Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 20(9): 1550-1560, 2021 09.
Article in English | MEDLINE | ID: mdl-34210825

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer characterized by early dissemination and poor drug response. Therefore, it is an unmet medical need to develop new strategies for treatment. As aberrant activation of ERK due to KRAS activating mutation is a driving force for PDAC, a brake system that can terminate ERK signaling represents an ideal druggable target. Herein, we demonstrate that forced expression of dual specificity phosphatase-2 (DUSP2), a specific ERK phosphatase, abrogated tumor formation and loss of Dusp2 facilitated Kras-driven PDAC progression. We report that a selective HDAC1/2 inhibitor (B390) has multifaceted therapeutic potential in PDAC by restoring the expression and function of DUSP2. In vitro study showed that treatment with B390 inhibited growth and migration abilities of PDAC cells, decreased extracellular vesicle-associated VEGF-C expression, and suppressed lymphatic endothelial cell proliferation. In vivo, B390 not only suppressed tumor growth by increasing tumor cell death, it also inhibited lymphangiogenesis and lymphovascular invasion. Taken together, our data demonstrate that B390 was able to alleviate loss of DUSP2-mediated pathologic processes, which provides the proof-of-concept evidence to demonstrate the potential of using selective HDAC1/2 inhibitors in PDAC treatment and suggests reinstating DUSP2 expression may be a strategy to subside PDAC progression.


Subject(s)
Dual Specificity Phosphatase 2/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Lymphangiogenesis , Pancreatic Neoplasms/drug therapy , Vascular Endothelial Growth Factor C/metabolism , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Dual Specificity Phosphatase 2/genetics , Extracellular Vesicles/metabolism , Humans , Mice , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor C/genetics
2.
Exp Biol Med (Maywood) ; 246(19): 2048-2056, 2021 10.
Article in English | MEDLINE | ID: mdl-34139872

ABSTRACT

Metastasis is not only one of the hallmarks of cancer but, unfortunately, it also is the most accurate biomarker for poor prognosis. Cancer cells metastasize through two different but eventually merged routes, the vasculature and lymphatic systems. The processes of cancer metastasis through blood vessel have been extensively studied and are well documented in the literature. In contrast, metastasis through the lymphatic system is less studied. Most people believe that cancer cells metastasize through lymphatic vessel are passive because the lymphatic system is thought to be a sewage draining system that collects whatever appears in the tissue fluid. It was recently found that cancer cells disseminated from lymphatic vessels are protected from being destroyed by our body's defense system. Furthermore, some cancer cells or cancer-associated immune cells secrete lymphangiogenic factors to recruit lymphatic vessel infiltration to the tumor region, a process known as lymphangiogenesis. To ensure the efficiency of lymphangiogenesis, the lymphangiogenic mediators are carried or packed by nanometer-sized particles named extracellular vesicles. Extracellular vesicles are lipid bilayer particles released from eventually every single cell, including bacterium, with diameters ranging from 30 nm (exosome) to several micrometers (apoptotic body). Components carried by extracellular vesicles include but are not limited to DNA, RNA, protein, fatty acid, and other metabolites. Recent studies suggest that cancer cells not only secrete more extracellular vesicles but also upload critical mediators required for lymphatic metastasis onto extracellular vesicles. This review will summarize recent advances in cancer lymphatic metastasis and how cancer cells regulate this process via extracellular vesicle-dependent lymphangiogenesis.


Subject(s)
Extracellular Vesicles/pathology , Lymphangiogenesis/physiology , Lymphatic Metastasis/pathology , Neoplasms/pathology , Animals , Humans
3.
Int J Mol Sci ; 22(9)2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33922657

ABSTRACT

BRCA mutation, one of the most common types of mutations in breast and ovarian cancer, has been suggested to be synthetically lethal with depletion of RAD52. Pharmacologically inhibiting RAD52 specifically eradicates BRCA-deficient cancer cells. In this study, we demonstrated that curcumin, a plant polyphenol, sensitizes BRCA2-deficient cells to CPT-11 by impairing RAD52 recombinase in MCF7 cells. More specifically, in MCF7-siBRCA2 cells, curcumin reduced homologous recombination, resulting in tumor growth suppression. Furthermore, a BRCA2-deficient cell line, Capan1, became resistant to CPT-11 when BRCA2 was reintroduced. In vivo, xenograft model studies showed that curcumin combined with CPT-11 reduced the growth of BRCA2-knockout MCF7 tumors but not MCF7 tumors. In conclusion, our data indicate that curcumin, which has RAD52 inhibitor activity, is a promising candidate for sensitizing BRCA2-deficient cells to DNA damage-based cancer therapies.


Subject(s)
BRCA2 Protein/deficiency , Breast Neoplasms/drug therapy , Curcumin/pharmacology , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , Homologous Recombination , Rad52 DNA Repair and Recombination Protein/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis , BRCA2 Protein/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , DNA Repair , Female , Humans , Irinotecan/pharmacology , Mice , Mice, Nude , Mutation , Topoisomerase I Inhibitors/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 117(41): 25859-25868, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33004630

ABSTRACT

Endometriosis is a highly prevalent gynecological disease with severe negative impacts on life quality and financial burden. Unfortunately, there is no cure for this disease, which highlights the need for further investigation about the pathophysiology of this disease to provide clues for developing novel therapeutic regimens. Herein, we identified that vascular endothelial growth factor (VEGF)-C, a potent lymphangiogenic factor, is up-regulated in endometriotic cells and contributes to increased lymphangiogenesis. Bioinformatic analysis and molecular biological characterization revealed that VEGF-C is negatively regulated by an orphan nuclear receptor, chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII). Further studies demonstrated that proinflammatory cytokines, via suppression of COUP-TFII level, induce VEGF-C overexpression. More importantly, we show that functional VEGF-C is transported by extracellular vesicles (EVs) to enhance the lymphangiogenic ability of lymphatic endothelial cells. Autotransplanted mouse model of endometriosis showed lenvatinib treatment abrogated the increased lymphatic vessels development in the endometriotic lesion, enlarged retroperitoneal lymph nodes, and immune cells infiltration, indicating that blocking VEGF-C signaling can reduce local chronic inflammation and concomitantly endometriosis development. Evaluation of EV-transmitted VEGF-C from patients' sera demonstrates it is a reliable noninvasive way for clinical diagnosis. Taken together, we identify the vicious cycle of inflammation, COUP-TFII, VEGF-C, and lymphangiogenesis in the endometriotic microenvironment, which opens up new horizons in understanding the pathophysiology of endometriosis. VEGF-C not only can serve as a diagnostic biomarker but also a molecular target for developing therapeutic regimens.


Subject(s)
Endometriosis/immunology , Extracellular Vesicles/immunology , Immune System/immunology , Lymphangiogenesis , Vascular Endothelial Growth Factor C/immunology , Animals , COUP Transcription Factor II/genetics , COUP Transcription Factor II/immunology , Cytokines/genetics , Cytokines/immunology , Endometriosis/genetics , Endometriosis/physiopathology , Endothelial Cells/immunology , Extracellular Vesicles/genetics , Female , Humans , Lymphatic Vessels/immunology , Mice , Vascular Endothelial Growth Factor C/genetics
5.
J Extracell Vesicles ; 9(1): 1746529, 2020.
Article in English | MEDLINE | ID: mdl-32341770

ABSTRACT

Early dissemination is a unique characteristic and a detrimental process of pancreatic ductal adenocarcinoma (PDAC); however, the underlying mechanism remains largely unknown. Here, we investigate the role of dual-specificity phosphatase-2 (DUSP2)-vascular endothelial growth factor-C (VEGF-C) axis in mediating PDAC lymphangiogenesis and lymphovascular invasion. Expression of DUSP2 is greatly suppressed in PDAC, which results in increased aberrant expression of extracellular vesicle (EV)-associated VEGF-C secretion. EV-VEGF-C exerts paracrine effects on lymphatic endothelial cells and autocrine effects on cancer cells, resulting in the lymphovascular invasion of cancer cells. Tissue-specific knockout of Dusp2 in mouse pancreas recapitulates PDAC phenotype and lymphovascular invasion. Mechanistically, loss-of-DUSP2 enhances proprotein convertase activity and vesicle trafficking to promote the release of the mature form of EV-VEGF-C. Collectively, these findings represent a conceptual advance in understanding pancreatic cancer lymphovascular invasion and suggest that loss-of-DUSP2-mediated VEGF-C processing may play important roles in early dissemination of pancreatic cancer. Abbreviations: DUSP2: dual-specificity phosphatase-2; VEGF-C: vascular endothelial growth factor-C; EV: extracellular vesicles; PDAC: pancreatic ductal adenocarcinoma; KD: knockdown.

6.
Sensors (Basel) ; 18(11)2018 Nov 13.
Article in English | MEDLINE | ID: mdl-30428635

ABSTRACT

Using the drone base station (DBS) to alleviate the network coverage supply-demand mismatch is an attractive issue. Found in DBS-assisted cellular mobile networks, the deployment of DBSs to cope with the dynamic load requirements is an important problem. The authors propose a proactive DBS deployment method to enhance the DBS deployment flexibility based on network traffic. The proposed scheme uses potential value and minimum distance to decide the areas that most need to be covered, which are named as proactive coverage areas (PCAs), whereby the DBSs are assigned to cover those PCAs. Meanwhile, when the number of required DBSs is determined, the energy consumption is related to the coverage radius and the altitude of DBSs. Therefore, the proposed method further investigates the on-demand coverage radius and then obtains the altitude of DBSs. Simulations show that the proposed proactive DBS deployment method provides better coverage performance with a significant complexity reduction.

7.
Nat Commun ; 6: 10077, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26687066

ABSTRACT

TP53 is mutated in 50% of all cancers, and its function is often compromised in cancers where it is not mutated. Here we demonstrate that the pro-tumorigenic/metastatic Six1 homeoprotein decreases p53 levels through a mechanism that does not involve the negative regulator of p53, MDM2. Instead, Six1 regulates p53 via a dual mechanism involving upregulation of microRNA-27a and downregulation of ribosomal protein L26 (RPL26). Mutation analysis confirms that RPL26 inhibits miR-27a binding and prevents microRNA-mediated downregulation of p53. The clinical relevance of this interaction is underscored by the finding that Six1 expression strongly correlates with decreased RPL26 across numerous tumour types. Importantly, we find that Six1 expression leads to marked resistance to therapies targeting the p53-MDM2 interaction. Thus, we identify a competitive mechanism of p53 regulation, which may have consequences for drugs aimed at reinstating p53 function in tumours.


Subject(s)
Down-Regulation , Homeodomain Proteins/metabolism , MicroRNAs/genetics , Neoplasms/genetics , Ribosomal Proteins/genetics , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , MicroRNAs/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomal Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Exp Biol Med (Maywood) ; 240(6): 718-24, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25888649

ABSTRACT

It has been shown in many clinical studies that the level of vascular endothelial growth factor-C (VEGF-C) positively correlates with lymph node metastasis. Nevertheless, beyond the canonical role of VEGF-C in stimulating lymphangiogenesis and thus promoting lymph node/distant metastasis, emerging evidence indicates that expression of VEGF-C contributes to various aspects of carcinogenicity via autocrine regulation. The newly identified functions of VEGF-C include but are not limited to proliferation, migration, invasion, and chemo-resistance. Besides tumor cell autocrine regulation, VEGF-C can also modulate the immune system such that tumor cells more easily escape immune surveillance. Therefore, understanding the functional roles and regulatory mechanisms related to the VEGF-C axis may lead to alternative strategies for cancer treatment. This mini-review will focus on summarizing recent discoveries regarding the unconventional functions of VEGF-C in cancer progression.


Subject(s)
Autocrine Communication , Cell Proliferation , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor C/metabolism , Animals , Disease Progression , Humans , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , Vascular Endothelial Growth Factor C/genetics
9.
Cancer Res ; 75(9): 1908-21, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25716682

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a dynamic process that relies on cellular plasticity. Recently, the process of an oncogenic EMT, followed by a reverse mesenchymal-to-epithelial transition (MET), has been implicated as critical in the metastatic colonization of carcinomas. Unlike governance of epithelial programming, regulation of mesenchymal programming is not well understood in EMT. Here, we describe and characterize the first microRNA that enhances exclusively mesenchymal programming. We demonstrate that miR-424 is upregulated early during a TWIST1 or SNAI1-induced EMT, and that it causes cells to express mesenchymal genes without affecting epithelial genes, resulting in a mixed/intermediate EMT. Furthermore, miR-424 increases motility, decreases adhesion, and induces a growth arrest, changes associated with a complete EMT that can be reversed when miR-424 expression is lowered, concomitant with an MET-like process. Breast cancer patient miR-424 levels positively associate with TWIST1/2 and EMT-like gene signatures, and miR-424 is increased in primary tumors versus matched normal breast. However, miR-424 is downregulated in patient metastases versus matched primary tumors. Correspondingly, miR-424 decreases tumor initiation and is posttranscriptionally downregulated in macrometastases in mice, suggesting the need for biphasic expression of miR-424 to transit the EMT-MET axis. Next-generation RNA sequencing revealed miR-424 regulates numerous EMT and cancer stemness-associated genes, including TGFBR3, whose downregulation promotes mesenchymal phenotypes, but not tumor-initiating phenotypes. Instead, we demonstrate that increased MAPK-ERK signaling is critical for miR-424-mediated decreases in tumor-initiating phenotypes. These findings suggest miR-424 plays distinct roles in tumor progression, potentially facilitating earlier, but repressing later, stages of metastasis by regulating an EMT-MET axis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Female , Humans , Mice , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinase Kinases/genetics , Neoplasm Metastasis , Nuclear Proteins/metabolism , RNA Processing, Post-Transcriptional , Signal Transduction , Twist-Related Protein 1/metabolism , Up-Regulation
10.
Breast Cancer Res ; 16(5): 462, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25358638

ABSTRACT

INTRODUCTION: Triple-negative breast cancers, particularly the claudin-low subtype, are highly aggressive and exhibit increased tumor-initiating cell (TIC) characteristics. In this study, we demonstrate that vascular endothelial growth factor C (VEGF-C) is highly expressed in the claudin-low breast cancer subtype and also that it mediates tumor progression, not only through its role in lymphangiogenesis but also through regulating TIC characteristics and the response to reactive oxygen species (ROS). METHODS: VEGF C expression was examined in breast cancer subtypes, and a VEGF C expression signature was derived. VEGF C expression and/or its associated signature was correlated with TIC and chemoresistance signatures. In vitro and in vivo assays were performed to determine whether VEGF-C expression alters TIC characteristics and the response of breast cancer cells to chemotherapy and oxidative stress. Array analysis was used to identify a downstream effector of VEGF-C, superoxide dismutase 3 (Sod3), which was tested for its involvement in VEGF-C-mediated resistance to oxidative stress and enhancement of in vivo metastasis. The VEGF-C-associated receptor neuropilin 2 (Nrp2) was knocked down to determine whether it is required for the observed effects of VEGF-C. Expression of VEGF C and Sod3 was assessed in human breast cancers. RESULTS: VEGF C is highly expressed in claudin-low breast cancers, and VEGF C and the VEGF C signature are associated with TIC-related gene signatures. VEGF-C-knockdown in mammary carcinoma cells decreases TIC properties in vitro and in vivo, sensitizing cells to oxidative stress and chemotherapy. We identified Sod3 as a target of VEGF-C in breast cancer cells by demonstrating that it is required for VEGF-C-mediated cell survival in response to oxidative stress and for VEGF-C-mediated metastasis. We demonstrate that Nrp2 is the VEGF-C-associated receptor that mediates alterations in Sod3 expression and the response of tumor cells to oxidative stress. We show that VEGF C and Sod3 are positively associated in human breast cancer. CONCLUSIONS: We describe a novel mechanism by which VEGF-C contributes to metastasis via its ability to enhance TIC-associated characteristics, particularly the response to ROS. We identified Sod3 as a critical mediator of VEGF-C-induced metastasis, and we provide evidence that the VEGF-C-Sod3 axis plays a role in human breast cancers.


Subject(s)
Breast Neoplasms/enzymology , Lung Neoplasms/enzymology , Superoxide Dismutase/metabolism , Vascular Endothelial Growth Factor C/physiology , Aldehyde Dehydrogenase/metabolism , Animals , Antioxidants/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Claudins/metabolism , Disease Progression , Disease-Free Survival , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
11.
Cancer Res ; 74(24): 7357-70, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25348955

ABSTRACT

Misexpression of developmental transcription factors occurs often in human cancers, where embryonic programs may be reinstated in a context that promotes or sustains malignant development. In this study, we report the involvement of the kidney development transcription factor Six2 in the metastatic progression of human breast cancer. We found that Six2 promoted breast cancer metastasis by a novel mechanism involving both transcriptional and epigenetic regulation of E-cadherin. Downregulation of E-cadherin by Six2 was necessary for its ability to increase soft agar growth and in vivo metastasis in an immunocompetent mouse model of breast cancer. Mechanistic investigations showed that Six2 represses E-cadherin expression by upregulating Zeb2, in part, through a microRNA-mediated mechanism and by stimulating promoter methylation of the E-cadherin gene (Cdh1). Clinically, SIX2 expression correlated inversely with CDH1 expression in human breast cancer specimens, corroborating the disease relevance of their interaction. Our findings establish Six2 as a regulator of metastasis in human breast cancers and demonstrate an epigenetic function for SIX family transcription factors in metastatic progression through the regulation of E-cadherin.


Subject(s)
Breast Neoplasms/genetics , Cadherins/biosynthesis , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Antigens, CD , Breast Neoplasms/pathology , Cadherins/genetics , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Repressor Proteins/metabolism , Transcription Factors/genetics , Zinc Finger E-box Binding Homeobox 2
12.
Breast Cancer Res ; 14(4): R100, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22765220

ABSTRACT

INTRODUCTION: Mammary-specific overexpression of Six1 in mice induces tumors that resemble human breast cancer, some having undergone epithelial to mesenchymal transition (EMT) and exhibiting stem/progenitor cell features. Six1 overexpression in human breast cancer cells promotes EMT and metastatic dissemination. We hypothesized that Six1 plays a role in the tumor initiating cell (TIC) population specifically in certain subtypes of breast cancer, and that by understanding its mechanism of action, we could potentially develop new means to target TICs. METHODS: We examined gene expression datasets to determine the breast cancer subtypes with Six1 overexpression, and then examined its expression in the CD24low/CD44+ putative TIC population in human luminal breast cancers xenografted through mice and in luminal breast cancer cell lines. Six1 overexpression, or knockdown, was performed in different systems to examine how Six1 levels affect TIC characteristics, using gene expression and flow cytometric analysis, tumorsphere assays, and in vivo TIC assays in immunocompromised and immune-competent mice. We examined the molecular pathways by which Six1 influences TICs using genetic/inhibitor approaches in vitro and in vivo. Finally, we examined the expression of Six1 and phosphorylated extracellular signal-regulated kinase (p-ERK) in human breast cancers. RESULTS: High levels of Six1 are associated with adverse outcomes in luminal breast cancers, particularly the luminal B subtype. Six1 levels are enriched in the CD24low/CD44+ TIC population in human luminal breast cancers xenografted through mice, and in tumorsphere cultures in MCF7 and T47D luminal breast cancer cells. When overexpressed in MCF7 cells, Six1expands the TIC population through activation of transforming growth factor-beta (TGF-ß) and mitogen activated protein kinase (MEK)/ERK signaling. Inhibition of ERK signaling in MCF7-Six1 cells with MEK1/2 inhibitors, U0126 and AZD6244, restores the TIC population of luminal breast cancer cells back to that observed in control cells. Administration of AZD6244 dramatically inhibits tumor formation efficiency and metastasis in cells that express high levels of Six1 ectopically or endogenously. Finally, we demonstrate that Six1 significantly correlates with phosphorylated ERK in human breast cancers. CONCLUSIONS: Six1 plays an important role in the TIC population in luminal breast cancers and induces a TIC phenotype by enhancing both TGF-ß and ERK signaling. MEK1/2 kinase inhibitors are potential candidates for targeting TICs in breast tumors.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Homeodomain Proteins/genetics , Neoplastic Stem Cells/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cluster Analysis , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Mice , Prognosis
13.
J Clin Invest ; 122(5): 1895-906, 2012 May.
Article in English | MEDLINE | ID: mdl-22466647

ABSTRACT

An association between lymph node metastasis and poor prognosis in breast cancer was observed decades ago. However, the mechanisms by which tumor cells infiltrate the lymphatic system are not completely understood. Recently, it has been proposed that the lymphatic system has an active role in metastatic dissemination and that tumor-secreted growth factors stimulate lymphangiogenesis. We therefore investigated whether SIX1, a homeodomain-containing transcription factor previously associated in breast cancer with lymph node positivity, was involved in lymphangiogenesis and lymphatic metastasis. In a model in which human breast cancer cells were injected into immune-compromised mice, we found that SIX1 expression promoted peritumoral and intratumoral lymphangiogenesis, lymphatic invasion, and distant metastasis of breast cancer cells. SIX1 induced transcription of the prolymphangiogenic factor VEGF-C, and this was required for lymphangiogenesis and lymphatic metastasis. Using a mouse mammary carcinoma model, we found that VEGF-C was not sufficient to mediate all the metastatic effects of SIX1, indicating that SIX1 acts through additional, VEGF-C-independent pathways. Finally, we verified the clinical significance of this prometastatic SIX1/VEGF-C axis by demonstrating coexpression of SIX1 and VEGF-C in human breast cancer. These data define a critical role for SIX1 in lymphatic dissemination of breast cancer cells, providing a direct mechanistic explanation for how VEGF-C expression is upregulated in breast cancer, resulting in lymphangiogenesis and metastasis.


Subject(s)
Gene Expression Regulation, Neoplastic , Homeodomain Proteins/physiology , Lung Neoplasms/secondary , Lymphangiogenesis , Mammary Neoplasms, Experimental/pathology , Vascular Endothelial Growth Factor C/genetics , Animals , Cell Line, Tumor , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lymphatic Metastasis , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Transcription, Genetic , Tumor Burden , Up-Regulation , Vascular Endothelial Growth Factor C/metabolism
14.
Cancer Res ; 70(24): 10371-80, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21056993

ABSTRACT

The Six1 homeodomain protein is a developmental transcription factor that has been implicated in tumor onset and progression. Our recent work shows that Six1 overexpression in human breast cancer cell lines is sufficient to induce epithelial-to-mesenchymal transition (EMT) and metastasis. Importantly, Six1-induced EMT and metastasis are dependent on TGF-ß signaling. The TGF-ß pathway plays a dual role in cancer, acting as a tumor suppressor in early lesions but enhancing metastatic spread in more advanced tumors. Our previous work indicated that Six1 may be a critical mediator of the switch in TGF-ß signaling from tumor suppressive to tumor promotional. However, the mechanism by which Six1 impinges on the TGF-ß pathway was, until now, unclear. In this work, we identify the TGF-ß type I receptor (TßRI) as a target of Six1 and a critical effector of Six1-induced TGF-ß signaling and EMT. We show that Six1-induced upregulation of TßRI is both necessary and sufficient to activate TGF-ß signaling and induce properties of EMT. Interestingly, increased TßRI expression is not sufficient to induce experimental metastasis, providing in vivo evidence that Six1 overexpression is required to switch TGF-ß signaling to the prometastatic phenotype and showing that induction of EMT is not sufficient to induce experimental metastasis. Together, these results show a novel mechanism for the activation of TGF-ß signaling, identify TßRI as a new target of Six1, and implicate Six1 as a determinant of TGF-ß function in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Homeodomain Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Metastasis , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Transcription, Genetic , Up-Regulation
15.
J Clin Endocrinol Metab ; 90(1): 286-95, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15483103

ABSTRACT

Aberrant production of cyclooxygenase-2 (COX-2) plays pivotal roles in many pathological processes including tumorigenesis and endometriosis, although the underlying mechanism remains obscure. Herein we report evidence to demonstrate that COX-2 is distinctly regulated by IL-1beta in normal and endometriotic stroma. Ectopic endometriotic stromal cell is at least 100 times more sensitive to IL-1beta treatment, compared with its eutopic counterpart. Induction of COX-2 expression in normal endometrial stroma by IL-1beta is primary due to enhancement of COX-2 mRNA stability. In contrast, IL-1beta not only increases COX-2 mRNA stability but also up-regulates COX-2 promoter activity in ectopic endometriotic stroma. Induction of COX-2 promoter activity by IL-1beta is mediated via MAPK-dependent phosphorylation of cAMP-responding element binding protein. Promoter activity and EMSAs demonstrate that a cAMP response element site located at -571/-564 of COX-2 promoter is critical for IL-1beta-induced COX-2 gene expression. Our results indicate that elevation of COX-2 expression in endometriotic tissues may result from increased sensitivity to proinflammatory cytokines such as IL-1beta, which is consistently present in the peritoneal fluid of endometriosis patients. Distinct regulation of COX-2 gene by IL-1beta may play a critical role in pathophysiological processes such as cancer formation and endometriosis.


Subject(s)
Endometriosis/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Interleukin-1/pharmacology , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Stromal Cells/enzymology , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclooxygenase 2 , Female , Humans , Membrane Proteins , Phosphorylation , Promoter Regions, Genetic , Receptors, Interleukin-1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...