Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Cancer Res Commun ; 2(12): 1545-1557, 2022 12.
Article in English | MEDLINE | ID: mdl-36561929

ABSTRACT

We report the inverse association between the expression of androgen receptor (AR) and interleukin-1beta (IL-1ß) in a cohort of patients with metastatic castration resistant prostate cancer (mCRPC). We also discovered that AR represses the IL-1ß gene by binding an androgen response element (ARE) half-site located within the promoter, which explains the IL-1ß expression in AR-negative (ARNEG) cancer cells. Consistently, androgen-depletion or AR-pathway inhibitors (ARIs) de-repressed IL-1ß in ARPOS cancer cells, both in vitro and in vivo. The AR transcriptional repression is sustained by histone de-acetylation at the H3K27 mark in the IL-1ß promoter. Notably, patients' data suggest that DNA methylation prevents IL-1ß expression, even if the AR-signaling axis is inactive. Our previous studies show that secreted IL-1ß supports metastatic progression in mice by altering the transcriptome of tumor-associated bone stroma. Thus, in prostate cancer patients harboring ARNEG tumor cells or treated with ADT/ARIs, and with the IL-1ß gene unmethylated, IL-1ß could condition the metastatic microenvironment to sustain disease progression.


Subject(s)
Bone Neoplasms , Prostatic Neoplasms , Humans , Male , Animals , Mice , Receptors, Androgen/genetics , Interleukin-1beta/genetics , Androgens , Prostatic Neoplasms/genetics , Signal Transduction/genetics , Bone Neoplasms/genetics , Tumor Microenvironment
2.
J Cancer ; 13(11): 3160-3176, 2022.
Article in English | MEDLINE | ID: mdl-36118530

ABSTRACT

Tumor-initiating cells (TICs) are a rare sub-population of cells within the bulk of a tumor that are major contributors to tumor initiation, metastasis, and chemoresistance. TICs have a stem-cell-like phenotype that is dictated by the expression of master regulator transcription factors, including OCT4, NANOG, and SOX2. These transcription factors are expressed via activation of multiple signaling pathways that drive cancer initiation and progression. Importantly, these same signaling pathways can be activated by select chemokine receptors. Chemokine receptors are increasingly being revealed as major drivers of the TIC phenotype, as their signaling can lead to activation of stemness-controlling transcription factors. Additionally, the cell surface expression of chemokine receptors provides a unique therapeutic target to disrupt signaling pathways that control the expression of master regulator transcription factors and the TIC phenotype. This review summarizes the master regulator transcription factors known to dictate the TIC phenotype, along with the complex signaling pathways that can mediate their expression and the chemokine receptors that are most upstream of this phenotype.

3.
Oncogene ; 41(9): 1337-1351, 2022 02.
Article in English | MEDLINE | ID: mdl-34999735

ABSTRACT

Metastasis-initiating cells (MICs) display stem cell-like features, cause metastatic recurrences and defy chemotherapy, which leads to patients' demise. Here we show that prostate and breast cancer patients harbor contingents of tumor cells with high expression of CX3CR1, OCT4a (POU5F1), and NANOG. Impairing CX3CR1 expression or signaling hampered the formation of tumor spheroids by cell lines from which we isolated small subsets co-expressing CX3CR1 and stemness-related markers, similarly to patients' tumors. These rare CX3CR1High cells show transcriptomic profiles enriched in pathways that regulate pluripotency and endowed with metastasis-initiating behavior in murine models. Cancer cells lacking these features (CX3CR1Low) were capable of re-acquiring CX3CR1-associated features over time, implying that MICs can continuously emerge from non-stem cancer cells. CX3CR1 expression also conferred resistance to docetaxel, and prolonged treatment with docetaxel selected CX3CR1High phenotypes with de-enriched transcriptomic profiles for apoptotic pathways. These findings nominate CX3CR1 as a novel marker of stem-like tumor cells and provide conceptual ground for future development of approaches targeting CX3CR1 signaling and (re)expression as therapeutic means to prevent or contain metastasis initiation.


Subject(s)
Octamer Transcription Factor-3
4.
Pharmacol Ther ; 199: 117-128, 2019 07.
Article in English | MEDLINE | ID: mdl-30877019

ABSTRACT

Here we propose that therapeutic targeting of circulating tumor cells (CTCs), which are widely understood to be the seeds of metastasis, would represent an effective strategy towards limiting numerical expansion of secondary lesions and containing overall tumor burden in cancer patients. However, the molecular mediators of tumor seeding have not been well characterized. This is in part due to the limited number of pre-clinical in vivo approaches that appropriately interrogate the mechanisms by which cancer cells home to arresting organs. It is critical that we continue to investigate the mediators of tumor seeding as it is evident that the ability of CTCs to colonize in distant sites is what drives disease progression even after the primary tumor has been ablated by local modalities. In addition to slowing disease progression, containing metastatic spread by impeding tumor cell seeding may also provide a clinical benefit by increasing the duration of the residence of CTCs in systemic circulation thereby increasing their exposure to pharmacological agents commonly used in the treatment of patients such as chemotherapy and immunotherapies. In this review we will examine the current state of knowledge about the mechanisms of tumor cells seeding as well as explore how targeting this stage of metastatic spreading may provide therapeutic benefit to patients with advanced disease.


Subject(s)
Neoplasm Metastasis/prevention & control , Neoplasms/pathology , Animals , Humans , Neoplasms/therapy , Neoplastic Cells, Circulating
5.
Adv Exp Med Biol ; 1210: 171-184, 2019.
Article in English | MEDLINE | ID: mdl-31900910

ABSTRACT

The propensity of prostate cancer cells to seed the skeleton and then progress into clinically relevant metastatic tumors is widely recognized and a major cause of morbidity and mortality for patients. The natural history of prostate adenocarcinoma most frequently begins with a tumor diagnosed at a localized stage, which is successfully treated by surgical and/or radiation therapy modalities. A relevant percentage of patients are clinically cured but approximately 20-30% will develop biochemical signs of recurrence, which respond to the inhibition of androgen receptor (AR) signaling by hormone-deprivation and receptor antagonists, before the inevitable transition into castration-resistant prostate cancer (CRPC). This stage simultaneously presents with or is rapidly followed by secondary tumors, which involve the skeleton in more than 90% of cases (mCRPC). While generalization in clinical practice is always unwise, it is indisputable that bone-metastatic prostate cancer is virtually incurable. Decades of research have revealed that the tissue microenvironment provided by the bone marrow is as important as the cell-autonomous features of tumor cells in fostering the right conditions that lead to establishment and progression of metastatic tumors in the skeleton.


Subject(s)
Bone Neoplasms/secondary , Prostatic Neoplasms/pathology , Tumor Microenvironment , Bone Neoplasms/metabolism , Bone and Bones/metabolism , Bone and Bones/pathology , Humans , Male
6.
Mol Cancer Res ; 16(12): 1844-1854, 2018 12.
Article in English | MEDLINE | ID: mdl-30115759

ABSTRACT

Circulating tumor cells (CTCs) are commonly detected in the systemic blood of patients with cancer with metastatic tumors. However, the mechanisms controlling the viability of cancer cells in blood and length of time spent in circulation, as well as their potential for generating additional tumors are still undefined. Here, it is demonstrated that CX3CR1, a chemokine receptor, drives reseeding of breast CTCs to multiple organs. Antagonizing this receptor dramatically impairs the progression of breast cancer cells in a relevant model of human metastatic disease, by affecting both tumor growth and numerical expansion. Notably, therapeutic targeting of CX3CR1 prolongs CTC permanence in the blood, both promoting their spontaneous demise by apoptosis and counteracting metastatic reseeding. These effects lead to containment of metastatic progression and extended survival. Finally, targeting CX3CR1 improves blood exposure of CTCs to doxorubicin and in combination with docetaxel shows synergistic effects in containing overall tumor burden. IMPLICATIONS: The current findings shed light on CTCs reseeding dynamics and support the development of CX3CR1 antagonism as a viable strategy to counteract metastatic progression.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , CX3C Chemokine Receptor 1/metabolism , Neoplasm Metastasis/drug therapy , Neoplastic Cells, Circulating/metabolism , Small Molecule Libraries/administration & dosage , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , CX3C Chemokine Receptor 1/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Disease Progression , Docetaxel/administration & dosage , Docetaxel/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Female , Humans , Neoplasm Transplantation , Neoplastic Cells, Circulating/drug effects , Prognosis , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays
7.
Cell ; 174(5): 1200-1215.e20, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30100187

ABSTRACT

Nuclear pore complexes (NPCs) regulate nuclear-cytoplasmic transport, transcription, and genome integrity in eukaryotic cells. However, their functional roles in cancer remain poorly understood. We interrogated the evolutionary transcriptomic landscape of NPC components, nucleoporins (Nups), from primary to advanced metastatic human prostate cancer (PC). Focused loss-of-function genetic screen of top-upregulated Nups in aggressive PC models identified POM121 as a key contributor to PC aggressiveness. Mechanistically, POM121 promoted PC progression by enhancing importin-dependent nuclear transport of key oncogenic (E2F1, MYC) and PC-specific (AR-GATA2) transcription factors, uncovering a pharmacologically targetable axis that, when inhibited, decreased tumor growth, restored standard therapy efficacy, and improved survival in patient-derived pre-clinical models. Our studies molecularly establish a role of NPCs in PC progression and give a rationale for NPC-regulated nuclear import targeting as a therapeutic strategy for lethal PC. These findings may have implications for understanding how NPC deregulation contributes to the pathogenesis of other tumor types.


Subject(s)
E2F1 Transcription Factor/metabolism , Membrane Glycoproteins/metabolism , Nuclear Pore/physiology , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Carcinogenesis , Cell Nucleus/metabolism , Cell Proliferation , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Nuclear Envelope , Nuclear Pore Complex Proteins , Signal Transduction
8.
Cancer Res ; 78(7): 1657-1671, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29358169

ABSTRACT

The functional significance of the chemokine receptor CCR5 in human breast cancer epithelial cells is poorly understood. Here, we report that CCR5 expression in human breast cancer correlates with poor outcome. CCR5+ breast cancer epithelial cells formed mammospheres and initiated tumors with >60-fold greater efficiency in mice. Reintroduction of CCR5 expression into CCR5-negative breast cancer cells promoted tumor metastases and induced DNA repair gene expression and activity. CCR5 antagonists Maraviroc and Vicriviroc dramatically enhanced cell killing mediated by DNA-damaging chemotherapeutic agents. Single-cell analysis revealed CCR5 governs PI3K/Akt, ribosomal biogenesis, and cell survival signaling. As CCR5 augments DNA repair and is reexpressed selectively on cancerous, but not normal breast epithelial cells, CCR5 inhibitors may enhance the tumor-specific activities of DNA damage response-based treatments, allowing a dose reduction of standard chemotherapy and radiation.Significance: This study offers a preclinical rationale to reposition CCR5 inhibitors to improve the treatment of breast cancer, based on their ability to enhance the tumor-specific activities of DNA-damaging chemotherapies administered in that disease. Cancer Res; 78(7); 1657-71. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , CCR5 Receptor Antagonists/pharmacology , DNA Damage/genetics , DNA Repair/immunology , Neoplastic Stem Cells/metabolism , Receptors, CCR5/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Maraviroc/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Transplantation, Heterologous
9.
Cancer Res ; 76(22): 6723-6734, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27634768

ABSTRACT

Proteomic analysis of castration-resistant prostate cancer demonstrated the enrichment of Src tyrosine kinase activity in approximately 90% of patients. Src is known to induce cyclin D1, and a cyclin D1-regulated gene expression module predicts poor outcome in human prostate cancer. The tumor-associated calcium signal transducer 2 (TACSTD2/Trop2/M1S1) is enriched in the prostate, promoting prostate stem cell self-renewal upon proteolytic activation via a γ-secretase cleavage complex (PS1, PS2) and TACE (ADAM17), which releases the Trop2 intracellular domain (Trop2 ICD). Herein, v-Src transformation of primary murine prostate epithelial cells increased the proportion of prostate cancer stem cells as characterized by gene expression, epitope characteristics, and prostatosphere formation. Cyclin D1 was induced by v-Src, and Src kinase induction of Trop2 ICD nuclear accumulation required cyclin D1. Cyclin D1 induced abundance of the Trop2 proteolytic cleavage activation components (PS2, TACE) and restrained expression of the inhibitory component of the Trop2 proteolytic complex (Numb). Patients with prostate cancer with increased nuclear Trop2 ICD and cyclin D1, and reduced Numb, had reduced recurrence-free survival probability (HR = 4.35). Cyclin D1, therefore, serves as a transducer of v-Src-mediated induction of Trop2 ICD by enhancing abundance of the Trop2 proteolytic activation complex. Cancer Res; 76(22); 6723-34. ©2016 AACR.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cyclin D1/metabolism , src-Family Kinases/metabolism , Animals , Humans , Mice , Signal Transduction , Transfection
10.
Mol Cancer Res ; 14(6): 518-27, 2016 06.
Article in English | MEDLINE | ID: mdl-27001765

ABSTRACT

UNLABELLED: Recent evidence indicates that cancer cells, even in the absence of a primary tumor, recirculate from established secondary lesions to further seed and colonize skeleton and soft tissues, thus expanding metastatic dissemination and precipitating the clinical progression to terminal disease. Recently, we reported that breast cancer cells utilize the chemokine receptor CX3CR1 to exit the blood circulation and lodge to the skeleton of experimental animals. Now, we show that CX3CR1 is overexpressed in human breast tumors and skeletal metastases. To assess the clinical potential of targeting CX3CR1 in breast cancer, a functional role of CX3CR1 in metastatic seeding and progression was first validated using a neutralizing antibody for this receptor and transcriptional suppression by CRISPR interference (CRISPRi). Successively, we synthesized and characterized JMS-17-2, a potent and selective small-molecule antagonist of CX3CR1, which was used in preclinical animal models of seeding and established metastasis. Importantly, counteracting CX3CR1 activation impairs the lodging of circulating tumor cells to the skeleton and soft-tissue organs and also negatively affects further growth of established metastases. Furthermore, nine genes were identified that were similarly altered by JMS-17-2 and CRISPRi and could sustain CX3CR1 prometastatic activity. In conclusion, these data support the drug development of CX3CR1 antagonists, and promoting their clinical use will provide novel and effective tools to prevent or contain the progression of metastatic disease in breast cancer patients. IMPLICATIONS: This work conclusively validates the instrumental role of CX3CR1 in the seeding of circulating cancer cells and is expected to pave the way for pairing novel inhibitors of this receptor with current standards of care for the treatment of breast cancer patients. Mol Cancer Res; 14(6); 518-27. ©2016 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Receptors, Chemokine/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CX3C Chemokine Receptor 1 , Cell Line, Tumor , Female , Humans , Mice
11.
Transl Oncol ; 8(5): 358-367, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26500026

ABSTRACT

Inflammatory breast cancer (IBC) is an aggressive and invasive tumor, accounting for 2.5% of all breast cancer cases, and characterized by rapid progression, regional and distant metastases, younger age of onset, and lower overall survival. Presently, there are no effective therapies against IBC and a paucity of model systems. Our aim was to develop a clinically relevant IBC model that would allow investigations on the role of chemokine receptors in IBC metastasis. Primary cultures of tumor cells were isolated from pleural exudates of an IBC patient and grown as spheres or monolayers. We developed a human xenograft model where patient-derived IBC cells, stably transduced with lentiviral vectors expressing fluorescent and bioluminescent markers, were inoculated directly into the left ventricle of mice. Our in vivo data show that these IBC cells (FC-IBC02A) are able to seed and proliferate into various organs, including brain, lungs, lymph nodes, and bone, closely replicating the metastatic spread observed in IBC patients. Moreover, cells were able to generate tumors when grafted in the mammary fat pad of mice. RT-PCR and microscopy studies revealed expression of both CXCR4 and ACKR3 receptors in FC-IBC02A cells. Furthermore, CXCL12 (the endogenous chemokine ligand of these receptors) induced transendothelial migration of these cells and stimulated signaling pathways involved in cell survival and migration - an effect reduced by CXCR4 or ACKR3 antagonists. This new model can be used to develop chemokine-based pharmacological approaches against the IBC metastatic process. This work also provides the first evidence of ACKR3 expression in IBC cells.

12.
Am J Pathol ; 185(9): 2505-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26362718

ABSTRACT

Active Stat5a/b predicts early recurrence and disease-specific death in prostate cancer (PC), which both typically are caused by development of metastatic disease. Herein, we demonstrate that Stat5a/b induces epithelial-to-mesenchymal transition (EMT) of PC cells, as shown by Stat5a/b regulation of EMT marker expression (Twist1, E-cadherin, N-cadherin, vimentin, and fibronectin) in PC cell lines, xenograft tumors in vivo, and patient-derived PCs ex vivo using organ explant cultures. Jak2-Stat5a/b signaling induced functional end points of EMT as well, indicated by disruption of epithelial cell monolayers and increased migration and adhesion of PC cells to fibronectin. Knockdown of Twist1 suppressed Jak2-Stat5a/b-induced EMT properties of PC cells, which were rescued by re-introduction of Twist1, indicating that Twist1 mediates Stat5a/b-induced EMT in PC cells. While promoting EMT, Jak2-Stat5a/b signaling induced stem-like properties in PC cells, such as sphere formation and expression of cancer stem cell markers, including BMI1. Mechanistically, both Twist1 and BMI1 were critical for Stat5a/b induction of stem-like features, because genetic knockdown of Twist1 suppressed Stat5a/b-induced BMI1 expression and sphere formation in stem cell culture conditions, which were rescued by re-introduction of BMI1. By using human prolactin knock-in mice, we demonstrate that prolactin-Stat5a/b signaling promoted metastases formation of PC cells in vivo. In conclusion, our data support the concept that Jak2-Stat5a/b signaling promotes metastatic progression of PC by inducing EMT and stem cell properties in PC cells.


Subject(s)
Epithelial-Mesenchymal Transition , Janus Kinase 2/metabolism , Prostatic Neoplasms/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Cadherins/metabolism , Humans , Male , Mice , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Prostatic Neoplasms/pathology , Recurrence , Signal Transduction/physiology , Twist-Related Protein 1/metabolism
13.
Mol Cancer Res ; 13(9): 1336-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26023164

ABSTRACT

UNLABELLED: The bone is a preferred site for metastatic homing of prostate cancer cells. Once prostate cancer patients develop skeletal metastases, they eventually succumb to the disease; therefore, it is imperative to identify key molecular drivers of this process. This study examines the involvement of protein kinase C epsilon (PKCε), an oncogenic protein that is abnormally overexpressed in human tumor specimens and cell lines, on prostate cancer cell bone metastasis. PC3-ML cells, a highly invasive prostate cancer PC3 derivative with bone metastatic colonization properties, failed to induce skeletal metastatic foci upon inoculation into nude mice when PKCε expression was silenced using shRNA. Interestingly, while PKCε depletion had only marginal effects on the proliferative, adhesive, and migratory capacities of PC3-ML cells in vitro or in the growth of xenografts upon s.c. inoculation, it caused a significant reduction in cell invasiveness. Notably, PKCε was required for transendothelial cell migration (TEM) as well as for the growth of PC3-ML cells in a bone biomimetic environment. At a mechanistic level, PKCε depletion abrogates the expression of IL1ß, a cytokine implicated in skeletal metastasis. Taken together, PKCε is a key factor for driving the formation of bone metastasis by prostate cancer cells and is a potential therapeutic target for advanced stages of the disease. IMPLICATIONS: This study uncovers an important new function of PKCε in the dissemination of cancer cells to the bone; thus, highlighting the promising potential of this oncogenic kinase as a therapeutic target for skeletal metastasis.


Subject(s)
Bone Neoplasms/secondary , Mediator Complex , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase C-epsilon/metabolism , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Heterografts , Humans , Interleukin-1beta/metabolism , Male , Mice , Mice, Nude , RNA, Small Interfering/metabolism
14.
Cancer Res ; 74(23): 7103-14, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25452256

ABSTRACT

Src family kinases (SFK) integrate signal transduction for multiple receptors, regulating cellular proliferation, invasion, and metastasis in human cancer. Although Src is rarely mutated in human prostate cancer, SFK activity is increased in the majority of human prostate cancers. To determine the molecular mechanisms governing prostate cancer bone metastasis, FVB murine prostate epithelium was transduced with oncogenic v-Src. The prostate cancer cell lines metastasized in FVB mice to brain and bone. Gene expression profiling of the tumors identified activation of a CCR5 signaling module when the prostate epithelial cell lines were grown in vivo versus tissue cultures. The whole body, bone, and brain metastatic prostate cancer burden was reduced by oral CCR5 antagonist. Clinical trials of CCR5 inhibitors may warrant consideration in patients with CCR5 activation in their tumors.


Subject(s)
Bone Neoplasms/prevention & control , CCR5 Receptor Antagonists/pharmacology , Genes, src , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Receptors, CCR5/genetics , src-Family Kinases/genetics , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Brain Neoplasms/metabolism , Brain Neoplasms/prevention & control , Brain Neoplasms/secondary , Cell Line, Transformed , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , Gene Expression Profiling/methods , Humans , Male , Mice , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Receptors, CCR5/metabolism , src-Family Kinases/metabolism
15.
Neoplasia ; 16(12): 1047-58, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25499218

ABSTRACT

Most triple-negative breast cancers (TNBCs) exhibit gene expression patterns associated with epithelial-to-mesenchymal transition (EMT), a feature that correlates with a propensity for metastatic spread. Overexpression of the EMT regulator Slug is detected in basal and mesenchymal-type TNBCs and is associated with reduced E-cadherin expression and aggressive disease. The effects of Slug depend, in part, on the interaction of its N-terminal SNAG repressor domain with the chromatin-modifying protein lysine demethylase 1 (LSD1); thus, we investigated whether tranylcypromine [also known as trans-2-phenylcyclopropylamine hydrochloride (PCPA) or Parnate], an inhibitor of LSD1 that blocks its interaction with Slug, suppresses the migration, invasion, and metastatic spread of TNBC cell lines. We show here that PCPA treatment induces the expression of E-cadherin and other epithelial markers and markedly suppresses migration and invasion of TNBC cell lines MDA-MB-231 and BT-549. These effects were phenocopied by Slug or LSD1 silencing. In two models of orthotopic breast cancer, PCPA treatment reduced local tumor growth and the number of lung metastases. In mice injected directly in the blood circulation with MDA-MB-231 cells, PCPA treatment or Slug silencing markedly inhibited bone metastases but had no effect on lung infiltration. Thus, blocking Slug activity may suppress the metastatic spread of TNBC and, perhaps, specifically inhibit homing/colonization to the bone.


Subject(s)
Gene Silencing/physiology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tranylcypromine/pharmacology , Triple Negative Breast Neoplasms/pathology , Animals , Cadherins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Histone Demethylases/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Microscopy, Fluorescence , Monoamine Oxidase Inhibitors/pharmacology , Neoplasm Invasiveness , Neoplasm Metastasis/prevention & control , Plasmids , Real-Time Polymerase Chain Reaction , Snail Family Transcription Factors , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
16.
J Clin Invest ; 123(7): 2907-20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23921130

ABSTRACT

Metabolic reprogramming is an important driver of tumor progression; however, the metabolic regulators of tumor cell motility and metastasis are not understood. Here, we show that tumors maintain energy production under nutrient deprivation through the function of HSP90 chaperones compartmentalized in mitochondria. Using cancer cell lines, we found that mitochondrial HSP90 proteins, including tumor necrosis factor receptor-associated protein-1 (TRAP-1), dampen the activation of the nutrient-sensing AMPK and its substrate UNC-51-like kinase (ULK1), preserve cytoskeletal dynamics, and release the cell motility effector focal adhesion kinase (FAK) from inhibition by the autophagy initiator FIP200. In turn, this results in enhanced tumor cell invasion in low nutrients and metastatic dissemination to bone or liver in disease models in mice. Moreover, we found that phosphorylated ULK1 levels were correlated with shortened overall survival in patients with non-small cell lung cancer. These results demonstrate that mitochondrial HSP90 chaperones, including TRAP-1, overcome metabolic stress and promote tumor cell metastasis by limiting the activation of the nutrient sensor AMPK and preventing autophagy.


Subject(s)
Bone Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cytoskeleton/metabolism , Liver Neoplasms, Experimental/metabolism , Lung Neoplasms/metabolism , Stress, Physiological , Adenylate Kinase/metabolism , Animals , Antineoplastic Agents/pharmacology , Autophagy-Related Protein-1 Homolog , Bone Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Cell Movement , Female , Gene Knockdown Techniques , Guanidines/pharmacology , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Hexokinase/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kaplan-Meier Estimate , Lactams, Macrocyclic/pharmacology , Liver Neoplasms, Experimental/secondary , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mice , Mice, SCID , Mitochondria/metabolism , Mitochondrial Membranes/enzymology , NIH 3T3 Cells , Neoplasm Transplantation , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics
17.
Cancer Res ; 73(11): 3297-305, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23536554

ABSTRACT

Despite the progress made in the early detection and treatment of prostate adenocarcinoma, the metastatic lesions from this tumor are incurable. We used genome-wide expression analysis of human prostate cancer cells with different metastatic behavior in animal models to reveal that bone-tropic phenotypes upregulate three genes encoding for the cytokine interleukin-1ß (IL-1ß), the chemokine CXCL6 (GCP-2), and the protease inhibitor elafin (PI3). The Oncomine database revealed that these three genes are significantly upregulated in human prostate cancer versus normal tissue and correlate with Gleason scores ≥7. This correlation was further validated for IL-1ß by immunodetection in prostate tissue arrays. Our study also shows that the exogenous overexpression of IL-1ß in nonmetastatic cancer cells promotes their growth into large skeletal lesions in mice, whereas its knockdown significantly impairs the bone progression of highly metastatic cells. In addition, IL-1ß secreted by metastatic cells induced the overexpression of COX-2 (PTGS2) in human bone mesenchymal cells treated with conditioned media from bone metastatic prostate cancer cells. Finally, we inspected human tissue specimens from skeletal metastases and detected prostate cancer cells positive for both IL-1ß and synaptophysin while concurrently lacking prostate-specific antigen (PSA, KLK3) expression. Collectively, these findings indicate that IL-1ß supports the skeletal colonization and metastatic progression of prostate cancer cells with an acquired neuroendocrine phenotype.


Subject(s)
Bone Neoplasms/secondary , Carcinoma, Neuroendocrine/pathology , Interleukin-1beta/biosynthesis , Prostatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Immunocompromised Host , Interleukin-1beta/genetics , Male , Mice , NIH 3T3 Cells , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Synaptophysin/biosynthesis , Up-Regulation
18.
Cancer Res ; 73(10): 3155-67, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23536555

ABSTRACT

The molecular mechanisms underlying metastatic dissemination are still not completely understood. We have recently shown that ß(1) integrin-dependent cell adhesion to fibronectin and signaling is affected by a transmembrane molecule, Trop-2, which is frequently upregulated in human carcinomas. Here, we report that Trop-2 promotes metastatic dissemination of prostate cancer cells in vivo and is abundantly expressed in metastasis from human prostate cancer. We also show here that Trop-2 promotes prostate cancer cell migration on fibronectin, a phenomenon dependent on ß(1) integrins. Mechanistically, we demonstrate that Trop-2 and the α(5)ß(1) integrin associate through their extracellular domains, causing relocalization of α(5)ß(1) and the ß(1)-associated molecule talin from focal adhesions to the leading edges. Trop-2 effect is specific as this molecule does not modulate migration on vitronectin, does not associate with the major vitronectin receptor, α(v)ß(3) integrin, and does not affect localization of α(v)ß(3) integrin as well as vinculin in focal adhesions. We show that Trop-2 enhances directional prostate cancer cell migration, through modulation of Rac1 GTPase activity. Finally, we show that Trop-2 induces activation of PAK4, a kinase that has been reported to mediate cancer cell migration. In conclusion, we provide the first evidence that ß(1) integrin-dependent migratory and metastatic competence of prostate cancer cells is enhanced by Trop-2.


Subject(s)
Antigens, Neoplasm/physiology , Cell Adhesion Molecules/physiology , Integrin beta1/physiology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Humans , Male , Mice , Neoplasm Metastasis , Protein Multimerization , Receptors, Vitronectin/physiology
19.
J Clin Invest ; 123(1): 493-508, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23257359

ABSTRACT

Cyclin D1b is a splice variant of the cell cycle regulator cyclin D1 and is known to harbor divergent and highly oncogenic functions in human cancer. While cyclin D1b is induced during disease progression in many cancer types, the mechanisms underlying cyclin D1b function remain poorly understood. Herein, cell and human tumor xenograft models of prostate cancer were utilized to resolve the downstream pathways that are required for the protumorigenic functions of cyclin D1b. Specifically, cyclin D1b was found to modulate the expression of a large transcriptional network that cooperates with androgen receptor (AR) signaling to enhance tumor cell growth and invasive potential. Notably, cyclin D1b promoted AR-dependent activation of genes associated with metastatic phenotypes. Further exploration determined that transcriptional induction of SNAI2 (Slug) was essential for cyclin D1b-mediated proliferative and invasive properties, implicating Slug as a critical driver of disease progression. Importantly, cyclin D1b expression highly correlated with that of Slug in clinical samples of advanced disease. In vivo analyses provided strong evidence that Slug enhances both tumor growth and metastatic phenotypes. Collectively, these findings reveal the underpinning mechanisms behind the protumorigenic functions of cyclin D1b and demonstrate that the convergence of the cyclin D1b/AR and Slug pathways results in the activation of processes critical for the promotion of lethal tumor phenotypes.


Subject(s)
Cyclin D1/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Transcription Factors/metabolism , Alternative Splicing/genetics , Animals , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Transcriptional Activation/genetics , Transplantation, Heterologous
20.
Breast Cancer Res ; 13(5): R91, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21933397

ABSTRACT

INTRODUCTION: Skeletal metastases from breast adenocarcinoma are responsible for most of the morbidity and mortality associated with this tumor and represent a significant and unmet need for therapy. The arrival of circulating cancer cells to the skeleton depends first on the adhesive interactions with the endothelial cells lining the bone marrow sinusoids, and then the extravasation toward chemoattractant molecules produced by the surrounding bone stroma.We have previously shown that the membrane-bound and cell-adhesive form of the chemokine fractalkine is exposed on the luminal side of human bone marrow endothelial cells and that bone stromal cells release the soluble and chemoattractant form of this chemokine. The goal of this study was to determine the role of fractalkine and its specific receptor CX3CR1 in the homing of circulating breast cancer cells to the skeleton. METHODS: We employed a powerful pre-clinical animal model of hematogenous metastasis, in which fluorescent cancer cells are identified immediately after their arrival to the bone. We engineered cells to over-express either wild-type or functional mutants of CX3CR1 as well as employed transgenic mice knockout for fractalkine. RESULTS: CX3CR1 protein is detected in human tissue microarrays of normal and malignant mammary glands. We also found that breast cancer cells expressing high levels of this receptor have a higher propensity to spread to the skeleton. Furthermore, studies with fractalkine-null transgenic mice indicate that the ablation of the adhesive and chemotactic ligand of CX3CR1 dramatically impairs the skeletal dissemination of circulating cancer cells. Finally, we conclusively confirmed the crucial role of CX3CR1 on breast cancer cells for both adhesion to bone marrow endothelium and extravasation into the bone stroma. CONCLUSIONS: We provide compelling evidence that the functional interactions between fractalkine produced by both the endothelial and stromal cells of bone marrow and the CX3CR1 receptor on breast cancer cells are determinant in the arrest and initial lodging needed for skeletal dissemination.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Receptors, Chemokine/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CX3C Chemokine Receptor 1 , Cell Adhesion/genetics , Chemokine CX3CL1/genetics , Endothelium/cytology , Endothelium/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mutation , Receptors, Chemokine/genetics , Stromal Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...