Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.264
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(28): e2404210121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38954541

RÉSUMÉ

Mesenchymal stem cells (MSCs) are essential in regenerative medicine. However, conventional expansion and harvesting methods often fail to maintain the essential extracellular matrix (ECM) components, which are crucial for their functionality and efficacy in therapeutic applications. Here, we introduce a bone marrow-inspired macroporous hydrogel designed for the large-scale production of MSC-ECM spheroids. Through a soft-templating approach leveraging liquid-liquid phase separation, we engineer macroporous hydrogels with customizable features, including pore size, stiffness, bioactive ligand distribution, and enzyme-responsive degradability. These tailored environments are conducive to optimal MSC proliferation and ease of harvesting. We find that soft hydrogels enhance mechanotransduction in MSCs, establishing a standard for hydrogel-based 3D cell culture. Within these hydrogels, MSCs exist as both cohesive spheroids, preserving their innate vitality, and as migrating entities that actively secrete functional ECM proteins. Additionally, we also introduce a gentle, enzymatic harvesting method that breaks down the hydrogels, allowing MSCs and secreted ECM to naturally form MSC-ECM spheroids. These spheroids display heightened stemness and differentiation capacity, mirroring the benefits of a native ECM milieu. Our research underscores the significance of sophisticated materials design in nurturing distinct MSC subpopulations, facilitating the generation of MSC-ECM spheroids with enhanced therapeutic potential.


Sujet(s)
Matrice extracellulaire , Hydrogels , Cellules souches mésenchymateuses , Sphéroïdes de cellules , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Hydrogels/composition chimique , Matrice extracellulaire/métabolisme , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Humains , Différenciation cellulaire , Techniques de culture cellulaire/méthodes , Prolifération cellulaire , Porosité , Mécanotransduction cellulaire/physiologie , Cellules cultivées
2.
Biomolecules ; 14(6)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38927015

RÉSUMÉ

The production of nanomaterials through environmentally friendly methods is a top priority in the sustainable development of nanotechnology. This paper presents data on the synthesis of silver nanoparticles using an aqueous extract of Sphagnum fallax moss at room temperature. The morphology, stability, and size of the nanoparticles were analyzed using various techniques, including transmission electron microscopy, Doppler laser velocimetry, and UV-vis spectroscopy. In addition, Fourier transform infrared spectroscopy was used to analyze the presence of moss metabolites on the surface of nanomaterials. The effects of different concentrations of citrate-stabilized and moss extract-stabilized silver nanoparticles on cell viability, necrosis induction, and cell impedance were compared. The internalization of silver nanoparticles into both monolayers and three-dimensional cells spheroids was evaluated using dark-field microscopy and hyperspectral imaging. An eco-friendly method for the synthesis of silver nanoparticles at room temperature is proposed, which makes it possible to obtain spherical nanoparticles of 20-30 nm in size with high bioavailability and that have potential applications in various areas of human life.


Sujet(s)
Nanoparticules métalliques , Extraits de plantes , Argent , Argent/composition chimique , Nanoparticules métalliques/composition chimique , Nanoparticules métalliques/toxicité , Extraits de plantes/composition chimique , Extraits de plantes/pharmacologie , Humains , Survie cellulaire/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Taille de particule
3.
Cells ; 13(12)2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38920683

RÉSUMÉ

Over the past decade, the development of three-dimensional (3D) models has increased exponentially, facilitating the unravelling of fundamental and essential cellular mechanisms by which cells communicate with each other, assemble into tissues and organs and respond to biochemical and biophysical stimuli under both physiological and pathological conditions. This section presents a concise overview of the most recent updates on the significant contribution of different types of 3D cell cultures including spheroids, organoids and organ-on-chip and bio-printed tissues in advancing our understanding of cellular and molecular mechanisms. The case studies presented include the 3D cultures of breast cancer (BC), endometriosis, the liver microenvironment and infections. In BC, the establishment of 3D culture models has permitted the visualization of the role of cancer-associated fibroblasts in the delivery of exosomes, as well as the significance of the physical properties of the extracellular matrix in promoting cell proliferation and invasion. This approach has also become a valuable tool in gaining insight into general and specific mechanisms of drug resistance. Given the considerable heterogeneity of endometriosis, 3D models offer a more accurate representation of the in vivo microenvironment, thereby facilitating the identification and translation of novel targeted therapeutic strategies. The advantages provided by 3D models of the hepatic environment, in conjunction with the high throughput characterizing various platforms, have enabled the elucidation of complex molecular mechanisms underlying various threatening hepatic diseases. A limited number of 3D models for gut and skin infections have been developed. However, a more profound comprehension of the spatial and temporal interactions between microbes, the host and their environment may facilitate the advancement of in vitro, ex vivo and in vivo disease models. Additionally, it may pave the way for the development of novel therapeutic approaches in diverse research fields. The interested reader will also find concluding remarks on the challenges and prospects of using 3D cell cultures for discovering cellular and molecular mechanisms in the research areas covered in this review.


Sujet(s)
Tumeurs du sein , Techniques de cultures cellulaires tridimensionnelles , Endométriose , Humains , Endométriose/anatomopathologie , Endométriose/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Techniques de cultures cellulaires tridimensionnelles/méthodes , Maladies transmissibles/métabolisme , Maladies transmissibles/anatomopathologie , Techniques de culture cellulaire/méthodes , Sphéroïdes de cellules/anatomopathologie , Sphéroïdes de cellules/métabolisme , Foie/anatomopathologie , Foie/métabolisme , Organoïdes/métabolisme , Organoïdes/anatomopathologie , Maladies du foie/anatomopathologie , Maladies du foie/métabolisme , Animaux
4.
Adipocyte ; 13(1): 2347215, 2024 12.
Article de Anglais | MEDLINE | ID: mdl-38864486

RÉSUMÉ

Adipose tissue plays a crucial role in metabolic syndrome, autoimmune diseases, and many cancers. Because of adipose's role in so many aspects of human health, there is a critical need for in vitro models that replicate adipose architecture and function. Traditional monolayer models, despite their convenience, are limited, showing heterogeneity and functional differences compared to 3D models. While monolayer cultures struggle with detachment and inefficient differentiation, healthy adipocytes in 3D culture accumulate large lipid droplets, secrete adiponectin, and produce low levels of inflammatory cytokines. The shift from monolayer models to more complex 3D models aims to better replicate the physiology of healthy adipose tissue in culture. This study introduces a simple and accessible protocol for generating adipose organoids using a scaffold-free spheroid model. The method, utilizing either 96-well spheroid plates or agarose micromolds, demonstrates increased throughput, uniformity, and ease of handling compared to previous techniques. This protocol allows for diverse applications, including drug testing, toxin screening, tissue engineering, and co-culturing. The choice between the two methods depends on the experimental goals, with the 96-well plate providing individualized control and the micromold offering scale advantages. The outlined protocol covers isolation, expansion, and characterization of stromal vascular fraction cells, followed by detailed steps for spheroid formation and optional downstream analyses.


Sujet(s)
Adipocytes , Tissu adipeux , Sphéroïdes de cellules , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/cytologie , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Humains , Adipocytes/métabolisme , Adipocytes/cytologie , Techniques de culture cellulaire/méthodes , Animaux , Ingénierie tissulaire/méthodes , Cellules cultivées , Différenciation cellulaire , Souris
5.
J Control Release ; 371: 386-405, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38844177

RÉSUMÉ

Recently, the formation of three-dimensional (3D) cell aggregates known as embryoid bodies (EBs) grown in media supplemented with HSC-specific morphogens has been utilized for the directed differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), into clinically relevant hematopoietic stem cells (HSCs). However, delivering growth factors and nutrients have become ineffective in inducing synchronous differentiation of cells due to their 3D conformation. Moreover, irregularly sized EBs often lead to the formation of necrotic cores in larger EBs, impairing differentiation. Here, we developed two gelatin microparticles (GelMPs) with different release patterns and two HSC-related growth factors conjugated to them. Slow and fast releasing GelMPs were conjugated with bone morphogenic factor-4 (BMP-4) and stem cell factor (SCF), respectively. The sequential presentation of BMP-4 and SCF in GelMPs resulted in efficient and effective hematopoietic differentiation, shown by the enhanced gene and protein expression of several mesoderm and HSC-related markers, and the increased concentration of released HSC-related cytokines. In the present study, we were able to generate CD34+, CD133+, and FLT3+ cells with similar cellular and molecular morphology as the naïve HSCs that can produce colony units of different blood cells, in vitro.


Sujet(s)
Protéine morphogénétique osseuse de type 4 , Différenciation cellulaire , Gélatine , Cellules souches hématopoïétiques , Cellules souches pluripotentes induites , Sphéroïdes de cellules , Facteur de croissance des cellules souches , Protéine morphogénétique osseuse de type 4/métabolisme , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Facteur de croissance des cellules souches/métabolisme , Gélatine/composition chimique , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Animaux , Humains , Souris
6.
Biofabrication ; 16(3)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38865994

RÉSUMÉ

The lack of adequate humanin vitromodels that recapitulate the cellular composition and response of the human liver to injury hampers the development of anti-fibrotic drugs. The goal of this study was to develop a human spheroid culture model to study liver fibrosis by using induced pluripotent stem cell (iPSC)-derived liver cells. iPSCs were independently differentiated towards hepatoblasts (iHepatoblasts), hepatic stellate cells (iHSCs), endothelial cells (iECs) and macrophages (iMΦ), before assembly into free floating spheroids by culturing cells in 96-well U-bottom plates and orbital shaking for up to 21 days to allow further maturation. Through transcriptome analysis, we show further maturation of iECs and iMΦ, the differentiation of the iHepatoblasts towards hepatocyte-like cells (iHeps) and the inactivation of the iHSCs by the end of the 3D culture. Moreover, these cultures display a similar expression of cell-specific marker genes (CYP3A4, PDGFRß, CD31andCD68) and sensitivity to hepatotoxicity as spheroids made using freshly isolated primary human liver cells. Furthermore, we show the functionality of the iHeps and the iHSCs by mimicking liver fibrosis through iHep-induced iHSC activation, using acetaminophen. In conclusion, we have established a reproducible human iPSC-derived liver culture model that can be used to mimic fibrosisin vitroas a replacement of primary human liver derived 3D models. The model can be used to investigate pathways involved in fibrosis development and to identify new targets for chronic liver disease therapy.


Sujet(s)
Différenciation cellulaire , Techniques de coculture , Cellules souches pluripotentes induites , Cirrhose du foie , Foie , Sphéroïdes de cellules , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Sphéroïdes de cellules/anatomopathologie , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Foie/anatomopathologie , Foie/cytologie , Modèles biologiques , Hépatocytes/cytologie , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/cytologie , Cellules étoilées du foie/anatomopathologie , Cellules cultivées
7.
Int J Mol Sci ; 25(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38892210

RÉSUMÉ

The tumor suppressor gene F-box and WD repeat domain-containing (FBXW) 7 reduces cancer stemness properties by promoting the protein degradation of pluripotent stem cell markers. We recently demonstrated the transcriptional repression of FBXW7 by the three-dimensional (3D) spheroid formation of several cancer cells. In the present study, we found that the transcriptional activity of FBXW7 was promoted by the inhibition of the Ca2+-activated K+ channel, KCa1.1, in a 3D spheroid model of human prostate cancer LNCaP cells through the Akt-Nrf2 signaling pathway. The transcriptional activity of FBXW7 was reduced by the siRNA-mediated inhibition of the CCAAT-enhancer-binding protein C/EBP δ (CEBPD) after the transfection of miR223 mimics in the LNCaP spheroid model, suggesting the transcriptional regulation of FBXW7 through the Akt-Nrf2-CEBPD-miR223 transcriptional axis in the LNCaP spheroid model. Furthermore, the KCa1.1 inhibition-induced activation of FBXW7 reduced (1) KCa1.1 activity and protein levels in the plasma membrane and (2) the protein level of the cancer stem cell (CSC) markers, c-Myc, which is a molecule degraded by FBXW7, in the LNCaP spheroid model, indicating that KCa1.1 inhibition-induced FBXW7 activation suppressed CSC conversion in KCa1.1-positive cancer cells.


Sujet(s)
Protéine-7 contenant une boite F et des répétitions WD , Régulation de l'expression des gènes tumoraux , Facteur-2 apparenté à NF-E2 , Tumeurs de la prostate , Transduction du signal , Sphéroïdes de cellules , Humains , Protéine-7 contenant une boite F et des répétitions WD/métabolisme , Protéine-7 contenant une boite F et des répétitions WD/génétique , Mâle , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Sphéroïdes de cellules/métabolisme , Lignée cellulaire tumorale , Régulation positive , Canaux potassiques calcium-dépendants de conductance intermédiaire/métabolisme , Canaux potassiques calcium-dépendants de conductance intermédiaire/génétique , Canaux potassiques calcium-dépendants de conductance intermédiaire/antagonistes et inhibiteurs , Sous-unités alpha des canaux potassiques calcium-dépendants de grande conductance/métabolisme , Sous-unités alpha des canaux potassiques calcium-dépendants de grande conductance/génétique , microARN/génétique , microARN/métabolisme , Protéines proto-oncogènes c-akt/métabolisme
8.
ACS Biomater Sci Eng ; 10(7): 4463-4479, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38848471

RÉSUMÉ

Scaffold-free bone microtissues differentiated from mesenchymal stem cell (MSC) spheroids offer great potential for bottom-up bone tissue engineering as a direct supply of cells and osteogenic signals. Many biomaterials or biomolecules have been incorporated into bone microtissues to enhance their osteogenic abilities, but these materials are far from clinical approval. Here, we aimed to incorporate hydroxyapatite (HAP) nanoparticles, an essential component of bone matrix, into MSC spheroids to instruct their osteogenic differentiation into bone microtissues and further self-organization into bone organoids with a trabecular structure. Furthermore, the biological interaction between HAP nanoparticles and MSCs and the potential molecular mechanisms in the bone development of MSC spheroids were investigated by both in vitro and in vivo studies. As a result, improved cell viability and osteogenic abilities were observed for the MSC spheroids incorporated with HAP nanoparticles at a concentration of 30 µg/mL. HAP nanoparticles could promote the sequential expression of osteogenic markers (Runx2, Osterix, Sclerostin), promote the expression of bone matrix proteins (OPN, OCN, and Collagen I), promote the mineralization of the bone matrix, and thus promote the bone development of MSC spheroids. The differentiated bone microtissues could further self-organize into linear, lamellar, and spatial bone organoids with trabecular structures. More importantly, adding FAK or Akt inhibitors could decrease the level of HAP-induced osteogenic differentiation of bone microtissues. Finally, excellent new bone regeneration was achieved after injecting bone microtissues into cranial bone defect models, which could also be eliminated by the Akt inhibitor. In conclusion, HAP nanoparticles could promote the development of bone microtissues by promoting the osteogenic differentiation of MSCs and the formation and mineralization of the bone matrix via the FAK/Akt pathway. The bone microtissues could act as individual ossification centers and self-organize into macroscale bone organoids, and in this meaning, the bone microtissues could be called microscale bone organoids. Furthermore, the bone microtissues revealed excellent clinical perspectives for injectable cellular therapies for bone defects.


Sujet(s)
Régénération osseuse , Différenciation cellulaire , Durapatite , Cellules souches mésenchymateuses , Nanoparticules , Ostéogenèse , Protéines proto-oncogènes c-akt , Durapatite/composition chimique , Durapatite/pharmacologie , Régénération osseuse/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Protéines proto-oncogènes c-akt/métabolisme , Animaux , Ostéogenèse/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/cytologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Ingénierie tissulaire/méthodes , Focal adhesion kinase 1/métabolisme , Os et tissu osseux/effets des médicaments et des substances chimiques , Souris , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme
9.
Cells ; 13(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891032

RÉSUMÉ

The Fusarium fungi is found in cereals and feedstuffs and may produce mycotoxins, which are secondary metabolites, such as the T-2 toxin (T-2). In this work, we explored the hepatotoxicity of T-2 using microfluidic 3D hepatic cultures. The objectives were: (i) exploring the benefits of microfluidic 3D cultures compared to conventional 3D cultures available commercially (Aggrewell plates), (ii) establishing 3D co-cultures of hepatic cells (HepG2) and stellate cells (LX2) and assessing T-2 exposure in this model, (iii) characterizing the induction of metabolizing enzymes, and (iv) evaluating inflammatory markers upon T-2 exposure in microfluidic hepatic cultures. Our results demonstrated that, in comparison to commercial (large-volume) 3D cultures, spheroids formed faster and were more functional in microfluidic devices. The viability and hepatic function decreased with increasing T-2 concentrations in both monoculture and co-cultures. The RT-PCR analysis revealed that exposure to T-2 upregulates the expression of multiple Phase I and Phase II hepatic enzymes. In addition, several pro- and anti-inflammatory proteins were increased in co-cultures after exposure to T-2.


Sujet(s)
Foie , Sphéroïdes de cellules , Toxine T-2 , Toxine T-2/toxicité , Humains , Cellules HepG2 , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Foie/métabolisme , Techniques de coculture , Microfluidique/méthodes , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie , Survie cellulaire/effets des médicaments et des substances chimiques
10.
Int J Mol Sci ; 25(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891883

RÉSUMÉ

Articular cartilage damage still remains a major problem in orthopedical surgery. The development of tissue engineering techniques such as autologous chondrocyte implantation is a promising way to improve clinical outcomes. On the other hand, the clinical application of autologous chondrocytes has considerable limitations. Mesenchymal stromal cells (MSCs) from various tissues have been shown to possess chondrogenic differentiation potential, although to different degrees. In the present study, we assessed the alterations in chondrogenesis-related gene transcription rates and extracellular matrix deposition levels before and after the chondrogenic differentiation of MSCs in a 3D spheroid culture. MSCs were obtained from three different tissues: umbilical cord Wharton's jelly (WJMSC-Wharton's jelly mesenchymal stromal cells), adipose tissue (ATMSC-adipose tissue mesenchymal stromal cells), and the dental pulp of deciduous teeth (SHEDs-stem cells from human exfoliated deciduous teeth). Monolayer MSC cultures served as baseline controls. Newly formed 3D spheroids composed of MSCs previously grown in 2D cultures were precultured for 2 days in growth medium, and then, chondrogenic differentiation was induced by maintaining them in the TGF-ß1-containing medium for 21 days. Among the MSC types studied, WJMSCs showed the most similarities with primary chondrocytes in terms of the upregulation of cartilage-specific gene expression. Interestingly, such upregulation occurred to some extent in all 3D spheroids, even prior to the addition of TGF-ß1. These results confirm that the potential of Wharton's jelly is on par with adipose tissue as a valuable cell source for cartilage engineering applications as well as for the treatment of osteoarthritis. The 3D spheroid environment on its own acts as a trigger for the chondrogenic differentiation of MSCs.


