Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Publication year range
1.
Immunity ; 54(8): 1758-1771.e7, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34256013

ABSTRACT

Apoptosis can potently defend against intracellular pathogens by directly killing microbes and eliminating their replicative niche. However, the reported ability of Mycobacterium tuberculosis to restrict apoptotic pathways in macrophages in vitro has led to apoptosis being dismissed as a host-protective process in tuberculosis despite a lack of in vivo evidence. Here we define crucial in vivo functions of the death receptor-mediated and BCL-2-regulated apoptosis pathways in mediating protection against tuberculosis by eliminating distinct populations of infected macrophages and neutrophils and priming T cell responses. We further show that apoptotic pathways can be targeted therapeutically with clinical-stage compounds that antagonize inhibitor of apoptosis (IAP) proteins to promote clearance of M. tuberculosis in mice. These findings reveal that any inhibition of apoptosis by M. tuberculosis is incomplete in vivo, advancing our understanding of host-protective responses to tuberculosis (TB) and revealing host pathways that may be targetable for treatment of disease.


Subject(s)
Apoptosis/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Neutrophils/immunology , Tuberculosis, Pulmonary/immunology , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cell Line , Dipeptides/therapeutic use , Humans , Indoles/therapeutic use , Lymphocyte Activation/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/microbiology , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocytes/immunology , Thiazoles/therapeutic use , Tuberculosis, Pulmonary/drug therapy
2.
PLoS Comput Biol ; 20(6): e1011361, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38875302

ABSTRACT

Tumor microenvironments (TMEs) contain vast amounts of information on patient's cancer through their cellular composition and the spatial distribution of tumor cells and immune cell populations. Exploring variations in TMEs between patient groups, as well as determining the extent to which this information can predict outcomes such as patient survival or treatment success with emerging immunotherapies, is of great interest. Moreover, in the face of a large number of cell interactions to consider, we often wish to identify specific interactions that are useful in making such predictions. We present an approach to achieve these goals based on summarizing spatial relationships in the TME using spatial K functions, and then applying functional data analysis and random forest models to both predict outcomes of interest and identify important spatial relationships. This approach is shown to be effective in simulation experiments at both identifying important spatial interactions while also controlling the false discovery rate. We further used the proposed approach to interrogate two real data sets of Multiplexed Ion Beam Images of TMEs in triple negative breast cancer and lung cancer patients. The methods proposed are publicly available in a companion R package funkycells.


Subject(s)
Cell Communication , Tumor Microenvironment , Tumor Microenvironment/physiology , Humans , Cell Communication/physiology , Computational Biology/methods , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Algorithms , Computer Simulation , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Neoplasms/immunology , Neoplasms/pathology , Models, Biological , Female , Random Forest
3.
Development ; 148(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34121118

ABSTRACT

Development of a branching tree in the embryonic lung is crucial for the formation of a fully mature functional lung at birth. Sox9+ cells present at the tip of the primary embryonic lung endoderm are multipotent cells responsible for branch formation and elongation. We performed a genetic screen in murine primary cells and identified aurora kinase b (Aurkb) as an essential regulator of Sox9+ cells ex vivo. In vivo conditional knockout studies confirmed that Aurkb was required for lung development but was not necessary for postnatal growth and the repair of the adult lung after injury. Deletion of Aurkb in embryonic Sox9+ cells led to the formation of a stunted lung that retained the expression of Sox2 in the proximal airways, as well as Sox9 in the distal tips. Although we found no change in cell polarity, we showed that loss of Aurkb or chemical inhibition of Aurkb caused Sox9+ cells to arrest at G2/M, likely responsible for the lack of branch bifurcation. This work demonstrates the power of genetic screens in identifying novel regulators of Sox9+ progenitor cells and lung branching morphogenesis.


Subject(s)
Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Lung/embryology , SOX9 Transcription Factor/metabolism , Animals , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Organogenesis , SOX9 Transcription Factor/genetics
4.
Nat Methods ; 18(9): 997-1012, 2021 09.
Article in English | MEDLINE | ID: mdl-34341583

ABSTRACT

Understanding intratumoral heterogeneity-the molecular variation among cells within a tumor-promises to address outstanding questions in cancer biology and improve the diagnosis and treatment of specific cancer subtypes. Single-cell analyses, especially RNA sequencing and other genomics modalities, have been transformative in revealing novel biomarkers and molecular regulators associated with tumor growth, metastasis and drug resistance. However, these approaches fail to provide a complete picture of tumor biology, as information on cellular location within the tumor microenvironment is lost. New technologies leveraging multiplexed fluorescence, DNA, RNA and isotope labeling enable the detection of tens to thousands of cancer subclones or molecular biomarkers within their native spatial context. The expeditious growth in these techniques, along with methods for multiomics data integration, promises to yield a more comprehensive understanding of cell-to-cell variation within and between individual tumors. Here we provide the current state and future perspectives on the spatial technologies expected to drive the next generation of research and diagnostic and therapeutic strategies for cancer.


Subject(s)
Gene Expression Profiling/methods , Mass Spectrometry/methods , Neoplasms/diagnostic imaging , Proteins/analysis , Animals , Humans , Mice, Transgenic , Multimodal Imaging , Neoplasms/genetics , Neoplasms/pathology , Single-Cell Analysis/methods , Tumor Microenvironment
5.
Biochem Soc Trans ; 50(2): 825-837, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35343573

ABSTRACT

Evading immune destruction is one of the hallmarks of cancer. A key mechanism of immune evasion deployed by tumour cells is to reduce neoantigen presentation through down-regulation of the antigen presentation machinery. MHC-I and MHC-II proteins are key components of the antigen presentation machinery responsible for neoantigen presentation to CD8+ and CD4+ T lymphocytes, respectively. Their expression in tumour cells is modulated by a complex interplay of genomic, transcriptomic and post translational factors involving multiple intracellular antigen processing pathways. Ongoing research investigates mechanisms invoked by cancer cells to abrogate MHC-I expression and attenuate anti-tumour CD8+ cytotoxic T cell response. The discovery of MHC-II on tumour cells has been less characterized. However, this finding has triggered further interest in utilising tumour-specific MHC-II to harness sustained anti-tumour immunity through the activation of CD4+ T helper cells. Tumour-specific expression of MHC-I and MHC-II has been associated with improved patient survival in most clinical studies. Thus, their reactivation represents an attractive way to unleash anti-tumour immunity. This review provides a comprehensive overview of physiologically conserved or novel mechanisms utilised by tumour cells to reduce MHC-I or MHC-II expression. It outlines current approaches employed at the preclinical and clinical trial interface towards reversing these processes in order to improve response to immunotherapy and survival outcomes for patients with cancer.


Subject(s)
Antigen Presentation , Neoplasms , CD4-Positive T-Lymphocytes , Humans , Immunologic Surveillance , Immunotherapy
6.
PLoS Biol ; 15(1): e2000731, 2017 01.
Article in English | MEDLINE | ID: mdl-28125611

ABSTRACT

Lung squamous cell carcinoma (SqCC), the second most common subtype of lung cancer, is strongly associated with tobacco smoking and exhibits genomic instability. The cellular origins and molecular processes that contribute to SqCC formation are largely unexplored. Here we show that human basal stem cells (BSCs) isolated from heavy smokers proliferate extensively, whereas their alveolar progenitor cell counterparts have limited colony-forming capacity. We demonstrate that this difference arises in part because of the ability of BSCs to repair their DNA more efficiently than alveolar cells following ionizing radiation or chemical-induced DNA damage. Analysis of mice harbouring a mutation in the DNA-dependent protein kinase catalytic subunit (DNA-PKcs), a key enzyme in DNA damage repair by nonhomologous end joining (NHEJ), indicated that BSCs preferentially repair their DNA by this error-prone process. Interestingly, polyploidy, a phenomenon associated with genetically unstable cells, was only observed in the human BSC subset. Expression signature analysis indicated that BSCs are the likely cells of origin of human SqCC and that high levels of NHEJ genes in SqCC are correlated with increasing genomic instability. Hence, our results favour a model in which heavy smoking promotes proliferation of BSCs, and their predilection for error-prone NHEJ could lead to the high mutagenic burden that culminates in SqCC. Targeting DNA repair processes may therefore have a role in the prevention and therapy of SqCC.


Subject(s)
DNA Damage , DNA End-Joining Repair , Lung/cytology , Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Death , Cell Separation , DNA Breaks, Double-Stranded , Epithelial Cells/cytology , Epithelial Cells/ultrastructure , Humans , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasms, Squamous Cell/pathology , Pulmonary Alveoli/cytology , Smoking/adverse effects , Trachea/cytology
7.
Nature ; 572(7771): 589-590, 2019 08.
Article in English | MEDLINE | ID: mdl-31462800
8.
Dev Biol ; 443(1): 1-9, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30194919

ABSTRACT

The highly conserved transcription factor Grainyhead-like 2 (Grhl2) exhibits a dynamic expression pattern in lung epithelium throughout embryonic development. Using a conditional gene targeting approach to delete Grhl2 in the developing lung epithelium, our results demonstrate that Grhl2 plays multiple roles in lung morphogenesis that are essential for respiratory function. Loss of Grhl2 leads to impaired ciliated cell differentiation and perturbed formation of terminal saccules. Critically, a substantial increase in Sox9-positive distal tip progenitor cells was observed following loss of Grhl2, suggesting that Grhl2 plays an important role in branching morphogenesis. Gene transcription profiling of Grhl2-deficient lung epithelial cells revealed a significant down regulation of Elf5, a member of the Ets family of transcription factors. Furthermore, ChIP and comparative genomic analyzes confirmed that Elf5 is a direct transcriptional target of Grhl2. Taken together, these results support the hypothesis that Grhl2 controls normal lung morphogenesis by tightly regulating the activity of distal tip progenitor cells.


Subject(s)
Alveolar Epithelial Cells/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , Alveolar Epithelial Cells/metabolism , Animals , Cell Differentiation , DNA-Binding Proteins/metabolism , Down-Regulation , Epithelium/metabolism , Gene Expression Profiling , Lung/embryology , Lung/metabolism , Lung/physiology , Mice/embryology , Respiratory Function Tests/methods , SOX9 Transcription Factor , Saccule and Utricle/metabolism
9.
Nucleic Acids Res ; 45(5): e30, 2017 03 17.
Article in English | MEDLINE | ID: mdl-27899618

ABSTRACT

Carefully designed control experiments provide a gold standard for benchmarking different genomics research tools. A shortcoming of many gene expression control studies is that replication involves profiling the same reference RNA sample multiple times. This leads to low, pure technical noise that is atypical of regular studies. To achieve a more realistic noise structure, we generated a RNA-sequencing mixture experiment using two cell lines of the same cancer type. Variability was added by extracting RNA from independent cell cultures and degrading particular samples. The systematic gene expression changes induced by this design allowed benchmarking of different library preparation kits (standard poly-A versus total RNA with Ribozero depletion) and analysis pipelines. Data generated using the total RNA kit had more signal for introns and various RNA classes (ncRNA, snRNA, snoRNA) and less variability after degradation. For differential expression analysis, voom with quality weights marginally outperformed other popular methods, while for differential splicing, DEXSeq was simultaneously the most sensitive and the most inconsistent method. For sample deconvolution analysis, DeMix outperformed IsoPure convincingly. Our RNA-sequencing data set provides a valuable resource for benchmarking different protocols and data pre-processing workflows. The extra noise mimics routine lab experiments more closely, ensuring any conclusions are widely applicable.


Subject(s)
Benchmarking , RNA/analysis , Sequence Analysis, RNA/standards , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Library , Genomics/instrumentation , Genomics/methods , Humans , RNA/classification , RNA/genetics , RNA Cleavage , Reagent Kits, Diagnostic/standards , Reference Standards , Sequence Analysis, RNA/methods , Sequence Analysis, RNA/statistics & numerical data
10.
Development ; 142(8): 1458-69, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25790853

ABSTRACT

Epigenetic mechanisms involved in the establishment of lung epithelial cell lineage identities during development are largely unknown. Here, we explored the role of the histone methyltransferase Ezh2 during lung lineage determination. Loss of Ezh2 in the lung epithelium leads to defective lung formation and perinatal mortality. We show that Ezh2 is crucial for airway lineage specification and alveolarization. Using optical projection tomography imaging, we found that branching morphogenesis is affected in Ezh2 conditional knockout mice and the remaining bronchioles are abnormal, lacking terminally differentiated secretory club cells. Remarkably, RNA-seq analysis revealed the upregulation of basal genes in Ezh2-deficient epithelium. Three-dimensional imaging for keratin 5 further showed the unexpected presence of a layer of basal cells from the proximal airways to the distal bronchioles in E16.5 embryos. ChIP-seq analysis indicated the presence of Ezh2-mediated repressive marks on the genomic loci of some but not all basal genes, suggesting an indirect mechanism of action of Ezh2. We found that loss of Ezh2 de-represses insulin-like growth factor 1 (Igf1) expression and that modulation of IGF1 signaling ex vivo in wild-type lungs could induce basal cell differentiation. Altogether, our work reveals an unexpected role for Ezh2 in controlling basal cell fate determination in the embryonic lung endoderm, mediated in part by repression of Igf1 expression.


Subject(s)
Cell Differentiation/physiology , Insulin-Like Growth Factor I/metabolism , Lung/cytology , Lung/metabolism , Polycomb Repressive Complex 2/metabolism , Animals , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Enhancer of Zeste Homolog 2 Protein , Flow Cytometry , Insulin-Like Growth Factor I/genetics , Keratin-5/genetics , Keratin-5/metabolism , Lung/embryology , Mice , Polycomb Repressive Complex 2/genetics , Polymerase Chain Reaction
11.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 89-101, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29038050

ABSTRACT

Maintenance of genomic integrity in tissue-specific stem cells is critical for tissue homeostasis and the prevention of deleterious diseases such as cancer. Stem cells are subject to DNA damage induced by endogenous replication mishaps or exposure to exogenous agents. The type of DNA lesion and the cell cycle stage will invoke different DNA repair mechanisms depending on the intrinsic DNA repair machinery of a cell. Inappropriate DNA repair in stem cells can lead to cell death, or to the formation and accumulation of genetic alterations that can be transmitted to daughter cells and so is linked to cancer formation. DNA mutational signatures that are associated with DNA repair deficiencies or exposure to carcinogenic agents have been described in cancer. Here we review the most recent findings on DNA repair pathways activated in epithelial tissue stem and progenitor cells and their implications for cancer mutational signatures. We discuss how deep knowledge of early molecular events leading to carcinogenesis provides insights into DNA repair mechanisms operating in tumours and how these could be exploited therapeutically.


Subject(s)
Adult Stem Cells/physiology , Carcinogenesis/genetics , DNA Repair/physiology , Neoplastic Stem Cells/pathology , Animals , Carcinogenesis/pathology , DNA Damage/genetics , Humans , Neoplastic Stem Cells/physiology
12.
Bioinformatics ; 33(13): 2050-2052, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28203714

ABSTRACT

MOTIVATION: graphics for RNA-sequencing and microarray gene expression analyses may contain upwards of tens of thousands of points. Details about certain genes or samples of interest are easily obscured in such dense summary displays. Incorporating interactivity into summary plots would enable additional information to be displayed on demand and facilitate intuitive data exploration. RESULTS: The open-source Glimma package creates interactive graphics for exploring gene expression analysis with a few simple R commands. It extends popular plots found in the limma package, such as multi-dimensional scaling plots and mean-difference plots, to allow individual data points to be queried and additional annotation information to be displayed upon hovering or selecting particular points. It also offers links between plots so that more information can be revealed on demand. Glimma is widely applicable, supporting data analyses from a number of well-established Bioconductor workflows ( limma , edgeR and DESeq2 ) and uses D3/JavaScript to produce HTML pages with interactive displays that enable more effective data exploration by end-users. Results from Glimma can be easily shared between bioinformaticians and biologists, enhancing reporting capabilities while maintaining reproducibility. AVAILABILITY AND IMPLEMENTATION: The Glimma R package is available from http://bioconductor.org/packages/Glimma/ . CONTACT: su.s@wehi.edu.au , law@wehi.edu.au or mritchie@wehi.edu.au.


Subject(s)
Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Software , Animals , Mice
13.
Nucleic Acids Res ; 43(15): e97, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-25925576

ABSTRACT

Variations in sample quality are frequently encountered in small RNA-sequencing experiments, and pose a major challenge in a differential expression analysis. Removal of high variation samples reduces noise, but at a cost of reducing power, thus limiting our ability to detect biologically meaningful changes. Similarly, retaining these samples in the analysis may not reveal any statistically significant changes due to the higher noise level. A compromise is to use all available data, but to down-weight the observations from more variable samples. We describe a statistical approach that facilitates this by modelling heterogeneity at both the sample and observational levels as part of the differential expression analysis. At the sample level this is achieved by fitting a log-linear variance model that includes common sample-specific or group-specific parameters that are shared between genes. The estimated sample variance factors are then converted to weights and combined with observational level weights obtained from the mean-variance relationship of the log-counts-per-million using 'voom'. A comprehensive analysis involving both simulations and experimental RNA-sequencing data demonstrates that this strategy leads to a universally more powerful analysis and fewer false discoveries when compared to conventional approaches. This methodology has wide application and is implemented in the open-source 'limma' package.


Subject(s)
Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Animals , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Humans , Linear Models , Mice , Reproducibility of Results
14.
Nature ; 465(7299): 798-802, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20383121

ABSTRACT

The ovarian hormones oestrogen and progesterone profoundly influence breast cancer risk, underpinning the benefit of endocrine therapies in the treatment of breast cancer. Modulation of their effects through ovarian ablation or chemoprevention strategies also significantly decreases breast cancer incidence. Conversely, there is an increased risk of breast cancer associated with pregnancy in the short term. The cellular mechanisms underlying these observations, however, are poorly defined. Here we demonstrate that mouse mammary stem cells (MaSCs) are highly responsive to steroid hormone signalling, despite lacking the oestrogen and progesterone receptors. Ovariectomy markedly diminished MaSC number and outgrowth potential in vivo, whereas MaSC activity increased in mice treated with oestrogen plus progesterone. Notably, even three weeks of treatment with the aromatase inhibitor letrozole was sufficient to reduce the MaSC pool. In contrast, pregnancy led to a transient 11-fold increase in MaSC numbers, probably mediated through paracrine signalling from RANK ligand. The augmented MaSC pool indicates a cellular basis for the short-term increase in breast cancer incidence that accompanies pregnancy. These findings further indicate that breast cancer chemoprevention may be achieved, in part, through suppression of MaSC function.


Subject(s)
Estrogens/metabolism , Mammary Glands, Animal/cytology , Progesterone/metabolism , Stem Cells/cytology , Animals , Aromatase/metabolism , Aromatase Inhibitors/pharmacology , CD24 Antigen/metabolism , Cell Count , ErbB Receptors/metabolism , Estrogens/pharmacology , Female , Humans , Integrin beta1/metabolism , Integrin beta3/metabolism , Letrozole , Mice , Nitriles/pharmacology , Ovariectomy , Paracrine Communication/drug effects , Pregnancy , Pregnancy, Animal/physiology , Progesterone/antagonists & inhibitors , Progesterone/pharmacology , RANK Ligand/metabolism , Receptors, Estrogen/deficiency , Receptors, Progesterone/deficiency , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Triazoles/pharmacology
15.
Respir Res ; 16: 67, 2015 Jun 07.
Article in English | MEDLINE | ID: mdl-26048572

ABSTRACT

BACKGROUND: The lung is constantly exposed to environmental challenges and must rapidly respond to external insults. Mechanisms involved in the repair of the damaged lung involve expansion of different epithelial cells to repopulate the injured cellular compartment. However, factors regulating cell proliferation following lung injury remain poorly understood. Here we studied the role of the transcriptional regulator Lmo4 during lung development, in the regulation of adult lung epithelial cell proliferation following lung damage and in the context of oncogenic transformation. METHODS: To study the role of Lmo4 in embryonic lung development, lung repair and tumorigenesis, we used conditional knock-out mice to delete Lmo4 in lung epithelial cells from the first stages of lung development. The role of Lmo4 in lung repair was evaluated using two experimental models of lung damage involving chemical and viral injury. The role of Lmo4 in lung tumorigenesis was measured using a mouse model of lung adenocarcinoma in which the oncogenic K-Ras protein has been knocked into the K-Ras locus. Overall survival difference between genotypes was tested by log rank test. Difference between means was tested using one-way ANOVA after assuring that assumptions of normality and equality of variance were satisfied. RESULTS: We found that Lmo4 was not required for normal embryonic lung morphogenesis. In the adult lung, loss of Lmo4 reduced epithelial cell proliferation and delayed repair of the lung following naphthalene or flu-mediated injury, suggesting that Lmo4 participates in the regulation of epithelial cell expansion in response to cellular damage. In the context of K-Ras(G12D)-driven lung tumor formation, Lmo4 loss did not alter overall survival but delayed initiation of lung hyperplasia in K-Ras(G12D) mice sensitized by naphthalene injury. Finally, we evaluated the expression of LMO4 in tissue microarrays of early stage non-small cell lung cancer and observed that LMO4 is more highly expressed in lung squamous cell carcinoma compared to adenocarcinoma. CONCLUSIONS: Together these results show that the transcriptional regulator Lmo4 participates in the regulation of lung epithelial cell proliferation in the context of injury and oncogenic transformation but that Lmo4 depletion is not sufficient to prevent lung repair or tumour formation.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Cell Proliferation/physiology , Disease Progression , LIM Domain Proteins/deficiency , Lung Neoplasms/metabolism , Lung/metabolism , Respiratory Mucosa/metabolism , Animals , Humans , Lung/pathology , Lung Neoplasms/pathology , Mice , Mice, Knockout , Respiratory Mucosa/pathology
16.
Nat Cell Biol ; 9(2): 201-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17187062

ABSTRACT

The transcription factor Gata-3 is a defining marker of the 'luminal' subtypes of breast cancer. To gain insight into the role of Gata-3 in breast epithelial development and oncogenesis, we have explored its normal function within the mammary gland by conditionally deleting Gata-3 at different stages of development. We report that Gata-3 has essential roles in the morphogenesis of the mammary gland in both the embryo and adult. Through the discovery of a novel marker (beta3-integrin) of luminal progenitor cells and their purification, we demonstrate that Gata-3 deficiency leads to an expansion of luminal progenitors and a concomitant block in differentiation. Remarkably, introduction of Gata-3 into a stem cell-enriched population induced maturation along the alveolar luminal lineage. These studies provide evidence for the existence of an epithelial hierarchy within the mammary gland and establish Gata-3 as a critical regulator of luminal differentiation.


Subject(s)
GATA3 Transcription Factor/metabolism , Mammary Glands, Animal/metabolism , Morphogenesis/drug effects , Animals , Cell Differentiation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , GATA3 Transcription Factor/deficiency , GATA3 Transcription Factor/pharmacology , Integrin beta3/biosynthesis , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/growth & development , Mice , Pregnancy , Stem Cells/metabolism
17.
Genome Biol ; 25(1): 99, 2024 04 18.
Article in English | MEDLINE | ID: mdl-38637899

ABSTRACT

Spatial molecular data has transformed the study of disease microenvironments, though, larger datasets pose an analytics challenge prompting the direct adoption of single-cell RNA-sequencing tools including normalization methods. Here, we demonstrate that library size is associated with tissue structure and that normalizing these effects out using commonly applied scRNA-seq normalization methods will negatively affect spatial domain identification. Spatial data should not be specifically corrected for library size prior to analysis, and algorithms designed for scRNA-seq data should be adopted with caution.


Subject(s)
Gene Expression Profiling , Single-Cell Analysis , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Gene Expression Profiling/methods , Algorithms , Biology
18.
Am J Pathol ; 181(6): 2225-38, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23063660

ABSTRACT

Despite the key roles of lymphatic vessels in homeostasis and disease, the cellular sources of signals that direct lymphatic vascular growth and patterning remain unknown. Using high-resolution imaging in two and three dimensions, we demonstrated that postnatal mouse mammary gland lymphatic vessels share an intimate spatial association with epithelial ducts and large blood vessels. We further demonstrated that the lymphatic vasculature is remodeled together with the mammary epithelial tree and blood vasculature during postnatal mouse mammary gland morphogenesis. Neither estrogen receptor α nor progesterone receptor were detected in lymphatic endothelial cells in the mouse mammary gland, suggesting that mammary gland lymphangiogenesis is not likely regulated directly by these steroid hormones. Epithelial cells, especially myoepithelial cells, were determined to be a rich source of prolymphangiogenic stimuli including VEGF-C and VEGF-D with temporally regulated expression levels during mammary gland morphogenesis. Blockade of VEGFR-3 signaling using a small-molecule inhibitor inhibited the proliferation of primary lymphatic endothelial cells promoted by mammary gland conditioned medium, suggesting that lymphangiogenesis in the mammary gland is likely driven by myoepithelial-derived VEGF-C and/or VEGF-D. These findings provide new insight into the architecture of the lymphatic vasculature in the mouse mammary gland and, by uncovering the proximity of lymphatic vessels to the epithelial tree, suggest a potential mechanism by which metastatic tumor cells access the lymphatic vasculature.


Subject(s)
Epithelium/growth & development , Epithelium/metabolism , Lymphangiogenesis , Lymphatic Vessels/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Animals , Animals, Newborn , Blood Vessels/cytology , Blood Vessels/growth & development , Blood Vessels/metabolism , Cell Proliferation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Developmental , Lymphatic Vessels/cytology , Mammary Glands, Animal/cytology , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Progesterone/metabolism , Time Factors , Vascular Endothelial Growth Factor D/genetics , Vascular Endothelial Growth Factor D/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
19.
Front Oncol ; 13: 1150349, 2023.
Article in English | MEDLINE | ID: mdl-36994206

ABSTRACT

Introduction: Tumour mutational burden (TMB) is an important emerging biomarker for immune checkpoint inhibitors (ICI). The stability of TMB values across distinct EBUS tumour regions is not well defined in advanced lung cancer patients. Methods: This study included a whole-genome sequencing cohort (n=11, LxG cohort) and a targeted Oncomine TML panel cohort (n=10, SxD cohort), where paired primary and metastatic samples were obtained by endobronchial ultrasound transbronchial needle aspiration (EBUS-TBNA). Results: The LxG cohort displayed a strong correlation between the paired primary and metastatic sites, with a median TMB score of 7.70 ± 5.39 and 8.31 ± 5.88 respectively. Evaluation of the SxD cohort demonstrated greater inter-tumoural TMB heterogeneity, where Spearman correlation between the primary and metastatic sites fell short of significance. Whilst median TMB scores were not significantly different between the two sites, 3 out of 10 paired samples were discordant when using a TMB cut-off of 10 mutations per Mb. In addition, PD-L1 copy number and KRAS mutations were assessed, demonstrating the feasibility of performing multiple molecular tests relevant to ICI treatment using a single EBUS sample. We also observed good consistency in PD-L1 copy number and KRAS mutation, where cut-off estimates were consistent across the primary and metastatic sites. Conclusions: Assessment of TMB acquired by EBUS from multiple sites is highly feasible and has the potential to improve accuracy of TMB panels as a companion diagnostic test. We demonstrate similar TMB values across primary and metastatic sites, however 3 out of 10 samples displayed inter-tumoural heterogeneity that would alter clinical management.

20.
Front Oncol ; 13: 1129195, 2023.
Article in English | MEDLINE | ID: mdl-37143952

ABSTRACT

Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer death worldwide. Immunotherapy with immune checkpoint inhibitors (ICI) has significantly improved outcomes in some patients, however 80-85% of patients receiving immunotherapy develop primary resistance, manifesting as a lack of response to therapy. Of those that do have an initial response, disease progression may occur due to acquired resistance. The make-up of the tumour microenvironment (TME) and the interaction between tumour infiltrating immune cells and cancer cells can have a large impact on the response to immunotherapy. Robust assessment of the TME with accurate and reproducible methods is vital to understanding mechanisms of immunotherapy resistance. In this paper we will review the evidence of several methodologies to assess the TME, including multiplex immunohistochemistry, imaging mass cytometry, flow cytometry, mass cytometry and RNA sequencing.

SELECTION OF CITATIONS
SEARCH DETAIL