Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Nat Commun ; 14(1): 1867, 2023 04 04.
Article in English | MEDLINE | ID: mdl-37015919

ABSTRACT

Metastatic melanoma develops once transformed melanocytic cells begin to de-differentiate into migratory and invasive melanoma cells with neural crest cell (NCC)-like and epithelial-to-mesenchymal transition (EMT)-like features. However, it is still unclear how transformed melanocytes assume a metastatic melanoma cell state. Here, we define DNA methylation changes that accompany metastatic progression in melanoma patients and discover Nuclear Receptor Subfamily 2 Group F, Member 2 - isoform 2 (NR2F2-Iso2) as an epigenetically regulated metastasis driver. NR2F2-Iso2 is transcribed from an alternative transcriptional start site (TSS) and it is truncated at the N-terminal end which encodes the NR2F2 DNA-binding domain. We find that NR2F2-Iso2 expression is turned off by DNA methylation when NCCs differentiate into melanocytes. Conversely, this process is reversed during metastatic melanoma progression, when NR2F2-Iso2 becomes increasingly hypomethylated and re-expressed. Our functional and molecular studies suggest that NR2F2-Iso2 drives metastatic melanoma progression by modulating the activity of full-length NR2F2 (Isoform 1) over EMT- and NCC-associated target genes. Our findings indicate that DNA methylation changes play a crucial role during metastatic melanoma progression, and their control of NR2F2 activity allows transformed melanocytes to acquire NCC-like and EMT-like features. This epigenetically regulated transcriptional plasticity facilitates cell state transitions and metastatic spread.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Cell Line, Tumor , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , Epigenesis, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Gene Expression Regulation, Neoplastic , COUP Transcription Factor II/metabolism
2.
Nat Commun ; 13(1): 844, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35149678

ABSTRACT

The combination of ultrahigh-throughput screening and sequencing informs on function and intragenic epistasis within combinatorial protein mutant libraries. Establishing a droplet-based, in vitro compartmentalised approach for robust expression and screening of protein kinase cascades (>107 variants/day) allowed us to dissect the intrinsic molecular features of the MKK-ERK signalling pathway, without interference from endogenous cellular components. In a six-residue combinatorial library of the MKK1 docking domain, we identified 29,563 sequence permutations that allow MKK1 to efficiently phosphorylate and activate its downstream target kinase ERK2. A flexibly placed hydrophobic sequence motif emerges which is defined by higher order epistatic interactions between six residues, suggesting synergy that enables high connectivity in the sequence landscape. Through positive epistasis, MKK1 maintains function during mutagenesis, establishing the importance of co-dependent residues in mammalian protein kinase-substrate interactions, and creating a scenario for the evolution of diverse human signalling networks.


Subject(s)
Epistasis, Genetic , Mitogen-Activated Protein Kinases/chemistry , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphates/metabolism , Catalysis , Humans , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Docking Simulation , Phosphorylation , Protein Domains , Protein Kinases/chemistry , Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Substrate Specificity
3.
Nature ; 595(7868): 578-584, 2021 07.
Article in English | MEDLINE | ID: mdl-34135508

ABSTRACT

Macrophages have a key role in shaping the tumour microenvironment (TME), tumour immunity and response to immunotherapy, which makes them an important target for cancer treatment1,2. However, modulating macrophages has proved extremely difficult, as we still lack a complete understanding of the molecular and functional diversity of the tumour macrophage compartment. Macrophages arise from two distinct lineages. Tissue-resident macrophages self-renew locally, independent of adult haematopoiesis3-5, whereas short-lived monocyte-derived macrophages arise from adult haematopoietic stem cells, and accumulate mostly in inflamed lesions1. How these macrophage lineages contribute to the TME and cancer progression remains unclear. To explore the diversity of the macrophage compartment in human non-small cell lung carcinoma (NSCLC) lesions, here we performed single-cell RNA sequencing of tumour-associated leukocytes. We identified distinct populations of macrophages that were enriched in human and mouse lung tumours. Using lineage tracing, we discovered that these macrophage populations differ in origin and have a distinct temporal and spatial distribution in the TME. Tissue-resident macrophages accumulate close to tumour cells early during tumour formation to promote epithelial-mesenchymal transition and invasiveness in tumour cells, and they also induce a potent regulatory T cell response that protects tumour cells from adaptive immunity. Depletion of tissue-resident macrophages reduced the numbers and altered the phenotype of regulatory T cells, promoted the accumulation of CD8+ T cells and reduced tumour invasiveness and growth. During tumour growth, tissue-resident macrophages became redistributed at the periphery of the TME, which becomes dominated by monocyte-derived macrophages in both mouse and human NSCLC. This study identifies the contribution of tissue-resident macrophages to early lung cancer and establishes them as a target for the prevention and treatment of early lung cancer lesions.


Subject(s)
Carcinogenesis , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Macrophages/immunology , Tumor Microenvironment , Animals , CD8-Positive T-Lymphocytes/immunology , Epithelial-Mesenchymal Transition , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , T-Lymphocytes, Regulatory/immunology
4.
Nat Commun ; 10(1): 5023, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31685822

ABSTRACT

Melanoma, the deadliest skin cancer, remains largely incurable at advanced stages. Currently, there is a lack of animal models that resemble human melanoma initiation and progression. Recent studies using a Tyr-CreER driven mouse model have drawn contradictory conclusions about the potential of melanocyte stem cells (McSCs) to form melanoma. Here, we employ a c-Kit-CreER-driven model that specifically targets McSCs to show that oncogenic McSCs are a bona fide source of melanoma that expand in the niche, and then establish epidermal melanomas that invade into the underlying dermis. Further, normal Wnt and Endothelin niche signals during hair anagen onset are hijacked to promote McSC malignant transformation during melanoma induction. Finally, molecular profiling reveals strong resemblance of murine McSC-derived melanoma to human melanoma in heterogeneity and gene signatures. These findings provide experimental validation of the human melanoma progression model and key insights into the transformation and heterogeneity of McSC-derived melanoma.


Subject(s)
Carcinogenesis/pathology , Melanocytes/pathology , Melanoma/pathology , Neoplastic Stem Cells/pathology , Animals , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/pathology , Dermis/pathology , Disease Models, Animal , Epidermis/pathology , Homeostasis , Humans , Melanocytes/metabolism , Mice , Mutation/genetics , Neoplastic Stem Cells/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-kit/metabolism , Tumor Microenvironment , Wnt Signaling Pathway
6.
Oncogene ; 38(29): 5766-5777, 2019 07.
Article in English | MEDLINE | ID: mdl-31239516

ABSTRACT

We previously showed that KLF4, a gene highly expressed in murine prostate stem cells, blocks the progression of indolent intraepithelial prostatic lesions into aggressive and rapidly growing tumors. Here, we show that the anti-tumorigenic effect of KLF4 extends to PC3 human prostate cancer cells growing in the bone. We compared KLF4 null cells with cells transduced with a DOX-inducible KLF4 expression system, and find KLF4 function inhibits PC3 growth in monolayer and soft agar cultures. Furthermore, KLF4 null cells proliferate rapidly, forming large, invasive, and osteolytic tumors when injected into mouse femurs, whereas KLF4 re-expression immediately after their intra-femoral inoculation blocks tumor development and preserves a normal bone architecture. KLF4 re-expression in established KLF4 null bone tumors inhibits their osteolytic effects, preventing bone fractures and inducing an osteogenic response with new bone formation. In addition to these profound biological changes, KLF4 also induces a transcriptional shift from an osteolytic program in KLF4 null cells to an osteogenic program. Importantly, bioinformatic analysis shows that genes regulated by KLF4 overlap significantly with those expressed in metastatic prostate cancer patients and in three individual cohorts with bone metastases, strengthening the clinical relevance of the findings in our xenograft model.


Subject(s)
Bone Neoplasms/secondary , Kruppel-Like Transcription Factors/physiology , Osteolysis/physiopathology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cohort Studies , Heterografts , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism
7.
Cell Stem Cell ; 24(3): 390-404.e8, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30713093

ABSTRACT

Basal tumor propagating cells (TPCs) control squamous cell carcinoma (SCC) growth by self-renewing and differentiating into supra-basal SCC cells, which lack proliferative potential. While transcription factors such as SOX2 and KLF4 can drive these behaviors, their molecular roles and regulatory interactions with each other have remained elusive. Here, we show that PITX1 is specifically expressed in TPCs, where it co-localizes with SOX2 and TRP63 and determines cell fate in mouse and human SCC. Combining gene targeting with chromatin immunoprecipitation sequencing (ChIP-seq) and transcriptomic analyses reveals that PITX1 cooperates with SOX2 and TRP63 to sustain an SCC-specific transcriptional feed-forward circuit that maintains TPC-renewal, while inhibiting KLF4 expression and preventing KLF4-dependent differentiation. Conversely, KLF4 represses PITX1, SOX2, and TRP63 expression to prevent TPC expansion. This bi-stable, multi-input network reveals a molecular framework that explains self-renewal, aberrant differentiation, and SCC growth in mice and humans, providing clues for developing differentiation-inducing therapeutic strategies.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Differentiation , Gene Expression Regulation, Neoplastic , Paired Box Transcription Factors/genetics , Transcription, Genetic , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Female , Humans , Kruppel-Like Factor 4 , Mice , Mice, Nude , Paired Box Transcription Factors/metabolism , Tumor Cells, Cultured
8.
Cell Rep ; 25(11): 3006-3020.e7, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540935

ABSTRACT

There is a considerable need to identify those individuals with prostate cancer who have indolent disease. We propose that genes that control adult stem cell homeostasis in organs with slow turnover, such as the prostate, control cancer fate. One such gene, KLF4, overexpressed in murine prostate stem cells, regulates their homeostasis, blocks malignant transformation, and controls the self-renewal of tumor-initiating cells. KLF4 loss induces the molecular features of aggressive cancer and converts PIN lesions to invasive sarcomatoid carcinomas; its re-expression in vivo reverses this process. Bioinformatic analysis links these changes to human cancer. KLF4 and its downstream targets make up a gene signature that identifies indolent tumors and predicts recurrence-free survival. This approach may improve prognosis and identify therapeutic targets for advanced cancer.


Subject(s)
Disease Progression , Gene Expression Regulation, Neoplastic , Homeostasis , Kruppel-Like Transcription Factors/genetics , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cell Self Renewal/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , Phenotype , Prognosis
9.
Mol Cell Oncol ; 5(2): e1413495, 2018.
Article in English | MEDLINE | ID: mdl-29487897

ABSTRACT

Using a functional proliferation reporter we identified quiescent tumor propagating cancer cells (TPCs) in intact squamous cell carcinomas, and found that TGFß signaling controls their reversible entry into a growth arrested state, which protects TPCs from chemotherapy. TPCs with compromised TGFß/Smad signaling can't enter quiescence and subsequently die from chemotherapy.

10.
J Mol Biol ; 430(7): 1004-1023, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29458126

ABSTRACT

Hydrolysis of organic sulfate esters proceeds by two distinct mechanisms, water attacking at either sulfur (S-O bond cleavage) or carbon (C-O bond cleavage). In primary and secondary alkyl sulfates, attack at carbon is favored, whereas in aromatic sulfates and sulfated sugars, attack at sulfur is preferred. This mechanistic distinction is mirrored in the classification of enzymes that catalyze sulfate ester hydrolysis: arylsulfatases (ASs) catalyze S-O cleavage in sulfate sugars and arylsulfates, and alkyl sulfatases break the C-O bond of alkyl sulfates. Sinorhizobium meliloti choline sulfatase (SmCS) efficiently catalyzes the hydrolysis of alkyl sulfate choline-O-sulfate (kcat/KM=4.8×103s-1M-1) as well as arylsulfate 4-nitrophenyl sulfate (kcat/KM=12s-1M-1). Its 2.8-Å resolution X-ray structure shows a buried, largely hydrophobic active site in which a conserved glutamate (Glu386) plays a role in recognition of the quaternary ammonium group of the choline substrate. SmCS structurally resembles members of the alkaline phosphatase superfamily, being most closely related to dimeric ASs and tetrameric phosphonate monoester hydrolases. Although >70% of the amino acids between protomers align structurally (RMSDs 1.79-1.99Å), the oligomeric structures show distinctly different packing and protomer-protomer interfaces. The latter also play an important role in active site formation. Mutagenesis of the conserved active site residues typical for ASs, H218O-labeling studies and the observation of catalytically promiscuous behavior toward phosphoesters confirm the close relation to alkaline phosphatase superfamily members and suggest that SmCS is an AS that catalyzes S-O cleavage in alkyl sulfate esters with extreme catalytic proficiency.


Subject(s)
Sinorhizobium meliloti/enzymology , Sulfatases/chemistry , Biocatalysis , Catalytic Domain , Esters/metabolism , Models, Molecular , Protein Multimerization , Substrate Specificity , Sulfatases/classification , Sulfatases/metabolism
11.
Cell Stem Cell ; 21(5): 650-664.e8, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29100014

ABSTRACT

Squamous cell carcinomas (SCCs) are heterogeneous tumors sustained by tumor-propagating cancer cells (TPCs). SCCs frequently resist chemotherapy through still unknown mechanisms. Here, we combine H2B-GFP-based pulse-chasing with cell-surface markers to distinguish quiescent from proliferative TPCs within SCCs. We find that quiescent TPCs resist DNA damage and exhibit increased tumorigenic potential in response to chemotherapy, whereas proliferative TPCs undergo apoptosis. Quiescence is regulated by TGF-ß/SMAD signaling, which directly regulates cell-cycle gene transcription to control a reversible G1 cell-cycle arrest, independent of p21CIP function. Indeed, genetic or pharmacological TGF-ß inhibition increases the susceptibility of TPCs to chemotherapy because it prevents entry into a quiescent state. These findings provide direct evidence that TPCs can reversibly enter a quiescent, chemoresistant state and thereby underscore the need for combinatorial approaches to improve treatment of chemotherapy-resistant SCCs.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Drug Resistance, Neoplasm/drug effects , Head and Neck Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , Animals , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Chromatin/metabolism , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Humans , Mice , Signal Transduction/drug effects , Smad Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck , Staining and Labeling
12.
Cell Stem Cell ; 20(5): 575-577, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28475877

ABSTRACT

Head and neck squamous cell carcinomas (HNSCCs) are refractory to therapeutic interventions. Chen et al. (2017) show that mouse and human HNSCCs and their metastases depend on Bmi1-expressing cancer stem cells and AP1 signaling and that simultaneously inhibiting Bmi1 or AP1, combined with Cisplatin, reduces tumor growth effectively in preclinical models.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cisplatin/therapeutic use , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Polycomb Repressive Complex 1/metabolism , Animals , Antineoplastic Agents/therapeutic use , Humans , Mice , Models, Biological , Polycomb Repressive Complex 1/antagonists & inhibitors , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Tamoxifen/therapeutic use
13.
EMBO J ; 36(13): 1963-1980, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28507225

ABSTRACT

Tissue homeostasis of skin is sustained by epidermal progenitor cells localized within the basal layer of the skin epithelium. Post-translational modification of the proteome, such as protein phosphorylation, plays a fundamental role in the regulation of stemness and differentiation of somatic stem cells. However, it remains unclear how phosphoproteomic changes occur and contribute to epidermal differentiation. In this study, we survey the epidermal cell differentiation in a systematic manner by combining quantitative phosphoproteomics with mammalian kinome cDNA library screen. This approach identified a key signaling event, phosphorylation of a desmosome component, PKP1 (plakophilin-1) by RIPK4 (receptor-interacting serine-threonine kinase 4) during epidermal differentiation. With genome-editing and mouse genetics approach, we show that loss of function of either Pkp1 or Ripk4 impairs skin differentiation and enhances epidermal carcinogenesis in vivo Phosphorylation of PKP1's N-terminal domain by RIPK4 is essential for their role in epidermal differentiation. Taken together, our study presents a global view of phosphoproteomic changes that occur during epidermal differentiation, and identifies RIPK-PKP1 signaling as novel axis involved in skin stratification and tumorigenesis.


Subject(s)
Cell Differentiation , Keratinocytes/physiology , Plakophilins/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Skin/cytology , Stem Cells/physiology , Animals , Carcinogenesis , Cells, Cultured , Gene Expression Profiling , Mice , Mice, Knockout , Phosphorylation , Proteome/analysis , Skin Neoplasms , Tissue Transplantation
14.
Cell Stem Cell ; 19(6): 784-799, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27570068

ABSTRACT

Hematopoietic-specific transcription factors require coactivators to communicate with the general transcription machinery and establish transcriptional programs that maintain hematopoietic stem cell (HSC) self-renewal, promote differentiation, and prevent malignant transformation. Mediator is a large coactivator complex that bridges enhancer-localized transcription factors with promoters, but little is known about Mediator function in adult stem cell self-renewal and differentiation. We show that MED12, a member of the Mediator kinase module, is an essential regulator of HSC homeostasis, as in vivo deletion of Med12 causes rapid bone marrow aplasia leading to acute lethality. Deleting other members of the Mediator kinase module does not affect HSC function, suggesting kinase-independent roles of MED12. MED12 deletion destabilizes P300 binding at lineage-specific enhancers, resulting in H3K27Ac depletion, enhancer de-activation, and consequent loss of HSC stemness signatures. As MED12 mutations have been described recently in blood malignancies, alterations in MED12-dependent enhancer regulation may control both physiological and malignant hematopoiesis.


Subject(s)
Enhancer Elements, Genetic/genetics , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mediator Complex/metabolism , Animals , Apoptosis/genetics , Bone Marrow/pathology , Cell Survival/genetics , Chromatin/metabolism , Gene Deletion , Gene Expression Profiling , Mice , Protein Binding , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism
15.
PLoS One ; 9(12): e116114, 2014.
Article in English | MEDLINE | ID: mdl-25541984

ABSTRACT

Glioblastoma multiforme (GBM) is a deadly primary brain malignancy. Glioblastoma stem cells (GSC), which have the ability to self-renew and differentiate into tumor lineages, are believed to cause tumor recurrence due to their resistance to current therapies. A subset of GSCs is marked by cell surface expression of CD133, a glycosylated pentaspan transmembrane protein. The study of CD133-expressing GSCs has been limited by the relative paucity of genetic tools that specifically target them. Here, we present CD133-LV, a lentiviral vector presenting a single chain antibody against CD133 on its envelope, as a vehicle for the selective transduction of CD133-expressing GSCs. We show that CD133-LV selectively transduces CD133+ human GSCs in dose-dependent manner and that transduced cells maintain their stem-like properties. The transduction efficiency of CD133-LV is reduced by an antibody that recognizes the same epitope on CD133 as the viral envelope and by shRNA-mediated knockdown of CD133. Conversely, the rate of transduction by CD133-LV is augmented by overexpression of CD133 in primary human GBM cultures. CD133-LV selectively transduces CD133-expressing cells in intracranial human GBM xenografts in NOD.SCID mice, but spares normal mouse brain tissue, neurons derived from human embryonic stem cells and primary human astrocytes. Our findings indicate that CD133-LV represents a novel tool for the selective genetic manipulation of CD133-expressing GSCs, and can be used to answer important questions about how these cells contribute to tumor biology and therapy resistance.


Subject(s)
Antigens, CD/immunology , Brain Neoplasms/pathology , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Glioblastoma/pathology , Glycoproteins/immunology , Neoplastic Stem Cells/pathology , Peptides/immunology , Transduction, Genetic , AC133 Antigen , Animals , Antigens, CD/analysis , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Genetic Vectors/genetics , Genetic Vectors/immunology , Glioblastoma/genetics , Glioblastoma/immunology , Glycoproteins/analysis , Humans , Lentivirus/genetics , Lentivirus/immunology , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Peptides/analysis , Tumor Cells, Cultured
16.
European J Org Chem ; 2014(18): 3930-3034, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-25232289

ABSTRACT

The substrate scope of inverting alkylsulfatase Pisa1 was extended towards benzylic sec-sulfate esters by suppression of competing non-enzymatic autohydrolysis by addition of dimethyl sulfoxide as co-solvent. Detailed investigation of the mechanism of autohydrolysis in 18O-labeled buffer by using an enantiopure sec-benzylic sulfate ester as substrate revealed that from the three possible pathways (i) inverting SN2-type nucleophilic attack of [OH-] at the benzylic carbon represents the major pathway, whereas (ii) SN1-type formation of a planar benzylic carbenium ion leading to racemization was a minor event, and (iii) Retaining SN2-type nucleophilic attack at sulfur took place at the limits of detection. The data obtained are interpreted by analysis of Hammett constants of meta substituents.

17.
Nat Commun ; 5: 4511, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25077433

ABSTRACT

Although the principles that balance stem cell self-renewal and differentiation in normal tissue homeostasis are beginning to emerge, it is still unclear whether cancer cells with tumour initiating potential are similarly governed, or whether they have acquired distinct mechanisms to sustain self-renewal and long-term tumour growth. Here we show that the transcription factor Sox2, which is not expressed in normal skin epithelium and is dispensable for epidermal homeostasis, marks tumour initiating cells (TICs) in cutaneous squamous cell carcinomas (SCCs). We demonstrate that Sox2 is required for SCC growth in mouse and human, where it enhances Nrp1/Vegf signalling to promote the expansion of TICs along the tumour-stroma interface. Our findings suggest that distinct transcriptional programmes govern self-renewal and long-term growth of TICs and normal skin epithelial stem and progenitor cells. These programmes present promising diagnostic markers and targets for cancer-specific therapies.


Subject(s)
Carcinoma, Squamous Cell/genetics , Neoplastic Stem Cells/metabolism , Neuropilin-1/genetics , SOXB1 Transcription Factors/genetics , Skin Neoplasms/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/metabolism , Organ Specificity , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/metabolism , Signal Transduction , Skin/metabolism , Skin/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Stem Cells/cytology , Stem Cells/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , Transcription, Genetic , Tumor Microenvironment/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
PLoS One ; 9(3): e91343, 2014.
Article in English | MEDLINE | ID: mdl-24621654

ABSTRACT

Plastidic uracil salvage is essential for plant growth and development. So far, PLUTO, the plastidic nucleobase transporter from Arabidopsis thaliana is the only known uracil importer at the inner plastidic membrane which represents the permeability barrier of this organelle. We present the first homology model of PLUTO, the sole plant NCS1 member from Arabidopsis based on the crystal structure of the benzyl hydantoin transporter MHP1 from Microbacterium liquefaciens and validated by molecular dynamics simulations. Polar side chains of residues Glu-227 and backbones of Val-145, Gly-147 and Thr-425 are proposed to form the binding site for the three PLUTO substrates uracil, adenine and guanine. Mutational analysis and competition studies identified Glu-227 as an important residue for uracil and to a lesser extent for guanine transport. A differential response in substrate transport was apparent with PLUTO double mutants E227Q G147Q and E227Q T425A, both of which most strongly affected adenine transport, and in V145A G147Q, which markedly affected guanine transport. These differences could be explained by docking studies, showing that uracil and guanine exhibit a similar binding mode whereas adenine binds deep into the catalytic pocket of PLUTO. Furthermore, competition studies confirmed these results. The present study defines the molecular determinants for PLUTO substrate binding and demonstrates key differences in structure-function relations between PLUTO and other NCS1 family members.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis , Molecular Docking Simulation , Mutagenesis, Site-Directed , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/metabolism , Sequence Homology, Amino Acid , Amino Acid Sequence , Arabidopsis Proteins/genetics , Binding, Competitive , Molecular Sequence Data , Mutation , Nucleobase Transport Proteins/genetics , Structure-Activity Relationship , Substrate Specificity
19.
Appl Microbiol Biotechnol ; 98(4): 1485-96, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24352732

ABSTRACT

This review gives an overview on the occurrence of sulfatases in Prokaryota, Eukaryota and Archaea. The mechanism of enzymes acting with retention or inversion of configuration during sulfate ester hydrolysis is discussed taking two complementary examples. Methods for the discovery of novel alkyl sulfatases are described by way of sequence-based search and enzyme induction. A comprehensive list of organisms with their respective substrate scope regarding prim- and sec-alkyl sulfate esters allows to assess the capabilities and limitations of various biocatalysts employed as whole cell systems or as purified enzymes with respect to their activities and enantioselectivities. Methods for immobilization and selectivity enhancement by addition of metal ions or organic (co)solvents are summarised.


Subject(s)
Sulfatases/chemistry , Sulfatases/metabolism , Archaea/enzymology , Hydrolysis , Pseudomonas aeruginosa/enzymology , Stereoisomerism , Sulfuric Acid Esters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...