Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Proc Natl Acad Sci U S A ; 119(43): e2203180119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36269860

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway represents the most hyperactivated oncogenic pathway in triple-negative breast cancer (TNBC), a highly aggressive tumor subtype encompassing ∼15% of breast cancers and which possesses no targeted therapeutics. Despite critical contributions of its signaling arms to disease pathogenesis, PI3K pathway inhibitors have not achieved expected clinical responses in TNBC, owing largely to a still-incomplete understanding of the compensatory cascades that operate downstream of PI3K. Here, we investigated the contributions of long noncoding RNAs (lncRNAs) to PI3K activities in clinical and experimental TNBC and discovered a prominent role for LINC01133 as a PI3K-AKT signaling effector. We found that LINC01133 exerted protumorigenic roles in TNBC and that it governed a previously undescribed mTOR Complex 2 (mTORC2)-dependent pathway that activated AKT in a PI3K-independent manner. Mechanistically, LINC01133 induced the expression of the mTORC2 component PROTOR1/PRR5 by competitively coupling away its negative messenger RNA (mRNA) regulator, the heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2B1). PROTOR1/PRR5 in turn was sufficient and necessary for LINC01133-triggered functions, casting previously unappreciated roles for this Rictor-binding protein in cellular signaling and growth. Notably, LINC01133 antagonism undermined cellular growth, and we show that the LINC01133-PROTOR1/PRR5 pathway was tightly associated with TNBC poor patient survival. Altogether, our findings uncovered a lncRNA-driven signaling shunt that acts as a critical determinant of malignancy downstream of the PI3K pathway and as a potential RNA therapeutic target in clinical TNBC management.


Subject(s)
RNA, Long Noncoding , Triple Negative Breast Neoplasms , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Triple Negative Breast Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Cell Proliferation/genetics , Phosphoinositide-3 Kinase Inhibitors , Mechanistic Target of Rapamycin Complex 2/metabolism , RNA, Messenger , Heterogeneous-Nuclear Ribonucleoproteins , Cell Line, Tumor
2.
Semin Cancer Biol ; 86(Pt 2): 81-92, 2022 11.
Article in English | MEDLINE | ID: mdl-36087857

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) encompass a heterogeneous population of fibroblastic progenitor cells that reside in multiple tissues around the body. They are endowed with capacities to differentiate into multiple connective tissue lineages, including chondrocytes, adipocytes, and osteoblasts, and are thought to function as trophic cells recruited to sites of injury and inflammation where they contribute to tissue regeneration. In keeping with these roles, MSCs also to home to sites of breast tumorigenesis, akin to their migration to wounds, and participate in tumor stroma formation. Mounting evidence over the past two decades has described the critical regulatory roles for tumor-associated MSCs in various aspects of breast tumor pathogenesis, be it tumor initiation, growth, angiogenesis, tumor microenvironment formation, immune evasion, cancer cell migration, invasion, survival, therapeutic resistance, dissemination, and metastatic colonization. In this review, we present a brief summary of the role of MSCs in breast tumor development and progression, highlight some of the molecular frameworks underlying their pro-malignant contributions, and present evidence of their promising utility in breast cancer therapy.


Subject(s)
Breast Neoplasms , Mesenchymal Stem Cells , Humans , Female , Breast Neoplasms/etiology , Breast Neoplasms/therapy , Breast Neoplasms/pathology , Mesenchymal Stem Cells/pathology , Tumor Microenvironment , Cell Movement , Adipocytes , Cell Transformation, Neoplastic/pathology
3.
Stem Cells ; 37(10): 1281-1292, 2019 10.
Article in English | MEDLINE | ID: mdl-31283068

ABSTRACT

The fibrotic tumor microenvironment is a critical player in the pathogenesis of triple-negative breast cancers (TNBCs), with the presence of fibroblastic infiltrates particularly correlating with tumors that are clinically advanced. On this front, we previously demonstrated that TNBCs are highly enriched in fibroblastic stromal progenitor cells called mesenchymal stem/stromal cells (MSCs) and that such cells play critical roles in promoting TNBC initiation and progression. How TNBC cells respond to MSC stimulation, however, is not fully understood, and stands to reveal contextual signals used by TNBC cells during tumor development and provide biomarkers and therapeutic targets of pertinence to TNBC management. Here, we report that MSCs strongly induced the long noncoding RNA (lncRNA) LINC01133 in neighboring TNBC cells. Indeed, although lncRNAs have been tightly associated with cancer development, their contributions to breast cancer in general, and to TNBC pathogenesis in particular, have not been fully elucidated, and we set out to determine if LINC01133 regulated malignant traits in TNBC cells. We establish that LINC01133 is sufficient, on its own, in promoting phenotypic and growth characteristics of cancer stem cell-like cells, and that it is a direct mediator of the MSC-triggered miR-199a-FOXP2 pathway in TNBC models. Furthermore, we show that LINC01133 is a critical regulator of the pluripotency-determining gene Kruppel-Like Factor 4 (KLF4), and that it represents a biomarker and prognosticator of disease outcome in the clinic. Collectively, our findings introduce LINC01133 as a novel functional driver of malignancy and a potential theranostic in TNBC. Stem Cells 2019;37:1281-1292.


Subject(s)
Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/genetics , Triple Negative Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Female , Humans , Kruppel-Like Factor 4 , Neoplastic Stem Cells/pathology , Phenotype , RNA, Long Noncoding/metabolism , Triple Negative Breast Neoplasms/diet therapy , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
4.
Nat Rev Mol Cell Biol ; 9(7): 517-31, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18568040

ABSTRACT

Extensive research on the Ras proteins and their functions in cell physiology over the past 30 years has led to numerous insights that have revealed the involvement of Ras not only in tumorigenesis but also in many developmental disorders. Despite great strides in our understanding of the molecular and cellular mechanisms of action of the Ras proteins, the expanding roster of their downstream effectors and the complexity of the signalling cascades that they regulate indicate that much remains to be learnt.


Subject(s)
Genes, ras , Signal Transduction/physiology , ras Proteins/metabolism , Amino Acid Sequence , Animals , Cell Transformation, Neoplastic , DNA-Binding Proteins/metabolism , Enzyme Activation , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Phylogeny , Protein Conformation , Protein Processing, Post-Translational , Sequence Alignment , Transcription Factors/metabolism , ras Proteins/chemistry , ras Proteins/classification , ras Proteins/genetics
5.
Cancer Cell ; 12(2): 160-70, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17692807

ABSTRACT

We investigated the influence of normal cell phenotype on the neoplastic phenotype by comparing tumors derived from two different normal human mammary epithelial cell populations, one of which was isolated using a new culture medium. Transformation of these two cell populations with the same set of genetic elements yielded cells that formed tumor xenografts exhibiting major differences in histopathology, tumorigenicity, and metastatic behavior. While one cell type (HMECs) yielded squamous cell carcinomas, the other cell type (BPECs) yielded tumors closely resembling human breast adenocarcinomas. Transformed BPECs gave rise to lung metastases and were up to 10(4)-fold more tumorigenic than transformed HMECs, which are nonmetastatic. Hence, the pre-existing differences between BPECs and HMECs strongly influence the phenotypes of their transformed derivatives.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast/cytology , Cell Transformation, Neoplastic , Epithelial Cells/cytology , Adenocarcinoma/etiology , Adenocarcinoma/pathology , Adult , Animals , Antigens, Polyomavirus Transforming/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Cell Division , Cells, Cultured , Female , Gene Expression Profiling , Genes, ras/physiology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Middle Aged , Transplantation, Heterologous
6.
Proc Natl Acad Sci U S A ; 109(43): 17460-5, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23033492

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent progenitor cells with the ability to differentiate into multiple mesoderm lineages in the course of normal tissue homeostasis or during injury. We have previously shown that MSCs migrate to sites of tumorigenesis, where they become activated by cancer cells to promote metastasis. However, the molecular and phenotypic attributes of the MSC-induced metastatic state of the cancer cells remained undetermined. Here, we show that bone marrow-derived human MSCs promote de novo production of lysyl oxidase (LOX) from human breast carcinoma cells, which is sufficient to enhance the metastasis of otherwise weakly metastatic cancer cells to the lungs and bones. We also show that LOX is an essential component of the CD44-Twist signaling axis, in which extracellular hyaluronan causes nuclear translocation of CD44 in the cancer cells, thus triggering LOX transcription by associating with its promoter. Processed and enzymatically active LOX, in turn, stimulates Twist transcription, which mediates the MSC-triggered epithelial-to-mesenchymal transition (EMT) of carcinoma cells. Surprisingly, although induction of EMT in breast cancer cells has been tightly associated with the generation of cancer stem cells, we find that LOX, despite being critical for EMT, does not contribute to the ability of MSCs to promote the formation of cancer stem cells in the carcinoma cell populations. Collectively, our studies highlight a critical role for LOX in cancer metastasis and indicate that the signaling pathways controlling stroma-induced EMT are distinct from pathways regulating the development of cancer stem cells.


Subject(s)
Breast Neoplasms/enzymology , Mesenchymal Stem Cells/enzymology , Protein-Lysine 6-Oxidase/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplastic Stem Cells/enzymology , Protein-Lysine 6-Oxidase/genetics , RNA, Messenger/genetics
7.
Nature ; 449(7162): 557-63, 2007 Oct 04.
Article in English | MEDLINE | ID: mdl-17914389

ABSTRACT

Mesenchymal stem cells have been recently described to localize to breast carcinomas, where they integrate into the tumour-associated stroma. However, the involvement of mesenchymal stem cells (or their derivatives) in tumour pathophysiology has not been addressed. Here, we demonstrate that bone-marrow-derived human mesenchymal stem cells, when mixed with otherwise weakly metastatic human breast carcinoma cells, cause the cancer cells to increase their metastatic potency greatly when this cell mixture is introduced into a subcutaneous site and allowed to form a tumour xenograft. The breast cancer cells stimulate de novo secretion of the chemokine CCL5 (also called RANTES) from mesenchymal stem cells, which then acts in a paracrine fashion on the cancer cells to enhance their motility, invasion and metastasis. This enhanced metastatic ability is reversible and is dependent on CCL5 signalling through the chemokine receptor CCR5. Collectively, these data demonstrate that the tumour microenvironment facilitates metastatic spread by eliciting reversible changes in the phenotype of cancer cells.


Subject(s)
Breast Neoplasms/pathology , Mesenchymal Stem Cells/pathology , Neoplasm Metastasis , Stromal Cells/pathology , Animals , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Movement , Chemokine CCL5 , Chemokines, CC/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Paracrine Communication , Receptors, CCR5/metabolism , Stromal Cells/metabolism
8.
Nat Cell Biol ; 25(7): 950-962, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37400498

ABSTRACT

The prolyl hydroxylation of hypoxia-inducible factor 1α (HIF-1α) mediated by the EGLN-pVHL pathway represents a classic signalling mechanism that mediates cellular adaptation under hypoxia. Here we identify RIPK1, a known regulator of cell death mediated by tumour necrosis factor receptor 1 (TNFR1), as a target of EGLN1-pVHL. Prolyl hydroxylation of RIPK1 mediated by EGLN1 promotes the binding of RIPK1 with pVHL to suppress its activation under normoxic conditions. Prolonged hypoxia promotes the activation of RIPK1 kinase by modulating its proline hydroxylation, independent of the TNFα-TNFR1 pathway. As such, inhibiting proline hydroxylation of RIPK1 promotes RIPK1 activation to trigger cell death and inflammation. Hepatocyte-specific Vhl deficiency promoted RIPK1-dependent apoptosis to mediate liver pathology. Our findings illustrate a key role of the EGLN-pVHL pathway in suppressing RIPK1 activation under normoxic conditions to promote cell survival and a model by which hypoxia promotes RIPK1 activation through modulating its proline hydroxylation to mediate cell death and inflammation in human diseases, independent of TNFR1.


Subject(s)
Necroptosis , Receptors, Tumor Necrosis Factor, Type I , Humans , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Hydroxylation , Hypoxia , Proline/metabolism , Inflammation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
9.
STAR Protoc ; 3(2): 101413, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35634360

ABSTRACT

We provide a protocol for gain-of-function (GOF) cDNA screen of genes that foster cancer cell colonization of secondary tissues, the last and most lethal step of the metastasis cascade. We present techniques for cDNA viral library preparation and delivery leading up to the recovery of colonization-promoting sequences in a proof-of-concept DU145-based mouse model of pulmonary metastasis. Adapted to other cDNA libraries and cancer models, this approach would prove widely useful in enumerating intrinsic genetic determinants underlying metastatic colonization. For complete details on the use and execution of this protocol, please refer to Tu et al. (2021).


Subject(s)
Gain of Function Mutation , Lung Neoplasms , Animals , DNA, Complementary/genetics , Disease Models, Animal , Gene Library , Lung Neoplasms/genetics , Mice
10.
J Mammary Gland Biol Neoplasia ; 15(4): 399-409, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21170761

ABSTRACT

Mesenchymal stem cells (MSCs) are a heterogeneous mix of stromal stem cells that can give rise to cells of mesodermal lineages, namely adipocytes, osteocytes and chondrocytes. They can home to sites of injury where they promote the repair and regeneration of damaged tissues. MSCs also home to sites of tumorigenesis, and as such, are utilized as efficient cellular vehicles for the delivery of anti-neoplastic therapeutics. Recently, MSCs within the tumor microenvironment have been shown to contribute to the desmoplastic reaction and to facilitate tumor formation and progression, sparking renewed interest in their pro-tumorigenic attributes and their roles as tumor stromal cells. Here, we describe the evidence linking MSCs to inflammatory processes and breast cancer development, and discuss their newly discovered physiological roles in the context of the tumor microenvironment.


Subject(s)
Breast Neoplasms/pathology , Breast Neoplasms/therapy , Mesenchymal Stem Cells/pathology , Animals , Disease Progression , Female , Humans , Tumor Microenvironment
11.
Mol Cell Endocrinol ; 535: 111374, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34242715

ABSTRACT

Cancer cells within breast tumors exist within a hierarchy in which only a small and rare subset of cells is able to regenerate growths with the heterogeneity of the original tumor. These highly malignant cancer cells, which behave like stem cells for new cancers and are called "cancer stem cells" or CSCs, have also been shown to possess increased resistance to therapeutics, and represent the root cause underlying therapy failures, persistence of residual disease, and relapse. As >90% of cancer deaths are due to refractory tumors, identification of critical molecular drivers of the CSC-state would reveal vulnerabilities that can be leveraged in designing therapeutics that eradicate advanced disease and improve patient survival outcomes. An expanding and complex body of work has now described the exquisite susceptibility of CSC pools to the regulatory influences of local and systemic hormones. Indeed, breast CSCs express a plethora of hormonal receptors, which funnel hormonal influences over every aspect of breast neoplasia - be it tumor onset, growth, survival, invasion, metastasis, or therapy resistance - via directly impacting CSC behavior. This article is intended to shed light on this active area of investigation by attempting to provide a systematic and comprehensive overview of the available evidence directly linking hormones to breast CSC biology.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Neoplastic Stem Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Signal Transduction , Survival Analysis
12.
iScience ; 24(5): 102425, 2021 May 21.
Article in English | MEDLINE | ID: mdl-34036247

ABSTRACT

Elucidations of the factors that promote the growth of disseminated tumor cells (DTCs) into life-threatening lesions stand to provide much needed prognostic and therapeutic targets of translational utility for patients with metastatic cancer. To identify such regulators, we conducted gain-of-function cDNA library screening to discover genes that foster prostate cancer cell colonization of mouse lungs as an experimental model. Our efforts identified the metabolic enzyme aldolase A (ALDOA) as a driver of cancer cell motility, anchorage-independent growth, and metastatic colonization, and as a prognosticator of adverse patient outcome across many malignancies, including prostate, breast, pancreatic, and liver cancers. Metabolomics coupled with biochemical and functional analyses revealed that ALDOA triggered the activation of adenosine-5'-monophosphate (AMP)-activated protein kinase (AMPK), which we demonstrate played essential promalignant activities in ALDOA-expressing cells. Collectively, these findings unveiled vivo approaches to identify metastatic colonization regulators and uncovered previously undescribed roles for ALDOA-AMPK pathway in tumor progression.

13.
NPJ Breast Cancer ; 7(1): 69, 2021 May 31.
Article in English | MEDLINE | ID: mdl-34059683

ABSTRACT

The development of triple-negative breast cancer (TNBC) is critically regulated by certain tumor-microenvironment-associated cells called mesenchymal stem/stromal cells (MSCs), which we and others have shown promote TNBC progression by activating pro-malignant signaling in neighboring cancer cells. Characterization of these cascades would better our understanding of TNBC biology and bring about therapeutics that eliminate the morbidity and mortality associated with advanced disease. Here, we focused on the emerging class of RNAs called long non-coding RNAs or lncRNAs and utilized a MSC-supported TNBC progression model to identify specific family members of functional relevance to TNBC pathogenesis. Indeed, although some have been described to play functional roles in TNBC, activities of lncRNAs as mediators of tumor-microenvironment-driven TNBC development remain to be fully explored. We report that MSCs stimulate robust expression of LINC01119 in TNBC cells, which in turn induces suppressor of cytokine signaling 5 (SOCS5), leading to accelerated cancer cell growth and tumorigenesis. We show that LINC01119 and SOCS5 exhibit tight correlation across multiple breast cancer gene sets and that they are highly enriched in TNBC patient cohorts. Importantly, we present evidence that the LINC01119-SOCS5 axis represents a powerful prognostic indicator of adverse outcomes in TNBC patients, and demonstrate that its repression severely impairs cancer cell growth. Altogether, our findings identify LINC01119 as a major driver of TNBC development and delineate critical non-coding RNA theranostics of potential translational utility in the management of advanced TNBC, a class of tumors in most need of effective and targeted therapy.

14.
Sci Signal ; 10(467)2017 02 21.
Article in English | MEDLINE | ID: mdl-28223415

ABSTRACT

This Podcast features an interview with Antoine Karnoub, senior author of a Research Article that appears in the 21 February 2017 issue of Science Signaling, about how pentraxin-3 (PTX3) links increased phosphoinositide 3-kinase (PI3K) signaling to stem cell properties in basal-like breast cancer (BLBC). BLBC is an aggressive type of breast cancer in which the tumor cells exhibit increased PI3K signaling and have stem cell-like properties. Thomas et al found that aberrant PI3K signaling in BLBCs stimulated the expression of PTX3, which encodes a protein that functions in innate immunity. PTX3 promoted stem cell-like traits in mammary epithelial cells and stimulated the growth of breast cancer cells. Conversely, decreasing the abundance of PTX3 in breast cancer cells reduced both the growth of these cells and their expression of stem cell markers. The abundance of PTX3 transcripts in breast tumors negatively correlated with patient survival, suggesting that PTX3 may be a useful biomarker for stratifying BLBC patients and that targeting PTX3 may suppress tumor growth in some BLBC patients.Listen to Podcast.


Subject(s)
Breast Neoplasms , Phosphatidylinositol 3-Kinase , C-Reactive Protein , Humans , Phosphatidylinositol 3-Kinases , Signal Transduction , Stem Cells
15.
Oncol Hematol Rev ; 13(1): 45-55, 2017.
Article in English | MEDLINE | ID: mdl-33959299

ABSTRACT

Metastasis is often accompanied by radio- and chemotherapeutic resistance to anticancer treatments and is the major cause of death in cancer patients. Better understanding of how cancer cells circumvent therapeutic insults and how disseminated cancer clones generate life-threatening metastases would therefore be paramount to the development of effective therapeutic approaches for clinical management of malignant disease. Mounting reports over the past two decades have provided evidence for the existence of a minor population of highly malignant cells within liquid and solid tumors, which are capable of self-renewing and of regenerating secondary growths with the heterogeneity of the primary tumors from which they derive. These cells, called tumor-initiating cells or cancer stem cells (CSCs) exhibit increased resistance to standard radio- and chemotherapies and appear to have mechanisms that enable them to evade immune surveillance. CSCs are therefore considered to be responsible for systemic residual disease after cancer therapy, as well as for disease relapse. How CSCs develop, the nature of the interactions they establish with their microenvironment, their phenotypic and functional characteristics, as well as their molecular dependencies have all taken center stage in cancer therapy. Indeed, improved understanding of CSC biology is critical to the development of important CSC-based anti-neoplastic approaches that have the potential to radically improve cancer management. Here, we summarize some of the most pertinent elements regarding CSC development and properties, and highlight some of the clinical modalities in current development as anti-CSC therapeutics.

16.
Cancer Res ; 77(2): 268-278, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27742687

ABSTRACT

Lysyl oxidase (LOX) is a secreted copper-dependent amine oxidase whose primary function is to drive collagen crosslinking and extracellular matrix stiffness. LOX in colorectal cancer synergizes with hypoxia-inducible factor-1 (HIF-1) to promote tumor progression. Here we investigated whether LOX/HIF1 endows colorectal cancer cells with full competence for aggressive colonization in bone. We show that a high LOX expression in primary tumors from patients with colorectal cancer was associated with poor clinical outcome, irrespective of HIF-1 In addition, LOX was expressed by tumor cells in the bone marrow from colorectal cancer patients with bone metastases. In vivo experimental studies show that LOX overexpression in colorectal cancer cells or systemic delivery of the conditioned medium from LOX-overexpressing colorectal cancer cells promoted tumor cell dissemination in the bone marrow and enhanced osteolytic lesion formation, irrespective of HIF-1 Conversely, silencing or pharmacologic inhibition of LOX activity blocked dissemination of colorectal cancer cells in the bone marrow and tumor-driven osteolytic lesion formation. In vitro, tumor-secreted LOX supported the attachment and survival of colorectal cancer cells to and in the bone matrix, and inhibited osteoblast differentiation. LOX overexpression in colorectal cancer cells also induced a robust production of IL6. In turn, both LOX and IL6 were acting in concert to promote RANKL-dependent osteoclast differentiation, thereby creating an imbalance between bone resorption and bone formation. Collectively, our findings show that LOX supports colorectal cancer cell dissemination in the bone marrow and they reveal a novel mechanism through which LOX-driven IL6 production by colorectal cancer cells impairs bone homeostasis. Cancer Res; 77(2); 268-78. ©2016 AACR.


Subject(s)
Bone Neoplasms/secondary , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/secondary , Neoplasm Invasiveness/pathology , Protein-Lysine 6-Oxidase/metabolism , Animals , Blotting, Western , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Heterografts , Humans , Immunohistochemistry , Interleukin-6/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Real-Time Polymerase Chain Reaction
17.
Sci Signal ; 10(467)2017 02 21.
Article in English | MEDLINE | ID: mdl-28223411

ABSTRACT

Basal-like breast cancers (BLBCs) exhibit hyperactivation of the phosphoinositide 3-kinase (PI3K) signaling pathway because of the frequent mutational activation of the PIK3CA catalytic subunit and the genetic loss of its negative regulators PTEN (phosphatase and tensin homolog) and INPP4B (inositol polyphosphate-4-phosphatase type II). However, PI3K inhibitors have had limited clinical efficacy in BLBC management because of compensatory amplification of PI3K downstream signaling loops. Therefore, identification of critical PI3K mediators is paramount to the development of effective BLBC therapeutics. Using transcriptomic analysis of activated PIK3CA-expressing BLBC cells, we identified the gene encoding the humoral pattern recognition molecule pentraxin-3 (PTX3) as a critical target of oncogenic PI3K signaling. We found that PTX3 abundance is stimulated, in part, through AKT- and nuclear factor κB (NF-κB)-dependent pathways and that presence of PTX3 is necessary for PI3K-induced stem cell-like traits. We further showed that PTX3 expression is greater in tumor samples from patients with BLBC and that it is prognostic of poor patient survival. Our results thus reveal PTX3 as a newly identified PI3K-regulated biomarker and a potential therapeutic target in BLBC.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , C-Reactive Protein/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Neoplastic Stem Cells/metabolism , Serum Amyloid P-Component/metabolism , Signal Transduction , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/therapy , C-Reactive Protein/genetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Female , Humans , Neoplastic Stem Cells/pathology , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quantitative Trait Loci , Serum Amyloid P-Component/genetics
18.
Breast Dis ; 26: 75-85, 2006.
Article in English | MEDLINE | ID: mdl-17473367

ABSTRACT

Chemokines have been initially characterized as chemoattractants for leukocytes infiltrating into inflamed tissues. However, over the past few years, accumulating evidence has described the critical involvement of this superfamily of intercellular signaling proteins in a variety of biological processes, which include embryogenesis, organogenesis, and tissue homeostasis. Moreover, recent work has demonstrated novel roles for chemokines and their receptors in regulating various aspects of the transformed phenotype, such as tumor growth, angiogenesis, invasion, and metastasis. Here, we review the current knowledge regarding chemokine/chemokine receptor involvement in breast cancer pathogenesis with primary emphases on their role in the metastatic spread of cancer cells and in tumor-stroma interactions.


Subject(s)
Breast Neoplasms/pathology , Chemokines/metabolism , Signal Transduction , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cell Movement , Chemokines/antagonists & inhibitors , Humans , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Neovascularization, Pathologic/physiopathology , Stromal Cells/metabolism , Stromal Cells/pathology
19.
Mol Cell Oncol ; 3(3): e1019022, 2016 May.
Article in English | MEDLINE | ID: mdl-27314063

ABSTRACT

In a recent article in Cell Stem Cell, we showed that mesenchymal stem cells (MSCs), progenitor cells that populate the breast tumor stroma, induce microRNA-mediated FOXP2 repression in breast cancer cells (BCCs), thus promoting cancer stem cell (CSC) and metastatic traits. Here, we discuss the implications of these findings for understanding metastatic CSC genesis.

20.
Oncogene ; 23(58): 9369-80, 2004 Dec 16.
Article in English | MEDLINE | ID: mdl-15516977

ABSTRACT

A novel splice variant of Rac1, designated Rac1b, is expressed in human breast and colon carcinoma cells. Rac1b contains an additional 19 amino-acid insert immediately behind the switch II domain, a region important for Rac1 interaction with regulators and effectors. Recent studies showed that Rac1b exhibited the biochemical properties of a constitutively activated GTPase, yet it showed impaired interaction with downstream effectors, suggesting that Rac1b may be defective in biological activity. Whether Rac1b is a biologically active protein was not addressed. Therefore, we evaluated the biochemical, signaling and growth-promoting properties of authentic Rac1b. Similar to previous observations, we found that Rac1b showed enhanced intrinsic guanine nucleotide exchange activity, impaired intrinsic GTPase activity, and failed to interact with RhoGDI. Surprisingly, we found that Rac1b, like the constitutively-activated and transforming Rac1(Q61L) mutant, promoted growth transformation of NIH3T3 cells. Rac1b-expressing cells also showed a loss of density-dependent and anchorage-dependent growth. Surprisingly, unlike activated Rac1(61L), Rac1b did not show enhanced activation of the nuclear factor kappaB (NF-kappaB) transcription factor or stimulate cyclin D1 expression, the signaling activities that best correlate with Rac1 transforming activity. However, Rac1b did promote activation of the AKT serine/threonine kinase. Therefore, we suggest that Rac1b selectively activates a subset of Rac1 downstream signaling pathways to facilitate cellular transformation.


Subject(s)
Cell Transformation, Neoplastic , Neuropeptides/physiology , RNA Splicing/physiology , rac GTP-Binding Proteins/physiology , Animals , Base Sequence , Cell Division/physiology , DNA Primers , Guanine Nucleotide Exchange Factors , Mice , Microscopy, Confocal , Microscopy, Fluorescence , NIH 3T3 Cells , Proteins/physiology , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , rac1 GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL