Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38746089

ABSTRACT

We have identified a NMIIA and IIB-specific small molecule inhibitor, MT-125, and have studied its effects in GBM. MT-125 has high brain penetrance and retention and an excellent safety profile; blocks GBM invasion and cytokinesis, consistent with the known roles of NMII; and prolongs survival as a single agent in murine GBM models. MT-125 increases signaling along both the PDGFR- and MAPK-driven pathways through a mechanism that involves the upregulation of reactive oxygen species, and it synergizes with FDA-approved PDGFR and mTOR inhibitors in vitro . Combining MT-125 with sunitinib, a PDGFR inhibitor, or paxalisib, a combined PI3 Kinase/mTOR inhibitor significantly improves survival in orthotopic GBM models over either drug alone, and in the case of sunitinib, markedly prolongs survival in ∼40% of mice. Our results provide a powerful rationale for developing NMII targeting strategies to treat cancer and demonstrate that MT-125 has strong clinical potential for the treatment of GBM. Highlights: MT-125 is a highly specific small molecule inhibitor of non-muscle myosin IIA and IIB, is well-tolerated, and achieves therapeutic concentrations in the brain with systemic dosing.Treating preclinical models of glioblastoma with MT-125 produces durable improvements in survival.MT-125 stimulates PDGFR- and MAPK-driven signaling in glioblastoma and increases dependency on these pathways.Combining MT-125 with an FDA-approved PDGFR inhibitor in a mouse GBM model synergizes to improve median survival over either drug alone, and produces tumor free, prolonged survival in over 40% of mice.

2.
Cell Stem Cell ; 31(5): 676-693.e10, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38626772

ABSTRACT

Frontotemporal dementia (FTD) is an incurable group of early-onset dementias that can be caused by the deposition of hyperphosphorylated tau in patient brains. However, the mechanisms leading to neurodegeneration remain largely unknown. Here, we combined single-cell analyses of FTD patient brains with a stem cell culture and transplantation model of FTD. We identified disease phenotypes in FTD neurons carrying the MAPT-N279K mutation, which were related to oxidative stress, oxidative phosphorylation, and neuroinflammation with an upregulation of the inflammation-associated protein osteopontin (OPN). Human FTD neurons survived less and elicited an increased microglial response after transplantation into the mouse forebrain, which we further characterized by single nucleus RNA sequencing of microdissected grafts. Notably, downregulation of OPN in engrafted FTD neurons resulted in improved engraftment and reduced microglial infiltration, indicating an immune-modulatory role of OPN in patient neurons, which may represent a potential therapeutic target in FTD.


Subject(s)
Frontotemporal Dementia , Neurons , Osteopontin , tau Proteins , Osteopontin/metabolism , Osteopontin/genetics , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Frontotemporal Dementia/metabolism , Humans , Neurons/metabolism , Neurons/pathology , Animals , tau Proteins/metabolism , Mice , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Microglia/metabolism , Microglia/pathology , Mutation/genetics
3.
bioRxiv ; 2024 Jan 21.
Article in English | MEDLINE | ID: mdl-38293120

ABSTRACT

Gliomas are highly aggressive brain tumors characterized by poor prognosis and composed of diffusely infiltrating tumor cells that intermingle with non-neoplastic cells in the tumor microenvironment, including neurons. Neurons are increasingly appreciated as important reactive components of the glioma microenvironment, due to their role in causing hallmark glioma symptoms, such as cognitive deficits and seizures, as well as their potential ability to drive glioma progression. Separately, mTOR signaling has been shown to have pleiotropic effects in the brain tumor microenvironment, including regulation of neuronal hyperexcitability. However, the local cellular-level effects of mTOR inhibition on glioma-induced neuronal alterations are not well understood. Here we employed neuron-specific profiling of ribosome-bound mRNA via 'RiboTag,' morphometric analysis of dendritic spines, and in vivo calcium imaging, along with pharmacological mTOR inhibition to investigate the impact of glioma burden and mTOR inhibition on these neuronal alterations. The RiboTag analysis of tumor-associated excitatory neurons showed a downregulation of transcripts encoding excitatory and inhibitory postsynaptic proteins and dendritic spine development, and an upregulation of transcripts encoding cytoskeletal proteins involved in dendritic spine turnover. Light and electron microscopy of tumor-associated excitatory neurons demonstrated marked decreases in dendritic spine density. In vivo two-photon calcium imaging in tumor-associated excitatory neurons revealed progressive alterations in neuronal activity, both at the population and single-neuron level, throughout tumor growth. This in vivo calcium imaging also revealed altered stimulus-evoked somatic calcium events, with changes in event rate, size, and temporal alignment to stimulus, which was most pronounced in neurons with high-tumor burden. A single acute dose of AZD8055, a combined mTORC1/2 inhibitor, reversed the glioma-induced alterations on the excitatory neurons, including the alterations in ribosome-bound transcripts, dendritic spine density, and stimulus evoked responses seen by calcium imaging. These results point to mTOR-driven pathological plasticity in neurons at the infiltrative margin of glioma - manifested by alterations in ribosome-bound mRNA, dendritic spine density, and stimulus-evoked neuronal activity. Collectively, our work identifies the pathological changes that tumor-associated excitatory neurons experience as both hyperlocal and reversible under the influence of mTOR inhibition, providing a foundation for developing therapies targeting neuronal signaling in glioma.

4.
Nat Commun ; 14(1): 2586, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37142563

ABSTRACT

Glioblastoma (GBM) diffusely infiltrates the brain and intermingles with non-neoplastic brain cells, including astrocytes, neurons and microglia/myeloid cells. This complex mixture of cell types forms the biological context for therapeutic response and tumor recurrence. We used single-nucleus RNA sequencing and spatial transcriptomics to determine the cellular composition and transcriptional states in primary and recurrent glioma and identified three compositional 'tissue-states' defined by cohabitation patterns between specific subpopulations of neoplastic and non-neoplastic brain cells. These tissue-states correlated with radiographic, histopathologic, and prognostic features and were enriched in distinct metabolic pathways. Fatty acid biosynthesis was enriched in the tissue-state defined by the cohabitation of astrocyte-like/mesenchymal glioma cells, reactive astrocytes, and macrophages, and was associated with recurrent GBM and shorter survival. Treating acute slices of GBM with a fatty acid synthesis inhibitor depleted the transcriptional signature of this pernicious tissue-state. These findings point to therapies that target interdependencies in the GBM microenvironment.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Glioblastoma/pathology , Prognosis , Brain Neoplasms/pathology , Glioma/genetics , Astrocytes/metabolism , Tumor Microenvironment/genetics
5.
Nat Commun ; 14(1): 1187, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36864031

ABSTRACT

Ferroptosis is mediated by lipid peroxidation of phospholipids containing polyunsaturated fatty acyl moieties. Glutathione, the key cellular antioxidant capable of inhibiting lipid peroxidation via the activity of the enzyme glutathione peroxidase 4 (GPX-4), is generated directly from the sulfur-containing amino acid cysteine, and indirectly from methionine via the transsulfuration pathway. Herein we show that cysteine and methionine deprivation (CMD) can synergize with the GPX4 inhibitor RSL3 to increase ferroptotic cell death and lipid peroxidation in both murine and human glioma cell lines and in ex vivo organotypic slice cultures. We also show that a cysteine-depleted, methionine-restricted diet can improve therapeutic response to RSL3 and prolong survival in a syngeneic orthotopic murine glioma model. Finally, this CMD diet leads to profound in vivo metabolomic, proteomic and lipidomic alterations, highlighting the potential for improving the efficacy of ferroptotic therapies in glioma treatment with a non-invasive dietary modification.


Subject(s)
Ferroptosis , Glioma , Humans , Animals , Mice , Methionine , Cysteine , Proteomics , Racemethionine , Glioma/drug therapy
6.
bioRxiv ; 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36865302

ABSTRACT

Glioma cells hijack developmental transcriptional programs to control cell state. During neural development, lineage trajectories rely on specialized metabolic pathways. However, the link between tumor cell state and metabolic programs is poorly understood in glioma. Here we uncover a glioma cell state-specific metabolic liability that can be leveraged therapeutically. To model cell state diversity, we generated genetically engineered murine gliomas, induced by deletion of p53 alone (p53) or with constitutively active Notch signaling (N1IC), a pathway critical in controlling cellular fate. N1IC tumors harbored quiescent astrocyte-like transformed cell states while p53 tumors were predominantly comprised of proliferating progenitor-like cell states. N1IC cells exhibit distinct metabolic alterations, with mitochondrial uncoupling and increased ROS production rendering them more sensitive to inhibition of the lipid hydroperoxidase GPX4 and induction of ferroptosis. Importantly, treating patient-derived organotypic slices with a GPX4 inhibitor induced selective depletion of quiescent astrocyte-like glioma cell populations with similar metabolic profiles.

7.
Cell Rep ; 39(12): 110991, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35732128

ABSTRACT

Inhibitors of the mitotic kinesin Kif11 are anti-mitotics that, unlike vinca alkaloids or taxanes, do not disrupt microtubules and are not neurotoxic. However, development of resistance has limited their clinical utility. While resistance to Kif11 inhibitors in other cell types is due to mechanisms that prevent these drugs from disrupting mitosis, we find that in glioblastoma (GBM), resistance to the Kif11 inhibitor ispinesib works instead through suppression of apoptosis driven by activation of STAT3. This form of resistance requires dual phosphorylation of STAT3 residues Y705 and S727, mediated by SRC and epidermal growth factor receptor (EGFR), respectively. Simultaneously inhibiting SRC and EGFR reverses this resistance, and combined targeting of these two kinases in vivo with clinically available inhibitors is synergistic and significantly prolongs survival in ispinesib-treated GBM-bearing mice. We thus identify a translationally actionable approach to overcoming Kif11 inhibitor resistance that may work to block STAT3-driven resistance against other anti-cancer therapies as well.


Subject(s)
Antimitotic Agents , Glioblastoma , Animals , Antimitotic Agents/pharmacology , Cell Line, Tumor , ErbB Receptors/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Kinesins , Mice , STAT3 Transcription Factor/metabolism , Signal Transduction
8.
Brain ; 145(10): 3666-3680, 2022 10 21.
Article in English | MEDLINE | ID: mdl-35552612

ABSTRACT

While several studies have attributed the development of tumour-associated seizures to an excitatory-inhibitory imbalance, we have yet to resolve the spatiotemporal interplay between different types of neuron in glioma-infiltrated cortex. Herein, we combined methods for single unit analysis of microelectrode array recordings with wide-field optical mapping of Thy1-GCaMP pyramidal cells in an ex vivo acute slice model of diffusely infiltrating glioma. This enabled simultaneous tracking of individual neurons from both excitatory and inhibitory populations throughout seizure-like events. Moreover, our approach allowed for observation of how the crosstalk between these neurons varied spatially, as we recorded across an extended region of glioma-infiltrated cortex. In tumour-bearing slices, we observed marked alterations in single units classified as putative fast-spiking interneurons, including reduced firing, activity concentrated within excitatory bursts and deficits in local inhibition. These results were correlated with increases in overall excitability. Mechanistic perturbation of this system with the mTOR inhibitor AZD8055 revealed increased firing of putative fast-spiking interneurons and restoration of local inhibition, with concomitant decreases in overall excitability. Altogether, our findings suggest that diffusely infiltrating glioma affect the interplay between excitatory and inhibitory neuronal populations in a reversible manner, highlighting a prominent role for functional mechanisms linked to mTOR activation.


Subject(s)
Glioma , Pyramidal Cells , Humans , Action Potentials/physiology , Pyramidal Cells/physiology , Neurons/physiology , Seizures , TOR Serine-Threonine Kinases
9.
Genome Med ; 13(1): 82, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33975634

ABSTRACT

BACKGROUND: Preclinical studies require models that recapitulate the cellular diversity of human tumors and provide insight into the drug sensitivities of specific cellular populations. The ideal platform would enable rapid screening of cell type-specific drug sensitivities directly in patient tumor tissue and reveal strategies to overcome intratumoral heterogeneity. METHODS: We combine multiplexed drug perturbation in acute slice culture from freshly resected tumors with single-cell RNA sequencing (scRNA-seq) to profile transcriptome-wide drug responses in individual patients. We applied this approach to drug perturbations on slices derived from six glioblastoma (GBM) resections to identify conserved drug responses and to one additional GBM resection to identify patient-specific responses. RESULTS: We used scRNA-seq to demonstrate that acute slice cultures recapitulate the cellular and molecular features of the originating tumor tissue and the feasibility of drug screening from an individual tumor. Detailed investigation of etoposide, a topoisomerase poison, and the histone deacetylase (HDAC) inhibitor panobinostat in acute slice cultures revealed cell type-specific responses across multiple patients. Etoposide has a conserved impact on proliferating tumor cells, while panobinostat treatment affects both tumor and non-tumor populations, including unexpected effects on the immune microenvironment. CONCLUSIONS: Acute slice cultures recapitulate the major cellular and molecular features of GBM at the single-cell level. In combination with scRNA-seq, this approach enables cell type-specific analysis of sensitivity to multiple drugs in individual tumors. We anticipate that this approach will facilitate pre-clinical studies that identify effective therapies for solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/genetics , RNA-Seq , Single-Cell Analysis , Antineoplastic Agents/therapeutic use , Computational Biology/methods , Drug Screening Assays, Antitumor/methods , Humans , Immunohistochemistry , In Situ Hybridization , Microscopy , Neoplasms/drug therapy , Precision Medicine/methods , Sensitivity and Specificity , Single-Cell Analysis/methods , Treatment Outcome , Tumor Microenvironment/genetics , Whole Genome Sequencing
10.
Genome Med ; 13(1): 88, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011400

ABSTRACT

BACKGROUND: Macrophages are the most common infiltrating immune cells in gliomas and play a wide variety of pro-tumor and anti-tumor roles. However, the different subpopulations of macrophages and their effects on the tumor microenvironment remain poorly understood. METHODS: We combined new and previously published single-cell RNA-seq data from 98,015 single cells from a total of 66 gliomas to profile 19,331 individual macrophages. RESULTS: Unsupervised clustering revealed a pro-tumor subpopulation of bone marrow-derived macrophages characterized by the scavenger receptor MARCO, which is almost exclusively found in IDH1-wild-type glioblastomas. Previous studies have implicated MARCO as an unfavorable marker in melanoma and non-small cell lung cancer; here, we find that bulk MARCO expression is associated with worse prognosis and mesenchymal subtype. Furthermore, MARCO expression is significantly altered over the course of treatment with anti-PD1 checkpoint inhibitors in a response-dependent manner, which we validate with immunofluorescence imaging. CONCLUSIONS: These findings illustrate a novel macrophage subpopulation that drives tumor progression in glioblastomas and suggest potential therapeutic targets to prevent their recruitment.


Subject(s)
Biomarkers, Tumor , Glioblastoma/diagnosis , Glioblastoma/etiology , Receptors, Immunologic/genetics , Single-Cell Analysis , Tumor-Associated Macrophages/metabolism , Cell Communication/genetics , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/mortality , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Isocitrate Dehydrogenase/genetics , Mutation , Prognosis , Single-Cell Analysis/methods , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/pathology
11.
iScience ; 23(12): 101802, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33299973

ABSTRACT

Invasion and proliferation are defining phenotypes of cancer, and in glioblastoma blocking one stimulates the other, implying that effective therapy must inhibit both, ideally through a single target that is also dispensable for normal tissue function. The molecular motor myosin 10 meets these criteria. Myosin 10 knockout mice can survive to adulthood, implying that normal cells can compensate for its loss; its deletion impairs invasion, slows proliferation, and prolongs survival in murine models of glioblastoma. Myosin 10 deletion also enhances tumor dependency on the DNA damage and the metabolic stress responses and induces synthetic lethality when combined with inhibitors of these processes. Our results thus demonstrate that targeting myosin 10 is active against glioblastoma by itself, synergizes with other clinically available therapeutics, may have acceptable side effects in normal tissues, and has potential as a heretofore unexplored therapeutic approach for this disease.

12.
Cell Rep ; 31(2): 107500, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294436

ABSTRACT

Diffusely infiltrating gliomas are known to cause alterations in cortical function, vascular disruption, and seizures. These neurological complications present major clinical challenges, yet their underlying mechanisms and causal relationships to disease progression are poorly characterized. Here, we follow glioma progression in awake Thy1-GCaMP6f mice using in vivo wide-field optical mapping to monitor alterations in both neuronal activity and functional hemodynamics. The bilateral synchrony of spontaneous neuronal activity gradually decreases in glioma-infiltrated cortical regions, while neurovascular coupling becomes progressively disrupted compared to uninvolved cortex. Over time, mice develop diverse patterns of high amplitude discharges and eventually generalized seizures that appear to originate at the tumors' infiltrative margins. Interictal and seizure events exhibit positive neurovascular coupling in uninfiltrated cortex; however, glioma-infiltrated regions exhibit disrupted hemodynamic responses driving seizure-evoked hypoxia. These results reveal a landscape of complex physiological interactions occurring during glioma progression and present new opportunities for exploring novel biomarkers and therapeutic targets.


Subject(s)
Glioma/physiopathology , Neurovascular Coupling/physiology , Animals , Brain/physiopathology , Cerebral Cortex/metabolism , Disease Progression , Hemodynamics/physiology , Male , Mice , Mice, Inbred C57BL , Nerve Net/physiopathology , Neurons/metabolism , Seizures/physiopathology
13.
ACS Chem Biol ; 15(2): 469-484, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31899616

ABSTRACT

Although radiation is widely used to treat cancers, resistance mechanisms often develop and involve activation of DNA repair and inhibition of apoptosis. Therefore, compounds that sensitize cancer cells to radiation via alternative cell death pathways are valuable. We report here that ferroptosis, a form of nonapoptotic cell death driven by lipid peroxidation, is partly responsible for radiation-induced cancer cell death. Moreover, we found that small molecules activating ferroptosis through system xc- inhibition or GPX4 inhibition synergize with radiation to induce ferroptosis in several cancer types by enhancing cytoplasmic lipid peroxidation but not increasing DNA damage or caspase activation. Ferroptosis inducers synergized with cytoplasmic irradiation, but not nuclear irradiation. Finally, administration of ferroptosis inducers enhanced the antitumor effect of radiation in a murine xenograft model and in human patient-derived models of lung adenocarcinoma and glioma. These results suggest that ferroptosis inducers may be effective radiosensitizers that can expand the efficacy and range of indications for radiation therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Ferroptosis/drug effects , Lipid Peroxidation/radiation effects , Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiation-Sensitizing Agents/therapeutic use , Amino Acid Transport System y+/metabolism , Animals , Carbolines/therapeutic use , Cell Line, Tumor , Gamma Rays , Humans , Imidazoles/therapeutic use , Ketones/therapeutic use , Lipid Peroxidation/drug effects , Mice, Nude , Piperazines/therapeutic use , Sorafenib/therapeutic use , Xenograft Model Antitumor Assays
14.
Neurobiol Dis ; 134: 104676, 2020 02.
Article in English | MEDLINE | ID: mdl-31731042

ABSTRACT

The purpose of this study is to develop a platform in which the cellular and molecular underpinnings of chronic focal neocortical lesional epilepsy can be explored and use it to characterize seizure-like events (SLEs) in an ex vivo model of infiltrating high-grade glioma. Microelectrode arrays were used to study electrophysiologic changes in ex vivo acute brain slices from a PTEN/p53 deleted, PDGF-B driven mouse model of high-grade glioma. Electrode locations were co-registered to the underlying histology to ascertain the influence of the varying histologic landscape on the observed electrophysiologic changes. Peritumoral, infiltrated, and tumor sites were sampled in tumor-bearing slices. Following the addition of zero Mg2+ solution, all three histologic regions in tumor-bearing slices showed significantly greater increases in firing rates when compared to the control sites. Tumor-bearing slices demonstrated increased proclivity for SLEs, with 40 events in tumor-bearing slices and 5 events in control slices (p-value = .0105). Observed SLEs were characterized by either low voltage fast (LVF) onset patterns or short bursts of repetitive widespread, high amplitude low frequency discharges. Seizure foci comprised areas from all three histologic regions. The onset electrode was found to be at the infiltrated margin in 50% of cases and in the peritumoral region in 36.9% of cases. These findings reveal a landscape of histopathologic and electrophysiologic alterations associated with ictogenesis and spread of tumor-associated seizures.


Subject(s)
Brain Neoplasms/physiopathology , Brain/physiopathology , Glioma/physiopathology , Neurons/physiology , Seizures/physiopathology , Action Potentials , Animals , Brain Neoplasms/complications , Disease Models, Animal , Glioma/complications , Mice, Transgenic , Microelectrodes , Seizures/complications
15.
Proc Natl Acad Sci U S A ; 116(31): 15550-15559, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31235578

ABSTRACT

The ability of glioblastoma to disperse through the brain contributes to its lethality, and blocking this behavior has been an appealing therapeutic approach. Although a number of proinvasive signaling pathways are active in glioblastoma, many are redundant, so targeting one can be overcome by activating another. However, these pathways converge on nonredundant components of the cytoskeleton, and we have shown that inhibiting one of these-the myosin II family of cytoskeletal motors-blocks glioblastoma invasion even with simultaneous activation of multiple upstream promigratory pathways. Myosin IIA and IIB are the most prevalent isoforms of myosin II in glioblastoma, and we now show that codeleting these myosins markedly impairs tumorigenesis and significantly prolongs survival in a rodent model of this disease. However, while targeting just myosin IIA also impairs tumor invasion, it surprisingly increases tumor proliferation in a manner that depends on environmental mechanics. On soft surfaces myosin IIA deletion enhances ERK1/2 activity, while on stiff surfaces it enhances the activity of NFκB, not only in glioblastoma but in triple-negative breast carcinoma and normal keratinocytes as well. We conclude myosin IIA suppresses tumorigenesis in at least two ways that are modulated by the mechanics of the tumor and its stroma. Our results also suggest that inhibiting tumor invasion can enhance tumor proliferation and that effective therapy requires targeting cellular components that drive both proliferation and invasion simultaneously.


Subject(s)
Carcinogenesis/metabolism , Cytoskeleton/metabolism , Glioblastoma/metabolism , MAP Kinase Signaling System , Neoplasm Proteins/metabolism , Nonmuscle Myosin Type IIA/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cytoskeleton/genetics , Cytoskeleton/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Mice , Neoplasm Proteins/genetics , Nonmuscle Myosin Type IIA/genetics
16.
Genome Med ; 10(1): 57, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30041684

ABSTRACT

BACKGROUND: Despite extensive molecular characterization, we lack a comprehensive understanding of lineage identity, differentiation, and proliferation in high-grade gliomas (HGGs). METHODS: We sampled the cellular milieu of HGGs by profiling dissociated human surgical specimens with a high-density microwell system for massively parallel single-cell RNA-Seq. We analyzed the resulting profiles to identify subpopulations of both HGG and microenvironmental cells and applied graph-based methods to infer structural features of the malignantly transformed populations. RESULTS: While HGG cells can resemble glia or even immature neurons and form branched lineage structures, mesenchymal transformation results in unstructured populations. Glioma cells in a subset of mesenchymal tumors lose their neural lineage identity, express inflammatory genes, and co-exist with marked myeloid infiltration, reminiscent of molecular interactions between glioma and immune cells established in animal models. Additionally, we discovered a tight coupling between lineage resemblance and proliferation among malignantly transformed cells. Glioma cells that resemble oligodendrocyte progenitors, which proliferate in the brain, are often found in the cell cycle. Conversely, glioma cells that resemble astrocytes, neuroblasts, and oligodendrocytes, which are non-proliferative in the brain, are generally non-cycling in tumors. CONCLUSIONS: These studies reveal a relationship between cellular identity and proliferation in HGG and distinct population structures that reflects the extent of neural and non-neural lineage resemblance among malignantly transformed cells.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Single-Cell Analysis , Transcriptome , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation , Cell Transformation, Neoplastic , Glioma/metabolism , Glioma/pathology , Humans , Neuroglia/pathology , Neurons/pathology
17.
Proc Natl Acad Sci U S A ; 111(34): 12550-5, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25114226

ABSTRACT

Glioblastomas (GBMs) diffusely infiltrate the brain, making complete removal by surgical resection impossible. The mixture of neoplastic and nonneoplastic cells that remain after surgery form the biological context for adjuvant therapeutic intervention and recurrence. We performed RNA-sequencing (RNA-seq) and histological analysis on radiographically guided biopsies taken from different regions of GBM and showed that the tissue contained within the contrast-enhancing (CE) core of tumors have different cellular and molecular compositions compared with tissue from the nonenhancing (NE) margins of tumors. Comparisons with the The Cancer Genome Atlas dataset showed that the samples from CE regions resembled the proneural, classical, or mesenchymal subtypes of GBM, whereas the samples from the NE regions predominantly resembled the neural subtype. Computational deconvolution of the RNA-seq data revealed that contributions from nonneoplastic brain cells significantly influence the expression pattern in the NE samples. Gene ontology analysis showed that the cell type-specific expression patterns were functionally distinct and highly enriched in genes associated with the corresponding cell phenotypes. Comparing the RNA-seq data from the GBM samples to that of nonneoplastic brain revealed that the differentially expressed genes are distributed across multiple cell types. Notably, the patterns of cell type-specific alterations varied between the different GBM subtypes: the NE regions of proneural tumors were enriched in oligodendrocyte progenitor genes, whereas the NE regions of mesenchymal GBM were enriched in astrocytic and microglial genes. These subtype-specific patterns provide new insights into molecular and cellular composition of the infiltrative margins of GBM.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Adult , Aged , Aged, 80 and over , Brain Neoplasms/classification , Contrast Media , Female , Glioblastoma/classification , Humans , Image-Guided Biopsy , Magnetic Resonance Imaging , Male , Middle Aged , RNA, Neoplasm/genetics , Sequence Analysis, RNA , Transcriptome , Tumor Microenvironment
18.
J Biol Chem ; 289(11): 7897-906, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24482227

ABSTRACT

We have shown previously that tyrosine phosphorylation of Wiskott-Aldrich syndrome protein (WASP) is important for diverse macrophage functions including phagocytosis, chemotaxis, podosome dynamics, and matrix degradation. However, the specific tyrosine kinase mediating WASP phosphorylation is still unclear. Here, we provide evidence that Hck, which is predominantly expressed in leukocytes, can tyrosine phosphorylate WASP and regulates WASP-mediated macrophage functions. We demonstrate that tyrosine phosphorylation of WASP in response to stimulation with CX3CL1 or via Fcγ receptor ligation were severely reduced in Hck(-/-) bone marrow-derived macrophages (BMMs) or in RAW/LR5 macrophages in which Hck expression was silenced using RNA-mediated interference (Hck shRNA). Consistent with reduced WASP tyrosine phosphorylation, phagocytosis, chemotaxis, and matrix degradation are reduced in Hck(-/-) BMMs or Hck shRNA cells. In particular, WASP phosphorylation was primarily mediated by the p61 isoform of Hck. Our studies also show that Hck and WASP are required for passage through a dense three-dimensional matrix and transendothelial migration, suggesting that tyrosine phosphorylation of WASP by Hck may play a role in tissue infiltration of macrophages. Consistent with a role for this pathway in invasion, WASP(-/-) BMMs do not invade into tumor spheroids with the same efficiency as WT BMMs and cells expressing phospho-deficient WASP have reduced ability to promote carcinoma cell invasion. Altogether, our results indicate that tyrosine phosphorylation of WASP by Hck is required for proper macrophage functions.


Subject(s)
Macrophages/cytology , Proto-Oncogene Proteins c-hck/metabolism , Wiskott-Aldrich Syndrome Protein/chemistry , Wiskott-Aldrich Syndrome Protein/metabolism , Animals , Bone Marrow Cells/cytology , Cell Line , Cell Movement , Chemotaxis , Chemotaxis, Leukocyte , Collagen/chemistry , Crosses, Genetic , Endothelial Cells/cytology , Macrophages/metabolism , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Protein Isoforms/chemistry , RNA Interference , Transendothelial and Transepithelial Migration , Tyrosine/chemistry
19.
Cell Rep ; 4(3): 429-36, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911287

ABSTRACT

A paracrine interaction between epidermal growth factor (EGF)-secreting tumor-associated macrophages (TAMs) and colony-stimulating factor 1 (CSF-1)-secreting breast carcinoma cells promotes invasion and metastasis. Here, we show that mice deficient in the hematopoietic-cell-specific Wiskott-Aldrich syndrome protein (WASp) are unable to support TAM-dependent carcinoma cell invasion and metastasis in both orthotopic and transgenic models of mammary tumorigenesis. Motility and invasion defects of tumor cells were recapitulated ex vivo upon coculture with WASp(-/-) macrophages. Mechanistically, WASp is required for macrophages to migrate toward CSF-1-producing carcinoma cells, as well as for the release of EGF through metalloprotease-dependent shedding of EGF from the cell surface of macrophages. Our findings suggest that WASp acts to support both the migration of TAMs and the production of EGF, which in concert promote breast tumor metastasis.


Subject(s)
Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Wiskott-Aldrich Syndrome Protein/metabolism , Animals , Cell Line, Tumor , Cell Movement/physiology , Disease Progression , Female , Heterografts , Humans , Mice , Mice, Transgenic , Neoplasm Metastasis , Rats , Wiskott-Aldrich Syndrome Protein/genetics
20.
PLoS One ; 7(1): e30033, 2012.
Article in English | MEDLINE | ID: mdl-22279563

ABSTRACT

Wiskott-Aldrich syndrome protein (WASp) is an actin nucleation promoting factor that is required for macrophages to directionally migrate towards various chemoattractants. The chemotaxis defect of WASp-deficient cells and its activation by Cdc42 in vivo suggest that WASp plays a role in directional sensing, however, its precise role in macrophage chemotaxis is still unclear. Using shRNA-mediated downregulation of WASp in the murine monocyte/macrophage cell line RAW/LR5 (shWASp), we found that WASp was responsible for the initial wave of actin polymerization in response to global stimulation with CSF-1, which in Dictyostelium discoideum amoebae and carcinoma cells has been correlated with the ability to migrate towards chemoattractants. Real-time monitoring of shWASp cells, as well as WASp⁻/⁻ bone marrow-derived macrophages (BMMs), in response to a CSF-1 gradient revealed that the protrusions from WASp-deficient cells were directional, showing intact directional sensing. However, the protrusions from WASp-deficient cells demonstrated reduced persistence compared to their respective control shRNA and wild-type cells. Further examination showed that tyrosine phosphorylation of WASp was required for both the first wave of actin polymerization following global CSF-1 stimulation and proper directional responses towards CSF-1. Importantly, the PI3K, Rac1 and WAVE2 proteins were incorporated normally in CSF-1 - elicited protrusions in the absence of WASp, suggesting that membrane protrusion driven by the WAVE2 complex signaling is intact. Collectively, these results suggest that WASp and its phosphorylation play critical roles in coordinating the actin cytoskeleton rearrangements necessary for the persistence of protrusions required for directional migration of macrophages towards CSF-1.


Subject(s)
Chemotaxis/physiology , Macrophages/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/metabolism , Actins/metabolism , Animals , Blotting, Western , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Line , Chemotaxis/drug effects , Chemotaxis/genetics , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Mice , Mice, 129 Strain , Mice, Knockout , Microscopy, Fluorescence , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Pseudopodia/drug effects , Pseudopodia/metabolism , RNA Interference , Time Factors , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...