Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nat Immunol ; 18(6): 622-632, 2017 06.
Article in English | MEDLINE | ID: mdl-28459433

ABSTRACT

The high risk of neonatal death from sepsis is thought to result from impaired responses by innate immune cells; however, the clinical observation of hyperinflammatory courses of neonatal sepsis contradicts this concept. Using transcriptomic, epigenetic and immunological approaches, we demonstrated that high amounts of the perinatal alarmins S100A8 and S100A9 specifically altered MyD88-dependent proinflammatory gene programs. S100 programming prevented hyperinflammatory responses without impairing pathogen defense. TRIF-adaptor-dependent regulatory genes remained unaffected by perinatal S100 programming and responded strongly to lipopolysaccharide, but were barely expressed. Steady-state expression of TRIF-dependent genes increased only gradually during the first year of life in human neonates, shifting immune regulation toward the adult phenotype. Disruption of this critical sequence of transient alarmin programming and subsequent reprogramming of regulatory pathways increased the risk of hyperinflammation and sepsis. Collectively these data suggest that neonates are characterized by a selective, transient microbial unresponsiveness that prevents harmful hyperinflammation in the delicate neonate while allowing for sufficient immunological protection.


Subject(s)
Calgranulin A/immunology , Calgranulin B/immunology , Immunity, Innate/immunology , Monocytes/immunology , Neonatal Sepsis/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Calgranulin A/drug effects , Calgranulin B/drug effects , Epigenesis, Genetic , Fetal Blood , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunity, Innate/drug effects , Immunoblotting , Infant, Newborn , Inflammation , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Monocytes/drug effects , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Neonatal Sepsis/genetics , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 4/immunology
3.
Cell Mol Life Sci ; 79(1): 65, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35013790

ABSTRACT

Coronavirus disease 2019 (COVID-19), the illness caused by a novel coronavirus now called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to more than 260 million confirmed infections and 5 million deaths to date. While vaccination is a powerful tool to control pandemic spread, medication to relieve COVID-19-associated symptoms and alleviate disease progression especially in high-risk patients is still lacking. In this study, we explore the suitability of the rapid accelerated fibrosarcoma/mitogen-activated protein kinase/extracellular signal-regulated kinase (Raf/MEK/ERK) pathway as a druggable target in the treatment of SARS-CoV-2 infections. We find that SARS-CoV-2 transiently activates Raf/MEK/ERK signaling in the very early infection phase and that ERK1/2 knockdown limits virus replication in cell culture models. We demonstrate that ATR-002, a specific inhibitor of the upstream MEK1/2 kinases which is currently evaluated in clinical trials as an anti-influenza drug, displays strong anti-SARS-CoV-2 activity in cell lines as well as in primary air-liquid-interphase epithelial cell (ALI) cultures, with a safe and selective treatment window. We also observe that ATR-002 treatment impairs the SARS-CoV-2-induced expression of pro-inflammatory cytokines, and thus might prevent COVID-19-associated hyperinflammation, a key player in COVID-19 progression. Thus, our data suggest that the Raf/MEK/ERK signaling cascade may represent a target for therapeutic intervention strategies against SARS-CoV-2 infections and that ATR-002 is a promising candidate for further drug evaluation.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Fenamates/pharmacology , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , SARS-CoV-2/drug effects , A549 Cells , Adult , Animals , COVID-19/metabolism , Cell Line , Cells, Cultured , Chlorocebus aethiops , Cytokines/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/metabolism , SARS-CoV-2/physiology , Vero Cells , Virus Replication/drug effects
4.
Proc Natl Acad Sci U S A ; 117(31): 18649-18660, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32690687

