Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Trends Endocrinol Metab ; 31(8): 551-560, 2020 08.
Article in English | MEDLINE | ID: mdl-32600959

ABSTRACT

In addition to being a major metabolic hormone, insulin is also a growth factor with a mitogenic effect on all cells, more marked in malignant cells that often overexpress the insulin receptor. In patients with metabolic diseases characterized by hyperinsulinemia (obesity, type 2 diabetes, and metabolic syndrome), the incidence of several types of cancer is increased, as is cancer-related mortality. Because of the worldwide growing prevalence of metabolic diseases and the diffuse use of insulin and its analogs for treating diabetes, the relationship between insulin and cancer has become a clinically relevant issue. Clinical studies have not clarified the degree to which hyperinsulinemia can influence cancer occurrence and prognosis. To better understand this issue, an improved scientific approach is required, with more careful consideration of the mechanisms related to hyperinsulinemia and carcinogenesis.


Subject(s)
Insulin/metabolism , Neoplasms/metabolism , Obesity/metabolism , Animals , Diabetes Mellitus, Type 2/physiopathology , Humans , Hyperinsulinism/metabolism , Insulin Resistance/physiology
2.
Mol Cell Endocrinol ; 457: 73-80, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-27794445

ABSTRACT

In the last two decades thyroid cancer incidence has increased worldwide more than any other cancer. Overdiagnosis of subclinical microcarcinomas has certainly contributed to this increase but many evidences indicate that a true increase, possibly due to environmental factors, has also occurred. Thyroid cancer incidence is markedly increased in volcanic areas. Thus, the volcanic environment is a good model to investigate the possible factors favoring thyroid cancer. In the volcanic area of Mt. Etna in Sicily, as well as in other volcanic areas, a non-anthropogenic pollution with heavy metals has been documented, a consequence of gas, ash and lava emission. Soil, water and atmosphere contamination, via the food chain, biocontaminate the residents as documented by high levels in the urines and the scalp hair compared to individuals living in adjacent non-volcanic areas. Trace amounts of metals are essential nutrients but, at higher concentrations, can be toxic for living cells. Metals can behave both as endocrine disruptors, perturbing the hormonal system, and as carcinogens, promoting malignant transformation. Similarly to other carcinogens, the transforming effect of heavy metals is higher in developing organisms as the fetus (contaminated via the mother) and individuals in early childhood. In the last decades environment metal pollution has greatly increased in industrialized countries. Although still within the "normal" limits for each single metal the hormesis effect (heavy metal activity at very low concentration because of biphasic, non linear cell response) and the possible potentiation effect resulting from the mixture of different metals acting synergistically can explain cell damage at very low concentrations. The effect of metals on the human thyroid is poorly studied: for some heavy metals no data are available. The scarce studies that have been performed mainly focus on metal effect as thyroid endocrine disruptors. The metal concentration in tissues has been rarely measured in the thyroid. Heavy metal accumulation and metabolism in the thyroid or the carcinogenic activity of different doses and different speciation of metals has not been investigated. These studies are now warranted to better understand thyroid biology and heavy metal role in human thyroid carcinogenesis.


Subject(s)
Environment , Metals, Heavy/analysis , Thyroid Neoplasms/chemically induced , Volcanic Eruptions/analysis , Humans , Incidence , Thyroid Gland/pathology , Thyroid Neoplasms/epidemiology
3.
Nutr Metab Cardiovasc Dis ; 23(9): 808-15, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23932729

ABSTRACT

Cancer incidence and mortality are higher among diabetic patients. This review examines the mechanisms, both general and site-specific, for this increase. Hyperglycemia and hyperinsulinemia, which are the major abnormalities that characterize diabetes, can promote cancer via both independent and synergic mechanisms. Insulin is both a metabolic hormone and a growth factor that promotes cell proliferation. When insulin levels are increased due to either insulin resistance or insulin treatment, their mitogenic effect is more marked in malignant cells that frequently overexpress the insulin receptor and, more specifically, its A isoform that has predominant mitogenic activity. Hyperglycemia provides energy for malignant cell proliferation and, via the peculiar energy utilization of cancer cells, favors cancer growth and neoangiogenesis. Additionally, diabetes-associated obesity has cancer-promoting effects due to mechanisms that are specific to excess fat cells (such as increased peripheral estrogens, increased pro-mitogen cytokines and growth factors). Also fat-associated chronic inflammation can favor cancer via the cell damage caused by reactive oxygen species (ROS) and via the production of inflammatory cytokines and transcription factors that stimulate cancer growth and invasiveness. Finally, the multiple drugs involved in the treatment of diabetes can also play a role. Diabetes-associated comorbidities, tissue-specific inflammation, and organ-specific dysfunctions can explain why the risk of cancer can differ by tissue type among diabetic patients. The increased risk of cancer-related mortality is moderate among individual patients with diabetes (RR = 1.25), but the pandemic nature of the disease means that a considerable number of lives could be spared through a better understanding of the factors associating diabetes and cancer.


