Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 314
Filter
1.
Oncogene ; 36(46): 6462-6471, 2017 11 16.
Article in English | MEDLINE | ID: mdl-28759039

ABSTRACT

Obesity is associated with an increase in cancer-specific mortality in women with breast cancer. Elevated cholesterol, particularly low-density lipoprotein cholesterol (LDL-C), is frequently seen in obese women. Here, we aimed to determine the importance of elevated circulating LDL, and LDL receptor (LDLR) expression in tumor cells, on the growth of breast cancer using mouse models of hyperlipidemia. We describe two novel immunodeficient mouse models of hyperlipidemia (Rag1-/-/LDLR-/- and Rag1-/-/ApoE (apolipoprotein E)-/- mice) in addition to established immunocompetent LDLR-/- and ApoE-/- mice. The mice were used to study the effects of elevated LDL-C in human triple-negative (MDA-MB-231) and mouse Her2/Neu-overexpressing (MCNeuA) breast cancers. Tumors derived from MCNeuA and MDA-MB-231 cells had high LDLR expression and formed larger tumors in mice with high circulating LDL-C concentrations than in mice with lower LDL-C. Silencing the LDLR in the tumor cells led to decreased growth of Her2/Neu-overexpressing tumors in LDLR-/- and ApoE-/- mice, with increased Caspase 3 cleavage. Additionally, in vitro, silencing the LDLR led to decreased cell survival in serum-starved conditions, associated with Caspase 3 cleavage. Examining publically available human data sets, we found that high LDLR expression in human breast cancers was associated with decreased recurrence-free survival, particularly in patients treated with systemic therapies. Overall, our results highlight the importance of the LDLR in the growth of triple-negative and HER2-overexpressing breast cancers in the setting of elevated circulating LDL-C, which may be important contributing factors to the increased recurrence and mortality in obese women with breast cancer.


Subject(s)
Cholesterol, LDL/metabolism , Hyperlipidemias/metabolism , Mammary Neoplasms, Experimental/metabolism , Receptors, LDL/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Blotting, Western , Cell Line, Tumor , Cell Survival/genetics , Cholesterol, LDL/blood , Disease Models, Animal , Disease Progression , Female , Humans , Hyperlipidemias/blood , Hyperlipidemias/genetics , Kaplan-Meier Estimate , MCF-7 Cells , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice, Knockout , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, LDL/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
2.
Oncogene ; 36(10): 1394-1403, 2017 03.
Article in English | MEDLINE | ID: mdl-27568979

ABSTRACT

Increased breast cancer risk and mortality has been associated with obesity and type 2 diabetes (T2D). Hyperinsulinemia, a key factor in obesity, pre-diabetes and T2D, has been associated with decreased breast cancer survival. In this study, a mouse model of pre-diabetes (MKR mouse) was used to investigate the mechanisms through which endogenous hyperinsulinemia promotes mammary tumor metastases. The MKR mice developed larger primary tumors and greater number of pulmonary metastases compared with wild-type (WT) mice after injection with c-Myc/Vegf overexpressing MVT-1 cells. Analysis of the primary tumors showed significant increase in vimentin protein expression in the MKR mice compared with WT. We hypothesized that vimentin was an important mediator in the effect of hyperinsulinemia on breast cancer metastasis. Lentiviral short hairpin RNA knockdown of vimentin led to a significant decrease in invasion of the MVT-1 cells and abrogated the increase in cell invasion in response to insulin. In the pre-diabetic MKR mouse, vimentin knockdown led to a decrease in pulmonary metastases. In vitro, we found that insulin increased pAKT, prevented caspase 3 activation, and increased vimentin. Inhibiting the phosphatidylinositol 3 kinase/AKT pathway, using NVP-BKM120, increased active caspase 3 and decreased vimentin levels. This study is the first to show that vimentin has an important role in tumor metastasis in vivo in the setting of pre-diabetes and endogenous hyperinsulinemia. Vimentin targeting may be an important therapeutic strategy to reduce metastases in patients with obesity, pre-diabetes or T2D.


Subject(s)
Diabetes Mellitus, Experimental/complications , Gene Silencing , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Vimentin/genetics , Animals , Caspases/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Insulin/pharmacology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
3.
Oncogenesis ; 4: e156, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26053034

ABSTRACT

Estrogen receptor-α (ERα)-negative breast cancer is clinically aggressive and does not respond to conventional hormonal therapies. Strategies that lead to re-expression of ERα could sensitize ERα-negative breast cancers to selective ER modulators. FTY720 (fingolimod, Gilenya), a sphingosine analog, is the Food and Drug Administration (FDA)-approved prodrug for treatment of multiple sclerosis that also has anticancer actions that are not yet well understood. We found that FTY720 is phosphorylated in breast cancer cells by nuclear sphingosine kinase 2 and accumulates there. Nuclear FTY720-P is a potent inhibitor of class I histone deacetylases (HDACs) that enhances histone acetylations and regulates expression of a restricted set of genes independently of its known effects on canonical signaling through sphingosine-1-phosphate receptors. High-fat diet (HFD) and obesity, which is now endemic, increase breast cancer risk and have been associated with worse prognosis. HFD accelerated the onset of tumors with more advanced lesions and increased triple-negative spontaneous breast tumors and HDAC activity in MMTV-PyMT transgenic mice. Oral administration of clinically relevant doses of FTY720 suppressed development, progression and aggressiveness of spontaneous breast tumors in these mice, reduced HDAC activity and strikingly reversed HFD-induced loss of estrogen and progesterone receptors in advanced carcinoma. In ERα-negative human and murine breast cancer cells, FTY720 reactivated expression of silenced ERα and sensitized them to tamoxifen. Moreover, treatment with FTY720 also re-expressed ERα and increased therapeutic sensitivity of ERα-negative syngeneic breast tumors to tamoxifen in vivo more potently than a known HDAC inhibitor. Our work suggests that a multipronged attack with FTY720 is a novel combination approach for effective treatment of both conventional hormonal therapy-resistant breast cancer and triple-negative breast cancer.

4.
Horm Metab Res ; 45(9): 655-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23700320

ABSTRACT

Obesity is associated with hyperleptinemia and this has led to the suggestion that leptin maybe a factor in cancer progression. To study the effect of leptin on cancer progression we used a mouse model of diabetes that was shown to enhance tumor progression and thereby determine if leptin affects cancer progression despite improvements in metabolic status. Mammary tumors were allowed to develop in male and female mice following orthotopic injection of cells expressing oncogenes. After 2 weeks leptin was administered to the mice using Alzet pumps. In these mice leptin failed to stimulate tumor progression; indeed, in those studies where glucose tolerance improved tumor growth was actually inhibited. Thus, the possibility exists that the effect of leptin on tumor progression maybe opposed by improvements in metabolism.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Leptin/administration & dosage , Leptin/pharmacology , Mammary Neoplasms, Animal/complications , Mammary Neoplasms, Animal/pathology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Male , Mice
5.
Diabet Med ; 30(8): 994-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23398488

ABSTRACT

AIM: An online diabetes course for medical residents led to lower patient blood glucose, but also increased hypoglycaemia despite improved trainee confidence and knowledge. Based on these findings, we determined whether an optimized educational intervention delivered to hospitalists (corresponding to an Acute Physician or Specialist in Acute Hospital Medicine in the UK) improved inpatient glycaemia without concomitant hypoglycaemia. METHODS: All 22 hospitalists at an academic medical centre were asked to participate in an online curriculum on the management of inpatient dysglycaemia in autumn 2009 and a refresher course in spring 2010. RESULTS: All hospitalists completed the initial intervention. Median event blood glucose decreased from 9.3 mmol/l (168 mg/dl) pre-intervention to 7.8 mmol/l (141 mg/dl) post-intervention and 8.5 mmol/l (153 mg/dl) post-refresher (P < 0.001 for both). Hospitalizations categorized as hyperglycaemia decreased from 83.3 to 55.6% (P = 0.014), with a trend towards euglycaemia (10-28.9%, P = 0.08) and no change in hypoglycaemia. Hyperglycaemic patient-days decreased from 72.0 to 57.3% (P = 0.004), with greater target glycaemia (27.3-39.4%, P = 0.016) and no change in hypoglycaemia. CONCLUSIONS: An optimized online educational intervention delivered to hospitalists yielded significant improvements in inpatient glycaemia without increased hypoglycaemia.


Subject(s)
Diabetes Mellitus/therapy , Hospitalists/education , Hyperglycemia/prevention & control , Hypoglycemia/prevention & control , Academic Medical Centers , Attitude of Health Personnel , Blood Glucose/analysis , Curriculum , Diabetes Mellitus/blood , Diabetes Mellitus/drug therapy , Diabetes, Gestational/blood , Diabetes, Gestational/drug therapy , Diabetes, Gestational/therapy , Female , Humans , Hyperglycemia/epidemiology , Hypoglycemia/chemically induced , Hypoglycemia/epidemiology , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Internet , Male , New York City/epidemiology , Personal Satisfaction , Pregnancy , Pregnancy in Diabetics/blood , Pregnancy in Diabetics/drug therapy , Pregnancy in Diabetics/therapy
6.
Oncogene ; 32(8): 961-7, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-22469977

ABSTRACT

Dyslipidemia has been associated with an increased risk for developing cancer. However, the implicated mechanisms are largely unknown. To explore the role of dyslipidemia in breast cancer growth and metastasis, we used the apolipoprotein E (ApoE) knockout mice (ApoE(-/-)), which exhibit marked dyslipidemia, with elevated circulating cholesterol and triglyceride levels in the setting of normal glucose homeostasis and insulin sensitivity. Non-metastatic Met-1 and metastatic Mvt-1 mammary cancer cells derived from MMTV-PyVmT/FVB-N transgenic mice and c-Myc/vegf tumor explants respectively, were injected into the mammary fat pad of ApoE(-/-) and wild-type (WT) females consuming a high-fat/high-cholesterol diet and tumor growth was evaluated. ApoE(-/-) mice exhibited increased tumor growth and displayed a greater number of spontaneous metastases to the lungs. Furthermore, intravenous injection of Mvt-1 cells resulted in a greater number of pulmonary metastases in the lungs of ApoE(-/-) mice compared with WT controls. To unravel the molecular mechanism involved in enhanced tumor growth in ApoE(-/-) mice, we studied the response of Mvt-1 cells to cholesterol in vitro. We found that cholesterol increased Akt(S473) phosphorylation in Mvt-1 cells as well as cellular proliferation, whereas cholesterol depletion in the cell membrane abrogated Akt(S473) phosphorylation induced by exogenously added cholesterol. Furthermore, in vivo administration of BKM120, a small-molecule inhibitor of phosphatidylinositol 3-kinase (PI3K), alleviated dyslipidemia-induced tumor growth and metastasis in Mvt-1 model with a concomitant decrease in PI3K/Akt signaling. Collectively, we suggest that the hypercholesterolemic milieu in the ApoE(-/-) mice is a favorable setting for mammary tumor growth and metastasis.


Subject(s)
Hypercholesterolemia/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Lung Neoplasms/blood , Mammary Neoplasms, Experimental/blood , Mice , Mice, Transgenic , Neoplasm Metastasis , Signal Transduction
7.
Oncogene ; 31(27): 3213-22, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22037215

ABSTRACT

Women with type 2 diabetes mellitus (T2DM) are at a greater risk of developing and dying from breast cancer than women without T2DM. Insulin resistance and hyperinsulinemia underlie the pathogenesis of T2DM. In the MKR mouse model of insulin resistance, we have previously shown increased activation of the phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR pathway in association with accelerated mammary tumor growth. In this study, we demonstrate that inhibiting PI3K with the oral pan-class I PI3K inhibitor, NVP-BKM120 reduced the growth of Met-1 and MCNeuA mammary tumor orthografts in the MKR mouse. NVP-BKM120 treatment decreased phosphorylation of Akt and S6 ribosomal protein (S6rp); no change in Erk1/2 phosphorylation was seen. Hyperglycemia, hypertriglyceridemia and greater hyperinsulinemia developed in the MKR mice treated with NVP-BKM120. We previously reported reduced tumor growth using intraperitoneal rapamycin in the MKR mouse, with the development of hyperglycemia and hypertriglyceridemia. Therefore, we examined whether the oral PI3K/mTOR inhibitor NVP-BEZ235 augmented the tumor suppressing effects of PI3K inhibition. We also investigated the effect of targeted PI3K/mTOR inhibition on PI3K/Akt/mTOR and Erk1/2 signaling, and the potential effects on glycemia. NVP-BEZ235 suppressed the growth of Met-1 and MCNeuA tumor orthografts, and decreased Akt and S6rp phosphorylation, despite increased Erk1/2 phosphorylation in Met-1 orthografts of MKR mice. Less marked hyperglycemia and hyperinsulinemia developed with NVP-BEZ235 than NVP-BKM120. Overall, the results of this study demonstrated that inhibiting PI3K/Akt/mTOR signaling with the oral agents NVP-BKM120 and NVP-BEZ235 decreased mammary tumor growth in the hyperinsulinemic MKR mouse. Inhibiting PI3K alone led to more severe metabolic derangement than inhibiting both PI3K and mTOR. Therefore, PI3K may be an important target for the treatment of breast cancer in women with insulin resistance. Monitoring for hyperglycemia and dyslipidemia should be considered when using these agents in humans, given the metabolic changes detected in this study.


Subject(s)
Hyperglycemia/chemically induced , Hyperinsulinism/complications , Insulin Resistance , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Disease Progression , Drug Interactions , Female , Humans , Imidazoles/adverse effects , Imidazoles/pharmacology , Imidazoles/therapeutic use , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/adverse effects , Quinolines/pharmacology , Quinolines/therapeutic use , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
8.
Diabetologia ; 53(10): 2224-32, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20577711

ABSTRACT

AIMS/HYPOTHESIS: Recent work has shown that there can be significant differences when glucose disposal is assessed for high-fat induced insulin resistance by static clamp methods vs dynamic assessment during a stable isotope i.p. glucose tolerance test. MKR mice, though lean, have severe insulin resistance and decreased muscle fatty acid oxidation. Our goal was to assess dynamic vs static glucose disposal in MKR mice, and to correlate glucose disposal and muscle-adipose-liver flux interactions with metabolic flexibility (indirect calorimetry) and muscle characteristics. METHODS: Stable isotope flux phenotyping was performed using [6,6-(2)H(2)]glucose, [U-(13)C(6)]glucose and [2-(13)C]glycerol. Muscle triacylglycerol (TAG) and diacylglycerol (DAG) content was assessed by thin layer chromatography, and histological determination of fibre type and cytochrome c activity performed. Metabolic flexibility was assessed by indirect calorimetry. RESULTS: Indirect calorimetry showed that MKR mice used more glucose than FVB/N mice during fasting (respiratory exchange ratio [RER] 0.88 vs 0.77, respectively). Compared with FVB/N mice, MKR mice had faster dynamic glucose disposal, despite increased whole-muscle DAG and TAG, and similar hepatic glucose production with higher fasting insulin and unchanged basal glucose. Fed MKR muscle had more glycogen, and increased levels of GLUT1 and GLUT4 than FVB/N muscle. Histology indicated that MKR soleus had mildly decreased cytochrome c activity overall and more type II (glycolytic) fibres compared with that in FVB/N mice. CONCLUSIONS/INTERPRETATION: MKR muscle adapts to using glucose, with more type II fibres present in red muscle. Fasting RER is elevated and glucose disposal during an i.p. glucose tolerance test is accelerated despite increased muscle DAG and TAG. Metabolic inflexibility may result from the compensatory use of fuel that can be best utilised for energy requirements; static vs dynamic glucose disposal assessments may measure complementary aspects of metabolic flexibility and insulin sensitivity.


Subject(s)
Glucose/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Animals , Blotting, Western , Body Composition/physiology , Calorimetry, Indirect , Glucose Tolerance Test , Hyperglycemia/metabolism , Lipid Metabolism/physiology , Lipid Metabolism/radiation effects , Male , Mice , Phenotype
9.
Oncogene ; 28(39): 3477-86, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19617901

ABSTRACT

Polyoma virus middle T antigen (PyVmT) is a powerful viral oncogene; however, the mechanisms of PyVmT activation are poorly understood. The insulin-like growth factor I receptor (IGF-IR) and the insulin receptor (IR) are known to be implicated in the development of many cancers. Furthermore, PyVmT-overexpressing mouse mammary carcinoma Met-1 cells are highly responsive to IGF-I and insulin. Herein, we demonstrate that PyVmT physically interacts with IGF-IR and IR in Met-1 cells. Insulin and IGF-I increase association of the IR and IGF-IR with PyVmT, enhance tyrosine phosphorylation of PyVmT and augment the recruitment of Src and PLCgamma(1) to PyVmT. This is accompanied by robust and sustained phosphorylation of Akt and ERK1/2, which are implicated in both PyVmT and IGF-IR/IR signalling. Both ligands significantly increase proliferation, survival, migration and invasion of Met-1 cells. Furthermore, orthotopic inoculation of Met-1 cells with shRNAmir-mediated knockdown of IR or IGF-IR fails to initiate tumour growth in recipient mice. In conclusion, our data indicate that the physical and functional interaction between PyVmT and cellular receptor tyrosine kinases, including IR and IGF-IR, is critical for PyVmT activation and tumour initiation. These results also provide a novel mechanism for oncogene activation in the host cell.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Insulin/metabolism , Mammary Neoplasms, Animal/metabolism , Receptor, IGF Type 1/metabolism , Animals , Cell Line, Tumor , Gene Knockdown Techniques , MAP Kinase Signaling System , Mice , Mice, Inbred Strains , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Receptor, Insulin/metabolism
10.
Diabetologia ; 52(7): 1434-41, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19436992

ABSTRACT

AIMS/HYPOTHESIS: Although it is known that lipid metabolism plays a role in insulin resistance in type 2 diabetes and in obesity, the mechanism is still largely unknown. Apolipoprotein E (ApoE) regulates plasma lipid levels and also plays a role in the uptake of lipids into various tissues. To investigate whether the suppression of whole-particle lipoprotein uptake into tissues affects insulin responsiveness and the diabetic condition, we examined the effect of an ApoE (also known as Apoe) gene deletion in MKR mice, a mouse model of type 2 diabetes. METHODS: ApoE ( -/- ), MKR, ApoE ( -/- )/MKR and control mice were placed on a high-fat, high-cholesterol diet for 16 weeks. Glucose tolerance, serum insulin, blood glucose, insulin tolerance, tissue triacylglycerol content and atherosclerotic lesions were assessed. RESULTS: ApoE ( -/- )/MKR and ApoE ( -/- ) mice showed significantly improved blood glucose, glucose tolerance and insulin sensitivity. Reduced triacylglycerol content in liver and reduced fat accumulation in liver and adipose tissue were found in ApoE ( -/- )/MKR and ApoE ( -/- ) mice compared with control and MKR mice. ApoE ( -/- ) and ApoE ( -/- )/MKR mice demonstrated similarly large atherosclerotic lesions, whereas MKR and control mice had small atherosclerotic lesions. CONCLUSIONS/INTERPRETATION: We demonstrated that ApoE deficiency abrogates insulin resistance in a mouse model of type 2 diabetes, suggesting that lipid accumulation in tissue is a major cause of insulin resistance in this mouse model.


Subject(s)
Apolipoproteins E/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Insulin Resistance/physiology , Lipid Metabolism/physiology , Adipose Tissue/metabolism , Animals , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Blood Glucose/metabolism , Body Composition/physiology , Body Weight/physiology , Cholesterol, Dietary/blood , Cholesterol, Dietary/pharmacology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Disease Models, Animal , Fatty Acids, Nonesterified/blood , Female , Insulin/blood , Liver/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Triglycerides/blood
11.
Exp Clin Endocrinol Diabetes ; 116 Suppl 1: S4-6, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18777452

ABSTRACT

Both obesity and Type 2 diabetes are independently associated with an increased risk of developing cancer and an increased mortality. The etiology is yet to be determined but insulin resistance and hyperinsulinemia maybe important factors. Hyperglycemia, hyperlipidemia and inflammatory cytokines in addition to the insulin-like growth factors are also possible factors involved in the process.


Subject(s)
Diabetes Mellitus, Type 2/complications , Neoplasms/diagnosis , Neoplasms/epidemiology , Neoplasms/etiology , Obesity/complications , Signal Transduction/genetics , Animals , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Humans , Models, Biological , Neoplasms/genetics , Obesity/epidemiology , Obesity/genetics , Prognosis , Risk Factors , Signal Transduction/physiology
12.
Horm Metab Res ; 40(10): 661-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18711691

ABSTRACT

Human papillomavirus (HPV) is the main risk factor for cervical cancer; however, some carcinomas occur in the absence of the virus. IGF-IR and an isoform of the insulin receptor, IR-A, play important roles in cancer. In this study we assessed the role of the IGF/insulin receptors in cervical cancer cell lines with different HPV status, SiHa (HPV positive), and C33a (HPV negative). Different patterns of receptor expression were found; while SiHa expressed IGF-IR, IR-A and IR-B, and IR/IGF-IR hybrid receptors, C33a cells expressed the IR-A only. Tyrosine phosphorylation of these receptors in response to their corresponding ligands correlated with the expression level of these receptors in the cell lines. Activation of PI3-K and MAPK pathways was revealed in both cell lines, however, no effects on proliferation, migration, or invasion were observed. Here we show that cervical cancer cell lines--positive and negative for HPV--differ in the type of insulin and IGF-1 receptors expressed. Additional studies are needed for characterization of the role of IR-A in cervical carcinogenesis.


Subject(s)
Papillomaviridae/physiology , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/virology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Female , Humans , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/metabolism , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/pathology
13.
Minerva Med ; 99(3): 253-62, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18497723

ABSTRACT

The metabolic syndrome is a constellation of risk factors including glucose dysregulation, central obesity, dyslipidemia, and hypertension. There are multiple definitions that have been described by various health organizations. However, we do know that insulin resistance plays a major role as the underlying cause for the development and potentiation of the metabolic syndrome. At present, it is unclear if the diagnosis of metabolic syndrome is greater than the sum of its parts. However, the presence of more than one of the associated risk factors should indicate that a patient is at increased risk for developing diabetes, cardiovascular disease and death. Thus, the primary care physician should aggressively treat the metabolic risk factors in their patients to prevent the onset and progression to more severe disease.


Subject(s)
Insulin Resistance/physiology , Metabolic Syndrome/etiology , Cytokines/metabolism , Dyslipidemias/complications , Dyslipidemias/metabolism , Fatty Acids, Nonesterified/metabolism , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , Hypertension/complications , Hypertension/drug therapy , Inflammation/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Metabolic Syndrome/physiopathology , Obesity/complications , Obesity/metabolism
14.
Cell Death Differ ; 15(8): 1304-17, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18437163

ABSTRACT

Disruption of endoplasmic reticulum (ER) homeostasis causes accumulation of unfolded and misfolded proteins in the ER, triggering the ER stress response, which can eventually lead to apoptosis when ER dysfunction is severe or prolonged. Here we demonstrate that human MCF-7 breast cancer cells, as well as murine NIH/3T3 fibroblasts, are rescued from ER stress-initiated apoptosis by insulin-like growth factor-I (IGF-I). IGF-I significantly augments the adaptive capacity of the ER by enhancing compensatory mechanisms such as the IRE1 alpha-, PERK- and ATF6-mediated arms of ER stress signalling. During ER stress, IGF-I stimulates translational recovery and induces expression of the key molecular chaperone protein Grp78/BiP, thereby enhancing the folding capacity of the ER and promoting recovery from ER stress. We also demonstrate that the antiapoptotic activity of IGF-I during ER stress may be mediated by a novel, as yet unidentified, signalling pathway(s). Application of signal transduction inhibitors of MEK (U1026), PI3K (LY294002 and wortmannin), JNK (SP600125), p38 (SB203580), protein kinases A and C (H-89 and staurosporine) and STAT3 (Stattic) does not prevent IGF-I-mediated protection from ER stress-induced apoptosis. Taken together, these data demonstrate that IGF-I protects against ER stress-induced apoptosis by increasing adaptive mechanisms through enhancement of ER stress-signalling pathways, thereby restoring ER homeostasis and preventing apoptosis.


Subject(s)
Endoplasmic Reticulum/metabolism , Insulin-Like Growth Factor I/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cycloheximide/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Enzyme Inhibitors/pharmacology , Humans , Mice , NIH 3T3 Cells , Protein Kinase Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , Recombinant Proteins/metabolism , Signal Transduction , Thapsigargin/pharmacology
15.
Oral Dis ; 14(2): 105-14, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18302671

ABSTRACT

Diabetes mellitus is a prevalent disease that affects millions of people worldwide and has paralleled the growing population of overweight and obese individuals. Early detection of prediabetes and diabetes, as well as lifestyle interventions including diet and exercise, are the overarching objectives in preventing and managing diabetes. For individuals who do not achieve glycemic control with lifestyle modification, there are newer medication classes that assist with weight loss, more physiologic insulins with convenient delivery systems, and old standbys like metformin and thiazolidinediones. Glycemic control along with blood pressure and cholesterol management reduce microvascular and macrovascular disease including cardiovascular events. Mounting evidence demonstrates that diabetes is a risk factor for periodontitis and possibly oral premalignancies and oral cancer. The systemic inflammatory response generated by inflamed periodontal tissue may in turn exacerbate diabetes, worsen cardiovascular outcomes, and increase mortality. Thus, oral medical and surgical physicians are vital in treating oral pathology, recognizing new cases of diabetes, and counseling people with diabetes to promote oral health. This article presents updates in the diagnosis, risk factors, prevention, management, and peri-oral complications of diabetes to assist oral health professionals in providing optimal care to patients with diabetes.


Subject(s)
Cardiovascular Diseases/complications , Diabetes Complications/complications , Diabetes Mellitus , Mouth Diseases/complications , Blood Glucose/metabolism , Cardiovascular Diseases/metabolism , Diabetes Complications/metabolism , Diabetes Complications/prevention & control , Diabetes Mellitus/classification , Diabetes Mellitus/metabolism , Diabetes Mellitus/prevention & control , Diabetes Mellitus/therapy , Humans , Mouth Diseases/metabolism , Mouth Diseases/prevention & control
16.
Mol Psychiatry ; 12(12): 1118-28, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17848918

ABSTRACT

Increasing evidence indicates that circulating insulin-like growth factor I (IGF-I) acts as a peripheral neuroactive signal participating not only in protection against injury but also in normal brain function. Epidemiological studies in humans as well as recent evidence in experimental animals suggest that blood-borne IGF-I may be involved in cognitive performance. In agreement with observations in humans, we found that mice with low-serum IGF-I levels due to liver-specific targeted disruption of the IGF-I gene presented cognitive deficits, as evidenced by impaired performance in a hippocampal-dependent spatial-recognition task. Mice with serum IGF-I deficiency also have disrupted long-term potentiation (LTP) in the hippocampus, but not in cortex. Impaired hippocampal LTP was associated with a reduction in the density of glutamatergic boutons that led to an imbalance in the glutamatergic/GABAergic synapse ratio in this brain area. Behavioral and synaptic deficits were ameliorated in serum IGF-I-deficient mice by prolonged systemic administration of IGF-I that normalized the density of glutamatergic boutons in the hippocampus. Altogether these results indicate that liver-derived circulating IGF-I affects crucial aspects of mature brain function; that is, learning and synaptic plasticity, through its trophic effects on central glutamatergic synapses. Declining levels of serum IGF-I during aging may therefore contribute to age-associated cognitive loss.


Subject(s)
Brain/metabolism , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Insulin-Like Growth Factor I/metabolism , Liver Extracts/chemistry , Age Factors , Animals , Behavior, Animal , Brain/anatomy & histology , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/genetics , Glutamate Decarboxylase/metabolism , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/therapeutic use , Maze Learning/physiology , Mice , Mice, Transgenic , Vesicular Glutamate Transport Protein 1/metabolism
17.
Diabetologia ; 49(1): 90-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16362284

ABSTRACT

AIMS/HYPOTHESIS: Although insulin resistance induces compensatory increases in beta cell mass and function to maintain normoglycaemia, it is not clear whether insulin resistance can precipitate beta cell dysfunction and hyperglycaemia without a pre-existing beta cell susceptibility. We therefore examined the beta cell phenotype in the MKR mouse, a model in which expression of a dominant-negative IGF 1 receptor (IGF1R) in skeletal muscle leads to systemic insulin resistance and diabetes. MATERIALS AND METHODS: Circulating glucose, insulin and glucagon concentrations were measured. Insulin sensitivity, glucose tolerance and insulin release in vivo were assessed by i.p. insulin and glucose tolerance tests. Beta cell function was assessed via insulin secretion from isolated islets and the glucose gradient in the perfused pancreas. Beta cell morphology was examined via immunohistochemistry. MKR mice were fed a high-fat diet containing sucrose (HFSD) to test metabolic capacity and beta cell function. RESULTS: Insulin-resistant MKR mice developed hyperglycaemia and a loss of insulin responsiveness in vivo. Basal insulin secretion from the perfused pancreas was elevated, with no response to glucose. Despite the demand on insulin secretion, MKR mice had increased pancreatic insulin content and beta cell mass mediated through hyperplasia and hypertrophy. The HFSD worsened hyperglycaemia in MKR mice but, despite increased food intake in these mice, failed to induce the obesity observed in wild-type mice. CONCLUSIONS/INTERPRETATION: Our studies demonstrate that insulin resistance of sufficient severity can impair glucose-stimulated insulin secretion, thereby undermining beta cell compensation and leading to hyperglycaemia. Moreover, because insulin stores were intact, the secretory defects reflect an early stage of beta cell dysfunction.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Glucose/pharmacology , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Blood Glucose/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/chemically induced , Dietary Sucrose , Disease Models, Animal , Glucagon/blood , Glucose/metabolism , Homeostasis , Insulin/blood , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred Strains , Receptor, Insulin/genetics
18.
J Endocrinol Invest ; 28(5 Suppl): 19-22, 2005.
Article in English | MEDLINE | ID: mdl-16114270

ABSTRACT

The IGFs are ubiquitous and have pleoitropic effects. They are critical for normal growth and development, and for normal functioning of adult tissues. A liver-specific gene-deletion knockout of the IGF-I gene resulted in a mouse model with reduced circulating IGF-I levels, that led to insulin resistance due to the secondary elevation of circulating GH levels. The reduction in circulating IGF-I levels was also associated with a reduction in cancer growth and metastases in three cancer models, one for colon cancer and two for breast cancer. A second mouse model, using the transgenic approach, inhibited the IGF-I and insulin receptor function in skeletal muscle, and resulted in severe insulin resistance in muscle followed by insulin resistance in fat and liver and, eventually, beta-cell dysfunction and development of Type 2 diabetes. This progression from insulin resistance to Type 2 diabetes was most likely due to lipotoxicity with elevated serum and tissue triglyceride levels. Evidence supporting the hypothesis came from the use of fibrates and leptin injections, each of which enhanced fatty acid (FA) oxidation in liver and muscle and was associated with a reversal of the insulin resistance and diabetes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Growth Hormone/physiology , Insulin-Like Growth Factor I/physiology , Neoplasms/physiopathology , Receptor, IGF Type 1/genetics , Animals , Disease Models, Animal , Mice , Mice, Knockout
19.
Proc Natl Acad Sci U S A ; 101(26): 9833-8, 2004 Jun 29.
Article in English | MEDLINE | ID: mdl-15210967

ABSTRACT

Although vascular dysfunction is a major suspect in the etiology of several important neurodegenerative diseases, the signals involved in vessel homeostasis in the brain are still poorly understood. We have determined whether insulin-like growth factor I (IGF-I), a wide-spectrum growth factor with angiogenic actions, participates in vascular remodeling in the adult brain. IGF-I induces the growth of cultured brain endothelial cells through hypoxiainducible factor 1 alpha and vascular endothelial growth factor, a canonical angiogenic pathway. Furthermore, the systemic injection of IGF-I in adult mice increases brain vessel density. Physical exercise that stimulates widespread brain vessel growth in normal mice fails to do so in mice with low serum IGF-I. Brain injury that stimulates angiogenesis at the injury site also requires IGF-I to promote perilesion vessel growth, because blockade of IGF-I input by an anti-IGF-I abrogates vascular growth at the injury site. Thus, IGF-I participates in vessel remodeling in the adult brain. Low serum/brain IGF-I levels that are associated with old age and with several neurodegenerative diseases may be related to an increased risk of vascular dysfunction.


Subject(s)
Brain/blood supply , Insulin-Like Growth Factor I/metabolism , Neovascularization, Physiologic , Aging/physiology , Animals , Antibodies/pharmacology , Brain/drug effects , Brain/pathology , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Division/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/deficiency , Insulin-Like Growth Factor I/pharmacology , Male , Mice , Mice, Inbred C57BL , Mutation , Neovascularization, Physiologic/drug effects , Physical Conditioning, Animal/physiology , Rats , Rats, Wistar , Serum/chemistry
20.
Ann Endocrinol (Paris) ; 65(1): 99-103, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15122103

ABSTRACT

NICTH is a fairly uncommon disorder but has been well characterized and the molecular mechanisms involved have given insights into the IGF system, both normal and abnormal. NICTH has brought together the molecular biologists and endocrinologists in a classic cross cultural coordination to study the disorder from all angles and to further understand the disorder.


Subject(s)
Hypoglycemia/etiology , Hypoglycemia/physiopathology , Adult , Homeostasis , Humans , Insulin-Like Growth Factor I/physiology , Insulin-Like Growth Factor II/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...