Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Small ; 19(52): e2302280, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37649234

ABSTRACT

Glioblastoma (GBM) is the most common primary malignant brain cancer in adults with a dismal prognosis. Temozolomide (TMZ) is the first-in-line chemotherapeutic; however, resistance is frequent and multifactorial. While many molecular and genetic factors have been linked to TMZ resistance, the role of the solid tumor morphology and the tumor microenvironment, particularly the blood-brain barrier (BBB), is unknown. Here, the authors investigate these using a complex in vitro model for GBM and its surrounding BBB. The model recapitulates important clinical features such as a dense tumor core with tumor cells that invade along the perivascular space; and a perfusable BBB with a physiological permeability and morphology that is altered in the presence of a tumor spheroid. It is demonstrated that TMZ sensitivity decreases with increasing cancer cell spatial organization, and that the BBB can contribute to TMZ resistance. Proteomic analysis with next-generation low volume sample workflows of these cultured microtissues revealed potential clinically relevant proteins involved in tumor aggressiveness and TMZ resistance, demonstrating the utility of complex in vitro models for interrogating the tumor microenvironment and therapy validation.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Temozolomide/pharmacology , Temozolomide/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/metabolism , Blood-Brain Barrier/metabolism , Tumor Microenvironment , Proteomics , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Xenograft Model Antitumor Assays
2.
STAR Protoc ; 3(2): 101411, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35620071

ABSTRACT

Intestinal cells marked by Lgr5 function as tissue-resident stem cells that sustain the homeostatic replenishment of the epithelium. By incorporating a diphtheria toxin receptor (DTR) cassette linked to the Lgr5 coding region, native Lgr5-expressing cells are susceptible to ablation upon DT administration in vivo. A similar strategy can be used for Lgr5-expressing cells within organoids established from DTR models. Together, these in vivo and in vitro approaches will facilitate dissection of the roles of Lgr5-expressing cells residing in different tissue compartments. For complete details on the use and execution of this protocol, please refer to Tan et al. (2021).


Subject(s)
Organoids , Receptors, G-Protein-Coupled , Animals , Heparin-binding EGF-like Growth Factor/genetics , Intestines , Mice , Receptors, G-Protein-Coupled/genetics , Stem Cells
3.
Cell Rep ; 34(4): 108633, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33503423

ABSTRACT

Lgr5+ crypt base columnar cells, the operational intestinal stem cells (ISCs), are thought to be dispensable for small intestinal (SI) homeostasis. Using a Lgr5-2A-DTR (diphtheria toxin receptor) model, which ablates Lgr5+ cells with near-complete efficiency and retains endogenous levels of Lgr5 expression, we show that persistent depletion of Lgr5+ ISCs in fact compromises SI epithelial integrity and reduces epithelial turnover in vivo. In vitro, Lgr5-2A-DTR SI organoids are unable to establish or survive when Lgr5+ ISCs are continuously eliminated by adding DT to the media. However, transient exposure to DT at the start of culture allows organoids to form, and the rate of outgrowth reduces with the increasing length of DT presence. Our results indicate that intestinal homeostasis requires a constant pool of Lgr5+ ISCs, which is supplied by rapidly reprogrammed non-Lgr5+ crypt populations when preexisting Lgr5+ ISCs are ablated.


Subject(s)
Intestinal Mucosa/metabolism , Intestines/immunology , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Homeostasis , Humans
4.
Cell Rep ; 33(12): 108535, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33357435

ABSTRACT

Regeneration of adult skeletal muscle is driven largely by resident satellite cells, a stem cell population increasingly considered to display a high degree of molecular heterogeneity. In this study, we find that Lgr5, a receptor for Rspo and a potent mediator of Wnt/ß-catenin signaling, marks a subset of activated satellite cells that contribute to muscle regeneration. Lgr5 is found to be rapidly upregulated in purified myogenic progenitors following acute cardiotoxin-induced injury. In vivo lineage tracing using our Lgr5-2ACreERT2R26tdTomatoLSL reporter mouse model shows that Lgr5+ cells can reconstitute damaged muscle fibers following muscle injury, as well as replenish the quiescent satellite cell pool. Moreover, conditional mutation in Lgr52ACreERT2;KrasG12D;Trp53flox/flox mice drives undifferentiated pleomorphic sarcoma formation in adult mice, thereby substantiating Lgr5+ cells as a cell of origin of sarcomas. Our findings provide the groundwork for developing Rspo/Wnt-signaling-based therapeutics to potentially enhance regenerative outcomes of skeletal muscles in degenerative muscle diseases.


Subject(s)
Muscle, Skeletal/metabolism , Receptors, G-Protein-Coupled/metabolism , Sarcoma/physiopathology , Stem Cells/metabolism , Animals , Cell Differentiation , Mice , Regeneration , Up-Regulation
5.
Nat Commun ; 10(1): 5378, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31772170

ABSTRACT

Wnt signaling is critical for directing epithelial gland development within the uterine lining to ensure successful gestation in adults. Wnt-dependent, Lgr5-expressing stem/progenitor cells are essential for the development of glandular epithelia in the intestine and stomach, but their existence in the developing reproductive tract has not been investigated. Here, we employ Lgr5-2A-EGFP/CreERT2/DTR mouse models to identify Lgr5-expressing cells in the developing uterus and to evaluate their stem cell identity and function. Lgr5 is broadly expressed in the uterine epithelium during embryogenesis, but becomes largely restricted to the tips of developing glands after birth. In-vivo lineage tracing/ablation/organoid culture assays identify these gland-resident Lgr5high cells as Wnt-dependent stem cells responsible for uterine gland development. Adjacent Lgr5neg epithelial cells within the neonatal glands function as essential niche components to support the function of Lgr5high stem cells ex-vivo. These findings constitute a major advance in our understanding of uterine development and lay the foundations for investigating potential contributions of Lgr5+ stem/progenitor cells to uterine disorders.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Stem Cells , Uterus/growth & development , Wnt Proteins/metabolism , Animals , Cell Lineage , Endometrium/growth & development , Female , Gene Expression Regulation, Developmental , Mice, Transgenic , Mullerian Ducts/cytology , Organoids , Pregnancy , Receptors, G-Protein-Coupled/genetics , Stem Cells/cytology , Stem Cells/physiology , Wnt Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...