Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Publication year range
1.
Cell ; 172(5): 1050-1062.e14, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474906

ABSTRACT

While the preponderance of morbidity and mortality in medulloblastoma patients are due to metastatic disease, most research focuses on the primary tumor due to a dearth of metastatic tissue samples and model systems. Medulloblastoma metastases are found almost exclusively on the leptomeningeal surface of the brain and spinal cord; dissemination is therefore thought to occur through shedding of primary tumor cells into the cerebrospinal fluid followed by distal re-implantation on the leptomeninges. We present evidence for medulloblastoma circulating tumor cells (CTCs) in therapy-naive patients and demonstrate in vivo, through flank xenografting and parabiosis, that medulloblastoma CTCs can spread through the blood to the leptomeningeal space to form leptomeningeal metastases. Medulloblastoma leptomeningeal metastases express high levels of the chemokine CCL2, and expression of CCL2 in medulloblastoma in vivo is sufficient to drive leptomeningeal dissemination. Hematogenous dissemination of medulloblastoma offers a new opportunity to diagnose and treat lethal disseminated medulloblastoma.


Subject(s)
Medulloblastoma/blood supply , Medulloblastoma/pathology , Meningeal Neoplasms/blood supply , Meningeal Neoplasms/secondary , Allografts , Animals , Cell Line, Tumor , Chemokine CCL2/metabolism , Chromosomes, Human, Pair 10/genetics , Female , Humans , Male , Medulloblastoma/genetics , Mice, SCID , Neoplastic Cells, Circulating , Parabiosis
2.
Cell ; 170(3): 429-442.e11, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753423

ABSTRACT

Hunger, driven by negative energy balance, elicits the search for and consumption of food. While this response is in part mediated by neurons in the hypothalamus, the role of specific cell types in other brain regions is less well defined. Here, we show that neurons in the dorsal raphe nucleus, expressing vesicular transporters for GABA or glutamate (hereafter, DRNVgat and DRNVGLUT3 neurons), are reciprocally activated by changes in energy balance and that modulating their activity has opposite effects on feeding-DRNVgat neurons increase, whereas DRNVGLUT3 neurons suppress, food intake. Furthermore, modulation of these neurons in obese (ob/ob) mice suppresses food intake and body weight and normalizes locomotor activity. Finally, using molecular profiling, we identify druggable targets in these neurons and show that local infusion of agonists for specific receptors on these neurons has potent effects on feeding. These data establish the DRN as an important node controlling energy balance. PAPERCLIP.


Subject(s)
Appetite Regulation , Dorsal Raphe Nucleus/metabolism , Neurons/metabolism , Animals , Body Weight , Brain/physiology , Dorsal Raphe Nucleus/cytology , Electrophysiology , Fasting , Hunger , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Optogenetics
3.
Cell ; 161(4): 803-16, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25913192

ABSTRACT

Active neurons exert a mitogenic effect on normal neural precursor and oligodendroglial precursor cells, the putative cellular origins of high-grade glioma (HGG). By using optogenetic control of cortical neuronal activity in a patient-derived pediatric glioblastoma xenograft model, we demonstrate that active neurons similarly promote HGG proliferation and growth in vivo. Conditioned medium from optogenetically stimulated cortical slices promoted proliferation of pediatric and adult patient-derived HGG cultures, indicating secretion of activity-regulated mitogen(s). The synaptic protein neuroligin-3 (NLGN3) was identified as the leading candidate mitogen, and soluble NLGN3 was sufficient and necessary to promote robust HGG cell proliferation. NLGN3 induced PI3K-mTOR pathway activity and feedforward expression of NLGN3 in glioma cells. NLGN3 expression levels in human HGG negatively correlated with patient overall survival. These findings indicate the important role of active neurons in the brain tumor microenvironment and identify secreted NLGN3 as an unexpected mechanism promoting neuronal activity-regulated cancer growth.


Subject(s)
Brain Neoplasms/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Proliferation , Glioma/pathology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Adolescent , Amino Acid Sequence , Animals , Brain Neoplasms/metabolism , Glioma/metabolism , Heterografts , Humans , Male , Mice , Molecular Sequence Data , Neoplasm Transplantation , Neurons/metabolism
6.
Lancet Oncol ; 23(11): 1430-1440, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36244398

ABSTRACT

BACKGROUND: Outcomes are poor in patients with HER2-negative, advanced gastric or gastro-oesophageal junction adenocarcinomas. In this study, we investigated efficacy and safety of the first-in-class, afucosylated, humanised IgG1 anti-fibroblast growth factor receptor 2 isoform IIb (FGFR2b) monoclonal antibody bemarituzumab with modified 5-fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) in patients with FGFR2b-selected gastric or gastro-oesophageal junction adenocarcinoma. METHODS: In the randomised, double-blind, placebo-controlled phase 2 trial (FIGHT), patients aged 18 years and older with HER2 non-positive, FGFR2b-selected gastric or gastro-oesophageal junction adenocarcinoma, and an Eastern Cooperative Oncology Group performance status of 0-1 were recruited from 144 clinical sites across 17 countries. Patients with previous treatment with any selective inhibitor of the FGF-FGFR pathway were excluded. Eligible patients were randomly assigned (1:1), using permuted-block randomisation (block size of four) and a central interactive voice-web-based response system, stratified by geographical region, previous treatment with curative intent, and administration of mFOLFOX6 while being screened for FGFR2b status, to either bemarituzumab (15 mg/kg of bodyweight) or matched placebo intravenously every 2 weeks. All patients also received mFOLFOX6 (oxaliplatin 85 mg/m2, leucovorin 400 mg/m2, and 5-fluorouracil as a 400 mg/m2 bolus followed by 2400 mg/m2 over approximately 46 h) intravenously every 2 weeks. Patients were given treatment until disease progression (defined by Response Evaluation Criteria in Solid Tumours [RECIST] version 1.1), unacceptable toxicity, withdrawal of consent, or death. The primary endpoint was progression-free survival in the intention-to-treat population (defined as all patients randomly assigned to treatment). Safety was assessed in all patients who received at least one dose of assigned treatment. This study is registered with ClinicalTrials.gov, NCT03694522, and is now complete. FINDINGS: Between Nov 14, 2017, and May 8, 2020, 910 patients were screened and 155 were randomly assigned to the bemarituzumab (n=77) or placebo group (n=78). Median age was 60·0 years (IQR 51·0-67·0), 44 (28%) participants were women, 111 (72%) were men, 89 (57%) were Asian, and 61 (39%) were White. At the time of the primary analysis and at a median follow-up of 10·9 months (IQR 6·3-14·2), median progression-free survival was 9·5 months (95% CI 7·3-12·9) in the bemarituzumab group and 7·4 months (5·8-8·4) in the placebo group (hazard ratio [HR] 0·68 [95% CI 0·44-1·04; p=0·073). Common grade 3 or worse adverse events were decreased neutrophil count (23 [30%] of 76 in the bemarituzumab group vs 27 [35%] of 77 in the placebo group), cornea disorder (18 [24%] vs none), neutropenia (ten [13%] vs seven [9%]), stomatitis (seven [9%] vs one [1%]), and anaemia (six [8%] vs ten [13%]). Serious treatment-emergent adverse events were reported in 24 (32%) patients in the bemarituzumab group and 28 (36%) in the placebo group. Serious mFOLFOX6 treatment-related adverse events occurred in nine (12%) patients in the bemarituzumab group and in 15 (19%) patients in the placebo group. All-grade corneal events (adverse events of special interest) occurred in 51 (67%) patients in the bemarituzumab group and eight (10%) in the placebo group; grade 3 corneal events were reported only in 18 (24%) patients in the bemarituzumab group. Treatment-related deaths occurred in three patients in the bemarituzumab group (two due to sepsis, one due to pneumonia) and none in the placebo group. INTERPRETATION: In this exploratory phase 2 study, despite no statistically significant improvement in progression-free survival, treatment with bemarituzumab showed promising clinical efficacy. Confirmatory phase 3 trials of bemarituzumab plus mFOLFOX6 powered to demonstrate statistical significance are being investigated in patients with previously untreated, FGFR2b-overexpressing, advanced gastric or gastro-oesophageal junction adenocarcinoma. FUNDING: Five Prime Therapeutics.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Male , Humans , Female , Middle Aged , Esophagogastric Junction/pathology , Leucovorin/adverse effects , Receptor, Fibroblast Growth Factor, Type 2/genetics , Stomach Neoplasms/pathology , Oxaliplatin/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Fluorouracil , Double-Blind Method
7.
Proc Natl Acad Sci U S A ; 116(3): 997-1006, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30602457

ABSTRACT

Glioblastoma multiforme (GBM) is a highly aggressive malignant brain tumor with fatal outcome. Tumor-associated macrophages and microglia (TAMs) have been found to be major tumor-promoting immune cells in the tumor microenvironment. Hence, modulation and reeducation of tumor-associated macrophages and microglia in GBM is considered a promising antitumor strategy. Resident microglia and invading macrophages have been shown to have distinct origin and function. Whereas yolk sac-derived microglia reside in the brain, blood-derived monocytes invade the central nervous system only under pathological conditions like tumor formation. We recently showed that disruption of the SIRPα-CD47 signaling axis is efficacious against various brain tumors including GBM primarily by inducing tumor phagocytosis. However, most effects are attributed to macrophages recruited from the periphery but the role of the brain resident microglia is unknown. Here, we sought to utilize a model to distinguish resident microglia and peripheral macrophages within the GBM-TAM pool, using orthotopically xenografted, immunodeficient, and syngeneic mouse models with genetically color-coded macrophages (Ccr2RFP) and microglia (Cx3cr1GFP). We show that even in the absence of phagocytizing macrophages (Ccr2RFP/RFP), microglia are effector cells of tumor cell phagocytosis in response to anti-CD47 blockade. Additionally, macrophages and microglia show distinct morphological and transcriptional changes. Importantly, the transcriptional profile of microglia shows less of an inflammatory response which makes them a promising target for clinical applications.


Subject(s)
Brain Neoplasms/immunology , CD47 Antigen/immunology , Glioblastoma/immunology , Microglia/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Phagocytosis , Receptors, Immunologic/immunology , Signal Transduction/immunology , Animals , Brain Neoplasms/pathology , CD47 Antigen/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred NOD , Mice, Transgenic , Microglia/pathology , Monocytes/immunology , Monocytes/pathology , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Receptors, Immunologic/genetics , Signal Transduction/genetics
8.
Genes Dev ; 27(24): 2642-7, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24352421

ABSTRACT

The self-renewal versus differentiation choice of Drosophila and mammalian neural stem cells (NSCs) requires Notch (N) signaling. How N regulates NSC behavior is not well understood. Here we show that canonical N signaling cooperates with a noncanonical N signaling pathway to mediate N-directed NSC regulation. In the noncanonical pathway, N interacts with PTEN-induced kinase 1 (PINK1) to influence mitochondrial function, activating mechanistic target of rapamycin complex 2 (mTORC2)/AKT signaling. Importantly, attenuating noncanonical N signaling preferentially impaired the maintenance of Drosophila and human cancer stem cell-like tumor-forming cells. Our results emphasize the importance of mitochondria to N and NSC biology, with important implications for diseases associated with aberrant N signaling.


Subject(s)
Mitochondria/metabolism , Multiprotein Complexes/metabolism , Neoplastic Stem Cells/metabolism , Protein Kinases/metabolism , Receptors, Notch/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Brain Neoplasms/physiopathology , Cell Line, Tumor , Cell Proliferation , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation , Humans , Mechanistic Target of Rapamycin Complex 2 , Microscopy, Electron, Transmission , Mitochondria/enzymology , Mitochondria/ultrastructure , Multiprotein Complexes/genetics , Mutation , Protein Kinases/genetics , RNA Interference , TOR Serine-Threonine Kinases/genetics
9.
Adv Exp Med Biol ; 1273: 197-208, 2020.
Article in English | MEDLINE | ID: mdl-33119883

ABSTRACT

Microglia are the brain resident phagocytes that act as the primary form of the immune defense in the central nervous system. These cells originate from primitive macrophages that arise from the yolk sac. Advances in imaging and single-cell RNA-seq technologies provided new insights into the complexity of microglia biology.Microglia play an essential role in the brain development and maintenance of brain homeostasis. They are also crucial in injury repair in the central nervous system. The tumor microenvironment is complex and includes neoplastic cells as well as varieties of host and infiltrating immune cells. Microglia are part of the glioma microenvironment and play a critical part in initiating and maintaining tumor growth and spread. Microglia can also act as effector cells in treatments against gliomas. In this chapter, we summarize the current knowledge of how and where microglia are generated. We also discuss their functions during brain development, injury repair, and homeostasis. Moreover, we discuss the role of microglia in the tumor microenvironment of gliomas and highlight their therapeutic implications.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Microglia/cytology , Tumor Microenvironment/immunology , Brain/immunology , Brain Neoplasms/therapy , Glioma/therapy , Humans , Microglia/immunology
10.
Nature ; 501(7467): 380-4, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24025767

ABSTRACT

Down's syndrome results from full or partial trisomy of chromosome 21. However, the consequences of the underlying gene-dosage imbalance on adult tissues remain poorly understood. Here we show that in Ts65Dn mice, which are trisomic for 132 genes homologous to genes on human chromosome 21, triplication of Usp16 reduces the self-renewal of haematopoietic stem cells and the expansion of mammary epithelial cells, neural progenitors and fibroblasts. In addition, Usp16 is associated with decreased ubiquitination of Cdkn2a and accelerated senescence in Ts65Dn fibroblasts. Usp16 can remove ubiquitin from histone H2A on lysine 119, a critical mark for the maintenance of multiple somatic tissues. Downregulation of Usp16, either by mutation of a single normal Usp16 allele or by short interfering RNAs, largely rescues all of these defects. Furthermore, in human tissues overexpression of USP16 reduces the expansion of normal fibroblasts and postnatal neural progenitors, whereas downregulation of USP16 partially rescues the proliferation defects of Down's syndrome fibroblasts. Taken together, these results suggest that USP16 has an important role in antagonizing the self-renewal and/or senescence pathways in Down's syndrome and could serve as an attractive target to ameliorate some of the associated pathologies.


Subject(s)
Down Syndrome/metabolism , Down Syndrome/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Ubiquitin Thiolesterase/metabolism , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Cell Proliferation , Cellular Senescence , Chromosomes, Human, Pair 21/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Down Syndrome/genetics , Epithelium/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Dosage , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Molecular Targeted Therapy , Trisomy/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitination
11.
Proc Natl Acad Sci U S A ; 112(7): 2145-50, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25646432

ABSTRACT

Macrophage-mediated programmed cell removal (PrCR) is an important mechanism of eliminating diseased and damaged cells before programmed cell death. The induction of PrCR by eat-me signals on tumor cells is countered by don't-eat-me signals such as CD47, which binds macrophage signal-regulatory protein α to inhibit phagocytosis. Blockade of CD47 on tumor cells leads to phagocytosis by macrophages. Here we demonstrate that the activation of Toll-like receptor (TLR) signaling pathways in macrophages synergizes with blocking CD47 on tumor cells to enhance PrCR. Bruton's tyrosine kinase (Btk) mediates TLR signaling in macrophages. Calreticulin, previously shown to be an eat-me signal on cancer cells, is activated in macrophages for secretion and cell-surface exposure by TLR and Btk to target cancer cells for phagocytosis, even if the cancer cells themselves do not express calreticulin.


Subject(s)
Calreticulin/physiology , Macrophages/immunology , Neoplasms/pathology , Protein-Tyrosine Kinases/metabolism , Toll-Like Receptors/physiology , Agammaglobulinaemia Tyrosine Kinase , Humans , Neoplasms/enzymology , Neoplasms/metabolism
12.
Blood ; 126(25): 2686-94, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26472751

ABSTRACT

Idelalisib is a first-in-class oral inhibitor of PI3Kδ that has shown substantial activity in patients with relapsed/refractory chronic lymphocytic leukemia (CLL). To evaluate idelalisib as initial therapy, 64 treatment-naïve older patients with CLL or small lymphocytic leukemia (median age, 71 years; range, 65-90) were treated with rituximab 375 mg/m(2) weekly ×8 and idelalisib 150 mg twice daily continuously for 48 weeks. Patients completing 48 weeks without progression could continue to receive idelalisib on an extension study. The median time on treatment was 22.4 months (range, 0.8-45.8+). The overall response rate (ORR) was 97%, including 19% complete responses. The ORR was 100% in patients with del(17p)/TP53 mutations and 97% in those with unmutated IGHV. Progression-free survival was 83% at 36 months. The most frequent (>30%) adverse events (any grade) were diarrhea (including colitis) (64%), rash (58%), pyrexia (42%), nausea (38%), chills (36%), cough (33%), and fatigue (31%). Elevated alanine transaminase/aspartate transaminase was seen in 67% of patients (23% grade ≥3). The combination of idelalisib and rituximab was highly active, resulting in durable disease control in treatment-naïve older patients with CLL. These results support the further development of idelalisib as initial treatment of CLL. This study is registered at ClinicalTrials.gov as #NCT01203930.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Purines/administration & dosage , Purines/adverse effects , Quinazolinones/administration & dosage , Quinazolinones/adverse effects , Rituximab/administration & dosage , Rituximab/adverse effects , Treatment Outcome
13.
J Neurooncol ; 123(3): 449-57, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25682090

ABSTRACT

Advancements in immunotherapeutics promise new possibilities for the creation of glioblastoma (GBM) treatment options. Ongoing work in cancer stem cell biology has progressively elucidated the role of this tumor sub-population in oncogenesis and has distinguished them as prime therapeutic targets. Current clinical trials take a multifaceted approach with the intention of harnessing the intrinsic cytotoxic capabilities of the immune system to directly target glioblastoma cancer stem cells (gCSC) or indirectly disrupt their stromal microenvironment. Monoclonal antibodies (mAbs), dendritic cell (DC) vaccines, and chimeric antigen receptor (CAR) T cell therapies have emerged as the most common approaches, with particular iterations incorporating cancer stem cell antigenic markers in their treatment designs. Ongoing work to determine the comprehensive antigenic profile of the gCSC in conjunction with efforts to counter the immunosuppressive tumor microenvironment holds much promise in future immunotherapeutic strategies against GBM. Given recent advancements in these fields, we believe there is tremendous potential to improve outcomes of GBM patients in the continuing evolution of immunotherapies targeted to cancer stem cell populations in GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Immunotherapy/methods , Neoplastic Stem Cells/physiology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Glioblastoma/immunology , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Immunosuppression Therapy
14.
Proc Natl Acad Sci U S A ; 109(17): 6662-7, 2012 Apr 24.
Article in English | MEDLINE | ID: mdl-22451913

ABSTRACT

CD47, a "don't eat me" signal for phagocytic cells, is expressed on the surface of all human solid tumor cells. Analysis of patient tumor and matched adjacent normal (nontumor) tissue revealed that CD47 is overexpressed on cancer cells. CD47 mRNA expression levels correlated with a decreased probability of survival for multiple types of cancer. CD47 is a ligand for SIRPα, a protein expressed on macrophages and dendritic cells. In vitro, blockade of CD47 signaling using targeted monoclonal antibodies enabled macrophage phagocytosis of tumor cells that were otherwise protected. Administration of anti-CD47 antibodies inhibited tumor growth in orthotopic immunodeficient mouse xenotransplantation models established with patient tumor cells and increased the survival of the mice over time. Anti-CD47 antibody therapy initiated on larger tumors inhibited tumor growth and prevented or treated metastasis, but initiation of the therapy on smaller tumors was potentially curative. The safety and efficacy of targeting CD47 was further tested and validated in immune competent hosts using an orthotopic mouse breast cancer model. These results suggest all human solid tumor cells require CD47 expression to suppress phagocytic innate immune surveillance and elimination. These data, taken together with similar findings with other human neoplasms, show that CD47 is a commonly expressed molecule on all cancers, its function to block phagocytosis is known, and blockade of its function leads to tumor cell phagocytosis and elimination. CD47 is therefore a validated target for cancer therapies.


Subject(s)
Antigens, Differentiation/metabolism , CD47 Antigen/immunology , Neoplasms/immunology , RNA, Messenger/genetics , Receptors, Immunologic/metabolism , Antibodies/immunology , CD47 Antigen/genetics , Cell Division/immunology , Flow Cytometry , Humans , Neoplasms/pathology , Neoplasms/therapy , Phagocytosis/immunology , Prognosis , Survival Analysis
16.
Nucleic Acids Res ; 40(Database issue): D98-108, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22116062

ABSTRACT

GeneDB (http://www.genedb.org) is a genome database for prokaryotic and eukaryotic pathogens and closely related organisms. The resource provides a portal to genome sequence and annotation data, which is primarily generated by the Pathogen Genomics group at the Wellcome Trust Sanger Institute. It combines data from completed and ongoing genome projects with curated annotation, which is readily accessible from a web based resource. The development of the database in recent years has focused on providing database-driven annotation tools and pipelines, as well as catering for increasingly frequent assembly updates. The website has been significantly redesigned to take advantage of current web technologies, and improve usability. The current release stores 41 data sets, of which 17 are manually curated and maintained by biologists, who review and incorporate data from the scientific literature, as well as other sources. GeneDB is primarily a production and annotation database for the genomes of predominantly pathogenic organisms.


Subject(s)
Databases, Genetic , Genomics , Molecular Sequence Annotation , Animals , Arthropods/genetics , Genome, Bacterial , Genome, Helminth , Genome, Protozoan , Internet , Vocabulary, Controlled
17.
Proc Natl Acad Sci U S A ; 108(11): 4453-8, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21368213

ABSTRACT

Diffuse intrinsic pontine gliomas (DIPGs) are highly aggressive tumors of childhood that are almost universally fatal. Our understanding of this devastating cancer is limited by a dearth of available tissue for study and by the lack of a faithful animal model. Intriguingly, DIPGs are restricted to the ventral pons and occur during a narrow window of middle childhood, suggesting dysregulation of a postnatal neurodevelopmental process. Here, we report the identification of a previously undescribed population of immunophenotypic neural precursor cells in the human and murine brainstem whose temporal and spatial distributions correlate closely with the incidence of DIPG and highlight a candidate cell of origin. Using early postmortem DIPG tumor tissue, we have established in vitro and xenograft models and find that the Hedgehog (Hh) signaling pathway implicated in many developmental and oncogenic processes is active in DIPG tumor cells. Modulation of Hh pathway activity has functional consequences for DIPG self-renewal capacity in neurosphere culture. The Hh pathway also appears to be active in normal ventral pontine precursor-like cells of the mouse, and unregulated pathway activity results in hypertrophy of the ventral pons. Together, these findings provide a foundation for understanding the cellular and molecular origins of DIPG, and suggest that the Hh pathway represents a potential therapeutic target in this devastating pediatric tumor.


Subject(s)
Brain Stem Neoplasms/metabolism , Brain Stem Neoplasms/pathology , Cell Lineage , Hedgehog Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Aggregation , Cell Proliferation , Humans , Intermediate Filament Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Nestin , Oligodendrocyte Transcription Factor 2 , Pons/growth & development , Pons/pathology , Signal Transduction , Time Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Water Sci Technol ; 69(3): 663-71, 2014.
Article in English | MEDLINE | ID: mdl-24552742

ABSTRACT

On-site wastewater treatment systems (OWS) are a potentially significant non-point source of nutrients to groundwater and surface waters, and are extensively used in coastal North Carolina. The goal of this study was to determine the treatment efficiency of four OWS in reducing total dissolved nitrogen (TDN) and dissolved organic carbon (DOC) concentrations before discharge to groundwater and/or adjacent surface water. Piezometers were installed for groundwater sample collection and nutrient analysis at four separate residences that use OWS. Septic tank effluent, groundwater, and surface water samples (from an adjacent stream) were collected four times during 2012 for TDN and DOC analysis and pH, temperature, electrical conductivity, and dissolved oxygen measurements. Treatment efficiencies from the tank to the groundwater beneath the drainfields ranged from 33 to 95% for TDN and 45 to 82% for DOC, although dilution accounted for most of the concentration reductions. There was a significant positive correlation between nitrate concentration and separation distance from trench bottom to water table and a significant negative correlation between DOC concentration and separation distance. The TDN and DOC transport (>15 m) from two OWS with groundwater saturated drainfield trenches was significant.


Subject(s)
Carbon/analysis , Groundwater/analysis , Nitrogen/analysis , Waste Management , North Carolina
19.
Neuro Oncol ; 26(6): 1109-1123, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38334125

ABSTRACT

BACKGROUND: Cellular senescence can have positive and negative effects on the body, including aiding in damage repair and facilitating tumor growth. Adamantinomatous craniopharyngioma (ACP), the most common pediatric sellar/suprasellar brain tumor, poses significant treatment challenges. Recent studies suggest that senescent cells in ACP tumors may contribute to tumor growth and invasion by releasing a senesecence-associated secretory phenotype. However, a detailed analysis of these characteristics has yet to be completed. METHODS: We analyzed primary tissue samples from ACP patients using single-cell, single-nuclei, and spatial RNA sequencing. We performed various analyses, including gene expression clustering, inferred senescence cells from gene expression, and conducted cytokine signaling inference. We utilized LASSO to select essential gene expression pathways associated with senescence. Finally, we validated our findings through immunostaining. RESULTS: We observed significant diversity in gene expression and tissue structure. Key factors such as NFKB, RELA, and SP1 are essential in regulating gene expression, while senescence markers are present throughout the tissue. SPP1 is the most significant cytokine signaling network among ACP cells, while the Wnt signaling pathway predominantly occurs between epithelial and glial cells. Our research has identified links between senescence-associated features and pathways, such as PI3K/Akt/mTOR, MYC, FZD, and Hedgehog, with increased P53 expression associated with senescence in these cells. CONCLUSIONS: A complex interplay between cellular senescence, cytokine signaling, and gene expression pathways underlies ACP development. Further research is crucial to understand how these elements interact to create novel therapeutic approaches for patients with ACP.


Subject(s)
Cellular Senescence , Craniopharyngioma , Machine Learning , Pituitary Neoplasms , Humans , Craniopharyngioma/metabolism , Craniopharyngioma/pathology , Craniopharyngioma/genetics , Pituitary Neoplasms/pathology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Phenotype , Gene Expression Regulation, Neoplastic , Child , Male , Female
20.
Nat Chem Biol ; 7(12): 925-34, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-22037470

ABSTRACT

Polyglutamine (polyQ) stretches exceeding a threshold length confer a toxic function to proteins that contain them and cause at least nine neurological disorders. The basis for this toxicity threshold is unclear. Although polyQ expansions render proteins prone to aggregate into inclusion bodies, this may be a neuronal coping response to more toxic forms of polyQ. The exact structure of these more toxic forms is unknown. Here we show that the monoclonal antibody 3B5H10 recognizes a species of polyQ protein in situ that strongly predicts neuronal death. The epitope selectively appears among some of the many low-molecular-weight conformational states assumed by expanded polyQ and disappears in higher-molecular-weight aggregated forms, such as inclusion bodies. These results suggest that protein monomers and possibly small oligomers containing expanded polyQ stretches can adopt a conformation that is recognized by 3B5H10 and is toxic or closely related to a toxic species.


Subject(s)
Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Peptides/chemistry , Peptides/toxicity , Antibodies, Monoclonal/immunology , Antibody Specificity , Cell Death/drug effects , Cells, Cultured , Epitopes/chemistry , Epitopes/immunology , Epitopes/toxicity , HEK293 Cells , Humans , Inclusion Bodies/chemistry , Molecular Weight , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Peptides/immunology , Structure-Activity Relationship , Trinucleotide Repeat Expansion
SELECTION OF CITATIONS
SEARCH DETAIL