Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 14(6): 953-964, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38501975

ABSTRACT

Pediatric cancers are rare diseases, and children without known germline predisposing conditions who develop a second malignancy during developmental ages are extremely rare. We present four such clinical cases and, through whole-genome and error-correcting ultra-deep duplex sequencing of tumor and normal samples, we explored the origin of the second malignancy in four children, uncovering different routes of development. The exposure to cytotoxic therapies was linked to the emergence of a secondary acute myeloid leukemia. A common somatic mutation acquired early during embryonic development was the driver of two solid malignancies in another child. In two cases, the two tumors developed from completely independent clones diverging during embryogenesis. Importantly, we demonstrate that platinum-based therapies contributed at least one order of magnitude more mutations per day of exposure than aging to normal tissues in these children. SIGNIFICANCE: Using whole-genome and error-correcting ultra-deep duplex sequencing, we uncover different origins for second neoplasms in four children. We also uncover the presence of platinum-related mutations across 10 normal tissues of exposed individuals, highlighting the impact that the use of cytotoxic therapies may have on cancer survivors. See related commentary by Pacyna and Nangalia, p. 900. This article is featured in Selected Articles from This Issue, p. 897.


Subject(s)
Mutation , Neoplasms, Second Primary , Humans , Child , Male , Neoplasms, Second Primary/genetics , Female , Child, Preschool , Adolescent , Antineoplastic Agents/therapeutic use , Whole Genome Sequencing , High-Throughput Nucleotide Sequencing , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Infant
2.
Nat Cell Biol ; 25(12): 1833-1847, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37945904

ABSTRACT

MAF amplification increases the risk of breast cancer (BCa) metastasis through mechanisms that are still poorly understood yet have important clinical implications. Oestrogen-receptor-positive (ER+) BCa requires oestrogen for both growth and metastasis, albeit by ill-known mechanisms. Here we integrate proteomics, transcriptomics, epigenomics, chromatin accessibility and functional assays from human and syngeneic mouse BCa models to show that MAF directly interacts with oestrogen receptor alpha (ERα), thereby promoting a unique chromatin landscape that favours metastatic spread. We identify metastasis-promoting genes that are de novo licensed following oestrogen exposure in a MAF-dependent manner. The histone demethylase KDM1A is key to the epigenomic remodelling that facilitates the expression of the pro-metastatic MAF/oestrogen-driven gene expression program, and loss of KDM1A activity prevents this metastasis. We have thus determined that the molecular basis underlying MAF/oestrogen-mediated metastasis requires genetic, epigenetic and hormone signals from the systemic environment, which influence the ability of BCa cells to metastasize.


Subject(s)
Breast Neoplasms , Epigenesis, Genetic , Estrogen Receptor alpha , Gene Amplification , Proto-Oncogene Proteins c-maf , Animals , Female , Humans , Mice , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens , Histone Demethylases/genetics , Histone Demethylases/metabolism , Proto-Oncogene Proteins c-maf/genetics
3.
Cancer Res ; 82(16): 2904-2917, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35749591

ABSTRACT

Immune-checkpoint blockade (ICB) promotes antitumor immune responses and can result in durable patient benefit. However, response rates in breast cancer patients remain modest, stimulating efforts to discover novel treatment options. Cancer-associated fibroblasts (CAF) represent a major component of the breast tumor microenvironment and have known immunosuppressive functions in addition to their well-established roles in directly promoting tumor growth and metastasis. Here we utilized paired syngeneic mouse mammary carcinoma models to show that CAF abundance is associated with insensitivity to combination αCTLA4 and αPD-L1 ICB. CAF-rich tumors exhibited an immunologically cold tumor microenvironment, with transcriptomic, flow cytometric, and quantitative histopathologic analyses demonstrating a relationship between CAF density and a CD8+ T-cell-excluded tumor phenotype. The CAF receptor Endo180 (Mrc2) is predominantly expressed on myofibroblastic CAFs, and its genetic deletion depleted a subset of αSMA-expressing CAFs and impaired tumor progression in vivo. The addition of wild-type, but not Endo180-deficient, CAFs in coimplantation studies restricted CD8+ T-cell intratumoral infiltration, and tumors in Endo180 knockout mice exhibited increased CD8+ T-cell infiltration and enhanced sensitivity to ICB compared with tumors in wild-type mice. Clinically, in a trial of melanoma patients, high MRC2 mRNA levels in tumors were associated with a poor response to αPD-1 therapy, highlighting the potential benefits of therapeutically targeting a specific CAF subpopulation in breast and other CAF-rich cancers to improve clinical responses to immunotherapy. SIGNIFICANCE: Paired syngeneic models help unravel the interplay between CAF and tumor immune evasion, highlighting the benefits of targeting fibroblast subpopulations to improve clinical responses to immunotherapy.


Subject(s)
Cancer-Associated Fibroblasts , Neoplasms , Animals , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Immune Checkpoint Inhibitors , Mice , Tumor Microenvironment
4.
Cell Stem Cell ; 28(10): 1790-1804.e8, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34010627

ABSTRACT

The role of heterochromatin in cell fate specification during development is unclear. We demonstrate that loss of the lysine 9 of histone H3 (H3K9) methyltransferase G9a in the mammary epithelium results in de novo chromatin opening, aberrant formation of the mammary ductal tree, impaired stem cell potential, disrupted intraductal polarity, and loss of tissue function. G9a loss derepresses long terminal repeat (LTR) retroviral sequences (predominantly the ERVK family). Transcriptionally activated endogenous retroviruses generate double-stranded DNA (dsDNA) that triggers an antiviral innate immune response, and knockdown of the cytosolic dsDNA sensor Aim2 in G9a knockout (G9acKO) mammary epithelium rescues mammary ductal invasion. Mammary stem cell transplantation into immunocompromised or G9acKO-conditioned hosts shows partial dependence of the G9acKO mammary morphological defects on the inflammatory milieu of the host mammary fat pad. Thus, altering the chromatin accessibility of retroviral elements disrupts mammary gland development and stem cell activity through both cell-autonomous and non-autonomous mechanisms.


Subject(s)
Endogenous Retroviruses , Histone-Lysine N-Methyltransferase , Mammary Glands, Animal/growth & development , Adipose Tissue/growth & development , Adipose Tissue/immunology , Animals , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Female , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Immunity , Mammary Glands, Animal/immunology
5.
Cell Stem Cell ; 28(4): 600-602, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33798421

ABSTRACT

COVID-19 has unfortunately halted lab work, conferences, and in-person networking, which is especially detrimental to researchers just starting their labs. Through social media and our reviewer networks, we met some early-career stem cell investigators impacted by the closures. Here, they introduce themselves and their research to our readers.


Subject(s)
Research Personnel , Stem Cells , COVID-19 , Humans
6.
Nat Cell Biol ; 20(12): 1400-1409, 2018 12.
Article in English | MEDLINE | ID: mdl-30455462

ABSTRACT

Mutations in, and the altered expression of, epigenetic modifiers are pervasive in human tumours, making epigenetic factors attractive antitumour targets. The open-versus-closed chromatin state within the cells-of-origin of cancer correlates with the uneven distribution of mutations. However, the long-term effect of targeting epigenetic modifiers on mutability in patients with cancer is unclear. Here, we increased chromatin accessibility by deleting the histone H3 lysine 9 (H3K9) methyltransferase G9a in murine epidermis and show that this does not alter the single nucleotide variant burden or global genomic distribution in chemical mutagen-induced squamous tumours. G9a-depleted tumours develop after a prolonged latency compared with their wild-type counterparts, but are more aggressive and have an expanded cancer progenitor pool, pronounced genomic instability and frequent loss-of-function p53 mutations. Thus, we call for caution when assessing long-term therapeutic benefits of chromatin modifier inhibitors, which may promote more aggressive disease.


Subject(s)
Chromatin/genetics , Genomic Instability , Histone-Lysine N-Methyltransferase/genetics , Mutation , Skin Neoplasms/genetics , Animals , Cell Line , Chromatin/metabolism , Epidermis/metabolism , Epidermis/pathology , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Invasiveness , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Nat Commun ; 9(1): 1420, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29650963

ABSTRACT

The chronic skin inflammation psoriasis is crucially dependent on the IL-23/IL-17 cytokine axis. Although IL-23 is expressed by psoriatic keratinocytes and immune cells, only the immune cell-derived IL-23 is believed to be disease relevant. Here we use a genetic mouse model to show that keratinocyte-produced IL-23 is sufficient to cause a chronic skin inflammation with an IL-17 profile. Furthermore, we reveal a cell-autonomous nuclear function for the actin polymerizing molecule N-WASP, which controls IL-23 expression in keratinocytes by regulating the degradation of the histone methyltransferases G9a and GLP, and H3K9 dimethylation of the IL-23 promoter. This mechanism mediates the induction of IL-23 by TNF, a known inducer of IL-23 in psoriasis. Finally, in keratinocytes of psoriatic lesions a decrease in H3K9 dimethylation correlates with increased IL-23 expression, suggesting relevance for disease. Taken together, our data describe a molecular pathway where epigenetic regulation of keratinocytes can contribute to chronic skin inflammation.


Subject(s)
Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/genetics , Interleukin-23 Subunit p19/genetics , Psoriasis/genetics , Skin/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , Adult , Animals , Disease Models, Animal , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Humans , Inflammation , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-23 Subunit p19/deficiency , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Mice , Mice, Knockout , Middle Aged , Primary Cell Culture , Promoter Regions, Genetic , Psoriasis/metabolism , Psoriasis/pathology , Signal Transduction , Skin/pathology , Wiskott-Aldrich Syndrome Protein, Neuronal/deficiency
8.
Elife ; 62017 04 20.
Article in English | MEDLINE | ID: mdl-28425913

ABSTRACT

The DNA methyltransferase Dnmt3a suppresses tumorigenesis in models of leukemia and lung cancer. Conversely, deregulation of Dnmt3b is thought to generally promote tumorigenesis. However, the role of Dnmt3a and Dnmt3b in many types of cancer remains undefined. Here, we show that Dnmt3a and Dnmt3b are dispensable for homeostasis of the murine epidermis. However, loss of Dnmt3a-but not Dnmt3b-increases the number of carcinogen-induced squamous tumors, without affecting tumor progression. Only upon combined deletion of Dnmt3a and Dnmt3b, squamous carcinomas become more aggressive and metastatic. Mechanistically, Dnmt3a promotes the expression of epidermal differentiation genes by interacting with their enhancers and inhibits the expression of lipid metabolism genes, including PPAR-γ, by directly methylating their promoters. Importantly, inhibition of PPAR-γ partially prevents the increase in tumorigenesis upon deletion of Dnmt3a. Altogether, we demonstrate that Dnmt3a and Dnmt3b protect the epidermis from tumorigenesis and that squamous carcinomas are sensitive to inhibition of PPAR-γ.


Subject(s)
Carcinoma, Squamous Cell/physiopathology , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epidermis/physiology , Homeostasis , PPAR gamma/metabolism , Animals , DNA Methyltransferase 3A , Mice , DNA Methyltransferase 3B
9.
Nature ; 541(7635): 41-45, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27974793

ABSTRACT

The fact that the identity of the cells that initiate metastasis in most human cancers is unknown hampers the development of antimetastatic therapies. Here we describe a subpopulation of CD44bright cells in human oral carcinomas that do not overexpress mesenchymal genes, are slow-cycling, express high levels of the fatty acid receptor CD36 and lipid metabolism genes, and are unique in their ability to initiate metastasis. Palmitic acid or a high-fat diet specifically boosts the metastatic potential of CD36+ metastasis-initiating cells in a CD36-dependent manner. The use of neutralizing antibodies to block CD36 causes almost complete inhibition of metastasis in immunodeficient or immunocompetent orthotopic mouse models of human oral cancer, with no side effects. Clinically, the presence of CD36+ metastasis-initiating cells correlates with a poor prognosis for numerous types of carcinomas, and inhibition of CD36 also impairs metastasis, at least in human melanoma- and breast cancer-derived tumours. Together, our results indicate that metastasis-initiating cells particularly rely on dietary lipids to promote metastasis.


Subject(s)
Antibodies, Neutralizing/pharmacology , CD36 Antigens/antagonists & inhibitors , Mouth Neoplasms/pathology , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , CD36 Antigens/genetics , CD36 Antigens/immunology , CD36 Antigens/metabolism , Cell Proliferation , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Lipid Metabolism/genetics , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Mice , Mouth Neoplasms/diagnosis , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Neoplastic Stem Cells/metabolism , Palmitic Acid/administration & dosage , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Penetrance , Prognosis , Transcriptome , Xenograft Model Antitumor Assays
10.
Cell Stem Cell ; 19(4): 491-501, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27476967

ABSTRACT

The genome-wide localization and function of endogenous Dnmt3a and Dnmt3b in adult stem cells are unknown. Here, we show that in human epidermal stem cells, the two proteins bind in a histone H3K36me3-dependent manner to the most active enhancers and are required to produce their associated enhancer RNAs. Both proteins prefer super-enhancers associated to genes that either define the ectodermal lineage or establish the stem cell and differentiated states. However, Dnmt3a and Dnmt3b differ in their mechanisms of enhancer regulation: Dnmt3a associates with p63 to maintain high levels of DNA hydroxymethylation at the center of enhancers in a Tet2-dependent manner, whereas Dnmt3b promotes DNA methylation along the body of the enhancer. Depletion of either protein inactivates their target enhancers and profoundly affects epidermal stem cell function. Altogether, we reveal novel functions for Dnmt3a and Dnmt3b at enhancers that could contribute to their roles in disease and tumorigenesis.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Enhancer Elements, Genetic/genetics , Epidermal Cells , Homeostasis , Stem Cells/cytology , Stem Cells/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Base Sequence , Cell Differentiation , DNA Methylation/genetics , DNA Methyltransferase 3A , DNA-Binding Proteins/metabolism , Dioxygenases , Histones/metabolism , Humans , Keratinocytes/cytology , Lysine/metabolism , Protein Binding , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , DNA Methyltransferase 3B
11.
Nat Rev Mol Cell Biol ; 17(10): 643-58, 2016 10.
Article in English | MEDLINE | ID: mdl-27405257

ABSTRACT

Mammalian embryonic development is a tightly regulated process that, from a single zygote, produces a large number of cell types with hugely divergent functions. Distinct cellular differentiation programmes are facilitated by tight transcriptional and epigenetic regulation. However, the contribution of epigenetic regulation to tissue homeostasis after the completion of development is less well understood. In this Review, we explore the effects of epigenetic dysregulation on adult stem cell function. We conclude that, depending on the tissue type and the epigenetic regulator affected, the consequences range from negligible to stem cell malfunction and disruption of tissue homeostasis, which may predispose to diseases such as cancer.


Subject(s)
Adult Stem Cells/physiology , Epigenesis, Genetic , Animals , Cell Differentiation , DNA Methylation , Epidermal Cells , Epidermis/physiology , Gene Expression Regulation, Developmental , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/physiology , Regeneration
12.
Curr Opin Genet Dev ; 36: 8-15, 2016 02.
Article in English | MEDLINE | ID: mdl-26874045

ABSTRACT

Cancer stem cells (CSCs) have been identified in various tumours and are defined by their potential to initiate tumours upon transplantation, self-renew and reconstitute tumour heterogeneity. Modifications of the epigenome can favour tumour initiation by affecting genome integrity, DNA repair and tumour cell plasticity. Importantly, an in-depth understanding of the epigenomic alterations underlying neoplastic transformation may open new avenues for chromatin-targeted cancer treatment, as these epigenetic changes could be inherently more amenable to inhibition and reversal than hard-wired genomic alterations. Here we discuss how CSC function is affected by chromatin state and epigenomic instability.


Subject(s)
Chromatin/genetics , Epigenesis, Genetic/genetics , Neoplasms/genetics , Neoplastic Stem Cells , Cell Transformation, Neoplastic/genetics , DNA Repair/genetics , Humans
13.
Nat Commun ; 7: 10305, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26777421

ABSTRACT

Stromal fibroblast recruitment to tumours and activation to a cancer-associated fibroblast (CAF) phenotype has been implicated in promoting primary tumour growth and progression to metastatic disease. However, the mechanisms underlying the tumour:fibroblast crosstalk that drive the intertumoural stromal heterogeneity remain poorly understood. Using in vivo models we identify Wnt7a as a key factor secreted exclusively by aggressive breast tumour cells, which induces CAF conversion. Functionally, this results in extracellular matrix remodelling to create a permissive environment for tumour cell invasion and promotion of distant metastasis. Mechanistically, Wnt7a-mediated fibroblast activation is not dependent on classical Wnt signalling. Instead, we demonstrate that Wnt7a potentiates TGFß receptor signalling both in 3D in vitro and in vivo models, thus highlighting the interaction between two of the key signalling pathways in development and disease. Importantly, in clinical breast cancer cohorts, tumour cell Wnt7a expression correlates with a desmoplastic, poor-prognosis stroma and poor patient outcome.


Subject(s)
Breast Neoplasms/metabolism , Fibroblasts/metabolism , Wnt Proteins/metabolism , Animals , Female , Fibroblasts/cytology , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Wnt Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...