Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Biol Chem ; 284(34): 22544-8, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19561074

ABSTRACT

Wnt signaling plays important roles in various physiological and pathophysiological processes. The pathway that leads to beta-catenin stabilization is initiated by Wnt binding to its cell surface receptors, which induces the formation of phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P(2)) via activation of phosphatidylinositol 4-phosphate 5-kinase (PIP5K) type I. Here, we show that Wnt also stimulated the production of phosphatidylinositol 4-phosphate (PtdIns(4)P), which depended on Frizzled (Fz), Dishevelled (Dvl), and phosphatidylinositol 4-kinase (PI4K) type II alpha in HEK293T cells. Dvl directly interacted with and activated PI4KII alpha by increasing its V(max) for ATP and PtdIns. In addition, Dvl regulated PI4KII alpha and PIP5KI via different domains. Moreover, Dvl, PI4KII alpha, and PIP5KI appeared to form a ternary complex upon Wnt3a stimulation. This complex may allow efficient production of PtdIns(4,5)P(2) from PtdIns, which is far more abundant than PtdIns(4)P in cells. Therefore, this study provides new insights into the mechanism by which Wnt3a regulates the production of PtdIns(4,5)P(2).


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Wnt Proteins/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Dishevelled Proteins , Enzyme-Linked Immunosorbent Assay , Humans , Immunoprecipitation , Minor Histocompatibility Antigens , Phosphatidylinositol Phosphates/metabolism , Phosphoproteins/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Binding , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Wnt3 Protein , Wnt3A Protein
2.
Cancer Discov ; 9(9): 1208-1227, 2019 09.
Article in English | MEDLINE | ID: mdl-31217296

ABSTRACT

Immune checkpoint blockade (ICB) therapy, which targets T cell-inhibitory receptors, has revolutionized cancer treatment. Among the breast cancer subtypes, evaluation of ICB has been of greatest interest in triple-negative breast cancer (TNBC) due to its immunogenicity, as evidenced by the presence of tumor-infiltrating lymphocytes and elevated PD-L1 expression relative to other subtypes. TNBC incidence is equally distributed across the age spectrum, affecting 10% to 15% of women in all age groups. Here we report that increased immune dysfunction with age limits ICB efficacy in aged TNBC-bearing mice. The tumor microenvironment in both aged mice and patients with TNBC shows decreased IFN signaling and antigen presentation, suggesting failed innate immune activation with age. Triggering innate immune priming with a STING agonist restored response to ICB in aged mice. Our data implicate age-related immune dysfunction as a mechanism of ICB resistance in mice and suggest potential prognostic utility of assessing IFN-related genes in patients with TNBC receiving ICB therapy. SIGNIFICANCE: These data demonstrate for the first time that age determines the T cell-inflamed phenotype in TNBC and affects response to ICB in mice. Evaluating IFN-related genes from tumor genomic data may aid identification of patients for whom combination therapy including an IFN pathway activator with ICB may be required.This article is highlighted in the In This Issue feature, p. 1143.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Interferon-gamma/administration & dosage , Interferons/metabolism , Triple Negative Breast Neoplasms/drug therapy , Xanthones/administration & dosage , Age Factors , Animals , Antigen Presentation , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Female , Humans , Interferon-gamma/pharmacology , Mice , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment , Xanthones/pharmacology , Xenograft Model Antitumor Assays
3.
Nat Cell Biol ; 21(7): 879-888, 2019 07.
Article in English | MEDLINE | ID: mdl-31263265

ABSTRACT

Most human tumours are heterogeneous, composed of cellular clones with different properties present at variable frequencies. Highly heterogeneous tumours have poor clinical outcomes, yet the underlying mechanism remains poorly understood. Here, we show that minor subclones of breast cancer cells expressing IL11 and FIGF (VEGFD) cooperate to promote metastatic progression and generate polyclonal metastases composed of driver and neutral subclones. Expression profiling of the epithelial and stromal compartments of monoclonal and polyclonal primary and metastatic lesions revealed that this cooperation is indirect, mediated through the local and systemic microenvironments. We identified neutrophils as a leukocyte population stimulated by the IL11-expressing minor subclone and showed that the depletion of neutrophils prevents metastatic outgrowth. Single-cell RNA-seq of CD45+ cell populations from primary tumours, blood and lungs demonstrated that IL11 acts on bone-marrow-derived mesenchymal stromal cells, which induce pro-tumorigenic and pro-metastatic neutrophils. Our results indicate key roles for non-cell-autonomous drivers and minor subclones in metastasis.


Subject(s)
Breast Neoplasms/pathology , Lung Neoplasms/pathology , Neoplasm Metastasis/pathology , Neutrophils/metabolism , Tumor Microenvironment , Animals , Carcinogenesis/metabolism , Disease Progression , Humans , Lung/pathology , Lung Neoplasms/secondary , Mesenchymal Stem Cells/cytology
5.
PLoS One ; 13(6): e0198790, 2018.
Article in English | MEDLINE | ID: mdl-29897959

ABSTRACT

Gene editing protocols often require the use of a subcloning step to isolate successfully edited cells, the behavior of which is then compared to the aggregate parental population and/or other non-edited subclones. Here we demonstrate that the inherent functional heterogeneity present in many cell lines can render these populations inappropriate controls, resulting in erroneous interpretations of experimental findings. We describe a novel CRISPR/Cas9 protocol that incorporates a single-cell cloning step prior to gene editing, allowing for the generation of appropriately matched, functionally equivalent control and edited cell lines. As a proof of concept, we generated matched control and osteopontin-knockout Her2+ and Estrogen receptor-negative murine mammary carcinoma cell lines and demonstrated that the osteopontin-knockout cell lines exhibit the expected biological phenotypes, including unaffected primary tumor growth kinetics and reduced metastatic outgrowth in female FVB mice. Using these matched cell lines, we discovered that osteopontin-knockout mammary tumors were more sensitive than control tumors to chemotherapy in vivo. Our results demonstrate that heterogeneity must be considered during experimental design when utilizing gene editing protocols and provide a solution to account for it.


Subject(s)
Antineoplastic Agents/therapeutic use , CRISPR-Cas Systems/genetics , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Disease Progression , Gene Editing , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology , Osteopontin/analysis , Osteopontin/deficiency , Osteopontin/genetics , Phenotype , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Transplantation, Heterologous
6.
Cancer Res ; 78(18): 5300-5314, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30065048

ABSTRACT

The presence of disseminated tumor cells in breast cancer patient bone marrow aspirates predicts decreased recurrence-free survival. Although it is appreciated that physiologic, pathologic, and therapeutic conditions impact hematopoiesis, it remains unclear whether targeting hematopoiesis presents opportunities for limiting bone metastasis. Using preclinical breast cancer models, we discovered that marrow from mice treated with the bisphosphonate zoledronic acid (ZA) are metastasis-suppressive. Specifically, ZA modulated hematopoietic myeloid/osteoclast progenitor cell (M/OCP) lineage potential to activate metastasis-suppressive activity. Granulocyte-colony stimulating factor (G-CSF) promoted ZA resistance by redirecting M/OCP differentiation. We identified M/OCP and bone marrow transcriptional programs associated with metastasis suppression and ZA resistance. Analysis of patient blood samples taken at randomization revealed that women with high-plasma G-CSF experienced significantly worse outcome with adjuvant ZA than those with lower G-CSF levels. Our findings support discovery of therapeutic strategies to direct M/OCP lineage potential and biomarkers that stratify responses in patients at risk of recurrence.Significance: Bone marrow myeloid/osteoclast progenitor cell lineage potential has a profound impact on breast cancer bone metastasis and can be modulated by G-CSF and bone-targeting agents. Cancer Res; 78(18); 5300-14. ©2018 AACR.


Subject(s)
Bone Marrow Cells/cytology , Breast Neoplasms/pathology , Cell Lineage , Hematopoietic Stem Cells/cytology , Neoplasm Metastasis/prevention & control , Animals , Antineoplastic Agents/pharmacology , Biomarkers/metabolism , Bone Marrow/pathology , Bone Neoplasms/prevention & control , Cell Differentiation , Cell Line, Tumor , Female , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoiesis , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Recurrence, Local , Osteoclasts/cytology , Osteoclasts/metabolism , Zoledronic Acid/pharmacology
7.
Cancer Res ; 76(10): 2932-43, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27197230

ABSTRACT

Triple-negative breast cancer (TNBC) is considered an early onset subtype of breast cancer that carries with it a poorer prognosis in young rather than older women for reasons that remain poorly understood. Hematopoiesis in the bone marrow becomes altered with age and may therefore affect the composition of tumor-infiltrating hematopoietic cells and subsequent tumor progression. In this study, we investigated how age- and tumor-dependent changes to bone marrow-derived hematopoietic cells impact TNBC progression. Using multiple mouse models of TNBC tumorigenesis and metastasis, we found that a specific population of bone marrow cells (BMC) upregulated CSF-1R and secreted the growth factor granulin to support stromal activation and robust tumor growth in young mice. However, the same cell population in old mice expressed low levels of CSF1R and granulin and failed to promote tumor outgrowth, suggesting that age influences the tumorigenic capacity of BMCs in response to tumor-associated signals. Importantly, BMCs from young mice were sufficient to activate a tumor-supportive microenvironment and induce tumor progression in old mice. These results indicate that hematopoietic age is an important determinant of TNBC aggressiveness and provide rationale for investigating age-stratified therapies designed to prevent the protumorigenic effects of activated BMCs. Cancer Res; 76(10); 2932-43. ©2016 AACR.


Subject(s)
Bone Marrow Cells/pathology , Hematopoiesis/physiology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment , Age Factors , Age of Onset , Animals , Apoptosis , Blotting, Western , Bone Marrow Cells/metabolism , Cell Proliferation , Disease Progression , Female , Flow Cytometry , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Progranulins , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Nat Commun ; 6: 8205, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26365782

ABSTRACT

Type I phosphatidylinositol phosphate kinase (PIP5K1) phosphorylates the head group of phosphatidylinositol 4-phosphate (PtdIns4P) to generate PtdIns4,5P2, which plays important roles in a wide range of cellular functions including Wnt signalling. However, the lack of its structural information has hindered the understanding of its regulation. Here we report the crystal structure of the catalytic domain of zebrafish PIP5K1A at 3.3 Å resolution. This molecule forms a side-to-side dimer. Mutagenesis study of PIP5K1A reveals two adjacent interfaces for the dimerization and interaction with the DIX domain of the Wnt signalling molecule dishevelled. Although these interfaces are located distally to the catalytic/substrate-binding site, binding to these interfaces either through dimerization or the interaction with DIX stimulates PIP5K1 catalytic activity. DIX binding additionally enhances PIP5K1 substrate binding. Thus, this study elucidates regulatory mechanisms for this lipid kinase and provides a paradigm for the understanding of PIP5K1 regulation by their interacting molecules.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dimerization , Phosphoproteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Binding Sites , Calorimetry , Catalytic Domain , Circular Dichroism , Crystallization , Crystallography, X-Ray , Dishevelled Proteins , HEK293 Cells , Humans , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Protein Structure, Tertiary , Zebrafish
9.
Cancer Discov ; 4(7): 760-1, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25002612

ABSTRACT

SUMMARY: Disease recurrence is the most common cause of death for patients with breast cancer, yet little is known about the molecular mechanisms underlying this process. Using inducible transgenic mouse model systems, Feng and colleagues identified SPSB1 as a determinant of breast cancer recurrence by virtue of its ability to protect tumor cells from apoptosis through c-MET activation.


Subject(s)
Breast Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Proto-Oncogene Proteins c-met/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , Animals , Female , Humans
10.
J Mol Cell Biol ; 6(3): 231-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24748645

ABSTRACT

Thymine DNA glycosylase (TDG), an enzyme that initiates the repair of G/T and G/U mismatches, has been lately found crucial in embryonic development to maintain epigenetic stability and facilitate the active DNA demethylation. Here we report a novel role of TDG in Wnt signaling as a transcriptional coactivator of ß-catenin/TCFs complex. Our data show that TDG binds to the transcriptional factor family LEF1/TCFs and potentiates ß-catenin/TCFs transactivation, while TDG depletion suppresses Wnt3a-stimulated reporter activity or target gene transcription. Next, we show that CBP, a known coactivator, is also required for TDG function through forming a cooperative complex on target promoters. Moreover, there is an elevation of TDG levels in human colon cancer tissue, and knockdown of TDG inhibits proliferation of the colon cells. Overall, our results reveal that TDG, as a new coactivator, promotes ß-catenin/TCFs transactivation and functionally cooperates with CBP in canonical Wnt signaling.


Subject(s)
Lymphoid Enhancer-Binding Factor 1/genetics , Peptide Fragments/metabolism , Sialoglycoproteins/metabolism , Thymine DNA Glycosylase/metabolism , Transcriptional Activation/genetics , beta Catenin/genetics , Animals , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HEK293 Cells , HeLa Cells , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Up-Regulation/genetics , Wnt Signaling Pathway/genetics , beta Catenin/metabolism
11.
Science ; 321(5894): 1350-3, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18772438

ABSTRACT

The canonical Wnt-beta-catenin signaling pathway is initiated by inducing phosphorylation of one of the Wnt receptors, low-density lipoprotein receptor-related protein 6 (LRP6), at threonine residue 1479 (Thr1479) and serine residue 1490 (Ser1490). By screening a human kinase small interfering RNA library, we identified phosphatidylinositol 4-kinase type II alpha and phosphatidylinositol-4-phosphate 5-kinase type I (PIP5KI) as required for Wnt3a-induced LRP6 phosphorylation at Ser1490 in mammalian cells and confirmed that these kinases are important for Wnt signaling in Xenopus embryos. Wnt3a stimulates the formation of phosphatidylinositol 4,5-bisphosphates [PtdIns (4,5)P2] through frizzled and dishevelled, the latter of which directly interacted with and activated PIP5KI. In turn, PtdIns (4,5)P2 regulated phosphorylation of LRP6 at Thr1479 and Ser1490. Therefore, our study reveals a signaling mechanism for Wnt to regulate LRP6 phosphorylation.


Subject(s)
LDL-Receptor Related Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Axin Protein , Cell Line , Dishevelled Proteins , Frizzled Receptors/metabolism , Humans , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , Minor Histocompatibility Antigens , Models, Biological , Phosphoproteins/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA, Small Interfering , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Serine/metabolism , Signal Transduction , Threonine/metabolism , Wnt3 Protein , Wnt3A Protein , Xenopus/embryology , Xenopus Proteins
SELECTION OF CITATIONS
SEARCH DETAIL