Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
1.
BMC Cancer ; 23(1): 618, 2023 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-37400763

RÉSUMÉ

BACKGROUND: Gene fusions are important cancer drivers in pediatric cancer and their accurate detection is essential for diagnosis and treatment. Clinical decision-making requires high confidence and precision of detection. Recent developments show RNA sequencing (RNA-seq) is promising for genome-wide detection of fusion products but hindered by many false positives that require extensive manual curation and impede discovery of pathogenic fusions. METHODS: We developed Fusion-sq to overcome existing disadvantages of detecting gene fusions. Fusion-sq integrates and "fuses" evidence from RNA-seq and whole genome sequencing (WGS) using intron-exon gene structure to identify tumor-specific protein coding gene fusions. Fusion-sq was then applied to the data generated from a pediatric pan-cancer cohort of 128 patients by WGS and RNA sequencing. RESULTS: In a pediatric pan-cancer cohort of 128 patients, we identified 155 high confidence tumor-specific gene fusions and their underlying structural variants (SVs). This includes all clinically relevant fusions known to be present in this cohort (30 patients). Fusion-sq distinguishes healthy-occurring from tumor-specific fusions and resolves fusions in amplified regions and copy number unstable genomes. A high gene fusion burden is associated with copy number instability. We identified 27 potentially pathogenic fusions involving oncogenes or tumor-suppressor genes characterized by underlying SVs, in some cases leading to expression changes indicative of activating or disruptive effects. CONCLUSIONS: Our results indicate how clinically relevant and potentially pathogenic gene fusions can be identified and their functional effects investigated by combining WGS and RNA-seq. Integrating RNA fusion predictions with underlying SVs advances fusion detection beyond extensive manual filtering. Taken together, we developed a method for identifying candidate gene fusions that is suitable for precision oncology applications. Our method provides multi-omics evidence for assessing the pathogenicity of tumor-specific gene fusions for future clinical decision making.


Sujet(s)
Tumeurs , Enfant , Humains , Tumeurs/génétique , RNA-Seq , Séquençage nucléotidique à haut débit/méthodes , Médecine de précision , Analyse de séquence d'ARN/méthodes , Fusion de gènes , Séquençage du génome entier
2.
Cancers (Basel) ; 14(19)2022 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-36230794

RÉSUMÉ

Chromosomal alterations have recurrently been identified in Wilms tumors (WTs) and some are associated with poor prognosis. Gain of 1q (1q+) is of special interest given its high prevalence and is currently actively studied for its prognostic value. However, the underlying mutational mechanisms and functional effects remain unknown. In a national unbiased cohort of 30 primary WTs, we integrated somatic SNVs, CNs and SVs with expression data and distinguished four clusters characterized by affected biological processes: muscle differentiation, immune system, kidney development and proliferation. Combined genome-wide CN and SV profiles showed that tumors profoundly differ in both their types of 1q+ and genomic stability and can be grouped into WTs with co-occurring 1p-/1q+, multiple chromosomal gains or CN neutral tumors. We identified 1q+ in eight tumors that differ in mutational mechanisms, subsequent rearrangements and genomic contexts. Moreover, 1q+ tumors were present in all four expression clusters reflecting activation of various biological processes, and individual tumors overexpress different genes on 1q. In conclusion, by integrating CNs, SVs and gene expression, we identified subgroups of 1q+ tumors reflecting differences in the functional effect of 1q gain, indicating that expression data is likely needed for further risk stratification of 1q+ WTs.

3.
Eur J Cancer ; 175: 311-325, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36182817

RÉSUMÉ

iTHER is a Dutch prospective national precision oncology program aiming to define tumour molecular profiles in children and adolescents with primary very high-risk, relapsed, or refractory paediatric tumours. Between April 2017 and April 2021, 302 samples from 253 patients were included. Comprehensive molecular profiling including low-coverage whole genome sequencing (lcWGS), whole exome sequencing (WES), RNA sequencing (RNA-seq), Affymetrix, and/or 850k methylation profiling was successfully performed for 226 samples with at least 20% tumour content. Germline pathogenic variants were identified in 16% of patients (35/219), of which 22 variants were judged causative for a cancer predisposition syndrome. At least one somatic alteration was detected in 204 (90.3%), and 185 (81.9%) were considered druggable, with clinical priority very high (6.1%), high (21.3%), moderate (26.0%), intermediate (36.1%), and borderline (10.5%) priority. iTHER led to revision or refinement of diagnosis in 8 patients (3.5%). Temporal heterogeneity was observed in paired samples of 15 patients, indicating the value of sequential analyses. Of 137 patients with follow-up beyond twelve months, 21 molecularly matched treatments were applied in 19 patients (13.9%), with clinical benefit in few. Most relevant barriers to not applying targeted therapies included poor performance status, as well as limited access to drugs within clinical trial. iTHER demonstrates the feasibility of comprehensive molecular profiling across all ages, tumour types and stages in paediatric cancers, informing of diagnostic, prognostic, and targetable alterations as well as reportable germline variants. Therefore, WES and RNA-seq is nowadays standard clinical care at the Princess Máxima Center for all children with cancer, including patients at primary diagnosis. Improved access to innovative treatments within biology-driven combination trials is required to ultimately improve survival.


Sujet(s)
Tumeurs , Adolescent , Enfant , Séquençage nucléotidique à haut débit , Humains , Oncologie médicale , Mutation , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Médecine de précision , Études prospectives ,
4.
JCO Precis Oncol ; 6: e2000504, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-35085008

RÉSUMÉ

PURPOSE: Gene fusions play a significant role in cancer etiology, making their detection crucial for accurate diagnosis, prognosis, and determining therapeutic targets. Current diagnostic methods largely focus on either targeted or low-resolution genome-wide techniques, which may be unable to capture rare events or both fusion partners. We investigate if RNA sequencing can overcome current limitations with traditional diagnostic techniques to identify gene fusion events. METHODS: We first performed RNA sequencing on a validation cohort of 24 samples with a known gene fusion event, after which a prospective pan-pediatric cancer cohort (n = 244) was tested by RNA sequencing in parallel to existing diagnostic procedures. This cohort included hematologic malignancies, tumors of the CNS, solid tumors, and suspected neoplastic samples. All samples were processed in the routine diagnostic workflow and analyzed for gene fusions using standard-of-care methods and RNA sequencing. RESULTS: We identified a clinically relevant gene fusion in 83 of 244 cases in the prospective cohort. Sixty fusions were detected by both routine diagnostic techniques and RNA sequencing, and one fusion was detected only in routine diagnostics, but an additional 24 fusions were detected solely by RNA sequencing. RNA sequencing, therefore, increased the diagnostic yield by 38%-39%. In addition, RNA sequencing identified both gene partners involved in the gene fusion, in contrast to most routine techniques. For two patients, the newly identified fusion by RNA sequencing resulted in treatment with targeted agents. CONCLUSION: We show that RNA sequencing is sufficiently robust for gene fusion detection in routine diagnostics of childhood cancers and can make a difference in treatment decisions.


Sujet(s)
Fusion de gènes , Tumeurs/diagnostic , Tumeurs/génétique , Analyse de séquence d'ARN , Enfant , Humains , Études prospectives
5.
Sci Rep ; 5: 14060, 2015 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-26358404

RÉSUMÉ

Bloom syndrome is an autosomal recessive disorder characterized by chromosomal instability and increased cancer risk, caused by biallelic mutations in the RECQL-helicase gene BLM. Previous studies have led to conflicting conclusions as to whether carriers of heterozygous BLM mutations have an increased risk to develop colorectal cancer (CRC). We recently identified two carriers of a pathogenic BLM mutation in a cohort of 55 early-onset CRC patients (≤45 years of age), suggesting an overrepresentation compared to the normal population. Here, we performed targeted sequencing using molecular inversion probes to screen an additional cohort of 185 CRC patients (≤50 years of age) and 532 population-matched controls for deleterious BLM mutations. In total, we identified three additional CRC patients (1.6%) and one control individual (0.2%) that carried a known pathogenic BLM mutation, suggesting that these mutations are enriched in early-onset CRC patients (P = 0.05516). A comparison with local and publically available databases from individuals without suspicion for hereditary cancer confirmed this enrichment (P = 0.003534). Analysis of family members of the five BLM mutation carriers with CRC suggests an incomplete penetrance for CRC development. Therefore, these data indicate that carriers of deleterious BLM mutations are at increased risk to develop CRC, albeit with a moderate-to-low penetrance.


Sujet(s)
Tumeurs colorectales/épidémiologie , Tumeurs colorectales/génétique , Mutation germinale , RecQ helicases/génétique , Adulte , Âge de début , Études cas-témoins , Jeux de données comme sujet , Exome , Femelle , Fréquence d'allèle , Étude d'association pangénomique , Hétérozygote , Séquençage nucléotidique à haut débit , Humains , Perte d'hétérozygotie , Mâle , Taux de mutation , Pedigree , Surveillance de la population , Risque
6.
Nat Genet ; 47(6): 668-71, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25938944

RÉSUMÉ

The genetic cause underlying the development of multiple colonic adenomas, the premalignant precursors of colorectal cancer (CRC), frequently remains unresolved in patients with adenomatous polyposis. Here we applied whole-exome sequencing to 51 individuals with multiple colonic adenomas from 48 families. In seven affected individuals from three unrelated families, we identified a homozygous germline nonsense mutation in the base-excision repair (BER) gene NTHL1. This mutation was exclusively found in a heterozygous state in controls (minor allele frequency of 0.0036; n = 2,329). All three families showed recessive inheritance of the adenomatous polyposis phenotype and progression to CRC in at least one member. All three affected women developed an endometrial malignancy or premalignancy. Genetic analysis of three carcinomas and five adenomas from different affected individuals showed a non-hypermutated profile enriched for cytosine-to-thymine transitions. We conclude that a homozygous loss-of-function germline mutation in the NTHL1 gene predisposes to a new subtype of BER-associated adenomatous polyposis and CRC.


Sujet(s)
Polypose adénomateuse colique/génétique , Deoxyribonuclease (pyrimidine dimer)/génétique , Études cas-témoins , Codon non-sens , Analyse de mutations d'ADN , Réparation de l'ADN , Femelle , Études d'associations génétiques , Prédisposition génétique à une maladie , Mutation germinale , Homozygote , Humains , Adulte d'âge moyen , Pedigree
7.
Mol Cytogenet ; 8: 33, 2015.
Article de Anglais | MEDLINE | ID: mdl-26023320

RÉSUMÉ

BACKGROUND: Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematological malignancies. In MDS patients with a fibrotic bone marrow the aspiration of cells often fails (dry-tap), which hampers standard karyotyping. Obtaining genetic data from these fibrotic marrows is therefore challenging, and up till now in situ hybridization applied to bone marrow biopsies is the only option. The microarray-based genomic profiling technology has already proven its value for bone marrow aspirates and peripheral blood samples, but has never been applied to the technically challenging bone marrow biopsies. We describe an approach for microarray-based genomic profiling on bone marrow biopsies and demonstrate its ability to obtain clinically relevant cytogenetic aberrations. In addition the data were compared with those obtained by in situ hybridization and karyotyping. RESULTS: We have evaluated the success rate of microarray-based genomic profiling by studying twenty-one bone marrow biopsies (7 fibrotic MDS, 12 non-fibrotic MDS and 2 reactive), by microarray-based genomic profiling and in situ hybridization (12 of 21 cases). The data obtained with these techniques were compared with conventional karyotyping data on corresponding bone marrow aspirates. Of the 15 copy number aberrations that were detected by in situ hybridization, 13 were concordant with microarray-based genomic profiling and karyotyping, whereas two hybridizations were misinterpreted. In 20 of 21 patients, the data obtained by microarray-based genomic profiling and karyotyping were identical or differences could be explained by the presence of marker chromosomes, complex karyotypes, clonal heterogeneity or disease progression. CONCLUSIONS: We demonstrate that genome wide microarray-based genomic profiling performed on bone marrow biopsies has a similar success rate compared to in situ hybridization, and prevents misinterpretation of chromosomal losses as observed by FISH. In addition, equal to even higher resolutions were obtained with genomic profiling compared to conventional karyotyping. Our findings indicate that microarray-based profiling, even on bone marrow biopsies, is a valid approach for the identification of genetic abnormalities. This is a valuable substitution in cases of fibrotic MDS lacking cytogenetic results.

8.
J Pathol ; 236(2): 155-64, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25712196

RÉSUMÉ

Heritable genetic variants can significantly affect the lifetime risk of developing cancer, including polyposis and colorectal cancer (CRC). Variants in genes currently known to be associated with a high risk for polyposis or CRC, however, explain only a limited number of hereditary cases. The identification of additional genetic causes is, therefore, crucial to improve CRC prevention, detection and treatment. We have performed genome-wide and targeted DNA copy number profiling and resequencing in early-onset and familial polyposis/CRC patients, and show that deletions affecting the open reading frame of the tumour suppressor gene FOCAD are recurrent and significantly enriched in CRC patients compared with unaffected controls. All patients carrying FOCAD deletions exhibited a personal or family history of polyposis. RNA in situ hybridization revealed FOCAD expression in epithelial cells in the colonic crypt, the site of tumour initiation, as well as in colonic tumours and organoids. Our data suggest that monoallelic germline deletions in the tumour suppressor gene FOCAD underlie moderate genetic predisposition to the development of polyposis and CRC.


Sujet(s)
Polypose adénomateuse colique/génétique , Tumeurs colorectales/génétique , Délétion de gène , Mutation germinale/génétique , Protéines suppresseurs de tumeurs/génétique , Polypose adénomateuse colique/métabolisme , Adulte , Études cas-témoins , Chromosomes humains de la paire 9/génétique , Tumeurs colorectales/métabolisme , Variations de nombre de copies de segment d'ADN/génétique , Cellules épithéliales/métabolisme , Femelle , Régulation de l'expression des gènes tumoraux/génétique , Hétérozygote , Humains , Mâle , Adulte d'âge moyen , Récidive tumorale locale/génétique , Cadres ouverts de lecture/génétique , Protéines suppresseurs de tumeurs/métabolisme
9.
Eur J Med Genet ; 56(9): 471-4, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23851227

RÉSUMÉ

The European Cytogeneticists Association Register of Unbalanced Chromosome Aberrations (ECARUCA, www.ecaruca.net) is an online database initiated in 2003 that collects and provides detailed, curated clinical and molecular information on rare unbalanced chromosome aberrations. ECARUCA now contains over 4800 cases with a total of more than 6600 genetic aberrations and has over 3000 account holders worldwide. Recently, the ECARUCA web site was renewed, including the presentation of interesting case reports in collaboration with the European Journal of Medical Genetics. This article gives an overview of the current status and future plans of the online ECARUCA database.


Sujet(s)
Aberrations des chromosomes , Bases de données d'acides nucléiques , Europe , Génome humain , Humains , Systèmes en direct , Enregistrements
10.
Gastroenterology ; 145(3): 544-7, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23747338

RÉSUMÉ

The spindle assembly checkpoint controls proper chromosome segregation during mitosis and prevents aneuploidy-an important feature of cancer cells. We performed genome-wide and targeted copy number and mutation analyses of germline DNA from 208 patients with familial or early-onset (40 years of age or younger) colorectal cancer; we identified haploinsufficiency or heterozygous mutations in the spindle assembly checkpoint genes BUB1 and BUB3 in 2.9% of them. Besides colorectal cancer, these patients had variegated aneuploidies in multiple tissues and variable dysmorphic features. These results indicate that mutations in BUB1 and BUB3 cause mosaic variegated aneuploidy and increase the risk of colorectal cancer at a young age.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Protéines du cycle cellulaire/génétique , Tumeurs colorectales/génétique , Mutation germinale , Protein-Serine-Threonine Kinases/génétique , Adulte , Marqueurs biologiques tumoraux/métabolisme , Études cas-témoins , Protéines du cycle cellulaire/métabolisme , Tumeurs colorectales/métabolisme , Étude d'association pangénomique , Humains , Protéines liant le poly-adp-ribose , Protein-Serine-Threonine Kinases/métabolisme , Facteurs de risque
11.
Nat Commun ; 4: 1342, 2013.
Article de Anglais | MEDLINE | ID: mdl-23299892

RÉSUMÉ

Candida albicans is the most common human fungal pathogen causing mucosal and systemic infections. However, human antifungal immunity remains poorly defined. Here by integrating transcriptional analysis and functional genomics, we identified Candida-specific host defence mechanisms in humans. Candida induced significant expression of genes from the type I interferon pathway in human peripheral blood mononuclear cells. This unexpectedly prominent role of type I interferon pathway in anti-Candida host defence was supported by additional evidence. Polymorphisms in type I interferon genes modulated Candida-induced cytokine production and were correlated with susceptibility to systemic candidiasis. In in vitro experiments, type I interferons skewed Candida-induced inflammation from a Th17 response towards a Th1 response. Patients with chronic mucocutaneous candidiasis displayed defective expression of genes in the type I interferon pathway. These findings indicate that the type I interferon pathway is a main signature of Candida-induced inflammation and has a crucial role in anti-Candida host defence in humans.


Sujet(s)
Candida albicans/immunologie , Génomique , Interactions hôte-pathogène/génétique , Interactions hôte-pathogène/immunologie , Interféron de type I/génétique , Interféron de type I/immunologie , Transduction du signal/génétique , Candidémie/génétique , Candidémie/immunologie , Candidémie/microbiologie , Candidose mucocutanée chronique/génétique , Candidose mucocutanée chronique/immunologie , Candidose mucocutanée chronique/microbiologie , Études cas-témoins , Analyse de regroupements , Régulation de l'expression des gènes , Prédisposition génétique à une maladie , Humains , Mutation/génétique , Polymorphisme de nucléotide simple/génétique , Facteur de transcription STAT-1/génétique , Transduction du signal/immunologie , Lymphocytes auxiliaires Th1/immunologie , Cellules Th17/immunologie , Transcription génétique
12.
Exp Physiol ; 98(3): 710-21, 2013 Mar.
Article de Anglais | MEDLINE | ID: mdl-23064509

RÉSUMÉ

Physical inactivity and exercise training result in opposite adaptations of vascular structure. However, the molecular mechanisms behind these adaptations are not completely understood. We used a unique study design to examine both vascular characteristics of the superficial femoral artery (using ultrasound) and gene expression levels (from a muscle biopsy) in human models for physical deconditioning and exercise training. Initially, we compared able-bodied control subjects (n = 6) with spinal cord-injured individuals (n = 8) to assess the effects of long-term deconditioning. Subsequently, able-bodied control subjects underwent short-term lower limb deconditioning using 3 weeks of unilateral limb suspension. Spinal cord-injured individuals were examined before and after 6 weeks of functional electrical stimulation exercise training. Baseline femoral artery diameter and hyperaemic flow were lower after short- and long-term deconditioning and higher after exercise training, whilst intima-media thickness/lumen ratio was increased with short- and long-term deconditioning and decreased with exercise training. Regarding gene expression levels of vasculature-related genes, we found that groups of genes including the vascular endothelial growth factor pathway, transforming growth factor ß1 and extracellular matrix proteins were strongly associated with vascular adaptations in humans. This approach resulted in the identification of important genes that may be involved in vascular adaptations after physical deconditioning and exercise.


Sujet(s)
Adaptation physiologique/physiologie , Exercice physique/physiologie , Suspension des membres postérieurs/physiologie , Voies et réseaux métaboliques/génétique , Aptitude physique/physiologie , Traumatismes de la moelle épinière/physiopathologie , Adulte , Épaisseur intima-média carotidienne , Stimulation électrique , Protéines de la matrice extracellulaire/génétique , Artère fémorale/anatomie et histologie , Humains , Mâle , Muscles squelettiques/physiologie , Traumatismes de la moelle épinière/thérapie , Transcriptome/physiologie , Facteur de croissance transformant bêta-1/génétique , Facteur de croissance endothéliale vasculaire de type A/génétique , Jeune adulte
13.
Stem Cells Dev ; 22(7): 1086-96, 2013 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-23145933

RÉSUMÉ

Bone marrow (BM) stromal cells (MSCs), also known as mesenchymal stem cells, display a high degree of heterogeneity. To shed light on the causes of this heterogeneity, MSCs were collected from either human BM (n=5) or adipose tissue (AT) (n=5), and expanded using 2 different culture methods: one based on fetal calf serum, and one based on human platelet lysate. After initial expansion, MSCs were frozen, and the vials were transported to 3 different laboratories and grown for 1 passage using the same brand of culture plastic, medium, and supplements. Subsequently, the cells were harvested and assayed for their gene expression profile using the Affymetrix exon microarray platform. Based on gene expression profiles, the most discriminative feature was the anatomical harvesting site, followed by culture methodology. Remarkably, genes in the WNT pathway were expressed at higher levels in BM-derived MSCs than in AT-derived MSCs. Although differences were found between laboratories, cell culture location only slightly affects heterogeneity. Furthermore, individual donors contributed marginally to the observed differences in transcriptomes. Finally, BM-derived MSCs displayed the highest level of similarity, irrespective their culture conditions, when compared to AT-derived cells.


Sujet(s)
Tissu adipeux/cytologie , Cellules de la moelle osseuse/cytologie , Techniques de culture cellulaire , Analyse de profil d'expression de gènes , Cellules souches mésenchymateuses/cytologie , Adipocytes/cytologie , Tissu adipeux/métabolisme , Cellules de la moelle osseuse/métabolisme , Différenciation cellulaire , Cellules cultivées , Humains , Cellules souches mésenchymateuses/métabolisme , Voie de signalisation Wnt/génétique
14.
Nat Genet ; 44(6): 639-41, 2012 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-22544363

RÉSUMÉ

We show that haploinsufficiency of KANSL1 is sufficient to cause the 17q21.31 microdeletion syndrome, a multisystem disorder characterized by intellectual disability, hypotonia and distinctive facial features. The KANSL1 protein is an evolutionarily conserved regulator of the chromatin modifier KAT8, which influences gene expression through histone H4 lysine 16 (H4K16) acetylation. RNA sequencing studies in cell lines derived from affected individuals and the presence of learning deficits in Drosophila melanogaster mutants suggest a role for KANSL1 in neuronal processes.


Sujet(s)
Malformations multiples/génétique , Délétion de segment de chromosome , Protéines nucléaires/génétique , Sujet âgé , Vieillissement , Chromosomes humains de la paire 17 , Faciès , Femelle , Haploinsuffisance , Humains , Déficience intellectuelle/génétique , Mâle , Adulte d'âge moyen , Mutation , Syndrome de Smith-Magenis , Syndrome
15.
Antioxid Redox Signal ; 17(12): 1657-69, 2012 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-22559215

RÉSUMÉ

AIMS: Cell regulation by signaling reactive oxygen species (sROS) is often incorrectly studied through extracellular oxidant addition. Here, we used the membrane-permeable antioxidant Trolox to examine the role of sROS in mitochondrial morphology, oxidative phosphorylation (OXPHOS), and cytosolic calcium (Ca(2+)) handling in healthy human skin fibroblasts. RESULTS AND INNOVATION: Trolox treatment reduced the levels of 5-(and-6)-chloromethyl-2',7'-dichlorodihydro-fluorescein (CM-H(2)DCF) oxidizing ROS, lowered cellular lipid peroxidation, and induced a less oxidized mitochondrial thiol redox state. This was paralleled by increased glutathione- and mitofusin-dependent mitochondrial filamentation, increased expression of fully assembled mitochondrial complex I, elevated activity of citrate synthase and OXPHOS enzymes, and a higher cellular O(2) consumption. In contrast, Trolox did not alter hydroethidium oxidation, cytosolic thiol redox state, mitochondrial NAD(P)H levels, or mitochondrial membrane potential. Whole genome expression profiling revealed that Trolox did not trigger significant changes in gene expression, suggesting that Trolox acts downstream of this process. Cytosolic Ca(2+) transients, induced by the hormone bradykinin, were of a higher amplitude and decayed faster in Trolox-treated cells. These effects were dose-dependently antagonized by hydrogen peroxide. CONCLUSIONS: Our findings suggest that Trolox-sensitive sROS are upstream regulators of mitochondrial mitofusin levels, morphology, and function in healthy human skin fibroblasts. This information not only facilitates the interpretation of antioxidant effects in cell models (of oxidative-stress), but also contributes to a better understanding of ROS-related human pathologies, including mitochondrial disorders.


Sujet(s)
Calcium/métabolisme , Chromanes/pharmacologie , Cytosol/métabolisme , Mitochondries/métabolisme , Espèces réactives de l'oxygène/métabolisme , Animaux , Technique de Western , Cellules CHO , Cellules cultivées , Cricetinae , Cellules HeLa , Humains , Peroxydation lipidique/effets des médicaments et des substances chimiques , Phosphorylation oxydative/effets des médicaments et des substances chimiques
16.
Macromol Biosci ; 12(5): 675-91, 2012 May.
Article de Anglais | MEDLINE | ID: mdl-22416023

RÉSUMÉ

A papillary-structured collagen fibril membrane is created, mimicking the 3D-architecture of the human papillary dermis. Primary human keratinocytes cultured to confluency on papillar-structured films are compared to keratinocytes cultured on flat membranes. Microscopical evaluation reveals the presence of morphologically distinct cells at the base of the papillar structures that are not observed on flat membranes. Gene expression microarrays and RT-qPCR indicate that these cells are in a more proliferative/migrational state, whereas cells on flat membranes have a more differentiated expression profile. Immunohistochemical stainings confirm these results. In conclusion, specific collagen architecture can direct keratinocyte behavior, and this may be used to further improve skin regeneration.


Sujet(s)
Matériaux biomimétiques/composition chimique , Collagène/composition chimique , Expression des gènes/effets des médicaments et des substances chimiques , Kératinocytes/effets des médicaments et des substances chimiques , Matériaux biomimétiques/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Collagène/pharmacologie , Collagène/ultrastructure , Derme/cytologie , Derme/effets des médicaments et des substances chimiques , Derme/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/métabolisme , Analyse de profil d'expression de gènes , Humains , Immunohistochimie , Kératinocytes/cytologie , Kératinocytes/métabolisme , Membrane artificielle , Microscopie électronique à balayage , Séquençage par oligonucléotides en batterie , Culture de cellules primaires , Structures d'échafaudage tissulaires , Cicatrisation de plaie/physiologie
17.
Am J Hum Genet ; 90(3): 426-33, 2012 Mar 09.
Article de Anglais | MEDLINE | ID: mdl-22341970

RÉSUMÉ

Revertant mosaicism is an infrequently observed phenomenon caused by spontaneous correction of a pathogenic allele. We have observed such reversions caused by mitotic recombination of mutant TERC (telomerase RNA component) alleles in six patients from four families affected by dyskeratosis congenita (DC). DC is a multisystem disorder characterized by mucocutaneous abnormalities, dystrophic nails, bone-marrow failure, lung fibrosis, liver cirrhosis, and cancer. We identified a 4 nt deletion in TERC in a family with an autosomal-dominant form of DC. In two affected brothers without bone-marrow failure, sequence analysis revealed pronounced overrepresentation of the wild-type allele in blood cells, whereas no such skewing was observed in the other tissues tested. These observations suggest that this mosaic pattern might have resulted from somatic reversion of the mutated allele to the normal allele in blood-forming cells. SNP-microarray analysis on blood DNA from the two brothers indeed showed independent events of acquired segmental isodisomy of chromosome 3q, including TERC, indicating that the reversions must have resulted from mitotic recombination events. Subsequently, after developing a highly sensitive method of detecting mosaic homozygosity, we have found four additional cases with a mosaic-reversion pattern in blood cells; these four cases are part of a cohort of 17 individuals with germline TERC mutations. This shows that revertant mosaicism is a recurrent event in DC. This finding has important implications for improving diagnostic testing and understanding the variable phenotype of DC.


Sujet(s)
Dyskératose congénitale/génétique , Mitose/génétique , Mosaïcisme , ARN/génétique , Recombinaison génétique , Telomerase/génétique , Adolescent , Adulte , Sujet âgé , Allèles , Lignage cellulaire , Enfant , Enfant d'âge préscolaire , Études de cohortes , Femelle , Mutation germinale , Homozygote , Humains , Mâle , Adulte d'âge moyen , Pedigree , Polymorphisme de nucléotide simple , Analyse de séquence d'ADN/méthodes , Jeune adulte
18.
Lancet Oncol ; 12(1): 49-55, 2011 Jan.
Article de Anglais | MEDLINE | ID: mdl-21145788

RÉSUMÉ

BACKGROUND: Lynch syndrome is caused by germline mutations in MSH2, MLH1, MSH6, and PMS2 mismatch-repair genes and leads to a high risk of colorectal and endometrial cancer. We previously showed that constitutional 3' end deletions of EPCAM can cause Lynch syndrome through epigenetic silencing of MSH2 in EPCAM-expressing tissues, resulting in tissue-specific MSH2 deficiency. We aim to establish the risk of cancer associated with such EPCAM deletions. METHODS: We obtained clinical data for 194 carriers of a 3' end EPCAM deletion from 41 families known to us at the Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands and compared cancer risk with data from a previously described cohort of 473 carriers from 91 families with mutations in MLH1, MSH2, MSH6, or a combined EPCAM-MSH2 deletion. FINDINGS: 93 of the 194 EPCAM deletion carriers were diagnosed with colorectal cancer; three of the 92 women with EPCAM deletions were diagnosed with endometrial cancer. Carriers of an EPCAM deletion had a 75% (95% CI 65-85) cumulative risk of colorectal cancer before the age of 70 years (mean age at diagnosis 43 years [SD 12]), which did not differ significantly from that of carriers of combined EPCAM-MSH2 deletion (69% [95% CI 47-91], p=0·8609) or mutations in MSH2 (77% [64-90], p=0·5892) or MLH1 (79% [68-90], p=0·5492), but was higher than noted for carriers of MSH6 mutation (50% [38-62], p<0·0001). By contrast, women with EPCAM deletions had a 12% [0-27] cumulative risk of endometrial cancer, which was lower than was that noted for carriers of a combined EPCAM-MSH2 deletion (55% [20-90], p<0·0001) or of a mutation in MSH2 (51% [33-69], p=0·0006) or MSH6 (34% [20-48], p=0·0309), but did not differ significantly from that noted for MLH1 (33% [15-51], p=0·1193) mutation carriers. This risk seems to be restricted to deletions that extend close to the MSH2 gene promoter. Of 194 carriers of an EPCAM deletion, three had duodenal cancer and four had pancreatic cancer. INTERPRETATION: EPCAM deletion carriers have a high risk of colorectal cancer; only those with deletions extending close to the MSH2 promoter have an increased risk of endometrial cancer. These results underscore the effect of mosaic MSH2 deficiency, leading to variable cancer risks, and could form the basis of an optimised protocol for the recognition and targeted prevention of cancer in EPCAM deletion carriers.


Sujet(s)
Antigènes néoplasiques/génétique , Molécules d'adhérence cellulaire/génétique , Tumeurs colorectales/génétique , Tumeurs de l'endomètre/génétique , Délétion de séquence , Adolescent , Adulte , Sujet âgé , Études de cohortes , Tumeurs colorectales/étiologie , Tumeurs de l'endomètre/étiologie , Molécule d'adhérence des cellules épithéliales , Femelle , Délétion de gène , Humains , Mâle , Adulte d'âge moyen , Protéine-2 homologue de MutS/génétique , Régions promotrices (génétique) , Risque
19.
Cell Transplant ; 20(6): 925-40, 2011.
Article de Anglais | MEDLINE | ID: mdl-21054934

RÉSUMÉ

The aim of this study was to evaluate the potential of culture-expanded human auricular and nasoseptal chondrocytes as cell source for regeneration of stable cartilage and to analyze the differences in gene expression profile of expanded chondrocytes from these specific locations. Auricular chondrocytes in monolayer proliferated less and more slowly (two passages took 26.7 ± 2.1 days and were reached in 4.37 ± 0.30 population doublings) than nasoseptal chondrocytes (19.3 ± 2.5 days; 5.45 ± 0.20 population doublings). However, auricular chondrocytes produced larger pellets with more cartilage-like matrix than nasoseptal chondrocytes (2.2 ± 0.71 vs. 1.7 ± 0.13 mm in diameter after 35 days of culture). Although the matrix formed by auricular and nasoseptal chondrocytes contained collagen X, it did not mineralize in an in vitro model or after in vivo subcutaneous implantation. A DNA microarray study on expanded auricular and nasoseptal chondrocytes from the same donors revealed 1,090 differentially expressed genes. No difference was observed in the expression of known markers of chondrogenic capacity (e.g., collagen II, FGFR3, BMP2, and ALK1). The most striking differences were that the auricular chondrocytes had a higher expression of anabolic growth factors BMP5 and IGF1, while matrix-degrading enzymes MMP13 and ADAMTS5 were higher expressed in nasoseptal chondrocytes. This might offer a possible explanation for the observed higher matrix production by auricular chondrocytes. Moreover, chondrocytes isolated from auricular or nasoseptal cartilage had specific gene expression profiles even after expansion. These differently expressed genes were not restricted to known characterization of donor site subtype (e.g., elastic), but were also related to developmental processes.


Sujet(s)
Cartilage articulaire/cytologie , Chondrocytes/métabolisme , Cartilage de l'oreille/cytologie , Analyse de profil d'expression de gènes , Septum nasal/cytologie , Protéines ADAM/génétique , Protéines ADAM/métabolisme , Protéine ADAMTS5 , Adolescent , Adulte , Sujet âgé , Protéine morphogénétique osseuse de type 5/génétique , Protéine morphogénétique osseuse de type 5/métabolisme , Cartilage articulaire/métabolisme , Enfant , Enfant d'âge préscolaire , Chondrogenèse , Collagène de type X/métabolisme , Cartilage de l'oreille/métabolisme , Humains , Facteur de croissance IGF-I/génétique , Facteur de croissance IGF-I/métabolisme , Matrix Metalloproteinase 13/génétique , Matrix Metalloproteinase 13/métabolisme , Adulte d'âge moyen , Septum nasal/métabolisme , Séquençage par oligonucléotides en batterie , Ingénierie tissulaire
20.
Int J Cancer ; 129(7): 1635-42, 2011 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-21128281

RÉSUMÉ

In the majority of colorectal cancers (CRCs) under clinical suspicion for a hereditary cause, the disease-causing genetic factors are still to be discovered. To identify such genetic factors we stringently selected a discovery cohort of 41 CRC index patients with microsatellite-stable tumors. All patients were below 40 years of age at diagnosis and/or exhibited an overt family history. We employed genome-wide copy number profiling using high-resolution SNP arrays on germline DNA, which resulted in the identification of novel copy number variants (CNVs) in six patients (15%) encompassing, among others, the cadherin gene CDH18, the bone morphogenetic protein antagonist family gene GREM1, and the breakpoint cluster region gene BCR. In addition, two genomic deletions were encountered encompassing two microRNA genes, hsa-mir-491/KIAA1797 and hsa-mir-646/AK309218. None of these CNVs has previously been reported in relation to CRC predisposition in humans, nor were they encountered in large control cohorts (>1,600 unaffected individuals). Since several of these newly identified candidate genes may be functionally linked to CRC development, our results illustrate the potential of this approach for the identification of novel candidate genes involved in CRC predisposition.


Sujet(s)
Variations de nombre de copies de segment d'ADN , Prédisposition génétique à une maladie , Adulte , Cadhérines/génétique , Études de cohortes , Tumeurs colorectales/génétique , Humains , Protéines et peptides de signalisation intercellulaire/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...