Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Int J Mol Sci ; 24(15)2023 Jul 30.
Article in English | MEDLINE | ID: mdl-37569595

ABSTRACT

Venoms are a rich source of bioactive compounds, and among them is leberagin-C (Leb-C), a disintegrin-like protein derived from the venom of Macrovipera lebetina transmediterrannea snakes. Leb-C has shown promising inhibitory effects on platelet aggregation. Previous studies have demonstrated that this SECD protein specifically targets α5ß1, αvß3, and αvß6 integrins through a mimic mechanism of RGD disintegrins. In our current study, we focused on exploring the potential effects of Leb-C on metastatic breast cancer. Our findings revealed that Leb-C disrupted the adhesion, migration, and invasion capabilities of MDA-MB-231 breast cancer cells and its highly metastatic D3H2LN sub-population. Additionally, we observed significant suppression of adhesion, migration, and invasion of human umbilical vein endothelial cells (HUVECs). Furthermore, Leb-C demonstrated a strong inhibitory effect on fibroblast-growth-factor-2-induced proliferation of HUVEC. We conducted in vivo experiments using nude mice and found that treatment with 2 µM of Leb-C resulted in a remarkable 73% reduction in D3H2LN xenograft tumor size. Additionally, quantification of intratumor microvessels revealed a 50% reduction in tumor angiogenesis in xenograft after 21 days of twice-weekly treatment with 2 µM of Leb-C. Collectively, these findings suggest the potential utility of this disintegrin-like protein for inhibiting aggressive and resistant metastatic breast cancer.


Subject(s)
Disintegrins , Triple Negative Breast Neoplasms , Animals , Mice , Humans , Disintegrins/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Mice, Nude , Platelet Aggregation , Human Umbilical Vein Endothelial Cells
2.
Endocr Connect ; 9(10): 1042-1050, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33112832

ABSTRACT

OBJECTIVE: We previously described a family in which predisposition to pheochromocytoma (PCC) segregates with a germline heterozygous KIF1B nucleotide variant (c.4442G>A, p.Ser1481Asn) in three generations. During the clinical follow-up, one proband's brother, negative for the KIF1B nucleotide variant, developed a bilateral PCC at 31 years. This prompted us to reconsider the genetic analysis. DESIGN AND METHODS: Germline DNA was analyzed by next-generation sequencing (NGS) using a multi-gene panel plus MLPA or by whole exome sequencing (WES). Tumor-derived DNA was analyzed by SnapShot, Sanger sequencing or NGS to identify loss-of-heterozygosity (LOH) or additional somatic mutations. RESULTS: A germline heterozygous variant of unknown significance in MAX (c.145T>C, p.Ser49Pro) was identified in the proband's brother. Loss of the wild-type MAX allele occurred in his PCCs thus demonstrating that this variant was responsible for the bilateral PCC in this patient. The proband and her affected grandfather also carried the MAX variant but no second hit could be found at the somatic level. No other pathogenic mutations were detected in 36 genes predisposing to familial PCC/PGL or familial cancers by WES of the proband germline. Germline variants detected in other genes, TFAP2E and TMEM214, may contribute to the multiple tumors of the proband. CONCLUSION: In this family, the heritability of PCC is linked to the MAX germline variant and not to the KIF1B germline variant which, however, may have contributed to the occurrence of neuroblastoma (NB) in the proband.

3.
Nutr Rev ; 78(1): 65-76, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31407778

ABSTRACT

Although chemotherapy succeeds in reducing tumor burden, the efficacy is limited due to acquired drug resistance and often irreparable side effects. Studies show that antioxidants may influence the response to chemotherapy and its side effects, although their use remains controversial. The evidence shows that some chemo-drugs induce oxidative stress and lead to normal tissue apoptosis and the entry of cancer cells to a dormant G0 state. Through the suppression of oxidative stress, antioxidants could protect normal cells and bring the tumor out of dormancy so as to expose it to chemotherapies. This review is focused on the redox biology of cancer/normal cells and association of reactive oxygen species with drug resistance, cancer dormancy, and side effects. To this end, evidence from cellular, animal, and clinical studies is provided to better understand the conundrum of dietary antioxidants in cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Diet , Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/prevention & control , Primary Prevention , Reactive Oxygen Species/metabolism
4.
J Clin Endocrinol Metab ; 104(8): 3362-3366, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31046099

ABSTRACT

CONTEXT: Heterozygous germline pathogenic variants found in succinate dehydrogenase (SDH) complex genes predispose to hereditary paraganglioma (PGL) syndromes. No mosaicism has yet been reported in this setting. DESIGN AND PARTICIPANT: We describe the clinical history of a case of SDH complex, subunit B (SDHB) mosaicism. A 24-year-old woman who developed a cardiogenic shock during dental surgery was diagnosed with a functional para-aortic PGL, which produced predominantly norepinephrine and its metabolites. The tumor was removed and showed a loss of SDHB expression by immunohistochemistry. Four years after initial laparotomy, the patient had a rapid cardiac decompensation during her second pregnancy, despite negative imaging 10 months before. Two recurrent functional PGLs were found and surgically removed. Initial genetic analysis performed by Sanger sequencing did not reveal any germline pathogenic variant in SDHB, VHL, SDHD, SDHC, SDHAF2, RET, MAX, and TMEM127. Next-generation sequencing performed on tumor- and blood-extracted DNAs highlighted the presence of a mosaic rare variant in SDHB (c.557G>A, p.Cys186Tyr) with an allelic ratio of 15% in the blood DNA. CONCLUSIONS: We report the full clinical description of a proband with SDHB mosaicism associated with a functional, recurrent PGL. This case strengthens the necessity to complete the genetic analysis with methodologies able to identify germline mosaicism, especially in the case of early disease onset.


Subject(s)
Germ-Line Mutation , Paraganglioma/genetics , Succinate Dehydrogenase/genetics , Female , Humans , Phenotype , Young Adult
5.
J Mol Diagn ; 21(3): 462-470, 2019 05.
Article in English | MEDLINE | ID: mdl-30731206

ABSTRACT

Von Hippel-Lindau disease (VHL) is a monogenic disorder characterized by the development of tumors affecting the central nervous system, kidney, pancreas, or adrenal glands, and due to germline mutations in the VHL tumor suppressor gene. About 5% of patients with a typical VHL phenotype have no mutation detected by conventional techniques, so a postzygotic VHL mosaicism can be suspected. The aim of this study was therefore to implement a next-generation sequencing (NGS) strategy for VHL mosaic mutation detection, including an optimization of the original Personal Genome Machine design by enrichment with oligonucleotides corresponding to amplicons with insufficient depth of coverage. Two complementary strategies were developed for the confirmation of mosaic mutations identified by NGS, SNaPshot for variants present at an allelic ratio greater than 5%, and droplet digital PCR for allelic ratio above 1%. VHL mutant plasmids were generated to assess VHL mosaic mutation detection in different exons and to set up an internal quality control that could be included in each run or regularly to validate the assay. This strategy was applied to 47 patients with a suggestive or clinical VHL disease, and mosaic mutations were identified in 8.5% of patients. In conclusion, NGS technologies combined with SNaPshot or droplet digital PCR allow the detection and confirmation of mosaic mutations in a clinical laboratory setting.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Mosaicism , Mutation/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Adolescent , Adult , Humans , Limit of Detection , Male , Middle Aged , Plasmids/genetics , Reproducibility of Results
7.
Respir Med Case Rep ; 24: 58-62, 2018.
Article in English | MEDLINE | ID: mdl-29977761

ABSTRACT

Alpha-1 antitrypsin deficiency is an autosomal, codominant disorder caused by mutations of the SERPINA1 gene. This genetic disorder is mainly associated with development of pulmonary emphysema and/or chronic liver disease and cirrhosis. Here we report a very rare alpha-1 antitrypsin Null Q0cairo homozygous mutation characterized by a complete absence of alpha-1 antitrypsin in the plasma, in a non-consanguineous Moroccan family. This mutation has been previously described in heterozygosis in only three cases worldwide: an Italian/Egyptian family and two Italian families (Zorzetto et al., 2005). The main clinical features in two members of this Moroccan family were the severity and precocity of bronchiectasis, quickly spreading and seriously limiting respiratory function and physical activity by the second decade of age. Moreover, the index case presented with many episodes of pulmonary infections concomitant with severe neutropenia. The third member of the family presented with ankylosing spondyloarthritis and developed panniculitis later but had no respiratory symptoms. The presence of this alpha-1-antitrypsin Q0cairo homozygous mutation could explain the severity of clinical manifestations. Moreover, our observations highlight a great variability of clinical expression for the same mutation: early severe bronchiectasis, panniculitis, rheumatologic manifestations. This study further underlines the importance of genotyping by whole SERPINA1 gene sequencing in addition to serum alpha-1 antitrypsin determination, to enable detection of alpha-1 antitrypsin deficiency due to rare genotypes.

8.
Eur J Med Genet ; 60(3): 178-184, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28089742

ABSTRACT

A heterozygous germline variant in the HABP2 gene c.1601G > A (p.Gly534Glu), which negatively impacts its tumor suppressive activity in vitro, has been described in 4-14% of kindreds of European-American ancestry with familial papillary thyroid carcinoma (fPTC). But it is also found in ≈4% of Europeans and European/Americans from public databases that, however, did not provide information on the thyroid function of the controls. To get unbiased results, we decided to compare HABP2 genotypes of patients with fPTC with those of "thyroid-checked" controls. A control group consisting of 136 European patients who were thyroidectomised for medullary thyroid carcinoma and devoid of any histologically detectable PTC or follicular-deriving carcinoma was built. In parallel we recruited 20 patients with fPTC from eleven independent European kindreds. The entire coding region of HABP2 was analyzed by Sanger sequencing the germline DNAs of patients. Nucleotide variants were searched for by Snap Shot analysis in the controls. Two variants, c.1601G > A (p.Gly534Glu) and c.364C > T (p.Arg122Trp), were found in 2 and 3 patients at the heterozygous level respectively (minor allele frequency (MAF): 5.0% and 7.5%, respectively). In controls, the MAF was either similar for the c.1601G > A HABP2 variant (2.94%, ns) or significantly lower for the c.364C > T variant (0.73%, p = 0.016). The Arg122 residue lies in the EGF-3 domain of HABP2 which is important for its activation but, however, superposition of the predicted 3D structures of the wild type and mutated proteins suggests that this variant is tolerated at the protein level. In conclusion, our data do not support the pathogenicity of the HABP2 c.1601G > A variant but highlight the existence of a new one that should be more extensively searched for in fPTC patients and its pathogenicity more carefully evaluated.


Subject(s)
Carcinoma, Neuroendocrine/genetics , Carcinoma/genetics , Gene Frequency/genetics , Serine Endopeptidases/genetics , Thyroid Neoplasms/genetics , Aged , Base Sequence , Carcinoma/pathology , Carcinoma, Papillary , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Retrospective Studies , Sequence Analysis, DNA , Thyroid Cancer, Papillary , Thyroid Gland/physiology , Thyroid Neoplasms/pathology
9.
Int J Cancer ; 138(6): 1472-81, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26476272

ABSTRACT

The serrated neoplasia pathway accounts for 20-30% of colorectal cancers (CRC), which are characterized by extensive methylation (CpG island methylation phenotype, CIMP), frequent BRAF mutation and high microsatellite instability (MSI). We recently identified MUC5AC mucin gene hypomethylation as a specific marker of MSI CRC. The early identification of preneoplastic lesions among serrated polyps is currently challenging. Here, we performed a detailed pathological and molecular analysis of a large series of colorectal serrated polyps and evaluated the usefulness of mucin genes MUC2 and MUC5AC to differentiate serrated polyps and to identify lesions with malignant potential. A series of 330 colorectal polyps including 218 serrated polyps [42 goblet cell-rich hyperplastic polyps (GCHP), 68 microvesicular hyperplastic polyps (MVHP), 100 sessile serrated adenoma (SSA) and eight traditional serrated adenoma (TSA)] and 112 conventional adenomas was analyzed for BRAF/KRAS mutations, MSI, CIMP, MLH1 and MGMT methylation, and MUC2 and MUC5AC expression and methylation. We show that MUC5AC hypomethylation is an early event in the serrated neoplasia pathway, and specifically detects MVHP and SSA, arguing for a filiation between MVHP, SSA and CIMP-H/MSI CRC, whereas GCHP and TSA arise from a distinct pathway. Moreover, MUC5AC hypomethylation specifically identified serrated lesions with BRAF mutation, CIMP-H or MSI, suggesting that it may be useful to identify serrated neoplasia pathway-related precursor lesions. Our data suggest that MVHP should be recognized among HP and require particular attention.


Subject(s)
Biomarkers, Tumor , Colonic Polyps/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation , Mucin 5AC/genetics , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/genetics , Colonic Polyps/pathology , Colorectal Neoplasms/diagnosis , Female , Gene Expression , Humans , Male , Microsatellite Instability , Middle Aged , Mutation , Prognosis , Proto-Oncogene Proteins B-raf/genetics , ras Proteins/genetics
10.
Mar Drugs ; 13(5): 2857-74, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25962124

ABSTRACT

Astaxanthin, a powerful antioxidant, is a good candidate for the prevention of intracellular oxidative stress. The aim of the study was to compare the antioxidant activity of astaxanthin present in two natural extracts from Haematococcus pluvialis, a microalgae strain, with that of synthetic astaxanthin. Natural extracts were obtained either by solvent or supercritical extraction methods. UV, HPLC-DAD and (HPLC-(atmospheric pressure chemical ionization (APCI)+)/ion trap-MS) characterizations of both natural extracts showed similar compositions of carotenoids, but different percentages in free astaxanthin and its ester derivatives. The Trolox equivalent antioxidant capacity (TEAC) assay showed that natural extracts containing esters displayed stronger antioxidant activities than free astaxanthin. Their antioxidant capacities to inhibit intracellular oxidative stress were then evaluated on HUVEC cells. The intracellular antioxidant activity in natural extracts was approximately 90-times higher than synthetic astaxanthin (5 µM). No modification, neither in the morphology nor in the viability, of vascular human cells was observed by in vitro biocompatibility study up to 10 µM astaxanthin concentrations. Therefore, these results revealed the therapeutic potential of the natural extracts in vascular human cell protection against oxidative stress without toxicity, which could be exploited in prevention and/or treatment of cardiovascular diseases.


Subject(s)
Endothelial Cells/drug effects , Microalgae/metabolism , Oxidative Stress/drug effects , Antioxidants/pharmacology , Carotenoids/metabolism , Cells, Cultured , Human Umbilical Vein Endothelial Cells , Humans , Xanthophylls/pharmacology
11.
Int J Cancer ; 136(12): 2811-21, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25403854

ABSTRACT

Colorectal cancers (CRC) with microsatellite instability (MSI) display unique clinicopathologic features including a mucinous pattern with frequent expression of the secreted mucins MUC2 and MUC5AC. The mechanisms responsible for this altered pattern of expression remain largely unknown. We quantified DNA methylation of mucin genes (MUC2, MUC5AC, MUC4) in colonic cancers and examined the association with clinicopathological characteristics and molecular (MSI, KRAS, BRAF, and TP53 mutations) features. A control cohort was used for validation. We detected frequent hypomethylation of MUC2 and MUC5AC in CRC. MUC2 and MUC5AC hypomethylation was associated with MUC2 and MUC5AC protein expression (p = 0.004 and p < 0.001, respectively), poor differentiation (p = 0.001 and p = 0.007, respectively) and MSI status (p < 0.01 and p < 0.001, respectively). Interestingly, MUC5AC hypomethylation was specific to MSI cancers. Moreover, it was significantly associated with BRAF mutation and CpG island methylator phenotype (p < 0.001 and p < 0.001, respectively). All these results were confirmed in the control cohort. In the multivariate analysis, MUC5AC hypomethylation was a highly predictive biomarker for MSI cancers. MUC5AC demethylation appears to be a hallmark of MSI in CRC. Determination of MUC5AC methylation status may be useful for understanding and predicting the natural history of CRC.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Microsatellite Instability , Mucin 5AC/genetics , Adult , Aged , Aged, 80 and over , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , CpG Islands/genetics , Decitabine , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Immunohistochemistry , Male , Middle Aged , Mucin 5AC/metabolism , Mutation , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics
12.
Thyroid ; 24(9): 1375-84, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25012490

ABSTRACT

BACKGROUND: The incidence of papillary thyroid carcinoma (PTC) has increased over the past 30 years in Western countries. PTC is usually associated with a good prognosis, but there is a wide range of aggressiveness, and some patients develop distant metastasis and/or resistance to standard treatment. Early identification of these high-risk tumors is a current challenge for appropriate patient management. MUC1 expression has been studied previously in thyroid cancer, but its prognostic value remains controversial. Here, we correlated MUC1 expression in PTC with clinical and pathological features and with the presence of the BRAF(V600E) mutation. METHODS: We performed a clinical and morphological analysis of 190 thyroid tumors (95 PTCs and 95 adenomas). MUC1 immunohistochemistry was carried out on a tissue microarray using different antibodies. The presence of the BRAF(V600E) mutation was investigated by pyrosequencing. MUC1 mRNA levels were assessed by quantitative reverse transcription polymerase chain reaction on a subset of PTC. RESULTS: MUC1 expression was observed in 49% of PTCs and was found to correlate with the presence of papillary architecture, a stromal lymphoid infiltrate, aggressive histological subtypes, extrathyroidal extension, lymph node metastasis, nuclear pseudoinclusions, lymphovascular invasion, and the presence of the BRAF(V600E) mutation (p<0.0001). MUC1 was abundant in nuclear pseudoinclusions. Multivariate analysis showed a strong association of MUC1 expression with the presence of the BRAF(V600E) mutation and lymph node metastasis (p<0.0001). Lymph node metastasis was the most important risk factor of relapse. CONCLUSIONS: Our study shows an association between MUC1 expression and the presence of the BRAF(V600E) mutation in PTC. Analysis of MUC1 expression could improve the risk stratification of PTCs.


Subject(s)
Carcinoma, Papillary/metabolism , Lymphatic Metastasis/genetics , Mucin-1/metabolism , Mutation , Neoplasm Recurrence, Local/genetics , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/metabolism , Adolescent , Adult , Aged , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Child , Female , Humans , Lymphatic Metastasis/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Prognosis , Retrospective Studies , Risk Factors , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Young Adult
13.
Eur J Hum Genet ; 22(9): 1149-52, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24301059

ABSTRACT

The identification of Von Hippel-Lindau (VHL) mosaic mutations by conventional Sanger sequencing requires a labour-intensive enrichment step, thus explaining that mosaicism occurrence is underestimated in patients. Nowadays, it is possible to detect mutation in cell sub-populations by next-generation sequencing (NGS). Here, we described a diagnosis strategy using NGS with high coverage in a series of eight patients who were negative for a VHL abnormality by Sanger sequencing and deletion search. In two patients, a mosaic mutation in VHL was detected by NGS. One patient with a 5.7% mutated allele frequency had a severe phenotype and an early disease onset. In conclusion, clinical NGS in an hospital molecular oncogenetics laboratory is an efficient tool to identify VHL mosaic mutation. Its use may improve patient monitoring and genetic counseling.


Subject(s)
Genetic Testing/methods , High-Throughput Nucleotide Sequencing/methods , Mosaicism , Phenotype , Von Hippel-Lindau Tumor Suppressor Protein/genetics , von Hippel-Lindau Disease/genetics , Adolescent , Adult , Female , Gene Frequency , Humans , Male , Middle Aged , von Hippel-Lindau Disease/diagnosis
14.
Mol Pharmacol ; 82(3): 521-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22723339

ABSTRACT

Statins and bisphosphonates are two distinct classes of isoprenoid pathway inhibitors targeting downstream enzyme to HMG-CoA reductase (upstream enzyme) and farnesyl-pyrophosphate synthase, respectively. Here, we studied fluvastatin (Fluva) and zoledronate (Zol), representative molecules of each class, respectively. In vivo metastatic potentials of both molecules were assessed. For the first time, we observed a significant reduction in progression of established metastases with Fluva treatment. Treatment with both Zol at 100 µg/kg and Fluva at 15 mg/kg inhibited 80% of the metastasis bioluminescence signal and increased survival of mice. The Zol and Fluva transcriptomic profiles of treated MDA-MB-231 cells revealed analogous patterns of affected genes, but each of them reached with different kinetics. The observable changes in gene expression started after 24 h for Fluva IC(50 72 h) and only after 48 h for Zol IC(50 72 h). To obtain early changes in gene expression of Zol-treated cells, a 3 times higher dose of Zol IC(50 72 h) had to be applied. Combining Fluva and Zol in vivo showed no synergy, but a benefit of several days in survival of mice. This study demonstrated that Zol or Fluva is of potential clinical use for the treatment of established metastasis.


Subject(s)
Adenocarcinoma/drug therapy , Breast Neoplasms/drug therapy , Diphosphonates/pharmacology , Fatty Acids, Monounsaturated/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Transcriptome/drug effects , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Female , Fluvastatin , Gene Expression/drug effects , Gene Expression Profiling/methods , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Zoledronic Acid
15.
Hum Mutat ; 33(1): 180-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21953887

ABSTRACT

Constitutional epimutations of DNA mismatch repair (MMR) genes have been recently reported as a possible cause of Lynch syndrome. However, little is known about their prevalence, the risk of transmission through the germline and the risk for carriers to develop cancers. In this study, we evaluated the contribution of constitutional epimutations of MMR genes in Lynch syndrome. A cohort of 134 unrelated Lynch syndrome-suspected patients without MMR germline mutation was screened for constitutional epimutations of MLH1 and MSH2 by quantitative bisulfite pyrosequencing. Patients were also screened for the presence of EPCAM deletions, a possible cause of MSH2 methylation. Tumors from patients with constitutional epimutations were extensively analyzed. We identified a constitutional MLH1 epimutation in two proband patients. For one of them, we report for the first time evidence of transmission to two children who also developed early colonic tumors, indicating that constitutional MLH1 epimutations are associated to a real risk of transgenerational inheritance of cancer susceptibility. Moreover, a somatic BRAF mutation was detected in one affected child, indicating that tumors from patients carrying constitutional MLH1 epimutation can mimic MSI-high sporadic tumors. These findings may have important implications for future diagnostic strategies and genetic counseling.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Epigenesis, Genetic , Genetic Predisposition to Disease , MutS Homolog 2 Protein/genetics , Nuclear Proteins/genetics , Adult , Aged , Child , Cohort Studies , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , DNA Mismatch Repair/genetics , Female , Genetic Testing , Germ-Line Mutation , Heredity , Heterozygote , Humans , Male , Methylation , Middle Aged , MutL Protein Homolog 1 , Pedigree , Proto-Oncogene Proteins B-raf/genetics , Sequence Analysis, DNA
16.
PLoS One ; 6(8): e23334, 2011.
Article in English | MEDLINE | ID: mdl-21858074

ABSTRACT

INTRODUCTION: The poor efficacy of various anti-cancer treatments against metastatic cells has focused attention on the role of tumor microenvironment in cancer progression. To understand the contribution of the extracellular matrix (ECM) environment to this phenomenon, we isolated ECM surrogate invading cell populations from MDA-MB-231 breast cancer cells and studied their genotype and malignant phenotype. METHODS: We isolated invasive subpopulations (INV) from non invasive populations (REF) using a 2D-Matrigel assay, a surrogate of basal membrane passage. INV and REF populations were investigated by microarray assay and for their capacities to adhere, invade and transmigrate in vitro, and to form metastases in nude mice. RESULTS: REF and INV subpopulations were stable in culture and present different transcriptome profiles. INV cells were characterized by reduced expression of cell adhesion and cell-cell junction genes (44% of down regulated genes) and by a gain in expression of anti-apoptotic and pro-angiogenic gene sets. In line with this observation, in vitro INV cells showed reduced adhesion and increased motility through endothelial monolayers and fibronectin. When injected into the circulation, INV cells induced metastases formation, and reduced injected mice survival by up to 80% as compared to REF cells. In nude mice, INV xenografts grew rapidly inducing vessel formation and displaying resistance to apoptosis. CONCLUSION: Our findings reveal that the in vitro ECM microenvironment per se was sufficient to select for tumor cells with a stable metastatic phenotype in vivo characterized by loss of adhesion molecules expression and induction of pro-angiogenic and survival factors.


Subject(s)
Basement Membrane/metabolism , Breast Neoplasms/genetics , Mammary Neoplasms, Experimental/genetics , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Basement Membrane/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion/genetics , Cell Line, Tumor , Collagen , Drug Combinations , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Laminin , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Proteoglycans , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Transendothelial and Transepithelial Migration/genetics , Transplantation, Heterologous , Tumor Burden/genetics
17.
Anticancer Res ; 31(4): 1321-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21508382

ABSTRACT

BACKGROUND/AIM: Both the sulfated and non-sulfated derivatives (e.g. carboxymethyl benzylamide dextrans) of heparan sulfate were reported to have anticancer activity. On this basis, we introduced sulfates and phenyls in carboxymethyl benzylamide dextrans into chitosan, which is easily modified by different functional groups in any given position and then evaluated anticancer activity in breast cancer cells. MATERIALS AND METHODS: Chitosan derivatives were synthesized by introducing sulfate and phenyl groups into chitosan. Cell proliferation and apoptosis were assessed by (3)H-thymidine incorporation and fluorescence activated cell sorter analysis. Activation of Ras/MAPK signaling pathway downstream of fibroblast growth factor-2 (FGF-2) was analyzed by Western blot. RESULTS: The sulfated chitosan (SCS) and the sulfated benzaldehyde chitosan (SBCS) significantly inhibited cell proliferation, induced apoptosis and blocked the FGF-2-induced phosphorylation of ERK in MCF-7 cells, SBCS had better inhibitory effects and a lower IC(50) compared to SCS. CONCLUSION: The sulfated and benzaldehyde chitosans seem to be good potential compounds for anticancer drug design.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Chitosan/chemistry , Chitosan/pharmacology , Blotting, Western , Breast Neoplasms/metabolism , Female , Fibroblast Growth Factor 2/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Tumor Cells, Cultured , ras Proteins/metabolism
18.
Hemoglobin ; 34(4): 389-93, 2010.
Article in English | MEDLINE | ID: mdl-20642337

ABSTRACT

A 37-year-old man presented a slight debility. The hemogram showed a phenotype of beta-thalassemia minor: Hb (13.1 g/dL), mean corpuscular volume (MCV) (62 fL) with low mean corpuscular hemoglobin (MCH) (20.8 pg), associated with a high level of Hb A(2) of 5.3%. The serum ferritin level was 1,072 ng/mL. The sequencing of the mutated fragment revealed a duplication of four bases of codons 7/8 involving a shift in the open reading frame starting from codon 9 with a TGA stop codon at codon 23: codons 7/8/9 (+AGAA); GAG.AAG.TCT(Gly-Lys-Ser)>GAG.AAAGAAG. The human hemoglobin (Hb) instability tests were negative. The patient did not present the high iron Fe (HFE) mutation (C282Y, H63D). The same mutation was found in five other unrelated families (representing a total of 23 patients). All of their ancestors came from the north of France. This mutation has not been described before and could have its origins in the native populations of Northern France.


Subject(s)
Codon/genetics , Mutation , beta-Globins/genetics , beta-Thalassemia/genetics , Adult , Amino Acid Sequence , Base Sequence , DNA Mutational Analysis , Family Health , Female , France , Humans , Male , Middle Aged , Phenotype , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Siblings , beta-Thalassemia/pathology
19.
Biochimie ; 91(8): 1045-52, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19505527

ABSTRACT

This work aimed to investigate the role of the disintegrin domain of the human ADAM9 (ADAM9D) on the adhesion of breast tumor cells and platelets to collagen I, in a dynamic flow assay to simulate in vivo shear conditions. Recombinant ADAM9D was able to support tumor cell adhesion through binding to the beta1 integrin subunit and also to inhibit the invasion through matrigel in vitro. In a dynamic flow assay ADAM9D inhibited about 75% and 65% of MDA-MB-231 tumor cells and platelet adhesion to collagen I, respectively. In addition, it was demonstrated that alphaVbeta3 integrin is new interacting partner for ADAM9D. In conclusion, these results suggest a role for the disintegrin domain of ADAM9 in the metastatic process. Also, ADAM9D may be a tool for investigating the role of ADAMs in metastasis and cancer progression and for the design of selective inhibitors against the adhesion and extravasation of cancer cells.


Subject(s)
ADAM Proteins/chemistry , ADAM Proteins/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen Type I/metabolism , Disintegrins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/pharmacology , ADAM Proteins/genetics , ADAM Proteins/isolation & purification , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cloning, Molecular , Collagen/metabolism , Drug Combinations , Endothelium/metabolism , Humans , Integrin alphaVbeta3/metabolism , Laminin/metabolism , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Protein Structure, Tertiary , Proteoglycans/metabolism , Rats
20.
Anticancer Res ; 29(6): 2347-55, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19528501

ABSTRACT

Mutual interactions between human breast cancer cells and endothelial cells were studied in a model mimicking tumor cell intravasation. MDA-MB-231 tumor cells and human umbilical vein endothelial cells (HUVEC) were cocultured on opposite sides of a Transwell filter allowing tumor cell contacts with the basement membrane of the HUVEC forming endothelium and tumor cell transendothelial migration. Confocal microscopy analysis showed that transmigrating MDA-MB-231 cells lay under the HUVEC, thereby inducing HUVEC detachment and tumor cell-HUVEC contact-dependent apoptosis. GM6001 a matrix metalloproteinase (MMP) inhibitor inhibited almost completely, the MDA-MB-231 cell transendothelial migration and the anoikis process. In this intravasation model, a tumor cell invasive mechanism was demonstrated (i) induction of extensive endothelial anoikis induced by degradation of the extracellular matrix (ECM) components, (ii) activation of pro-matrix metalloproteinase (MMP)-2 into MMP-2 by the MT1-MMP-TIMP (tissue inhibitor metalloproteinase) 2-pro-MMP-2 membrane complex and (iii) attraction and migration of metastatic cell through apoptotic endothelium. These interactions could partly explain the necrosis-angiogenesis relationship in tumor angiogenesis.


Subject(s)
Anoikis , Breast Neoplasms/pathology , Cell Movement , Endothelium, Vascular/pathology , Extracellular Matrix/metabolism , Blotting, Western , Cells, Cultured , Coculture Techniques , Female , Humans , Immunoenzyme Techniques , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinases/metabolism , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...