Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 17(1): 20-29, 2019 01.
Article in English | MEDLINE | ID: mdl-30131447

ABSTRACT

The regulation of colorectal cancer cell survival pathways remains to be elucidated. Previously, it was demonstrated that endothelial cells (EC) from the liver (liver parenchymal ECs or LPEC), the most common site of colorectal cancer metastases, secrete soluble factors in the conditioned medium (CM) that, in turn, increase the cancer stem cell phenotype in colorectal cancer cells. However, the paracrine effects of LPECs on other colorectal cancer cellular functions have not been investigated. Here, results showed that CM from LPECs increased cell growth and chemoresistance by activating AKT in colorectal cancer cells in vitro. Using an unbiased receptor tyrosine kinase array, it was determined that human epidermal growth factor receptor 3 (ERBB3/HER3) was activated by CM from LPECs, and it mediated AKT activation, cell growth, and chemoresistance in colorectal cancer cells. Inhibition of HER3, either by an inhibitor AZD8931 or an antibody MM-121, blocked LPEC-induced HER3-AKT activation and cell survival in colorectal cancer cells. In addition, CM from LPECs increased in vivo tumor growth in a xenograft mouse model. Furthermore, inhibiting HER3 with AZD8931 significantly blocked tumor growth induced by EC CM. These results demonstrated a paracrine role of liver ECs in promoting cell growth and chemoresistance via activating HER3-AKT in colorectal cancer cells. IMPLICATIONS: This study suggested a potential of treating patients with metastatic colorectal cancer with HER3 antibodies/inhibitors that are currently being assessed in clinical trials for various cancer types.


Subject(s)
Cell Communication/physiology , Colorectal Neoplasms/metabolism , Endothelial Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-3/metabolism , Animals , Cell Line, Tumor , Cell Survival/physiology , Colorectal Neoplasms/pathology , Endothelial Cells/pathology , Enzyme Activation , HCT116 Cells , HT29 Cells , Heterografts , Humans , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-3/antagonists & inhibitors , Signal Transduction
2.
Clin Cancer Res ; 24(14): 3447-3455, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29643062

ABSTRACT

Purpose: The successful translation of laboratory research into effective therapies is dependent upon the validity of peer-reviewed publications. However, several publications in recent years suggested that published scientific findings could be reproduced only 11% to 45% of the time. Multiple surveys attempted to elucidate the fundamental causes of data irreproducibility and underscored potential solutions, more robust experimental designs, better statistics, and better mentorship. However, no prior survey has addressed the role of the review and publication process on honest reporting.Experimental Design: We developed an anonymous online survey intended for trainees involved in bench research. The survey included questions related to mentoring/career development, research practice, integrity, and transparency, and how the pressure to publish and the publication process itself influence their reporting practices.Results: Responses to questions related to mentoring and training practices were largely positive, although an average of approximately 25% did not seem to receive optimal mentoring. A total of 39.2% revealed having been pressured by a principle investigator or collaborator to produce "positive" data. About 62.8% admitted that the pressure to publish influences the way they report data. The majority of respondents did not believe that extensive revisions significantly improved the manuscript while adding to the cost and time invested.Conclusions: This survey indicates that trainees believe that the pressure to publish affects honest reporting, mostly emanating from our system of rewards and advancement. The publication process itself affects faculty and trainees and appears to influence a shift in their ethics from honest reporting ("negative data") to selective reporting, data falsification, or even fabrication. Clin Cancer Res; 24(14); 3447-55. ©2018 AACR.


Subject(s)
Ethics, Research , Publications , Reproducibility of Results , Research/statistics & numerical data , Research/standards , Humans , Internet , Professional Practice/ethics , Professional Practice/standards , Publications/statistics & numerical data , Research Personnel , Students , Surveys and Questionnaires
3.
PLoS One ; 13(1): e0190070, 2018.
Article in English | MEDLINE | ID: mdl-29293549

