Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
JCI Insight ; 9(2)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38258905

ABSTRACT

Pain of unknown etiology is frequent in individuals with the tumor predisposition syndrome neurofibromatosis 1 (NF1), even when tumors are absent. Nerve Schwann cells (SCs) were recently shown to play roles in nociceptive processing, and we find that chemogenetic activation of SCs is sufficient to induce afferent and behavioral mechanical hypersensitivity in wild-type mice. In mouse models, animals showed afferent and behavioral hypersensitivity when SCs, but not neurons, lacked Nf1. Importantly, hypersensitivity corresponded with SC-specific upregulation of mRNA encoding glial cell line-derived neurotrophic factor (GDNF), independently of the presence of tumors. Neuropathic pain-like behaviors in the NF1 mice were inhibited by either chemogenetic silencing of SC calcium or by systemic delivery of GDNF-targeting antibodies. Together, these findings suggest that alterations in SCs directly modulate mechanical pain and suggest cell-specific treatment strategies to ameliorate pain in individuals with NF1.


Subject(s)
Hypersensitivity , Neuralgia , Neurofibromatosis 1 , Animals , Mice , Neurofibromatosis 1/genetics , Nociception , Glial Cell Line-Derived Neurotrophic Factor/genetics , Schwann Cells
2.
J Pharmacol Exp Ther ; 385(2): 106-116, 2023 05.
Article in English | MEDLINE | ID: mdl-36849412

ABSTRACT

Individuals with neurofibromatosis type 1 develop rat sarcoma virus (RAS)-mitogen-activated protein kinase-mitogen-activated and extracellular signal-regulated kinase (RAS-MAPK-MEK)-driven nerve tumors called neurofibromas. Although MEK inhibitors transiently reduce volumes of most plexiform neurofibromas in mouse models and in neurofibromatosis type 1 (NF1) patients, therapies that increase the efficacy of MEK inhibitors are needed. BI-3406 is a small molecule that prevents Son of Sevenless (SOS)1 interaction with Kirsten rat sarcoma viral oncoprotein (KRAS)-GDP, interfering with the RAS-MAPK cascade upstream of MEK. Single agent SOS1 inhibition had no significant effect in the DhhCre;Nf1 fl/fl mouse model of plexiform neurofibroma, but pharmacokinetics (PK)-driven combination of selumetinib with BI-3406 significantly improved tumor parameters. Tumor volumes and neurofibroma cell proliferation, reduced by MEK inhibition, were further reduced by the combination. Neurofibromas are rich in ionized calcium binding adaptor molecule 1 (Iba1)+ macrophages; combination treatment resulted in small and round macrophages, with altered cytokine expression indicative of altered activation. The significant effects of MEK inhibitor plus SOS1 inhibition in this preclinical study suggest potential clinical benefit of dual targeting of the RAS-MAPK pathway in neurofibromas. SIGNIFICANCE STATEMENT: Interfering with the RAS-mitogen-activated protein kinase (RAS-MAPK) cascade upstream of mitogen activated protein kinase kinase (MEK), together with MEK inhibition, augment effects of MEK inhibition on neurofibroma volume and tumor macrophages in a preclinical model system. This study emphasizes the critical role of the RAS-MAPK pathway in controlling tumor cell proliferation and the tumor microenvironment in benign neurofibromas.


Subject(s)
Neurofibroma, Plexiform , Neurofibroma , Neurofibromatosis 1 , Animals , Mice , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases , Neurofibroma/drug therapy , Neurofibroma, Plexiform/drug therapy , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/therapeutic use , Tumor Microenvironment , SOS1 Protein/metabolism
3.
Elife ; 112022 03 21.
Article in English | MEDLINE | ID: mdl-35311647

ABSTRACT

Neurofibromatosis type 1 (NF1) is characterized by nerve tumors called neurofibromas, in which Schwann cells (SCs) show deregulated RAS signaling. NF1 is also implicated in regulation of cAMP. We identified the G-protein-coupled receptor (GPCR) P2ry14 in human neurofibromas, neurofibroma-derived SC precursors (SCPs), mature SCs, and mouse SCPs. Mouse Nf1-/- SCP self-renewal was reduced by genetic or pharmacological inhibition of P2ry14. In a mouse model of NF1, genetic deletion of P2ry14 rescued low cAMP signaling, increased mouse survival, delayed neurofibroma initiation, and improved SC Remak bundles. P2ry14 signals via Gi to increase intracellular cAMP, implicating P2ry14 as a key upstream regulator of cAMP. We found that elevation of cAMP by either blocking the degradation of cAMP or by using a P2ry14 inhibitor diminished NF1-/- SCP self-renewal in vitro and neurofibroma SC proliferation in in vivo. These studies identify P2ry14 as a critical regulator of SCP self-renewal, SC proliferation, and neurofibroma initiation.


Subject(s)
Cyclic AMP/metabolism , Neurofibroma , Neurofibromatosis 1 , Receptors, Purinergic P2Y/metabolism , Animals , Cell Self Renewal , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Mice , Neurofibroma/genetics , Neurofibroma/metabolism , Neurofibroma/pathology , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Schwann Cells/metabolism
4.
Oncogene ; 40(24): 4229-4241, 2021 06.
Article in English | MEDLINE | ID: mdl-34079083

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNST) are aggressive soft-tissue sarcomas that cause significant mortality in adults with neurofibromatosis type 1. We compared gene expression of growth factors in normal human nerves to MPNST and normal human Schwann cells to MPNST cell lines. We identified WNT5A as the most significantly upregulated ligand-coding gene and verified its protein expression in MPNST cell lines and tumors. In many contexts WNT5A acts as an oncogene. However, inhibiting WNT5A expression using shRNA did not alter MPNST cell proliferation, invasion, migration, or survival in vitro. Rather, shWNT5A-treated MPNST cells upregulated mRNAs associated with the remodeling of extracellular matrix and with immune cell communication. In addition, these cells secreted increased amounts of the proinflammatory cytokines CXCL1, CCL2, IL6, CXCL8, and ICAM1. Versus controls, shWNT5A-expressing MPNST cells formed larger tumors in vivo. Grafted tumors contained elevated macrophage/stromal cells, larger and more numerous blood vessels, and increased levels of Mmp9, Cxcl13, Lipocalin-1, and Ccl12. In some MPNST settings, these effects were mimicked by targeting the WNT5A receptor ROR2. These data suggest that the non-canonical Wnt ligand WNT5A inhibits MPNST tumor formation by modulating the MPNST microenvironment, so that blocking WNT5A accelerates tumor growth in vivo.


Subject(s)
Cell Proliferation/genetics , Nerve Sheath Neoplasms/genetics , Tumor Microenvironment/genetics , Wnt-5a Protein/genetics , Cell Line, Tumor , Cell Movement/genetics , Extracellular Matrix/genetics , Humans , Nerve Sheath Neoplasms/pathology , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibrosarcoma/genetics , Neurofibrosarcoma/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Schwann Cells/pathology
6.
Cancer Res ; 80(21): 4720-4730, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32816910

ABSTRACT

Plexiform neurofibromas are benign nerve sheath Schwann cell tumors characterized by biallelic mutations in the neurofibromatosis type 1 (NF1) tumor suppressor gene. Atypical neurofibromas show additional frequent loss of CDKN2A/Ink4a/Arf and may be precursor lesions of aggressive malignant peripheral nerve sheath tumors (MPNST). Here we combined loss of Nf1 in developing Schwann cells with global Ink4a/Arf loss and identified paraspinal plexiform neurofibromas and atypical neurofibromas. Upon transplantation, atypical neurofibromas generated genetically engineered mice (GEM)-PNST similar to human MPNST, and tumors showed reduced p16INK4a protein and reduced senescence markers, confirming susceptibility to transformation. Superficial GEM-PNST contained regions of nerve-associated plexiform neurofibromas or atypical neurofibromas and grew rapidly on transplantation. Transcriptome analyses showed similarities to corresponding human tumors. Thus, we recapitulated nerve tumor progression in NF1 and provided preclinical platforms for testing therapies at each tumor grade. These results support a tumor progression model in which loss of NF1 in Schwann cells drives plexiform neurofibromas formation, additional loss of Ink4a/Arf contributes to atypical neurofibromas formation, and further changes underlie transformation to MPNST. SIGNIFICANCE: New mouse models recapitulate the stepwise progression of NF1 tumors and will be useful to define effective treatments that halt tumor growth and tumor progression in NF1.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Neurofibroma/genetics , Neurofibroma/pathology , Neurofibrosarcoma/genetics , Neurofibrosarcoma/pathology , Animals , Disease Models, Animal , Disease Progression , Genes, Neurofibromatosis 1 , Mice , Mice, Mutant Strains , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology
7.
JCI Insight ; 4(3)2019 Feb 07.
Article in English | MEDLINE | ID: mdl-30728335

ABSTRACT

Plexiform neurofibroma is a major contributor to morbidity in patients with neurofibromatosis type I (NF1). Macrophages and mast cells infiltrate neurofibroma, and data from mouse models implicate these leukocytes in neurofibroma development. Antiinflammatory therapy targeting these cell populations has been suggested as a means to prevent neurofibroma development. Here, we compare gene expression in Nf1-mutant nerves, which invariably form neurofibroma, and show disruption of neuron-glial cell interactions and immune cell infiltration to mouse models, which rarely progresses to neurofibroma with or without disruption of neuron-glial cell interactions. We find that the chemokine Cxcl10 is uniquely upregulated in NF1 mice that invariably develop neurofibroma. Global deletion of the CXCL10 receptor Cxcr3 prevented neurofibroma development in these neurofibroma-prone mice, and an anti-Cxcr3 antibody somewhat reduced tumor numbers. Cxcr3 expression localized to T cells and DCs in both inflamed nerves and neurofibromas, and Cxcr3 expression was necessary to sustain elevated macrophage numbers in Nf1-mutant nerves. To our knowledge, these data support a heretofore-unappreciated role for T cells and DCs in neurofibroma initiation.

8.
Oncogene ; 38(15): 2876-2884, 2019 04.
Article in English | MEDLINE | ID: mdl-30542122

ABSTRACT

Plexiform neurofibroma, a benign peripheral nerve tumor, is associated with the biallelic loss of function of the NF1 tumor suppressor in Schwann cells. Here, we show that FLLL32, a small molecule inhibitor of JAK2/STAT3 signaling, reduces neurofibroma growth in mice with conditional, biallelic deletion of Nf1 in the Schwann cell lineage. FLLL32 treatment or Stat3 deletion in tumor cells reduced inflammatory cytokine expression and tumor macrophage numbers in neurofibroma. Although STAT3 inhibition downregulated the chemokines CCL2 and CCL12, which can signal through CCR2 to recruit macrophages to peripheral nerves, deletion of Ccr2 did not improve survival or reduce macrophage numbers in neurofibroma-bearing mice. Interestingly, Iba1+; F4/80+;CD11b+ macrophages accounted for ~20-40% of proliferating cells in untreated tumors. FLLL32 suppressed macrophage proliferation, implicating STAT3-dependent, local proliferation in neurofibroma macrophage accumulation, and decreased Schwann cell proliferation and increased Schwann cell death. The functions of STAT3 signaling in neurofibroma Schwann cells and macrophages, and its relevance as a therapeutic target in neurofibroma, merit further investigation.


Subject(s)
Cell Proliferation/drug effects , Curcumin/analogs & derivatives , Neurofibroma, Plexiform/drug therapy , Neurofibroma, Plexiform/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Animals , Cell Death/drug effects , Chemokine CCL2/metabolism , Curcumin/pharmacology , Cytokines/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Humans , Janus Kinase 2/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Monocyte Chemoattractant Proteins/metabolism , Neurofibromatosis 1/metabolism , Schwann Cells/drug effects , Schwann Cells/metabolism , Signal Transduction/drug effects
9.
Commun Biol ; 1: 158, 2018.
Article in English | MEDLINE | ID: mdl-30302402

ABSTRACT

Neurofibromatosis Type 1 (NF1) is a genetic disease caused by mutations in Neurofibromin 1 (NF1). NF1 patients present with a variety of clinical manifestations and are predisposed to cancer development. Many NF1 animal models have been developed, yet none display the spectrum of disease seen in patients and the translational impact of these models has been limited. We describe a minipig model that exhibits clinical hallmarks of NF1, including café au lait macules, neurofibromas, and optic pathway glioma. Spontaneous loss of heterozygosity is observed in this model, a phenomenon also described in NF1 patients. Oral administration of a mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor suppresses Ras signaling. To our knowledge, this model provides an unprecedented opportunity to study the complex biology and natural history of NF1 and could prove indispensable for development of imaging methods, biomarkers, and evaluation of safety and efficacy of NF1-targeted therapies.

11.
Cancer Cell ; 33(2): 292-308.e7, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29438698

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive Schwann cell (SC)-lineage-derived sarcomas. Molecular events driving SC-to-MPNST transformation are incompletely understood. Here, we show that human MPNSTs exhibit elevated HIPPO-TAZ/YAP expression, and that TAZ/YAP hyperactivity in SCs caused by Lats1/2 loss potently induces high-grade nerve-associated tumors with full penetrance. Lats1/2 deficiency reprograms SCs to a cancerous, progenitor-like phenotype and promotes hyperproliferation. Conversely, disruption of TAZ/YAP activity alleviates tumor burden in Lats1/2-deficient mice and inhibits human MPNST cell proliferation. Moreover, genome-wide profiling reveals that TAZ/YAP-TEAD1 directly activates oncogenic programs, including platelet-derived growth factor receptor (PDGFR) signaling. Co-targeting TAZ/YAP and PDGFR pathways inhibits tumor growth. Thus, our findings establish a previously unrecognized convergence between Lats1/2-TAZ/YAP signaling and MPNST pathogenesis, revealing potential therapeutic targets in these untreatable tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Schwann Cells/cytology , Animals , Cell Cycle Proteins , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic , Humans , Mice , Signal Transduction/genetics , Transcription Factors , YAP-Signaling Proteins
12.
Glia ; 65(12): 1990-2002, 2017 12.
Article in English | MEDLINE | ID: mdl-28856719

ABSTRACT

Costello syndrome (CS) is a gain of function Rasopathy caused by heterozygous activating mutations in the HRAS gene. Patients show brain dysfunction that can include abnormal brain white matter. Transgenic activation of HRas in the entire mouse oligodendrocyte lineage resulted in myelin defects and behavioral abnormalities, suggesting roles for disrupted myelin in CS brain dysfunction. Here, we studied a mouse model in which the endogenous HRas gene is conditionally replaced by mutant HRasG12V in mature oligodendrocytes, to separate effects in mature myelinating cells from developmental events. Increased myelin thickness due to decompaction was detectable within one month of HRasG12V expression in the corpus callosum of adult mice. Increases in active ERK and Nitric Oxide (NO) were present in HRas mutants and inhibition of NO synthase (NOS) or MEK each partially rescued myelin decompaction. In addition, genetic or pharmacologic inhibition of Notch signaling improved myelin compaction. Complete rescue of myelin structure required dual drug treatments combining MAPK, NO, or Notch inhibition; with MEK + NOS blockade producing the most robust effect. We suggest that individual or concomitant blockade of these pathways in CS patients may improve aspects of brain function.


Subject(s)
MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Myelin Sheath/metabolism , Nitric Oxide/metabolism , Oligodendroglia/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Notch/metabolism , Animals , Corpus Callosum/pathology , Corpus Callosum/ultrastructure , Enzyme Inhibitors/pharmacology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Mutation/genetics , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/ultrastructure , NG-Nitroarginine Methyl Ester/pharmacology , Oligodendroglia/ultrastructure , Proto-Oncogene Proteins p21(ras)/genetics , Tamoxifen/pharmacology
13.
Cell Rep ; 19(3): 545-557, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423318

ABSTRACT

The RASopathy neurofibromatosis type 1 (NF1) is one of the most common autosomal dominant genetic disorders. In NF1 patients, neurological issues may result from damaged myelin, and mice with a neurofibromin gene (Nf1) mutation show white matter (WM) defects including myelin decompaction. Using mouse genetics, we find that altered Nf1 gene-dose in mature oligodendrocytes results in progressive myelin defects and behavioral abnormalities mediated by aberrant Notch activation. Blocking Notch, upstream mitogen-activated protein kinase (MAPK), or nitric oxide signaling rescues myelin defects in hemizygous Nf1 mutants, and pharmacological gamma secretase inhibition rescues aberrant behavior with no effects in wild-type (WT) mice. Concomitant pathway inhibition rescues myelin abnormalities in homozygous mutants. Notch activation is also observed in Nf1+/- mouse brains, and cells containing active Notch are increased in NF1 patient WM. We thus identify Notch as an Nf1 effector regulating myelin structure and behavior in a RASopathy and suggest that inhibition of Notch signaling may be a therapeutic strategy for NF1.


Subject(s)
Myelin Sheath/metabolism , Neurofibromin 1/metabolism , Receptors, Notch/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Behavior, Animal , Cell Count , Claudins/metabolism , Gene Dosage , Humans , MAP Kinase Signaling System , Mice, Inbred C57BL , Models, Biological , Mutation/genetics , Neuroglia/metabolism , Nitric Oxide/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Signal Transduction , ras Proteins/metabolism
14.
Oncotarget ; 8(11): 17412-17427, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28147331

ABSTRACT

Malignant peripheral nerve sheath tumor (MPNST) and neuroblastoma models respond to the investigational small molecule Aurora A kinase inhibitor, alisertib. We previously reported that MPNST and neuroblastomas are also susceptible to oncolytic herpes virus (oHSV) therapy. Herein, we show that combination of alisertib and HSV1716, a virus derived from HSV-1 and attenuated by deletion of RL1, exhibits significantly increased antitumor efficacy compared to either monotherapy. Alisertib and HSV1716 reduced tumor growth and increased survival in two xenograft models of MPNST and neuroblastoma. We found the enhanced antitumor effect was due to multiple mechanisms that likely each contribute to the combination effect. First, oncolytic herpes virus increased the sensitivity of uninfected cells to alisertib cytotoxicity, a process we term virus-induced therapeutic adjuvant (VITA). Second, alisertib increased peak virus production and slowed virus clearance from tumors, both likely a consequence of it preventing virus-mediated increase of intratumoral NK cells. We also found that alisertib inhibited virus-induced accumulation of intratumoral myeloid derived suppressor cells, which normally are protumorigenic. Our data suggest that clinical trials of the combination of oHSV and alisertib are warranted in patients with neuroblastoma or MPNST.


Subject(s)
Antineoplastic Agents/administration & dosage , Azepines/administration & dosage , Neurilemmoma/pathology , Neuroblastoma/pathology , Oncolytic Virotherapy/methods , Pyrimidines/administration & dosage , Animals , Aurora Kinase A/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Combined Modality Therapy , Cytotoxicity, Immunologic/immunology , Female , Flow Cytometry , Herpesvirus 1, Human , Humans , Immunity, Innate/immunology , Immunohistochemistry , Mice , Mice, Nude , Xenograft Model Antitumor Assays
15.
N Engl J Med ; 375(26): 2550-2560, 2016 12 29.
Article in English | MEDLINE | ID: mdl-28029918

ABSTRACT

BACKGROUND: Effective medical therapies are lacking for the treatment of neurofibromatosis type 1-related plexiform neurofibromas, which are characterized by elevated RAS-mitogen-activated protein kinase (MAPK) signaling. METHODS: We conducted a phase 1 trial of selumetinib (AZD6244 or ARRY-142886), an oral selective inhibitor of MAPK kinase (MEK) 1 and 2, in children who had neurofibromatosis type 1 and inoperable plexiform neurofibromas to determine the maximum tolerated dose and to evaluate plasma pharmacokinetics. Selumetinib was administered twice daily at a dose of 20 to 30 mg per square meter of body-surface area on a continuous dosing schedule (in 28-day cycles). We also tested selumetinib using a mouse model of neurofibromatosis type 1-related neurofibroma. Response to treatment (i.e., an increase or decrease from baseline in the volume of plexiform neurofibromas) was monitored by using volumetric magnetic resonance imaging analysis to measure the change in size of the plexiform neurofibroma. RESULTS: A total of 24 children (median age, 10.9 years; range, 3.0 to 18.5) with a median tumor volume of 1205 ml (range, 29 to 8744) received selumetinib. Patients were able to receive selumetinib on a long-term basis; the median number of cycles was 30 (range, 6 to 56). The maximum tolerated dose was 25 mg per square meter (approximately 60% of the recommended adult dose). The most common toxic effects associated with selumetinib included acneiform rash, gastrointestinal effects, and asymptomatic creatine kinase elevation. The results of pharmacokinetic evaluations of selumetinib among the children in this trial were similar to those published for adults. Treatment with selumetinib resulted in confirmed partial responses (tumor volume decreases from baseline of ≥20%) in 17 of the 24 children (71%) and decreases from baseline in neurofibroma volume in 12 of 18 mice (67%). Disease progression (tumor volume increase from baseline of ≥20%) has not been observed to date. Anecdotal evidence of decreases in tumor-related pain, disfigurement, and functional impairment was observed. CONCLUSIONS: Our early-phase data suggested that children with neurofibromatosis type 1 and inoperable plexiform neurofibromas benefited from long-term dose-adjusted treatment with selumetinib without having excess toxic effects. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT01362803 .).


Subject(s)
Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurofibroma, Plexiform/drug therapy , Neurofibromatosis 1/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Adolescent , Animals , Benzimidazoles/adverse effects , Child , Child, Preschool , Disease Models, Animal , Disease Progression , Female , Humans , Magnetic Resonance Imaging , Male , Mice , Neurofibroma, Plexiform/diagnostic imaging , Protein Kinase Inhibitors/adverse effects
16.
Oncotarget ; 7(33): 53191-53203, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27448963

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are soft tissue sarcomas that are a major cause of mortality of Neurofibromatosis type 1 (NF1) patients. MPNST patients have few therapeutic options available and only complete surgical resection can be curative. MPNST formation and survival are dependent on activated ß-catenin signaling. The goal of this study was to determine if inhibition of the CK2 enzyme can be therapeutically exploited in MPNSTs, given CK2's role in mainta ining oncogenic phenotypes including stabilization of ß-catenin. We found that CK2α is over-expressed in MPNSTs and is critical for maintaining cell survival, as the CK2 inhibitor, CX-4945 (Silmitasertib), and shRNA targeting CK2α each significantly reduce MPNST cell viability. These effects were preceded by loss of critical signaling pathways in MPNSTs, including destabilization of ß-catenin and TCF8. CX-4945 administration in vivo slowed tumor growth and extends survival time. We conclude that CK2 inhibition is a promising approach to blocking ß-catenin in MPNST cells, although combinatorial therapies may be required for maximal efficacy.


Subject(s)
Apoptosis/drug effects , Casein Kinase II/antagonists & inhibitors , Naphthyridines/pharmacology , Nerve Sheath Neoplasms/drug therapy , beta Catenin/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/genetics , Benzamides/administration & dosage , Benzamides/pharmacology , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Diphenylamine/administration & dosage , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Drug Synergism , Female , Humans , Mice, Nude , Naphthyridines/administration & dosage , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/metabolism , Phenazines , Proteolysis/drug effects , RNA Interference , Xenograft Model Antitumor Assays , beta Catenin/genetics
17.
J Neurosci ; 36(21): 5724-35, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27225763

ABSTRACT

UNLABELLED: Spinal reflex circuit development requires the precise regulation of axon trajectories, synaptic specificity, and synapse formation. Of these three crucial steps, the molecular mechanisms underlying synapse formation between group Ia proprioceptive sensory neurons and motor neurons is the least understood. Here, we show that the Rho GTPase Cdc42 controls synapse formation in monosynaptic sensory-motor connections in presynaptic, but not postsynaptic, neurons. In mice lacking Cdc42 in presynaptic sensory neurons, proprioceptive sensory axons appropriately reach the ventral spinal cord, but significantly fewer synapses are formed with motor neurons compared with wild-type mice. Concordantly, electrophysiological analyses show diminished EPSP amplitudes in monosynaptic sensory-motor circuits in these mutants. Temporally targeted deletion of Cdc42 in sensory neurons after sensory-motor circuit establishment reveals that Cdc42 does not affect synaptic transmission. Furthermore, addition of the synaptic organizers, neuroligins, induces presynaptic differentiation of wild-type, but not Cdc42-deficient, proprioceptive sensory neurons in vitro Together, our findings demonstrate that Cdc42 in presynaptic neurons is required for synapse formation in monosynaptic sensory-motor circuits. SIGNIFICANCE STATEMENT: Group Ia proprioceptive sensory neurons form direct synapses with motor neurons, but the molecular mechanisms underlying synapse formation in these monosynaptic sensory-motor connections are unknown. We show that deleting Cdc42 in sensory neurons does not affect proprioceptive sensory axon targeting because axons reach the ventral spinal cord appropriately, but these neurons form significantly fewer presynaptic terminals on motor neurons. Electrophysiological analysis further shows that EPSPs are decreased in these mice. Finally, we demonstrate that Cdc42 is involved in neuroligin-dependent presynaptic differentiation of proprioceptive sensory neurons in vitro These data suggest that Cdc42 in presynaptic sensory neurons is essential for proper synapse formation in the development of monosynaptic sensory-motor circuits.


Subject(s)
Axon Guidance/physiology , Motor Neurons/physiology , Neurogenesis/physiology , Presynaptic Terminals/physiology , Sensory Receptor Cells/physiology , cdc42 GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Cells, Cultured , Female , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Male , Mice , Mice, Knockout , Motor Neurons/cytology , Presynaptic Terminals/ultrastructure , Sensory Receptor Cells/cytology , Spinal Cord/cytology , Spinal Cord/physiology
18.
J Clin Invest ; 126(2): 571-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26690703

ABSTRACT

Airway hyperresponsiveness (AHR) affects 55%-77% of children with sickle cell disease (SCD) and occurs even in the absence of asthma. While asthma increases SCD morbidity and mortality, the mechanisms underlying the high AHR prevalence in a hemoglobinopathy remain unknown. We hypothesized that placenta growth factor (PlGF), an erythroblast-secreted factor that is elevated in SCD, mediates AHR. In allergen-exposed mice, loss of Plgf dampened AHR, reduced inflammation and eosinophilia, and decreased expression of the Th2 cytokine IL-13 and the leukotriene-synthesizing enzymes 5-lipoxygenase and leukotriene-C4-synthase. Plgf-/- mice treated with leukotrienes phenocopied the WT response to allergen exposure; conversely, anti-PlGF Ab administration in WT animals blunted the AHR. Notably, Th2-mediated STAT6 activation further increased PlGF expression from lung epithelium, eosinophils, and macrophages, creating a PlGF/leukotriene/Th2-response positive feedback loop. Similarly, we found that the Th2 response in asthma patients is associated with increased expression of PlGF and its downstream genes in respiratory epithelial cells. In an SCD mouse model, we observed increased AHR and higher leukotriene levels that were abrogated by anti-PlGF Ab or the 5-lipoxygenase inhibitor zileuton. Overall, our findings indicate that PlGF exacerbates AHR and uniquely links the leukotriene and Th2 pathways in asthma. These data also suggest that zileuton and anti-PlGF Ab could be promising therapies to reduce pulmonary morbidity in SCD.


Subject(s)
Anemia, Sickle Cell/metabolism , Asthma/metabolism , Interleukin-13/metabolism , Leukotrienes/metabolism , Pregnancy Proteins/metabolism , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/pathology , Animals , Asthma/etiology , Asthma/genetics , Asthma/pathology , Disease Models, Animal , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Interleukin-13/genetics , Leukotrienes/genetics , Mice , Mice, Knockout , Placenta Growth Factor , Pregnancy Proteins/genetics , Th2 Cells/metabolism , Th2 Cells/pathology
19.
Blood ; 126(15): 1844-55, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26286849

ABSTRACT

Sickle cell disease (SCD) results in vascular occlusions, chronic hemolytic anemia, and cumulative organ damage. A conspicuous feature of SCD is chronic inflammation and coagulation system activation. Thrombin (factor IIa [FIIa]) is both a central protease in hemostasis and a key modifier of inflammatory processes. To explore the hypothesis that reduced prothrombin (factor II [FII]) levels in SCD will limit vaso-occlusion, vasculopathy, and inflammation, we used 2 strategies to suppress FII in SCD mice. Weekly administration of FII antisense oligonucleotide "gapmer" to Berkeley SCD mice to selectively reduce circulating FII levels to ∼10% of normal for 15 weeks significantly diminished early mortality. More comprehensive, long-term comparative studies were done using mice with genetic diminution of circulating FII. Here, cohorts of FII(lox/-) mice (constitutively carrying ∼10% normal FII) and FII(WT) mice were tracked in parallel for a year following the imposition of SCD via hematopoietic stem cell transplantation. This genetically imposed suppression of FII levels resulted in an impressive reduction in inflammation (reduction in leukocytosis, thrombocytosis, and circulating interleukin-6 levels), reduced endothelial cell dysfunction (reduced endothelial activation and circulating soluble vascular cell adhesion molecule), and a significant improvement in SCD-associated end-organ damage (nephropathy, pulmonary hypertension, pulmonary inflammation, liver function, inflammatory infiltration, and microinfarctions). Notably, all of these benefits were achieved with a relatively modest 1.25-fold increase in prothrombin times, and in the absence of hemorrhagic complications. Taken together, these data establish that prothrombin is a powerful modifier of SCD-induced end-organ damage, and present a novel therapeutic target to ameliorate SCD pathologies.


Subject(s)
Anemia, Sickle Cell/complications , Genetic Therapy , Hypertension, Pulmonary/prevention & control , Inflammation/prevention & control , Prothrombin/physiology , Vascular Diseases/prevention & control , Anemia, Sickle Cell/mortality , Anemia, Sickle Cell/physiopathology , Animals , Blood Coagulation , Cells, Cultured , Hypertension, Pulmonary/etiology , Immunoenzyme Techniques , Inflammation/etiology , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Oligoribonucleotides, Antisense/pharmacology , Prothrombin/antagonists & inhibitors , Survival Rate , Thrombin/metabolism , Vascular Diseases/etiology
20.
Pediatr Blood Cancer ; 62(10): 1709-16, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25907661

ABSTRACT

BACKGROUND: Neurofibromatosis type 1 (NF1) is a genetic disorder that predisposes affected individuals to formation of benign neurofibromas, peripheral nerve tumors that can be associated with significant morbidity. Loss of the NF1 Ras-GAP protein causes increased Ras-GTP, and we previously found that inhibiting MEK signaling downstream of Ras can shrink established neurofibromas in a genetically engineered murine model. PROCEDURES: We studied effects of MEK inhibition using 1.5 mg/kg/day PD-0325901 prior to neurofibroma onset in the Nf1 (flox/flox); Dhh-Cre mouse model. We also treated mice with established tumors at 0.5 and 1.5 mg/kg/day doses of PD-0325901. We monitored tumor volumes using MRI and volumetric measurements, and measured pharmacokinetic and pharmacodynamic endpoints. RESULTS: Early administration significantly delayed neurofibroma development as compared to vehicle controls. When treatment was discontinued neurofibromas grew, but no rebound effect was observed and neurofibromas remained significantly smaller than controls. Low dose treatment of mice with PD-0325901 resulted in neurofibroma shrinkage equivalent to that observed at higher doses. Tumor cell proliferation decreased, although less than at higher doses with drug. Tumor blood vessels per area correlated with tumor shrinkage. CONCLUSIONS: Neurofibroma development was not prevented by MEK inhibition, beginning at 1 month of age, but tumor size was controlled by early treatment. Moreover, treatment with PD-0325901 at very low doses may shrink neurofibromas while minimizing toxicity. These studies highlight how genetically engineered mouse models can guide clinical trial design.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Enzyme Inhibitors/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Neurofibromatosis 1/pathology , Animals , Diphenylamine/pharmacology , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Electron
SELECTION OF CITATIONS
SEARCH DETAIL
...