Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
Cell ; 186(19): 4007-4037, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37714133

ABSTRACT

The TGF-ß regulatory system plays crucial roles in the preservation of organismal integrity. TGF-ß signaling controls metazoan embryo development, tissue homeostasis, and injury repair through coordinated effects on cell proliferation, phenotypic plasticity, migration, metabolic adaptation, and immune surveillance of multiple cell types in shared ecosystems. Defects of TGF-ß signaling, particularly in epithelial cells, tissue fibroblasts, and immune cells, disrupt immune tolerance, promote inflammation, underlie the pathogenesis of fibrosis and cancer, and contribute to the resistance of these diseases to treatment. Here, we review how TGF-ß coordinates multicellular response programs in health and disease and how this knowledge can be leveraged to develop treatments for diseases of the TGF-ß system.


Subject(s)
Signal Transduction , Transforming Growth Factor beta , Animals , Adaptation, Physiological , Cell Proliferation , Embryonic Development
2.
Nat Rev Mol Cell Biol ; 21(6): 341-352, 2020 06.
Article in English | MEDLINE | ID: mdl-32300252

ABSTRACT

Epithelial-mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by 'the EMT International Association' (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT.


Subject(s)
Biomedical Research/standards , Epithelial-Mesenchymal Transition , Animals , Cell Movement , Cell Plasticity , Consensus , Developmental Biology/standards , Humans , Neoplasms/pathology , Terminology as Topic
3.
Cell ; 168(6): 1101-1113.e13, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283064

ABSTRACT

We molecularly dissected leptomeningeal metastasis, or spread of cancer to the cerebrospinal fluid (CSF), which is a frequent and fatal condition mediated by unknown mechanisms. We selected lung and breast cancer cell lines for the ability to infiltrate and grow in CSF, a remarkably acellular, mitogen-poor metastasis microenvironment. Complement component 3 (C3) was upregulated in four leptomeningeal metastatic models and proved necessary for cancer growth within the leptomeningeal space. In human disease, cancer cells within the CSF produced C3 in correlation with clinical course. C3 expression in primary tumors was predictive of leptomeningeal relapse. Mechanistically, we found that cancer-cell-derived C3 activates the C3a receptor in the choroid plexus epithelium to disrupt the blood-CSF barrier. This effect allows plasma components, including amphiregulin, and other mitogens to enter the CSF and promote cancer cell growth. Pharmacologic interference with C3 signaling proved therapeutically beneficial in suppressing leptomeningeal metastasis in these preclinical models.


Subject(s)
Complement C3/metabolism , Meningeal Neoplasms/secondary , Neoplasm Metastasis/pathology , Animals , Breast Neoplasms/pathology , Cerebrospinal Fluid , Choroid Plexus/blood supply , Complement C3/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/pathology , Macrophage-1 Antigen/metabolism , Mice , Signal Transduction , Tumor Microenvironment , Up-Regulation
4.
Cell ; 165(1): 45-60, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27015306

ABSTRACT

Metastasis frequently develops years after the removal of a primary tumor, from a minority of disseminated cancer cells that survived as latent entities through unknown mechanisms. We isolated latency competent cancer (LCC) cells from early stage human lung and breast carcinoma cell lines and defined the mechanisms that suppress outgrowth, support long-term survival, and maintain tumor-initiating potential in these cells during the latent metastasis stage. LCC cells show stem-cell-like characteristics and express SOX2 and SOX9 transcription factors, which are essential for their survival in host organs under immune surveillance and for metastatic outgrowth under permissive conditions. Through expression of the WNT inhibitor DKK1, LCC cells self-impose a slow-cycling state with broad downregulation of ULBP ligands for NK cells and evasion of NK-cell-mediated clearance. By expressing a Sox-dependent stem-like state and actively silencing WNT signaling, LCC cells can enter quiescence and evade innate immunity to remain latent for extended periods.


Subject(s)
Autocrine Communication , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Tumor Escape , Wnt Signaling Pathway , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Immunologic Surveillance , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice , Mice, Nude , SOX9 Transcription Factor/metabolism , SOXB1 Transcription Factors/metabolism
5.
Cell ; 164(5): 1015-30, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26898331

ABSTRACT

TGF-ß signaling can be pro-tumorigenic or tumor suppressive. We investigated this duality in pancreatic ductal adenocarcinoma (PDA), which, with other gastrointestinal cancers, exhibits frequent inactivation of the TGF-ß mediator Smad4. We show that TGF-ß induces an epithelial-mesenchymal transition (EMT), generally considered a pro-tumorigenic event. However, in TGF-ß-sensitive PDA cells, EMT becomes lethal by converting TGF-ß-induced Sox4 from an enforcer of tumorigenesis into a promoter of apoptosis. This is the result of an EMT-linked remodeling of the cellular transcription factor landscape, including the repression of the gastrointestinal lineage-master regulator Klf5. Klf5 cooperates with Sox4 in oncogenesis and prevents Sox4-induced apoptosis. Smad4 is required for EMT but dispensable for Sox4 induction by TGF-ß. TGF-ß-induced Sox4 is thus geared to bolster progenitor identity, whereas simultaneous Smad4-dependent EMT strips Sox4 of an essential partner in oncogenesis. Our work demonstrates that TGF-ß tumor suppression functions through an EMT-mediated disruption of a lineage-specific transcriptional network.


Subject(s)
Carcinoma, Ductal/genetics , Epithelial-Mesenchymal Transition , Gene Regulatory Networks , Pancreatic Neoplasms/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis , Carcinoma, Ductal/pathology , Kruppel-Like Transcription Factors/metabolism , Mice , Organoids/metabolism , Organoids/pathology , Pancreatic Neoplasms/pathology , SOXC Transcription Factors/metabolism , Smad4 Protein/metabolism
6.
Immunity ; 54(5): 1037-1054.e7, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33756102

ABSTRACT

Immune cells identify and destroy tumors by recognizing cellular traits indicative of oncogenic transformation. In this study, we found that myocardin-related transcription factors (MRTFs), which promote migration and metastatic invasion, also sensitize cancer cells to the immune system. Melanoma and breast cancer cells with high MRTF expression were selectively eliminated by cytotoxic lymphocytes in mouse models of metastasis. This immunosurveillance phenotype was further enhanced by treatment with immune checkpoint blockade (ICB) antibodies. We also observed that high MRTF signaling in human melanoma is associated with ICB efficacy in patients. Using biophysical and functional assays, we showed that MRTF overexpression rigidified the filamentous actin cytoskeleton and that this mechanical change rendered mouse and human cancer cells more vulnerable to cytotoxic T lymphocytes and natural killer cells. Collectively, these results suggest that immunosurveillance has a mechanical dimension, which we call mechanosurveillance, that is particularly relevant for the targeting of metastatic disease.


Subject(s)
Lymphocytes/immunology , Neoplasms/immunology , Actin Cytoskeleton/immunology , Actins/immunology , Animals , Cell Communication/immunology , Cell Line , Cell Line, Tumor , Cell Movement/immunology , Female , HEK293 Cells , Humans , Killer Cells, Natural/immunology , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Signal Transduction/immunology , Transcription Factors/immunology
7.
Nat Rev Mol Cell Biol ; 19(7): 419-435, 2018 07.
Article in English | MEDLINE | ID: mdl-29643418

ABSTRACT

Few cell signals match the impact of the transforming growth factor-ß (TGFß) family in metazoan biology. TGFß cytokines regulate cell fate decisions during development, tissue homeostasis and regeneration, and are major players in tumorigenesis, fibrotic disorders, immune malfunctions and various congenital diseases. The effects of the TGFß family are mediated by a combinatorial set of ligands and receptors and by a common set of receptor-activated mothers against decapentaplegic homologue (SMAD) transcription factors, yet the effects can differ dramatically depending on the cell type and the conditions. Recent progress has illuminated a model of TGFß action in which SMADs bind genome-wide in partnership with lineage-determining transcription factors and additionally integrate inputs from other pathways and the chromatin to trigger specific cellular responses. These new insights clarify the operating logic of the TGFß pathway in physiology and disease.

8.
Nat Rev Mol Cell Biol ; 19(7): 479, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29740128

ABSTRACT

In the section 'Combinatorial ligand perception' of the original article, DMP1 was incorrectly used in place of BMP. This has now been corrected in the HTML and PDF versions of the article.

9.
Nature ; 626(8000): 864-873, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38326607

ABSTRACT

Macrophage activation is controlled by a balance between activating and inhibitory receptors1-7, which protect normal tissues from excessive damage during infection8,9 but promote tumour growth and metastasis in cancer7,10. Here we report that the Kupffer cell lineage-determining factor ID3 controls this balance and selectively endows Kupffer cells with the ability to phagocytose live tumour cells and orchestrate the recruitment, proliferation and activation of natural killer and CD8 T lymphoid effector cells in the liver to restrict the growth of a variety of tumours. ID3 shifts the macrophage inhibitory/activating receptor balance to promote the phagocytic and lymphoid response, at least in part by buffering the binding of the transcription factors ELK1 and E2A at the SIRPA locus. Furthermore, loss- and gain-of-function experiments demonstrate that ID3 is sufficient to confer this potent anti-tumour activity to mouse bone-marrow-derived macrophages and human induced pluripotent stem-cell-derived macrophages. Expression of ID3 is therefore necessary and sufficient to endow macrophages with the ability to form an efficient anti-tumour niche, which could be harnessed for cell therapy in cancer.


Subject(s)
Inhibitor of Differentiation Proteins , Kupffer Cells , Neoplasms , Animals , Humans , Mice , Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage , Induced Pluripotent Stem Cells/cytology , Inhibitor of Differentiation Proteins/deficiency , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Kupffer Cells/cytology , Kupffer Cells/immunology , Kupffer Cells/metabolism , Liver/immunology , Liver/pathology , Macrophage Activation , Neoplasm Proteins , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phagocytosis
10.
Cell ; 156(5): 1002-16, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24581498

ABSTRACT

Brain metastasis is an ominous complication of cancer, yet most cancer cells that infiltrate the brain die of unknown causes. Here, we identify plasmin from the reactive brain stroma as a defense against metastatic invasion, and plasminogen activator (PA) inhibitory serpins in cancer cells as a shield against this defense. Plasmin suppresses brain metastasis in two ways: by converting membrane-bound astrocytic FasL into a paracrine death signal for cancer cells, and by inactivating the axon pathfinding molecule L1CAM, which metastatic cells express for spreading along brain capillaries and for metastatic outgrowth. Brain metastatic cells from lung cancer and breast cancer express high levels of anti-PA serpins, including neuroserpin and serpin B2, to prevent plasmin generation and its metastasis-suppressive effects. By protecting cancer cells from death signals and fostering vascular co-option, anti-PA serpins provide a unifying mechanism for the initiation of brain metastasis in lung and breast cancers.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Brain/metabolism , Fibrinolysin/metabolism , Neuropeptides/metabolism , Plasminogen Activator Inhibitor 2/metabolism , Serpins/metabolism , Adenocarcinoma/secondary , Animals , Astrocytes/metabolism , Brain/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/secondary , Cell Line, Tumor , Cell Survival , Disease Models, Animal , Fas Ligand Protein/metabolism , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Neural Cell Adhesion Molecule L1/metabolism , Neuropeptides/genetics , Plasminogen Activator Inhibitor 2/genetics , Plasminogen Activators/metabolism , Serpins/genetics , Neuroserpin
11.
Nature ; 616(7958): 806-813, 2023 04.
Article in English | MEDLINE | ID: mdl-36991128

ABSTRACT

Metastasis frequently develops from disseminated cancer cells that remain dormant after the apparently successful treatment of a primary tumour. These cells fluctuate between an immune-evasive quiescent state and a proliferative state liable to immune-mediated elimination1-6. Little is known about the clearing of reawakened metastatic cells and how this process could be therapeutically activated to eliminate residual disease in patients. Here we use models of indolent lung adenocarcinoma metastasis to identify cancer cell-intrinsic determinants of immune reactivity during exit from dormancy. Genetic screens of tumour-intrinsic immune regulators identified the stimulator of interferon genes (STING) pathway as a suppressor of metastatic outbreak. STING activity increases in metastatic progenitors that re-enter the cell cycle and is dampened by hypermethylation of the STING promoter and enhancer in breakthrough metastases or by chromatin repression in cells re-entering dormancy in response to TGFß. STING expression in cancer cells derived from spontaneous metastases suppresses their outgrowth. Systemic treatment of mice with STING agonists eliminates dormant metastasis and prevents spontaneous outbreaks in a T cell- and natural killer cell-dependent manner-these effects require cancer cell STING function. Thus, STING provides a checkpoint against the progression of dormant metastasis and a therapeutically actionable strategy for the prevention of disease relapse.


Subject(s)
Lung Neoplasms , Neoplasm Metastasis , Animals , Mice , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Cell Cycle , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local/drug therapy , T-Lymphocytes/immunology , Transforming Growth Factor beta , Killer Cells, Natural/immunology
12.
Immunity ; 50(4): 924-940, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995507

ABSTRACT

Transforming growth factor (TGF)-ß is a crucial enforcer of immune homeostasis and tolerance, inhibiting the expansion and function of many components of the immune system. Perturbations in TGF-ß signaling underlie inflammatory diseases and promote tumor emergence. TGF-ß is also central to immune suppression within the tumor microenvironment, and recent studies have revealed roles in tumor immune evasion and poor responses to cancer immunotherapy. Here, we present an overview of the complex biology of the TGF-ß family and its context-dependent nature. Then, focusing on cancer, we discuss the roles of TGF-ß signaling in distinct immune cell types and how this knowledge is being leveraged to unleash the immune system against the tumor.


Subject(s)
Neoplasms/immunology , Transforming Growth Factor beta/physiology , Adaptive Immunity , Animals , Dendritic Cells/immunology , Disease Progression , Epithelial-Mesenchymal Transition , Fibroblasts/immunology , Humans , Immunity, Innate , Inflammation , Macrophages/immunology , Mice, Knockout , Neutrophils/immunology , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/immunology , Tumor Escape , Tumor Microenvironment
14.
Cell ; 154(5): 1060-1073, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23993096

ABSTRACT

How organ-specific metastatic traits arise in primary tumors remains unknown. Here, we show a role of the breast tumor stroma in selecting cancer cells that are primed for metastasis in bone. Cancer-associated fibroblasts (CAFs) in triple-negative (TN) breast tumors skew heterogeneous cancer cell populations toward a predominance of clones that thrive on the CAF-derived factors CXCL12 and IGF1. Limiting concentrations of these factors select for cancer cells with high Src activity, a known clinical predictor of bone relapse and an enhancer of PI3K-Akt pathway activation by CXCL12 and IGF1. Carcinoma clones selected in this manner are primed for metastasis in the CXCL12-rich microenvironment of the bone marrow. The evidence suggests that stromal signals resembling those of a distant organ select for cancer cells that are primed for metastasis in that organ, thus illuminating the evolution of metastatic traits in a primary tumor and its distant metastases.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Neoplasm Metastasis , Signal Transduction , Animals , Bone Marrow/metabolism , Bone Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Chemokine CXCL12/metabolism , Fibroblasts/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Neoplasm Transplantation , Transcription, Genetic , Transplantation, Heterologous , src-Family Kinases/genetics , src-Family Kinases/metabolism
15.
Immunity ; 48(4): 626-628, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29669246

ABSTRACT

Immune checkpoint therapy can induce durable remissions, but many tumors demonstrate resistance. In a recent issue of Nature, Mariathasan et al. (2018) and Tauriello et al. (2018) identify stromal TGF-ß signaling as a determinant of immune exclusion. Combination TGF-ß inhibition and immunotherapy induces complete responses in mouse models.


Subject(s)
T-Lymphocytes , Transforming Growth Factor beta , Animals , Disease Models, Animal , Immunotherapy , Mice , Neoplasms
16.
Cell ; 149(6): 1179-81, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682238

ABSTRACT

Mutations in keratinocyte and melanocyte precursors that are caused by extensive sun exposure are well-established contributors to skin cancer. Now Hu et al. provide evidence that the sun's harmful rays may also cause tumor-promoting epigenetic modifications in dermal fibroblasts, highlighting further the importance of tumor-stroma interactions in cancer.

17.
Cell ; 150(1): 165-78, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770218

ABSTRACT

Metastasis and chemoresistance in cancer are linked phenomena, but the molecular basis for this link is unknown. We uncovered a network of paracrine signals between carcinoma, myeloid, and endothelial cells that drives both processes in breast cancer. Cancer cells that overexpress CXCL1 and 2 by transcriptional hyperactivation or 4q21 amplification are primed for survival in metastatic sites. CXCL1/2 attract CD11b(+)Gr1(+) myeloid cells into the tumor, which produce chemokines including S100A8/9 that enhance cancer cell survival. Although chemotherapeutic agents kill cancer cells, these treatments trigger a parallel stromal reaction leading to TNF-α production by endothelial and other stromal cells. TNF-α via NF-kB heightens the CXCL1/2 expression in cancer cells, thus amplifying the CXCL1/2-S100A8/9 loop and causing chemoresistance. CXCR2 blockers break this cycle, augmenting the efficacy of chemotherapy against breast tumors and particularly against metastasis. This network of endothelial-carcinoma-myeloid signaling interactions provides a mechanism linking chemoresistance and metastasis, with opportunities for intervention.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Chemokine CXCL1/metabolism , Drug Resistance, Neoplasm , Neoplasm Metastasis , Paracrine Communication , Animals , Breast Neoplasms/metabolism , Calgranulin A/metabolism , Calgranulin B/metabolism , Carcinoma/metabolism , Chemokine CXCL1/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Lymph Nodes/pathology , Lymphatic Metastasis , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Neoplasm Transplantation , Transplantation, Heterologous
18.
Genes Dev ; 33(21-22): 1506-1524, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31582430

ABSTRACT

TGF-ß receptors phosphorylate SMAD2 and SMAD3 transcription factors, which then form heterotrimeric complexes with SMAD4 and cooperate with context-specific transcription factors to activate target genes. Here we provide biochemical and structural evidence showing that binding of SMAD2 to DNA depends on the conformation of the E3 insert, a structural element unique to SMAD2 and previously thought to render SMAD2 unable to bind DNA. Based on this finding, we further delineate TGF-ß signal transduction by defining distinct roles for SMAD2 and SMAD3 with the forkhead pioneer factor FOXH1 as a partner in the regulation of differentiation genes in mouse mesendoderm precursors. FOXH1 is prebound to target sites in these loci and recruits SMAD3 independently of TGF-ß signals, whereas SMAD2 remains predominantly cytoplasmic in the basal state and set to bind SMAD4 and join SMAD3:FOXH1 at target promoters in response to Nodal TGF-ß signals. The results support a model in which signal-independent binding of SMAD3 and FOXH1 prime mesendoderm differentiation gene promoters for activation, and signal-driven SMAD2:SMAD4 binds to promoters that are preloaded with SMAD3:FOXH1 to activate transcription.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Models, Molecular , Signal Transduction , Smad2 Protein , Smad3 Protein , Transforming Growth Factor beta/metabolism , Animals , Embryo, Mammalian , Mice , Mice, Inbred C57BL , Protein Binding , Protein Structure, Tertiary , Smad2 Protein/chemistry , Smad2 Protein/metabolism , Smad3 Protein/chemistry , Smad3 Protein/metabolism
19.
Cell ; 147(7): 1511-24, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22196728

ABSTRACT

Specific chromatin marks keep master regulators of differentiation silent yet poised for activation by extracellular signals. We report that nodal TGF-ß signals use the poised histone mark H3K9me3 to trigger differentiation of mammalian embryonic stem cells. Nodal receptors induce the formation of companion Smad4-Smad2/3 and TRIM33-Smad2/3 complexes. The PHD-Bromo cassette of TRIM33 facilitates binding of TRIM33-Smad2/3 to H3K9me3 and H3K18ac on the promoters of mesendoderm regulators Gsc and Mixl1. The crystal structure of this cassette, bound to histone H3 peptides, illustrates that PHD recognizes K9me3, and Bromo binds an adjacent K18ac. The interaction between TRIM33-Smad2/3 and H3K9me3 displaces the chromatin-compacting factor HP1γ, making nodal response elements accessible to Smad4-Smad2/3 for Pol II recruitment. In turn, Smad4 increases K18 acetylation to augment TRIM33-Smad2/3 binding. Thus, nodal effectors use the H3K9me3 mark as a platform to switch master regulators of stem cell differentiation from the poised to the active state.


Subject(s)
Chromatin Assembly and Disassembly , Embryonic Stem Cells/metabolism , Histones/metabolism , Smad Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , Goosecoid Protein/genetics , Homeodomain Proteins/genetics , Humans , Mice , Models, Molecular , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Alignment
20.
Nature ; 578(7793): E11, 2020 02.
Article in English | MEDLINE | ID: mdl-31937917

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

SELECTION OF CITATIONS
SEARCH DETAIL