Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
J Am Chem Soc ; 145(24): 12992-12997, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37294668

ABSTRACT

An expansion of the hexanucleotide (GGGGCC) repeat sequence in chromosome 9 open frame 72 (c9orf72) is the most common genetic mutation in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The mutation leads to the production of toxic dipeptide repeat proteins (DPRs) that induce neurodegeneration. However, the fundamental physicochemical properties of DPRs remain largely unknown due to their limited availability. Here, we synthesized the c9orf72 DPRs poly-glycine-arginine (poly-GR), poly-proline-arginine (poly-PR), poly-glycine-proline (poly-GP), poly-proline-alanine (poly-PA), and poly-glycine-alanine (poly-GA) using automated fast-flow peptide synthesis (AFPS) and achieved single-domain chemical synthesis of proteins with up to 200 amino acids. Circular dichroism spectroscopy of the synthetic DPRs revealed that proline-containing poly-PR, poly-GP, and poly-PA could adopt polyproline II-like helical secondary structures. In addition, structural analysis by size-exclusion chromatography indicated that longer poly-GP and poly-PA might aggregate. Furthermore, cell viability assays showed that human neuroblastoma cells cultured with poly-GR and poly-PR with longer repeat lengths resulted in reduced cell viability, while poly-GP and poly-PA did not, thereby reproducing the cytotoxic property of endogenous DPRs. This research demonstrates the potential of AFPS to synthesize low-complexity peptides and proteins necessary for studying their pathogenic mechanisms and constructing disease models.


Subject(s)
Dipeptides , Proteins , Humans , Dipeptides/chemistry , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Open Reading Frames , Proteins/chemistry , Glycine , Alanine , Proline , Arginine/genetics , Chromosomes, Human, Pair 9/metabolism
2.
J Neuropathol Exp Neurol ; 80(9): 821-829, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34363682

ABSTRACT

Polymorphous low-grade neuroepithelial tumor of the young (PLNTY) is a recently described epileptogenic tumor characterized by oligodendroglioma-like components, aberrant CD34 expression, and frequent mitogen-activated protein kinase (MAPK) pathway activation. We molecularly profiled 13 cases with diagnostic histopathological features of PLNTY (10 female; median age, 16 years; range, 5-52). Patients frequently presented with seizures (9 of 12 with available history) and temporal lobe tumors (9 of 13). MAPK pathway activating alterations were identified in all 13 cases. Fusions were present in the 7 youngest patients: FGFR2-CTNNA3 (n = 2), FGFR2-KIAA1598 (FGFR2-SHTN1) (n = 1), FGFR2-INA (n = 1), FGFR2-MPRIP (n = 1), QKI-NTRK2 (n = 1), and KIAA1549-BRAF (n = 1). BRAF V600E mutation was present in 6 patients (17 years or older). Two fusion-positive cases additionally harbored TP53/RB1 abnormalities suggesting biallelic inactivation. Copy number changes predominantly involving whole chromosomes were observed in all 10 evaluated cases, with losses of chromosome 10q occurring with FGFR2-KIAA1598 (SHTN1)/CTNNA3 fusions. The KIAA1549-BRAF and QKI-NTRK2 fusions were associated respectively with a 7q34 deletion and 9q21 duplication. This study shows that despite its name, PLNTY also occurs in older adults, who frequently show BRAF V600E mutation. It also expands the spectrum of the MAPK pathway activating alterations associated with PLNTY and demonstrates recurrent chromosomal copy number changes consistent with chromosomal instability.


Subject(s)
Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasms, Neuroepithelial/metabolism , Receptor, trkB/metabolism , Seizures/metabolism , Adult , Aneuploidy , Chromosomes, Human, Pair 9/metabolism , Female , Gene Fusion/physiology , Humans , Recurrence , Seizures/genetics , Transcription Factors/metabolism
3.
Hematol Oncol ; 39(3): 419-422, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33369766