Subject(s)
Breast Neoplasms/physiopathology , Colorectal Neoplasms/physiopathology , Diabetes Mellitus, Type 2/drug therapy , Endometrial Neoplasms/physiopathology , Liver Neoplasms/physiopathology , Prostatic Neoplasms/physiopathology , Breast Neoplasms/etiology , Colorectal Neoplasms/etiology , Cytokines/blood , Diabetes Mellitus, Type 2/complications , Endometrial Neoplasms/etiology , Female , Humans , Hyperglycemia/complications , Hyperglycemia/drug therapy , Hyperinsulinism/complications , Hyperinsulinism/drug therapy , Hypoglycemic Agents/therapeutic use , Inflammation/complications , Inflammation/drug therapy , Insulin/blood , Insulin/therapeutic use , Insulin Resistance , Liver Neoplasms/etiology , Male , Obesity/complications , Obesity/drug therapy , Pancreas/drug effects , Pancreas/metabolism , Prostatic Neoplasms/etiology , Risk Factors
4.
Acta Haematol ; 126(4): 205-10, 2011.
Article in English | MEDLINE | ID: mdl-21934293

ABSTRACT

BRIT1 (BRCT-repeat inhibitor of hTERT expression), also known as microcephalin (MCPH1), is a crucial gene in the complex cellular machine that is devoted to DNA repair and acts as a regulator of both the intra-S and G2/M checkpoints. The most important role of BRIT1/MCPH1 in the regulation of cell cycle progression appears to be the G2/M checkpoint. The K562 and peripheral blood cells of chronic myeloid leukemia (CML) patients at diagnosis were found to downregulate BRIT1/MCPH1. However, we could not find any correlation between bcr/abl activity and the BRIT1/MCPH1 level. In order to study the genomic instability of CML cells, we evaluated the ability of these cells to arrest mitotic division after exposure to hydroxyurea, a known genotoxic agent. We showed that CML cells continue to proliferate without the activation of the G2/M cell cycle checkpoint arrest or of the apoptotic mechanism. This behavior may predispose the cells to accumulate genomic defects. In conclusion, we found that CML cells have a low BRIT1/MCPH1 level and show a defective G2/M arrest, confirming that these cells have a constitutive genomic instability.


Subject(s)
G2 Phase Cell Cycle Checkpoints , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Actins/antagonists & inhibitors , Adult , Aged , Cell Cycle Proteins , Cell Proliferation/drug effects , Cells, Cultured , Cytochalasin B/toxicity , Cytokinesis/drug effects , Cytoskeletal Proteins , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Hydroxyurea/antagonists & inhibitors , Hydroxyurea/toxicity , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukocytes/drug effects , Leukocytes/metabolism , Male , Middle Aged , Mutagens/toxicity , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , RNA, Messenger/metabolism
10.
Oncogene ; 20(29): 3845-56, 2001 Jun 28.
Article in English | MEDLINE | ID: mdl-11439348