Sujet(s)
Différenciation cellulaire , Chondrocytes , Chondrogenèse , Matrice extracellulaire , Cellules souches mésenchymateuses , Sphéroïdes de cellules , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Humains , Chondrogenèse/génétique , Matrice extracellulaire/métabolisme , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Chondrocytes/cytologie , Chondrocytes/métabolisme , Cellules cultivées , Gelée de Wharton/cytologie , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Techniques de culture cellulaire/méthodes , Ingénierie tissulaire/méthodes , Cartilage/cytologie , Cartilage/métabolisme , Dent de lait/cytologie , Dent de lait/métabolisme , Pulpe dentaire/cytologie , Pulpe dentaire/métabolisme
11.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892032

RÉSUMÉ

Keloids, marked by abnormal cellular proliferation and excessive extracellular matrix (ECM) accumulation, pose significant therapeutic challenges. Ethyl pyruvate (EP), an inhibitor of the high-mobility group box 1 (HMGB1) and TGF-ß1 pathways, has emerged as a potential anti-fibrotic agent. Our research evaluated EP's effects on keloid fibroblast (KF) proliferation and ECM production, employing both in vitro cell cultures and ex vivo patient-derived keloid spheroids. We also analyzed the expression levels of ECM components in keloid tissue spheroids treated with EP through immunohistochemistry. Findings revealed that EP treatment impedes the nuclear translocation of HMGB1 and diminishes KF proliferation. Additionally, EP significantly lowered mRNA and protein levels of collagen I and III by attenuating TGF-ß1 and pSmad2/3 complex expression in both human dermal fibroblasts and KFs. Moreover, metalloproteinase I (MMP-1) and MMP-3 mRNA levels saw a notable increase following EP administration. In keloid spheroids, EP induced a dose-dependent reduction in ECM component expression. Immunohistochemical and western blot analyses confirmed significant declines in collagen I, collagen III, fibronectin, elastin, TGF-ß, AKT, and ERK 1/2 expression levels. These outcomes underscore EP's antifibrotic potential, suggesting its viability as a therapeutic approach for keloids.


Sujet(s)
Fibroblastes , Chéloïde , Pyruvates , Sphéroïdes de cellules , Humains , Chéloïde/métabolisme , Chéloïde/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Pyruvates/pharmacologie , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Matrix metalloproteinase 1/métabolisme , Matrix metalloproteinase 1/génétique , Facteur de croissance transformant bêta-1/métabolisme , Protéine HMGB1/métabolisme , Protéine HMGB1/génétique , Collagène/métabolisme , Collagène/biosynthèse , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Matrix metalloproteinase 3/métabolisme , Matrix metalloproteinase 3/génétique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Collagène de type I/métabolisme , Collagène de type I/génétique , Protéine Smad2/métabolisme , Protéine Smad2/génétique , Protéine Smad-3/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Mâle
12.
J Transl Med ; 22(1): 530, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38831317

RÉSUMÉ

BACKGROUND: Cancer stem-like cells (CSCs) have been extensively researched as the primary drivers of therapy resistance and tumor relapse in patients with breast cancer. However, due to lack of specific molecular markers, increased phenotypic plasticity and no clear clinicopathological features, the assessment of CSCs presence and functionality in solid tumors is challenging. While several potential markers, such as CD24/CD44, have been proposed, the extent to which they truly represent the stem cell potential of tumors or merely provide static snapshots is still a subject of controversy. Recent studies have highlighted the crucial role of the tumor microenvironment (TME) in influencing the CSC phenotype in breast cancer. The interplay between the tumor and TME induces significant changes in the cancer cell phenotype, leading to the acquisition of CSC characteristics, therapeutic resistance, and metastatic spread. Simultaneously, CSCs actively shape their microenvironment by evading immune surveillance and attracting stromal cells that support tumor progression. METHODS: In this study, we associated in vitro mammosphere formation assays with bulk tumor microarray profiling and deconvolution algorithms to map CSC functionality and the microenvironmental landscape in a large cohort of 125 breast tumors. RESULTS: We found that the TME score was a significant factor associated with CSC functionality. CSC-rich tumors were characterized by an immune-suppressed TME, while tumors devoid of CSC potential exhibited high immune infiltration and activation of pathways involved in the immune response. Gene expression analysis revealed IFNG, CXCR5, CD40LG, TBX21 and IL2RG to be associated with the CSC phenotype and also displayed prognostic value for patients with breast cancer. CONCLUSION: These results suggest that the characterization of CSCs content and functionality in tumors can be used as an attractive strategy to fine-tune treatments and guide clinical decisions to improve patients therapy response.


Sujet(s)
Tumeurs du sein , Régulation de l'expression des gènes tumoraux , Cellules souches tumorales , Microenvironnement tumoral , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Femelle , Transcription génétique , Analyse de profil d'expression de gènes , Lignée cellulaire tumorale , Sphéroïdes de cellules/anatomopathologie , Sphéroïdes de cellules/métabolisme , Phénotype
13.
STAR Protoc ; 5(2): 103111, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38833372

RÉSUMÉ

Currently, there is no effective treatment for obesity and alcohol-associated liver diseases, partially due to the lack of translational human models. Here, we present a protocol to generate 3D human liver spheroids that contain all the liver cell types and mimic "livers in a dish." We describe strategies to induce metabolic and alcohol-associated hepatic steatosis, inflammation, and fibrosis. We outline potential applications, including using human liver spheroids for experimental and translational research and drug screening to identify potential anti-fibrotic therapies.


Sujet(s)
Cirrhose du foie , Foie , Sphéroïdes de cellules , Humains , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/anatomopathologie , Cirrhose du foie/métabolisme , Cirrhose du foie/anatomopathologie , Foie/métabolisme , Foie/anatomopathologie , Stress physiologique/physiologie , Techniques de culture cellulaire/méthodes , Hépatocytes/métabolisme , Hépatocytes/anatomopathologie
14.
Biofabrication ; 16(3)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38934608

RÉSUMÉ

Breast cancer develops in close proximity to mammary adipose tissue and interactions with the local adipose environment have been shown to drive tumor progression. The specific role, however, of this complex tumor microenvironment in cancer cell migration still needs to be elucidated. Therefore, in this study, a 3D bioprinted breast cancer model was developed that allows for a comprehensive analysis of individual tumor cell migration parameters in dependence of adjacent adipose stroma. In this co-culture model, a breast cancer compartment with MDA-MB-231 breast cancer cells embedded in collagen is surrounded by an adipose tissue compartment consisting of adipose-derived stromal cell (ASC) or adipose spheroids in a printable bioink based on thiolated hyaluronic acid. Printing parameters were optimized for adipose spheroids to ensure viability and integrity of the fragile lipid-laden cells. Preservation of the adipogenic phenotype after printing was demonstrated by quantification of lipid content, expression of adipogenic marker genes, the presence of a coherent adipo-specific extracellular matrix, and cytokine secretion. The migration of tumor cells as a function of paracrine signaling of the surrounding adipose compartment was then analyzed using live-cell imaging. The presence of ASC or adipose spheroids substantially increased key migration parameters of MDA-MB-231 cells, namely motile fraction, persistence, invasion distance, and speed. These findings shed new light on the role of adipose tissue in cancer cell migration. They highlight the potential of our 3D printed breast cancer-stroma model to elucidate mechanisms of stroma-induced cancer cell migration and to serve as a screening platform for novel anti-cancer drugs targeting cancer cell dissemination.


Sujet(s)
Tissu adipeux , Bio-impression , Tumeurs du sein , Mouvement cellulaire , Impression tridimensionnelle , Sphéroïdes de cellules , Cellules stromales , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Sphéroïdes de cellules/anatomopathologie , Sphéroïdes de cellules/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Tissu adipeux/cytologie , Femelle , Lignée cellulaire tumorale , Cellules stromales/anatomopathologie , Cellules stromales/métabolisme , Cellules stromales/cytologie , Techniques de coculture , Microenvironnement tumoral
15.
Mol Cell Proteomics ; 23(6): 100782, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38705386

RÉSUMÉ

Cellular communication within the brain is imperative for maintaining homeostasis and mounting effective responses to pathological triggers like hypoxia. However, a comprehensive understanding of the precise composition and dynamic release of secreted molecules has remained elusive, confined primarily to investigations using isolated monocultures. To overcome these limitations, we utilized the potential of TurboID, a non-toxic biotin ligation enzyme, to capture and enrich secreted proteins specifically originating from human brain pericytes in spheroid cocultures with human endothelial cells and astrocytes. This approach allowed us to characterize the pericyte secretome within a more physiologically relevant multicellular setting encompassing the constituents of the blood-brain barrier. Through a combination of mass spectrometry and multiplex immunoassays, we identified a wide spectrum of different secreted proteins by pericytes. Our findings demonstrate that the pericytes secretome is profoundly shaped by their intercellular communication with other blood-brain barrier-residing cells. Moreover, we identified substantial differences in the secretory profiles between hypoxic and normoxic pericytes. Mass spectrometry analysis showed that hypoxic pericytes in coculture increase their release of signals related to protein secretion, mTOR signaling, and the complement system, while hypoxic pericytes in monocultures showed an upregulation in proliferative pathways including G2M checkpoints, E2F-, and Myc-targets. In addition, hypoxic pericytes show an upregulation of proangiogenic proteins such as VEGFA but display downregulation of canonical proinflammatory cytokines such as CXCL1, MCP-1, and CXCL6. Understanding the specific composition of secreted proteins in the multicellular brain microvasculature is crucial for advancing our knowledge of brain homeostasis and the mechanisms underlying pathology. This study has implications for the identification of targeted therapeutic strategies aimed at modulating microvascular signaling in brain pathologies associated with hypoxia.


Sujet(s)
Hypoxie cellulaire , Techniques de coculture , Péricytes , Sphéroïdes de cellules , Péricytes/métabolisme , Humains , Sphéroïdes de cellules/métabolisme , Sécrétome/métabolisme , Cellules endothéliales/métabolisme , Astrocytes/métabolisme , Protéomique/méthodes , Communication cellulaire , Barrière hémato-encéphalique/métabolisme , Cellules cultivées , Encéphale/métabolisme , Spectrométrie de masse , Transduction du signal
16.
ACS Synth Biol ; 13(6): 1705-1715, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38726686

RÉSUMÉ

The spatial sorting of cells into appropriate tissue compartments is essential for embryogenesis and tissue development. Spatial cell sorting is controlled by the interplay between cell surface affinity and intracellular mechanical properties. However, intracellular signaling that can sufficiently sort cell populations remains unexplored. In this study, we engineered chimeric cadherins by replacing the cadherin intracellular domain with cytoskeletal regulators to test their ability to induce spatial cell sorting. Using a fibroblast-based reconstitution system, we observed that Rac1 and RhoA activity in the cadherin tail induced outward and inward sorting, respectively. In particular, RhoA activity embedded cells toward the inside of E-cadherin-expressing spheroids and tumor spheroids, leading to tissue invagination. Despite the simplicity of chimeric cadherin design, our results indicate that differences in cadherin intracellular activities can determine the direction of spatial cell sorting, even when cell surface affinity is not different, and provide new molecular tools to engineer tissue architectures.


Sujet(s)
Cadhérines , Protéine G rac1 , Protéine G RhoA , Cadhérines/métabolisme , Cadhérines/génétique , Protéine G rac1/métabolisme , Protéine G rac1/génétique , Humains , Protéine G RhoA/métabolisme , Protéine G RhoA/génétique , Animaux , Souris , Fibroblastes/métabolisme , Fibroblastes/cytologie , Sphéroïdes de cellules/métabolisme
17.
Int J Mol Sci ; 25(10)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38791178

RÉSUMÉ

Three-dimensional cell cultures have improved the evaluation of drugs for cancer therapy, due to their high similarity to solid tumors. In melanoma, autophagy appears to show a dual role depending on the progression of the disease. p62 protein has been proposed for the evaluation of autophagic flux since its expression is an indicator of the state of autophagy. Pentoxifylline (PTX) and Norcantharidin (NCTD) are drugs that have been shown to possess anticancer effects. In this work, we used B16F1 mouse melanoma cells in two-dimensional (2D) monolayer cultures and three-dimensional (3D) spheroids to test the effect of PTX and NCTD over the p62 expression. We analyzed the effect on p62 expression through Western blot and immunofluorescence assays. Our results indicate that PTX decreases p62 expression in both cell culture models, while Norcantharidin increases its expression in 3D cultures at 24 h. Therefore, these drugs could have a potential therapeutic use for the regulation of autophagy in melanoma, depending on the state of evolution of the disease.


Sujet(s)
Autophagie , Composés hétérocycliques bicycliques , Pentoxifylline , Composés hétérocycliques bicycliques/pharmacologie , Animaux , Souris , Pentoxifylline/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mélanome expérimental/métabolisme , Mélanome expérimental/traitement médicamenteux , Mélanome expérimental/anatomopathologie , Techniques de culture cellulaire , Séquestosome-1/métabolisme , Séquestosome-1/génétique , Antinéoplasiques/pharmacologie , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme
18.
Biomed Microdevices ; 26(2): 26, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38806765

RÉSUMÉ

Three-dimensional (3D) cell culture models have been extensively utilized in various mechanistic studies as well as for drug development studies as superior in vitro platforms than conventional two-dimensional (2D) cell culture models. This is especially the case in cancer biology, where 3D cancer models, such as spheroids or organoids, have been utilized extensively to understand the mechanisms of cancer development. Recently, many sophisticated 3D models such as organ-on-a-chip models are emerging as advanced in vitro models that can more accurately mimic the in vivo tissue functions. Despite such advancements, spheroids are still considered as a powerful 3D cancer model due to the relatively simple structure and compatibility with existing laboratory instruments, and also can provide orders of magnitude higher throughput than complex in vitro models, an extremely important aspects for drug development. However, creating well-defined spheroids remain challenging, both in terms of throughputs in generation as well as reproducibility in size and shape that can make it challenging for drug testing applications. In the past decades, droplet microfluidics utilizing hydrogels have been highlighted due to their potentials. Importantly, core-shell structured gel droplets can avoid spheroid-to-spheroid adhesion that can cause large variations in assays while also enabling long-term cultivation of spheroids with higher uniformity by protecting the core organoid area from external environment while the outer porous gel layer still allows nutrient exchange. Hence, core-shell gel droplet-based spheroid formation can improve the predictivity and reproducibility of drug screening assays. This review paper will focus on droplet microfluidics-based technologies for cancer spheroid production using various gel materials and structures. In addition, we will discuss emerging technologies that have the potential to advance the production of spheroids, prospects of such technologies, and remaining challenges.


Sujet(s)
Hydrogels , Sphéroïdes de cellules , Sphéroïdes de cellules/cytologie , Sphéroïdes de cellules/métabolisme , Humains , Hydrogels/composition chimique , Laboratoires sur puces , Techniques de culture cellulaire/instrumentation , Techniques de culture cellulaire/méthodes , Techniques de cultures cellulaires tridimensionnelles/instrumentation , Techniques de cultures cellulaires tridimensionnelles/méthodes , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Microfluidique/instrumentation , Microfluidique/méthodes , Animaux
19.
STAR Protoc ; 5(2): 103058, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38748881

RÉSUMÉ

Three-dimensional (3D) models play an increasingly important role in preclinical drug testing as they faithfully mimic interactions between cancer cells and the tumor microenvironment (TME). Here, we present a protocol for generating scaffold-free 3D multicomponent human melanoma spheroids. We describe steps for characterizing models using live-cell imaging and histology, followed by drug testing and assessment of cell death through various techniques such as imaging, luminescence-based assays, and flow cytometry. Finally, we demonstrate the models' adaptability for co-cultures with immune cells.


Sujet(s)
Mélanome , Sphéroïdes de cellules , Humains , Sphéroïdes de cellules/anatomopathologie , Sphéroïdes de cellules/métabolisme , Mélanome/anatomopathologie , Mélanome/métabolisme , Évaluation préclinique de médicament/méthodes , Microenvironnement tumoral , Techniques de coculture/méthodes , Lignée cellulaire tumorale , Tests de criblage d'agents antitumoraux/méthodes , Techniques de culture cellulaire/méthodes
20.
Chem Biol Interact ; 397: 111046, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38735451

RÉSUMÉ

Cylindrospermopsin, a potent hepatotoxin produced by harmful cyanobacterial blooms, poses environmental and human health concerns. We used a 3D human liver in vitro model based on spheroids of HepG2 cells, in combination with molecular and biochemical assays, automated imaging, targeted LC-MS-based proteomics, and lipidomics, to explore cylindrospermopsin effects on lipid metabolism and the processes implicated in hepatic steatosis. Cylindrospermopsin (1 µM, 48 h) did not significantly affect cell viability but partially reduced albumin secretion. However, it increased neutral lipid accumulation in HepG2 spheroids while decreasing phospholipid levels. Simultaneously, cylindrospermopsin upregulated genes for lipogenesis regulation (SREBF1) and triacylglycerol synthesis (DGAT1/2) and downregulated genes for fatty acid synthesis (ACLY, ACCA, FASN, SCD1). Fatty acid uptake, oxidation, and lipid efflux genes were not significantly affected. Targeted proteomics revealed increased levels of perilipin 2 (adipophilin), a major hepatocyte lipid droplet-associated protein. Lipid profiling quantified 246 lipid species in the spheroids, with 28 significantly enriched and 15 downregulated by cylindrospermopsin. Upregulated species included neutral lipids, sphingolipids (e.g., ceramides and dihexosylceramides), and some glycerophospholipids (phosphatidylethanolamines, phosphatidylserines), while phosphatidylcholines and phosphatidylinositols were mostly reduced. It suggests that cylindrospermopsin exposures might contribute to developing and progressing towards hepatic steatosis or metabolic dysfunction-associated steatotic liver disease (MASLD).


Sujet(s)
Alcaloïdes , Toxines bactériennes , Toxines de cyanobactéries , Métabolisme lipidique , Foie , Uracile , Humains , Alcaloïdes/pharmacologie , Toxines bactériennes/métabolisme , Uracile/analogues et dérivés , Uracile/métabolisme , Métabolisme lipidique/effets des médicaments et des substances chimiques , Cellules HepG2 , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Homéostasie/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Protéomique , Lipidomique , Lipogenèse/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...