Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Anal Chem ; 92(24): 15693-15698, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33232116

ABSTRACT

The extracellular matrix (ECM) provides an architectural meshwork that surrounds and supports cells. The dysregulation of heavily post-translationally modified ECM proteins directly contributes to various diseases. Mass spectrometry (MS)-based proteomics is an ideal tool to identify ECM proteins and characterize their post-translational modifications, but ECM proteomics remains challenging owing to the extremely low solubility of the ECM. Herein, enabled by effective solubilization of ECM proteins using our recently developed photocleavable surfactant, Azo, we have developed a streamlined ECM proteomic strategy that allows fast tissue decellularization, efficient extraction and enrichment of ECM proteins, and rapid digestion prior to reversed-phase liquid chromatography (RPLC)-MS analysis. A total of 173 and 225 unique ECM proteins from mouse mammary tumors have been identified using 1D and 2D RPLC-MS/MS, respectively. Moreover, 87 (from 1DLC-MS/MS) and 229 (from 2DLC-MS/MS) post-translational modifications of ECM proteins, including glycosylation, phosphorylation, and hydroxylation, were identified and localized. This Azo-enabled ECM proteomics strategy will streamline the analysis of ECM proteins and promote the study of ECM biology.


Subject(s)
Azo Compounds/chemistry , Extracellular Matrix/chemistry , Neoplasm Proteins/analysis , Proteomics , Surface-Active Agents/chemistry , Animals , Antigens, Polyomavirus Transforming/chemistry , Extracellular Matrix/metabolism , Mammary Tumor Virus, Mouse/chemistry , Mass Spectrometry , Mice , Mice, Transgenic , Neoplasm Proteins/metabolism , Photochemical Processes , Solubility
2.
Breast Cancer Res ; 20(1): 66, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29976229

ABSTRACT

BACKGROUND: Syndecan-1 (Sdc1), a cell surface heparan sulfate proteoglycan normally expressed primarily by epithelia and plasma cells, is aberrantly induced in stromal fibroblasts of breast carcinomas. Stromal fibroblast-derived Sdc1 participates in paracrine growth stimulation of breast carcinoma cells and orchestrates stromal extracellular matrix fiber alignment, thereby creating a migration and invasion-permissive microenvironment. Here, we specifically tested the role of stromal Sdc1 in metastasis. METHODS: The metastatic potential of the aggressive mouse mammary carcinoma cell lines, 4T1 and E0776, was tested in wild-type and genetically Sdc1-deficient host animals. Metastatic lesions were characterized by immunohistochemical analysis. RESULTS: After orthotopic inoculation, the lung metastatic burden was reduced in Sdc1-/- animals by 97% and more than 99%, in BALB/cJ and C57BL/6 animals, respectively. The difference in metastatic efficiency was maintained when the tumor cells were injected into the tail vein, suggesting that host Sdc1 exerts its effect during later stages of the metastatic cascade. Co-localization studies identified Sdc1 expression in stromal fibroblasts within the metastatic microenvironment and in normal airway epithelial cells but not in other cells (endothelial cells, α-smooth muscle actin positive cells, leucocytes, macrophages). The Ki67 proliferation index and the rate of apoptosis of the metastatic tumor cells were diminished in Sdc1-/- vs. Sdc1+/+ animals, and leucocyte density was indistinguishable. Sdc1-mediated metastatic efficiency was abolished when the animals were housed at a thermoneutral ambient temperature of 31 °C, suggesting that the host Sdc1 effect on metastasis requires mild cold stress. CONCLUSIONS: In summary, Sdc1 is induced in the lung microenvironment after mammary carcinoma cell dissemination and promotes outgrowth of metastases in a temperature-dependent manner.


Subject(s)
Breast Neoplasms/genetics , Lung Neoplasms/genetics , Mammary Neoplasms, Animal/genetics , Syndecan-1/genetics , Tumor Microenvironment/genetics , Animals , Apoptosis/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Metastasis
3.
Cancer Res ; 78(7): 1672-1684, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29363543

ABSTRACT

Although antiestrogen therapies are successful in many patients with estrogen receptor alpha-positive (ERα+) breast cancer, 25% to 40% fail to respond. Although multiple mechanisms underlie evasion of these treatments, including tumor heterogeneity and drug-resistant cancer stem cells (CSC), further investigations have been limited by the paucity of preclinical ERα+ tumor models. Here, we examined a mouse model of prolactin-induced aggressive ERα+ breast cancer, which mimics the epidemiologic link between prolactin exposure and increased risk for metastatic ERα+ tumors. Like a subset of ERα+ patient cancers, the prolactin-induced adenocarcinomas contained two major tumor subpopulations that expressed markers of normal luminal and basal epithelial cells. CSC activity was distributed equally across these two tumor subpopulations. Treatment with the selective estrogen receptor downregulator (SERD), ICI 182,780 (ICI), did not slow tumor growth, but induced adaptive responses in CSC activity, increased markers of plasticity including target gene reporters of Wnt/Notch signaling and epithelial-mesenchymal transition, and increased double-positive (K8/K5) cells. In primary tumorsphere cultures, ICI stimulated CSC self-renewal and was able to overcome the dependence of self-renewal upon Wnt or Notch signaling individually, but not together. Our findings demonstrate that treatment of aggressive mixed lineage ERα+ breast cancers with a SERD does not inhibit growth, but rather evokes tumor cell plasticity and regenerative CSC activity, predicting likely negative impacts on patient tumors with these characteristics.Significance: This study suggests that treatment of a subset of ERα+ breast cancers with antiestrogen therapies may not only fail to slow growth but also promote aggressive behavior by evoking tumor cell plasticity and regenerative CSC activity. Cancer Res; 78(7); 1672-84. ©2018 AACR.


Subject(s)
Adenocarcinoma/drug therapy , Breast Neoplasms/drug therapy , Cell Plasticity/drug effects , Estrogen Receptor Antagonists/therapeutic use , Estrogen Receptor alpha/antagonists & inhibitors , Mammary Neoplasms, Animal/drug therapy , Neoplastic Stem Cells/pathology , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Estrogen Receptor alpha/metabolism , Female , Fulvestrant/therapeutic use , Humans , Mammary Neoplasms, Animal/chemically induced , Mammary Neoplasms, Animal/pathology , Mice , Prolactin/adverse effects , Tumor Cells, Cultured , Wnt Signaling Pathway/physiology
4.
Cancer Epidemiol Biomarkers Prev ; 27(2): 138-145, 2018 02.
Article in English | MEDLINE | ID: mdl-29141852

ABSTRACT

Background: Collagen fibers surrounding breast ducts may influence breast cancer progression. Syndecan-1 interacts with constituents in the extracellular matrix, including collagen fibers, and may contribute to cancer cell migration. Thus, the orientation of collagen fibers surrounding ductal carcinoma in situ (DCIS) lesions and stromal syndecan-1 expression may predict recurrence.Methods: We evaluated collagen fiber alignment and syndecan-1 expression in 227 women diagnosed with DCIS in 1995 to 2006 followed through 2014 (median, 14.5 years; range, 0.7-17.6). Stromal collagen alignment was evaluated from diagnostic tissue slides using second harmonic generation microscopy and fiber analysis software. Univariate analysis was conducted using χ2 tests and ANOVA. The association between collagen alignment z-scores, syndecan-1 staining intensity, and time to recurrence was evaluated using HRs and 95% confidence intervals (CIs).Results: Greater fiber angles surrounding DCIS lesions, but not syndecan-1 staining intensity, were related to positive HER2 (P = 0.002) status, comedo necrosis (P = 0.03), and negative estrogen receptor (P = 0.002) and progesterone receptor (P = 0.02) status. Fiber angle distributions surrounding lesions included more angles closer to 90 degrees than normal ducts (P = 0.06). Collagen alignment z-scores for DCIS lesions were positively related to recurrence (HR = 1.25; 95% CI, 0.84-1.87 for an interquartile range increase in average fiber angles).Conclusions: Although collagen alignment and stromal syndecan-1 expression did not predict recurrence, collagen fibers perpendicular to the duct perimeter were more frequent in DCIS lesions with features typical of poor prognosis.Impact: Follow-up studies are warranted to examine whether additional features of the collagen matrix may more strongly predict patient outcomes. Cancer Epidemiol Biomarkers Prev; 27(2); 138-45. ©2017 AACR.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Collagen/metabolism , Disease Progression , Neoplasm Recurrence, Local , Syndecan-1/metabolism , Adult , Aged , Analysis of Variance , Biomarkers, Tumor/metabolism , Collagen/chemistry , Disease-Free Survival , Female , Humans , Longitudinal Studies , Middle Aged , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
7.
PLoS One ; 11(2): e0150132, 2016.
Article in English | MEDLINE | ID: mdl-26909794

ABSTRACT

Expression of the cell surface proteoglycan syndecan-1 (Sdc1) is frequently induced in stromal fibroblasts of invasive breast carcinomas. We have recently identified a correlation between stromal Sdc1 expression and extracellular matrix (ECM) fiber alignment, both in vitro and in vivo. ECMs derived from Sdc1-positive human mammary fibroblasts (HMF) showed an aligned fiber architecture, which contrasted markedly with the more random fiber arrangement in the ECM produced by Sdc1-negative HMFs. We further demonstrated that aligned fiber architecture promotes the directional migration and invasion of breast carcinoma cells. To decipher the molecular mechanisms governing the formation of an aligned, invasion-permissive ECM, a series of Sdc1 mutants was introduced into HMF. We found that both the ectodomain and heparan sulfate chains of Sdc1 were required for full activity of Sdc1 in regulating ECM alignment, while transmembrane and cytoplasmic domains were dispensable. Sdc1 regulates the activities of several integrins via its ectodomain. Integrins are key players in the assembly of fibronectin-rich ECM. In addition, integrins are capable of regulating cell morphology and cell shape and orientation may affect ECM architecture. Therefore, we investigated the role of integrins in Sdc1-mediated ECM fiber alignment. Sdc1-overexpressing HMF gained an enhanced spindle-shaped morphology when cultured in an overconfluent state under conditions permissive for ECM production, which was partially reversed by siRNA-mediated silencing of ß3 integrin expression. Moreover, suppression of αvß3 integrin activity by a function-blocking antibody or ß3 knockdown largely abolished the aligned ECM fiber architecture and consequently the invasion-permissive properties of the ECM induced by Sdc1. The results suggest that Sdc1 may modulate fibronectin fibrillogenesis and/or alter cell morphology during ECM production through αvß3 integrin, thereby mediating ECM fiber alignment. Understanding the mechanisms governing ECM organization may lead to the development of novel stroma-targeted therapy for breast cancer, aiming at converting an invasion-permissive to an invasion-restrictive microenvironment.


Subject(s)
Extracellular Matrix/metabolism , Fibroblasts/metabolism , Heparitin Sulfate/metabolism , Integrin alphaVbeta3/metabolism , Syndecan-1/metabolism , Cell Line , Extracellular Matrix/genetics , Female , Fibronectins/genetics , Fibronectins/metabolism , Heparitin Sulfate/genetics , Humans , Integrin alphaVbeta3/genetics , Syndecan-1/genetics
8.
Adv Exp Med Biol ; 846: 265-80, 2015.
Article in English | MEDLINE | ID: mdl-25472543

ABSTRACT

The signal transduction events that orchestrate cellular activities required for angiogenesis remain incompletely understood. We and others recently described that proangiogenic mediators such as fibroblast growth factors can activate members of the signal transducers and activators of transcription (STAT) family. STAT5 activation is necessary and sufficient to induce migration, invasion and tube formation of endothelial cells. STAT5 effects on endothelial cells require the secretion of the prolactin (PRL) family member proliferin-1 (PLF1) in mice and PRL in humans. In human endothelial cells, PRL activates the PRL receptor (PRLR) resulting in MAPK and STAT5 activation, thus closing a positive feedback loop. In vivo, endothelial cell-derived PRL is expected to combine with PRL of tumor cell and pituitary origin to raise the concentration of this polypeptide hormone in the tumor microenvironment. Thus, PRL may stimulate tumor angiogenesis via autocrine, paracrine, and endocrine pathways. The disruption of tumor angiogenesis by interfering with PRL signaling may offer an attractive target for therapeutic intervention.


Subject(s)
Feedback, Physiological/physiology , Neovascularization, Pathologic , Neovascularization, Physiologic , Prolactin/metabolism , STAT5 Transcription Factor/metabolism , Animals , Cell Movement/drug effects , Cell Movement/genetics , Endothelial Cells/drug effects , Endothelial Cells/physiology , Fibroblast Growth Factors/pharmacology , Humans , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/genetics , Prolactin/genetics , STAT5 Transcription Factor/genetics
9.
PLoS One ; 8(10): e76373, 2013.
Article in English | MEDLINE | ID: mdl-24124550

ABSTRACT

The utilization of 3D, physiologically relevant in vitro cancer models to investigate complex interactions between tumor and stroma has been increasing. Prior work has generally focused on the cancer cells and, the role of fibroblast culture conditions on tumor-stromal cell interactions is still largely unknown. Here, we focus on the stroma by comparing functional behaviors of human mammary fibroblasts (HMFs) cultured in 2D and 3D and their effects on the invasive progression of breast cancer cells (MCF10DCIS.com). We identified increased levels of several paracrine factors from HMFs cultured in 3D conditions that drive the invasive transition. Using a microscale co-culture model with improved compartmentalization and sensitivity, we demonstrated that HMFs cultured in 3D intensify the promotion of the invasive progression through the HGF/c-Met interaction. This study highlights the importance of the 3D stromal microenvironment in the development of multiple cell type in vitro cancer models.


Subject(s)
Breast Neoplasms/metabolism , Coculture Techniques , Fibroblasts/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Fibroblasts/pathology , Hepatocyte Growth Factor/biosynthesis , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Microfluidic Analytical Techniques/methods , Neoplasm Invasiveness , Phenotype , Stromal Cells/metabolism , Stromal Cells/pathology
10.
Mol Cell Biol ; 33(22): 4408-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24019070

ABSTRACT

Malignant gliomas are highly lethal neoplasms with limited treatment options. We previously found that the heparan sulfate proteoglycan glypican 1 (GPC1) is universally and highly expressed in human gliomas. In this study, we investigated the biological activity of GPC1 expression in both human glioma cells and normal astrocytes in vitro. Expression of GPC1 inactivates the G1/S checkpoint and strongly stimulates DNA replication. Constitutive expression of GPC1 causes DNA rereplication and DNA damage, suggesting a mutagenic activity for GPC1. GPC1 expression leads to a significant downregulation of the tumor suppressors pRb, Cip/Kip cyclin-dependent kinase inhibitors (CKIs), and CDH1, and upregulation of the pro-oncogenic proteins cyclin E, cyclin-dependent kinase 2 (CDK2), Skp2, and Cdt1. These GPC1-induced changes are accompanied by a significant reduction in all types of D cyclins, which is independent of serum supplementation. It is likely that GPC1 stimulates the so-called Skp2 autoinduction loop, independent of cyclin D-CDK4/6. Knockdown of Skp2, CDK2, or cyclin E, three key elements within the network modulated by GPC1, results in a reduction of the S phase and aneuploid fractions, implying a functional role for these regulators in GPC1-induced S phase entry and DNA rereplication. In addition, a significant activation of both the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathways by GPC1 is seen in normal human astrocytes even in the presence of growth factor supplement. Both pathways are constitutively activated in human gliomas. The surprising magnitude and the mitogenic and mutagenic nature of the effect exerted by GPC1 on the cell cycle imply that GPC1 may play an important role in both glioma tumorigenesis and growth.


Subject(s)
Brain Neoplasms/metabolism , DNA Replication , Glioblastoma/metabolism , Glypicans/metabolism , S Phase , Aneuploidy , Astrocytes/cytology , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , DNA/genetics , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Glypicans/genetics , Humans
11.
J Biol Chem ; 288(29): 21184-21196, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23729680

ABSTRACT

We have shown previously that the murine prolactin/growth hormone family member proliferin plays a pivotal role in angiogenesis induced by the FGF2/STAT5 signaling cascade. To delineate the signaling pathway downstream of STAT5 in the human system, where proliferin does not exist, we expressed constitutively active (CA) or dominant-negative (DN) mutant STAT5A in hCMEC/D3 human brain endothelial cells. We found that conditioned medium from CA-STAT5A- but not from DN-STAT5A-overexpressing endothelial cells (EC) is sufficient to induce EC migration and tube formation but not proliferation, indicating that STAT5A regulates the secretion of autocrine proangiogenic factors. We identified prolactin (PRL) as a candidate autocrine factor. CA-STAT5A expression stimulates PRL production at the RNA and protein level, and STAT5A binds to the PRL promoter region, suggesting direct transcriptional regulation. Medium conditioned by CA-STAT5A-overexpressing EC induces phosphorylation of the PRL receptor and activates MAPK. Knockdown of PRL expression by shRNA or blocking of PRL activity with neutralizing antibodies removed the CA-STAT5A-dependent proangiogenic activity from the conditioned medium of EC. The addition of recombinant PRL restores this activity. STAT5A-induced PRL in the conditioned medium can activate STAT5, STAT1, and to a lesser extent STAT3 in hCMEC/D3 cells, suggesting the existence of a positive feedback loop between STAT5 and PRL that promotes angiogenesis. Furthermore, we find that VEGF, a potent proangiogenic factor, is induced by activation of STAT5A, and VEGF induction depends on PRL expression. These observations demonstrate a STAT5/PRL/VEGF signaling cascade in human brain EC and implicate PRL and VEGF as autocrine regulators of EC migration, invasion, and tube formation.


Subject(s)
Autocrine Communication , Feedback, Physiological , Neovascularization, Physiologic , Prolactin/metabolism , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , Angiogenesis Inducing Agents/metabolism , Animals , Autocrine Communication/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological/drug effects , Gene Silencing/drug effects , Humans , Mice , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , Prolactin/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , RNA, Small Interfering/metabolism , Receptors, Prolactin/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Anticancer Res ; 33(4): 1525-36, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23564794

ABSTRACT

BACKGROUND/AIM: The tumor microenvironment plays a major role in tumor growth and progression. Its manipulation can lead to a reversion of the malignant phenotype. Here we explored the ability of normal mammary fibroblasts (HMFs) to induce reversion of the malignant phenotype of primary breast carcinoma cells (PBCs) in a three-dimensional (3D) context. MATERIALS AND METHODS: PBCs were isolated from 13 primary breast carcinomas and cultured in 3D collagen-I gels as mono- or co-culture with HMFs. RESULTS: In five co-cultures, PBCs exhibited reversion of their malignant phenotype, whereas PBCs in matched monocultures exhibited disorganized growth. Reversion, defined as the restoration of the complete baso-apical polarity axis, was confirmed with established polarity markers. Secretion of the tissue-specific glycoprotein MAM-6 into the acinar lumens and deposition of basement membrane indicated functional differentiation. Gene expression analysis revealed a set of differentially regulated genes which possibly affect the reversion process. These included MAL, ELF5, MAP6, ZMYND11 and SQLE. CONCLUSION: These findings highlight the significant role of fibroblasts in regulating the carcinoma phenotype.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Breast/cytology , Cell Transformation, Neoplastic , Fibroblasts/cytology , Tumor Microenvironment , Breast/metabolism , Breast Neoplasms/metabolism , Cell Communication , Cell Culture Techniques , Coculture Techniques , Female , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Phenotype , Tumor Cells, Cultured
13.
PLoS One ; 7(10): e46685, 2012.
Article in English | MEDLINE | ID: mdl-23056402

ABSTRACT

Stromal fibroblasts actively participate in normal mammary gland homeostasis and in breast carcinoma growth and progression by secreting paracrine factors; however, little is known about the identity of paracrine mediators in individual patients. The purpose of this study was to characterize paracrine signaling pathways between breast carcinoma cells and breast carcinoma-associated fibroblasts (CAF) or normal mammary fibroblasts (NF), respectively. CAF and NF were isolated from breast carcinoma tissue samples and adjacent normal mammary gland tissue of 28 patients. The fibroblasts were grown in 3D collagen gel co-culture with T47D human breast carcinoma cells and T47D cell growth was measured. CAF stimulated T47D cell growth to a significantly greater degree than NF. We detected a considerable inter-individual heterogeneity of paracrine interactions but identified FGF2, HB-EGF, heparanase-1 and SDF1 as factors that were consistently responsible for the activity of carcinoma-associated fibroblasts. CAF from low-grade but not high-grade carcinomas required insulin-like growth factor 1 and transforming growth factor beta 1 to stimulate carcinoma growth. Paradoxically, blocking of membrane-type 1 matrix metalloprotease stimulated T47D cell growth in co-culture with NF. The results were largely mirrored by treating the fibroblasts with siRNA oligonucleotides prior to co-culture, implicating the fibroblasts as principal production site for the secreted mediators. In summary, we identify a paracrine signaling network with inter-individual commonalities and differences. These findings have significant implications for the design of stroma-targeted therapies.


Subject(s)
Breast Neoplasms/metabolism , Fibroblasts/metabolism , Breast , Breast Neoplasms/genetics , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Female , Humans , Paracrine Communication/genetics , Paracrine Communication/physiology , Tumor Cells, Cultured
14.
Nat Genet ; 44(8): 910-5, 2012 Jul 08.
Article in English | MEDLINE | ID: mdl-22772369

ABSTRACT

Chronic kidney disease (CKD) represents a major health burden. Its central feature of renal fibrosis is not well understood. By exome sequencing, we identified mutations in FAN1 as a cause of karyomegalic interstitial nephritis (KIN), a disorder that serves as a model for renal fibrosis. Renal histology in KIN is indistinguishable from that of nephronophthisis, except for the presence of karyomegaly. The FAN1 protein has nuclease activity and acts in DNA interstrand cross-link (ICL) repair within the Fanconi anemia DNA damage response (DDR) pathway. We show that cells from individuals with FAN1 mutations have sensitivity to the ICL-inducing agent mitomycin C but do not exhibit chromosome breakage or cell cycle arrest after diepoxybutane treatment, unlike cells from individuals with Fanconi anemia. We complemented ICL sensitivity with wild-type FAN1 but not with cDNA having mutations found in individuals with KIN. Depletion of fan1 in zebrafish caused increased DDR, apoptosis and kidney cysts. Our findings implicate susceptibility to environmental genotoxins and inadequate DNA repair as novel mechanisms contributing to renal fibrosis and CKD.


Subject(s)
DNA Repair/genetics , Exodeoxyribonucleases/genetics , Mutation , Nephritis, Interstitial/genetics , Renal Insufficiency, Chronic/genetics , Animals , Cell Line , DNA Damage , Endodeoxyribonucleases , Fanconi Anemia Complementation Group D2 Protein/genetics , Gene Knockdown Techniques , Genes, Recessive , Genetic Complementation Test , Humans , Multifunctional Enzymes , Nephritis, Interstitial/complications , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Zebrafish/abnormalities , Zebrafish/genetics
15.
Cancer Lett ; 325(2): 175-88, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-22776560

ABSTRACT

Stromal factors play a critical role in the development of the mammary gland. Using a three dimensional-coculture model we demonstrate a significant role for stromal fibroblasts in the regulation of normal mammary epithelial morphogenesis and the control of tumor growth. Both soluble factors secreted by fibroblasts and fibroblast-derived modifications of the matrix compliance contribute to the regulation of epithelial cell morphogenesis. Readjustment of matrix tension by fibroblasts can even induce a phenotypic reversion of breast carcinoma cells. These data offer a basis to develop new strategies for the normalization of the tumor stroma as an innovative target in cancer therapy.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Epithelial Cells/cytology , Fibroblasts/physiology , Tumor Microenvironment/physiology , Breast/pathology , Cell Differentiation , Cell Division , Cell Shape , Cells, Cultured/physiology , Coculture Techniques , Compliance , Epithelial Cells/pathology , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Humans , Morphogenesis , Paracrine Communication , Phenotype , Stress, Mechanical , Stromal Cells/physiology , Tumor Cells, Cultured/ultrastructure
16.
Cancer Res ; 72(13): 3207-16, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22532168

ABSTRACT

The transcription factor RE1 silencing transcription factor (REST) is lost in approximately 20% of breast cancers. Although it is known that these RESTless tumors are highly aggressive and include all tumor subtypes, the underlying tumorigenic mechanisms remain unknown. In this study, we show that loss of REST results in upregulation of LIN28A, a known promoter of tumor development, in breast cancer cell lines and human breast tumors. We found that LIN28A was a direct transcriptional target of REST in cancer cells and that loss of REST resulted in increased LIN28A expression and enhanced tumor growth both in vitro and in vivo, effects that were dependent on heightened LIN28A expression. Tumors lacking REST expression were locally invasive, consistent with the increased lymph node involvement observed in human RESTless tumors. Clinically, human RESTless breast tumors also displayed significantly enhanced LIN28A expression when compared with non-RESTless tumors. Our findings therefore show a critical role for the REST-LIN28A axis in tumor aggression and suggest a causative relationship between REST loss and tumorigenicity in vivo.


Subject(s)
Breast Neoplasms/pathology , Cell Division/physiology , DNA-Binding Proteins/biosynthesis , Repressor Proteins/metabolism , Animals , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/physiology , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Mice , RNA-Binding Proteins , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics
17.
J Biol Chem ; 287(9): 6490-502, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22199350

ABSTRACT

Multiple secreted factors induce the formation of new blood vessels (angiogenesis). The signal transduction events that orchestrate the numerous cellular activities required for angiogenesis remain incompletely understood. We have shown previously that STAT5 plays a pivotal role in angiogenesis induced by FGF2 and FGF8b. To delineate the signaling pathway downstream of STAT5, we expressed constitutively active (CA) or dominant-negative (DN) mutant STAT5A in mouse brain endothelial cells (EC). We found that the conditioned medium from CA-STAT5A but not from dominant-negative STAT5A overexpressing EC is sufficient to induce EC invasion and tube formation, indicating that STAT5A regulates the secretion of autocrine proangiogenic factors. Conversely, CA-STAT5A-induced conditioned medium had no effect on EC proliferation. Using a comparative genome-wide transcription array screen, we identified the prolactin family member proliferin (PLF1 and PLF4) as a candidate autocrine factor. The CA-STAT5A-dependent transcription and secretion of PLF by EC was confirmed by quantitative RT-PCR and Western blotting, respectively. CA-STAT5A binds to the PLF1 promoter region, suggesting a direct transcriptional regulation. Knockdown of PLF expression by shRNA or by blocking of PLF activity with neutralizing antibodies removed the CA-STAT5A-dependent proangiogenic activity from the conditioned medium of EC. Similarly, the ability of concentrated conditioned medium from CA-STAT5A transfected EC to induce angiogenesis in Matrigel plugs in vivo was abolished when PLF was depleted from the medium. These observations demonstrate a FGF/STAT5/PLF signaling cascade in EC and implicate PLF as autocrine regulator of EC invasion and tube formation.


Subject(s)
Autocrine Communication/physiology , Endothelial Cells/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/physiology , STAT5 Transcription Factor/metabolism , Animals , Brain/blood supply , Cell Line, Transformed , Cell Movement/physiology , Culture Media, Conditioned/pharmacology , Endothelial Cells/cytology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mice , Oligonucleotide Array Sequence Analysis , Prolactin , Promoter Regions, Genetic/physiology , RNA, Small Interfering/genetics , STAT5 Transcription Factor/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
J Biol Chem ; 287(8): 5898-909, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22203671

ABSTRACT

The heparan sulfate proteoglycan glypican-1 (GPC1) is involved in tumorigenesis and angiogenesis and is overexpressed frequently in tumor and endothelial cells (ECs) in human gliomas. We demonstrated previously that in brain EC, GPC1 regulates mitotic cyclins and securin as well as mitosis and that GPC1 is required for progression through the cell cycle. To characterize the molecular mechanism underlying cell cycle regulation by GPC1, we systematically investigated its effects on key G(1)/S checkpoint regulators and on major signaling pathways reportedly activated by Dally (Division abnormally delayed) the Drosophila GPC1 homologue. We found that elevated GPC1 affected a wide range of G(1)/S checkpoint regulators, leading to inactivation of the G(1)/S checkpoint and increased S phase entry, apparently by activating the mitogen-independent Skp2 autoinduction loop. Specifically, GPC1 suppressed CDK inhibitors (CKIs), including p21, p27, p16, and p19, and the D cyclins, and induced CDK2 and Skp2. GPC1 may trigger the Skp2 autoinduction loop at least partially by suppressing p21 transcription as knockdown of p21 by RNAi can mimic the effect of GPC1 on the cell cycle regulators related to the loop. Moreover, multiple mitogenic signaling pathways, including ERK MAPK, Wnt and BMP signaling, were significantly stimulated by GPC1 as has been reported for Dally in Drosophila. Notably, the c-Myc oncoprotein, which is frequently up-regulated by both ERK and Wnt signaling and functions as a potent transcription repressor for CKIs as well as D cyclins, was also significantly induced by GPC1. These findings provide mechanistic insights into how GPC1 regulates the cell cycle and proliferation.


Subject(s)
Endothelial Cells/cytology , G1 Phase Cell Cycle Checkpoints , Glypicans/metabolism , S Phase Cell Cycle Checkpoints , S-Phase Kinase-Associated Proteins/metabolism , Animals , Cell Division , Cell Proliferation , Cyclin D/metabolism , Cyclin-Dependent Kinases/metabolism , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Signal Transduction/drug effects
19.
Cell Tissue Res ; 346(2): 223-36, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987222

ABSTRACT

During cancer cell growth many tumors exhibit various grades of desmoplasia, unorganized production of fibrous or connective tissue, composed mainly of collagen fibers and myofibroblasts. The accumulation of an extracellular matrix (ECM) surrounding tumors directly affects cancer cell proliferation, migration and spread; therefore the study of desmoplasia is of vital importance. Stromal fibroblasts surrounding tumors are activated to myofibroblasts and become the primary producers of ECM during desmoplasia. The composition, density and organization of this ECM accumulation play a major role on the influence desmoplasia has upon tumor cells. In this study, we analyzed desmoplasia in vivo in human colorectal carcinoma tissue, detecting an up-regulation of collagen I, collagen IV and collagen V in human colorectal cancer desmoplastic reaction. These components were then analyzed in vitro co-cultivating colorectal cancer cells (Caco-2 and HCT116) and fibroblasts utilizing various co-culture techniques. Our findings demonstrate that direct cell-cell contact between fibroblasts and colorectal cancer cells evokes an increase in ECM density, composed of unorganized collagens (I, III, IV and V) and proteoglycans (biglycan, fibromodulin, perlecan and versican). The desmoplastic collagen fibers were thick, with an altered orientation, as well as deposited as bundles. This increased ECM density inhibited the migration and invasion of the colorectal tumor cells in both 2D and 3D co-culture systems. Therefore this study sheds light on a possible restricting role desmoplasia could play in colorectal cancer invasion.


Subject(s)
Collagen/biosynthesis , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Up-Regulation , Aged , Cell Line, Tumor , Coculture Techniques , Collagen/genetics , Colorectal Neoplasms/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Invasiveness , Proteoglycans/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology
20.
Am J Pathol ; 178(3): 1221-32, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21356373

ABSTRACT

Evidence for the potent influence of stromal organization and function on invasion and metastasis of breast tumors is ever growing. We have performed a rigorous examination of the relationship of a tumor-associated collagen signature-3 (TACS-3) to the long-term survival rate of human patients. TACS-3 is characterized by bundles of straightened and aligned collagen fibers that are oriented perpendicular to the tumor boundary. An evaluation of TACS-3 was performed in biopsied tissue sections from 196 patients by second harmonic generation imaging of the backscattered signal generated by collagen. Univariate analysis of a Cox proportional hazard model demonstrated that the presence of TACS-3 was associated with poor disease-specific and disease-free survival, resulting in hazard ratios between 3.0 and 3.9. Furthermore, TACS-3 was confirmed to be an independent prognostic indicator regardless of tumor grade and size, estrogen or progesterone receptor status, human epidermal growth factor receptor-2 status, node status, and tumor subtype. Interestingly, TACS-3 was positively correlated to expression of stromal syndecan-1, a receptor for several extracellular matrix proteins including collagens. Because of the strong statistical evidence for poor survival in patients with TACS, and because the assessment can be performed in routine histopathological samples imaged via second harmonic generation or using picrosirius, we propose that quantifying collagen alignment is a viable, novel paradigm for the prediction of human breast cancer survival.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Collagen/metabolism , Biopsy , Breast Neoplasms/classification , Diagnostic Imaging , Female , Humans , Multivariate Analysis , Prognosis , Regression Analysis , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...