Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cells ; 11(11)2022 05 24.
Article in English | MEDLINE | ID: mdl-35681428

ABSTRACT

Inflammation and fibrosis are important components of diseases that contribute to the malfunction of epithelia and endothelia. The Rho guanine nucleotide exchange factor (GEF) GEF-H1/ARHGEF-2 is induced in disease and stimulates inflammatory and fibrotic processes, cell migration, and metastasis. Here, we have generated peptide inhibitors to block the function of GEF-H1. Inhibitors were designed using a structural in silico approach or by isolating an inhibitory sequence from the autoregulatory C-terminal domain. Candidate inhibitors were tested for their ability to block RhoA/GEF-H1 binding in vitro, and their potency and specificity in cell-based assays. Successful inhibitors were then evaluated in models of TGFß-induced fibrosis, LPS-stimulated endothelial cell-cell junction disruption, and cell migration. Finally, the most potent inhibitor was successfully tested in an experimental retinal disease mouse model, in which it inhibited blood vessel leakage and ameliorated retinal inflammation when treatment was initiated after disease diagnosis. Thus, an antagonist that blocks GEF-H1 signaling effectively inhibits disease features in in vitro and in vivo disease models, demonstrating that GEF-H1 is an effective therapeutic target and establishing a new therapeutic approach.


Subject(s)
Retinal Diseases , Signal Transduction , Animals , Fibrosis , Inflammation , Mice , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism
2.
Int J Pharm ; 621: 121755, 2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35447226

ABSTRACT

Treatment of posterior eye diseases with intravitreal injections of drugs, while effective, is invasive and associated with side effects such as retinal detachment and endophthalmitis. In this work, we have formulated a model compound, rapamycin (RAP), in nanoparticle-based eye drops and evaluated the delivery of RAP to the posterior eye tissues in a healthy rabbit. We have also studied the formulation in experimental autoimmune uveitis (EAU) mouse model with retinal inflammation. Aqueous RAP eye drops were prepared using N-palmitoyl-N-monomethyl-N,N-dimethyl-N,N,N-trimethyl-6-O-glycolchitosan (Molecular Envelope Technology - MET) containing 0.23 ± 0.001% w/v RAP with viscosity, osmolarity, and pH within the ocular comfort range, and the formulation (MET-RAP) was stable in terms of drug content at both refrigeration and room temperature for one month. The MET-RAP eye drops delivered RAP to the choroid-retina with a Cmax of 145 ± 49 ng/g (tmax = 1 h). The topical application of the MET-RAP eye drops to the EAU mouse model resulted in significant disease suppression compared to controls, with activity similar to dexamethasone eye drops. The MET-RAP eye drops also resulted in a reduction of RORγt and an increase in both Foxp3 expression and IL-10 secretion, indicating a mechanism involving the inhibition of Th17 cells and the up-regulation of T-reg cells. The MET-RAP formulation delivers RAP to the posterior eye segments, and the formulation is active in EAU.


Subject(s)
Posterior Eye Segment , Uveitis , Animals , Mice , Ophthalmic Solutions/pharmacology , Rabbits , Retina , Sirolimus/pharmacology , Uveitis/drug therapy
3.
J Vis Exp ; (179)2022 01 12.
Article in English | MEDLINE | ID: mdl-35098942

ABSTRACT

Experimental Autoimmune Uveitis (EAU) is driven by immune cells responding to self-antigens. Many features of this non-infectious, intraocular inflammatory disease model recapitulate the clinical phenotype of posterior uveitis affecting humans. EAU has been used reliably to study the efficacy of novel inflammatory therapeutics, their mode of action and to further investigate the mechanisms that underpin disease progression of intraocular disorders. Here, we provide a detailed protocol on EAU induction in the C57BL/6J mouse - the most widely used model organism with susceptibility to this disease. Clinical assessment of disease severity and progression will be demonstrated using fundoscopy, histological examination and fluorescein angiography. The induction procedure involves subcutaneous injection of an emulsion containing a peptide (IRBP1-20) from the ocular protein interphotoreceptor retinoid binding protein (also known as retinol binding protein 3), Complete Freund's Adjuvant (CFA) and supplemented with killed Mycobacterium tuberculosis. Injection of this viscous emulsion on the back of the neck is followed by a single intraperitoneal injection of Bordetella pertussis toxin. At the onset of symptoms (day 12-14) and under general anesthesia, fundoscopic images are taken to assess disease progression through clinical examination. These data can be directly compared with those at later timepoints and peak disease (day 20-22) with differences analyzed. At the same time, this protocol allows the investigator to assess potential differences in vessel permeability and damage using fluorescein angiography. EAU can be induced in other mouse strains - both wildtype or genetically modified - and combined with novel therapies offering flexibility for studying drug efficacy and/or disease mechanisms.


Subject(s)
Autoimmune Diseases , Uveitis , Animals , Disease Models, Animal , Eye Proteins/therapeutic use , Inflammation , Mice , Mice, Inbred C57BL , Uveitis/drug therapy , Uveitis/pathology
4.
Int J Mol Sci ; 23(1)2022 Jan 03.
Article in English | MEDLINE | ID: mdl-35008929

ABSTRACT

Non-infectious uveitis (NIU) is an inflammatory eye disease initiated via CD4+ T-cell activation and transmigration, resulting in focal retinal tissue damage and visual acuity disturbance. Cell adhesion molecules (CAMs) are activated during the inflammatory process to facilitate the leukocyte recruitment cascade. Our review focused on CAM-targeted therapies in experimental autoimmune uveitis (EAU) and NIU. We concluded that CAM-based therapies have demonstrated benefits for controlling EAU severity with decreases in immune cell migration, especially via ICAM-1/LFA-1 and VCAM-1/VLA-4 (integrin) pathways. P-selectin and E-selectin are more involved specifically in uveitis related to vasculitis. These therapies have potential clinical applications for the development of a more personalized and specific treatment. Localized therapies are the future direction to avoid serious systemic side effects.


Subject(s)
Cell Adhesion Molecules , Molecular Targeted Therapy , Uveitis/therapy , Humans , Inflammation , Uveitis/metabolism
6.
Cells ; 10(2)2021 02 15.
Article in English | MEDLINE | ID: mdl-33671954

ABSTRACT

Retinal vascular diseases have distinct, complex and multifactorial pathogeneses yet share several key pathophysiological aspects including inflammation, vascular permeability and neovascularisation. In non-infectious posterior uveitis (NIU), retinal vasculitis involves vessel leakage leading to retinal enlargement, exudation, and macular oedema. Neovascularisation is not a common feature in NIU, however, detection of the major angiogenic factor-vascular endothelial growth factor A (VEGF-A)-in intraocular fluids in animal models of uveitis may be an indication for a role for this cytokine in a highly inflammatory condition. Suppression of VEGF-A by directly targeting the leukotriene B4 (LTB4) receptor (BLT1) pathway indicates a connection between leukotrienes (LTs), which have prominent roles in initiating and propagating inflammatory responses, and VEGF-A in retinal inflammatory diseases. Further research is needed to understand how LTs interact with intraocular cytokines in retinal inflammatory diseases to guide the development of novel therapeutic approaches targeting both inflammatory mediator pathways.


Subject(s)
Inflammation/drug therapy , Receptors, Leukotriene B4/metabolism , Retinal Vasculitis/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Disease Models, Animal , Humans , Receptors, Leukotriene B4/immunology , Retinal Vasculitis/immunology , Uveitis/drug therapy , Uveitis/immunology
7.
J Neuroinflammation ; 18(1): 49, 2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33602234

ABSTRACT

BACKGROUND: The integrin VLA-4 (α4ß1) plays an important role in leukocyte trafficking. This study investigated the efficacy of a novel topical α4ß1 integrin inhibitor (GW559090, GW) in a mouse model for non-infectious posterior uveitis (experimental autoimmune uveitis; EAU) and its effect on intraocular leukocyte subsets. METHODS: Mice (female; B10.RIII or C57Bl/6; aged 6-8 weeks) were immunized with specific interphotoreceptor retinoid-binding protein (IRBP) peptides to induce EAU. Topically administered GW (3, 10, and 30 mg/ml) were given twice daily either therapeutically once disease was evident, or prophylactically, and compared with vehicle-treated (Veh) and 0.1% dexamethasone-treated (Dex) controls. Mice were sacrificed at peak disease. The retinal T cell subsets were investigated by immunohistochemistry and immunofluorescence staining. The immune cells within the retina, blood, and draining lymph nodes (dLNs) were phenotyped by flow cytometry. The effect of GW559090 on non-adherent, adherent, and migrated CD4+ T cell subsets across a central nervous system (CNS) endothelium was further assayed in vitro and quantitated by flow cytometry. RESULTS: There was a significant reduction in clinical and histological scores in GW10- and Dex-treated groups as compared to controls either administered therapeutically or prophylactically. There were fewer CD45+ leukocytes infiltrating the retinae and vitreous fluids in the treated GW10 group (P < 0.05). Immunofluorescence staining and flow cytometry data identified decreased levels of retinal Th17 cells (P ≤ 0.001) in the GW10-treated eyes, leaving systemic T cell subsets unaffected. In addition, fewer Ly6C+ inflammatory monocyte/macrophages (P = 0.002) and dendritic cells (P = 0.017) crossed the BRB following GW10 treatment. In vitro migration assays confirmed that Th17 cells were selectively suppressed by GW559090 in adhering to endothelial monolayers. CONCLUSIONS: This α4ß1 integrin inhibitor may exert a modulatory effect in EAU progression by selectively blocking Th17 cell migration across the blood-retinal barrier without affecting systemic CD4+ T cell subsets. Local α4ß1 integrin-directed inhibition could be clinically relevant in treating a Th17-dominant form of uveitis.


Subject(s)
Autoimmune Diseases/drug therapy , Blood-Retinal Barrier/drug effects , Integrin alpha4beta1/antagonists & inhibitors , Phenylalanine/analogs & derivatives , Piperidines/administration & dosage , Th17 Cells/drug effects , Uveitis/drug therapy , Animals , Autoimmune Diseases/metabolism , Blood-Retinal Barrier/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Female , Humans , Integrin alpha4beta1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenylalanine/administration & dosage , Phenylalanine/metabolism , Piperidines/metabolism , Th17 Cells/metabolism , Uveitis/metabolism
8.
Am J Pathol ; 191(2): 320-334, 2021 02.
Article in English | MEDLINE | ID: mdl-33159884

ABSTRACT

Nomacopan, a drug originally derived from tick saliva, has dual functions of sequestering leukotriene B4 (LTB4) and inhibiting complement component 5 (C5) activation. Nomacopan has been shown to provide therapeutic benefit in experimental autoimmune uveitis (EAU). Longer acting forms of nomacopan were more efficacious in mouse EAU models, and the long-acting variant that inhibited only LTB4 was at least as effective as the long-acting variant that inhibited both C5 and LTB4, preventing structural damage to the retina and a significantly reducing effector T helper 17 cells and inflammatory macrophages. Increased levels of LTB4 and C5a (produced upon C5 activation) were detected during disease progression. Activated retinal lymphocytes were shown to express LTB4 receptors (R) in vitro and in inflamed draining lymph nodes. Levels of LTB4R-expressing active/inflammatory retinal macrophages were also increased. Within the draining lymph node CD4+ T-cell population, 30% expressed LTB4R+ following activation in vitro, whereas retinal infiltrating cells expressed LTB4R and C5aR. Validation of expression of those receptors in human uveitis and healthy tissues suggests that infiltrating cells could be targeted by inhibitors of the LTB4-LTB4 receptor 1 (BLT1) pathway as a novel therapeutic approach. This study provides novel data on intraocular LTB4 and C5a in EAU, their associated receptor expression by retinal infiltrating cells in mouse and human tissues, and in attenuating EAU via the dual inhibitor nomacopan.


Subject(s)
Leukotriene B4/metabolism , Receptors, Leukotriene B4/metabolism , Retina/metabolism , Uveitis/immunology , Uveitis/metabolism , Animals , Biological Products/pharmacology , Complement C5a/antagonists & inhibitors , Complement C5a/metabolism , Disease Models, Animal , Female , Humans , Leukotriene B4/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Receptors, Leukotriene B4/antagonists & inhibitors , Retina/immunology , Th17 Cells/immunology
9.
Eur J Immunol ; 50(12): 1941-1951, 2020 12.
Article in English | MEDLINE | ID: mdl-32652562

ABSTRACT

Immunopathogenic roles for both Th1 (CD4+ IFN-γ+ ) and Th17 (CD4+ IL-17A+ ) cells have been demonstrated in experimental autoimmune uveitis (EAU). However, the role for Th17/Th1 (CD4+ T cells co-expressing IFN-γ and IL-17A) cells in EAU is not yet understood. Using interphotoreceptor retinoid-binding protein peptide-induced EAU in mice, we found increased levels of Th17/Th1 cells in EAU retinae (mean 9.6 ± 4.2%) and draining LNs (mean 8.4 ± 3.9%; p = 0.01) relative to controls. Topical dexamethasone treatment effectively reduced EAU severity and decreased retinal Th1 cells (p = 0.01), but had no impact on retinal Th17/Th1 or Th17 cells compared to saline controls. Using in vitro migration assays with mouse CNS endothelium, we demonstrated that Th17/Th1 cells were significantly increased within the migrated population relative to controls (mean 15.6 ± 9.5% vs. 1.9 ± 1.5%; p = 0.01). Chemokine receptor profiles of Th17/Th1 cells (CXCR3 and CCR6) did not change throughout the transendothelial migration process and were unaffected by dexamethasone treatment. These findings support a role for Th17/Th1 cells in EAU and their resistance to steroid inhibition suggests the importance of targeting both Th17 and Th17/Th1 cells for improving therapy.


Subject(s)
Autoimmune Diseases/immunology , Cell Movement/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL
10.
Br J Cancer ; 117(3): 398-408, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28632727

ABSTRACT

BACKGROUND: The most prevalent form of bone cancer is osteosarcoma (OS), which is associated with poor prognosis in case of metastases formation. Mice harbouring liver kinase B1 (LKB1+/-) develop osteoblastoma-like tumours. Therefore, we asked whether loss of LKB1 gene has a role in the pathogenesis of human OS. METHODS: Osteosarcomas (n=259) were screened for LKB1 and sirtuin 1 (SIRT1) protein expression using immunohistochemistry and western blot. Those cases were also screened for LKB1 genetic alterations by next-generation sequencing, Sanger sequencing, restriction fragment length polymorphism and fluorescence in situ hybridisation approaches. We studied LKB1 protein degradation through SIRT1 expression. MicroRNA expression investigations were also conducted to identify the microRNAs involved in the SIRT1/LKB1 pathway. RESULTS: Forty-one per cent (106 out of 259) OS had lost LKB1 protein expression with no evident genetic anomalies. We obtained evidence that SIRT1 impairs LKB1 protein stability, and that SIRT1 depletion leads to accumulation of LKB1 in OS cell lines resulting in growth arrest. Further investigations revealed the role of miR-204 in the regulation of SIRT1 expression, which impairs LKB1 stability. CONCLUSIONS: We demonstrated the involvement of sequential regulation of miR-204/SIRT1/LKB1 in OS cases and showed a mechanism for the loss of expression of LKB1 tumour suppressor in this malignancy.


Subject(s)
Bone Neoplasms/genetics , Osteosarcoma/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Sirtuin 1/metabolism , AMP-Activated Protein Kinase Kinases , Anoikis/genetics , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Bone Neoplasms/metabolism , Bortezomib/pharmacology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Humans , MicroRNAs/genetics , Naphthols/pharmacology , Osteosarcoma/metabolism , Protein Biosynthesis/drug effects , Signal Transduction , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , TOR Serine-Threonine Kinases/metabolism , Transfection
11.
J Immunol ; 198(3): 1093-1103, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28039300

ABSTRACT

Experimental autoimmune uveitis (EAU), in which CD4+ Th1 and/or Th17 cells are immunopathogenic, mimics various clinical features of noninfectious uveitis in humans. The impact of bromodomain extraterminal (BET) inhibitors on Th17 cell function was studied in a mouse model of EAU in vivo and in mouse and human Th17 cells in vitro. Two BET inhibitors (GSK151 and JQ1) were able to ameliorate the progression of inflammation in EAU and in mouse CD4+ T cells in vitro, downregulating levels of Th17 cells. Additionally, the uveitogenic capacity of Th17 cells to transfer EAU was abrogated by BET inhibitors in an adoptive transfer model. In human CD4+ T cells, a 5-d exposure to BET inhibitors was accompanied by a significant downregulation of Th17-associated genes IL-17A, IL-22, and retinoic acid-related orphan receptor γt. However, in vitro, the inhibitors had no effect on already polarized Th17 cells. The key finding is that, in response to BET inhibitors, Th17-enriched cultures developed a regulatory phenotype, upregulated FOXP3 expression and IL-10 secretion, and lost pathogenicity in vivo. We conclude that BET targeting of Th17 cells is a potential therapeutic opportunity for a wide range of inflammatory and autoimmune diseases, including uveitis.


Subject(s)
Autoimmune Diseases/drug therapy , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Retina/drug effects , Th17 Cells/drug effects , Transcription Factors/antagonists & inhibitors , Uveitis/drug therapy , Animals , Cytokines/biosynthesis , Down-Regulation , Female , Forkhead Transcription Factors/analysis , Mice , Nuclear Proteins/physiology , Nuclear Receptor Subfamily 1, Group F, Member 3/analysis , Receptors, CCR6/analysis , Retina/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Transcription Factors/physiology , Tumor Necrosis Factor-alpha/analysis
12.
Cell Rep ; 15(12): 2756-70, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27292648

ABSTRACT

The transcription factor T-bet directs Th1 cell differentiation, but the molecular mechanisms that underlie this lineage-specific gene regulation are not completely understood. Here, we show that T-bet acts through enhancers to allow the recruitment of Mediator and P-TEFb in the form of the super elongation complex (SEC). Th1 genes are occupied by H3K4me3 and RNA polymerase II in Th2 cells, while T-bet-mediated recruitment of P-TEFb in Th1 cells activates transcriptional elongation. P-TEFb is recruited to both genes and enhancers, where it activates enhancer RNA transcription. P-TEFb inhibition and Mediator and SEC knockdown selectively block activation of T-bet target genes, and P-TEFb inhibition abrogates Th1-associated experimental autoimmune uveitis. T-bet activity is independent of changes in NF-κB RelA and Brd4 binding, with T-bet- and NF-κB-mediated pathways instead converging to allow P-TEFb recruitment. These data provide insight into the mechanism through which lineage-specifying factors promote differentiation of alternative T cell fates.


Subject(s)
Gene Expression Regulation , T-Box Domain Proteins/metabolism , Th1 Cells/metabolism , Transcription Elongation, Genetic , Animals , Cell Lineage/genetics , Enhancer Elements, Genetic/genetics , Humans , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding/genetics , RNA/genetics , RNA/metabolism , Th2 Cells/metabolism , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Uveitis/genetics
13.
Nat Commun ; 4: 2166, 2013.
Article in English | MEDLINE | ID: mdl-23863747

ABSTRACT

Isocitrate dehydrogenase (IDH) genes 1 and 2 are frequently mutated in acute myeloid leukaemia (AML), low-grade glioma, cholangiocarcinoma (CC) and chondrosarcoma (CS). For AML, low-grade glioma and CC, mutant IDH status is associated with a DNA hypermethylation phenotype, implicating altered epigenome dynamics in the aetiology of these cancers. Here we show that the IDH variants in CS are also associated with a hypermethylation phenotype and display increased production of the oncometabolite 2-hydroxyglutarate, supporting the role of mutant IDH-produced 2-hydroxyglutarate as an inhibitor of TET-mediated DNA demethylation. Meta-analysis of the acute myeloid leukaemia, low-grade glioma, cholangiocarcinoma and CS methylation data identifies cancer-specific effectors within the retinoic acid receptor activation pathway among the hypermethylated targets. By analysing sequence motifs surrounding hypermethylated sites across the four cancer types, and using chromatin immunoprecipitation and western blotting, we identify the transcription factor EBF1 (early B-cell factor 1) as an interaction partner for TET2, suggesting a sequence-specific mechanism for regulating DNA methylation.


Subject(s)
DNA-Binding Proteins/genetics , Isocitrate Dehydrogenase/genetics , Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Glutarates/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction , Trans-Activators/metabolism
14.
Nat Genet ; 43(12): 1262-5, 2011 Nov 06.
Article in English | MEDLINE | ID: mdl-22057236

ABSTRACT

Ollier disease and Maffucci syndrome are characterized by multiple central cartilaginous tumors that are accompanied by soft tissue hemangiomas in Maffucci syndrome. We show that in 37 of 40 individuals with these syndromes, at least one tumor has a mutation in isocitrate dehydrogenase 1 (IDH1) or in IDH2, 65% of which result in a R132C substitution in the protein. In 18 of 19 individuals with more than one tumor analyzed, all tumors from a given individual shared the same IDH1 mutation affecting Arg132. In 2 of 12 subjects, a low level of mutated DNA was identified in non-neoplastic tissue. The levels of the metabolite 2HG were measured in a series of central cartilaginous and vascular tumors, including samples from syndromic and nonsyndromic subjects, and these levels correlated strongly with the presence of IDH1 mutations. The findings are compatible with a model in which IDH1 or IDH2 mutations represent early post-zygotic occurrences in individuals with these syndromes.


Subject(s)
Enchondromatosis/genetics , Isocitrate Dehydrogenase/genetics , Mutation, Missense , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Genetic Association Studies , Humans , Male , Middle Aged , Mosaicism , Sequence Analysis, DNA , Young Adult
15.
J Pathol ; 224(3): 334-43, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21598255

ABSTRACT

Somatic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 occur in gliomas and acute myeloid leukaemia (AML). Since patients with multiple enchondromas have occasionally been reported to have these conditions, we hypothesized that the same mutations would occur in cartilaginous neoplasms. Approximately 1200 mesenchymal tumours, including 220 cartilaginous tumours, 222 osteosarcomas and another ∼750 bone and soft tissue tumours, were screened for IDH1 R132 mutations, using Sequenom(®) mass spectrometry. Cartilaginous tumours and chondroblastic osteosarcomas, wild-type for IDH1 R132, were analysed for IDH2 (R172, R140) mutations. Validation was performed by capillary sequencing and restriction enzyme digestion. Heterozygous somatic IDH1/IDH2 mutations, which result in the production of a potential oncometabolite, 2-hydroxyglutarate, were only detected in central and periosteal cartilaginous tumours, and were found in at least 56% of these, ∼40% of which were represented by R132C. IDH1 R132H mutations were confirmed by immunoreactivity for this mutant allele. The ratio of IDH1:IDH2 mutation was 10.6 : 1. No IDH2 R140 mutations were detected. Mutations were detected in enchondromas through to conventional central and dedifferentiated chondrosarcomas, in patients with both solitary and multiple neoplasms. No germline mutations were detected. No mutations were detected in peripheral chondrosarcomas and osteochondromas. In conclusion, IDH1 and IDH2 mutations represent the first common genetic abnormalities to be identified in conventional central and periosteal cartilaginous tumours. As in gliomas and AML, the mutations appear to occur early in tumourigenesis. We speculate that a mosaic pattern of IDH-mutation-bearing cells explains the reports of diverse tumours (gliomas, AML, multiple cartilaginous neoplasms, haemangiomas) occurring in the same patient.


Subject(s)
Bone Neoplasms/genetics , Chondroma/genetics , Chondrosarcoma/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Bone Neoplasms/diagnosis , Bone Neoplasms/pathology , Chondroma/diagnosis , Chondroma/pathology , Chondrosarcoma/pathology , Enchondromatosis/genetics , Enchondromatosis/pathology , Female , Follow-Up Studies , Germ-Line Mutation , Humans , Magnetic Resonance Imaging , Male , Osteosarcoma/genetics , Osteosarcoma/pathology
17.
Cancer Res ; 69(9): 3746-54, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19383913

ABSTRACT

For clinically relevant studies on melanoma progression and invasiveness, in vivo experimental systems with a human cellular microenvironment would be advantageous. We have compared tumor formation from a human cutaneous malignant melanoma cell line (BL), after injection as conventional xenografts in the mouse, or when injected into a predominantly species-specific environment of human embryonic stem cell-derived teratoma induced in the mouse (the hEST model). The resulting melanoma histology was generally analogous, both systems showing delimited densely packed areas with pleomorphic cells of malignant appearance. A specificity of the integration process into the human embryonic teratoma tissues was indicated by the melanoma exclusively being found in areas compatible with condensed mesenchyme, similar to neural crest development. Here, also enhanced neovascularization was seen within the human mesenchymal tissues facing the BL melanoma growth. Furthermore, in the hEST model an additional melanoma cell phenotype occurred, located at the border of, or infiltrating into, the surrounding human loose mesenchymal fibrous stroma. This BL population had a desmoplastic spindle-like appearance, with markers indicative of dedifferentiation and migration. The appearance of this apparently more aggressive phenotype, as well as the induction of human angiogenesis, shows specific interactions with the human embryonic microenvironment in the hEST model. In conclusion, these data provide exciting options for using the hEST model in molecular in vivo studies on differentiation, invasiveness, and malignancy of human melanoma, while analyzing species-specific reactions in vivo.


Subject(s)
Melanoma/pathology , Neoplasm Transplantation/pathology , Transplantation, Heterologous/pathology , Animals , Cell Differentiation/physiology , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/pathology , Embryonic Stem Cells/pathology , Humans , Immunohistochemistry , Male , Melanoma/metabolism , Mice , Mice, SCID , Phenotype , S100 Calcium-Binding Protein A4 , S100 Proteins/biosynthesis , Species Specificity , Teratoma/metabolism , Teratoma/pathology
18.
Int J Cancer ; 124(1): 16-26, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18814281

ABSTRACT

Activating mutations in the NRAS gene, which occur predominantly in codon 61 (Q61R, Q61K) are among the most common genetic events in malignant melanoma. NRAS protein with oncogenic codon 61 mutations may therefore be good therapeutic targets. In the present study, we used gene expression profiling as a method for global characterization of gene expression alterations that resulted from treatment of melanoma cells with siRNA specifically targeting NRAS(Q61R). Sixteen probe sets representing 15 unique genes were identified whose expression was significantly altered by siRNA against NRAS(Q61R) in 2 melanoma cell lines. The genes with altered expression are involved in several functions, including modulation of cell growth, invasion and migration. The results suggest that downregulation of cyclin E2 and cyclin D1 and also upregulation of the negative cell-cycle regulator HBP1 in NRAS(Q61R) knockdown cells contribute to the inhibition of cell proliferation. Furthermore, suppression of oncogenic NRAS results in reduced migration and invasion, which is accompanied by downregulation of EphA2 (a receptor tyrosine kinase), uPAR (urokinase receptor) and cytoskeleton proteins such as leupaxin, paxillin and vinculin. These studies support the concept that suppression of oncogenic NRAS by siRNA can induce growth arrest and inhibit invasion of human melanoma cells by modulating the levels of these gene products.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, ras , Melanoma/metabolism , Mutation , Oncogene Protein p21(ras)/metabolism , Skin Neoplasms/metabolism , Cell Adhesion Molecules/pharmacology , Cell Line, Tumor , Cyclin D1/metabolism , Cyclins/metabolism , Cytoskeleton/metabolism , Humans , Paxillin/pharmacology , Phenotype , Phosphoproteins/pharmacology , Vinculin/pharmacology
19.
Int J Cancer ; 115(1): 65-73, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15688405

ABSTRACT

The majority of human melanomas harbor activating mutations in either the BRAF or NRAS gene. To date, the role of oncogenic NRAS in melanoma remains poorly defined and no current therapies are directed at specifically suppressing oncogenic NRAS in human melanoma tumors. The aim of our study, therefore, was to investigate the effects of suppressing oncogenic NRAS in human melanoma cell lines in vitro. Using both small interfering RNA- and plasmid based-RNA interference techniques, oncogenic NRAS was specifically suppressed in 2 human melanoma cell lines, 224 and BL, which harbor a codon 61 CAA (glutamine) to CGA (arginine) NRAS mutation. Suppression of oncogenic NRAS in these cell lines resulted in increased apoptosis. Furthermore, in 224 cells we demonstrated decreased phosphorylation of extracellular signal-regulated kinase (ERK) and Akt, and reduced expression of NF-kappaB and cyclin D1 in the N-Ras signaling pathway. In contrast, RNA interference directed at wild-type (WT) NRAS had no significant effect on apoptosis of 224 cells or 2 human melanoma cell lines (A375 and 397) containing WT NRAS but a codon 600 GTG (valine) to GAG (glutamate) mutation in BRAF. These data suggest that oncogenic NRAS is important for avoidance of apoptosis in melanomas that harbor the codon 61 NRAS mutation and emphasizes oncogenic NRAS as a therapeutic target in patients with tumors that harbor this mutation.


Subject(s)
Apoptosis , Genes, ras/genetics , Melanoma/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , Cell Line, Tumor , Cell Proliferation , Codon , Coloring Agents/pharmacology , Cyclin D1/metabolism , Down-Regulation , Ethidium/pharmacology , Genetic Vectors , Humans , Immunoblotting , In Situ Nick-End Labeling , Melanoma/metabolism , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Plasmids/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , Time Factors , Transfection
20.
Cancer Res ; 64(3): 807-11, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871803

ABSTRACT

Exposure to UV radiation likely plays a key role in melanoma development, whereas other etiologic agents remain unknown. Here we show that in normal human skin an increased expression of a combination of three growth factors, basic fibroblast growth factor, stem cell factor, and endothelin-3, along with exposure to UVB can transform normal melanocytes into a melanoma phenotype within 4 weeks. Invasion of melanoma lesions was found in skin from newborn donors, whereas melanomas in adult skin were of a noninvasive in situ type only. This suggests that susceptibility of skin to exogenous tumor promoters is dependent on age. This is the first report on human cancer initiation in vivo in which an imbalance of physiological factors combined with an environmental carcinogen can lead to transformation of normal tissue.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/radiation effects , Growth Substances/biosynthesis , Melanoma/etiology , Neoplasms, Radiation-Induced/etiology , Skin/radiation effects , Ultraviolet Rays/adverse effects , Adult , Endothelin-3/biosynthesis , Fibroblast Growth Factor 2/biosynthesis , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Infant, Newborn , Melanocytes/metabolism , Melanocytes/radiation effects , Melanoma/metabolism , Neoplasms, Radiation-Induced/metabolism , Skin/cytology , Skin/metabolism , Stem Cell Factor/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...