ABSTRACT

Primary mediastinal large B-cell lymphoma (PMBCL) is a rare aggressive B-cell lymphoma characterized by the frequent presence of amplification and translocation events at 9p24.1, resulting in the expression of the programmed cell death-1 (PD-1) ligands PD-L1 and PD-L2. Pembrolizumab, a humanized anti-PD-1 monoclonal antibody, binds PD-1 and blocks this interaction, enhancing the activity of the immune system against tumor cells, and has shown activity in PMBCL and in some cases of primary and secondary central nervous system (CNS) lymphoma. We report the case of a 40-year-old woman diagnosed with relapsed PMBCL and secondary CNS involvement who responded to pembrolizumab monotherapy, allowing for a later allogeneic stem cell transplant.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Central Nervous System Neoplasms/therapy , Lymphoma, B-Cell/therapy , Mediastinal Neoplasms/therapy , Stem Cell Transplantation , Adult , Allografts , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Female , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Mediastinal Neoplasms/genetics , Mediastinal Neoplasms/metabolism , Mediastinal Neoplasms/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Translocation, Genetic , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
4.
Leuk Res ; 98: 106453, 2020 11.
Article in English | MEDLINE | ID: mdl-33059120

ABSTRACT

We described the clinical features and outcomes for 63 adult patients with acute myeloid leukemia (AML) with a translocation involving the 11q23 locus (MLL) who were treated at Memorial Sloan Kettering Cancer Center (MSK). The population included 40 female (63 %) and 23 male (37 %) patients, with a median age of 51 years old (range 18-82 years). Of the 31 patients who had had an antecedent malignancy, 14 (45 %) had had breast cancer or DCIS and 22 (71 %) had received anthracycline-based systemic chemotherapy. The translocation partner for the 11q23 rearrangement was identified in 60 of the 63 patients (95 %) studied. The distribution of translocation partners differed for those who had previously received cytotoxic chemotherapy. Most patients with therapy-related disease had a 9p22 or 19p13 partner, as compared to those with de novo disease (95 % vs. 68 %, p = 0.023). Of the 30 patients who received all therapy under observation, 15 (50 %) patients had de novo disease and 15 (50 %) had received antecedent chemotherapy. No significant difference in survival was observed between groups (p = 0.44). Twenty-two patients received induction as up-front therapy, of whom 11 (50 %) achieved CR / CRi. The achievement of CR / CRi with one course of induction was associated with improved OS, with a 6-month OS of 73 % as compared to 23 % for those who did not (p = 0.018). The achievement of CR / CRi with a single course of induction may be a marker of favorable survival in this subtype of high-risk AML. KEY POINT: Response to a single induction was associated with favorable survival in this population.


Subject(s)
Anthracyclines/administration & dosage , Breast Neoplasms , Chromosomes, Human, Pair 11 , Leukemia, Myeloid, Acute , Neoplasms, Second Primary , Translocation, Genetic , Adolescent , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/metabolism , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 19/metabolism , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Neoplasms, Second Primary/drug therapy , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/metabolism , Neoplasms, Second Primary/pathology
5.
Hematol Oncol ; 38(4): 607-610, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32602167
6.
Circ Genom Precis Med ; 13(3): e002806, 2020 06.
Article in English | MEDLINE | ID: mdl-32396387

ABSTRACT

BACKGROUND: Common chromosome 9p21 single nucleotide polymorphisms (SNPs) increase coronary heart disease risk, independent of traditional lipid risk factors. However, lipids comprise large numbers of structurally related molecules not measured in traditional risk measurements, and many have inflammatory bioactivities. Here, we applied lipidomic and genomic approaches to 3 model systems to characterize lipid metabolic changes in common Chr9p21 SNPs, which confer ≈30% elevated coronary heart disease risk associated with altered expression of ANRIL, a long ncRNA. METHODS: Untargeted and targeted lipidomics was applied to plasma from NPHSII (Northwick Park Heart Study II) homozygotes for AA or GG in rs10757274, followed by correlation and network analysis. To identify candidate genes, transcriptomic data from shRNA downregulation of ANRIL in HEK-293 cells was mined. Transcriptional data from vascular smooth muscle cells differentiated from induced pluripotent stem cells of individuals with/without Chr9p21 risk, nonrisk alleles, and corresponding knockout isogenic lines were next examined. Last, an in-silico analysis of miRNAs was conducted to identify how ANRIL might control lysoPL (lysophosphospholipid)/lysoPA (lysophosphatidic acid) genes. RESULTS: Elevated risk GG correlated with reduced lysoPLs, lysoPA, and ATX (autotaxin). Five other risk SNPs did not show this phenotype. LysoPL-lysoPA interconversion was uncoupled from ATX in GG plasma, suggesting metabolic dysregulation. Significantly altered expression of several lysoPL/lysoPA metabolizing enzymes was found in HEK cells lacking ANRIL. In the vascular smooth muscle cells data set, the presence of risk alleles associated with altered expression of several lysoPL/lysoPA enzymes. Deletion of the risk locus reversed the expression of several lysoPL/lysoPA genes to nonrisk haplotype levels. Genes that were altered across both cell data sets were DGKA, MBOAT2, PLPP1, and LPL. The in-silico analysis identified 4 ANRIL-regulated miRNAs that control lysoPL genes as miR-186-3p, miR-34a-3p, miR-122-5p, and miR-34a-5p. CONCLUSIONS: A Chr9p21 risk SNP associates with complex alterations in immune-bioactive phospholipids and their metabolism. Lipid metabolites and genomic pathways associated with coronary heart disease pathogenesis in Chr9p21 and ANRIL-associated disease are demonstrated.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Coronary Disease , Lysophospholipids , Phosphoric Diester Hydrolases , Polymorphism, Single Nucleotide , Chromosomes, Human, Pair 9/metabolism , Coronary Disease/genetics , Coronary Disease/metabolism , HEK293 Cells , Humans , Lysophospholipids/genetics , Lysophospholipids/metabolism , Male , Middle Aged , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism
7.
Nat Commun ; 10(1): 4928, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666522

ABSTRACT

Kleefstra syndrome (KS) is a neurodevelopmental disorder caused by mutations in the histone methyltransferase EHMT1. To study the impact of decreased EHMT1 function in human cells, we generated excitatory cortical neurons from induced pluripotent stem (iPS) cells derived from KS patients. Neuronal networks of patient-derived cells exhibit network bursting with a reduced rate, longer duration, and increased temporal irregularity compared to control networks. We show that these changes are mediated by upregulation of NMDA receptor (NMDAR) subunit 1 correlating with reduced deposition of the repressive H3K9me2 mark, the catalytic product of EHMT1, at the GRIN1 promoter. In mice EHMT1 deficiency leads to similar neuronal network impairments with increased NMDAR function. Finally, we rescue the KS patient-derived neuronal network phenotypes by pharmacological inhibition of NMDARs. Summarized, we demonstrate a direct link between EHMT1 deficiency and NMDAR hyperfunction in human neurons, providing a potential basis for more targeted therapeutic approaches for KS.


Subject(s)
Craniofacial Abnormalities/genetics , Heart Defects, Congenital/genetics , Histone-Lysine N-Methyltransferase/genetics , Intellectual Disability/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Cerebral Cortex/cytology , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Craniofacial Abnormalities/metabolism , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Heart Defects, Congenital/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Induced Pluripotent Stem Cells , Intellectual Disability/metabolism , Loss of Function Mutation , Male , Mice , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Primary Cell Culture , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Up-Regulation
8.
Clin Biochem ; 73: 70-76, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31386834

ABSTRACT

BACKGROUND: Chromosomal region 9p21.3 is most robustly associated with coronary artery disease (CAD) in western European populations. However, heterogeneity in CAD phenotypes leads to uncertainty whether 9p21.3 is associated with stable and/or acute clinical presentations of CAD. 9p21.3 is rich in regulatory elements, but the underlying mechanisms of its actions in CAD remain unclear. We investigate the association of 9p21.3 two haplotype blocks lead variants (rs10757278 and rs518394) with first-ever non-fatal myocardial infarction (MI) in CAD patients and their association with CDKN2B mRNA expression in peripheral blood mononuclear cells 6 months after the event. METHODS: We included CAD patients with sustained first MI (n = 523) and controls (n = 583). Gene expression was assessed in 72 patients 6 months after MI and 43 healthy controls. TaqMan® technology was used for the gene expression and genotyping analysis. RESULTS: CDKN2B mRNA was significantly lower in MI patients compared with the controls (p = 0.002) and in patients carrying the rs10757278 G risk allele versus AA homozygotes (p = 0.012) 6 months after the event. While we confirmed the association of rs10757278 with CDKN2B expression in MI patients, we failed to find an association between the investigated variants and MI or disease burden. CONCLUSIONS: We suggest a dysregulation of gene expression in the 9p21.3 region six months after acute MI, which is affected by a genetic variant in patients. The rs10757278 rare allele is one factor that might lead to prolonged risk for proatherogenic complications.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Coronary Artery Disease , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , Gene Expression Regulation , Haplotypes , Myocardial Infarction , Response Elements , Adult , Aged , Alleles , Chromosomes, Human, Pair 9/metabolism , Coronary Artery Disease/enzymology , Coronary Artery Disease/genetics , Cyclin-Dependent Kinase Inhibitor p15/genetics , Female , Follow-Up Studies , Homozygote , Humans , Male , Middle Aged , Myocardial Infarction/enzymology , Myocardial Infarction/genetics
9.
J Interferon Cytokine Res ; 39(10): 599-608, 2019 10.
Article in English | MEDLINE | ID: mdl-31070498

ABSTRACT

Humans are polymorphic in their ability to produce type-III interferons. Most individuals of African ancestry are genetically capable of generating all 4 type-III interferons (IFN-λ1, 2, 3, and 4), whereas the majority of individuals of European and Asian ancestry lack IFN-λ4 and thus can generate only IFN-λ1, 2, and 3. All 4 type-III IFNs are encoded by genes located within a ∼55 kb genomic region on human chromosome 19. Although IFN-λ4 appears to be important in animals, genetic alterations acquired in the Hominidae lineage, and particularly in humans, resulted in the elimination of IFN-λ4 or restriction of its activity, suggesting that IFN-λ4 function might be detrimental to human health. Genetic variants within the IFNL region, including those controlling production and activity of IFN-λ4, have been strongly associated with clearance of hepatitis C virus (HCV) infection. There is growing evidence for association of the same genetic variants with a multitude of other disease conditions. This article reviews the genetic landscape of the human IFNL genetic locus, with an emphasis on the genetic control of IFN-λ4 production and activity, and its association with viral clearance.


Subject(s)
Chromosomes, Human, Pair 9 , Genetic Loci , Genetic Variation , Hepacivirus/metabolism , Hepatitis C , Interferons , Animals , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatitis C/pathology , Humans , Interferons/genetics , Interferons/metabolism , Interferon Lambda
11.
Nat Commun ; 10(1): 368, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30664630

ABSTRACT

The molecular pathogenesis of salivary gland acinic cell carcinoma (AciCC) is poorly understood. The secretory Ca-binding phosphoprotein (SCPP) gene cluster at 4q13 encodes structurally related phosphoproteins of which some are specifically expressed at high levels in the salivary glands and constitute major components of saliva. Here we report on recurrent rearrangements [t(4;9)(q13;q31)] in AciCC that translocate active enhancer regions from the SCPP gene cluster to the region upstream of Nuclear Receptor Subfamily 4 Group A Member 3 (NR4A3) at 9q31. We show that NR4A3 is specifically upregulated in AciCCs, and that active chromatin regions and gene expression signatures in AciCCs are highly correlated with the NR4A3 transcription factor binding motif. Overexpression of NR4A3 in mouse salivary gland cells increases expression of known NR4A3 target genes and has a stimulatory functional effect on cell proliferation. We conclude that NR4A3 is upregulated through enhancer hijacking and has important oncogenic functions in AciCC.


Subject(s)
Carcinoma, Acinar Cell/genetics , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Salivary Gland Neoplasms/genetics , Salivary Proteins and Peptides/genetics , Translocation, Genetic , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Carcinoma, Acinar Cell/metabolism , Carcinoma, Acinar Cell/pathology , Cell Proliferation , Chromatin/chemistry , Chromatin/metabolism , Chromosomes, Human, Pair 4/chemistry , Chromosomes, Human, Pair 4/metabolism , Chromosomes, Human, Pair 9/chemistry , Chromosomes, Human, Pair 9/metabolism , Cohort Studies , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Female , Genetic Loci , Humans , Male , Mice , Multigene Family , Primary Cell Culture , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Salivary Gland Neoplasms/metabolism , Salivary Gland Neoplasms/pathology , Salivary Glands/metabolism , Salivary Glands/pathology , Salivary Proteins and Peptides/metabolism
13.
Cell ; 175(7): 1796-1810.e20, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30528432

ABSTRACT

The 9p21.3 cardiovascular disease locus is the most influential common genetic risk factor for coronary artery disease (CAD), accounting for ∼10%-15% of disease in non-African populations. The ∼60 kb risk haplotype is human-specific and lacks coding genes, hindering efforts to decipher its function. Here, we produce induced pluripotent stem cells (iPSCs) from risk and non-risk individuals, delete each haplotype using genome editing, and generate vascular smooth muscle cells (VSMCs). Risk VSMCs exhibit globally altered transcriptional networks that intersect with previously identified CAD risk genes and pathways, concomitant with aberrant adhesion, contraction, and proliferation. Unexpectedly, deleting the risk haplotype rescues VSMC stability, while expressing the 9p21.3-associated long non-coding RNA ANRIL induces risk phenotypes in non-risk VSMCs. This study shows that the risk haplotype selectively predisposes VSMCs to adopt a cell state associated with CAD phenotypes, defines new VSMC-based networks of CAD risk genes, and establishes haplotype-edited iPSCs as powerful tools for functionally annotating the human genome.


Subject(s)
Chromosomes, Human, Pair 9 , Coronary Artery Disease , Gene Editing , Haplotypes , Induced Pluripotent Stem Cells , Polymorphism, Single Nucleotide , Aged , Aged, 80 and over , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Female , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription, Genetic
14.
Hematology Am Soc Hematol Educ Program ; 2018(1): 213-220, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504313

ABSTRACT

Genetic alterations of the PD-L1/PD-L2 locus on chromosome 9p24.1 are a defining biological feature of classical Hodgkin lymphoma (HL). The resulting programmed death-ligand 1 (PD-L1) expression on Hodgkin Reed-Sternberg cells as well as the PD-L1 expressed in the HL microenvironment result in an ineffective host antitumor immune response and make HL a ripe target for programmed cell death-1 (PD-1) blockade. Anti-PD-1 antibody monotherapy has been effective and well tolerated in patients with relapsed or refractory (rel/ref) HL, with the majority of patients experiencing an objective response (approximately two-thirds of patients) and a median duration of response of 16.6 months in the study with the longest follow-up. Based on these data, nivolumab and pembrolizumab were approved by the US Food and Drug Administration (FDA) for the treatment of advanced rel/ref HL. Evidence has emerged that patients with HL benefit from continued PD-1 blockade beyond disease progression according to traditionally defined response criteria, and that the addition of, or switch to, chemotherapy after anti-PD-1 antibody failure can potentially re-induce clinical response. Subsequent studies have evaluated novel anti-PD-1-based combination regimens as well as the use of anti-PD-1 antibody therapy earlier in the course of a HL patient's therapy, including first salvage therapy for rel/ref disease (eg, nivolumab plus brentuximab vedotin) and even first-line treatment (eg, nivolumab added to doxorubicin, vinblastine, dacarbazine chemotherapy). The current role of PD-1 blockade in HL is as monotherapy in patients with advanced rel/ref disease, but the results of ongoing studies and the evolving treatment landscape in HL will determine the role of PD-1 blockade in the future.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Hodgkin Disease , Neoplasm Proteins/antagonists & inhibitors , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Hodgkin Disease/drug therapy , Hodgkin Disease/genetics , Hodgkin Disease/metabolism , Hodgkin Disease/pathology , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Reed-Sternberg Cells/metabolism , Reed-Sternberg Cells/pathology
15.
PLoS One ; 13(8): e0202365, 2018.
Article in English | MEDLINE | ID: mdl-30138332

ABSTRACT

BACKGROUND: Single nucleotide polymorphisms (SNPs) in the 9p21 region have been associated with cardiovascular disease (CVD), but previous studies have focussed on older populations. The objective of this study was to determine the association between 9p21 genotypes and biomarkers of CVD risk in young adults from different ethnocultural groups. METHODS: Subjects were 1,626 participants aged 20-29 years from the Toronto Nutrigenomics and Health Study. Fasting blood was collected to measure glucose, insulin, c-reactive protein and serum lipids, as well as to isolate DNA for genotyping subjects for five SNPs in 9p21. Analyses were conducted for the entire population and separately for women (n = 1,109), men (n = 517), Caucasians (n = 771), East Asians (n = 561) South Asians (n = 175) and Others (n = 119). ANOVA and ANCOVA were used to examine if 9p21 genotypes were associated with biomarkers of CVD risk. RESULTS: In the entire group, the risk alleles of rs10757278 and rs2383206 were associated with higher mean insulin (p = 0.01). Risk alleles for rs4977574, rs10757278, rs2383206, rs1333049 and rs10757274 were associated with higher serum insulin in women (p = 0.008, p = 0.008, p = 0.0003, p = 0.002, and p = 0.001, respectively), but not in men (p = 0.41, p = 0.13, p = 0.31, p = 0.34, and 0.35, respectively). The association between 9p21 and insulin remained significant among women not taking hormonal contraceptives (HC), but was not significant among women taking HCs. CONCLUSION: Our findings suggest that 9p21 genotypes may play a role in the development of insulin resistance in early adulthood among women, but not men, and the effects appear to be attenuated by HC use.


Subject(s)
Alleles , Chromosomes, Human, Pair 9/genetics , Fasting/blood , Insulin , Polymorphism, Single Nucleotide , Sex Characteristics , Adult , Cardiovascular Diseases/blood , Cardiovascular Diseases/genetics , Chromosomes, Human, Pair 9/metabolism , Female , Humans , Insulin/blood , Insulin/genetics , Insulin Resistance/genetics , Male , Risk Factors
16.
Curr Hematol Malig Rep ; 13(4): 275-288, 2018 08.
Article in English | MEDLINE | ID: mdl-29951888

ABSTRACT

PURPOSE OF REVIEW: Acute lymphoblastic leukemias (ALL) are malignant disorders of immature B or T cells that occur characteristically in children, usually under the age of 6 (75%). Approximately 6000 new cases of ALL are diagnosed each year in the USA, 80-85% of which represent B-ALL forms. Most presentations of B-ALL are leukemic, whereas T-ALL presents with a mediastinal mass, with or without leukemic involvement. The revised fourth edition of the World Health Organization (WHO) classification (2017) has introduced some changes in both B and T-ALL. Here, we summarize the categories of lymphoblastic leukemia/lymphomas as defined by the WHO and recent developments in the understanding of this group of hematologic malignancy. RECENT FINDINGS: Two provisional categories of B-ALL have now been identified including B-ALL, BCR-ABL1-like, and B-ALL with iAMP21. The Philadelphia chromosome-like B-ALL includes forms of the disease that shares the expression profiling of B-ALL with t(9;22) but lack such rearrangement. The second one shows amplification of part of the chromosome 21. Both entities are associated with worse prognosis. Within the T-ALL group, an early precursor T cell form has now been introduced as a provisional category. Such group demonstrates expression of stem cell and myeloid markers in conjunction with the T cell antigens. The current review summarizes the recent updates to the WHO classification.


Subject(s)
Hematopoiesis , Lymphoid Tissue , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Child, Preschool , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 21/metabolism , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Female , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Infant , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Male , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/classification , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/classification , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Translocation, Genetic , World Health Organization
17.
Nucleic Acids Res ; 46(10): 4950-4965, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29554304

ABSTRACT

Kleefstra syndrome, a disease with intellectual disability, autism spectrum disorders and other developmental defects is caused in humans by haploinsufficiency of EHMT1. Although EHMT1 and its paralog EHMT2 were shown to be histone methyltransferases responsible for deposition of the di-methylated H3K9 (H3K9me2), the exact nature of epigenetic dysfunctions in Kleefstra syndrome remains unknown. Here, we found that the epigenome of Ehmt1+/- adult mouse brain displays a marked increase of H3K9me2/3 which correlates with impaired expression of protocadherins, master regulators of neuronal diversity. Increased H3K9me3 was present already at birth, indicating that aberrant methylation patterns are established during embryogenesis. Interestingly, we found that Ehmt2+/- mice do not present neither the marked increase of H3K9me2/3 nor the cognitive deficits found in Ehmt1+/- mice, indicating an evolutionary diversification of functions. Our finding of increased H3K9me3 in Ehmt1+/- mice is the first one supporting the notion that EHMT1 can quench the deposition of tri-methylation by other Histone methyltransferases, ultimately leading to impaired neurocognitive functioning. Our insights into the epigenetic pathophysiology of Kleefstra syndrome may offer guidance for future developments of therapeutic strategies for this disease.


Subject(s)
Cadherins/genetics , Cognitive Dysfunction/metabolism , Craniofacial Abnormalities/metabolism , Heart Defects, Congenital/metabolism , Histones/metabolism , Intellectual Disability/metabolism , Animals , Animals, Newborn , Cadherins/metabolism , Chromosome Deletion , Chromosomes, Human, Pair 9/metabolism , Cognitive Dysfunction/genetics , Craniofacial Abnormalities/psychology , Disease Models, Animal , Gene Expression Regulation , Heart Defects, Congenital/psychology , Hippocampus/metabolism , Histone-Lysine N-Methyltransferase/genetics , Intellectual Disability/psychology , Lysine/metabolism , Male , Methylation , Mice, Knockout
19.
PLoS One ; 13(2): e0193213, 2018.
Article in English | MEDLINE | ID: mdl-29489901

ABSTRACT

OBJECTIVE: To study the presence of 9p deletion and p16, cyclin D1 and Myc expression and their respective diagnostic and prognostic interest in oligodendrogliomas. METHODS: We analyzed a retrospective series of 40 consecutive anaplastic oligodendrogliomas (OIII) from a single institution and compared them to a control series of 10 low grade oligodendrogliomas (OII). Automated FISH analysis of chromosome 9p status and immunohistochemistry for p16, cyclin D1 and Myc was performed for all cases and correlated with clinical and histological data, event free survival (EFS) and overall survival (OS). RESULTS: Chromosome 9p deletion was observed in 55% of OIII (22/40) but not in OII. Deletion was highly correlated to EFS (median = 29 versus 53 months, p<0.0001) and OS (median = 48 versus 83 months, p<0.0001) in both the total cohort and the OIII population. In 9p non-deleted oligodendrogliomas, p16 hyperexpression correlated with a shorter OS (p = 0.02 in OII and p = 0.0001 in OIII) whereas lack of p16 expression was correlated to a shorter EFS and OS in 9p deleted OIII (p = 0.001 and p = 0.0002 respectively). Expression of Cyclin D1 was significantly higher in OIII (median expression 45% versus 14% for OII, p = 0.0006) and was correlated with MIB-1 expression (p<0.0001), vascular proliferation (p = 0.002), tumor necrosis (p = 0.04) and a shorter EFS in the total cohort (p = 0.05). Hyperexpression of Myc was correlated to grade (median expression 27% in OII versus 35% in OIII, p = 0.03), and to a shorter EFS in 9p non-deleted OIII (p = 0.01). CONCLUSION: Chromosome 9p deletion identifies a subset of OIII with significantly worse prognosis. The combination of 9p status and p16 expression level identifies two distinct OIII populations with divergent prognosis. Hyperexpression of Bcl1 and Myc appears highly linked to anaplasia but the prognostic value is unclear and should be investigated further.


Subject(s)
Brain Neoplasms , Chromosome Deletion , Cyclin D1 , Cyclin-Dependent Kinase Inhibitor p16 , Gene Expression Regulation, Neoplastic , Oligodendroglioma , Proto-Oncogene Proteins c-myc , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease-Free Survival , Female , Humans , Male , Middle Aged , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , Oligodendroglioma/mortality , Oligodendroglioma/pathology , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Survival Rate
20.
J Hum Genet ; 63(5): 555-562, 2018 May.
Article in English | MEDLINE | ID: mdl-29459631

ABSTRACT

Kleefstra syndrome (KS) (9q34 deletion syndrome) is a rare autosomal dominant disorder characterized by intellectual disability, frequently coupled with a spectrum of complex physical and clinical manifestations. As the euchromatic histone methyltransferase-1 gene (EHMT1, GLP, or KMT1D) within the 9q34 region is deleted or mutated in most of the individuals with KS, its absence or defect in one allele is speculated to cause the major symptoms of the syndrome. Most of the EHMT1 mutations are frameshift or nonsense mutations, but two individuals with KS were reported to possess EHMT1 missense mutations. These two mutations have been predicted to cause a defective enzymatic function, but precise biochemical validation was not conducted. Therefore, we validated these two mutations by performing in vitro histone methyltransferase (HMT) activity assay and found that C1073Y and R1197W mutations severely affected the HMT activity. Additionally, the same amino-acid substitutions in mouse GLP induced impairment of in vivo GLP function. Furthermore, these two EHMT1 mutants showed defective heterocomplex formation with G9a (partner HMT) which is essential for their in vivo HMT function. Conclusively, our biochemical characterization clearly demonstrates that the previously reported two missense mutations of EHMT1 deteriorate HMT activity and GLP function, which presumably cause KS.


Subject(s)
Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Histone-Lysine N-Methyltransferase/genetics , Intellectual Disability/genetics , Intellectual Disability/metabolism , Mutation, Missense , Amino Acid Substitution , Animals , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Chromosomes, Human, Pair 9/metabolism , Disease Models, Animal , Embryonic Stem Cells/metabolism , Female , Histocompatibility Antigens , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Methylation , Mice , Mice, Knockout , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...