in dentistry. Also, such models could be challenged by setting a pathological environment (peri-implantitis) to better understand the disease and eventually serve as a platform to evaluate novel treatment modalities. The aim of this systematic literature review was to cover all the in vitro three-dimensional (3D) complex models available for research in implant dentistry. To accomplish this, a comprehensive search of the literature present on Scopus and PubMed databases was done using specific keywords, as well as inclusion/exclusion criteria. Out of 1334 articles found, we have finally included 27 articles in this review with publication dates between 2001 and 2022. In those articles, the 3D models were designed to study tissue-implant interface behavior in bone or gingival tissue. The articles focused on simulating implant integration, evaluating the effect of different conditions on implant integration, or developing an infection model for the implant integration process. The methods used involved implant material and cells organized in a specific 3D structure. The 3D models developed were able to simulate the process of dental implant osseo- and soft tissue integration and lead to results comparable with conventional in vitro and in vivo models. A relatively limited number of articles were obtained, which indicates that this is an emerging field, highly dependent on progresses made in biotechnologies and tissue engineering, and that further investigation is needed to enhance these 3D in vitro models.


Asunto(s)
Implantes Dentales , Humanos , Animales , Modelos Biológicos
9.
Int J Mol Sci ; 25(8)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38673911

RESUMEN

One of the most significant challenges in human health risk assessment is to evaluate hazards from exposure to environmental chemical mixtures. Polycyclic aromatic hydrocarbons (PAHs) are a class of ubiquitous contaminants typically found as mixtures in gaseous and particulate phases in ambient air pollution associated with petrochemicals from Superfund sites and the burning of fossil fuels. However, little is understood about how PAHs in mixtures contribute to toxicity in lung cells. To investigate mixture interactions and component additivity from environmentally relevant PAHs, two synthetic mixtures were created from PAHs identified in passive air samplers at a legacy creosote site impacted by wildfires. The primary human bronchial epithelial cells differentiated at the air-liquid interface were treated with PAH mixtures at environmentally relevant proportions and evaluated for the differential expression of transcriptional biomarkers related to xenobiotic metabolism, oxidative stress response, barrier integrity, and DNA damage response. Component additivity was evaluated across all endpoints using two independent action (IA) models with and without the scaling of components by toxic equivalence factors. Both IA models exhibited trends that were unlike the observed mixture response and generally underestimated the toxicity across dose suggesting the potential for non-additive interactions of components. Overall, this study provides an example of the usefulness of mixture toxicity assessment with the currently available methods while demonstrating the need for more complex yet interpretable mixture response evaluation methods for environmental samples.


Asunto(s)
Células Epiteliales , Hidrocarburos Policíclicos Aromáticos , Humanos , Hidrocarburos Policíclicos Aromáticos/toxicidad , Hidrocarburos Policíclicos Aromáticos/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Modelos Biológicos , Contaminantes Atmosféricos/toxicidad , Células Cultivadas , Bronquios/metabolismo , Bronquios/citología , Bronquios/efectos de los fármacos , Biomarcadores
10.
Biofabrication ; 16(2)2024 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-38306674

RESUMEN

Glandular cancers are amongst the most prevalent types of cancer, which can develop in many different organs, presenting challenges in their detection as well as high treatment variability and failure rates. For that purpose, anticancer drugs are commonly tested in cancer cell lines grown in 2D tissue culture on plastic dishesin vitro, or in animal modelsin vivo. However, 2D culture models diverge significantly from the 3D characteristics of living tissues and animal models require extensive animal use and time. Glandular cancers, such as prostate cancer-the second leading cause of male cancer death-typically exist in co-centrical architectures where a cell layer surrounds an acellular lumen. Herein, this spatial cellular position and 3D architecture, containing dual compartments with different hydrogel materials, is engineered using a simple co-axial nozzle setup, in a single step utilizing prostate as a model of glandular cancer. The resulting hydrogel soft structures support viable prostate cancer cells of different cell lines and enable over-time maturation into cancer-mimicking aggregates surrounding the acellular core. The biofabricated cancer mimicking structures are then used as a model to predict the inhibitory efficacy of the poly ADP ribose polymerase inhibitor, Talazoparib, and the antiandrogen drug, Enzalutamide, in the growth of the cancer cell layer. Our results show that the obtained hydrogel constructs can be adapted to quickly obtain 3D cancer models which combine 3D physiological architectures with high-throughput screening to detect and optimize anti-cancer drugs in prostate and potentially other glandular cancer types.


Asunto(s)
Antineoplásicos , Neoplasias de la Próstata , Humanos , Animales , Masculino , Hidrogeles/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Línea Celular
11.
Biomaterials ; 305: 122450, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38169190

RESUMEN

In vitro atherosclerosis models are essential to evaluate therapeutics before in vivo and clinical studies, but significant limitations remain, such as the lack of three-layer vascular architecture and limited atherosclerotic features. Moreover, no scalable 3D atherosclerosis model is available for making high-throughput assays for therapeutic evaluation. Herein, we report an in vitro 3D three-layer nanomatrix vascular sheet with critical atherosclerosis multi-features (VSA), including endothelial dysfunction, monocyte recruitment, macrophages, extracellular matrix remodeling, smooth muscle cell phenotype transition, inflammatory cytokine secretion, foam cells, and calcification initiation. Notably, we present the creation of high-throughput functional assays with VSAs and the use of these assays for evaluating therapeutics for atherosclerosis treatment. The therapeutics include conventional drugs (statin and sirolimus), candidates for treating atherosclerosis (curcumin and colchicine), and potential gene therapy (miR-146a-loaded liposomes). The high efficiency and flexibility of the scalable VSA functional assays should facilitate drug discovery and development for atherosclerosis.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Humanos , Aterosclerosis/tratamiento farmacológico , Macrófagos , Células Espumosas , Monocitos , Expresión Génica , Miocitos del Músculo Liso
12.
Macromol Biosci ; 24(2): e2300162, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37716014

RESUMEN

Collagen-type I gels are widely used for the fabrication of 3D in vitro gingival models. Unfortunately, their long-term stability is low, which limits the variety of in vitro applications. To overcome this problem and achieve better hydrolytic stability of 3D gingival models, fibrin-based hydrogel blends with increased long-term stability in vitro are investigated. Two different fibrin-based hydrogels are tested: fibrin 2.5% (w/v) and fibrin 1% (w/v)/gelatin 5% (w/v). Appropriate numbers of primary human gingival fibroblasts (HGFs) and OKG4/bmi1/TERT (OKG) keratinocytes are optimized to achieve a homogeneous distribution of cells under the assumed 3D conditions. Both hydrogels support the viability of HGFs and the stability of the hydrogel over 28 days. In vitro cultivation at the air-liquid interface triggers keratinization of the epithelium and increases its thickness, allowing the formation of multiple tissue-like layers. The presence of HGFs in the hydrogel further enhances epithelial differentiation. In conclusion, a fibrin-based 3D gingival model mimics the histology of native gingiva in vitro and ensures its long-term stability in comparison with the previously reported collagen paralogs. These results open new perspectives for extending the period within which specific biological or pathological conditions of artificial gingival tissue can be evaluated.


Asunto(s)
Fibrina , Encía , Humanos , Colágeno , Colágeno Tipo I , Hidrogeles/farmacología , Fibroblastos , Ingeniería de Tejidos/métodos
13.
Tissue Eng Part B Rev ; 30(2): 217-229, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37830183

RESUMEN

In the past decades, anticancer drug development brought the field of tumor engineering to a new level by the need of robust test systems. Simulating tumor microenvironment in vitro remains a challenge, and osteosarcoma-the most common primary bone cancer-is no exception. The growing evidence points to the inevitable connection between biomechanical stimuli and tumor chemosensitivity and aggressiveness, thus making this component of the microenvironment a mandatory requirement to the developed models. In this review, we addressed the question: is the "in vivo - in vitro" gap in osteosarcoma engineering bridged from the perspective of biomechanical stimuli? The most notable biomechanical cues in the tumor cell microenvironment are observed and compared in the contexts of in vivo conditions and engineered three-dimensional in vitro models. Impact statement The importance of biomechanical stimuli in three-dimensional in vitro models for drug testing is becoming more pronounced nowadays. This review might assist in understanding the key players of the biophysical environment of primary bone cancer and the current state of bone tumor engineering from this perspective.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Neoplasias Óseas/patología , Osteosarcoma/patología , Microambiente Tumoral , Microambiente Celular , Modelos Biológicos
14.
J Biomed Mater Res A ; 112(4): 524-533, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-37029655

RESUMEN

With new daily discoveries about the long-term impacts of COVID-19, there is a clear need to develop in vitro models that can be used to better understand the pathogenicity and impact of COVID-19. Here, we demonstrate the utility of developing a model of endothelial dysfunction that utilizes human induced pluripotent stem cell-derived endothelial progenitors encapsulated in collagen hydrogels to study the effects of COVID-19 on the endothelium. These cells form capillary-like vasculature within 1 week after encapsulation and treating these cell-laden hydrogels with SARS-CoV-2 spike protein resulted in a significant decrease in the number of vessel-forming cells as well as vessel network connectivity quantified by our computational pipeline. This vascular dysfunction is a unique phenomenon observed upon treatment with SARS-CoV-2 SP and is not seen upon treatment with other coronaviruses, indicating that these effects were specific to SARS-CoV-2. We show that this vascular dysfunction is caused by an increase in inflammatory cytokines, associated with the COVID-19 cytokine storm, released from SARS-CoV-2 spike protein treated endothelial cells. Following treatment with the corticosteroid dexamethasone, we were able to prevent SARS-CoV-2 spike protein-induced endothelial dysfunction. Our results highlight the importance of understanding the interactions between SARS-CoV-2 spike protein and the endothelium and show that even in the absence of immune cells, the proposed 3D in vitro model for angiogenesis can reproduce COVID-19-induced endothelial dysfunction seen in clinical settings. This model represents a significant step in creating physiologically relevant disease models to further study the impact of long COVID and potentially identify mitigating therapeutics.


Asunto(s)
COVID-19 , Células Madre Pluripotentes Inducidas , Humanos , Glicoproteína de la Espiga del Coronavirus , Células Endoteliales , Síndrome Post Agudo de COVID-19 , SARS-CoV-2 , Hidrogeles/farmacología
15.
J Pathol ; 262(1): 90-104, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37929635

RESUMEN

Crosstalk between cancer and stellate cells is pivotal in pancreatic cancer, resulting in differentiation of stellate cells into myofibroblasts that drives tumour progression. To assess cooperative mechanisms in a 3D context, we generated chimeric spheroids using human and mouse cancer and stellate cells. Species-specific deconvolution of bulk-RNA sequencing data revealed cell type-specific transcriptomes underpinning invasion. This dataset highlighted stellate-specific expression of transcripts encoding the collagen-processing enzymes ADAMTS2 and ADAMTS14. Strikingly, loss of ADAMTS2 reduced, while loss of ADAMTS14 promoted, myofibroblast differentiation and invasion independently of their primary role in collagen-processing. Functional and proteomic analysis demonstrated that these two enzymes regulate myofibroblast differentiation through opposing roles in the regulation of transforming growth factor ß availability, acting on the protease-specific substrates, Serpin E2 and fibulin 2, for ADAMTS2 and ADAMTS14, respectively. Showcasing a broader complexity for these enzymes, we uncovered a novel regulatory axis governing malignant behaviour of the pancreatic cancer stroma. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Miofibroblastos , Neoplasias Pancreáticas , Animales , Humanos , Ratones , Proteínas ADAMTS/genética , Proteínas ADAMTS/metabolismo , Diferenciación Celular , Colágeno/metabolismo , Miofibroblastos/metabolismo , Neoplasias Pancreáticas/patología , Proteómica
16.
J Exp Clin Cancer Res ; 42(1): 341, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38102692

RESUMEN

BACKGROUND: Chimeric antigen receptor (CAR) T-cells are a promising approach in cancer immunotherapy, particularly for treating hematologic malignancies. Yet, their effectiveness is limited when tackling solid tumors, where immune cell infiltration and immunosuppressive tumor microenvironments (TME) are major hurdles. Fibroblast activation protein (FAP) is highly expressed on cancer-associated fibroblasts (CAFs) and various tumor cells, playing an important role in tumor growth and immunosuppression. Aiming to modulate the TME with increased clinical safety and effectiveness, we developed novel small and size-extended immunotheranostic UniCAR target modules (TMs) targeting FAP. METHODS: The specific binding and functionality of the αFAP-scFv TM and the size-extended αFAP-IgG4 TM were assessed using 2D and 3D in vitro models as well as in vivo. Their specific tumor accumulation and diagnostic potential were evaluated using PET studies after functionalization with a chelator and suitable radionuclide. RESULTS: The αFAP-scFv and -IgG4 TMs effectively and specifically redirected UniCAR T-cells using 2D, 3D, and in vivo models. Moreover, a remarkably high and specific accumulation of radiolabeled FAP-targeting TMs at the tumor site of xenograft mouse models was observed. CONCLUSIONS: These findings demonstrate that the novel αFAP TMs are promising immunotheranostic tools to foster cancer imaging and treatment, paving the way for a more convenient, individualized, and safer treatment of cancer patients.


Asunto(s)
Neoplasias , Linfocitos T , Humanos , Animales , Ratones , Microambiente Tumoral , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Inmunoterapia/métodos , Modelos Animales de Enfermedad , Inmunoglobulina G/metabolismo , Línea Celular Tumoral
17.
Front Bioeng Biotechnol ; 11: 1260397, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38026882

RESUMEN

Many neurodegenerative diseases are identified but their causes and cure are far from being well-known. The problem resides in the complexity of the neural tissue and its location which hinders its easy evaluation. Although necessary in the drug discovery process, in vivo animal models need to be reduced and show relevant differences with the human tissues that guide scientists to inquire about other possible options which lead to in vitro models being explored. From organoids to organ-on-a-chips, 3D models are considered the cutting-edge technology in cell culture. Cell choice is a big parameter to take into consideration when planning an in vitro model and cells capable of mimicking both healthy and diseased tissue, such as induced pluripotent stem cells (iPSC), are recognized as good candidates. Hence, we present a critical review of the latest models used to study neurodegenerative disease, how these models have evolved introducing microfluidics platforms, 3D cell cultures, and the use of induced pluripotent cells to better mimic the neural tissue environment in pathological conditions.

18.
Adv Healthc Mater ; 12(30): e2301422, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37703581

RESUMEN

During orthodontic tooth movement (OTM), the periodontal ligament (PDL) plays a crucial role in regulating the tissue remodeling process. To decipher the cellular and molecular mechanisms underlying this process in vitro, suitable 3D models are needed that more closely approximate the situation in vivo. Here, a customized bioreactor is developed that allows dynamic loading of PDL-derived fibroblasts (PDLF). A collagen-based hydrogel mixture is optimized to maintain structural integrity and constant cell growth during stretching. Numerical simulations show a uniform stress distribution in the hydrogel construct under stretching. Compared to static conditions, controlled cyclic stretching results in directional alignment of collagen fibers and enhances proliferation and spreading ability of the embedded PDLF cells. Effective force transmission to the embedded cells is demonstrated by a more than threefold increase in Periostin protein expression. The cyclic stretch conditions also promote extensive remodeling of the extracellular matrix, as confirmed by increased glycosaminoglycan production. These results highlight the importance of dynamic loading over an extended period of time to determine the behavior of PDLF and to identify in vitro mechanobiological cues triggered during OTM-like stimulus. The introduced dynamic bioreactor is therefore a useful in vitro tool to study these mechanisms.


Asunto(s)
Matriz Extracelular , Ligamento Periodontal , Ligamento Periodontal/fisiología , Matriz Extracelular/metabolismo , Colágeno/metabolismo , Reactores Biológicos , Hidrogeles/farmacología , Hidrogeles/metabolismo , Estrés Mecánico
19.
Adv Healthc Mater ; 12(26): e2300903, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37589373

RESUMEN

Modeling the heterogeneity of the tumor microenvironment (TME) in vitro is essential to investigating fundamental cancer biology and developing novel treatment strategies that holistically address the factors affecting tumor progression and therapeutic response. Thus, the development of new tools for both in vitro modeling, such as patient-derived organoids (PDOs) and complex 3D in vitro models, and single cell omics analysis, such as single-cell RNA-sequencing, represents a new frontier for investigating tumor heterogeneity. Specifically, the integration of PDO-based 3D in vitro models and single cell analysis offers a unique opportunity to explore the intersecting effects of interpatient, microenvironmental, and tumor cell heterogeneity on cell phenotypes in the TME. In this review, the current use of PDOs in complex 3D in vitro models of the TME is discussed and the emerging directions in the development of these models are highlighted. Next, work that has successfully applied single cell analysis to PDO-based models is examined and important experimental considerations are identified for this approach. Finally, open questions are highlighted that may be amenable to exploration using the integration of PDO-based models and single cell analysis. Ultimately, such investigations may facilitate the identification of novel therapeutic targets for cancer that address the significant influence of tumor-TME interactions.


Asunto(s)
Neoplasias , Humanos , Biología , Organoides , Fenotipo , Análisis de la Célula Individual , Microambiente Tumoral
20.
Phytochemistry ; 212: 113713, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37169138

RESUMEN

The potential antiviral effects of indole-3-carbinol (I3C), a phytochemical found in Cruciferous vegetables, were investigated. Fibroblasts and epithelial cells were co-cultured on Alvetex® scaffolds, to obtain ad hoc 3D in vitro platforms able to mimic the trachea and intestinal mucosae, which represent the primary structures involved in the coronavirus pathogenesis. The two barriers generated in vitro were treated with various concentrations of I3C for different incubation periods. A protective effect of I3C on both intestinal and trachea models was demonstrated. A significant reduction in the transcription of the two main genes belonging to the Homologous to E6AP C-terminus (HECT)-E3 ligase family members, namely NEDD4 E3 Ubiquitin Protein Ligase (NEDD4) and WW Domain Containing E3 Ubiquitin Protein Ligase 1 (WWP1), which promote virus matrix protein ubiquitination and inhibit viral egression, were detected. These findings indicate I3C potential effect in preventing coronavirus cell egression processes that inhibit viral production. Although further studies are needed to clarify the molecular mechanisms whereby HECT family members control virus life cycle, this work paves the way to the possible therapeutic use of new natural compounds that may reduce the clinical severity of future pandemics.


Asunto(s)
Antivirales , Brassicaceae , Coronavirus , Intestinos , Modelos Biológicos , Fitoquímicos , Tráquea , Verduras , Antivirales/farmacología , Brassicaceae/química , Coronavirus/efectos de los fármacos , Coronavirus/metabolismo , Técnicas In Vitro , Intestinos/efectos de los fármacos , Intestinos/metabolismo , Intestinos/virología , Fitoquímicos/farmacología , Tráquea/efectos de los fármacos , Tráquea/metabolismo , Tráquea/virología , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Verduras/química , Proteínas de la Matriz Viral/metabolismo , Reproducibilidad de los Resultados , Porcinos , Animales , Humanos , Técnicas de Cultivo Tridimensional de Células
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...