ABSTRACT

The Hepatocyte Growth Factor (HGF) and its receptor Met are physiological regulators of cell migration. HGF and Met have also been implicated in tumor progression and metastasis. We show here that the tyrosine kinase inhibitor STI571 has a stimulatory effect on HGF-induced migration and branching morphogenesis in thyroid cancer but not in primary or immortalized thyroid epithelial cells. These stimulatory effects of STI571 are observed at a concentration that is clinically relevant. The STI571-enhanced motile response can be correlated with an increase in the Met receptor tyrosine phosphorylation as well as ERK and Akt activation by HGF. Interestingly, one of the targets of STI571, namely the c-Abl tyrosine kinase, is activated by HGF and is recruited at the migrating edge of thyroid cancer cells. These data suggests that c-Abl and/or STI571-inhibited tyrosine kinases can negatively regulate the Met receptor to restrain the motile response in thyroid cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Hepatocyte Growth Factor/pharmacology , Piperazines/pharmacology , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Benzamides , Cells, Cultured , Enzyme Activation , Extracellular Matrix , Hepatocyte Growth Factor/metabolism , Humans , Imatinib Mesylate , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Morphogenesis , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-met/metabolism , Thyroid Gland/cytology , Thyroid Neoplasms , Tumor Cells, Cultured , Tyrosine/metabolism
11.
Nat Med ; 7(2): 228-34, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11175855

ABSTRACT

The chimeric BCR-ABL oncoprotein is the molecular hallmark of chronic myelogenous leukemia (CML). BCR-ABL contains nuclear import and export signals but it is localized only in the cytoplasm where it activates mitogenic and anti-apoptotic pathways. We have found that inhibition of the BCR-ABL tyrosine kinase, either by mutation or by the drug STI571, can stimulate its nuclear entry. By combining STI571 with leptomycin B (LMB) to block nuclear export, we trapped BCR-ABL in the nucleus and the nuclear BCR-ABL tyrosine kinase activates apoptosis. As a result, the combined treatment with STI571 and LMB causes the irreversible and complete killing of BCR-ABL transformed cells, whereas the effect of either drug alone is fully reversible. The combined treatment with STI571 and LMB also preferentially eliminates mouse bone marrow cells that express BCR-ABL. These results indicate that nuclear entrapment of BCR-ABL can be used as a therapeutic strategy to selectively kill chronic myelogenous leukemia cells.


Subject(s)
Apoptosis , Cell Nucleus/metabolism , Fusion Proteins, bcr-abl/metabolism , Active Transport, Cell Nucleus , Animals , Benzamides , Bone Marrow Cells/cytology , Cell Line, Transformed , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated/pharmacology , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Mice , Piperazines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Tumor Cells, Cultured
12.
Am J Pathol ; 155(6): 1967-76, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10595926

ABSTRACT

Experimental evidence has shown, both in vitro and in animal models, that neoplastic growth and subsequent metastasis formation depend on the tumor's ability to induce an angiogenic switch. This requires a change in the balance of angiogenic stimulators and inhibitors. To assess the potential role of angiogenesis factors in human thyroid tumor growth and spread, we analyzed their expression by semiquantitative RT-PCR and immunohistochemistry in normal thyroid tissues, benign lesions, and different thyroid carcinomas. Compared to normal tissues, in thyroid neoplasias we observed a consistent increase in vascular endothelial growth factor (VEGF), VEGF-C, and angiopoietin-2 and in their tyrosine kinase receptors KDR, Flt-4, and Tek. In particular, we report the overexpression of angiopoietin-2 and VEGF in thyroid tumor progression from a prevascular to a vascular phase. In fact, we found a strong association between tumor size and high levels of VEGF and angiopoietin-2. Furthermore, our results show an increased expression of VEGF-C in lymph node invasive thyroid tumors and, on the other hand, a decrease of thrombospondin-1, an angioinhibitory factor, in thyroid malignancies capable of hematic spread. These results suggest that, in human thyroid tumors, angiogenesis factors seem involved in neoplastic growth and aggressiveness. Moreover, our findings are in keeping with a recent hypothesis that in the presence of VEGF, angiopoietin-2 may collaborate at the front of invading vascular sprouts, serving as an initial angiogenic signal that accompanies tumor growth.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Angiogenesis Inhibitors/metabolism , Neovascularization, Pathologic/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Angiopoietin-1 , Angiopoietin-2 , Blotting, Northern , Down-Regulation , Endothelial Growth Factors/metabolism , Humans , Immunohistochemistry , Lymphatic Metastasis , Lymphokines/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Invasiveness , Neoplasm Staging , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , RNA/analysis , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondins/metabolism , Thyroid Gland/metabolism , Thyroid Gland/pathology , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
13.
J Clin Endocrinol Metab ; 83(7): 2493-6, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9661633

ABSTRACT

Expression of the Na+/I- symporter (NIS) gene was investigated by RT-PCR in a selected series of 26 primary thyroid carcinomas (19 papillary, 5 follicular, and 2 anaplastic). Fifteen follicular adenomas (11 "cold" and 4 "hot" adenomas) were also studied. Five of 19 papillary thyroid cancer did not express NIS messenger ribonucleic acid (mRNA). In all but 1 follicular cancer, NIS transcript was fully detected. In anaplastic tissue, NIS mRNA was only barely detected in 1 case. All of the follicular thyroid adenomas except 1 expressed the NIS gene. In contrast, all tumors studied excluding the anaplastic histotype fully expressed thyroglobulin and thyroid peroxidase mRNA transcripts. In 2 patients, a lower expression (3- to 5-fold) of NIS mRNA was found in metastasis by dot blot analysis compared with those in both normal and primary neoplastic thyroid tissue. Four of 8 differentiated thyroid cancer patients selected for the presence of metastases with negative posttherapy 131I total body scan showed the lack of NIS gene expression in their primary cancer. This defect, at least in these cases, is a somatic and intrinsic lesion of the primary cancer cells and is not due to a dedifferentiation process in the metastatic tissue. The early detection of the loss of NIS gene expression in the primary cancer, therefore, may provide useful information for the management of differentiated thyroid cancer patients.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic/physiology , Iodides/metabolism , Sodium/metabolism , Thyroid Neoplasms/metabolism , Adenocarcinoma, Follicular/metabolism , Adenoma/metabolism , Adult , Aged , Carcinoma/metabolism , Carcinoma, Papillary/metabolism , Female , Humans , Male , Middle Aged , RNA, Messenger/biosynthesis , Thyroid Neoplasms/secondary
14.
Clin Cancer Res ; 4(1): 223-8, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9516975

ABSTRACT

The papillary carcinoma family (PCF) of thyroid tumors includes a wide variety of neoplastic entities regarded as well-differentiated, poorly differentiated, and undifferentiated papillary thyroid carcinomas. Recent studies have established the presence of alternative oncogenic rearrangements of the RET and NTRK1 genes in a consistent fraction (< or = 50%) of papillary thyroid tumors. RET oncogenic rearrangements are also very frequent (approximately 60%) in Chernobyl radiation-associated papillary thyroid neoplasias, which show an increased aggressiveness in terms of pathological stage at disease onset. These observations prompted us to study the relationship between the presence or absence of RET and NTRK1 oncogenes and the clinicopathological features (age, sex, histopathology, and pTNMC2 staging) of 76 consecutive, non-radiation-related tumors of the PCF. As previously reported, statistical univariate analysis revealed a correlation between the combination of RET and NTRK1 (RET/NTRK1) positivity and young age of patients at diagnosis. In addition, a significant association was found between RET/NTRK1 positivity and locally advanced stage of disease at presentation (pT4: P < 0.015). The multivariate analysis confirmed that RET/NTRK1 activation parallels an unfavorable disease presentation, which may correlate with a less favorable disease outcome. Furthermore, within the PCF, the frequency of RET/NTRK1 positivity was not influenced by the different neoplastic subtypes or the tumor versus degree of differentiation.


Subject(s)
Carcinoma, Papillary/genetics , Drosophila Proteins , Gene Rearrangement , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Nerve Growth Factor/genetics , Thyroid Neoplasms/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Papillary/pathology , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-ret , Receptor, trkA , Thyroid Neoplasms/pathology
15.
Recent Results Cancer Res ; 154: 237-47, 1998.
Article in English | MEDLINE | ID: mdl-10027004

ABSTRACT

A combined cytogenetic and molecular analysis of thyroid tumours has indicated that these neoplasms might represent a significant model for analysing human epithelial cell multi-step cancerogenesis. Thyroid tumours comprise a broad spectrum of lesions with different phenotypes and variable biological and clinical behaviour. Molecular analysis has detected specific genetic alterations in these different tumour types. In particular, the well-differentiated carcinomas of the papillary type are characterised by the activation of the tyrosine kinase receptors (TKRs) RET and NTRK1 proto-oncogenes. Cytogenetic analysis of these tumours has contributed to defining the chromosomal mechanisms leading to the TKRs' oncogenic activation. The results have shown that, in the majority of the cases, intra-chromosomal inversions of chromosome 10 and of chromosome 1 lead to the formation of RET-derived and NTRK1-derived oncogenes, respectively. Exposure to ionizing radiation is associated with papillary carcinomas, and RET activation has been suggested to be related to this event. All these findings are contributing to the definition of genetic and environmental factors relevant to the pathogenesis of thyroid tumours. Moreover, the molecular characterisation of specific genetic lesions could provide significant information about the association between ionising radiation and RET oncogene activation.


Subject(s)
Carcinoma, Papillary/genetics , Drosophila Proteins , Gene Rearrangement , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Nerve Growth Factor/genetics , Thyroid Neoplasms/genetics , Adolescent , Adult , Age Factors , Carcinoma, Papillary/pathology , Child , Child, Preschool , Humans , Infant , Proto-Oncogene Proteins c-ret , Receptor, trkA , Thyroid Neoplasms/pathology
16.
Am J Surg Pathol ; 21(12): 1466-73, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9414190

ABSTRACT

We reclassified 720 nonmedullary invasive thyroid carcinomas diagnosed and treated between 1975 and 1993. Twenty-seven cases met the criteria of insular carcinoma and 29 cases those of widely invasive follicular carcinoma. Comparison of these histotypes with respect to pathologic stage and overall, relative, and visceral metastasis-free survival showed a significant association between histotype and pT and pN categories. In particular, pT4 (p < 0.001) and pN1 (p < 0.001) categories were more frequent in the insular carcinoma histotype. By contrast, no significant differences in overall, relative, or visceral metastasis-free survival were observed between insular carcinoma and widely invasive follicular carcinoma. Molecular analysis by polymerase chain reaction-single-strand conformation polymorphism demonstrated RAS gene family point mutations in five of eight cases analyzed in each of the two histotypes, with a high proportion of CAA-->AAA transversion at codon 61 of the N-RAS gene in insular carcinoma. These findings suggest that insular carcinoma represents a de novo entity distinct from widely invasive follicular carcinoma, that widely invasive follicular carcinoma has biologic characteristics more consistent with poorly differentiated than well-differentiated carcinomas, and that both insular carcinoma and widely invasive follicular carcinoma share similar molecular alterations.


Subject(s)
Adenocarcinoma, Follicular/classification , Adenocarcinoma, Follicular/pathology , Genes, ras , Point Mutation , Thyroid Neoplasms/classification , Thyroid Neoplasms/pathology , Abdominal Neoplasms/classification , Abdominal Neoplasms/pathology , Abdominal Neoplasms/secondary , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/mortality , Adolescent , Adult , Aged , Codon , DNA Primers , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Recurrence , Retrospective Studies , Survival Rate , Thyroid Neoplasms/genetics , Thyroid Neoplasms/mortality , Time Factors
17.
J Clin Endocrinol Metab ; 82(3): 735-8, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9062474

ABSTRACT

Thyroid carcinomas, even when well differentiated, usually appear as hypofunctioning at scintigraphy. We report a case of an aggressive insular thyroid carcinoma presenting as an autonomously functioning thyroid nodule and causing severe thyrotoxicosis. The tumor was metastatic to a cervical lymph node and both lungs. An activating mutation of the TSH receptor gene in both the primary tumor and the lymph node metastasis was found, due to a base substitution at codon 633 (normal guanine at position 1896 replaced by cytosine CAC for GAC causing aspartic acid substitution by histidine). Other known oncogenes (gsp, ras, PTC/ret, trk, met, and p53) were not involved. This is the first description of an activating TSH receptor mutation in a thyroid hyperfunctioning carcinoma in which an aggressive malignant phenotype coexisted with activation of the cAMP cascade and differentiated thyroid functions.


Subject(s)
Carcinoma/genetics , Point Mutation , Receptors, Thyrotropin/genetics , Thyroid Neoplasms/genetics , Base Sequence , Carcinoma/diagnostic imaging , Carcinoma/pathology , Female , Genes , Humans , Lung Neoplasms/secondary , Lymphatic Metastasis , Middle Aged , Polymerase Chain Reaction , Radionuclide Imaging , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/pathology , Thyrotoxicosis/etiology
18.
Mol Endocrinol ; 10(11): 1318-26, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8923458

ABSTRACT

MDA-MB231 human breast cancer cells are unresponsive to insulin and contain a glycoprotein inhibitor of insulin-stimulated insulin receptor (IR) tyrosine kinase activity. Prior studies in both fibroblasts from insulin- resistant non-insulin-dependent diabetes mellitus patients and transfected cells indicate that overexpression of membrane glycoprotein PC-1 reduces IR tyrosine kinase activity. In the present study, we measured PC-1 content and activity in MDA-MB231 and four other human breast cancer cell lines. We observed that PC-1 expression was 3- to 30-fold higher in MDA-MB231 cells when compared with the other breast cell lines. Wheat germ agglutinin extracts of MDA-MB231 cells inhibited IR tyrosine kinase activity. Treatment of these extracts with an antibody to PC-1 significantly reduced their ability to inhibit insulin-stimulated IR tyrosine kinase activity. In addition, when cell clones with different PC-1 activity were selected from MDA-MB231 cells, we found an inverse correlation (r = -0.741, P = 0.006) between the PC-1 activity and the insulin-stimulated IR autophosphorylation. A similar inverse correlation was observed in cell clones derived from the insulin-responsive breast cancer cell line MCF-7. By both immunoprecipitation and cross-linking studies we found PC-1 to be associated with IR. These studies indicate, therefore, that overexpression of PC-1 in MDA-MB231 cells may account, at least in part, for the reduced IR tyrosine kinase activity and suggest that PC-1 is a specific modulator of the IR activity in breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Phosphoric Diester Hydrolases , Pyrophosphatases , Receptor, Insulin/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Division/drug effects , Cell Extracts/pharmacology , Chromatography, Affinity , Clone Cells , Female , Humans , Insulin/pharmacology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/drug effects , Phosphorylation , Receptor, Insulin/drug effects , Receptor, Insulin/isolation & purification , Tumor Cells, Cultured
19.
J Clin Endocrinol Metab ; 81(5): 2006-9, 1996 May.
Article in English | MEDLINE | ID: mdl-8626874

ABSTRACT

Oncogenic rearrangements of RET and NTRK1 proto-oncogenes are frequently detected in papillary thyroid carcinomas. Several studies have shown an association between ionizing radiation and development of this tumor type. In addition in vitro irradiation of tumor cell lines induced rearrangements of RET similar to those observed in human papillary thyroid carcinomas. These two observations could be related to the reported increased incidence of papillary thyroid carcinomas in children living in contaminated areas around Chernobyl, given that it has been demonstrated that about 60% of them presents a RET oncogenic activation. However, this high frequency of RET positivity in radiation exposed children does not rule out the possibility that age could also play a role in the development of RET positive tumors. To assess this possibility we looked for a relationship between the presence of RET and NTRK1 oncogenic rearrangements and age at surgery in a sample of 92 consecutive patients. Our results show that, in papillary thyroid carcinoma, the frequency of RET and NTRK1 activation is significantly higher in the group of patients aged 4-30 years, thas supporting the concept that age could be contributing to this thyroid specific carcinogenic process.


Subject(s)
Aging , Carcinoma, Papillary/genetics , Drosophila Proteins , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Nerve Growth Factor/genetics , Thyroid Neoplasms/genetics , 3T3 Cells , Adolescent , Adult , Animals , Base Sequence , Blotting, Southern , Child , Child, Preschool , Humans , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Proto-Oncogene Proteins c-ret , Receptor, trkA , Transfection
20.
Int J Radiat Oncol Biol Phys ; 11(6): 1209-13, 1985 Jun.
Article in English | MEDLINE | ID: mdl-3997602

ABSTRACT

Irradiation treatment portals of the upper abdomen must limit the dose to the kidneys. Sparing one-third of the parenchyma of each kidney will prevent late clinical sequelae. One hundred CT scans of the abdomen were studied to evaluate using the vertebrae as landmark for treatment planning. In lateral fields, using the anterior border of the vertebral column as a landmark for the posterior high isodose line will limit treatment to less than 60% (mean 22%) of a single kidney. Placing the edge of an anterior/posterior field 2 cm lateral to the vertebral column will limit the dose to less than 44% of a single kidney (mean 11%).


Subject(s)
Kidney/diagnostic imaging , Neoplasms/radiotherapy , Patient Care Planning/methods , Tomography, X-Ray Computed , Adult , Aged , Female , Humans , Kidney/radiation effects , Male , Middle Aged , Radiation Injuries/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...