ABSTRACT

Starting at birth, the immune system of newborns and children encounters and is influenced by environmental challenges. It is still not completely understood how γδ T cells emerge and adapt during early life. Studying the composition of T cell receptors (TCRs) using next-generation sequencing (NGS) in neonates, infants, and children can provide valuable insights into the adaptation of T cell subsets. To investigate how neonatal γδ T cell repertoires are shaped by microbial exposure after birth, we monitored the γ-chain (TRG) and δ-chain (TRD) repertoires of peripheral blood T cells in newborns, infants, and young children from Europe and sub-Saharan Africa. We identified a set of TRG and TRD sequences that were shared by all children from Europe and Africa. These were primarily public clones, characterized by simple rearrangements of Vγ9 and Vδ2 chains with low junctional diversity and usage of non-TRDJ1 gene segments, reminiscent of early ontogenetic subsets of γδ T cells. Further profiling revealed that these innate, public Vγ9Vδ2+ T cells underwent an immediate TCR-driven polyclonal proliferation within the first 4 wk of life. In contrast, γδ T cells using Vδ1+ and Vδ3+TRD rearrangements did not significantly expand after birth. However, different environmental cues may lead to the observed increase of Vδ1+ and Vδ3+TRD sequences in the majority of African children. In summary, we show how dynamic γδ TCR repertoires develop directly after birth and present important differences among γδ T cell subsets.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets/immunology , Africa South of the Sahara , Bacteria/immunology , Child , Child, Preschool , Europe , Gene Rearrangement, T-Lymphocyte/genetics , Gene Rearrangement, T-Lymphocyte/immunology , Humans , Infant , Infant, Newborn , Longitudinal Studies , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology
5.
EMBO Rep ; 21(12): e50421, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33180976

ABSTRACT

Pyroptosis is a fulminant form of macrophage cell death, contributing to release of pro-inflammatory cytokines. In humans, it depends on caspase 1/4-activation of gasdermin D and is characterized by the release of cytoplasmic content. Pathogens apply strategies to avoid or antagonize this host response. We demonstrate here that a small accessory protein (PB1-F2) of contemporary H5N1 and H3N2 influenza A viruses (IAV) curtails fulminant cell death of infected human macrophages. Infection of macrophages with a PB1-F2-deficient mutant of a contemporary IAV resulted in higher levels of caspase-1 activation, cleavage of gasdermin D, and release of LDH and IL-1ß. Mechanistically, PB1-F2 limits transition of NLRP3 from its auto-repressed and closed confirmation into its active state. Consequently, interaction of a recently identified licensing kinase NEK7 with NLRP3 is diminished, which is required to initiate inflammasome assembly.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A virus , Humans , Inflammasomes/genetics , Influenza A Virus, H3N2 Subtype , Influenza A virus/genetics , Macrophages , NIMA-Related Kinases , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pyroptosis
6.
Genes Dev ; 28(8): 858-74, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24736844

ABSTRACT

Although regulation of stem cell homeostasis by microRNAs (miRNAs) is well studied, it is unclear how individual miRNAs genomically encoded within an organized polycistron can interact to induce an integrated phenotype. miR-99a/100, let-7, and miR-125b paralogs are encoded in two tricistrons on human chromosomes 11 and 21. They are highly expressed in hematopoietic stem cells (HSCs) and acute megakaryoblastic leukemia (AMKL), an aggressive form of leukemia with poor prognosis. Here, we show that miR-99a/100∼125b tricistrons are transcribed as a polycistronic message transactivated by the homeobox transcription factor HOXA10. Integrative analysis of global gene expression profiling, miRNA target prediction, and pathway architecture revealed that miR-99a/100, let-7, and miR-125b functionally converge at the combinatorial block of the transforming growth factor ß (TGFß) pathway by targeting four receptor subunits and two SMAD signaling transducers. In addition, down-regulation of tumor suppressor genes adenomatous polyposis coli (APC)/APC2 stabilizes active ß-catenin and enhances Wnt signaling. By switching the balance between Wnt and TGFß signaling, the concerted action of these tricistronic miRNAs promoted sustained expansion of murine and human HSCs in vitro or in vivo while favoring megakaryocytic differentiation. Hence, our study explains the high phylogenetic conservation of the miR-99a/100∼125b tricistrons controlling stem cell homeostasis, the deregulation of which contributes to the development of AMKL.


Subject(s)
Hematopoietic Stem Cells/metabolism , Homeostasis/genetics , MicroRNAs , Signal Transduction , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism , Animals , Apoptosis/genetics , Down-Regulation , Erythropoiesis/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, APC/physiology , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Binding , Thrombopoiesis/genetics , Wnt Proteins/genetics
7.
Gastroenterology ; 159(6): 2130-2145.e5, 2020 12.
Article in English | MEDLINE | ID: mdl-32805279

ABSTRACT

BACKGROUND & AIMS: After birth, the immune system matures via interactions with microbes in the gut. The S100 calcium binding proteins S100A8 and S100A9, and their extracellular complex form, S100A8-A9, are found in high amounts in human breast milk. We studied levels of S100A8-A9 in fecal samples (also called fecal calprotectin) from newborns and during infancy, and their effects on development of the intestinal microbiota and mucosal immune system. METHODS: We collected stool samples (n = 517) from full-term (n = 72) and preterm infants (n = 49) at different timepoints over the first year of life (days 1, 3, 10, 30, 90, 180, and 360). We measured levels of S100A8-A9 by enzyme-linked immunosorbent assay and analyzed fecal microbiomes by 16S sRNA gene sequencing. We also obtained small and large intestine biopsies from 8 adults and 10 newborn infants without inflammatory bowel diseases (controls) and 8 infants with necrotizing enterocolitis and measured levels of S100A8 by immunofluorescence microscopy. Children were followed for 2.5 years and anthropometric data and medical information on infections were collected. We performed studies with newborn C57BL/6J wild-type and S100a9-/- mice (which also lack S100A8). Some mice were fed or given intraperitoneal injections of S100A8 or subcutaneous injections of Staphylococcus aureus. Blood and intestine, mesenterial and celiac lymph nodes were collected; cells and cytokines were measured by flow cytometry and studied in cell culture assays. Colon contents from mice were analyzed by culture-based microbiology assays. RESULTS: Loss of S100A8 and S100A9 in mice altered the phenotypes of colonic lamina propria macrophages, compared with wild-type mice. Intestinal tissues from neonatal S100-knockout mice had reduced levels of CX3CR1 protein, and Il10 and Tgfb1 mRNAs, compared with wild-type mice, and fewer T-regulatory cells. S100-knockout mice weighed 21% more than wild-type mice at age 8 weeks and a higher proportion developed fatal sepsis during the neonatal period. S100-knockout mice had alterations in their fecal microbiomes, with higher abundance of Enterobacteriaceae. Feeding mice S100 at birth prevented the expansion of Enterobacteriaceae, increased numbers of T-regulatory cells and levels of CX3CR1 protein and Il10 mRNA in intestine tissues, and reduced body weight and death from neonatal sepsis. Fecal samples from term infants, but not preterm infants, had significantly higher levels of S100A8-A9 during the first 3 months of life than fecal samples from adults; levels decreased to adult levels after weaning. Fecal samples from infants born by cesarean delivery had lower levels of S100A8-A9 than from infants born by vaginal delivery. S100 proteins were expressed by lamina propria macrophages in intestinal tissues from infants, at higher levels than in intestinal tissues from adults. High fecal levels of S100 proteins, from 30 days to 1 year of age, were associated with higher abundance of Actinobacteria and Bifidobacteriaceae, and lower abundance of Gammaproteobacteria-particularly opportunistic Enterobacteriaceae. A low level of S100 proteins in infants' fecal samples associated with development of sepsis and obesity by age 2 years. CONCLUSION: S100A8 and S100A9 regulate development of the intestinal microbiota and immune system in neonates. Nutritional supplementation with these proteins might aide in development of preterm infants and prevent microbiota-associated disorders in later years.


Subject(s)
Calgranulin A/metabolism , Calgranulin B/metabolism , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Adult , Animals , Biopsy , Calgranulin A/administration & dosage , Calgranulin A/analysis , Calgranulin B/analysis , Calgranulin B/genetics , Child, Preschool , Colon/microbiology , Colon/pathology , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Dysbiosis/microbiology , Dysbiosis/prevention & control , Enterocolitis, Necrotizing/epidemiology , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/microbiology , Enterocolitis, Necrotizing/prevention & control , Feces/chemistry , Feces/microbiology , Female , Follow-Up Studies , Gastrointestinal Microbiome/genetics , Humans , Immunity, Mucosal , Infant , Infant, Newborn , Infant, Premature/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Obesity/epidemiology , Obesity/immunology , Obesity/microbiology , Obesity/prevention & control , RNA, Ribosomal, 16S/genetics , Sepsis/epidemiology , Sepsis/immunology , Sepsis/microbiology , Sepsis/prevention & control
8.
Eur J Immunol ; 50(7): 1078-1080, 2020 07.
Article in English | MEDLINE | ID: mdl-32181500

ABSTRACT

AMPK (adenosine monophosphate-activated protein kinase) is phosphorylated (AMPK-P) in response to low energy through allosteric activation by Adenosine mono- or diphosphate (AMP/ADP). Folliculin (FLCN) and the FLCN-interacting proteins 1 and 2 (FNIP1, 2) modulate AMPK. FNIP1 deficiency patients have a AMPK-P gain of function phenotype with hypertrophic cardiomyopathy, Wolff-Parkinson-White pre-excitation syndrome, myopathy of skeletal muscles and combined immunodeficiency.


Subject(s)
Cardiomyopathies , Carrier Proteins , Genes, Recessive , Immunologic Deficiency Syndromes , Mutation , Pre-Excitation Syndromes , Cardiomyopathies/genetics , Cardiomyopathies/immunology , Cardiomyopathies/pathology , Carrier Proteins/genetics , Carrier Proteins/immunology , Female , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Male , Pre-Excitation Syndromes/genetics , Pre-Excitation Syndromes/immunology , Pre-Excitation Syndromes/pathology
9.
Clin Exp Pharmacol Physiol ; 47(5): 838-847, 2020 05.
Article in English | MEDLINE | ID: mdl-31883392

ABSTRACT

The ability of cancer cells to form clusters is a characteristic feature in the development of metastatic tumours with drug resistance. Several studies demonstrated that clusters of circulating tumour cells (CTCs) have a greater metastatic potential to establish new tumours at secondary sites than single CTCs. However, the mechanism of cluster formation is not well understood. In this study, we investigated whether cancer stemness would contribute to cluster formation. We used a tumour sphere culture method to enrich cancer stem cells (CSCs) from colon cancer cells and found that during the second generation of sphere culture, clusters (between 3 and 5 cells) formed within the first 24 hours, whereas the rest remained as single cells. The clusters were analysed for stemness and metastatic potential, including gene expressions for cancer stemness (CD133 and Lgr5), epithelial-mesenchymal transition (E-cadherin and TGF-ß 1-3) and hypoxia-induced factors (HIF-1α and HIF-2α). The results showed that the clusters expressed higher levels of these genes and colon CSC surface markers (including CD24, CD44 and CD133) than the single cells. Among these markers, CD24 seemed the major contributor linking the cells into the clusters. These clusters also showed a stronger ability to both form colonies and migrate. Our data collectively suggest that colon cancer stemness contributes to cluster formation and that clustered cells exhibit a great metastatic potential. Our study thus provides a method to study the CTC clusters and derive insight into oncogenesis and metastasis.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Biomarkers, Tumor/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation , HCT116 Cells , HT29 Cells , Humans , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Phenotype , Spheroids, Cellular
10.
Biomacromolecules ; 20(2): 625-634, 2019 02 11.
Article in English | MEDLINE | ID: mdl-30157649

ABSTRACT

Targeting the spleen with nanoparticles could increase the efficacy of vaccines and cancer immunotherapy, and have the potential to treat intracellular infections including leishmaniasis, trypanosome, splenic TB, AIDS, malaria, and hematological disorders. Although, nanoparticle capture in both the liver and spleen has been well documented, there are only a few examples of specific capture in the spleen alone. It is proposed that the larger the nanoparticle size (>400 nm) the greater the specificity and capture within the spleen. Here, we synthesized five nanostructures with different shapes (ranging from spheres, worms, rods, nanorattles, and toroids) and poly( N-isopropylacrylamide), PNIPAM, surface coating using the temperature-directed morphology transformation (TDMT) method. Globular PNIPAM (i.e., water insoluble) surface coatings have been shown to significantly increase cell uptake and enhanced enzyme activity. We incorporated a globular component of PNIPAM on the nanostructure surface and examined the in vivo biodistribution of these nanostructures and accumulation in various tissues and organs in a mouse model. The in vivo biodistribution as a function of time was influenced by the shape and PNIPAM surface composition, in which organ capture and retention was the highest in the spleen. The rods (∼150 nm in length and 15 nm in width) showed the highest capture and retention of greater than 35% to the initial injection amount compared to all other nanostructures. It was found that the rods specifically targeted the cells in the red pulp region of the spleen due to the shape and PNIPAM coating of the rod. This remarkable accumulation and selectively into the spleen represents new nanoparticle design parameters to develop new splenotropic effects for vaccines and other therapeutics.


Subject(s)
Acrylic Resins/chemistry , Nanoparticles/chemistry , Animals , Female , Hot Temperature , Mice , Mice, Inbred C57BL , Nanoparticles/metabolism , Nanoparticles/ultrastructure , RAW 264.7 Cells , Spleen/metabolism , Stimuli Responsive Polymers/chemistry , Tissue Distribution
11.
Clin Exp Pharmacol Physiol ; 44(2): 153-161, 2017 02.
Article in English | MEDLINE | ID: mdl-27809360

ABSTRACT

Chemotherapy resistance is a major contributor to poor treatment responses and tumour relapse, the development of which has been strongly linked to the action of cancer stem cells (CSCs). Mounting evidence suggests that CSCs are reliant on low oxygen conditions and hypoxia-inducible factors 1α and 2α (HIF1α and HIF2α) to maintain their stem cell features. Research in the last decade has begun to clarify the functional differences between the two HIFα subtypes (HIFαs). Here, we review and discuss these differences in relation to CSC-associated drug resistance. Both HIFαs contribute to CSC survival but play different roles -HIF1α being more responsible for survival functions and HIF2α for stemness traits such as self-renewal - and are sensitive to different degrees of hypoxia. Failure to account for physiologically relevant oxygen concentrations in many studies may influence the current understanding of the roles of HIFαs. We also discuss how hypoxia and HIFαs contribute to CSC drug resistance via promotion of ABC drug transporters Breast cancer resistance protein (BCRP), MDR1, and MRP1 and through maintenance of quiescence. Additionally, we explore the PI3K/AKT cell survival pathway that may support refractory cancer by promoting CSCs and activating both HIF1α and HIF2α. Accordingly, HIF1α and HIF2α inhibition, potentially via PI3K/AKT inhibitors, could reduce chemotherapy resistance and prevent cancer relapse.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Drug Resistance, Neoplasm/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Tumor Hypoxia/physiology , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/physiology , Cell Survival/drug effects , Cell Survival/physiology , Drug Resistance, Neoplasm/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Tumor Hypoxia/drug effects
12.
J Exp Med ; 221(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38753245

ABSTRACT

Preterm infants are at high risk of developing neonatal sepsis. γδ T cells are thought to be an important set of effector cells in neonates. Here, γδ T cells were investigated in a longitudinal cohort of preterm neonates using next-generation sequencing, flow cytometry, and functional assays. During the first year of life, the Vγ9Vδ2 T cell subset showed dynamic phenotypic changes and elevated levels of fetal-derived Vγ9Vδ2 T cells were evident in infants with sepsis. Single-cell transcriptomics identified HLA-DRhiCD83+ γδ T cells in neonatal sepsis, which expressed genes related to antigen presentation. In vitro assays showed that CD83 was expressed on activated Vγ9Vδ2 T cells in preterm and term neonates, but not in adults. In contrast, activation of adult Vγ9Vδ2 T cells enhanced CD86 expression, which was presumably the key receptor to induce CD4 T cell proliferation. Together, we provide a map of the maturation of γδ T cells after preterm birth and highlight their phenotypic diversity in infections.


Subject(s)
Antigens, CD , CD83 Antigen , Infant, Premature , Receptors, Antigen, T-Cell, gamma-delta , Humans , Infant, Newborn , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Infant, Premature/immunology , Antigens, CD/metabolism , Antigens, CD/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Female , Male , Sepsis/immunology , Cohort Studies , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adult , Lymphocyte Activation/immunology , Neonatal Sepsis/immunology , Infant
13.
Sci Rep ; 7(1): 8761, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28821767

ABSTRACT

Devil facial tumour (DFT) disease, a transmissible cancer where the infectious agent is the tumour itself, has caused a dramatic decrease in Tasmanian devil numbers in the wild. The purpose of this study was to take a candidate gene/pathway approach to identify potentially perturbed genes or pathways in DFT. A fusion of chromosome 1 and X is posited as the initial event leading to the development of DFT, with the rearranged chromosome 1 material now stably maintained as the tumour spreads through the population. This hypothesis makes chromosome 1 a prime chromosome on which to search for mutations involved in tumourigenesis. As DFT1 has a Schwann cell origin, we selected genes commonly implicated in tumour pathways in human nerve cancers, or cancers more generally, to determine whether they were rearranged in DFT1, and mapped them using molecular cytogenetics. Many cancer-related genes were rearranged, such as the region containing the tumour suppressor NF2 and a copy gain for ERBB3, a member of the epidermal growth factor receptor family of receptor tyrosine kinases implicated in proliferation and invasion of tumours in humans. Our mapping results have provided strong candidates not previously detected by sequencing DFT1 genomes.


Subject(s)
Biomarkers, Tumor , Cell Transformation, Neoplastic/genetics , Facial Neoplasms/etiology , Genetic Predisposition to Disease , Cell Transformation, Neoplastic/metabolism , Chromosome Aberrations , Chromosome Mapping , Facial Neoplasms/metabolism , Facial Neoplasms/pathology , Genetic Association Studies , Humans , Karyotype
SELECTION OF CITATIONS
SEARCH DETAIL