Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Cancer Cell ; 42(5): 904-914.e9, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38579724

ABSTRACT

A subset of patients with IDH-mutant glioma respond to inhibitors of mutant IDH (IDHi), yet the molecular underpinnings of such responses are not understood. Here, we profiled by single-cell or single-nucleus RNA-sequencing three IDH-mutant oligodendrogliomas from patients who derived clinical benefit from IDHi. Importantly, the tissues were sampled on-drug, four weeks from treatment initiation. We further integrate our findings with analysis of single-cell and bulk transcriptomes from independent cohorts and experimental models. We find that IDHi treatment induces a robust differentiation toward the astrocytic lineage, accompanied by a depletion of stem-like cells and a reduction of cell proliferation. Furthermore, mutations in NOTCH1 are associated with decreased astrocytic differentiation and may limit the response to IDHi. Our study highlights the differentiating potential of IDHi on the cellular hierarchies that drive oligodendrogliomas and suggests a genetic modifier that may improve patient stratification.


Subject(s)
Brain Neoplasms , Cell Differentiation , Isocitrate Dehydrogenase , Mutation , Oligodendroglioma , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Oligodendroglioma/drug therapy , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/antagonists & inhibitors , Humans , Cell Differentiation/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Cell Lineage/drug effects , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Cell Proliferation/drug effects , Animals , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Mice , Single-Cell Analysis/methods
2.
Proc Natl Acad Sci U S A ; 120(6): e2219199120, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36724255

ABSTRACT

Immune checkpoint blockers (ICBs) have failed in all phase III glioblastoma trials. Here, we found that ICBs induce cerebral edema in some patients and mice with glioblastoma. Through single-cell RNA sequencing, intravital imaging, and CD8+ T cell blocking studies in mice, we demonstrated that this edema results from an inflammatory response following antiprogrammed death 1 (PD1) antibody treatment that disrupts the blood-tumor barrier. Used in lieu of immunosuppressive corticosteroids, the angiotensin receptor blocker losartan prevented this ICB-induced edema and reprogrammed the tumor microenvironment, curing 20% of mice which increased to 40% in combination with standard of care treatment. Using a bihemispheric tumor model, we identified a "hot" tumor immune signature prior to losartan+anti-PD1 therapy that predicted long-term survival. Our findings provide the rationale and associated biomarkers to test losartan with ICBs in glioblastoma patients.


Subject(s)
Glioblastoma , Animals , Mice , Glioblastoma/pathology , Losartan/pharmacology , Losartan/therapeutic use , Immune Checkpoint Inhibitors/adverse effects , CD8-Positive T-Lymphocytes , Edema , Tumor Microenvironment
4.
Cancer ; 128(1): 47-58, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34633681

ABSTRACT

The 2016 revised fourth edition of the World Health Organization (WHO) classification of central nervous system (CNS) tumors incorporated molecular features with histologic grading, revolutionizing how oncologists conceptualize primary brain and spinal cord tumors as well as providing new insights into their management and prognosis. The 2021 revised fifth edition of the WHO classification further integrates molecular alterations for CNS tumor categorization, updating current understanding of the pathophysiology of many of these disease entities. Here, the authors review changes in the new classification for the most common primary adult tumors-gliomas (including astrocytomas, oligodendrogliomas, and ependymomas) and meningiomas-highlighting the key genomic alterations for each group classification to help clinicians interpret them as they consider therapeutic options-including clinical trials and targeted therapies-and discuss the prognosis of these tumors with their patients. The revised, updated 2021 WHO classification also further integrates molecular alterations in the classification of pediatric CNS tumors, but those are not covered in the current review.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Glioma , Adult , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Central Nervous System/pathology , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/therapy , Child , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Prognosis , World Health Organization
5.
Nat Genet ; 53(10): 1469-1479, 2021 10.
Article in English | MEDLINE | ID: mdl-34594037

ABSTRACT

Single-cell RNA sequencing has revealed extensive transcriptional cell state diversity in cancer, often observed independently of genetic heterogeneity, raising the central question of how malignant cell states are encoded epigenetically. To address this, here we performed multiomics single-cell profiling-integrating DNA methylation, transcriptome and genotype within the same cells-of diffuse gliomas, tumors characterized by defined transcriptional cell state diversity. Direct comparison of the epigenetic profiles of distinct cell states revealed key switches for state transitions recapitulating neurodevelopmental trajectories and highlighted dysregulated epigenetic mechanisms underlying gliomagenesis. We further developed a quantitative framework to directly measure cell state heritability and transition dynamics based on high-resolution lineage trees in human samples. We demonstrated heritability of malignant cell states, with key differences in hierarchal and plastic cell state architectures in IDH-mutant glioma versus IDH-wild-type glioblastoma, respectively. This work provides a framework anchoring transcriptional cancer cell states in their epigenetic encoding, inheritance and transition dynamics.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Plasticity/genetics , Epigenesis, Genetic , Glioma/genetics , Glioma/pathology , Inheritance Patterns/genetics , Transcription, Genetic , Cell Line, Tumor , CpG Islands/genetics , DNA Copy Number Variations/genetics , DNA Methylation/genetics , Humans , Isocitrate Dehydrogenase/genetics , Phylogeny , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic/genetics , Single-Cell Analysis , Transcriptome/genetics
6.
Cell ; 184(5): 1281-1298.e26, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33592174

ABSTRACT

T cells are critical effectors of cancer immunotherapies, but little is known about their gene expression programs in diffuse gliomas. Here, we leverage single-cell RNA sequencing (RNA-seq) to chart the gene expression and clonal landscape of tumor-infiltrating T cells across 31 patients with isocitrate dehydrogenase (IDH) wild-type glioblastoma and IDH mutant glioma. We identify potential effectors of anti-tumor immunity in subsets of T cells that co-express cytotoxic programs and several natural killer (NK) cell genes. Analysis of clonally expanded tumor-infiltrating T cells further identifies the NK gene KLRB1 (encoding CD161) as a candidate inhibitory receptor. Accordingly, genetic inactivation of KLRB1 or antibody-mediated CD161 blockade enhances T cell-mediated killing of glioma cells in vitro and their anti-tumor function in vivo. KLRB1 and its associated transcriptional program are also expressed by substantial T cell populations in other human cancers. Our work provides an atlas of T cells in gliomas and highlights CD161 and other NK cell receptors as immunotherapy targets.


Subject(s)
Glioma/immunology , NK Cell Lectin-Like Receptor Subfamily B/genetics , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm , Disease Models, Animal , Gene Expression Profiling , Glioma/genetics , Killer Cells, Natural/immunology , Lectins, C-Type/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Receptors, Cell Surface/genetics , Single-Cell Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocytes/cytology , Tumor Escape
7.
Nat Med ; 27(2): 289-300, 2021 02.
Article in English | MEDLINE | ID: mdl-33495604

ABSTRACT

Synovial sarcoma (SyS) is an aggressive neoplasm driven by the SS18-SSX fusion, and is characterized by low T cell infiltration. Here, we studied the cancer-immune interplay in SyS using an integrative approach that combines single-cell RNA sequencing (scRNA-seq), spatial profiling and genetic and pharmacological perturbations. scRNA-seq of 16,872 cells from 12 human SyS tumors uncovered a malignant subpopulation that marks immune-deprived niches in situ and is predictive of poor clinical outcomes in two independent cohorts. Functional analyses revealed that this malignant cell state is controlled by the SS18-SSX fusion, is repressed by cytokines secreted by macrophages and T cells, and can be synergistically targeted with a combination of HDAC and CDK4/CDK6 inhibitors. This drug combination enhanced malignant-cell immunogenicity in SyS models, leading to induced T cell reactivity and T cell-mediated killing. Our study provides a blueprint for investigating heterogeneity in fusion-driven malignancies and demonstrates an interplay between immune evasion and oncogenic processes that can be co-targeted in SyS and potentially in other malignancies.


Subject(s)
Carcinogenesis/genetics , Molecular Targeted Therapy , Oncogene Proteins, Fusion/genetics , Sarcoma, Synovial/drug therapy , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/genetics , Histone Deacetylases/therapeutic use , Humans , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogenes/genetics , RNA-Seq , Sarcoma, Synovial/genetics , Sarcoma, Synovial/pathology , Single-Cell Analysis
9.
Nat Med ; 26(5): 792-802, 2020 05.
Article in English | MEDLINE | ID: mdl-32405060

ABSTRACT

Single-cell genomics is essential to chart tumor ecosystems. Although single-cell RNA-Seq (scRNA-Seq) profiles RNA from cells dissociated from fresh tumors, single-nucleus RNA-Seq (snRNA-Seq) is needed to profile frozen or hard-to-dissociate tumors. Each requires customization to different tissue and tumor types, posing a barrier to adoption. Here, we have developed a systematic toolbox for profiling fresh and frozen clinical tumor samples using scRNA-Seq and snRNA-Seq, respectively. We analyzed 216,490 cells and nuclei from 40 samples across 23 specimens spanning eight tumor types of varying tissue and sample characteristics. We evaluated protocols by cell and nucleus quality, recovery rate and cellular composition. scRNA-Seq and snRNA-Seq from matched samples recovered the same cell types, but at different proportions. Our work provides guidance for studies in a broad range of tumors, including criteria for testing and selecting methods from the toolbox for other tumors, thus paving the way for charting tumor atlases.


Subject(s)
Algorithms , Cell Nucleus/genetics , Genomics/methods , Neoplasms/genetics , RNA-Seq/methods , Single-Cell Analysis/methods , Adult , Animals , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Child , Computational Biology/methods , Female , Freezing , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Knockout , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Sequence Analysis, RNA/methods , Tumor Cells, Cultured , Exome Sequencing/methods
10.
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
11.
Cell ; 177(6): 1583-1599.e16, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31150624

ABSTRACT

T and B cells are the two known lineages of adaptive immune cells. Here, we describe a previously unknown lymphocyte that is a dual expresser (DE) of TCR and BCR and key lineage markers of both B and T cells. In type 1 diabetes (T1D), DEs are predominated by one clonotype that encodes a potent CD4 T cell autoantigen in its antigen binding site. Molecular dynamics simulations revealed that this peptide has an optimal binding register for diabetogenic HLA-DQ8. In concordance, a synthetic version of the peptide forms stable DQ8 complexes and potently stimulates autoreactive CD4 T cells from T1D patients, but not healthy controls. Moreover, mAbs bearing this clonotype are autoreactive against CD4 T cells and inhibit insulin tetramer binding to CD4 T cells. Thus, compartmentalization of adaptive immune cells into T and B cells is not absolute, and violators of this paradigm are likely key drivers of autoimmune diseases.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Adolescent , Adult , Autoantigens/immunology , Child , Child, Preschool , Diabetes Mellitus, Type 1/metabolism , Epitopes/immunology , Female , HEK293 Cells , HLA-DQ Antigens/immunology , HLA-DQ Antigens/ultrastructure , Humans , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Middle Aged , Molecular Dynamics Simulation , Peptides , Protein Binding/immunology
12.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27030713

ABSTRACT

BACKGROUND: Stroke patients often suffer from a central neuropathic pain syndrome called central post-stroke pain. This syndrome is characterized by evoked pain hypersensitivity as well as spontaneous, on-going pain in the body area affected by the stroke. Clinical evidence strongly suggests a dysfunction in central pain pathways as an important pathophysiological factor in the development of central post-stroke pain, but the exact underlying mechanisms remain poorly understood. To elucidate the underlying pathophysiology of central post-stroke pain, we generated a mouse model that is based on a unilateral stereotactic lesion of the thalamic ventral posterolateral nucleus, which typically causes central post-stroke pain in humans. RESULTS: Behavioral analysis showed that the sensory changes in our model are comparable to the sensory abnormalities observed in patients suffering from central post-stroke pain. Surprisingly, pharmacological inhibition of spinal and peripheral key components of the pain system had no effect on the induction or maintenance of the evoked hypersensitivity observed in our model. In contrast, microinjection of lidocaine into the thalamic lesion completely reversed injury-induced hypersensitivity. CONCLUSIONS: These results suggest that the evoked hypersensitivity observed in central post-stroke pain is causally linked to on-going neuronal activity in the lateral thalamus.


Subject(s)
Pain/etiology , Pain/physiopathology , Stroke/complications , Stroke/physiopathology , Animals , Collagenases/administration & dosage , Disease Models, Animal , Hyperalgesia/complications , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Kainic Acid/administration & dosage , Lidocaine/administration & dosage , Mice, Inbred C57BL , Microinjections , Nerve Fibers, Unmyelinated/pathology , Sensation , Spinal Cord/pathology , Spinal Cord/physiopathology , TRPV Cation Channels/metabolism , Thalamus/pathology , Thalamus/physiopathology , Ventral Thalamic Nuclei/pathology , Ventral Thalamic Nuclei/physiopathology
13.
Nat Commun ; 5: 5472, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25434649

ABSTRACT

Mechanisms underlying central neuropathic pain are poorly understood. Although glial dysfunction has been functionally linked with neuropathic pain, very little is known about modulation of pain by oligodendrocytes. Here we report that genetic ablation of oligodendrocytes rapidly triggers a pattern of sensory changes that closely resemble central neuropathic pain, which are manifest before overt demyelination. Primary oligodendrocyte loss is not associated with autoreactive T- and B-cell infiltration in the spinal cord and neither activation of microglia nor reactive astrogliosis contribute functionally to central pain evoked by ablation of oligodendrocytes. Instead, light and electron microscopic analyses reveal axonal pathology in the spinal dorsal horn and spinothalamic tract concurrent with the induction and maintenance of nociceptive hypersensitivity. These data reveal a role for oligodendrocytes in modulating pain and suggest that perturbation of oligodendrocyte functions that maintain axonal integrity can lead to central neuropathic pain independent of immune contributions.


Subject(s)
Adaptive Immunity/immunology , Axons/pathology , Immunity, Innate/immunology , Neuralgia/pathology , Oligodendroglia/physiology , Spinal Cord Dorsal Horn/pathology , Spinothalamic Tracts/pathology , Animals , Astrocytes , Axons/ultrastructure , B-Lymphocytes , Mice , Microglia , Microscopy, Electron , Neuralgia/immunology , Nociception , Oligodendroglia/metabolism , Spinal Cord/pathology , Spinal Cord/ultrastructure , Spinal Cord Dorsal Horn/immunology , Spinal Cord Dorsal Horn/ultrastructure , Spinothalamic Tracts/immunology , Spinothalamic Tracts/ultrastructure , T-Lymphocytes
14.
Pain ; 154(12): 2801-2812, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23973358

ABSTRACT

The rich diversity of lipids and the specific signalling pathways they recruit provides tremendous scope for modulation of biological functions. Lysophosphatidylinositol (LPI) is emerging as a key modulator of cell proliferation, migration, and function, and holds important pathophysiological implications due to its high levels in diseased tissues, such as in cancer. Here we report a novel role for LPI in sensitization of peripheral sensory neurons, which was evident as exaggerated sensitivity to painful and innocuous pressure. Histopathological analyses indicated lack of involvement of myelin pathology and immune cell recruitment by LPI. Using pharmacological and conditional genetic tools in mice, we delineated receptor-mediated from non-receptor-mediated effects of LPI and we observed that GPR55, which functions as an LPI receptor when heterologously expressed in mammalian cells, only partially mediates LPI-induced actions in the context of pain sensitization in vivo; we demonstrate that, in vivo, LPI functions by activating Gα(13) as well as Gα(q/11) arms of G-protein signalling in sensory neurons. This study thus reports a novel pathophysiological function for LPI and elucidates underlying molecular mechanisms.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Lysophospholipids/physiology , Nociception/physiology , Sensory Receptor Cells/physiology , Signal Transduction/physiology , Animals , Dose-Response Relationship, Drug , GTP-Binding Proteins/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociception/drug effects , Phospholipids/pharmacology , Phospholipids/physiology , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL