Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 30
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Cell Rep ; 43(10): 114837, 2024 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-39368089

RÉSUMÉ

The major lactiferous ducts of the human breast branch out and end at terminal ductal lobular units (TDLUs). Despite their functional and clinical importance, the three-dimensional (3D) architecture of TDLUs has remained undetermined. Our quantitative and volumetric imaging of healthy human breast tissue demonstrates that highly branched TDLUs, which exhibit increased proliferation, are uncommon in the resting tissue regardless of donor age, parity, or hormonal contraception. Overall, TDLUs have a consistent shape and branch parameters, and they contain a main subtree that dominates in bifurcation events and exhibits a more duct-like keratin expression pattern. Simulation of TDLU branching morphogenesis in three dimensions suggests that evolutionarily conserved mechanisms regulate mammary gland branching in humans and mice despite their anatomical differences. In all, our data provide structural insight into 3D anatomy and branching of the human breast and exemplify the power of volumetric imaging in gaining a deeper understanding of breast biology.

2.
Nature ; 633(8028): 198-206, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39232148

RÉSUMÉ

Oncogenic mutations are abundant in the tissues of healthy individuals, but rarely form tumours1-3. Yet, the underlying protection mechanisms are largely unknown. To resolve these mechanisms in mouse mammary tissue, we use lineage tracing to map the fate of wild-type and Brca1-/-;Trp53-/- cells, and find that both follow a similar pattern of loss and spread within ducts. Clonal analysis reveals that ducts consist of small repetitive units of self-renewing cells that give rise to short-lived descendants. This offers a first layer of protection as any descendants, including oncogenic mutant cells, are constantly lost, thereby limiting the spread of mutations to a single stem cell-descendant unit. Local tissue remodelling during consecutive oestrous cycles leads to the cooperative and stochastic loss and replacement of self-renewing cells. This process provides a second layer of protection, leading to the elimination of most mutant clones while enabling the minority that by chance survive to expand beyond the stem cell-descendant unit. This leads to fields of mutant cells spanning large parts of the epithelial network, predisposing it for transformation. Eventually, clone expansion becomes restrained by the geometry of the ducts, providing a third layer of protection. Together, these mechanisms act to eliminate most cells that acquire somatic mutations at the expense of driving the accelerated expansion of a minority of cells, which can colonize large areas, leading to field cancerization.


Sujet(s)
Transformation cellulaire néoplasique , Glandes mammaires animales , Mutation , Animaux , Femelle , Souris , Protéine BRCA1/déficit , Protéine BRCA1/génétique , Protéine BRCA1/métabolisme , Lignage cellulaire/génétique , Auto-renouvellement cellulaire/génétique , Transformation cellulaire néoplasique/génétique , Clones cellulaires/cytologie , Clones cellulaires/métabolisme , Clones cellulaires/anatomopathologie , Glandes mammaires animales/cytologie , Glandes mammaires animales/anatomopathologie , Glandes mammaires animales/métabolisme , Protéine p53 suppresseur de tumeur/déficit , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Cycle oestral , Cellules souches/cytologie , Cellules souches/métabolisme , Cellules souches/anatomopathologie
3.
Cardiovasc Res ; 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39253943

RÉSUMÉ

AIMS: During embryonic development, arteriovenous (AV) differentiation ensures proper blood vessel formation and maturation. Defects in arterial or venous identity cause inappropriate fusion of vessels, resulting in atypical shunts, so-called arteriovenous malformations (AVM). Currently, the mechanism behind AVM formation remains unclear and treatment options are fairly limited. Mammalian AV differentiation is initiated before the onset of blood flow in the embryo; however, this pre-flow mechanism is poorly understood. Here, we aimed to unravel the role of Smad1/5 signalling in pre-flow arterial identity, and in the process uncovered an unexpected control mechanism of Smad1/5 signalling. METHODS AND RESULTS: We establish that despite Notch1 being expressed in the pre-flow mouse embryo, it is not activated, nor is it necessary for the expression of the earliest arterial genes in the dorsal aortae (i.e., Hey1 and Gja4). Furthermore, interrupting blood flow by using the Ncx1 KO model completely prevents the activation of Notch1 signalling, suggesting a strong role of shear stress in maintaining arterial identity. We demonstrate that early expression of Hey1 and Gja4 requires SMAD1/5 signalling. Using embryo cultures, we show that Smad1/5 signalling is activated through the Alk1/Alk5/TGFßR2 receptor complex, with TGFß1 as a necessary ligand. Furthermore, our findings demonstrate that early arterial gene expression requires the acetylation of Smad1/5 proteins, rendering them more sensitive to TGFß1 stimulation. Blocking acetyl-CoA production prevents pre-flow arterial expression of Hey1 and Gja4, while stabilizing acetylation rescues their expression. CONCLUSIONS: Our findings highlight the importance of the acetyl-CoA production in the cell and provide a novel control mechanism of Smad1/5 signalling involving protein acetylation. As disturbed canonical Smad1/5 signalling is involved in several vascular conditions, our results offer new insights in treatment options for circumventing canonical Smad1/5 signalling.

4.
NPJ Breast Cancer ; 10(1): 31, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38658604

RÉSUMÉ

Research on metastatic cancer has been hampered by limited sample availability. Here we present the breast cancer post-mortem tissue donation program UPTIDER and show how it enabled sampling of a median of 31 (range: 5-90) metastases and 5-8 liquids per patient from its first 20 patients. In a dedicated experiment, we show the mild impact of increasing time after death on RNA quality, transcriptional profiles and immunohistochemical staining in tumor tissue samples. We show that this impact can be counteracted by organ cooling. We successfully generated ex vivo models from tissue and liquid biopsies from distinct histological subtypes of breast cancer. We anticipate these and future findings of UPTIDER to elucidate mechanisms of disease progression and treatment resistance and to provide tools for the exploration of precision medicine strategies in the metastatic setting.

5.
Cell Rep ; 43(4): 114020, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38554280

RÉSUMÉ

Lymphatic endothelial cells (LECs) of the lymph node (LN) parenchyma orchestrate leukocyte trafficking and peripheral T cell dynamics. T cell responses to immunotherapy largely rely on peripheral T cell recruitment in tumors. Yet, a systematic and molecular understanding of how LECs within the LNs control T cell dynamics under steady-state and tumor-bearing conditions is lacking. Intravital imaging combined with immune phenotyping shows that LEC-specific deletion of the essential autophagy gene Atg5 alters intranodal positioning of lymphocytes and accrues their persistence in the LNs by increasing the availability of the main egress signal sphingosine-1-phosphate. Single-cell RNA sequencing of tumor-draining LNs shows that loss of ATG5 remodels niche-specific LEC phenotypes involved in molecular pathways regulating lymphocyte trafficking and LEC-T cell interactions. Functionally, loss of LEC autophagy prevents recruitment of tumor-infiltrating T and natural killer cells and abrogates response to immunotherapy. Thus, an LEC-autophagy program boosts immune-checkpoint responses by guiding systemic T cell dynamics.


Sujet(s)
Autophagie , Inhibiteurs de points de contrôle immunitaires , Noeuds lymphatiques , Sphingosine/analogues et dérivés , Lymphocytes T , Autophagie/effets des médicaments et des substances chimiques , Animaux , Noeuds lymphatiques/immunologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Souris de lignée C57BL , Protéine-5 associée à l'autophagie/métabolisme , Protéine-5 associée à l'autophagie/génétique , Cellules endothéliales/métabolisme , Sphingosine/pharmacologie , Sphingosine/métabolisme , Humains , Lysophospholipides/métabolisme , Immunothérapie/méthodes , Mouvement cellulaire
6.
Breast Cancer Res ; 26(1): 29, 2024 02 19.
Article de Anglais | MEDLINE | ID: mdl-38374113

RÉSUMÉ

BACKGROUND: Neoadjuvant chemotherapy (NAC) is the standard of care for patients with early-stage triple negative breast cancers (TNBC). However, more than half of TNBC patients do not achieve a pathological complete response (pCR) after NAC, and residual cancer burden (RCB) is associated with dismal long-term prognosis. Understanding the mechanisms underlying differential treatment outcomes is therefore critical to limit RCB and improve NAC efficiency. METHODS: Human TNBC cell lines and patient-derived organoids were used in combination with real-time metabolic assays to evaluate the effect of NAC (paclitaxel and epirubicin) on tumor cell metabolism, in particular glycolysis. Diagnostic biopsies (pre-NAC) from patients with early TNBC were analyzed by bulk RNA-sequencing to evaluate the predictive value of a glycolysis-related gene signature. RESULTS: Paclitaxel induced a consistent metabolic switch to glycolysis, correlated with a reduced mitochondrial oxidative metabolism, in TNBC cells. In pre-NAC diagnostic biopsies from TNBC patients, glycolysis was found to be upregulated in non-responders. Furthermore, glycolysis inhibition greatly improved response to NAC in TNBC organoid models. CONCLUSIONS: Our study pinpoints a metabolic adaptation to glycolysis as a mechanism driving resistance to NAC in TNBC. Our data pave the way for the use of glycolysis-related genes as predictive biomarkers for NAC response, as well as the development of inhibitors to overcome this glycolysis-driven resistance to NAC in human TNBC patients.


Sujet(s)
Tumeurs du sein triple-négatives , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Traitement néoadjuvant , Pronostic , Résultat thérapeutique , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
7.
Methods Mol Biol ; 2764: 77-105, 2024.
Article de Anglais | MEDLINE | ID: mdl-38393590

RÉSUMÉ

Over the past 50 years, researchers from the mammary gland field have launched a collection of distinctive 3D cell culture systems to study multiple aspects of mammary gland physiology and disease. As our knowledge about the mammary gland evolves, more sophisticated 3D cell culture systems are required to answer more and more complex questions. Nowadays, morphologically complex mammary organoids can be generated in distinct 3D settings, along with reproduction of multiple aspects of the gland microenvironment. Yet, each 3D culture protocol comes with its advantages and limitations, where some culture systems are best suited to study stemness potential, whereas others are tailored towards the study of mammary gland morphogenesis. Therefore, prior to starting a 3D mammary culture experiment, it is important to consider and select the ideal culture model to address the biological question of interest. The number and technical requirements of novel 3D cell culture methods vastly increased over the past decades, making it currently challenging and time consuming to identify the best experimental testing. In this chapter, we provide a summary of the most promising murine and human 3D organoid models that are currently used in mammary gland biology research. For each model, we will provide a brief description of the protocol and an overview of the expected morphological outcome, the advantages of the model, and the potential pitfalls, to guide the reader to the best model of choice for specific applications.


Sujet(s)
Glandes mammaires animales , Glandes mammaires humaines , Humains , Souris , Animaux , Région mammaire , Organoïdes , Techniques de culture cellulaire/méthodes , Arbres de décision
8.
STAR Protoc ; 4(3): 102526, 2023 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-37651235

RÉSUMÉ

Mouse intraductal modeling enables efficient in vivo propagation of pre-invasive breast cancer lesions and provides a suitable micro-environment for creating patient-derived tumor xenograft models of estrogen-receptor-positive breast cancer. Here, we present a protocol for mouse intraductal modeling of primary ductal carcinoma in situ (DCIS). We describe steps for processing primary DCIS tissues and performing intraductal injections. We then detail procedures for processing intraductal lesions for 3D whole-mount imaging or serial transplantation using magnetic bead sorting. For complete details on the use and execution of this protocol, please refer to Hutten et al. (2023).1.


Sujet(s)
Tumeurs du sein , Carcinome intracanalaire non infiltrant , Humains , Souris , Animaux , Femelle , Carcinome intracanalaire non infiltrant/anatomopathologie , Tumeurs du sein/imagerie diagnostique , Tumeurs du sein/anatomopathologie , Modèles animaux de maladie humaine , Microenvironnement tumoral
9.
Sci Adv ; 9(17): eadf9063, 2023 04 28.
Article de Anglais | MEDLINE | ID: mdl-37126544

RÉSUMÉ

Aberrant AKT activation occurs in a number of cancers, metabolic syndrome, and immune disorders, making it an important target for the treatment of many diseases. To monitor spatial and temporal AKT activity in a live setting, we generated an Akt-FRET biosensor mouse that allows longitudinal assessment of AKT activity using intravital imaging in conjunction with image stabilization and optical window technology. We demonstrate the sensitivity of the Akt-FRET biosensor mouse using various cancer models and verify its suitability to monitor response to drug targeting in spheroid and organotypic models. We also show that the dynamics of AKT activation can be monitored in real time in diverse tissues, including in individual islets of the pancreas, in the brown and white adipose tissue, and in the skeletal muscle. Thus, the Akt-FRET biosensor mouse provides an important tool to study AKT dynamics in live tissue contexts and has broad preclinical applications.


Sujet(s)
Techniques de biocapteur , Protéines proto-oncogènes c-akt , Souris , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Transfert d'énergie par résonance de fluorescence/méthodes , Techniques de biocapteur/méthodes
10.
Cancer Cell ; 41(5): 986-1002.e9, 2023 05 08.
Article de Anglais | MEDLINE | ID: mdl-37116492

RÉSUMÉ

Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive breast cancer (IBC). Due to a lack of biomarkers able to distinguish high- from low-risk cases, DCIS is treated similar to early IBC even though the minority of untreated cases eventually become invasive. Here, we characterized 115 patient-derived mouse-intraductal (MIND) DCIS models reflecting the full spectrum of DCIS observed in patients. Utilizing the possibility to follow the natural progression of DCIS combined with omics and imaging data, we reveal multiple prognostic factors for high-risk DCIS including high grade, HER2 amplification, expansive 3D growth, and high burden of copy number aberrations. In addition, sequential transplantation of xenografts showed minimal phenotypic and genotypic changes over time, indicating that invasive behavior is an intrinsic phenotype of DCIS and supporting a multiclonal evolution model. Moreover, this study provides a collection of 19 distributable DCIS-MIND models spanning all molecular subtypes.


Sujet(s)
Tumeurs du sein , Carcinome intracanalaire non infiltrant , Humains , Animaux , Souris , Femelle , Carcinome intracanalaire non infiltrant/génétique , Carcinome intracanalaire non infiltrant/anatomopathologie , Biobanques , Hétérogreffes , Marqueurs biologiques tumoraux/génétique , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Facteurs de risque , Évolution de la maladie
11.
Sci Adv ; 9(12): eadd5028, 2023 03 22.
Article de Anglais | MEDLINE | ID: mdl-36947620

RÉSUMÉ

Endothelial cells (ECs) grant access of disseminated cancer cells to distant organs. However, the molecular players regulating the activation of quiescent ECs at the premetastatic niche (PMN) remain elusive. Here, we find that ECs at the PMN coexpress tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its cognate death receptor 5 (DR5). Unexpectedly, endothelial TRAIL interacts intracellularly with DR5 to prevent its signaling and preserve a quiescent vascular phenotype. In absence of endothelial TRAIL, DR5 activation induces EC death and nuclear factor κB/p38-dependent EC stickiness, compromising vascular integrity and promoting myeloid cell infiltration, breast cancer cell adhesion, and metastasis. Consistently, both down-regulation of endothelial TRAIL at the PMN by proangiogenic tumor-secreted factors and the presence of the endogenous TRAIL inhibitors decoy receptor 1 (DcR1) and DcR2 favor metastasis. This study discloses an intracrine mechanism whereby TRAIL blocks DR5 signaling in quiescent endothelia, acting as gatekeeper of the vascular barrier that is corrupted by the tumor during cancer cell dissemination.


Sujet(s)
Tumeurs du sein , Cellules endothéliales , Humains , Femelle , Cellules endothéliales/métabolisme , Ligands , Récepteurs de TRAIL/génétique , Récepteurs de TRAIL/métabolisme , Ligand TRAIL , Apoptose/génétique , Facteur de nécrose tumorale alpha/pharmacologie
12.
Methods Mol Biol ; 2608: 183-205, 2023.
Article de Anglais | MEDLINE | ID: mdl-36653709

RÉSUMÉ

The mammary gland consists of a bilayered epithelial structure with an extensively branched morphology. The majority of this epithelial tree is laid down during puberty, during which actively proliferating terminal end buds repeatedly elongate and bifurcate to form the basic structure of the ductal tree. Mammary ducts consist of a basal and luminal cell layer with a multitude of identified sub-lineages within both layers. The understanding of how these different cell lineages are cooperatively driving branching morphogenesis is a problem of crossing multiple scales, as this requires information on the macroscopic branched structure of the gland, as well as data on single-cell dynamics driving the morphogenic program. Here we describe a method to combine genetic lineage tracing with whole-gland branching analysis. Quantitative data on the global organ structure can be used to derive a model for mammary gland branching morphogenesis and provide a backbone on which the dynamics of individual cell lineages can be simulated and compared to lineage-tracing approaches. Eventually, these quantitative models and experiments allow to understand the couplings between the macroscopic shape of the mammary gland and the underlying single-cell dynamics driving branching morphogenesis.


Sujet(s)
Cellules épithéliales , Glandes mammaires animales , Animaux , Morphogenèse/génétique , Lignage cellulaire
13.
Nat Commun ; 14(1): 183, 2023 01 12.
Article de Anglais | MEDLINE | ID: mdl-36635273

RÉSUMÉ

Cancer-associated fibroblasts (CAFs) are abundantly present in the microenvironment of virtually all tumors and strongly impact tumor progression. Despite increasing insight into their function and heterogeneity, little is known regarding the origin of CAFs. Understanding the origin of CAF heterogeneity is needed to develop successful CAF-based targeted therapies. Through various transplantation studies in mice, we show that CAFs in both invasive lobular breast cancer and triple-negative breast cancer originate from mammary tissue-resident normal fibroblasts (NFs). Single-cell transcriptomics, in vivo and in vitro studies reveal the transition of CD26+ and CD26- NF populations into inflammatory CAFs (iCAFs) and myofibroblastic CAFs (myCAFs), respectively. Functional co-culture experiments show that CD26+ NFs transition into pro-tumorigenic iCAFs which recruit myeloid cells in a CXCL12-dependent manner and enhance tumor cell invasion via matrix-metalloproteinase (MMP) activity. Together, our data suggest that CD26+ and CD26- NFs transform into distinct CAF subpopulations in mouse models of breast cancer.


Sujet(s)
Tumeurs du sein , Fibroblastes associés au cancer , Tumeurs du sein triple-négatives , Humains , Animaux , Souris , Femelle , Dipeptidyl peptidase 4/génétique , Fibroblastes , Fibroblastes associés au cancer/anatomopathologie , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/anatomopathologie , Myofibroblastes/anatomopathologie , Microenvironnement tumoral , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale
14.
Nat Commun ; 14(1): 151, 2023 01 11.
Article de Anglais | MEDLINE | ID: mdl-36631478

RÉSUMÉ

Oriented cell divisions are critical for the formation and maintenance of structured epithelia. Proper mitotic spindle orientation relies on polarised anchoring of force generators to the cell cortex by the evolutionarily conserved protein complex formed by the Gαi subunit of heterotrimeric G proteins, the Leucine-Glycine-Asparagine repeat protein (LGN) and the nuclear mitotic apparatus protein. However, the polarity cues that control cortical patterning of this ternary complex remain largely unknown in mammalian epithelia. Here we identify the membrane-associated protein Annexin A1 (ANXA1) as an interactor of LGN in mammary epithelial cells. Annexin A1 acts independently of Gαi to instruct the accumulation of LGN and nuclear mitotic apparatus protein at the lateral cortex to ensure cortical anchoring of Dynein-Dynactin and astral microtubules and thereby planar alignment of the mitotic spindle. Loss of Annexin A1 randomises mitotic spindle orientation, which in turn disrupts epithelial architecture and luminogenesis in three-dimensional cultures of primary mammary epithelial cells. Our findings establish Annexin A1 as an upstream cortical cue that regulates LGN to direct planar cell divisions during mammalian epithelial morphogenesis.


Sujet(s)
Annexine A1 , Polarité de la cellule , Cellules épithéliales , Appareil du fuseau , Animaux , Humains , Souris , Annexine A1/métabolisme , Protéines du cycle cellulaire/métabolisme , Division cellulaire/génétique , Division cellulaire/physiologie , Polarité de la cellule/génétique , Polarité de la cellule/physiologie , Cellules épithéliales/métabolisme , Mammifères/métabolisme , Morphogenèse , Appareil du fuseau/génétique , Appareil du fuseau/métabolisme
15.
Article de Anglais | MEDLINE | ID: mdl-36167726

RÉSUMÉ

Breast cancer is a pathological condition characterized by high morphological and molecular heterogeneity. Not only the breast cancer cells, but also their tumor micro-environment consists of a multitude of cell types and states, which continuously evolve throughout progression of the disease. To understand breast cancer evolution within this complex environment, in situ analysis of breast cancer and their co-evolving cells and structures in space and time are essential. In this review, recent technical advances in three-dimensional (3D) and intravital imaging of breast cancer are discussed. Moreover, we highlight the resulting new knowledge on breast cancer biology obtained through these innovative imaging technologies. Finally, we discuss how multidimensional imaging technologies can be integrated with molecular profiling to understand the full complexity of breast cancer and the tumor micro-environment during tumor progression and treatment response.


Sujet(s)
Tumeurs du sein , Humains , Femelle , Tumeurs du sein/imagerie diagnostique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Imagerie diagnostique , Microenvironnement tumoral
16.
Dev Cell ; 57(20): 2350-2364.e7, 2022 10 24.
Article de Anglais | MEDLINE | ID: mdl-36283390

RÉSUMÉ

Ductal carcinoma in situ (DCIS) is a pre-invasive stage of breast cancer. During invasion, the encapsulating DCIS basement membrane (BM) is compromised, and tumor cells invade the surrounding stroma. The mechanisms that regulate functional epithelial BMs in vivo are poorly understood. Myosin-X (MYO10) is a filopodia-inducing protein associated with metastasis and poor clinical outcome in invasive breast cancer (IBC). We identify elevated MYO10 expression in human DCIS and IBC, and this suggests links with disease progression. MYO10 promotes filopodia formation and cell invasion in vitro and cancer-cell dissemination from progressively invasive human DCIS xenografts. However, MYO10-depleted xenografts are more invasive. These lesions exhibit compromised BMs, poorly defined borders, and increased cancer-cell dispersal and EMT-marker-positive cells. In addition, cancer spheroids are dependent on MYO10-filopodia to generate a near-continuous extracellular matrix boundary. Thus, MYO10 is protective in early-stage breast cancer, correlating with tumor-limiting BMs, and pro-invasive at later stages, facilitating cancer-cell dissemination.


Sujet(s)
Tumeurs du sein , Carcinome canalaire du sein , Carcinome intracanalaire non infiltrant , Humains , Femelle , Carcinome intracanalaire non infiltrant/métabolisme , Carcinome intracanalaire non infiltrant/anatomopathologie , Pseudopodes/métabolisme , Tumeurs du sein/anatomopathologie , Myosines/métabolisme , Membrane basale/métabolisme , Carcinome canalaire du sein/métabolisme
17.
Front Physiol ; 13: 826107, 2022.
Article de Anglais | MEDLINE | ID: mdl-35399282

RÉSUMÉ

Branching morphogenesis is the process that gives rise to branched structures in several organs, such as the lung, the kidney, and the mammary gland. Although morphologically well described, the exact mechanisms driving branch elongation and bifurcation are still poorly understood. Signaling cues from the stroma and extracellular matrix have an important role in driving branching morphogenesis. Organoid models derived from primary mammary epithelial cells have emerged as a powerful tool to gain insight into branching morphogenesis of the mammary gland. However, current available mammary organoid culture protocols result in morphologically simple structures which do not resemble the complex branched structure of the in vivo mammary gland. Supplementation of growth factors to mammary organoids cultured in basement membrane extract or collagen I were shown to induce bud formation and elongation but are not sufficient to drive true branching events. Here, we present an improved culture approach based on 3D primary mammary epithelial cell culture to develop branched organoids with a complex morphology. By alternating the addition of fibroblast growth factor 2 and epidermal growth factor to mammary organoids cultured in a basement membrane extract matrix enriched with collagen type I fibers, we obtain complex mammary organoid structures with primary, secondary, and tertiary branches over a period of 15-20 days. Mammary organoid structures grow >1 mm in size and show an elongated and branched shape which resembles in vivo mammary gland morphology. This novel branched mammary organoid model offers many possibilities to study the mechanisms of branching in the developing mammary gland.

18.
J Vis Exp ; (179)2022 01 20.
Article de Anglais | MEDLINE | ID: mdl-35129183

RÉSUMÉ

The branched structure of the mammary gland is highly dynamic and undergoes several phases of growth and remodeling after birth. Intravital microscopy in combination with skin flap surgery or implantation of imaging windows has been used to study the dynamics of the healthy mammary gland at different developmental stages. Most mammary imaging technologies are limited to a time frame of hours to days, whereas the majority of mammary gland remodeling processes occur in time frames of days to weeks. To study mammary gland remodeling, methods that allow optical access to the tissue of interest for extended time frames are required. Here, an improved version of the titanium mammary imaging window with a replaceable lid (R.MIW) is described that allows high-resolution imaging of the mammary gland with a cellular resolution for up to several weeks. Importantly, the R.MIW provides tissue access over the entire duration of the intravital imaging experiment and could therefore be used for local tissue manipulation, labeling, drug administration, or image-guided microdissection. Taken together, the R.MIW enables high-resolution characterization of the cellular dynamics during mammary gland development, homeostasis, and disease.


Sujet(s)
Microscopie intravitale , Glandes mammaires animales , Animaux , Homéostasie , Microscopie intravitale/méthodes , Glandes mammaires animales/imagerie diagnostique
19.
Article de Anglais | MEDLINE | ID: mdl-37621948

RÉSUMÉ

Tissues are heterogeneous with respect to cellular and non-cellular components and in the dynamic interactions between these elements. To study the behaviour and fate of individual cells in these complex tissues, intravital microscopy (IVM) techniques such as multiphoton microscopy have been developed to visualize intact and live tissues at cellular and subcellular resolution. IVM experiments have revealed unique insights into the dynamic interplay between different cell types and their local environment, and how this drives morphogenesis and homeostasis of tissues, inflammation and immune responses, and the development of various diseases. This Primer introduces researchers to IVM technologies, with a focus on multiphoton microscopy of rodents, and discusses challenges, solutions and practical tips on how to perform IVM. To illustrate the unique potential of IVM, several examples of results are highlighted. Finally, we discuss data reproducibility and how to handle big imaging data sets.

20.
Nat Chem Biol ; 17(11): 1139-1147, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34504322

RÉSUMÉ

The functional activity and differentiation potential of cells are determined by their interactions with surrounding cells. Approaches that allow unbiased characterization of cell states while at the same time providing spatial information are of major value to assess this environmental influence. However, most current techniques are hampered by a tradeoff between spatial resolution and cell profiling depth. Here, we develop a photocage-based technology that allows isolation and in-depth analysis of live cells from regions of interest in complex ex vivo systems, including primary human tissues. The use of a highly sensitive 4-nitrophenyl(benzofuran) cage coupled to a set of nanobodies allows high-resolution photo-uncaging of different cell types in areas of interest. Single-cell RNA-sequencing of spatially defined CD8+ T cells is used to exemplify the feasibility of identifying location-dependent cell states. The technology described here provides a valuable tool for the analysis of spatially defined cells in diverse biological systems, including clinical samples.


Sujet(s)
Benzofuranes/composition chimique , Lymphocytes T CD8+/cytologie , Nitrophénols/composition chimique , Analyse sur cellule unique , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE