Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nat Genet ; 54(12): 1881-1894, 2022 12.
Article in English | MEDLINE | ID: mdl-36471067

ABSTRACT

Histone 3 lysine27-to-methionine (H3-K27M) mutations most frequently occur in diffuse midline gliomas (DMGs) of the childhood pons but are also increasingly recognized in adults. Their potential heterogeneity at different ages and midline locations is vastly understudied. Here, through dissecting the single-cell transcriptomic, epigenomic and spatial architectures of a comprehensive cohort of patient H3-K27M DMGs, we delineate how age and anatomical location shape glioma cell-intrinsic and -extrinsic features in light of the shared driver mutation. We show that stem-like oligodendroglial precursor-like cells, present across all clinico-anatomical groups, display varying levels of maturation dependent on location. We reveal a previously underappreciated relationship between mesenchymal cancer cell states and age, linked to age-dependent differences in the immune microenvironment. Further, we resolve the spatial organization of H3-K27M DMG cell populations and identify a mitotic oligodendroglial-lineage niche. Collectively, our study provides a powerful framework for rational modeling and therapeutic interventions.


Subject(s)
Glioma , Humans , Child , Glioma/genetics , Histones/genetics , Methionine , Mutation , Racemethionine , Tumor Microenvironment/genetics
2.
Cancer Discov ; 12(12): 2880-2905, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36305736

ABSTRACT

Diffuse midline gliomas are uniformly fatal pediatric central nervous system cancers that are refractory to standard-of-care therapeutic modalities. The primary genetic drivers are a set of recurrent amino acid substitutions in genes encoding histone H3 (H3K27M), which are currently undruggable. These H3K27M oncohistones perturb normal chromatin architecture, resulting in an aberrant epigenetic landscape. To interrogate for epigenetic dependencies, we performed a CRISPR screen and show that patient-derived H3K27M-glioma neurospheres are dependent on core components of the mammalian BAF (SWI/SNF) chromatin remodeling complex. The BAF complex maintains glioma stem cells in a cycling, oligodendrocyte precursor cell-like state, in which genetic perturbation of the BAF catalytic subunit SMARCA4 (BRG1), as well as pharmacologic suppression, opposes proliferation, promotes progression of differentiation along the astrocytic lineage, and improves overall survival of patient-derived xenograft models. In summary, we demonstrate that therapeutic inhibition of the BAF complex has translational potential for children with H3K27M gliomas. SIGNIFICANCE: Epigenetic dysregulation is at the core of H3K27M-glioma tumorigenesis. Here, we identify the BRG1-BAF complex as a critical regulator of enhancer and transcription factor landscapes, which maintain H3K27M glioma in their progenitor state, precluding glial differentiation, and establish pharmacologic targeting of the BAF complex as a novel treatment strategy for pediatric H3K27M glioma. See related commentary by Beytagh and Weiss, p. 2730. See related article by Mo et al., p. 2906.


Subject(s)
Epigenome , Glioma , Animals , Humans , Mutation , Glioma/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Neoplastic Stem Cells/metabolism , Mammals/genetics , Mammals/metabolism , DNA Helicases/genetics , Nuclear Proteins/genetics
3.
Cancer Immunol Res ; 10(8): 996-1012, 2022 08 03.
Article in English | MEDLINE | ID: mdl-35706413

ABSTRACT

Melanoma-derived brain metastases (MBM) represent an unmet clinical need because central nervous system progression is frequently an end stage of the disease. Immune checkpoint inhibitors (ICI) provide a clinical opportunity against MBM; however, the MBM tumor microenvironment (TME) has not been fully elucidated in the context of ICI. To dissect unique elements of the MBM TME and correlates of MBM response to ICI, we collected 32 fresh MBM and performed single-cell RNA sequencing of the MBM TME and T-cell receptor clonotyping on T cells from MBM and matched blood and extracranial lesions. We observed myeloid phenotypic heterogeneity in the MBM TME, most notably multiple distinct neutrophil states, including an IL8-expressing population that correlated with malignant cell epithelial-to-mesenchymal transition. In addition, we observed significant relationships between intracranial T-cell phenotypes and the distribution of T-cell clonotypes intracranially and peripherally. We found that the phenotype, clonotype, and overall number of MBM-infiltrating T cells were associated with response to ICI, suggesting that ICI-responsive MBMs interact with peripheral blood in a manner similar to extracranial lesions. These data identify unique features of the MBM TME that may represent potential targets to improve clinical outcomes for patients with MBM.


Subject(s)
Brain Neoplasms , Melanoma , Humans , Immune Checkpoint Inhibitors , Tumor Microenvironment
4.
Cancer Cell ; 38(1): 44-59.e9, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32663469

ABSTRACT

Ependymoma is a heterogeneous entity of central nervous system tumors with well-established molecular groups. Here, we apply single-cell RNA sequencing to analyze ependymomas across molecular groups and anatomic locations to investigate their intratumoral heterogeneity and developmental origins. Ependymomas are composed of a cellular hierarchy initiating from undifferentiated populations, which undergo impaired differentiation toward three lineages of neuronal-glial fate specification. While prognostically favorable groups of ependymoma predominantly harbor differentiated cells, aggressive groups are enriched for undifferentiated cell populations. The delineated transcriptomic signatures correlate with patient survival and define molecular dependencies for targeted treatment approaches. Taken together, our analyses reveal a developmental hierarchy underlying ependymomas relevant to biological and clinical behavior.


Subject(s)
Central Nervous System Neoplasms/genetics , Ependymoma/genetics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Cell Differentiation/genetics , Cell Proliferation/genetics , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/therapy , Child , Ependymoma/pathology , Ependymoma/therapy , Genomics/methods , Humans , Neurons/metabolism , Neurons/pathology , Prognosis , Survival Analysis
5.
Cell ; 178(4): 835-849.e21, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31327527

ABSTRACT

Diverse genetic, epigenetic, and developmental programs drive glioblastoma, an incurable and poorly understood tumor, but their precise characterization remains challenging. Here, we use an integrative approach spanning single-cell RNA-sequencing of 28 tumors, bulk genetic and expression analysis of 401 specimens from the The Cancer Genome Atlas (TCGA), functional approaches, and single-cell lineage tracing to derive a unified model of cellular states and genetic diversity in glioblastoma. We find that malignant cells in glioblastoma exist in four main cellular states that recapitulate distinct neural cell types, are influenced by the tumor microenvironment, and exhibit plasticity. The relative frequency of cells in each state varies between glioblastoma samples and is influenced by copy number amplifications of the CDK4, EGFR, and PDGFRA loci and by mutations in the NF1 locus, which each favor a defined state. Our work provides a blueprint for glioblastoma, integrating the malignant cell programs, their plasticity, and their modulation by genetic drivers.


Subject(s)
Brain Neoplasms/genetics , Cell Plasticity/genetics , Glioblastoma/genetics , Adolescent , Aged , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Lineage/genetics , Child , Cohort Studies , Disease Models, Animal , Female , Genetic Heterogeneity , Glioblastoma/pathology , Heterografts , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Middle Aged , Mutation , RNA-Seq , Single-Cell Analysis/methods , Tumor Microenvironment/genetics
6.
Nature ; 572(7767): 74-79, 2019 08.
Article in English | MEDLINE | ID: mdl-31341285

ABSTRACT

Medulloblastoma is a malignant childhood cerebellar tumour type that comprises distinct molecular subgroups. Whereas genomic characteristics of these subgroups are well defined, the extent to which cellular diversity underlies their divergent biology and clinical behaviour remains largely unexplored. Here we used single-cell transcriptomics to investigate intra- and intertumoral heterogeneity in 25 medulloblastomas spanning all molecular subgroups. WNT, SHH and Group 3 tumours comprised subgroup-specific undifferentiated and differentiated neuronal-like malignant populations, whereas Group 4 tumours consisted exclusively of differentiated neuronal-like neoplastic cells. SHH tumours closely resembled granule neurons of varying differentiation states that correlated with patient age. Group 3 and Group 4 tumours exhibited a developmental trajectory from primitive progenitor-like to more mature neuronal-like cells, the relative proportions of which distinguished these subgroups. Cross-species transcriptomics defined distinct glutamatergic populations as putative cells-of-origin for SHH and Group 4 subtypes. Collectively, these data provide insights into the cellular and developmental states underlying subtype-specific medulloblastoma biology.


Subject(s)
Genomics , Medulloblastoma/genetics , Medulloblastoma/pathology , Single-Cell Analysis , Transcriptome , Adolescent , Adult , Animals , Cell Lineage , Cerebellum/metabolism , Cerebellum/pathology , Child , Child, Preschool , DNA Copy Number Variations , Gene Expression Regulation, Neoplastic , Glutamic Acid/metabolism , Humans , Infant , Medulloblastoma/classification , Mice , Neurons/metabolism , Neurons/pathology
7.
Science ; 360(6386): 331-335, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29674595

ABSTRACT

Gliomas with histone H3 lysine27-to-methionine mutations (H3K27M-glioma) arise primarily in the midline of the central nervous system of young children, suggesting a cooperation between genetics and cellular context in tumorigenesis. Although the genetics of H3K27M-glioma are well characterized, their cellular architecture remains uncharted. We performed single-cell RNA sequencing in 3321 cells from six primary H3K27M-glioma and matched models. We found that H3K27M-glioma primarily contain cells that resemble oligodendrocyte precursor cells (OPC-like), whereas more differentiated malignant cells are a minority. OPC-like cells exhibit greater proliferation and tumor-propagating potential than their more differentiated counterparts and are at least in part sustained by PDGFRA signaling. Our study characterizes oncogenic and developmental programs in H3K27M-glioma at single-cell resolution and across genetic subclones, suggesting potential therapeutic targets in this disease.


Subject(s)
Brain Neoplasms/pathology , Carcinogenesis/genetics , Glioma/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oncogenes , Brain Neoplasms/genetics , Cell Proliferation , Glioma/genetics , Histones/metabolism , Humans , Mitogen-Activated Protein Kinase 7/genetics , Mutation , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
8.
J Hum Genet ; 63(4): 459-471, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29371683

ABSTRACT

The apolipoprotein E (APOE) ε4 allele is the major genetic risk factor for Alzheimer's disease (AD). Multiple regulatory elements, spanning the extended TOMM40-APOE-APOC2 region, regulate gene expression at this locus. Regulatory element DNA methylation changes occur under different environmental conditions, such as disease. Our group and others have described an APOE CpG island as hypomethylated in AD, compared to cognitively normal controls. However, little is known about methylation of the larger TOMM40-APOE-APOC2 region. The hypothesis of this investigation was that regulatory element methylation levels of the larger TOMM40-APOE-APOC2 region are associated with AD. The aim was to determine whether DNA methylation of the TOMM40-APOE-APOC2 region differs in AD compared to cognitively normal controls in post-mortem brain and peripheral blood. DNA was extracted from human brain (n = 12) and peripheral blood (n = 67). A methylation array was used for this analysis. Percent methylation within the TOMM40-APOE-APOC2 region was evaluated for differences according to tissue type, disease state, AD-related biomarkers, and gene expression. Results from this exploratory analysis suggest that regulatory element methylation levels within the larger TOMM40-APOE-APOC2 gene region correlate with AD-related biomarkers and TOMM40 or APOE gene expression in AD.


Subject(s)
Alzheimer Disease/genetics , Apolipoprotein C-II/genetics , Apolipoproteins E/genetics , DNA Methylation , Genetic Association Studies , Genetic Predisposition to Disease , Membrane Transport Proteins/genetics , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Biomarkers , Biopsy , Case-Control Studies , Cerebellum/metabolism , Cerebellum/pathology , CpG Islands , Female , Gene Expression , Genetic Loci , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mitochondrial Precursor Protein Import Complex Proteins , Organ Specificity/genetics , Promoter Regions, Genetic
9.
Neurobiol Aging ; 63: 110-119, 2018 03.
Article in English | MEDLINE | ID: mdl-29253717

ABSTRACT

Recent reports in Alzheimer's disease (AD) research suggest that alterations in microRNA (miRNA) expression are associated with disease pathology. Our previous studies suggest that A Disintegrin and Metalloproteinase 10 (ADAM10) expression is important in AD and could be modulated by an extended regulatory region that includes the 3' untranslated region. In this study, we have investigated the role of trans-acting factors in ADAM10 gene regulation. Our study shows that miRNA-140-5p has enhanced expression in the AD postmortem brain hippocampus using high-throughput miRNA arrays and quantitative real-time polymerase chain reaction. Interestingly, we have also seen that miRNA-140-5p seed sequence is present on 3' untranslated region of both ADAM10 and its transcription factor SOX2. The specific interaction of miRNA-140-5p with both ADAM10 and SOX2 signifies high regulatory importance of this miRNA in controlling ADAM10 expression. Thus, this investigation unravels mechanisms underlying ADAM10 downregulation by miR-140-5p and suggests that dysfunctional regulation of ADAM10 expression is exacerbated by AD-related neurotoxic effects. These findings underscore the importance of understanding the impact of trans-acting factors in the modulation of AD pathophysiology.


Subject(s)
ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Gene Expression Regulation/genetics , Genetic Association Studies , Hippocampus/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/physiology , 3' Untranslated Regions , Cell Line , Down-Regulation/genetics , Humans , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction/methods , SOXB1 Transcription Factors/genetics
10.
J Exp Med ; 214(10): 2875-2887, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28878000

ABSTRACT

Recent advances in single-cell, transcriptomic profiling have provided unprecedented access to investigate cell heterogeneity during tissue and organ development. In this study, we used massively parallel, single-cell RNA sequencing to define cell heterogeneity within the zebrafish kidney marrow, constructing a comprehensive molecular atlas of definitive hematopoiesis and functionally distinct renal cells found in adult zebrafish. Because our method analyzed blood and kidney cells in an unbiased manner, our approach was useful in characterizing immune-cell deficiencies within DNA-protein kinase catalytic subunit (prkdc), interleukin-2 receptor γ a (il2rga), and double-homozygous-mutant fish, identifying blood cell losses in T, B, and natural killer cells within specific genetic mutants. Our analysis also uncovered novel cell types, including two classes of natural killer immune cells, classically defined and erythroid-primed hematopoietic stem and progenitor cells, mucin-secreting kidney cells, and kidney stem/progenitor cells. In total, our work provides the first, comprehensive, single-cell, transcriptomic analysis of kidney and marrow cells in the adult zebrafish.


Subject(s)
Hematopoiesis, Extramedullary/genetics , Kidney/cytology , RNA/genetics , Zebrafish/anatomy & histology , Animals , Animals, Genetically Modified , Cell Lineage/genetics , Cell Lineage/physiology , Gene Expression Profiling , Hematopoiesis, Extramedullary/physiology , Hematopoietic Stem Cells , Kidney/metabolism , Sequence Analysis, RNA , Zebrafish/genetics , Zebrafish/metabolism
11.
Science ; 355(6332)2017 03 31.
Article in English | MEDLINE | ID: mdl-28360267

ABSTRACT

Tumor subclasses differ according to the genotypes and phenotypes of malignant cells as well as the composition of the tumor microenvironment (TME). We dissected these influences in isocitrate dehydrogenase (IDH)-mutant gliomas by combining 14,226 single-cell RNA sequencing (RNA-seq) profiles from 16 patient samples with bulk RNA-seq profiles from 165 patient samples. Differences in bulk profiles between IDH-mutant astrocytoma and oligodendroglioma can be primarily explained by distinct TME and signature genetic events, whereas both tumor types share similar developmental hierarchies and lineages of glial differentiation. As tumor grade increases, we find enhanced proliferation of malignant cells, larger pools of undifferentiated glioma cells, and an increase in macrophage over microglia expression programs in TME. Our work provides a unifying model for IDH-mutant gliomas and a general framework for dissecting the differences among human tumor subclasses.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Tumor Microenvironment , Brain Neoplasms/classification , Cell Lineage , Glioma/classification , Humans , Macrophages , Microglia/metabolism , Microglia/pathology , Neoplasm Grading , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Principal Component Analysis , Sequence Analysis, RNA , Single-Cell Analysis
12.
Neurobiol Aging ; 51: 43-53, 2017 03.
Article in English | MEDLINE | ID: mdl-28033507

ABSTRACT

Neurofibrillary tangles (NFTs), composed of hyperphosphorylated tau, are a key pathologic feature of Alzheimer's disease (AD). Tau phosphorylation is under the control of multiple kinases and phosphatases, including Fyn. Previously, our group found an association between 2 regulatory single nucleotide polymorphisms in the FYN gene with increased tau levels in the cerebrospinal fluid. In this study, we hypothesized that Fyn expression in the brain is influenced by AD status and genetic content. We found that Fyn protein, but not messenger RNA, levels were increased in AD patients compared to cognitively normal controls and are associated with regulatory region single nucleotide polymorphisms. In addition, the expression of the FYN 3'UTR can decrease expression in multiple cell lines, suggesting this regulatory region plays an important role in FYN expression. Taken together, these data suggest that FYN expression is regulated according to AD status and regulatory region haplotype, and genetic variants may be instrumental in the development of neurofibrillary tangles in AD and other tauopathies.


Subject(s)
Alzheimer Disease/genetics , Gene Expression/genetics , Genetic Association Studies , Genetic Variation/genetics , Proto-Oncogene Proteins c-fyn/genetics , Regulatory Sequences, Nucleic Acid/genetics , 3' Untranslated Regions , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Brain/metabolism , Cell Line , Female , Haplotypes , Humans , Male , Phosphorylation , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-fyn/metabolism , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...