Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 15(4): e0231711, 2020.
Article in English | MEDLINE | ID: mdl-32298357

ABSTRACT

Actively growing tumors are often histologically associated with Ki67 positivity, while the detection of invasiveness relies on non-quantitative pathologic evaluation of mostly advanced tumors. We recently reported that reduced expression of the Ca2+-dependent membrane-binding annexin A6 (AnxA6) is associated with increased expression of the Ca2+ activated RasGRF2 (GRF2), and that the expression status of these proteins inversely influence the growth and motility of triple negative breast cancer (TNBC) cells. Here, we establish that the reciprocal expression of AnxA6 and GRF2 is at least in part, dependent on inhibition of non-selective Ca2+ channels in AnxA6-low but not AnxA6-high TNBC cells. Immunohistochemical staining of breast cancer tissues revealed that compared to non-TNBC tumors, TNBC tumors express lower levels of AnxA6 and higher Ki67 expression. GRF2 expression levels strongly correlated with high Ki67 in pretreatment biopsies from patients with residual disease and with residual tumor size following chemotherapy. Elevated AnxA6 expression more reliably identified patients who responded to chemotherapy, while low AnxA6 levels were significantly associated with shorter distant relapse-free survival. Finally, the reciprocal expression of AnxA6 and GRF2 can delineate GRF2-low/AnxA6-high invasive from GRF2-high/AnxA6-low rapidly growing TNBCs. These data suggest that AnxA6 may be a reliable biomarker for distant relapse-free survival and response of TNBC patients to chemotherapy, and that the reciprocal expression of AnxA6 and GRF2 can reliably delineate TNBCs into rapidly growing and invasive subsets which may be more relevant for subset-specific therapeutic interventions.


Subject(s)
Annexin A6/metabolism , Calcium Channels/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , ras Guanine Nucleotide Exchange Factors/metabolism , Animals , Annexin A6/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Calcium Channel Blockers/pharmacology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Humans , Ki-67 Antigen/metabolism , Mice , Neoplasm Metastasis/genetics , Prognosis , Transplantation, Heterologous , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , ras Guanine Nucleotide Exchange Factors/genetics
2.
Cancer Med ; 5(12): 3532-3543, 2016 12.
Article in English | MEDLINE | ID: mdl-27882696

ABSTRACT

Glioblastomas (high-grade astrocytomas) are highly aggressive brain tumors with poor prognosis and limited treatment options. In the present studies, we have defined the role of fetuin-A, a liver-derived multifunctional serum protein, in the growth of an established glioblastoma cell line, LN229. We hereby demonstrate that these cells synthesize ectopic fetuin-A which supports their growth in culture in the absence of serum. We have demonstrated that a panel of tissue microarray (TMA) of glioblastomas also express ectopic fetuin-A. Knocking down fetuin-A using shRNA approach in LN229, significantly reduced their in vitro growth as well as growth and invasion in vivo. The fetuin-A knockdown subclones of LN229 (A and D) also had reduced motility and invasive capacity. Treatment of LN229 cells with asialofetuin (ASF), attenuated their uptake of labeled fetuin-A, and induced senescence in them. Interestingly, the D subclone that had ~90% reduction in ectopic fetuin-A, underwent senescence in serum-free medium which was blunted in the presence of purified fetuin-A. Uptake of labeled exosomes was attenuated in fetuin-A knockdown subclones A and D. Taken together, the studies demonstrate the impact of fetuin-A as significant node of growth, motility, and invasion signaling in glioblastomas that can be targeted for therapy.


Subject(s)
Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , alpha-2-HS-Glycoprotein/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cellular Senescence/genetics , Disease Models, Animal , Exosomes/metabolism , Gene Expression , Gene Knockdown Techniques , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Grading , alpha-2-HS-Glycoprotein/metabolism
3.
Carcinogenesis ; 36 Suppl 1: S128-59, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106135

ABSTRACT

The purpose of this review is to stimulate new ideas regarding low-dose environmental mixtures and carcinogens and their potential to promote invasion and metastasis. Whereas a number of chapters in this review are devoted to the role of low-dose environmental mixtures and carcinogens in the promotion of invasion and metastasis in specific tumors such as breast and prostate, the overarching theme is the role of low-dose carcinogens in the progression of cancer stem cells. It is becoming clearer that cancer stem cells in a tumor are the ones that assume invasive properties and colonize distant organs. Therefore, low-dose contaminants that trigger epithelial-mesenchymal transition, for example, in these cells are of particular interest in this review. This we hope will lead to the collaboration between scientists who have dedicated their professional life to the study of carcinogens and those whose interests are exclusively in the arena of tissue invasion and metastasis.


Subject(s)
Carcinogens, Environmental/adverse effects , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Animals , Disease Progression , Environmental Exposure/adverse effects , Epithelial-Mesenchymal Transition/drug effects , Humans
4.
Carcinogenesis ; 36 Suppl 1: S254-96, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106142

ABSTRACT

Lifestyle factors are responsible for a considerable portion of cancer incidence worldwide, but credible estimates from the World Health Organization and the International Agency for Research on Cancer (IARC) suggest that the fraction of cancers attributable to toxic environmental exposures is between 7% and 19%. To explore the hypothesis that low-dose exposures to mixtures of chemicals in the environment may be combining to contribute to environmental carcinogenesis, we reviewed 11 hallmark phenotypes of cancer, multiple priority target sites for disruption in each area and prototypical chemical disruptors for all targets, this included dose-response characterizations, evidence of low-dose effects and cross-hallmark effects for all targets and chemicals. In total, 85 examples of chemicals were reviewed for actions on key pathways/mechanisms related to carcinogenesis. Only 15% (13/85) were found to have evidence of a dose-response threshold, whereas 59% (50/85) exerted low-dose effects. No dose-response information was found for the remaining 26% (22/85). Our analysis suggests that the cumulative effects of individual (non-carcinogenic) chemicals acting on different pathways, and a variety of related systems, organs, tissues and cells could plausibly conspire to produce carcinogenic synergies. Additional basic research on carcinogenesis and research focused on low-dose effects of chemical mixtures needs to be rigorously pursued before the merits of this hypothesis can be further advanced. However, the structure of the World Health Organization International Programme on Chemical Safety 'Mode of Action' framework should be revisited as it has inherent weaknesses that are not fully aligned with our current understanding of cancer biology.


Subject(s)
Carcinogenesis/chemically induced , Carcinogens, Environmental/adverse effects , Environmental Exposure/adverse effects , Hazardous Substances/adverse effects , Neoplasms/chemically induced , Neoplasms/etiology , Animals , Humans
5.
Mol Cancer ; 12(1): 167, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24354805

ABSTRACT

BACKGROUND: The expression of annexin A6 (AnxA6) in AnxA6-deficient non-invasive tumor cells has been shown to terminate epidermal growth factor receptor (EGFR) activation and downstream signaling. However, as a scaffolding protein, AnxA6 may stabilize activated cell-surface receptors to promote cellular processes such as tumor cell motility and invasiveness. In this study, we investigated the contribution of AnxA6 in the activity of EGFR in invasive breast cancer cells and examined whether the expression status of AnxA6 influences the response of these cells to EGFR-targeted tyrosine kinase inhibitors (TKIs) and/or patient survival. RESULTS: We demonstrate that in invasive BT-549 breast cancer cells AnxA6 expression is required for sustained membrane localization of activated (phosho-Y1068) EGFR and consequently, persistent activation of MAP kinase ERK1/2 and phosphoinositide 3-kinase/Akt pathways. Depletion of AnxA6 in these cells was accompanied by rapid degradation of activated EGFR, attenuated downstream signaling and as expected enhanced anchorage-independent growth. Besides inhibition of cell motility and invasiveness, AnxA6-depleted cells were also more sensitive to the EGFR-targeted TKIs lapatinib and PD153035. We also provide evidence suggesting that reduced AnxA6 expression is associated with a better relapse-free survival but poorer distant metastasis-free and overall survival of basal-like breast cancer patients. CONCLUSIONS: Together this demonstrates that the rapid degradation of activated EGFR in AnxA6-depleted invasive tumor cells underlies their sensitivity to EGFR-targeted TKIs and reduced motility. These data also suggest that AnxA6 expression status may be useful for the prediction of the survival and likelihood of basal-like breast cancer patients to respond to EGFR-targeted therapies.


Subject(s)
Annexin A6/genetics , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Quinazolines/pharmacology , Annexin A6/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease-Free Survival , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , Female , Gene Expression , Humans , Inhibitory Concentration 50 , Kaplan-Meier Estimate , Lapatinib , Lysosomes/metabolism , Neoplasm Recurrence, Local/metabolism , Protein Kinase Inhibitors/pharmacology , Proteolysis
6.
Biochem Biophys Res Commun ; 438(4): 660-5, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23933250

ABSTRACT

The present study was conducted to determine whether fetuin-A, a dominant serum protein plays a role in chemo-attraction and chemo-invasion of carcinoma cells in vitro. Serum is normally used as positive chemotaxis control in Boyden chamber motility assays, prompting the need to identify the factor/s in serum that contributes the bulk of chemo-taxis and invasion. Serum has a plethora of chemotactic factors including stromal derived factor 1 also known as CXCL12. Using highly purified fetuin-A, we compared its chemo-attraction potential to culture medium containing 10% fetal bovine serum. We also investigated its ability to attract tumor cells through a bed of Matrigel (invasion assay). We demonstrated, using similar concentration range of fetuin-A found in blood, that it robustly supports both directed chemo-attraction and invasion of breast tumor cells. More importantly, we showed that at low concentrations (fetuin-A coated wells) itinteracts synergistically with CXCL12 to promote chemotaxis. The presence of plasminogen (PL) blunted the fetuin-A mediated chemotaxis. Taken together, the data suggest an in vivo chemotaxis/invasion role for fetuin-A.


Subject(s)
Breast Neoplasms/pathology , Breast/pathology , Chemokine CXCL12/metabolism , Chemotaxis , alpha-2-HS-Glycoprotein/metabolism , Animals , Biocompatible Materials/metabolism , Breast/metabolism , Breast Neoplasms/metabolism , Cattle , Cell Line, Tumor , Collagen/metabolism , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Combinations , Female , Humans , Laminin/metabolism , Neoplasm Invasiveness/pathology , Plasminogen/metabolism , Proteoglycans/metabolism
7.
In Vivo ; 23(5): 827-33, 2009.
Article in English | MEDLINE | ID: mdl-19779119

ABSTRACT

BACKGROUND: The purpose of this study was to investigate the immunological impact of combining neoadjuvant total androgen suppression (TAS) with radiotherapy (xRT) in the treatment of prostate cancer by monitoring blood cytokine levels. PATIENTS AND METHODS: Participants were stage I-II prostate cancer patients receiving xRT alone (n=18) or TAS+xRT (n=19) under the procedures outlined in RTOG protocols #94-08 and #94-13. Peripheral blood samples were collected immediately prior to TAS (xRT+TAS group), immediately prior to xRT, 24 hours after initiation of xRT, and weekly during xRT. Samples were monitored for the immunoregulatory cytokines interleukin (IL)-1beta, IL-6 and transforming growth factor (TGF)beta using ELISA procedures. RESULTS: Following initiation of xRT, both patient groups demonstrated an immediate elevation of the proinflammatory cytokines IL-1beta and IL-6 in their plasma. These cytokine levels appeared to peak after 1-2 weeks of xRT before returning toward pre xRT levels. In contrast, the profibrotic cytokine TGFbeta appeared to decrease immediately following initiation of xRT, but, subsequently, underwent two distinct waves of elevation, occurring at 1-2 weeks and 5-6 weeks into the xRT. Surprisingly, while the temporal pattern of plasma cytokine response was similar in both treatment groups, the magnitude of cytokine expression was noticeably different, appearing to be significantly affected by the addition of TAS. Indeed, administration of neoadjuvant TAS appeared to bring about a marked elevation of IL-1beta and IL-6 and a significant reduction in TGFbeta when compared to patients receiving xRT alone. CONCLUSION: The precise mechanisms underlying this TAS-related increase of the proinflammatory cytokines IL-1beta and IL-6 and decrease of the profibrotic cytokine TGFbeta remain unclear. However, previous reports have documented that androgens tend to be immunosuppressive in nature. It is conceivable, therefore, that administration of TAS shifts the ratio of proinflammatory and profibrotic cytokines toward a more immunostimulatory state.


Subject(s)
Adenocarcinoma/blood , Androgen Antagonists/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Cytokines/blood , Prostatic Neoplasms/blood , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Aged , Humans , Male , Middle Aged , Neoadjuvant Therapy , Neoplasm Staging , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Radiotherapy, Adjuvant
8.
Anticancer Res ; 28(6A): 3601-7, 2008.
Article in English | MEDLINE | ID: mdl-19189640

ABSTRACT

BACKGROUND: Interleukin-1alpha (IL-1) is known to radioprotect the gastrointestinal tract, but the mechanism by which this protection occurs remains unclear. These studies were undertaken to investigate whether the radioprotective potential of IL-1 may be linked to an ability to reduce apoptosis within the gastrointestinal crypts. MATERIALS AND METHODS: IL-1 was administered to C57Bl/6 mice 24 hours prior to receiving 8 Gy abdominal X-irradiation (xRT). At designated times, experimental mice were sacrificed, jejunal tissue removed, and paraffin-embedded sections analyzed for apoptosis indices (AI) and immunohistochemical determination of active caspase-3, -8 and -9. RESULTS: AI data demonstrated that 8 Gy irradiation resulted in a marked jejunal apoptotic response, but IL-1 pretreatment significantly attenuated this response. Concomitant with this attenuation, reduced levels of caspase-3 and 9, but not caspase-8, activation were observed, particularly within goblet cells. CONCLUSION: The results outlined herein suggest that radioprotection by IL-1 is mediated, at least in part, through a reduction in the apoptotic response which appears to involve down-regulation of the intrinsic apoptotic pathway.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , Interleukin-1alpha/pharmacology , Jejunum/drug effects , Jejunum/radiation effects , Radiation-Protective Agents/pharmacology , Animals , Caspases/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/radiation effects , Enzyme Activation/radiation effects , Female , In Vitro Techniques , Isoenzymes/metabolism , Jejunum/enzymology , Jejunum/pathology , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...