Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cancer Commun (Lond) ; 41(5): 414-431, 2021 05.
Article in English | MEDLINE | ID: mdl-33609419

ABSTRACT

BACKGROUND: Type II diabetes mellitus (DM2) is a significant risk factor for cancers, including breast cancer. However, a proper diabetic breast cancer mouse model is not well-established for treatment strategy design. Additionally, the precise diabetic signaling pathways that regulate cancer growth remain unresolved. In the present study, we established a suitable mouse model and demonstrated the pathogenic role of diabetes on breast cancer progression. METHODS: We successfully generated a transgenic mouse model of human epidermal growth factor receptor 2 positive (Her2+ or ERBB2) breast cancer with DM2 by crossing leptin receptor mutant (Leprdb/+ ) mice with MMTV-ErbB2/neu) mice. The mouse models were administrated with antidiabetic drugs to assess the impacts of controlling DM2 in affecting tumor growth. Magnetic resonance spectroscopic imaging was employed to analyze the tumor metabolism. RESULTS: Treatment with metformin/rosiglitazone in MMTV-ErbB2/Leprdb/db mouse model reduced serum insulin levels, prolonged overall survival, decreased cumulative tumor incidence, and inhibited tumor progression. Anti-insulin resistance medications also inhibited glycolytic metabolism in tumors in vivo as indicated by the reduced metabolic flux of hyperpolarized 13 C pyruvate-to-lactate reaction. The tumor cells from MMTV-ErbB2/Leprdb/db transgenic mice treated with metformin had reprogrammed metabolism by reducing levels of both oxygen consumption and lactate production. Metformin decreased the expression of Myc and pyruvate kinase isozyme 2 (PKM2), leading to metabolism reprogramming. Moreover, metformin attenuated the mTOR/AKT signaling pathway and altered adipokine profiles. CONCLUSIONS: MMTV-ErbB2/Leprdb/db mouse model was able to recapitulate diabetic HER2+ human breast cancer. Additionally, our results defined the signaling pathways deregulated in HER2+ breast cancer under diabetic condition, which can be intervened by anti-insulin resistance therapy.


Subject(s)
Breast Neoplasms , Diabetes Mellitus, Type 2 , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic , Signal Transduction
2.
Gastroenterol Rep (Oxf) ; 8(4): 261-276, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32843973

ABSTRACT

BACKGROUND: Diabetes mellitus type 2 (DM2) is a modifiable risk factor associated with pancreatic carcinogenesis and tumor progression on the basis of epidemiology studies, but the biological mechanisms are not completely understood. The purpose of this study is to demonstrate direct evidence for the mechanisms mediating these epidemiologic phenomena. Our hypothesis is that DM2 accelerates pancreatic cancer growth and that metformin treatment has a beneficial impact. METHODS: To determine the effect of glucose and insulin in pancreatic cancer proliferation, we used conditioned media to mimic DM2 conditions. Also, we studied the effect of anti-diabetic drugs, particularly metformin and rosiglitazone on pancreatic cancer growth. We established orthotopic/syngeneic (Lepr db/db ) mouse cancer models to evaluate the effect of diabetes on pancreatic tumor growth and aggressiveness. RESULTS: Our results showed that diabetes promotes pancreatic tumor growth. Furthermore, enhanced tumor growth and aggressiveness (e.g. epithelial-mesenchymal transition) can be explained by functional transcriptomic and metabolomic changes in the mice with diabetes, namely via activation of the AKT/mTOR pathway. Metformin treatment suppressed the diabetes-induced AKT/mTOR pathway activation and tumor growth. The metabolic profile determined by mass spectrum showed important changes of metabolites in the pancreatic cancer derived from diabetic mice treated with metformin. CONCLUSIONS: Diabetes mellitus type 2 has critical effects that promote pancreatic cancer progression via transcriptomic and metabolomic changes. Our animal models provide strong evidence for the causal relationship between diabetes and accelerated pancreatic cancers. This study sheds a new insight into the effects of metformin and its potential as part of therapeutic interventions for pancreatic cancer in diabetic patients.

3.
Nat Commun ; 9(1): 461, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29386624

ABSTRACT

Previously we have reported that metastatic melanoma cell lines and tumor specimens have reduced expression of ADAR1 and consequently are impaired in their ability to perform A-to-I microRNA (miRNA) editing. The effects of A-to-I miRNAs editing on melanoma growth and metastasis are yet to be determined. Here we report that miR-378a-3p is undergoing A-to-I editing only in the non-metastatic but not in metastatic melanoma cells. The function of the edited form is different from its wild-type counterpart. The edited form of miR-378a-3p preferentially binds to the 3'-UTR of the PARVA oncogene and inhibits its expression, thus preventing the progression of melanoma towards the malignant phenotype. Indeed, edited miR-378a-3p but not its WT form inhibits melanoma metastasis in vivo. These results further emphasize the role of RNA editing in melanoma progression.


Subject(s)
Adenosine/genetics , Gene Expression Regulation, Neoplastic , Inosine/genetics , Melanoma/pathology , MicroRNAs/genetics , Microfilament Proteins/genetics , RNA Editing , Skin Neoplasms/pathology , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Epigenesis, Genetic , Female , Humans , Melanoma/genetics , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Oncogenes , Skin Neoplasms/genetics
4.
Sci Rep ; 7: 40714, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28102366

ABSTRACT

The effects of AURKA overexpression associated with poor clinical outcomes have been attributed to increased cell cycle progression and the development of genomic instability with aneuploidy. We used RNA interference to examine the effects of AURKA overexpression in human bladder cancer cells. Knockdown had minimal effects on cell proliferation but blocked tumor cell invasion. Whole genome mRNA expression profiling identified nicotinamide N-methyltransferase (NNMT) as a downstream target that was repressed by AURKA. Chromatin immunoprecipitation and NNMT promoter luciferase assays revealed that AURKA's effects on NNMT were caused by PAX3-mediated transcriptional repression and overexpression of NNMT blocked tumor cell invasion in vitro. Overexpression of AURKA and activation of its downstream pathway was enriched in the basal subtype in primary human tumors and was associated with poor clinical outcomes. We also show that the FISH test for the AURKA gene copy number in urine yielded a specificity of 79.7% (95% confidence interval [CI] = 74.2% to 84.1%), and a sensitivity of 79.6% (95% CI = 74.2% to 84.1%) with an AUC of 0.901 (95% CI = 0.872 to 0.928; P < 0.001). These results implicate AURKA as an effective biomarker for bladder cancer detection as well as therapeutic target especially for its basal type.


Subject(s)
Aurora Kinase A/genetics , Biomarkers, Tumor , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/genetics , Aurora Kinase A/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Progression , Early Detection of Cancer , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Prognosis , Transcription, Genetic , Urinary Bladder Neoplasms/mortality
5.
Cancer Res ; 76(11): 3145-55, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27013197

ABSTRACT

Nuclear factor of activated T cell (NFAT1, NFATC2) is a transcription factor that binds and positively regulates IL2 expression during T-cell activation. NFAT1 has important roles in both innate and adaptive immune responses, but its involvement in cancer is not completely understood. We previously demonstrated that NFAT1 contributes to melanoma growth and metastasis by regulating the autotaxin gene (Enpp2). Here, we report a strong correlation between NFAT1 expression and metastatic potential in melanoma cell lines and tumor specimens. To elucidate the mechanisms underlying NFAT1 overexpression during melanoma progression, we conducted a microarray on a highly metastatic melanoma cell line in which NFAT1 expression was stably silenced. We identified and validated two downstream targets of NFAT1, IL8, and MMP3. Accordingly, NFAT1 depletion in metastatic melanoma cell lines was associated with reduced IL8 and MMP3 expression, whereas NFAT1 overexpression in a weakly metastatic cell line induced expression of these targets. Restoration of NFAT1 expression recovered IL8 and MMP3 expression levels back to baseline, indicating that both are direct targets of NFAT1. Moreover, in vivo studies demonstrated that NFAT1 and MMP3 promoted melanoma tumor growth and lung metastasis. Collectively, our findings assign a new role for NFAT1 in melanoma progression, underscoring the multifaceted functions that immunomodulatory factors may acquire in an unpredictable tumor microenvironment. Cancer Res; 76(11); 3145-55. ©2016 AACR.


Subject(s)
Interleukin-8/metabolism , Lung Neoplasms/secondary , Matrix Metalloproteinase 3/metabolism , Melanoma/pathology , NFATC Transcription Factors/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation , Female , Humans , Immunoenzyme Techniques , Interleukin-8/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Matrix Metalloproteinase 3/genetics , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , NFATC Transcription Factors/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cancer Res ; 75(19): 4131-42, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26282167

ABSTRACT

Adrenocortical carcinoma is a rare malignancy with poor prognosis and limited response to chemotherapy. Hepatocyte growth factor (HGF) and its receptor cMET augment cancer growth and resistance to chemotherapy, but their role in adrenocortical carcinoma has not been examined. In this study, we investigated the association between HGF/cMET expression and cancer hallmarks of adrenocortical carcinoma. Transcriptomic and immunohistochemical analyses indicated that increased HGF/cMET expression in human adrenocortical carcinoma samples was positively associated with cancer-related biologic processes, including proliferation and angiogenesis, and negatively correlated with apoptosis. Accordingly, treatment of adrenocortical carcinoma cells with exogenous HGF resulted in increased cell proliferation in vitro and in vivo while short hairpin RNA-mediated knockdown or pharmacologic inhibition of cMET suppressed cell proliferation and tumor growth. Moreover, exposure of cells to mitotane, cisplatin, or radiation rapidly induced pro-cMET expression and was associated with an enrichment of genes (e.g., CYP450 family) related to therapy resistance, further implicating cMET in the anticancer drug response. Together, these data suggest an important role for HGF/cMET signaling in adrenocortical carcinoma growth and resistance to commonly used treatments. Targeting cMET, alone or in combination with other drugs, could provide a breakthrough in the management of this aggressive cancer.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Carcinoma/metabolism , Hepatocyte Growth Factor/physiology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-met/physiology , Signal Transduction/physiology , Adenoma/drug therapy , Adenoma/metabolism , Adenoma/pathology , Adrenal Cortex Neoplasms/blood supply , Adrenal Cortex Neoplasms/drug therapy , Adrenal Cortex Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Anilides/pharmacology , Anilides/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma/blood supply , Carcinoma/drug therapy , Carcinoma/pathology , Cell Division , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/radiation effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Male , Mice , Mice, Nude , Middle Aged , Mitotane/pharmacology , Molecular Targeted Therapy , Neovascularization, Pathologic/physiopathology , Pyridines/pharmacology , Pyridines/therapeutic use , RNA Interference , RNA, Small Interfering/pharmacology , Transcriptome , Xenograft Model Antitumor Assays
7.
Nat Commun ; 6: 7530, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26179207

ABSTRACT

Extensive reprogramming of cellular energy metabolism is a hallmark of cancer. Despite its importance, the molecular mechanism controlling this tumour metabolic shift remains not fully understood. Here we show that 14-3-3σ regulates cancer metabolic reprogramming and protects cells from tumorigenic transformation. 14-3-3σ opposes tumour-promoting metabolic programmes by enhancing c-Myc poly-ubiquitination and subsequent degradation. 14-3-3σ demonstrates the suppressive impact on cancer glycolysis, glutaminolysis, mitochondrial biogenesis and other major metabolic processes of tumours. Importantly, 14-3-3σ expression levels predict overall and recurrence-free survival rates, tumour glucose uptake and metabolic gene expression in breast cancer patients. Thus, these results highlight that 14-3-3σ is an important regulator of tumour metabolism, and loss of 14-3-3σ expression is critical for cancer metabolic reprogramming. We anticipate that pharmacologically elevating the function of 14-3-3σ in tumours could be a promising direction for targeted anticancer metabolism therapy development in future.


Subject(s)
14-3-3 Proteins/genetics , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Energy Metabolism/genetics , Exoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-myc/metabolism , 14-3-3 Proteins/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease-Free Survival , Exoribonucleases/metabolism , Female , Gene Knockout Techniques , Glutamine/metabolism , Glycolysis/genetics , HCT116 Cells , Humans , Middle Aged , Organelle Biogenesis , Prognosis , Proteolysis , Ubiquitination/genetics , Young Adult
8.
Nat Cell Biol ; 17(3): 311-21, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25686251

ABSTRACT

Although recent studies have shown that adenosine-to-inosine (A-to-I) RNA editing occurs in microRNAs (miRNAs), its effects on tumour growth and metastasis are not well understood. We present evidence of CREB-mediated low expression of ADAR1 in metastatic melanoma cell lines and tumour specimens. Re-expression of ADAR1 resulted in the suppression of melanoma growth and metastasis in vivo. Consequently, we identified three miRNAs undergoing A-to-I editing in the weakly metastatic melanoma but not in strongly metastatic cell lines. One of these miRNAs, miR-455-5p, has two A-to-I RNA-editing sites. The biological function of edited miR-455-5p is different from that of the unedited form, as it recognizes a different set of genes. Indeed, wild-type miR-455-5p promotes melanoma metastasis through inhibition of the tumour suppressor gene CPEB1. Moreover, wild-type miR-455 enhances melanoma growth and metastasis in vivo, whereas the edited form inhibits these features. These results demonstrate a previously unrecognized role for RNA editing in melanoma progression.


Subject(s)
Adenosine/metabolism , Gene Expression Regulation, Neoplastic , Inosine/metabolism , Melanoma/genetics , RNA Editing , Skin Neoplasms/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Progression , Female , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , MicroRNAs , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Transplantation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
9.
Clin Cancer Res ; 21(9): 2127-37, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25595279

ABSTRACT

PURPOSE: Zoledronic acid is being increasingly recognized for its antitumor properties, but the underlying functions are not well understood. In this study, we hypothesized that zoledronic acid inhibits ovarian cancer angiogenesis preventing Rac1 activation. EXPERIMENTAL DESIGN: The biologic effects of zoledronic acid were examined using a series of in vitro [cell invasion, cytokine production, Rac1 activation, reverse-phase protein array, and in vivo (orthotopic mouse models)] experiments. RESULTS: There was significant inhibition of ovarian cancer (HeyA8-MDR and OVCAR-5) cell invasion as well as reduced production of proangiogenic cytokines in response to zoledronic acid treatment. Furthermore, zoledronic acid inactivated Rac1 and decreased the levels of Pak1/p38/matrix metalloproteinase-2 in ovarian cancer cells. In vivo, zoledronic acid reduced tumor growth, angiogenesis, and cell proliferation and inactivated Rac1 in both HeyA8-MDR and OVCAR-5 models. These in vivo antitumor effects were enhanced in both models when zoledronic acid was combined with nab-paclitaxel. CONCLUSIONS: Zoledronic acid has robust antitumor and antiangiogenic activity and merits further clinical development as ovarian cancer treatment.


Subject(s)
Neovascularization, Pathologic/pathology , Ovarian Neoplasms/pathology , Signal Transduction/physiology , Albumins/pharmacology , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Line, Tumor , Diphosphonates/pharmacology , Female , High-Throughput Screening Assays , Humans , Imidazoles/pharmacology , Immunohistochemistry , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Nude , Paclitaxel/pharmacology , Signal Transduction/drug effects , Transfection , Xenograft Model Antitumor Assays , Zoledronic Acid , p21-Activated Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism
10.
Nat Commun ; 5: 5384, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25395170

ABSTRACT

Cullin-RING ubiquitin ligases (CRLs) are critical in ubiquitinating Myc, while COP9 signalosome (CSN) controls neddylation of Cullin in CRL. The mechanistic link between Cullin neddylation and Myc ubiquitination/degradation is unclear. Here we show that Myc is a target of the CSN subunit 6 (CSN6)-Cullin signalling axis and that CSN6 is a positive regulator of Myc. CSN6 enhanced neddylation of Cullin-1 and facilitated autoubiquitination/degradation of Fbxw7, a component of CRL involved in Myc ubiquitination, thereby stabilizing Myc. Csn6 haplo-insufficiency decreased Cullin-1 neddylation but increased Fbxw7 stability to compromise Myc stability and activity in an Eµ-Myc mouse model, resulting in decelerated lymphomagenesis. We found that CSN6 overexpression, which leads to aberrant expression of Myc target genes, is frequent in human cancers. Together, these results define a mechanism for the regulation of Myc stability through the CSN-Cullin-Fbxw7 axis and provide insights into the correlation of CSN6 overexpression with Myc stabilization/activation during tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Carcinogenesis/genetics , Peptide Hydrolases/physiology , Proto-Oncogene Proteins c-myc/physiology , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , COP9 Signalosome Complex , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Lymphoma/metabolism , Lymphoma/physiopathology , Mice , Mice, Transgenic/genetics , Neoplasms, Experimental/genetics , Peptide Hydrolases/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , SKP Cullin F-Box Protein Ligases/physiology , Transcription, Genetic/physiology , Ubiquitination
11.
J Natl Cancer Inst ; 106(7)2014 Jul.
Article in English | MEDLINE | ID: mdl-24957076

ABSTRACT

BACKGROUND: Obesity increases the risk of cancer death among postmenopausal women with estrogen receptor-positive (ER+) breast cancer, but the direct evidence for the mechanisms is lacking. The purpose of this study is to demonstrate direct evidence for the mechanisms mediating this epidemiologic phenomenon. METHODS: We analyzed transcriptomic profiles of pretreatment biopsies from a prospective cohort of 137 ER+ breast cancer patients. We generated transgenic (MMTV-TGFα;A (y) /a) and orthotopic/syngeneic (A (y) /a) obese mouse models to investigate the effect of obesity on tumorigenesis and tumor progression and to determine biological mechanisms using whole-genome transcriptome microarrays and protein analyses. We used a coculture system to examine the impact of adipocytes/adipokines on breast cancer cell proliferation. All statistical tests were two-sided. RESULTS: Functional transcriptomic analysis of patients revealed the association of obesity with 59 biological functional changes (P < .05) linked to cancer hallmarks. Gene enrichment analysis revealed enrichment of AKT-target genes (P = .04) and epithelial-mesenchymal transition genes (P = .03) in patients. Our obese mouse models demonstrated activation of the AKT/mTOR pathway in obesity-accelerated mammary tumor growth (3.7- to 7.0-fold; P < .001; n = 6-7 mice per group). Metformin or everolimus can suppress obesity-induced secretion of adipokines and breast tumor formation and growth (0.5-fold, P = .04; 0.3-fold, P < .001, respectively; n = 6-8 mice per group). The coculture model revealed that adipocyte-secreted adipokines (eg, TIMP-1) regulate adipocyte-induced breast cancer cell proliferation and invasion. Metformin suppress adipocyte-induced cell proliferation and adipocyte-secreted adipokines in vitro. CONCLUSIONS: Adipokine secretion and AKT/mTOR activation play important roles in obesity-accelerated breast cancer aggressiveness in addition to hyperinsulinemia, estrogen signaling, and inflammation. Metformin and everolimus have potential for therapeutic interventions of ER+ breast cancer patients with obesity.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Breast Neoplasms/etiology , Breast Neoplasms/metabolism , Metformin/pharmacology , Obesity/complications , Obesity/metabolism , Receptors, Estrogen/metabolism , Sirolimus/analogs & derivatives , Transcriptome , Adipocytes , Adipokines/metabolism , Aged , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Everolimus , Female , Humans , Kaplan-Meier Estimate , Mice , Mice, Transgenic , Middle Aged , Obesity/epidemiology , Obesity/genetics , Postmenopause , Prospective Studies , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
12.
J Immigr Minor Health ; 16(5): 865-73, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23828626

ABSTRACT

This study assessed the association of cardiometabolic risk factors with systemic inflammation, insulin resistance, and adypocytokines in a Hispanic adolescent subgroup. A clinic-based sample of 101 Puerto Rican adolescents, 48 of whom were overweight or obese based on body mass index percentiles for age and sex, was recruited during 2010. Data were collected through interviews, blood pressure and anthropometric measurements, and blood drawing. Overall prevalence of the metabolic syndrome was 16.8 % and increased to 37.5 % among overweight/obese youth. The overweight/obese group exhibited significantly (p < 0.05) higher values for abdominal obesity measures, systolic blood pressure, triglycerides, insulin resistance, C peptide, high-sensitivity C reactive protein, fibrinogen, leptin, and IL-6 and lower levels of high density lipoprotein cholesterol, adiponectin, and IGF-1. Total adiponectin significantly correlated with most cardiovascular risk factors independent of sex, Tanner stage, and adiposity. Altered cardiometabolic and adipocytokine profiles were present in this Hispanic subgroup, reinforcing the need to strengthen strategies addressing childhood obesity.


Subject(s)
Adipokines/blood , Hispanic or Latino/statistics & numerical data , Inflammation/epidemiology , Metabolic Syndrome/ethnology , Adolescent , Biomarkers/blood , Blood Pressure , Cross-Sectional Studies , Female , Hispanic or Latino/ethnology , Humans , Inflammation/blood , Inflammation/ethnology , Male , Metabolic Syndrome/epidemiology , Obesity/blood , Obesity/epidemiology , Obesity/ethnology , Overweight/blood , Overweight/epidemiology , Overweight/ethnology , Prevalence , Risk Factors
13.
Cell Cycle ; 11(21): 4059-68, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23095493

ABSTRACT

FBXW7, a component of E3 ubiquitin ligase, plays an important role in mitotic checkpoint, but its role remains unclear. Aurora B is a mitotic checkpoint kinase that plays a pivotal role in mitosis by ensuring correct chromosome segregation and normal progression through mitosis. Whether Aurora B and FBXW7 are coordinately regulated during mitosis is not known. Here, we show that FBXW7 is a negative regulator for Aurora B. Ectopic expression of FBXW7 can suppress the expression of Aurora B. Accordingly, FBXW7 deficiency leads to Aurora B elevation. Mechanistic studies show that all FBXW7 isoforms are negative regulators of Aurora B expression through ubiquitination-mediated protein degradation. Aurora B interacts with R465 and R505 residues of WD 40 domain of FBXW7. Significantly, inverse correlation between FBXW7 and Aurora B elevation is translated into the deregulation of mitosis. FBWX7 expression mitigates Aurora B-mediated cell growth and mitotic deregulation. In addition, FBXW7 reduces the percentage of multinucleated cells caused by Aurora B overexpression. These data suggest that FBXW7 is an important negative regulator of Aurora B, and that the loss or mutation of FBXW7 as seen in many types of cancer could lead to an abnormal elevation of Aurora B and result in deregulated mitosis, which accelerates cancer cell growth.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Aurora Kinase B , Aurora Kinases , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , F-Box Proteins/chemistry , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , HCT116 Cells , HEK293 Cells , Humans , Leupeptins/pharmacology , Mitosis/drug effects , Protease Inhibitors/pharmacology , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Stability , Protein Structure, Tertiary , Transfection , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination
14.
Proc Natl Acad Sci U S A ; 109(24): E1513-22, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22611192

ABSTRACT

Aurora B is a mitotic checkpoint kinase that plays a pivotal role in the cell cycle, ensuring correct chromosome segregation and normal progression through mitosis. Aurora B is overexpressed in many types of human cancers, which has made it an attractive target for cancer therapies. Tumor suppressor p53 is a genome guardian and important negative regulator of the cell cycle. Whether Aurora B and p53 are coordinately regulated during the cell cycle is not known. We report that Aurora B directly interacts with p53 at different subcellular localizations and during different phases of the cell cycle (for instance, at the nucleus in interphase and the centromeres in prometaphase of mitosis). We show that Aurora B phosphorylates p53 at S183, T211, and S215 to accelerate the degradation of p53 through the polyubiquitination-proteasome pathway, thus functionally suppressing the expression of p53 target genes involved in cell cycle inhibition and apoptosis (e.g., p21 and PUMA). Pharmacologic inhibition of Aurora B in cancer cells with WT p53 increased p53 protein level and expression of p53 target genes to inhibit tumor growth. Together, these results define a mechanism of p53 inactivation during the cell cycle and imply that oncogenic hyperactivation or overexpression of Aurora B may compromise the tumor suppressor function of p53. We have elucidated the antineoplastic mechanism for Aurora B kinase inhibitors in cancer cells with WT p53.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Aurora Kinase B , Aurora Kinases , Humans , Interphase , Mitosis , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proteolysis , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism , Transcription, Genetic , Ubiquitination
15.
Diabetes Metab Syndr Obes ; 4: 61-5, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21448323

ABSTRACT

Exenatide is an incretin mimetic that is recently available in the US for the treatment of diabetes. There is a paucity of information on the effects of exenatide in glucocorticoid (GC)-induced diabetes. Although the effect of continuous intravenous infusion of exenatide on GC-induced glucose intolerance has been investigated before in healthy human males receiving oral prednisolone, we investigated the efficacy of a single subcutaneous dose of exenatide (3 µg/kg) in lowering blood glucose in GC-induced glucose intolerance in C57BL/6 mice. In a longitudinal experiment, the area under the curve (AUC) of oral glucose tolerance tests (OGTT) significantly increased after dexamethasone (P = 0.004), which was subsequently decreased by exenatide (P < 0.001). A cross-sectional experiment showed that exenatide improved glucose tolerance compared with placebo in a mouse model of dexamethasone-induced glucose intolerance. AUC of OGTT in the exenatide group were significantly (P < 0.001) lower than in the placebo group. Insulin tolerance tests (ITT) demonstrated that exenatide decreased the ability of the mice to tolerate insulin compared with placebo. The AUC of ITT in the exenatide group were also significantly (P = 0.006) lower than in the placebo group. In conclusion, a single dose of exenatide was able to decrease glucose intolerance and insulin resistance in these placebo-controlled experiments. Future clinical trials are justified to investigate the role of exenatide in the treatment of GC-induced glucose intolerance/diabetes.

16.
J Cell Mol Med ; 15(4): 825-36, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20455996

ABSTRACT

Despite investigations into mechanisms linking type 2 diabetes and cancer, there is a gap in knowledge about pharmacotherapy for diabetes in cancer patients. Epidemiological studies have shown that diabetic cancer patients on different antidiabetic treatments have different survival. The clinically relevant question is whether some antidiabetic pharmacotherapeutic agents promote cancer whereas others inhibit cancer progression. We investigated the hypothesis that various antidiabetic drugs had differential direct impact on cancer cells using four human cell lines (pancreatic cancer: MiaPaCa2, Panc-1; breast cancer: MCF7, HER18). We found that insulin and glucose promoted cancer cell proliferation and contributed to chemoresistance. Metformin and rosiglitazone suppressed cancer cell growth and induced apoptosis. Both drugs affected signalling in the protein kinases B (AKT)/mammalian target of rapamycin pathway; metformin activated adenosine monophosphate (AMP)-activated protein kinase whereas rosiglitazone increased chromosome ten level. Although high insulin and glucose concentrations promoted chemoresistance, the combination of metformin or rosiglitazone with gemcitabine or doxorubicin, resulted in an additional decrease in live cancer cells and increase in apoptosis. In contrast, exenatide did not have direct effect on cancer cells. In conclusion, different types of antidiabetic pharmacotherapy had a differential direct impact on cancer cells. This study provides experimental evidence to support further investigation of metformin and rosiglitazone as first-line therapies for type 2 diabetes in cancer patients.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Hypoglycemic Agents/therapeutic use , Neoplasms/pathology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Glucose/pharmacology , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Metformin/pharmacology , Pancreatic Neoplasms/pathology , Rosiglitazone , Thiazolidinediones/pharmacology , Gemcitabine
17.
Mol Cancer ; 9: 243, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20843328

ABSTRACT

Mammalian constitutive photomorphogenic 1 (COP1) is a p53 E3 ubiquitin ligase involved in regulating p53 protein level. In plants, the dynamic cytoplasm/nucleus distribution of COP1 is important for its function in terms of catalyzing the degradation of target proteins. In mammalian cells, the biological consequence of cytoplasmic distribution of COP1 is not well characterized. Here, we show that DNA damage leads to the redistribution of COP1 to the cytoplasm and that 14-3-3σ, a p53 target gene product, controls COP1 subcellular localization. Investigation of the underlying mechanism suggests that COP1 S387 phosphorylation is required for COP1 to bind 14-3-3σ. Significantly, upon DNA damage, 14-3-3σ binds to phosphorylated COP1 at S387, resulting in COP1's accumulation in the cytoplasm. Cytoplasmic COP1 localization leads to its enhanced ubiquitination. We also show that N-terminal 14-3-3σ interacts with COP1 and promotes COP1 nuclear export through its NES sequence. Further, we show that COP1 is important in causing p53 nuclear exclusion. Finally, we demonstrate that 14-3-3σ targets COP1 for nuclear export, thereby preventing COP1-mediated p53 nuclear export. Together, these results define a novel, detailed mechanism for the subcellular localization and regulation of COP1 after DNA damage and provide a mechanistic explanation for the notion that 14-3-3σ's impact on the inhibition of p53 E3 ligases is an important step for p53 stabilization after DNA damage.


Subject(s)
14-3-3 Proteins/metabolism , DNA Damage/physiology , Ubiquitin-Protein Ligases/metabolism , 14-3-3 Proteins/genetics , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/physiology , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Damage/genetics , Humans , Immunoblotting , Immunoprecipitation , Protein Binding , Ubiquitin-Protein Ligases/genetics , Ubiquitination
18.
Mol Cancer ; 9: 42, 2010 Feb 22.
Article in English | MEDLINE | ID: mdl-20175926

ABSTRACT

BACKGROUND: Aurora B kinase is an important mitotic kinase involved in chromosome segregation and cytokinesis. It is overexpressed in many cancers and thus may be an important molecular target for chemotherapy. AZD1152 is the prodrug for AZD1152-HQPA, which is a selective inhibitor of Aurora B kinase activity. Preclinical antineoplastic activity of AZD1152 against acute myelogenous leukemia, multiple myeloma and colorectal cancer has been reported. However, this compound has not been evaluated in breast cancer, the second leading cause of cancer deaths among women. RESULTS: The antineoplastic activity of AZD1152-HQPA in six human breast cancer cell lines, three of which overexpress HER2, is demonstrated. AZD1152-HQPA specifically inhibited Aurora B kinase activity in breast cancer cells, thereby causing mitotic catastrophe, polyploidy and apoptosis, which in turn led to apoptotic death. AZD1152 administration efficiently suppressed the tumor growth in a breast cancer cell xenograft model. In addition, AZD1152 also inhibited pulmonary metastatic nodule formation in a metastatic breast cancer model. Notably, it was also found that the protein level of Aurora B kinase declined after inhibition of Aurora B kinase activity by AZD1152-HQPA in a time- and dose-dependent manner. Investigation of the underlying mechanism suggested that AZD1152-HQPA accelerated protein turnover of Aurora B via enhancing its ubiquitination. CONCLUSIONS: It was shown that AZD1152 is an effective antineoplastic agent for breast cancer, and our results define a novel mechanism for posttranscriptional regulation of Aurora B after AZD1152 treatment and provide insight into dosing regimen design for this kinase inhibitor in metastatic breast cancer treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Organophosphates/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/therapeutic use , Aneuploidy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aurora Kinase B , Aurora Kinases , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , G2 Phase/drug effects , Humans , Mice , Mitosis/drug effects , Neoplasm Metastasis , Organophosphates/pharmacology , Polyploidy , Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/metabolism , Quinazolines/pharmacology , Tumor Stem Cell Assay , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...