Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; : e2307963, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38602451

ABSTRACT

In recent decades, the role of tumor biomechanics on cancer cell behavior at the primary site has been increasingly appreciated. However, the effect of primary tumor biomechanics on the latter stages of the metastatic cascade, such as metastatic seeding of secondary sites and outgrowth remains underappreciated. This work sought to address this in the context of triple negative breast cancer (TNBC), a cancer type known to aggressively disseminate at all stages of disease progression. Using mechanically tuneable model systems, mimicking the range of stiffness's typically found within breast tumors, it is found that, contrary to expectations, cancer cells exposed to softer microenvironments are more able to colonize secondary tissues. It is shown that heightened cell survival is driven by enhanced metabolism of fatty acids within TNBC cells exposed to softer microenvironments. It is demonstrated that uncoupling cellular mechanosensing through integrin ß1 blocking antibody effectively causes stiff primed TNBC cells to behave like their soft counterparts, both in vitro and in vivo. This work is the first to show that softer tumor microenvironments may be contributing to changes in disease outcome by imprinting on TNBC cells a greater metabolic flexibility and conferring discrete cell survival advantages.

2.
Nat Commun ; 13(1): 6539, 2022 11 07.
Article in English | MEDLINE | ID: mdl-36344500

ABSTRACT

Cancers evade the immune system through the process of cancer immunoediting. While immune checkpoint inhibitors are effective for reactivating tumour immunity in some cancer types, many other solid cancers, including breast cancer, remain largely non-responsive. Understanding how non-responsive cancers evade immunity and whether this occurs at the clonal level will improve immunotherapeutic design. Here we use DNA barcoding to track murine mammary cancer cell clones during immunoediting and determine clonal transcriptional profiles that allow immune evasion following anti-PD1 plus anti-CTLA4 immunotherapy. Clonal diversity is significantly restricted by immunotherapy treatment in both primary tumours and metastases, demonstrating selection for pre-existing breast cancer cell populations and ongoing immunoediting during metastasis and treatment. Immunotherapy resistant clones express a common gene signature associated with poor survival of basal-like breast cancer patient cohorts. At least one of these genes has an existing small molecule that can potentially be used to improve immunotherapy response.


Subject(s)
Breast Neoplasms , DNA Barcoding, Taxonomic , Humans , Mice , Animals , Female , Immunotherapy , Immunologic Factors , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Longitudinal Studies
3.
Nat Genet ; 53(9): 1334-1347, 2021 09.
Article in English | MEDLINE | ID: mdl-34493872

ABSTRACT

Breast cancers are complex cellular ecosystems where heterotypic interactions play central roles in disease progression and response to therapy. However, our knowledge of their cellular composition and organization is limited. Here we present a single-cell and spatially resolved transcriptomics analysis of human breast cancers. We developed a single-cell method of intrinsic subtype classification (SCSubtype) to reveal recurrent neoplastic cell heterogeneity. Immunophenotyping using cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) provides high-resolution immune profiles, including new PD-L1/PD-L2+ macrophage populations associated with clinical outcome. Mesenchymal cells displayed diverse functions and cell-surface protein expression through differentiation within three major lineages. Stromal-immune niches were spatially organized in tumors, offering insights into antitumor immune regulation. Using single-cell signatures, we deconvoluted large breast cancer cohorts to stratify them into nine clusters, termed 'ecotypes', with unique cellular compositions and clinical outcomes. This study provides a comprehensive transcriptional atlas of the cellular architecture of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Single-Cell Analysis , Transcriptome/genetics , B-Lymphocytes/immunology , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Breast Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , Endothelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Macrophages/cytology , Macrophages/immunology , Membrane Proteins/genetics , Myeloid Cells/immunology , Myeloid Cells/metabolism , Sequence Analysis, RNA , Tumor Microenvironment , Tumor Suppressor Proteins/genetics
4.
Genome Med ; 13(1): 81, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33971952

ABSTRACT

BACKGROUND: High throughput single-cell RNA sequencing (scRNA-Seq) has emerged as a powerful tool for exploring cellular heterogeneity among complex human cancers. scRNA-Seq studies using fresh human surgical tissue are logistically difficult, preclude histopathological triage of samples, and limit the ability to perform batch processing. This hindrance can often introduce technical biases when integrating patient datasets and increase experimental costs. Although tissue preservation methods have been previously explored to address such issues, it is yet to be examined on complex human tissues, such as solid cancers and on high throughput scRNA-Seq platforms. METHODS: Using the Chromium 10X platform, we sequenced a total of ~ 120,000 cells from fresh and cryopreserved replicates across three primary breast cancers, two primary prostate cancers and a cutaneous melanoma. We performed detailed analyses between cells from each condition to assess the effects of cryopreservation on cellular heterogeneity, cell quality, clustering and the identification of gene ontologies. In addition, we performed single-cell immunophenotyping using CITE-Seq on a single breast cancer sample cryopreserved as solid tissue fragments. RESULTS: Tumour heterogeneity identified from fresh tissues was largely conserved in cryopreserved replicates. We show that sequencing of single cells prepared from cryopreserved tissue fragments or from cryopreserved cell suspensions is comparable to sequenced cells prepared from fresh tissue, with cryopreserved cell suspensions displaying higher correlations with fresh tissue in gene expression. We showed that cryopreservation had minimal impacts on the results of downstream analyses such as biological pathway enrichment. For some tumours, cryopreservation modestly increased cell stress signatures compared to freshly analysed tissue. Further, we demonstrate the advantage of cryopreserving whole-cells for detecting cell-surface proteins using CITE-Seq, which is impossible using other preservation methods such as single nuclei-sequencing. CONCLUSIONS: We show that the viable cryopreservation of human cancers provides high-quality single-cells for multi-omics analysis. Our study guides new experimental designs for tissue biobanking for future clinical single-cell RNA sequencing studies.


Subject(s)
Biological Specimen Banks , Cryopreservation , Genomics , Neoplasms/diagnosis , Single-Cell Analysis , Biomarkers, Tumor , Cryopreservation/methods , Cryopreservation/standards , Gene Expression Profiling/methods , Gene Expression Regulation , Genomics/methods , High-Throughput Nucleotide Sequencing , Humans , Immunophenotyping , Neoplasms/etiology , Organ Specificity/genetics , Sequence Analysis, RNA/methods , Signal Transduction , Single-Cell Analysis/methods
5.
iScience ; 24(2): 102072, 2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33554073

ABSTRACT

Inhibitor of differentiation (ID) proteins dimerize with basic HLH (bHLH) transcription factors, repressing transcription of lineage-specification genes across diverse cellular lineages. ID4 is a key regulator of mammary stem cells; however, the mechanism by which it achieves this is unclear. Here, we show that ID4 has a cell autonomous role in preventing myoepithelial differentiation of basal cells in mammary organoids and in vivo. ID4 positively regulates proliferative genes and negatively regulates genes involved in myoepithelial function. Mass spectrometry reveals that ID4 interacts with the bHLH protein HEB, which binds to E-box motifs in regulatory elements of basal developmental genes involved in extracellular matrix and the contractile cytoskeleton. We conclude that high ID4 expression in mammary basal stem cells antagonizes HEB transcriptional activity, preventing myoepithelial differentiation and allowing for appropriate tissue morphogenesis. Downregulation of ID4 during pregnancy modulates gene regulated by HEB, promoting specialization of basal cells into myoepithelial cells.

