Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell ; 186(8): 1689-1707, 2023 04 13.
Article in English | MEDLINE | ID: mdl-37059069

ABSTRACT

The nervous system governs both ontogeny and oncology. Regulating organogenesis during development, maintaining homeostasis, and promoting plasticity throughout life, the nervous system plays parallel roles in the regulation of cancers. Foundational discoveries have elucidated direct paracrine and electrochemical communication between neurons and cancer cells, as well as indirect interactions through neural effects on the immune system and stromal cells in the tumor microenvironment in a wide range of malignancies. Nervous system-cancer interactions can regulate oncogenesis, growth, invasion and metastatic spread, treatment resistance, stimulation of tumor-promoting inflammation, and impairment of anti-cancer immunity. Progress in cancer neuroscience may create an important new pillar of cancer therapy.


Subject(s)
Neoplasms , Neurosciences , Humans , Immune System , Neoplasms/pathology , Neurons/pathology , Tumor Microenvironment
2.
Cell ; 181(7): 1445-1449, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32533917

ABSTRACT

The COVID19 crisis has magnified the issues plaguing academic science, but it has also provided the scientific establishment with an unprecedented opportunity to reset. Shoring up the foundation of academic science will require a concerted effort between funding agencies, universities, and the public to rethink how we support scientists, with a special emphasis on early career researchers.


Subject(s)
Career Mobility , Research Personnel/trends , Research/trends , Achievement , Biomedical Research , Humans , Research Personnel/education , Science/education , Science/trends , Universities
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
4.
Nature ; 617(7961): 599-607, 2023 May.
Article in English | MEDLINE | ID: mdl-37138086

ABSTRACT

Gliomas synaptically integrate into neural circuits1,2. Previous research has demonstrated bidirectional interactions between neurons and glioma cells, with neuronal activity driving glioma growth1-4 and gliomas increasing neuronal excitability2,5-8. Here we sought to determine how glioma-induced neuronal changes influence neural circuits underlying cognition and whether these interactions influence patient survival. Using intracranial brain recordings during lexical retrieval language tasks in awake humans together with site-specific tumour tissue biopsies and cell biology experiments, we find that gliomas remodel functional neural circuitry such that task-relevant neural responses activate tumour-infiltrated cortex well beyond the cortical regions that are normally recruited in the healthy brain. Site-directed biopsies from regions within the tumour that exhibit high functional connectivity between the tumour and the rest of the brain are enriched for a glioblastoma subpopulation that exhibits a distinct synaptogenic and neuronotrophic phenotype. Tumour cells from functionally connected regions secrete the synaptogenic factor thrombospondin-1, which contributes to the differential neuron-glioma interactions observed in functionally connected tumour regions compared with tumour regions with less functional connectivity. Pharmacological inhibition of thrombospondin-1 using the FDA-approved drug gabapentin decreases glioblastoma proliferation. The degree of functional connectivity between glioblastoma and the normal brain negatively affects both patient survival and performance in language tasks. These data demonstrate that high-grade gliomas functionally remodel neural circuits in the human brain, which both promotes tumour progression and impairs cognition.


Subject(s)
Brain Neoplasms , Glioblastoma , Neural Pathways , Humans , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Thrombospondin 1/antagonists & inhibitors , Gabapentin/pharmacology , Gabapentin/therapeutic use , Disease Progression , Cognition , Survival Rate , Wakefulness , Biopsy , Cell Proliferation/drug effects
5.
Nature ; 623(7986): 366-374, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37914930

ABSTRACT

The role of the nervous system in the regulation of cancer is increasingly appreciated. In gliomas, neuronal activity drives tumour progression through paracrine signalling factors such as neuroligin-3 and brain-derived neurotrophic factor1-3 (BDNF), and also through electrophysiologically functional neuron-to-glioma synapses mediated by AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) receptors4,5. The consequent glioma cell membrane depolarization drives tumour proliferation4,6. In the healthy brain, activity-regulated secretion of BDNF promotes adaptive plasticity of synaptic connectivity7,8 and strength9-15. Here we show that malignant synapses exhibit similar plasticity regulated by BDNF. Signalling through the receptor tropomyosin-related kinase B16 (TrkB) to CAMKII, BDNF promotes AMPA receptor trafficking to the glioma cell membrane, resulting in increased amplitude of glutamate-evoked currents in the malignant cells. Linking plasticity of glioma synaptic strength to tumour growth, graded optogenetic control of glioma membrane potential demonstrates that greater depolarizing current amplitude promotes increased glioma proliferation. This potentiation of malignant synaptic strength shares mechanistic features with synaptic plasticity17-22 that contributes to memory and learning in the healthy brain23-26. BDNF-TrkB signalling also regulates the number of neuron-to-glioma synapses. Abrogation of activity-regulated BDNF secretion from the brain microenvironment or loss of glioma TrkB expression robustly inhibits tumour progression. Blocking TrkB genetically or pharmacologically abrogates these effects of BDNF on glioma synapses and substantially prolongs survival in xenograft models of paediatric glioblastoma and diffuse intrinsic pontine glioma. Together, these findings indicate that BDNF-TrkB signalling promotes malignant synaptic plasticity and augments tumour progression.


Subject(s)
Adaptation, Physiological , Glioma , Neuronal Plasticity , Synapses , Animals , Child , Humans , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Proliferation , Disease Progression , Glioma/metabolism , Glioma/pathology , Glutamic Acid/metabolism , Neurons/cytology , Neurons/metabolism , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptors, AMPA/metabolism , Signal Transduction , Synapses/metabolism , Tumor Microenvironment , Optogenetics
6.
Nature ; 573(7775): 539-545, 2019 09.
Article in English | MEDLINE | ID: mdl-31534222

ABSTRACT

High-grade gliomas are lethal brain cancers whose progression is robustly regulated by neuronal activity. Activity-regulated release of growth factors promotes glioma growth, but this alone is insufficient to explain the effect that neuronal activity exerts on glioma progression. Here we show that neuron and glioma interactions include electrochemical communication through bona fide AMPA receptor-dependent neuron-glioma synapses. Neuronal activity also evokes non-synaptic activity-dependent potassium currents that are amplified by gap junction-mediated tumour interconnections, forming an electrically coupled network. Depolarization of glioma membranes assessed by in vivo optogenetics promotes proliferation, whereas pharmacologically or genetically blocking electrochemical signalling inhibits the growth of glioma xenografts and extends mouse survival. Emphasizing the positive feedback mechanisms by which gliomas increase neuronal excitability and thus activity-regulated glioma growth, human intraoperative electrocorticography demonstrates increased cortical excitability in the glioma-infiltrated brain. Together, these findings indicate that synaptic and electrical integration into neural circuits promotes glioma progression.


Subject(s)
Brain/physiopathology , Electrical Synapses/pathology , Electrophysiological Phenomena , Glioma/physiopathology , Animals , Brain/cytology , Cell Membrane/pathology , Cell Proliferation , Gap Junctions/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Mice , Mice, Inbred NOD , Neurons/pathology , Optogenetics , Potassium/metabolism , Synaptic Transmission , Tumor Cells, Cultured
7.
Nature ; 549(7673): 533-537, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28959975

ABSTRACT

High-grade gliomas (HGG) are a devastating group of cancers, and represent the leading cause of brain tumour-related death in both children and adults. Therapies aimed at mechanisms intrinsic to glioma cells have translated to only limited success; effective therapeutic strategies will need also to target elements of the tumour microenvironment that promote glioma progression. Neuronal activity promotes the growth of a range of molecularly and clinically distinct HGG types, including adult and paediatric glioblastoma (GBM), anaplastic oligodendroglioma, and diffuse intrinsic pontine glioma (DIPG). An important mechanism that mediates this neural regulation of brain cancer is activity-dependent cleavage and secretion of the synaptic adhesion molecule neuroligin-3 (NLGN3), which promotes glioma proliferation through the PI3K-mTOR pathway. However, the necessity of NLGN3 for glioma growth, the proteolytic mechanism of NLGN3 secretion, and the further molecular consequences of NLGN3 secretion in glioma cells remain unknown. Here we show that HGG growth depends on microenvironmental NLGN3, identify signalling cascades downstream of NLGN3 binding in glioma, and determine a therapeutically targetable mechanism of secretion. Patient-derived orthotopic xenografts of paediatric GBM, DIPG and adult GBM fail to grow in Nlgn3 knockout mice. NLGN3 stimulates several oncogenic pathways, such as early focal adhesion kinase activation upstream of PI3K-mTOR, and induces transcriptional changes that include upregulation of several synapse-related genes in glioma cells. NLGN3 is cleaved from both neurons and oligodendrocyte precursor cells via the ADAM10 sheddase. ADAM10 inhibitors prevent the release of NLGN3 into the tumour microenvironment and robustly block HGG xenograft growth. This work defines a promising strategy for targeting NLGN3 secretion, which could prove transformative for HGG therapy.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Glioma/metabolism , Glioma/pathology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/metabolism , Adult , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Cell Proliferation , Child , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glioma/genetics , Heterografts , Humans , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Neoplasm Transplantation , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/pathology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Microenvironment
8.
Cancer Discov ; 14(4): 669-673, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38571430

ABSTRACT

SUMMARY: The field of cancer neuroscience has begun to define the contributions of nerves to cancer initiation and progression; here, we highlight the future directions of basic and translational cancer neuroscience for malignancies arising outside of the central nervous system.


Subject(s)
Neoplasms , Neurosciences , Humans , Central Nervous System , Forecasting , Proteomics
9.
Sci Transl Med ; 15(706): eadi5170, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37494471

ABSTRACT

Neuronal activity drives cancer progression through functional integration of malignant cell networks into neural circuitry.


Subject(s)
Neoplasms , Neurons , Humans , Neurons/physiology
10.
bioRxiv ; 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36711554

ABSTRACT

Neural activity is increasingly recognized as a critical regulator of cancer growth. In the brain, neuronal activity robustly influences glioma growth both through paracrine mechanisms and through electrochemical integration of malignant cells into neural circuitry via neuron-to-glioma synapses, while perisynaptic neurotransmitter signaling drives breast cancer brain metastasis growth. Outside of the CNS, innervation of tumors such as prostate, breast, pancreatic and gastrointestinal cancers by peripheral nerves similarly regulates cancer progression. However, the extent to which the nervous system regulates lung cancer progression, either in the lung or when metastatic to brain, is largely unexplored. Small cell lung cancer (SCLC) is a lethal high-grade neuroendocrine tumor that exhibits a strong propensity to metastasize to the brain. Here we demonstrate that, similar to glioma, metastatic SCLC cells in the brain co-opt neuronal activity-regulated mechanisms to stimulate growth and progression. Optogenetic stimulation of cortical neuronal activity drives proliferation and invasion of SCLC brain metastases. In the brain, SCLC cells exhibit electrical currents and consequent calcium transients in response to neuronal activity, and direct SCLC cell membrane depolarization is sufficient to promote the growth of SCLC tumors. In the lung, vagus nerve transection markedly inhibits primary lung tumor formation, progression and metastasis, highlighting a critical role for innervation in overall SCLC initiation and progression. Taken together, these studies illustrate that neuronal activity plays a crucial role in dictating SCLC pathogenesis in both primary and metastatic sites.

12.
Neuro Oncol ; 14(3): 315-25, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22156546

ABSTRACT

The phosphatidylinositol-3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway is activated in more than88% of glioblastomas (GBM). New drugs targeting this pathway are currently in clinical trials. However, noninvasive assessment of treatment response remains challenging. By using magnetic resonance spectroscopy (MRS), PI3K/Akt/mTOR pathway inhibition was monitored in 3 GBM cell lines (GS-2, GBM8, and GBM6; each with a distinct pathway activating mutation) through the measurement of 2 mechanistically linked MR biomarkers: phosphocholine (PC) and hyperpolarized lactate.(31)P MRS studies showed that treatment with the PI3K inhibitor LY294002 induced significant decreases in PC to 34 %± 9% of control in GS-2 cells, 48% ± 5% in GBM8, and 45% ± 4% in GBM6. The mTOR inhibitor everolimus also induced a significant decrease in PC to 62% ± 14%, 57% ± 1%, and 58% ± 1% in GS-2, GBM8, and GBM6 cells, respectively. Using hyperpolarized (13)C MRS, we demonstrated that hyperpolarized lactate levels were significantly decreased following PI3K/Akt/mTOR pathway inhibition in all 3 cell lines to 51% ± 10%, 62% ± 3%, and 58% ± 2% of control with LY294002 and 72% ± 3%, 61% ± 2%, and 66% ± 3% of control with everolimus in GS-2, GBM8, and GBM6 cells, respectively. These effects were mediated by decreases in the activity and expression of choline kinase α and lactate dehydrogenase, which respectively control PC and lactate production downstream of HIF-1. Treatment with the DNA damaging agent temozolomide did not have an effect on either biomarker in any cell line. This study highlights the potential of PC and hyperpolarized lactate as noninvasive MR biomarkers of response to targeted inhibitors in GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Glioblastoma/enzymology , Lactic Acid/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylcholine/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Biomarkers/metabolism , Cell Line, Tumor , Chromones/pharmacology , Everolimus , Glioblastoma/metabolism , Humans , Magnetic Resonance Spectroscopy , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
13.
Cancer Res ; 70(19): 7400-10, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20858719

ABSTRACT

The recent development of hyperpolarized (13)C magnetic resonance spectroscopic imaging provides a novel method for in vivo metabolic imaging with potential applications for detection of cancer and response to treatment. Chemotherapy-induced apoptosis was shown to decrease the flux of hyperpolarized (13)C label from pyruvate to lactate due to depletion of NADH, the coenzyme of lactate dehydrogenase. In contrast, we show here that in PC-3MM2 tumors, inhibition of platelet-derived growth factor receptor with imatinib reduces the conversion of hyperpolarized pyruvate to lactate by lowering the expression of lactate dehydrogenase itself. This was accompanied by reduced expression of vascular endothelial growth factor and glutaminase, and is likely mediated by reduced expression of their transcriptional factors hypoxia-inducible factor-1 and c-Myc. Our results indicate that hyperpolarized (13)C MRSI could potentially detect the molecular effect of various cell signaling inhibitors, thus providing a radiation-free method to predict tumor response.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Animals , Benzamides , Capillary Permeability/drug effects , Carbon Isotopes , Humans , Imatinib Mesylate , L-Lactate Dehydrogenase/biosynthesis , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Magnetic Resonance Spectroscopy/methods , Male , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Paclitaxel/pharmacology , Piperazines/pharmacology , Prostatic Neoplasms/blood supply , Pyrimidines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/biosynthesis
14.
Cancer Res ; 70(4): 1296-305, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20145128

ABSTRACT

Numerous mechanism-based anticancer drugs that target the phosphatidylinositol 3-kinase (PI3K) pathway are in clinical trials. However, it remains challenging to assess responses by traditional imaging methods. Here, we show for the first time the efficacy of hyperpolarized (13)C magnetic resonance spectroscopy (MRS) in detecting the effect of PI3K inhibition by monitoring hyperpolarized [1-(13)C]lactate levels produced from hyperpolarized [1-(13)C]pyruvate through lactate dehydrogenase (LDH) activity. In GS-2 glioblastoma cells, PI3K inhibition by LY294002 or everolimus caused hyperpolarized lactate to drop to 42 +/- 12% and to 76 +/- 5%, respectively. In MDA-MB-231 breast cancer cells, hyperpolarized lactate dropped to 71 +/- 15% after treatment with LY294002. These reductions were correlated with reductions in LDH activity to 48 +/- 4%, 63 +/- 4%, and 69 +/- 12%, respectively, and were associated with a drop in levels of LDHA mRNA and LDHA and hypoxia-inducible factor-1alpha proteins. Supporting these findings, tumor growth inhibition achieved by everolimus in murine GS-2 xenografts was associated with a drop in the hyperpolarized lactate-to-pyruvate ratio detected by in vivo MRS imaging, whereas an increase in this ratio occurred with tumor growth in control animals. Taken together, our findings illustrate the application of hyperpolarized (13)C MRS of pyruvate to monitor alterations in LDHA activity and expression caused by PI3K pathway inhibition, showing the potential of this method for noninvasive imaging of drug target modulation.


Subject(s)
Carbon Isotopes , Enzyme Inhibitors/therapeutic use , Magnetic Resonance Spectroscopy/methods , Monitoring, Physiologic/methods , Neoplasms/diagnosis , Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Animals , Carbon Isotopes/pharmacokinetics , Chromones/pharmacology , Chromones/therapeutic use , Drug Delivery Systems , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Everolimus , Glioblastoma/diagnosis , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Lactic Acid/pharmacokinetics , Mice , Mice, Nude , Morpholines/pharmacology , Morpholines/therapeutic use , Neoplasms/pathology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Sirolimus/therapeutic use , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL