Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 6578, 2023 10 18.
Article in English | MEDLINE | ID: mdl-37852949

ABSTRACT

Large genes including several CRISPR-Cas modules like gene activators (CRISPRa) require dual adeno-associated viral (AAV) vectors for an efficient in vivo delivery and expression. Current dual AAV vector approaches have important limitations, e.g., low reconstitution efficiency, production of alien proteins, or low flexibility in split site selection. Here, we present a dual AAV vector technology based on reconstitution via mRNA trans-splicing (REVeRT). REVeRT is flexible in split site selection and can efficiently reconstitute different split genes in numerous in vitro models, in human organoids, and in vivo. Furthermore, REVeRT can functionally reconstitute a CRISPRa module targeting genes in various mouse tissues and organs in single or multiplexed approaches upon different routes of administration. Finally, REVeRT enabled the reconstitution of full-length ABCA4 after intravitreal injection in a mouse model of Stargardt disease. Due to its flexibility and efficiency REVeRT harbors great potential for basic research and clinical applications.


Subject(s)
Gene Editing , Trans-Splicing , Humans , Animals , Mice , Trans-Splicing/genetics , Genetic Therapy , Stargardt Disease , Genetic Vectors/genetics , Dependovirus/genetics , Dependovirus/metabolism , ATP-Binding Cassette Transporters/metabolism
3.
RSC Chem Biol ; 2(4): 1291-1295, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34458843

ABSTRACT

Templated chemistry offers the prospect of addressing specificity challenges occurring in bioconjugation reactions. Here, we show two peptide-templated amide-bond forming reactions that enable the concurrent labelling of two different membrane proteins with two different peptide nucleic acid (PNA) barcodes. The reaction system is based on the mutually selective coiled coil interaction between two thioester-linked PNA-peptide conjugates and two cysteine peptides serving as genetically encoded peptide tags. Orthogonal coiled coil templated covalent labelling is highly specific, quantitative and proceeds within a minute. To demonstrate the usefulness, we evaluated receptor internalisation of two membranous receptors EGFR (epidermal growth factor) and ErbB2 (epidermal growth factor receptor 2) by first staining PNA-tagged proteins with fluorophore-DNA conjugates and then erasing signals from non-internalized receptors via toehold-mediated strand displacement.

4.
Nucleic Acids Res ; 49(11): e62, 2021 06 21.
Article in English | MEDLINE | ID: mdl-33684219

ABSTRACT

The genetic code of mammalian cells can be expanded to allow the incorporation of non-canonical amino acids (ncAAs) by suppressing in-frame amber stop codons (UAG) with an orthogonal pyrrolysyl-tRNA synthetase (PylRS)/tRNAPylCUA (PylT) pair. However, the feasibility of this approach is substantially hampered by unpredictable variations in incorporation efficiencies at different stop codon positions within target proteins. Here, we apply a proteomics-based approach to quantify ncAA incorporation rates at hundreds of endogenous amber stop codons in mammalian cells. With these data, we compute iPASS (Identification of Permissive Amber Sites for Suppression; available at www.bultmannlab.eu/tools/iPASS), a linear regression model to predict relative ncAA incorporation efficiencies depending on the surrounding sequence context. To verify iPASS, we develop a dual-fluorescence reporter for high-throughput flow-cytometry analysis that reproducibly yields context-specific ncAA incorporation efficiencies. We show that nucleotides up- and downstream of UAG synergistically influence ncAA incorporation efficiency independent of cell line and ncAA identity. Additionally, we demonstrate iPASS-guided optimization of ncAA incorporation rates by synonymous exchange of codons flanking the amber stop codon. This combination of in silico analysis followed by validation in living mammalian cells substantially simplifies identification as well as adaptation of sites within a target protein to confer high ncAA incorporation rates.


Subject(s)
Amino Acids/metabolism , Genetic Code , Animals , Cell Line , Codon , Codon, Terminator , Computer Simulation , Embryonic Stem Cells/metabolism , Flow Cytometry , Genes, Reporter , HEK293 Cells , Humans , Linear Models , Mice , Mutation , Proteomics
5.
Sci Rep ; 11(1): 5838, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712646

ABSTRACT

Chemotherapy resistance is the main impediment in the treatment of acute myeloid leukaemia (AML). Despite rapid advances, the various mechanisms inducing resistance development remain to be defined in detail. Here we report that loss-of-function mutations (LOF) in the histone methyltransferase EZH2 have the potential to confer resistance against the chemotherapeutic agent cytarabine. We identify seven distinct EZH2 mutations leading to loss of H3K27 trimethylation via multiple mechanisms. Analysis of matched diagnosis and relapse samples reveal a heterogenous regulation of EZH2 and a loss of EZH2 in 50% of patients. We confirm that loss of EZH2 induces resistance against cytarabine in the cell lines HEK293T and K562 as well as in a patient-derived xenograft model. Proteomics and transcriptomics analysis reveal that resistance is conferred by upregulation of multiple direct and indirect EZH2 target genes that are involved in apoptosis evasion, augmentation of proliferation and alteration of transmembrane transporter function. Our data indicate that loss of EZH2 results in upregulation of its target genes, providing the cell with a selective growth advantage, which mediates chemotherapy resistance.


Subject(s)
Drug Resistance, Neoplasm/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Loss of Function Mutation/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, Myeloid, Acute/diagnosis , Neoplasm Recurrence, Local/pathology , Up-Regulation/drug effects , Up-Regulation/genetics , Xenograft Model Antitumor Assays
6.
Nat Chem ; 13(1): 15-23, 2021 01.
Article in English | MEDLINE | ID: mdl-33288896

ABSTRACT

DNA nanotechnology is an emerging field that promises fascinating opportunities for the manipulation and imaging of proteins on a cell surface. The key to progress is the ability to create a nucleic acid-protein junction in the context of living cells. Here we report a covalent labelling reaction that installs a biostable peptide nucleic acid (PNA) tag. The reaction proceeds within minutes and is specific for proteins carrying a 2 kDa coiled-coil peptide tag. Once installed, the PNA label serves as a generic landing platform that enables the recruitment of fluorescent dyes via nucleic acid hybridization. We demonstrate the versatility of this approach by recruiting different fluorophores, assembling multiple fluorophores for increased brightness and achieving reversible labelling by way of toehold-mediated strand displacement. Additionally, we show that labelling can be carried out using two different coiled-coil systems, with epidermal growth factor receptor and endothelin receptor type B, on both HEK293 and CHO cells. Finally, we apply the method to monitor internalization of epidermal growth factor receptor on CHO cells.


Subject(s)
ErbB Receptors/metabolism , Microscopy, Fluorescence , Peptide Nucleic Acids/chemistry , Receptor, Endothelin B/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , ErbB Receptors/chemistry , ErbB Receptors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Nucleic Acid Hybridization , Peptide Nucleic Acids/chemical synthesis , Peptide Nucleic Acids/metabolism , Peptides/chemical synthesis , Peptides/chemistry , Peptides/metabolism , Protein Binding , Receptor, Endothelin B/chemistry , Receptor, Endothelin B/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification
8.
Nat Commun ; 11(1): 5972, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33235224

ABSTRACT

Genome-wide DNA demethylation is a unique feature of mammalian development and naïve pluripotent stem cells. Here, we describe a recently evolved pathway in which global hypomethylation is achieved by the coupling of active and passive demethylation. TET activity is required, albeit indirectly, for global demethylation, which mostly occurs at sites devoid of TET binding. Instead, TET-mediated active demethylation is locus-specific and necessary for activating a subset of genes, including the naïve pluripotency and germline marker Dppa3 (Stella, Pgc7). DPPA3 in turn drives large-scale passive demethylation by directly binding and displacing UHRF1 from chromatin, thereby inhibiting maintenance DNA methylation. Although unique to mammals, we show that DPPA3 alone is capable of inducing global DNA demethylation in non-mammalian species (Xenopus and medaka) despite their evolutionary divergence from mammals more than 300 million years ago. Our findings suggest that the evolution of Dppa3 facilitated the emergence of global DNA demethylation in mammals.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone , DNA Demethylation , Mammals/genetics , Pluripotent Stem Cells/metabolism , Animals , Biological Evolution , CCAAT-Enhancer-Binding Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Methylation , DNA-Directed DNA Polymerase/metabolism , Epigenomics , Evolution, Molecular , Gene Expression Regulation , Genes, Regulator , Germ Cells/metabolism , Mice , Ubiquitin-Protein Ligases/metabolism
9.
Sci Rep ; 10(1): 12066, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32694513

ABSTRACT

Cytosine DNA bases can be methylated by DNA methyltransferases and subsequently oxidized by TET proteins. The resulting 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) are considered demethylation intermediates as well as stable epigenetic marks. To dissect the contributions of these cytosine modifying enzymes, we generated combinations of Tet knockout (KO) embryonic stem cells (ESCs) and systematically measured protein and DNA modification levels at the transition from naive to primed pluripotency. Whereas the increase of genomic 5-methylcytosine (5mC) levels during exit from pluripotency correlated with an upregulation of the de novo DNA methyltransferases DNMT3A and DNMT3B, the subsequent oxidation steps turned out to be far more complex. The strong increase of oxidized cytosine bases (5hmC, 5fC, and 5caC) was accompanied by a drop in TET2 levels, yet the analysis of KO cells suggested that TET2 is responsible for most 5fC formation. The comparison of modified cytosine and enzyme levels in Tet KO cells revealed distinct and differentiation-dependent contributions of TET1 and TET2 to 5hmC and 5fC formation arguing against a processive mechanism of 5mC oxidation. The apparent independent steps of 5hmC and 5fC formation suggest yet to be identified mechanisms regulating TET activity that may constitute another layer of epigenetic regulation.


Subject(s)
Cell Differentiation , Cytosine/metabolism , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Oxidation-Reduction , Proto-Oncogene Proteins/genetics , Animals , CRISPR-Cas Systems , Chromatography, High Pressure Liquid , DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases , Epigenesis, Genetic , Mice , Mice, Knockout , Proteome , Proteomics , Proto-Oncogene Proteins/metabolism , Tandem Mass Spectrometry
10.
Methods Mol Biol ; 2175: 109-121, 2020.
Article in English | MEDLINE | ID: mdl-32681487

ABSTRACT

Biotin proximity labeling has largely extended the toolbox of mass spectrometry-based interactomics. To date, BirA, engineered BirA variants, or other biotinylating enzymes have been widely applied to characterize protein interactions. By implementing chromatin purification-based methods the genome-wide interactome of proteins can be defined. However, acquiring a high-resolution interactome of a single genomic locus preferably by multiplexed measurements of several distinct genomic loci in parallel remains challenging. We recently developed CasID, a novel approach where the catalytically inactive Cas9 (dCas9) is coupled to the promiscuous biotin ligase BirA (BirA∗). With CasID, first the local proteome at repetitive telomeric, major satellite, and minor satellite regions was determined. With more efficient biotin ligases and sensitive mass spectrometry, others have successfully identified the chromatin composition at even smaller genomic, non-repetitive regions of a few hundred base pairs in length. Here, we summarize the most recent developments towards interactomics at a single genomic locus and provide a step-by-step protocol based on the CasID approach.


Subject(s)
Chromatin/metabolism , Mass Spectrometry/methods , Proteome/metabolism , Proteomics/methods , Animals , Biotin , Biotinylation , Carbon-Nitrogen Ligases , Cell Line , Escherichia coli Proteins , Genomics , Mice , Repressor Proteins
11.
Cell Rep ; 31(5): 107522, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32330423

ABSTRACT

Tumor cells orchestrate their microenvironment. Here, we provide biochemical, structural, functional, and clinical evidence that Cathepsin S (CTSS) alterations induce a tumor-promoting immune microenvironment in follicular lymphoma (FL). We found CTSS mutations at Y132 in 6% of FL (19/305). Another 13% (37/286) had CTSS amplification, which was associated with higher CTSS expression. CTSS Y132 mutations lead to accelerated autocatalytic conversion from an enzymatically inactive profrom to active CTSS and increased substrate cleavage, including CD74, which regulates major histocompatibility complex class II (MHC class II)-restricted antigen presentation. Lymphoma cells with hyperactive CTSS more efficiently activated antigen-specific CD4+ T cells in vitro. Tumors with hyperactive CTSS showed increased CD4+ T cell infiltration and proinflammatory cytokine perturbation in a mouse model and in human FLs. In mice, this CTSS-induced immune microenvironment promoted tumor growth. Clinically, patients with CTSS-hyperactive FL had better treatment outcomes with standard immunochemotherapies, indicating that these immunosuppressive regimens target both the lymphoma cells and the tumor-promoting immune microenvironment.


Subject(s)
Antigen Presentation/immunology , Cathepsins/metabolism , Lymphoma, Follicular/metabolism , Tumor Microenvironment/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Cytokines/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Immunosuppression Therapy , Lymphoma, Follicular/pathology , Mice
12.
Nat Commun ; 11(1): 304, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949141

ABSTRACT

Biological processes in development and disease are controlled by the abundance, localization and modification of cellular proteins. We have developed versatile tools based on recombinant E3 ubiquitin ligases that are controlled by light or drug induced heterodimerization for nanobody or DARPin targeted depletion of endogenous proteins in cells and organisms. We use this rapid, tunable and reversible protein depletion for functional studies of essential proteins like PCNA in DNA repair and to investigate the role of CED-3 in apoptosis during Caenorhabditis elegans development. These independent tools can be combined for spatial and temporal depletion of different sets of proteins, can help to distinguish immediate cellular responses from long-term adaptation effects and can facilitate the exploration of complex networks.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cytological Techniques , Light , Ubiquitin-Protein Ligases/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/radiation effects , Animals , Apoptosis , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/drug effects , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/radiation effects , Caspases/drug effects , Caspases/metabolism , Caspases/radiation effects , Cell Engineering/methods , DNA Damage , DNA Ligase ATP , DNA Repair , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins , HeLa Cells , Humans , Lamin Type A/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
13.
Leukemia ; 34(1): 50-62, 2020 01.
Article in English | MEDLINE | ID: mdl-31201358

ABSTRACT

Acute myeloid leukemia (AML) is an aggressive hematologic neoplasm resulting from the malignant transformation of myeloid progenitors. Despite intensive chemotherapy leading to initial treatment responses, relapse caused by intrinsic or acquired drug resistance represents a major challenge. Here, we report that histone 3 lysine 27 demethylase KDM6A (UTX) is targeted by inactivating mutations and mutation-independent regulation in relapsed AML. Analyses of matched diagnosis and relapse specimens from individuals with KDM6A mutations showed an outgrowth of the KDM6A mutated tumor population at relapse. KDM6A expression is heterogeneously regulated and relapse-specific loss of KDM6A was observed in 45.7% of CN-AML patients. KDM6A-null myeloid leukemia cells were more resistant to treatment with the chemotherapeutic agents cytarabine (AraC) and daunorubicin. Inducible re-expression of KDM6A in KDM6A-null cell lines suppressed proliferation and sensitized cells again to AraC treatment. RNA expression analysis and functional studies revealed that resistance to AraC was conferred by downregulation of the nucleoside membrane transporter ENT1 (SLC29A1) by reduced H3K27 acetylation at the ENT1 locus. Our results show that loss of KDM6A provides cells with a selective advantage during chemotherapy, which ultimately leads to the observed outgrowth of clones with KDM6A mutations or reduced KDM6A expression at relapse.


Subject(s)
Drug Resistance, Neoplasm/physiology , Histone Demethylases/genetics , Histone Demethylases/metabolism , Leukemia, Myeloid, Acute/pathology , Animals , Heterografts , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mutation
14.
Angiogenesis ; 22(1): 117-131, 2019 02.
Article in English | MEDLINE | ID: mdl-30132150

ABSTRACT

Cancer cells sustain their metabolic needs through nutrients and oxygen supplied by the bloodstream. The requirement for tumor angiogenesis has been therapeutically exploited in the clinical setting mainly by means of inhibition of the vascular endothelial growth factor family of ligands and receptors. Despite promising results in preclinical models, the benefits for patients proved to be limited. Inadequate efficacy similarly halted the development of agents impinging on the activity of the activin receptor-like kinase (ALK)1, a member of the transforming growth factor-ß superfamily. Notwithstanding its characterization as an endothelial cell marker, the full spectrum of biological processes associated with ALK1 is essentially unexplored. Here, we present data revealing the genetic network associated with ACVRL1 (the gene encoding for ALK1) expression in human cancer tissues. Computational analysis unveiled a hitherto unknown role for ACVRL1 in relation to genes modulating the functionality of the immune cell compartment. Moreover, we generated a signature of 8 genes co-expressed with ACVRL1 across different tumor types and characterized the c-type lectin domain containing protein (CLEC)14A as a potential downstream target of ACVRL1. Considering the lack of reagents for ALK1 detection that has hampered the field to date, our work provides the opportunity to validate the 8-gene signature and CLEC14A as biomarkers for ALK1 activity. Ultimately, this may help revisit the clinical development of already existing ALK1-blocking compounds as precision medicines for cancer.


Subject(s)
Activin Receptors, Type II/immunology , Biomarkers, Tumor/immunology , Cell Adhesion Molecules/immunology , Gene Expression Regulation, Neoplastic/immunology , Lectins, C-Type/immunology , Neoplasms/immunology , Transcription, Genetic/immunology , Activin Receptors, Type II/genetics , Animals , Biomarkers, Tumor/genetics , Cell Adhesion Molecules/genetics , Female , Humans , Lectins, C-Type/genetics , Male , Mice , Neoplasms/genetics , Neoplasms/pathology
15.
Matrix Biol ; 81: 107-121, 2019 08.
Article in English | MEDLINE | ID: mdl-30502484

ABSTRACT

Cancer stem cell populations are important for the initiation, progression and metastasis of tumors. The mechanisms governing cancer stem cell control are only partially understood, but activation of the Notch3 pathway plays a crucial role in the maintenance of breast cancer stem cells. Expression of Cartilage Oligomeric Matrix Protein (COMP) in breast cancer cells is correlated with poor survival and higher recurrence rates in patients. In this study, we provide in vivo and in vitro evidence that COMP expression increases the proportion of cancer stem cells in breast cancer. Thus, MDA-MB-231 and BT-20 cells expressing COMP formed larger tumorspheres in vivo and in vitro and displayed higher ALDH-activity than cells lacking COMP. Additionally, BT-20 COMP-expressing cells displayed higher expression of CD133 compared with the control cells. Furthermore, among the different Notch receptors, Notch3 is specifically activated in COMP-expressing cells. Mechanistically, activation of Notch3 is mediated by secreted, polymeric COMP, which interacts with both Notch3 and its ligand Jagged1, bridging the receptor and ligand together, enhancing Notch3-specific signaling. COMP-dependent Notch3 activation also leads to cross-talk with ß-Catenin and AKT pathways. Using the model of MMTV-PyMT mouse breast tumorigenesis, we observed a decrease in the size of tumors and the amount of cancer stem cells as well as reduced Notch3 activation, in COMP knockout mice in comparison to wild type mice. In conclusion, we reveal a novel molecular mechanism whereby COMP regulates the cancer stem cell population through increasing the interaction between Notch3 and Jagged1, leading to increased activation of Notch3 signaling.


Subject(s)
Breast Neoplasms/metabolism , Cartilage Oligomeric Matrix Protein/metabolism , Jagged-1 Protein/metabolism , Neoplastic Stem Cells/metabolism , Receptor, Notch3/metabolism , Animals , Breast Neoplasms/genetics , Cartilage Oligomeric Matrix Protein/genetics , Cell Line, Tumor , Female , Gene Knockout Techniques , Humans , Mice , Neoplasm Transplantation , Receptor, Notch3/genetics , Signal Transduction
16.
Oncoimmunology ; 7(12): e1500671, 2018.
Article in English | MEDLINE | ID: mdl-30524892

ABSTRACT

Mutated proteins arising from somatic mutations in tumors are promising targets for cancer immunotherapy. They represent true tumor-specific antigens (TSAs) as they are exclusively expressed in tumors, reduce the risk of autoimmunity and are more likely to overcome tolerance compared to wild-type (wt) sequences. Hence, we designed a panel of long peptides (LPs, 28-35 aa) comprising driver gene mutations in TP35 and KRAS frequently found in gastrointestinal tumors to test their combined immunotherapeutic potential. We found increased numbers of T cells responsive against respective mutated and wt peptides in colorectal cancer patients that carry the tested mutations in their tumors than patients with other mutations. Further, active immunization of HLA(-A2/DR1)-humanized mice with mixes of the same mutated LPs yielded simultaneous, polyvalent CD8+/CD4+ T cell responses against the majority of peptides. Peptide-specific T cells possessed a multifunctional cytokine profile with CD4+ T cells showing a TH1-like phenotype. Two mutated peptides (Kras[G12V], p53[R248W]) induced significantly higher T cell responses than corresponding wt sequences and comprised HLA-A2/DR1-restricted mutated epitopes. However, vaccination with the same highly immunogenic LPs strongly increased systemic regulatory T cells (Treg) numbers in a syngeneic sarcoma model over-expressing these mutated protein variants and resulted in accelerated tumor outgrowth. In contrast, tumor outgrowth was delayed when vaccination was directed against tumor-intrinsic Kras/Tp53 mutations of lower immunogenicity. Conclusively, we show that LP vaccination targeting multiple mutated TSAs elicits polyvalent, multifunctional, and mutation-specific effector T cells capable of targeting tumors. However, the success of this therapeutic approach can be hampered by vaccination-induced, TSA-specific Tregs.

17.
Nat Commun ; 9(1): 5150, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30514914

ABSTRACT

Cancer-associated fibroblasts (CAFs) are a major constituent of the tumor microenvironment, although their origin and roles in shaping disease initiation, progression and treatment response remain unclear due to significant heterogeneity. Here, following a negative selection strategy combined with single-cell RNA sequencing of 768 transcriptomes of mesenchymal cells from a genetically engineered mouse model of breast cancer, we define three distinct subpopulations of CAFs. Validation at the transcriptional and protein level in several experimental models of cancer and human tumors reveal spatial separation of the CAF subclasses attributable to different origins, including the peri-vascular niche, the mammary fat pad and the transformed epithelium. Gene profiles for each CAF subtype correlate to distinctive functional programs and hold independent prognostic capability in clinical cohorts by association to metastatic disease. In conclusion, the improved resolution of the widely defined CAF population opens the possibility for biomarker-driven development of drugs for precision targeting of CAFs.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts , Sequence Analysis, RNA , Transcriptome , Adipose Tissue/metabolism , Animals , Biomarkers, Tumor/genetics , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/classification , Cell Cycle/genetics , Cell Line, Tumor , Cluster Analysis , Disease Progression , Epithelium/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intercellular Junctions/genetics , Logistic Models , Mice , Mice, Transgenic , Prognosis , Transcription Factors/genetics , Transcriptome/genetics
18.
Biochem Biophys Res Commun ; 503(2): 984-990, 2018 09 05.
Article in English | MEDLINE | ID: mdl-29932922

ABSTRACT

The development and progression of a tumor depends on the close interaction of malignant cells and the supportive and suppressive tumor microenvironment. Paracrine signaling enables tumor cells to shape the surrounding tissue in order to decrease recognition by the immune system, attract blood vessels to fuel growth, change metabolic programs, and induce wound healing programs. In this study, we investigate the role of the platelet-derived growth factor (PDGF) family members PDGFA, PDGFB, PDGFC and PDGFD and their cognate tyrosine kinase receptors PDGFRA and PDGFRB, using publicly available data from The Cancer Genome Atlas and the Human Protein Atlas. Large scale analysis of expression correlation in RNA sequencing data from 7616 samples derived from 16 tumor types, revealed conserved functional programs in PDGF signaling in the majority of solid tumor types. Besides the well-known effects of PDGF signaling in mesenchymal cells, our analyses revealed a potential role of PDGF signaling in the composition of the immune microenvironment. We furthermore derived gene signatures with increased prognostic value for each PDGF family member. This study emphasizes the potential to impinge on specific paracrine signaling events to interfere with the crosstalk between malignant cells and their microenvironment.


Subject(s)
Neoplasms/genetics , Platelet-Derived Growth Factor/genetics , Signal Transduction , Transcriptome , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/diagnosis , Neoplasms/metabolism , Platelet-Derived Growth Factor/analysis , Platelet-Derived Growth Factor/metabolism , Prognosis
19.
Nat Med ; 24(4): 463-473, 2018 05.
Article in English | MEDLINE | ID: mdl-29529015

ABSTRACT

Breast tumors of the basal-like, hormone receptor-negative subtype remain an unmet clinical challenge, as there is high rate of recurrence and poor survival in patients following treatment. Coevolution of the malignant mammary epithelium and its underlying stroma instigates cancer-associated fibroblasts (CAFs) to support most, if not all, hallmarks of cancer progression. Here we delineate a previously unappreciated role for CAFs as determinants of the molecular subtype of breast cancer. We identified paracrine crosstalk between cancer cells expressing platelet-derived growth factor (PDGF)-CC and CAFs expressing the cognate receptors in human basal-like mammary carcinomas. Genetic or pharmacological intervention of PDGF-CC activity in mouse models of cancer resulted in conversion of basal-like breast cancers into a hormone receptor-positive state that enhanced sensitivity to endocrine therapy in previously resistant tumors. We conclude that specification of breast cancer to the basal-like subtype is under microenvironmental control and is therapeutically actionable.


Subject(s)
Breast Neoplasms/pathology , Lymphokines/metabolism , Paracrine Communication , Platelet-Derived Growth Factor/metabolism , Tumor Microenvironment , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/blood supply , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Proliferation , Epithelial Cells/metabolism , Estrogen Receptor alpha/metabolism , Female , Fibrosis , Humans , Lymphokines/deficiency , Mice, Inbred C57BL , Middle Aged , Neovascularization, Pathologic/pathology , Platelet-Derived Growth Factor/deficiency , Prognosis , Proportional Hazards Models , Signal Transduction , Stromal Cells/pathology , Survival Analysis , Treatment Outcome
20.
Oncotarget ; 7(47): 76920-76933, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27764775

ABSTRACT

Human CUB and Sushi multiple domains 1 (CSMD1) is a membrane-bound complement inhibitor suggested to act as a putative tumor suppressor gene, since allelic loss of this region encompassing 8p23 including CSMD1 characterizes various malignancies. Here, we assessed the role of CSMD1 as a tumor suppressor gene in the development of breast cancer in vitro and in vivo. We found that human breast tumor tissues expressed CSMD1 at lower levels compared to that in normal mammary tissues. The decreased expression of CSMD1 was linked to a shorter overall survival of breast cancer patients. We also revealed that expression of CSMD1 in human breast cancer cells BT-20 and MDA-MB-231 significantly inhibited their malignant phenotypes, including migration, adhesion and invasion. Conversely, stable silencing of CSMD1 expression in T47D cells enhanced cancer cell migratory, adherent and clonogenic abilities. Moreover, expression of CSMD1 in the highly invasive MDA-MB-231 cells diminished their signaling potential as well as their stem cell-like properties as assessed by measurement of aldehyde dehydrogenase activity. In a xenograft model, expression of CSMD1 blocked the ability of cancer cells to metastasize to secondary sites in vivo, likely via inhibiting local invasion but not the extravasation into distant tissues. Taken together, these findings demonstrate the role of CSMD1 as a tumor suppressor gene in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Down-Regulation , Membrane Proteins/genetics , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Prognosis , Survival Analysis , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...