6.
Front Cell Dev Biol ; 8: 552, 2020.
Article in English | MEDLINE | ID: mdl-32766238

ABSTRACT

Breast cancers display phenotypic and functional heterogeneity and several lines of evidence support the existence of cancer stem cells (CSCs) in certain breast cancers, a minor population of cells capable of tumor initiation and metastatic dissemination. Identifying factors that regulate the CSC phenotype is therefore important for developing strategies to treat metastatic disease. The Inhibitor of Differentiation Protein 1 (Id1) and its closely related family member Inhibitor of Differentiation 3 (Id3) (collectively termed Id) are expressed by a diversity of stem cells and are required for metastatic dissemination in experimental models of breast cancer. In this study, we show that ID1 is expressed in rare neoplastic cells within ER-negative breast cancers. To address the function of Id1 expressing cells within tumors, we developed independent murine models of Triple Negative Breast Cancer (TNBC) in which a genetic reporter permitted the prospective isolation of Id1+ cells. Id1+ cells are enriched for self-renewal in tumorsphere assays in vitro and for tumor initiation in vivo. Conversely, depletion of Id1 and Id3 in the 4T1 murine model of TNBC demonstrates that Id1/3 are required for cell proliferation and self-renewal in vitro, as well as primary tumor growth and metastatic colonization of the lung in vivo. Using combined bioinformatic analysis, we have defined a novel mechanism of Id protein function via negative regulation of the Roundabout Axon Guidance Receptor Homolog 1 (Robo1) leading to activation of a Myc transcriptional programme.

7.
EMBO J ; 39(19): e104063, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32790115

ABSTRACT

The tumour stroma regulates nearly all stages of carcinogenesis. Stromal heterogeneity in human triple-negative breast cancers (TNBCs) remains poorly understood, limiting the development of stromal-targeted therapies. Single-cell RNA sequencing of five TNBCs revealed two cancer-associated fibroblast (CAF) and two perivascular-like (PVL) subpopulations. CAFs clustered into two states: the first with features of myofibroblasts and the second characterised by high expression of growth factors and immunomodulatory molecules. PVL cells clustered into two states consistent with a differentiated and immature phenotype. We showed that these stromal states have distinct morphologies, spatial relationships and functional properties in regulating the extracellular matrix. Using cell signalling predictions, we provide evidence that stromal-immune crosstalk acts via a diverse array of immunoregulatory molecules. Importantly, the investigation of gene signatures from inflammatory-CAFs and differentiated-PVL cells in independent TNBC patient cohorts revealed strong associations with cytotoxic T-cell dysfunction and exclusion, respectively. Such insights present promising candidates to further investigate for new therapeutic strategies in the treatment of TNBCs.


Subject(s)
Triple Negative Breast Neoplasms/immunology , Tumor Escape , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Female , Humans , RNA-Seq , Stromal Cells/immunology , Stromal Cells/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Triple Negative Breast Neoplasms/pathology
8.
Science ; 368(6495): 1127-1131, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32499442

ABSTRACT

In microorganisms, evolutionarily conserved mechanisms facilitate adaptation to harsh conditions through stress-induced mutagenesis (SIM). Analogous processes may underpin progression and therapeutic failure in human cancer. We describe SIM in multiple in vitro and in vivo models of human cancers under nongenotoxic drug selection, paradoxically enhancing adaptation at a competing intrinsic fitness cost. A genome-wide approach identified the mechanistic target of rapamycin (MTOR) as a stress-sensing rheostat mediating SIM across multiple cancer types and conditions. These observations are consistent with a two-phase model for drug resistance, in which an initially rapid expansion of genetic diversity is counterbalanced by an intrinsic fitness penalty, subsequently normalizing to complete adaptation under the new conditions. This model suggests synthetic lethal strategies to minimize resistance to anticancer therapy.


Subject(s)
Adaptation, Physiological/genetics , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Mutagenesis , Neoplasms/drug therapy , Neoplasms/genetics , TOR Serine-Threonine Kinases/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , DNA Repair/genetics , Genetic Fitness , Genome-Wide Association Study , Humans , Selection, Genetic , Signal Transduction , TOR Serine-Threonine Kinases/genetics
9.
Breast Cancer Res ; 22(1): 63, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32527287

ABSTRACT

BACKGROUND: Basal-like breast cancer (BLBC) is a poorly characterised, heterogeneous disease. Patients are diagnosed with aggressive, high-grade tumours and often relapse with chemotherapy resistance. Detailed understanding of the molecular underpinnings of this disease is essential to the development of personalised therapeutic strategies. Inhibitor of differentiation 4 (ID4) is a helix-loop-helix transcriptional regulator required for mammary gland development. ID4 is overexpressed in a subset of BLBC patients, associating with a stem-like poor prognosis phenotype, and is necessary for the growth of cell line models of BLBC through unknown mechanisms. METHODS: Here, we have defined unique molecular insights into the function of ID4 in BLBC and the related disease high-grade serous ovarian cancer (HGSOC), by combining RIME proteomic analysis, ChIP-seq mapping of genomic binding sites and RNA-seq. RESULTS: These studies reveal novel interactions with DNA damage response proteins, in particular, mediator of DNA damage checkpoint protein 1 (MDC1). Through MDC1, ID4 interacts with other DNA repair proteins (γH2AX and BRCA1) at fragile chromatin sites. ID4 does not affect transcription at these sites, instead binding to chromatin following DNA damage. Analysis of clinical samples demonstrates that ID4 is amplified and overexpressed at a higher frequency in BRCA1-mutant BLBC compared with sporadic BLBC, providing genetic evidence for an interaction between ID4 and DNA damage repair deficiency. CONCLUSIONS: These data link the interactions of ID4 with MDC1 to DNA damage repair in the aetiology of BLBC and HGSOC.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Animals , Apoptosis/physiology , Breast Neoplasms/pathology , Carcinoma, Basal Cell/pathology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Chromatin/genetics , Chromatin/metabolism , DNA Damage , Female , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Prognosis , Proteogenomics , Tumor Cells, Cultured
10.
Nat Commun ; 10(1): 3120, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31311926

ABSTRACT

High-throughput single-cell RNA sequencing is a powerful technique but only generates short reads from one end of a cDNA template, limiting the reconstruction of highly diverse sequences such as antigen receptors. To overcome this limitation, we combined targeted capture and long-read sequencing of T-cell-receptor (TCR) and B-cell-receptor (BCR) mRNA transcripts with short-read transcriptome profiling of barcoded single-cell libraries generated by droplet-based partitioning. We show that Repertoire and Gene Expression by Sequencing (RAGE-Seq) can generate accurate full-length antigen receptor sequences at nucleotide resolution, infer B-cell clonal evolution and identify alternatively spliced BCR transcripts. We apply RAGE-Seq to 7138 cells sampled from the primary tumor and draining lymph node of a breast cancer patient to track transcriptome profiles of expanded lymphocyte clones across tissues. Our results demonstrate that RAGE-Seq is a powerful method for tracking the clonal evolution from large numbers of lymphocytes applicable to the study of immunity, autoimmunity and cancer.


Subject(s)
Clonal Evolution/genetics , Lymphocytes/metabolism , Single-Cell Analysis/methods , Clonal Evolution/immunology , Gene Expression Profiling , High-Throughput Nucleotide Sequencing/methods , Humans , Primary Cell Culture , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Sequence Analysis, RNA/methods
11.
Breast Cancer Res ; 20(1): 100, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30176939

ABSTRACT

Differentiation of stem cells into highly specialised cells requires gene expression changes brought about by remodelling of the chromatin architecture. During this lineage-commitment process, the majority of DNA needs to be packaged into inactive heterochromatin, allowing only a subset of regulatory elements to remain open and functionally required genes to be expressed. Epigenetic mechanisms such as DNA methylation, post-translational modifications to histone tails, and nucleosome positioning all potentially contribute to the changes in higher order chromatin structure during differentiation. The mammary gland is a particularly useful model to study these complex epigenetic processes since the majority of its development is postnatal, the gland is easily accessible, and development occurs in a highly reproducible manner. Inappropriate epigenetic remodelling can also drive tumourigenesis; thus, insights into epigenetic remodelling during mammary gland development advance our understanding of breast cancer aetiology. We review the current literature surrounding DNA methylation and histone modifications in the developing mammary gland and its implications for breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast/growth & development , Carcinogenesis/genetics , Epigenesis, Genetic/physiology , Animals , Breast/pathology , Breast Neoplasms/pathology , Cell Differentiation/genetics , Chromatin/metabolism , DNA Methylation/physiology , Female , Histone Code/physiology , Histones/metabolism , Humans , Mammary Glands, Animal/growth & development , Models, Animal , Stem Cells/physiology
12.
PLoS Genet ; 13(11): e1007072, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29117179

ABSTRACT

We identified a non-synonymous mutation in Oas2 (I405N), a sensor of viral double-stranded RNA, from an ENU-mutagenesis screen designed to discover new genes involved in mammary development. The mutation caused post-partum failure of lactation in healthy mice with otherwise normally developed mammary glands, characterized by greatly reduced milk protein synthesis coupled with epithelial cell death, inhibition of proliferation and a robust interferon response. Expression of mutant but not wild type Oas2 in cultured HC-11 or T47D mammary cells recapitulated the phenotypic and transcriptional effects observed in the mouse. The mutation activates the OAS2 pathway, demonstrated by a 34-fold increase in RNase L activity, and its effects were dependent on expression of RNase L and IRF7, proximal and distal pathway members. This is the first report of a viral recognition pathway regulating lactation.


Subject(s)
2',5'-Oligoadenylate Synthetase/genetics , Lactation/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Adenine Nucleotides/metabolism , Animals , Cell Culture Techniques , Endoribonucleases/metabolism , Female , Humans , Mammary Glands, Animal/metabolism , Mice , Milk , Mutation/genetics , Oligoribonucleotides/metabolism , RNA, Double-Stranded/metabolism , Signal Transduction/genetics
13.
Nat Commun ; 6: 6548, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25813983

ABSTRACT

Basal-like breast cancer (BLBC) is a heterogeneous disease with poor prognosis; however, its cellular origins and aetiology are poorly understood. In this study, we show that inhibitor of differentiation 4 (ID4) is a key regulator of mammary stem cell self-renewal and marks a subset of BLBC with a putative mammary basal cell of origin. Using an ID4GFP knock-in reporter mouse and single-cell transcriptomics, we show that ID4 marks a stem cell-enriched subset of the mammary basal cell population. ID4 maintains the mammary stem cell pool by suppressing key factors required for luminal differentiation. Furthermore, ID4 is specifically expressed by a subset of human BLBC that possess a very poor prognosis and a transcriptional signature similar to a mammary stem cell. These studies identify ID4 as a mammary stem cell regulator, deconvolute the heterogeneity of BLBC and link a subset of mammary stem cells to the aetiology of BLBC.


Subject(s)
Breast Neoplasms/genetics , Inhibitor of Differentiation Proteins/genetics , Mammary Glands, Animal/cytology , RNA, Messenger/metabolism , Stem Cells/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Knock-In Techniques , Humans , Inhibitor of Differentiation Proteins/metabolism , Mammary Glands, Animal/metabolism , Mice , Neoplasm Transplantation , Phenotype , Real-Time Polymerase Chain Reaction
14.
Cancer Discov ; 5(1): 35-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25312016

ABSTRACT

UNLABELLED: Recent clinical trials have shown that bisphosphonate drugs improve breast cancer patient survival independent of their antiresorptive effects on the skeleton. However, because bisphosphonates bind rapidly to bone mineral, the exact mechanisms of their antitumor action, particularly on cells outside of bone, remain unknown. Here, we used real-time intravital two-photon microscopy to show extensive leakage of fluorescent bisphosphonate from the vasculature in 4T1 mouse mammary tumors, where it initially binds to areas of small, granular microcalcifications that are engulfed by tumor-associated macrophages (TAM), but not tumor cells. Importantly, we also observed uptake of radiolabeled bisphosphonate in the primary breast tumor of a patient and showed the resected tumor to be infiltrated with TAMs and to contain similar granular microcalcifications. These data represent the first compelling in vivo evidence that bisphosphonates can target cells in tumors outside the skeleton and that their antitumor activity is likely to be mediated via TAMs. SIGNIFICANCE: Bisphosphonates are assumed to act solely in bone. However, mouse models and clinical trials show that they have surprising antitumor effects outside bone. We provide unequivocal evidence that bisphosphonates target TAMs, but not tumor cells, to exert their extraskeletal effects, offering a rationale for use in patients with early disease.


Subject(s)
Bone Density Conservation Agents/metabolism , Diphosphonates/metabolism , Macrophages/metabolism , Neoplasms/diagnosis , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Animals , Bone Density Conservation Agents/therapeutic use , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Calcinosis , Carbocyanines , Diphosphonates/therapeutic use , Disease Models, Animal , Female , Humans , Macrophages/drug effects , Macrophages/immunology , Mice , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasms/drug therapy , Phagocytosis/immunology , Xenograft Model Antitumor Assays
15.
Oncoimmunology ; 2(8): e25409, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-24083081

ABSTRACT

We have recently been the first to demonstrate that interleukin (IL)-27 protects against the emergence and progression of autochthonous tumors. Accumulating evidence suggests that IL-27 might be uniquely well positioned to amplify beneficial TH1 anticancer immune responses while suppressing the unwanted accumulation of regulatory T cells.

16.
Genes Dev ; 27(19): 2086-98, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24065739

ABSTRACT

Serine and cysteine cathepsin (Cts) proteases are an important class of intracellular and pericellular enzymes mediating multiple aspects of tumor development. Emblematic of these is CtsB, reported to play functionally significant roles during pancreatic islet and mammary carcinogenesis. CtsC, on the other hand, while up-regulated during pancreatic islet carcinogenesis, lacks functional significance in mediating neoplastic progression in that organ. Given that protein expression and enzymatic activity of both CtsB and CtsC are increased in numerous tumors, we sought to understand how tissue specificity might factor into their functional significance. Thus, whereas others have reported that CtsB regulates metastasis of mammary carcinomas, we found that development of squamous carcinomas occurs independently of CtsB. In contrast to these findings, our studies found no significant role for CtsC during mammary carcinogenesis but revealed squamous carcinogenesis to be functionally dependent on CtsC. In this context, dermal/stromal fibroblasts and bone marrow-derived cells expressed increased levels of enzymatically active CtsC that regulated the complexity of infiltrating immune cells in neoplastic skin, development of angiogenic vasculature, and overt squamous cell carcinoma growth. These studies highlight the important contribution of tissue/microenvironment context to solid tumor development and indicate that tissue specificity defines functional significance for these two members of the cysteine protease family.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/physiopathology , Cathepsin C/metabolism , Skin Neoplasms/physiopathology , Animals , Cathepsin B/genetics , Cathepsin B/metabolism , Cathepsin C/genetics , Cell Line, Tumor , Chymases/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Leukocytes/metabolism , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/genetics , Pancreatic Elastase/metabolism
17.
PLoS One ; 8(3): e57469, 2013.
Article in English | MEDLINE | ID: mdl-23554861

ABSTRACT

Interleukin-27 (IL-27) is a pleiotropic cytokine but its immunosuppressive effects predominate during many in vivo immunological challenges. Despite this, evidence from tumor cell line transfer models suggested that IL-27 could promote immune responses in the tumor context. However, the role of IL-27 in immunity against tumors that develop in situ and in tumor immunosurveillance remain undefined. In this study, we demonstrate that tumor development and growth are accelerated in IL-27 receptor α (Il27ra)-deficient mice. Enhanced tumor growth in both carcinogen-induced fibrosarcoma and oncogene-driven mammary carcinoma was associated with decreased interferon-γ production by CD4 and CD8 T cells and increased numbers of regulatory T-cells (Treg). This is the first study to show that IL-27 promotes protective immune responses against endogenous tumors, which is critical as the basis for future development of an IL-27 based therapeutic agent.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interleukins/immunology , Neoplasms, Experimental/immunology , Receptors, Cytokine/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Immune Tolerance/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukins/genetics , Male , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Receptors, Cytokine/genetics , Receptors, Interleukin , Signal Transduction/genetics , T-Lymphocytes, Regulatory/pathology
18.
PLoS One ; 5(8): e11947, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20689821

ABSTRACT

The accumulation of poorly differentiated cells is a hallmark of breast neoplasia and progression. Thus an understanding of the factors controlling mammary differentiation is critical to a proper understanding of breast tumourigenesis. The Inhibitor of Differentiation 1 (Id1) protein has well documented roles in the control of mammary epithelial differentiation and proliferation in vitro and breast cancer progression in vivo. However, it has not been determined whether Id1 expression is sufficient for the inhibition of mammary epithelial differentiation or the promotion of neoplastic transformation in vivo. We now show that Id1 is not commonly expressed by the luminal mammary epithelia, as previously reported. Generation and analysis of a transgenic mouse model of Id1 overexpression in the mammary gland reveals that Id1 is insufficient for neoplastic progression in virgin animals or to prevent terminal differentiation of the luminal epithelia during pregnancy and lactation. Together, these data demonstrate that there is no luminal cell-autonomous role for Id1 in mammary epithelial cell fate determination, ductal morphogenesis and terminal differentiation.


Subject(s)
Gene Expression Regulation, Developmental , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Animals , Cell Differentiation , Female , Lactation/genetics , Lactation/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/physiology , Mice , Mice, Transgenic , Pregnancy
19.
Dis Model Mech ; 3(5-6): 317-32, 2010.
Article in English | MEDLINE | ID: mdl-20223936

ABSTRACT

Innate regulatory networks within organs maintain tissue homeostasis and facilitate rapid responses to damage. We identified a novel pathway regulating vessel stability in tissues that involves matrix metalloproteinase 14 (MMP14) and transforming growth factor beta 1 (TGFbeta(1)). Whereas plasma proteins rapidly extravasate out of vasculature in wild-type mice following acute damage, short-term treatment of mice in vivo with a broad-spectrum metalloproteinase inhibitor, neutralizing antibodies to TGFbeta(1), or an activin-like kinase 5 (ALK5) inhibitor significantly enhanced vessel leakage. By contrast, in a mouse model of age-related dermal fibrosis, where MMP14 activity and TGFbeta bioavailability are chronically elevated, or in mice that ectopically express TGFbeta in the epidermis, cutaneous vessels are resistant to acute leakage. Characteristic responses to tissue damage are reinstated if the fibrotic mice are pretreated with metalloproteinase inhibitors or TGFbeta signaling antagonists. Neoplastic tissues, however, are in a constant state of tissue damage and exhibit altered hemodynamics owing to hyperleaky angiogenic vasculature. In two distinct transgenic mouse tumor models, inhibition of ALK5 further enhanced vascular leakage into the interstitium and facilitated increased delivery of high molecular weight compounds into premalignant tissue and tumors. Taken together, these data define a central pathway involving MMP14 and TGFbeta that mediates vessel stability and vascular response to tissue injury. Antagonists of this pathway could be therapeutically exploited to improve the delivery of therapeutics or molecular contrast agents into tissues where chronic damage or neoplastic disease limits their efficient delivery.


Subject(s)
Blood Vessels/enzymology , Blood Vessels/pathology , Matrix Metalloproteinase 14/metabolism , Transforming Growth Factor beta/metabolism , Aging/pathology , Animals , Fibrillar Collagens/metabolism , Homeostasis , Matrix Metalloproteinase 14/deficiency , Mice , Models, Biological , Mustard Plant , Plant Oils , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Stromal Cells/enzymology , Stromal Cells/pathology , Vascular Resistance
20.
Cancer Cell ; 17(2): 121-34, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20138013

ABSTRACT

Chronically activated leukocytes recruited to premalignant tissues functionally contribute to cancer development; however, mechanisms underlying pro- versus anti-tumor programming of neoplastic tissues by immune cells remain obscure. Using the K14-HPV16 mouse model of squamous carcinogenesis, we report that B cells and humoral immunity foster cancer development by activating Fcgamma receptors (FcgammaRs) on resident and recruited myeloid cells. Stromal accumulation of autoantibodies in premalignant skin, through their interaction with activating FcgammaRs, regulate recruitment, composition, and bioeffector functions of leukocytes in neoplastic tissue, which in turn promote neoplastic progression and subsequent carcinoma development. These findings support a model in which B cells, humoral immunity, and activating FcgammaRs are required for establishing chronic inflammatory programs that promote de novo carcinogenesis.


Subject(s)
B-Lymphocytes/immunology , Carcinoma, Squamous Cell/immunology , Neoplasms, Glandular and Epithelial/immunology , Receptors, IgG/physiology , Animals , B-Lymphocytes/metabolism , CD11b Antigen/metabolism , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Immunity, Humoral/physiology , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Transgenic , Models, Biological , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms, Glandular and Epithelial/blood supply , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neovascularization, Pathologic , Receptors, IgG/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...