ABSTRACT

BACKGROUND: There is conflicting data on the role of macrophages in colorectal cancer (CRC); some studies have shown that macrophages can exert an anti-tumor effect whereas others show that macrophages are tumor promoting. We sought to determine the role of conditioned medium (CM) from macrophages, in particular classically activated macrophages, on the development of the CSC phenotype in CRC cells, which is believed to mediate tumor growth and chemoresistance. METHODS: Murine (CT26) and human (HCP-1) CRC cell lines were treated with CM from lipopolysaccharide (LPS)-activated murine macrophages. The CSC population was assessed using the sphere-forming assay and aldehyde dehydrogenase assay. Chemoresistance studies were performed using the MTT assay. CSC transcription factors and SHH protein were analyzed by Western blotting. RESULTS: The results showed that LPS-activated macrophage CM induced the CSC phenotype in CRC cells. Further studies showed that the CSC phenotype was mediated by the sonic hedgehog (SHH)-Gli signaling pathway, which is known to drive self-renewal; these effects were blocked by depletion of SHH in macrophage CM. In addition, LPS-activated macrophage CM enhanced chemoresistance. CONCLUSIONS: LPS-activated macrophages play an active role in promoting the CSC phenotype through activation of the SHH-Gli signaling pathway in CRC cells.


Subject(s)
Colorectal Neoplasms/pathology , Hedgehog Proteins/metabolism , Macrophages/metabolism , Neoplastic Stem Cells/pathology , Animals , Colorectal Neoplasms/metabolism , Culture Media, Conditioned , Humans , Mice , Neoplastic Stem Cells/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
4.
Cancer Prev Res (Phila) ; 10(7): 398-409, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28483840

ABSTRACT

Chronic infection and associated inflammation have long been suspected to promote human carcinogenesis. Recently, certain gut bacteria, including some in the Fusobacterium genus, have been implicated in playing a role in human colorectal cancer development. However, the Fusobacterium species and subspecies involved and their oncogenic mechanisms remain to be determined. We sought to identify the specific Fusobacterium spp. and ssp. in clinical colorectal cancer specimens by targeted sequencing of Fusobacterium 16S ribosomal RNA gene. Five Fusobacterium spp. were identified in clinical colorectal cancer specimens. Additional analyses confirmed that Fusobacterium nucleatum ssp. animalis was the most prevalent F. nucleatum subspecies in human colorectal cancers. We also assessed inflammatory cytokines in colorectal cancer specimens using immunoassays and found that expression of the cytokines IL17A and TNFα was markedly increased but IL21 decreased in the colorectal tumors. Furthermore, the chemokine (C-C motif) ligand 20 was differentially expressed in colorectal tumors at all stages. In in vitro co-culture assays, F. nucleatum ssp. animalis induced CCL20 protein expression in colorectal cancer cells and monocytes. It also stimulated the monocyte/macrophage activation and migration. Our observations suggested that infection with F. nucleatum ssp. animalis in colorectal tissue could induce inflammatory response and promote colorectal cancer development. Further studies are warranted to determine if F. nucleatum ssp. animalis could be a novel target for colorectal cancer prevention and treatment. Cancer Prev Res; 10(7); 398-409. ©2017 AACR.


Subject(s)
Adenocarcinoma/immunology , Carcinogenesis/immunology , Chemokine CCL20/metabolism , Colorectal Neoplasms/immunology , Fusobacterium Infections/immunology , Fusobacterium nucleatum/immunology , Monocytes/immunology , Adenocarcinoma/microbiology , Adenocarcinoma/pathology , Adenocarcinoma/prevention & control , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Movement/immunology , Coculture Techniques , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Disease Progression , Female , Fusobacterium Infections/microbiology , Fusobacterium Infections/pathology , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/isolation & purification , Humans , Interleukin-17/metabolism , Interleukins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Macrophage Activation/immunology , Male , Middle Aged , Monocytes/metabolism , Neoplasm Staging , Primary Cell Culture , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Th17 Cells/immunology , Th17 Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Mol Oncol ; 11(8): 1023-1034, 2017 08.
Article in English | MEDLINE | ID: mdl-28453235

ABSTRACT

In colorectal cancer (CRC), cancer stem cells (CSCs) have been hypothesized to mediate cell survival and chemoresistance. Previous studies from our laboratory described a role for liver parenchymal endothelial cells (LPECs) in mediating the CSC phenotype in CRC cells in a paracrine/angiocrine fashion. The objectives of this study were to determine whether endothelial cells (ECs) from different organs can induce the CSC phenotype in CRC cells and to elucidate the signaling pathways involved. We treated a newly developed CRC cell line (HCP-1) and established CRC cell lines (HT29 and SW480) with conditioned medium (CM) from primary ECs isolated from nonmalignant liver, lung, colon mucosa, and kidney. Our results showed that CM from ECs from all organs increased the number of CSCs, as determined by sphere formation, and protein levels of NANOG and OCT4 in CRC cells. With the focus of further elucidating the role of the liver vascular network in mediating the CSC phenotype, we demonstrated that CM from LPECs increased resistance to 5-fluorouracil in CRC cells. Moreover, we showed that LPEC CM specifically induced NANOGP8 expression in CRC cells by specific enzyme digestion and a luciferase reporter assay using a vector containing the NANOGP8 promoter. Lastly, we found that LPEC CM-induced NANOGP8 expression and sphere formation were mediated by AKT activation. Our studies demonstrated a paracrine role for ECs in regulating the CSC phenotype and chemoresistance in CRC cells by AKT-mediated induction of NANOGP8. These studies suggest a more specific approach to target CSCs by blocking the expression of NANOGP8 in cancer cells.


Subject(s)
Colorectal Neoplasms/metabolism , Endothelial Cells/metabolism , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/metabolism , Paracrine Communication , Signal Transduction , Cell Line, Tumor , Colorectal Neoplasms/pathology , Endothelial Cells/pathology , Humans , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-akt/metabolism
6.
Stem Cells Transl Med ; 5(3): 331-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26744411

ABSTRACT

Evidence is accumulating for the role of cancer stem cells (CSCs) in mediating chemoresistance in patients with metastatic colorectal cancer (mCRC). A disintegrin and metalloproteinase domain 17 (ADAM17; also known as tumor necrosis factor-α-converting enzyme [TACE]) was shown to be overexpressed and to mediate cell proliferation and chemoresistance in CRC cells. However, its role in mediating the CSC phenotype in CRC has not been well-characterized. The objective of the present study was to determine whether ADAM17 regulates the CSC phenotype in CRC and to elucidate the downstream signaling mechanism that mediates cancer stemness. We treated established CRC cell lines and a newly established human CRC cell line HCP-1 with ADAM17-specific small interfering RNA (siRNA) or the synthetic peptide inhibitor TAPI-2. The effects of ADAM17 inhibition on the CSC phenotype and chemosensitivity to 5-fluorouracil (5-FU) in CRC cells were examined. siRNA knockdown and TAPI-2 decreased the protein levels of cleaved Notch1 (Notch1 intracellular domain) and HES-1 in CRC cells. A decrease in the CSC phenotype was determined by sphere formation and ALDEFLUOR assays. Moreover, TAPI-2 sensitized CRC cells to 5-FU by decreasing cell viability and the median lethal dose of 5-FU and increasing apoptosis. We also showed the cleavage and release of soluble Jagged-1 and -2 by ADAM17 in CRC cells. Our studies have elucidated a role of ADAM17 in regulating the CSC phenotype and chemoresistance in CRC cells. The use of drugs that inhibit ADAM17 activity might increase the therapeutic benefit to patients with mCRC and, potentially, those with other solid malignancies.


Subject(s)
ADAM Proteins/genetics , Colorectal Neoplasms/genetics , Neoplastic Stem Cells/drug effects , Receptor, Notch1/genetics , ADAM Proteins/biosynthesis , ADAM17 Protein , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Fluorouracil/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxamic Acids/administration & dosage , Neoplastic Stem Cells/pathology , Receptor, Notch1/biosynthesis , Signal Transduction/drug effects
7.
Mol Cancer Res ; 13(9): 1274-82, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26041938

ABSTRACT

UNLABELLED: A large number of pseudogenes have been found to be transcribed in human cancers. However, only a few pseudogenes are functionally characterized. Here, we identified a transcribed pseudogene of VEGFR1, or fms-related tyrosine kinase 1 (FLT1), in human colorectal cancer cells. Interestingly, this pseudogene (designated as FLT1P1) was found to be transcribed bidirectionally and functionally modulated cognate VEGFR1 protein expression in the cells. Mechanistically, expression of FLT1P1 antisense transcript not only inhibited the VEGFR1 expression, but also inhibited non-cognate VEGF-A expression through interaction with miR-520a. Perturbation of FLT1P1 expression by RNA interference (RNAi) markedly inhibited tumor cell proliferation and xenograft tumor growth. This study identifies FLT1P1 antisense as a critical regulator of VEGFR1 and VEGF-A expression in colorectal cancer cells, and highlights its role in regulation of the pathogenesis of colorectal cancer. IMPLICATIONS: The VEGFR1 pseudogene, FLT1P1, is a novel and functional regulator of VEGF signaling and its targeting could be an alternative strategy to modulate its cognate/target gene expression and downstream activity in cancer.


Subject(s)
Colorectal Neoplasms/metabolism , Pseudogenes , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Female , Heterografts , Humans , Mice, Nude , MicroRNAs/metabolism , RNA, Antisense/metabolism , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Breast Cancer Res Treat ; 151(3): 501-13, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25971596

ABSTRACT

Resistance to HER2-targeted therapies remains a major obstacle in the treatment of HER2-overexpressing breast cancer. CD44, a putative breast cancer stem cell (CSC) marker, is overexpressed in trastuzumab-resistant breast cancer cells. While CSC-related genes may play a role in the development of trastuzumab resistance, conflicting results have been published about CSC response to trastuzumab. We hypothesized that CD44 contributes to trastuzumab resistance independently of its role as a CSC marker. We used trastuzumab-sensitive breast cancer cell lines and their trastuzumab-resistant isogenic counterparts to evaluate the role of CD44 in response to trastuzumab. miRNA and mRNA expression were analyzed using microarray chips. A gene set enrichment analysis was created and matched with response to trastuzumab in cells and patient samples. The proportions of CSC in trastuzumab-resistant cells were similar to or lower than in the trastuzumab-sensitive cells. However, CD44 expression levels were significantly higher in both trastuzumab-resistant cell lines and its knockdown led to an increased response to trastuzumab. The invasiveness and anchorage-independent growth of trastuzumab-resistant cells were higher and blocked by downregulation of CD44. Results also showed that CD44-related resistance to trastuzumab is regulated by miRNAs. We identified a CD44-related gene expression profile that correlated with response to trastuzumab in cell lines and breast cancer patients. CD44 mediates trastuzumab resistance in HER2-positive breast cancer cells independently of its role as a CSC marker and that this role of CD44 is partly regulated by miRNA.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Gene Expression , Hyaluronan Receptors/genetics , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , MicroRNAs/genetics , RNA, Small Interfering/genetics
9.
Mol Oncol ; 9(3): 586-600, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25435280

ABSTRACT

Resistance to HER2-targeted therapies remains a major obstacle in the treatment of HER2-overexpressing breast cancer. Understanding the molecular pathways that contribute to the development of drug resistance is needed to improve the clinical utility of novel agents, and to predict the success of targeted personalized therapy based on tumor-specific mutations. Little is known about the clinical significance of HER family mutations in breast cancer. Because mutations within HER1/EGFR are predictive of response to tyrosine kinase inhibitors (TKI) in lung cancer, we investigated whether mutations in HER family kinase domains are predictive of response to targeted therapy in HER2-overexpressing breast cancer. We sequenced the HER family kinase domains from 76 HER2-overexpressing invasive carcinomas and identified 12 missense variants. Patients whose tumors carried any of these mutations did not respond to HER2 directed therapy in the metastatic setting. We developed mutant cell lines and used structural analyses to determine whether changes in protein conformation could explain the lack of response to therapy. We also functionally studied all HER2 mutants and showed that they conferred an aggressive phenotype and altered effects of the TKI lapatinib. Our data demonstrate that mutations in the finely tuned HER kinase domains play a critical function in breast cancer progression and may serve as prognostic and predictive markers.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Disease Progression , ErbB Receptors/chemistry , ErbB Receptors/genetics , Mutation/genetics , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Computational Biology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Lapatinib , Mice, Nude , Mutant Proteins/chemistry , Phenotype , Phosphorylation/drug effects , Prognosis , Protein Structure, Tertiary , Quinazolines/pharmacology , Treatment Outcome
10.
Biochem Biophys Res Commun ; 413(1): 46-52, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21867682

ABSTRACT

The Akt kinase is a critical effector in growth factor signaling. Activation of Akt driven by the growth factor dependent PI3K (phosphatidylinositol-3-OH kinase) is coupled to the plasma membrane translocation and phosphorylation of Akt on two sites by PDK1 (phosphoinositide-dependent protein kinase-1) on Thr-308 and by mTORC2 (mammalian Target of Rapamycin Complex 2) on Ser-473. In our study we examined the sub-cellular localization of mTORC2 and identified that this kinase complex predominantly resides on endoplasmic reticulum (ER). Our immunostaining analysis did not show a substantial co-localization of the mTORC2 component rictor with Golgi, lysosome, clathrin-coated vesicles, early endosomes, or plasma membrane but indicated a strong co-localization of rictor with ribosomal protein S6 and ER marker. Our biochemical study also identified the mTORC2 components rictor, SIN1, and mTOR as the highly abundant proteins in the ER fraction, whereas only small amount of these proteins are detected in the plasma membrane and cytosolic fractions. We found that growth factor signaling does not alter the ER localization of mTORC2 and also does not induce its translocation to the plasma membrane. Based on our study we suggest that the mTORC2-dependent phosphorylation of Akt on Ser-473 takes place on the surface of ER.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Endoplasmic Reticulum/enzymology , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/analysis , Carrier Proteins/analysis , Cell Line , Cell Membrane/enzymology , Clathrin-Coated Vesicles/enzymology , Endoplasmic Reticulum/chemistry , Endosomes/enzymology , Golgi Apparatus/enzymology , Humans , Immunohistochemistry , Lysosomes/enzymology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Staining and Labeling , TOR Serine-Threonine Kinases/analysis
11.
Breast Cancer Res Treat ; 130(3): 1029-36, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21830016

ABSTRACT

CD44 is an adhesion molecule involved in tumor cell invasion and metastasis. The function of CD44 in breast cancer is not understood completely, or is its role as a predictive or prognostic factor. In this study, we tested for the hypothesis that the concentration of soluble CD44 (sCD44) in serum is correlated with clinicopathological factors, especially HER2, and survival in patients with breast cancer. We retrospectively identified 110 patients with breast cancer who had been treated at The University of Texas MD Anderson Cancer Center (MDACC) from September 2001 to May 2004. Sera were collected before definitive surgery in patients with stage I or II breast cancer, before initiation of neoadjuvant chemotherapy (if indicated) for patients with stage I-III breast cancer, and before initiation of systemic therapy in patients with stage IV breast cancer. sCD44 levels were determined using an enzyme-linked immunosorbent assay. The median age at diagnosis was 51 years (range, 28.6-87.1 years). sCD44 concentration was correlated with tumor stage (P = 0.0308). sCD44 serum concentration did not predict pathological response in patients treated with neoadjuvant chemotherapy. Among patients with distant metastases, sCD44 levels were significantly higher in patients with liver involvement than in patients with metastases at other sites. The overall survival rate did not differ between patients with high sCD44 concentration and patients with low sCD44 concentration. However, sCD44 concentration was a significant predictor of overall survival for patients with HER2-positive breast cancer, while no difference in overall survival rates was observed in patients with HER2-negative breast cancer. To the best of our knowledge, this is the first study to show an association between circulating sCD44 levels and survival in HER2-positive breast cancer patients. Our results suggest a role for sCD44 as a prognostic marker. Furthermore, sCD44 level may offer a new clinical therapeutic target in HER2-positive breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Hyaluronan Receptors/blood , Receptor, ErbB-2/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cohort Studies , Female , Humans , Middle Aged , Neoadjuvant Therapy , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL