Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nature ; 633(8028): 165-173, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39143209

RESUMEN

The intimate relationship between the epithelium and immune system is crucial for maintaining tissue homeostasis, with perturbations therein linked to autoimmune disease and cancer1-3. Whereas stem cell-derived organoids are powerful models of epithelial function4, they lack tissue-resident immune cells that are essential for capturing organ-level processes. We describe human intestinal immuno-organoids (IIOs), formed through self-organization of epithelial organoids and autologous tissue-resident memory T (TRM) cells, a portion of which integrate within the epithelium and continuously survey the barrier. TRM cell migration and interaction with epithelial cells was orchestrated by TRM cell-enriched transcriptomic programs governing cell motility and adhesion. We combined IIOs and single-cell transcriptomics to investigate intestinal inflammation triggered by cancer-targeting biologics in patients. Inflammation was associated with the emergence of an activated population of CD8+ T cells that progressively acquired intraepithelial and cytotoxic features. The appearance of this effector population was preceded and potentiated by a T helper-1-like CD4+ population, which initially produced cytokines and subsequently became cytotoxic itself. As a system amenable to direct perturbation, IIOs allowed us to identify the Rho pathway as a new target for mitigation of immunotherapy-associated intestinal inflammation. Given that they recapitulate both the phenotypic outcomes and underlying interlineage immune interactions, IIOs can be used to study tissue-resident immune responses in the context of tumorigenesis and infectious and autoimmune diseases.


Asunto(s)
Intestinos , Organoides , Femenino , Humanos , Masculino , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/citología , Movimiento Celular/inmunología , Células Epiteliales/inmunología , Células Epiteliales/citología , Inmunoterapia/efectos adversos , Inflamación/inmunología , Inflamación/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/citología , Intestinos/inmunología , Intestinos/citología , Células T de Memoria/citología , Células T de Memoria/inmunología , Organoides/citología , Organoides/inmunología , Análisis de la Célula Individual , Transcriptoma , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años
2.
Development ; 141(9): 1794-804, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24757002

RESUMEN

During organogenesis, various molecular and physical signals are orchestrated in space and time to sculpt multiple cell types into functional tissues and organs. The complex and dynamic nature of the process has hindered studies aimed at delineating morphogenetic mechanisms in vivo, particularly in mammals. Recent demonstrations of stem cell-driven tissue assembly in culture offer a powerful new tool for modeling and dissecting organogenesis. However, despite the highly organotypic nature of stem cell-derived tissues, substantial differences set them apart from their in vivo counterparts, probably owing to the altered microenvironment in which they reside and the lack of mesenchymal influences. Advances in the biomaterials and microtechnology fields have, for example, afforded a high degree of spatiotemporal control over the cellular microenvironment, making it possible to interrogate the effects of individual microenvironmental components in a modular fashion and rapidly identify organ-specific synthetic culture models. Hence, bioengineering approaches promise to bridge the gap between stem cell-driven tissue formation in culture and morphogenesis in vivo, offering mechanistic insight into organogenesis and unveiling powerful new models for drug discovery, as well as strategies for tissue regeneration in the clinic. We draw on several examples of stem cell-derived organoids to illustrate how bioengineering can contribute to tissue formation ex vivo. We also discuss the challenges that lie ahead and potential ways to overcome them.


Asunto(s)
Bioingeniería/métodos , Organogénesis , Células Madre/citología , Animales , Materiales Biocompatibles/farmacología , Humanos , Organogénesis/efectos de los fármacos , Organoides/efectos de los fármacos , Células Madre/efectos de los fármacos , Ingeniería de Tejidos
3.
Nat Biomed Eng ; 8(4): 345-360, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38114742

RESUMEN

Predicting the toxicity of cancer immunotherapies preclinically is challenging because models of tumours and healthy organs do not typically fully recapitulate the expression of relevant human antigens. Here we show that patient-derived intestinal organoids and tumouroids supplemented with immune cells can be used to study the on-target off-tumour toxicities of T-cell-engaging bispecific antibodies (TCBs), and to capture clinical toxicities not predicted by conventional tissue-based models as well as inter-patient variabilities in TCB responses. We analysed the mechanisms of T-cell-mediated damage of neoplastic and donor-matched healthy epithelia at a single-cell resolution using multiplexed immunofluorescence. We found that TCBs that target the epithelial cell-adhesion molecule led to apoptosis in healthy organoids in accordance with clinical observations, and that apoptosis is associated with T-cell activation, cytokine release and intra-epithelial T-cell infiltration. Conversely, tumour organoids were more resistant to damage, probably owing to a reduced efficiency of T-cell infiltration within the epithelium. Patient-derived intestinal organoids can aid the study of immune-epithelial interactions as well as the preclinical and clinical development of cancer immunotherapies.


Asunto(s)
Anticuerpos Biespecíficos , Apoptosis , Organoides , Linfocitos T , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Humanos , Organoides/inmunología , Linfocitos T/inmunología , Intestinos/inmunología , Inmunoterapia/métodos , Molécula de Adhesión Celular Epitelial/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Femenino , Mucosa Intestinal/inmunología
4.
Bio Protoc ; 13(1)2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36789090

RESUMEN

Traditional drug safety assessments often fail to predict complications in humans, especially when the drug targets the immune system. Rodent-based preclinical animal models are often ill-suited for predicting immunotherapy-mediated adverse events in humans, in part because of the fundamental differences in immunological responses between species and the human relevant expression profile of the target antigen, if it is expected to be present in normal, healthy tissue. While human-relevant cell-based models of tissues and organs promise to bridge this gap, conventional in vitro two-dimensional models fail to provide the complexity required to model the biological mechanisms of immunotherapeutic effects. Also, like animal models, they fail to recapitulate physiologically relevant levels and patterns of organ-specific proteins, crucial for capturing pharmacology and safety liabilities. Organ-on-Chip models aim to overcome these limitations by combining micro-engineering with cultured primary human cells to recreate the complex multifactorial microenvironment and functions of native tissues and organs. In this protocol, we show the unprecedented capability of two human Organs-on-Chip models to evaluate the safety profile of T cell-bispecific antibodies (TCBs) targeting tumor antigens. These novel tools broaden the research options available for a mechanistic understanding of engineered therapeutic antibodies and for assessing safety in tissues susceptible to adverse events. Graphical abstract Figure 1. Graphical representation of the major steps in target-dependent T cell-bispecific antibodies engagement and immunomodulation, as performed in the Colon Intestine-Chip.

5.
Front Pharmacol ; 13: 837261, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35586042

RESUMEN

Cancer immunotherapy often involves the use of engineered molecules to selectively bind and activate T cells located within tumour tissue. Fundamental to the success of such treatments is the presence or recruitment of T cells localised within the tumour microenvironment. Advanced organ-on-a-chip systems provide an in vitro setting in which to investigate how novel molecules influence the spatiotemporal dynamics of T cell infiltration into tissue, both in the context of anti-tumour efficacy and off-tumour toxicity. While highly promising, the complexity of these systems is such that mathematical modelling plays a crucial role in the quantitative evaluation of experimental results and maximising the mechanistic insight derived. We develop a mechanistic, mathematical model of a novel microphysiological in vitro platform that recapitulates T cell infiltration into epithelial tissue, which may be normal or transformed. The mathematical model describes the spatiotemporal dynamics of infiltrating T cells in response to chemotactic cytokine signalling. We integrate the model with dynamic imaging data to optimise key model parameters. The mathematical model demonstrates a good fit to the observed experimental data and accurately describes the distribution of infiltrating T cells. This model is designed to complement the in vitro system; with the potential to elucidate complex biological mechanisms, including the mode of action of novel therapies and the drivers of safety events, and, ultimately, improve the efficacy-safety profile of T cell-targeted cancer immunotherapies.

6.
Cell Stem Cell ; 29(6): 905-917.e6, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35508177

RESUMEN

Patient-derived xenografts (PDXs) and patient-derived organoids (PDOs) have been shown to model clinical response to cancer therapy. However, it remains challenging to use these models to guide timely clinical decisions for cancer patients. Here, we used droplet emulsion microfluidics with temperature control and dead-volume minimization to rapidly generate thousands of micro-organospheres (MOSs) from low-volume patient tissues, which serve as an ideal patient-derived model for clinical precision oncology. A clinical study of recently diagnosed metastatic colorectal cancer (CRC) patients using an MOS-based precision oncology pipeline reliably assessed tumor drug response within 14 days, a timeline suitable for guiding treatment decisions in the clinic. Furthermore, MOSs capture original stromal cells and allow T cell penetration, providing a clinical assay for testing immuno-oncology (IO) therapies such as PD-1 blockade, bispecific antibodies, and T cell therapies on patient tumors.


Asunto(s)
Neoplasias del Colon , Medicina de Precisión , Neoplasias del Colon/patología , Humanos , Inmunoterapia , Organoides/patología
7.
Adv Mater ; 32(30): e1905366, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32548863

RESUMEN

Intestinal organoids are useful in vitro models for basic and translational studies aimed at understanding and treating disease. However, their routine culture relies on animal-derived matrices that limit translation to clinical applications. In fact, there are few fully defined, synthetic hydrogel systems that allow for the expansion of intestinal organoids. Here, an allyl sulfide photodegradable hydrogel is presented, achieving rapid degradation through radical addition-fragmentation chain transfer (AFCT) reactions, to support routine passaging of intestinal organoids. Shear rheology to first characterize the effect of thiol and allyl sulfide crosslink structures on degradation kinetics is used. Irradiation with 365 nm light (5 mW cm-2 ) in the presence of a soluble thiol (glutathione at 15 × 10-3 m), and a photoinitiator (lithium phenyl-2,4,6-trimethylbenzoylphosphinate at 1 × 10-3 m), leads to complete hydrogel degradation in less than 15 s. Allyl sulfide hydrogels are used to support the formation of epithelial colonies from single intestinal stem cells, and rapid photodegradation is used to achieve repetitive passaging of stem cell colonies without loss in morphology or organoid formation potential. This platform could support long-term culture of intestinal organoids, potentially replacing the need for animal-derived matrices, while also allowing systematic variations to the hydrogel properties tailored for the organoid of interest.


Asunto(s)
Compuestos Alílicos/química , Hidrogeles/química , Hidrogeles/metabolismo , Organoides/metabolismo , Fotólisis , Compuestos de Sulfhidrilo/química , Sulfuros/química , Animales , Mucosa Intestinal/citología , Luz , Ratones , Reología , Resistencia al Corte , Solubilidad
8.
Prog Mol Biol Transl Sci ; 126: 257-78, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25081622

RESUMEN

Multiple molecular and physical cues engage in complex interplay to afford the fine control over stem cell behavior that is required during tissue development, homeostasis, and repair. As a result of a close collaboration between biologists, engineers, and physicists over the past decade, the types of mechanical parameters which influence stem cells, along with the corresponding cellular outcomes, have started to emerge. Moreover, the field has begun to identify the molecular structures and mechanisms whereby stem cells sense physical signals and transduce them into biological response. In this chapter, we survey the existing knowledge of stem cell mechanobiology and discuss bioengineering approaches which hold the potential to address outstanding questions and concepts. In particular, we cover approaches which may help dissect the context-dependent influence of the physical environment, and resolve its interactions with molecular signals. We also describe platforms which could recreate the spatial and temporal variations in mechanics which are present in vivo and may play a role in patterning stem cell behaviors. Comprehensive understanding of the complex mechanical regulation of stem cell fate may bring us closer to taking full advantage of their vast regenerative potential.


Asunto(s)
Materiales Biocompatibles/farmacología , Biofisica , Células Madre/citología , Animales , Linaje de la Célula/efectos de los fármacos , Humanos , Mecanotransducción Celular/efectos de los fármacos , Modelos Biológicos , Células Madre/efectos de los fármacos
9.
Adv Drug Deliv Rev ; 69-70: 19-28, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24582599

RESUMEN

The development of new drugs is currently a long and costly process in large part due to the failure of promising drug candidates identified in initial in vitro screens to perform as intended in vivo. New approaches to drug screening are being developed which focus on providing more biomimetic platforms. This review surveys this new generation of drug screening technologies, and provides an overview of recent developments in organoid culture systems which could afford previously unmatched fidelity for testing bioactivity and toxicity. The challenges inherent in such approaches will also be discussed, with a view towards bridging the gap between proof-of-concept studies and a wider implementation within the drug development community.


Asunto(s)
Descubrimiento de Drogas/tendencias , Organoides/efectos de los fármacos , Organoides/fisiología , Células Madre/efectos de los fármacos , Células Madre/fisiología , Animales , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/tendencias , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Humanos , Organoides/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA