Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Clin Cancer Res ; 30(5): 1038-1053, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38127282

ABSTRACT

PURPOSE: Plexiform neurofibromas (PNF) are benign peripheral nerve sheath tumors (PNST) associated with neurofibromatosis type 1 (NF1). Despite similar histologic appearance, these neoplasms exhibit diverse evolutionary trajectories, with a subset progressing to malignant peripheral nerve sheath tumor (MPNST), the leading cause of premature death in individuals with NF1. Malignant transformation of PNF often occurs through the development of atypical neurofibroma (ANF) precursor lesions characterized by distinct histopathologic features and CDKN2A copy-number loss. Although genomic studies have uncovered key driver events promoting tumor progression, the transcriptional changes preceding malignant transformation remain poorly defined. EXPERIMENTAL DESIGN: Here we resolve gene-expression profiles in PNST across the neurofibroma-to-MPNST continuum in NF1 patients and mouse models, revealing early molecular features associated with neurofibroma evolution and transformation. RESULTS: Our findings demonstrate that ANF exhibit enhanced signatures of antigen presentation and immune response, which are suppressed as malignant transformation ensues. MPNST further displayed deregulated survival and mitotic fidelity pathways, and targeting key mediators of these pathways, CENPF and BIRC5, disrupted the growth and viability of human MPNST cell lines and primary murine Nf1-Cdkn2a-mutant Schwann cell precursors. Finally, neurofibromas contiguous with MPNST manifested distinct alterations in core oncogenic and immune surveillance programs, suggesting that early molecular events driving disease progression may precede histopathologic evidence of malignancy. CONCLUSIONS: If validated prospectively in future studies, these signatures may serve as molecular diagnostic tools to augment conventional histopathologic diagnosis by identifying neurofibromas at high risk of undergoing malignant transformation, facilitating risk-adapted care.


Subject(s)
Nerve Sheath Neoplasms , Neurofibroma , Neurofibromatosis 1 , Neurofibrosarcoma , Animals , Humans , Mice , Gene Expression Profiling , Nerve Sheath Neoplasms/genetics , Neurofibroma/genetics , Neurofibromatosis 1/genetics , Neurofibrosarcoma/genetics
3.
Clin Cancer Res ; 29(17): 3438-3456, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37406085

ABSTRACT

PURPOSE: Plexiform neurofibromas (PNF) are peripheral nerve sheath tumors that cause significant morbidity in persons with neurofibromatosis type 1 (NF1), yet treatment options remain limited. To identify novel therapeutic targets for PNF, we applied an integrated multi-omic approach to quantitatively profile kinome enrichment in a mouse model that has predicted therapeutic responses in clinical trials for NF1-associated PNF with high fidelity. EXPERIMENTAL DESIGN: Utilizing RNA sequencing combined with chemical proteomic profiling of the functionally enriched kinome using multiplexed inhibitor beads coupled with mass spectrometry, we identified molecular signatures predictive of response to CDK4/6 and RAS/MAPK pathway inhibition in PNF. Informed by these results, we evaluated the efficacy of the CDK4/6 inhibitor, abemaciclib, and the ERK1/2 inhibitor, LY3214996, alone and in combination in reducing PNF tumor burden in Nf1flox/flox;PostnCre mice. RESULTS: Converging signatures of CDK4/6 and RAS/MAPK pathway activation were identified within the transcriptome and kinome that were conserved in both murine and human PNF. We observed robust additivity of the CDK4/6 inhibitor, abemaciclib, in combination with the ERK1/2 inhibitor, LY3214996, in murine and human NF1(Nf1) mutant Schwann cells. Consistent with these findings, the combination of abemaciclib (CDK4/6i) and LY3214996 (ERK1/2i) synergized to suppress molecular signatures of MAPK activation and exhibited enhanced antitumor activity in Nf1flox/flox;PostnCre mice in vivo. CONCLUSIONS: These findings provide rationale for the clinical translation of CDK4/6 inhibitors alone and in combination with therapies targeting the RAS/MAPK pathway for the treatment of PNF and other peripheral nerve sheath tumors in persons with NF1.


Subject(s)
Nerve Sheath Neoplasms , Neurofibroma, Plexiform , Neurofibroma , Neurofibromatosis 1 , Humans , Mice , Animals , Neurofibroma, Plexiform/etiology , Neurofibroma, Plexiform/genetics , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics , MAP Kinase Signaling System , Proteomics , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Neurofibroma/complications , Cyclin-Dependent Kinase 4/genetics
4.
FEBS J ; 290(12): 3059-3075, 2023 06.
Article in English | MEDLINE | ID: mdl-35305060

ABSTRACT

Hedgehog signalling is essential for development, crucial for normal anatomical arrangement and activated during tissue damage repair. Dysregulation of hedgehog signalling is associated with cancer, developmental disorders and other diseases including osteoarthritis (OA). The hedgehog gene was first discovered in Drosophila melanogaster, and the pathway is evolutionarily conserved in most animals. Although there are several hedgehog ligands with different protein expression patterns, they share a common plasma membrane receptor, Patched1 and hedgehog signalling pathway activation is transduced through the G-protein-coupled receptor-like protein Smoothened (SMO) and downstream effectors. Functional assays revealed that activation of SMO is dependent on sterol binding, and cholesterol was observed bound to SMO in crystallography experiments. In vertebrates, hedgehog signalling coordinates endochondral ossification and balances osteoblast and osteoclast activation to maintain homeostasis. A recently discovered mutation of SMO in humans (SMOR173C ) is predicted to alter cholesterol binding and is associated with a higher risk of hip OA. Functional studies in mice and human tissue analysis provide evidence that hedgehog signalling is pathologically activated in chondrocytes of osteoarthritic cartilage.


Subject(s)
Drosophila Proteins , Osteoarthritis , Animals , Humans , Mice , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Cholesterol , Osteoarthritis/genetics
6.
PLoS One ; 16(7): e0252048, 2021.
Article in English | MEDLINE | ID: mdl-34264955

ABSTRACT

Neurofibromatosis Type 2 (NF2) is an autosomal dominant genetic syndrome caused by mutations in the NF2 tumor suppressor gene resulting in multiple schwannomas and meningiomas. There are no FDA approved therapies for these tumors and their relentless progression results in high rates of morbidity and mortality. Through a combination of high throughput screens, preclinical in vivo modeling, and evaluation of the kinome en masse, we identified actionable drug targets and efficacious experimental therapeutics for the treatment of NF2 related schwannomas and meningiomas. These efforts identified brigatinib (ALUNBRIG®), an FDA-approved inhibitor of multiple tyrosine kinases including ALK, to be a potent inhibitor of tumor growth in established NF2 deficient xenograft meningiomas and a genetically engineered murine model of spontaneous NF2 schwannomas. Surprisingly, neither meningioma nor schwannoma cells express ALK. Instead, we demonstrate that brigatinib inhibited multiple tyrosine kinases, including EphA2, Fer and focal adhesion kinase 1 (FAK1). These data demonstrate the power of the de novo unbiased approach for drug discovery and represents a major step forward in the advancement of therapeutics for the treatment of NF2 related malignancies.


Subject(s)
Meningeal Neoplasms/genetics , Meningioma/genetics , Neurilemmoma/genetics , Neurofibromin 2/deficiency , Neurofibromin 2/genetics , Organophosphorus Compounds/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Cell Proliferation , Humans , Mutation , Neurilemmoma/pathology
7.
Hum Mol Genet ; 30(17): 1607-1617, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34075397

ABSTRACT

Neurofibromatosis Type II (NF2) is an autosomal dominant cancer predisposition syndrome in which germline haploinsufficiency at the NF2 gene confers a greatly increased propensity for tumor development arising from tissues of neural crest derived origin. NF2 encodes the tumor suppressor, Merlin, and its biochemical function is incompletely understood. One well-established function of Merlin is as a negative regulator of group A serine/threonine p21-activated kinases (PAKs). In these studies we explore the role of PAK1 and its closely related paralog, PAK2, both pharmacologically and genetically, in Merlin-deficient Schwann cells and in a genetically engineered mouse model (GEMM) that develops spontaneous vestibular and spinal schwannomas. We demonstrate that PAK1 and PAK2 are both hyper activated in Merlin-deficient murine schwannomas. In preclinical trials, a pan Group A PAK inhibitor, FRAX-1036, transiently reduced PAK1 and PAK2 phosphorylation in vitro, but had insignificant efficacy in vivo. NVS-PAK1-1, a PAK1 selective inhibitor, had a greater but still minimal effect on our GEMM phenotype. However, genetic ablation of Pak1 but not Pak2 reduced tumor formation in our NF2 GEMM. Moreover, germline genetic deletion of Pak1 was well tolerated, while conditional deletion of Pak2 in Schwann cells resulted in significant morbidity and mortality. These data support the further development of PAK1-specific small molecule inhibitors and the therapeutic targeting of PAK1 in vestibular schwannomas and argue against PAK1 and PAK2 existing as functionally redundant protein isoforms in Schwann cells.


Subject(s)
Neurofibromatosis 2/genetics , p21-Activated Kinases/metabolism , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Genes, Tumor Suppressor/drug effects , Indoles , Longevity , Mice , Neurilemmoma/genetics , Neurofibromatosis 2/metabolism , Neurofibromin 2/genetics , Phosphorylation , Piperidines , Pyrimidines , Schwann Cells/metabolism , p21-Activated Kinases/genetics
8.
Neurooncol Adv ; 3(1): vdab063, 2021.
Article in English | MEDLINE | ID: mdl-34131650

ABSTRACT

BACKGROUND: Genetically susceptible individuals can develop malignancies after irradiation of normal tissues. In the context of therapeutic irradiation, it is not known whether irradiating benign neoplasms in susceptible individuals promotes neoplastic transformation and worse clinical outcomes. Individuals with Neurofibromatosis 1 (NF1) are susceptible to both radiation-induced second malignancies and spontaneous progression of plexiform neurofibromas (PNs) to malignant peripheral nerve sheath tumors (MPNSTs). The role of radiotherapy in the treatment of benign neoplasms such as PNs is unclear. METHODS: To test whether radiotherapy promotes neoplastic progression of PNs and reduces overall survival, we administered spinal irradiation (SI) to conditional knockout mouse models of NF1-associated PNs in 2 germline contexts: Nf1 fllfl ; PostnCre + and Nf1 fl/- ; PostnCre + . Both genotypes develop extensive Nf1 null spinal PNs, modeling PNs in NF1 patients. A total of 101 mice were randomized to 0 Gy, 15 Gy (3 Gy × 5), or 30 Gy (3 Gy × 10) of spine-focused, fractionated SI and aged until signs of illness. RESULTS: SI decreased survival in both Nf1 fllfl mice and Nf1 fl/- mice, with the worst overall survival occurring in Nf1 fl/- mice receiving 30 Gy. SI was also associated with increasing worrisome histologic features along the PN-MPNST continuum in PNs irradiated to higher radiation doses. CONCLUSIONS: This preclinical study provides experimental evidence that irradiation of pre-existing PNs reduces survival and may shift PNs to higher grade neoplasms.

9.
Br J Cancer ; 124(9): 1566-1580, 2021 04.
Article in English | MEDLINE | ID: mdl-33658640

ABSTRACT

BACKGROUND: MPNST is a rare soft-tissue sarcoma that can arise from patients with NF1. Existing chemotherapeutic and targeted agents have been unsuccessful in MPNST treatment, and recent findings implicate STAT3 and HIF1-α in driving MPNST. The DNA-binding and transcriptional activity of both STAT3 and HIF1-α is regulated by Redox factor-1 (Ref-1) redox function. A first-generation Ref-1 inhibitor, APX3330, is being tested in cancer clinical trials and could be applied to MPNST. METHODS: We characterised Ref-1 and p-STAT3 expression in various MPNST models. Tumour growth, as well as biomarkers of apoptosis and signalling pathways, were measured by qPCR and western blot following treatment with inhibitors of Ref-1 or STAT3. RESULTS: MPNSTs from Nf1-Arfflox/floxPostnCre mice exhibit significantly increased positivity of p-STAT3 and Ref-1 expression when malignant transformation occurs. Inhibition of Ref-1 or STAT3 impairs MPNST growth in vitro and in vivo and induces apoptosis. Genes highly expressed in MPNST patients are downregulated following inhibition of Ref-1 or STAT3. Several biomarkers downstream of Ref-1 or STAT3 were also downregulated following Ref-1 or STAT3 inhibition. CONCLUSIONS: Our findings implicate a unique therapeutic approach to target important MPNST signalling nodes in sarcomas using new first-in-class small molecules for potential translation to the clinic.


Subject(s)
Biomarkers, Tumor/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Gene Expression Regulation, Neoplastic , Neurofibrosarcoma/pathology , STAT3 Transcription Factor/metabolism , Adolescent , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neurofibrosarcoma/genetics , Neurofibrosarcoma/metabolism , Prognosis , STAT3 Transcription Factor/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Am J Health Syst Pharm ; 78(9): 800-805, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33599751

ABSTRACT

PURPOSE: The proceedings of an international summit on the current and desired future state of use of robotic systems to compound intravenous (IV) solutions are summarized. SUMMARY: The International IV Robotics Summit was held at the Cleveland Clinic main campus in Cleveland, OH, on April 29 and 30, 2019. The purpose of the summit was 2-fold: (1) to define the current state of robotic IV compounding and (2) to develop a guide for automation companies, pharmacy departments, and drug manufacturers to improve the technology and expand the use of IV robotics in health systems in the future. The first day of the summit included 45-minute presentations by each of the speakers. Each lecturer recounted a different hospital's experience implementing and using IV robotics. On day 2 of the summit, an expert panel dedicated to mapping the future of IV robotics was convened to determine barriers to widespread adoption of IV robotics in health systems and offer potential solutions to remove these barriers. The expert panel targeted 3 specific audiences: robot manufacturers, drug manufacturers, and fellow pharmacy leaders. CONCLUSION: It is the hope of the international faculty that the information that emerged from the summit can be used by others to successfully implement IV compounding robotics in their sterile products areas to maximize patient safety. The summit also served as a call to action for pharmacy leaders, drug manufacturers, and robotic companies to develop a safer, more efficient future for patients by working together to optimize the development and operation of IV robotics.


Subject(s)
Robotics , Automation , Humans
11.
Nat Med ; 27(1): 165-173, 2021 01.
Article in English | MEDLINE | ID: mdl-33442015

ABSTRACT

Neurofibromatosis type 1 (NF1) plexiform neurofibromas (PNs) are progressive, multicellular neoplasms that cause morbidity and may transform to sarcoma. Treatment of Nf1fl/fl;Postn-Cre mice with cabozantinib, an inhibitor of multiple tyrosine kinases, caused a reduction in PN size and number and differential modulation of kinases in cell lineages that drive PN growth. Based on these findings, the Neurofibromatosis Clinical Trials Consortium conducted a phase II, open-label, nonrandomized Simon two-stage study to assess the safety, efficacy and biologic activity of cabozantinib in patients ≥16 years of age with NF1 and progressive or symptomatic, inoperable PN ( NCT02101736 ). The trial met its primary outcome, defined as ≥25% of patients achieving a partial response (PR, defined as ≥20% reduction in target lesion volume as assessed by magnetic resonance imaging (MRI)) after 12 cycles of therapy. Secondary outcomes included adverse events (AEs), patient-reported outcomes (PROs) assessing pain and quality of life (QOL), pharmacokinetics (PK) and the levels of circulating endothelial cells and cytokines. Eight of 19 evaluable (42%) trial participants achieved a PR. The median change in tumor volume was 15.2% (range, +2.2% to -36.9%), and no patients had disease progression while on treatment. Nine patients required dose reduction or discontinuation of therapy due to AEs; common AEs included gastrointestinal toxicity, hypothyroidism, fatigue and palmar plantar erythrodysesthesia. A total of 11 grade 3 AEs occurred in eight patients. Patients with PR had a significant reduction in tumor pain intensity and pain interference in daily life but no change in global QOL scores. These data indicate that cabozantinib is active in NF1-associated PN, resulting in tumor volume reduction and pain improvement.


Subject(s)
Anilides/therapeutic use , Neurofibroma, Plexiform/drug therapy , Neurofibromatosis 1/drug therapy , Pyridines/therapeutic use , Adolescent , Adult , Anilides/adverse effects , Anilides/pharmacokinetics , Animals , Disease Models, Animal , Female , Genes, Neurofibromatosis 1 , Humans , Male , Mice , Mice, Mutant Strains , Neurofibroma, Plexiform/genetics , Neurofibroma, Plexiform/pathology , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Pain Measurement , Prospective Studies , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Pyridines/adverse effects , Pyridines/pharmacokinetics , Quality of Life , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Translational Research, Biomedical , Young Adult
13.
J Biol Chem ; 295(29): 9948-9958, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32471868

ABSTRACT

Neurofibromatosis type 1 (NF1) is a common cancer predisposition syndrome caused by mutations in the NF1 tumor suppressor gene. NF1 encodes neurofibromin, a GTPase-activating protein for RAS proto-oncogene GTPase (RAS). Plexiform neurofibromas are a hallmark of NF1 and result from loss of heterozygosity of NF1 in Schwann cells, leading to constitutively activated p21RAS. Given the inability to target p21RAS directly, here we performed an shRNA library screen of all human kinases and Rho-GTPases in a patient-derived NF1-/- Schwann cell line to identify novel therapeutic targets to disrupt PN formation and progression. Rho family members, including Rac family small GTPase 1 (RAC1), were identified as candidates. Corroborating these findings, we observed that shRNA-mediated knockdown of RAC1 reduces cell proliferation and phosphorylation of extracellular signal-regulated kinase (ERK) in NF1-/- Schwann cells. Genetically engineered Nf1flox/flox;PostnCre+ mice, which develop multiple PNs, also exhibited increased RAC1-GTP and phospho-ERK levels compared with Nf1flox/flox;PostnCre- littermates. Notably, mice in which both Nf1 and Rac1 loci were disrupted (Nf1flox/floxRac1flox/flox;PostnCre+) were completely free of tumors and had normal phospho-ERK activity compared with Nf1flox/flox ;PostnCre+ mice. We conclude that the RAC1-GTPase is a key downstream node of RAS and that genetic disruption of the Rac1 allele completely prevents PN tumor formation in vivo in mice.


Subject(s)
Gene Knockdown Techniques , Neoplasms, Second Primary , Neurofibroma, Plexiform , Neurofibromatosis 1 , Neuropeptides/deficiency , rac1 GTP-Binding Protein/deficiency , Animals , Mice , Mice, Knockout , Neoplasms, Second Primary/enzymology , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/pathology , Neoplasms, Second Primary/prevention & control , Neurofibroma, Plexiform/enzymology , Neurofibroma, Plexiform/genetics , Neurofibroma, Plexiform/prevention & control , Neurofibromatosis 1/enzymology , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibromin 1/deficiency , Neurofibromin 1/metabolism , Neuropeptides/metabolism , Proto-Oncogene Mas , rac1 GTP-Binding Protein/metabolism
14.
Pediatr Blood Cancer ; 67(8): e28372, 2020 08.
Article in English | MEDLINE | ID: mdl-32459399

ABSTRACT

BACKGROUND: Neurofibromatosis type 1 (NF1) is a common genetic disorder characterized by plexiform neurofibromas (pNF), which are thought to be congenital tumors that arise in utero and enlarge throughout life. Genetic studies in murine models delineated an indispensable role for the stem cell factor (SCF)/c-kit pathway in pNF initiation and progression. A subsequent phase 2 clinical trial using imatinib mesylate to inhibit SCF/c-kit demonstrated tumor shrinkage in a subset of preexisting pNF; however, imatinib's role on preventing pNF development has yet to be explored. PROCEDURE: We evaluated the effect of imatinib dosed at 10-100 mg/kg/day for 12 weeks to one-month-old Nf1flox/flox ;PostnCre(+) mice, prior to onset of pNF formation. To determine durability of response, we then monitored for pNF growth at later time points, comparing imatinib- with vehicle-treated mice. We assessed gross and histopathological analysis of tumor burden. RESULTS: Imatinib administered preventatively led to a significant decrease in pNF number, even at doses as low as 10 mg/kg/day. Tumor development continued to be significantly inhibited after cessation of imatinib dosed at 50 and 100 mg/kg/day. In the cohort of treated mice that underwent prolonged follow-up, the size of residual tumors was significantly reduced as compared with age-matched littermates that received vehicle control. CONCLUSIONS: Early administration of imatinib inhibits pNF genesis in vivo, and effects are sustained after discontinuation of therapy. These findings may guide clinical use of imatinib in young NF1 patients prior to the substantial development of pNF.


Subject(s)
Imatinib Mesylate/administration & dosage , Neoplasms, Experimental/prevention & control , Neurofibroma, Plexiform/prevention & control , Neurofibromatosis 1/prevention & control , Animals , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neurofibroma, Plexiform/genetics , Neurofibroma, Plexiform/metabolism , Neurofibroma, Plexiform/pathology , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/pathology
15.
Mol Cancer Ther ; 18(12): 2321-2330, 2019 12.
Article in English | MEDLINE | ID: mdl-31527226

ABSTRACT

Neurofibromatosis Type 1 (NF1) is one of the most common genetic tumor predisposition syndromes in humans. Mutant NF1 results in dysregulated RAS allowing neoplasms throughout the neuroaxis. Plexiform neurofibromas (pNF) afflict up to 50% of patients with NF1. They are complex tumors of the peripheral nerve that cause major morbidity via nerve dysregulation and mortality via conversion to malignant sarcoma. Genetically engineered mouse models (GEMM) of NF1 provide valuable insights for the identification of therapies that have utility in people with pNF. Preclinical studies in GEMMs implicate mast cells and the c-Kit/Kit ligand pathway in pNF tumorigenesis. Kit ligand is a potent chemokine secreted by tumorigenic, Nf1-deficient Schwann cells. Ketotifen is an FDA-approved drug for the treatment of allergic conjunctivitis and asthma that promotes mast cell stabilization and has been used in prior case studies to treat or prevent pNFs. This study investigated the effect of ketotifen on mast cell infiltration and degranulation in the presence and absence of Kit ligand provocation and the effect of ketotifen on shrinking or preventing pNF formation in the Nf1flox/flox ;PostnCre + GEMM. Ketotifen decreased mast cell infiltration in response to exogenous Kit ligand administration, but did not affect mast cell degranulation. Importantly, ketotifen did not reduce mast cells numbers or activity in pNF and did not prevent pNF formation or decrease the volume of established pNF despite administration of pharmacologically active doses. These findings suggest that ketotifen has limited use as monotherapy to prevent or reduce pNF burden in the setting of Nf1 mutations.


Subject(s)
Chemotaxis/physiology , Histamine H1 Antagonists/therapeutic use , Ketotifen/therapeutic use , Mast Cells/drug effects , Neurofibroma/genetics , Neurofibromin 1/deficiency , Animals , Histamine H1 Antagonists/pharmacology , Ketotifen/pharmacology , Mice , Stem Cell Factor
16.
Hum Mol Genet ; 28(16): 2752-2762, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31091306

ABSTRACT

Plexiform neurofibroma (PN) tumors are a hallmark manifestation of neurofibromatosis type 1 (NF1) that arise in the Schwann cell (SC) lineage. NF1 is a common heritable cancer predisposition syndrome caused by germline mutations in the NF1 tumor suppressor, which encodes a GTPase-activating protein called neurofibromin that negatively regulates Ras proteins. Whereas most PN are clinically indolent, a subset progress to atypical neurofibromatous neoplasms of uncertain biologic potential (ANNUBP) and/or to malignant peripheral nerve sheath tumors (MPNSTs). In small clinical series, loss of 9p21.3, which includes the CDKN2A locus, has been associated with the genesis of ANNUBP. Here we show that the Cdkn2a alternate reading frame (Arf) serves as a gatekeeper tumor suppressor in mice that prevents PN progression by inducing senescence-mediated growth arrest in aberrantly proliferating Nf1-/- SC. Conditional ablation of Nf1 and Arf in the neural crest-derived SC lineage allows escape from senescence, resulting in tumors that accurately phenocopy human ANNUBP and progress to MPNST with high penetrance. This animal model will serve as a platform to study the clonal development of ANNUBP and MPNST and to identify new therapies to treat existing tumors and to prevent disease progression.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Neurofibroma/genetics , Neurofibroma/pathology , Neurofibromatosis 1/genetics , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/genetics , Disease Models, Animal , Disease Progression , Genotype , Heterografts , Humans , Immunohistochemistry , Mice , Mutation , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Neurofibroma/metabolism , Neurofibroma/mortality , Neurofibromatosis 1/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , ras Proteins/metabolism
17.
Hum Mol Genet ; 28(4): 572-583, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30335132

ABSTRACT

Schwannomas are common, highly morbid and medically untreatable tumors that can arise in patients with germ line as well as somatic mutations in neurofibromatosis type 2 (NF2). These mutations most commonly result in the loss of function of the NF2-encoded protein, Merlin. Little is known about how Merlin functions endogenously as a tumor suppressor and how its loss leads to oncogenic transformation in Schwann cells (SCs). Here, we identify nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)-inducing kinase (NIK) as a potential drug target driving NF-κB signaling and Merlin-deficient schwannoma genesis. Using a genomic approach to profile aberrant tumor signaling pathways, we describe multiple upregulated NF-κB signaling elements in human and murine schwannomas, leading us to identify a caspase-cleaved, proteasome-resistant NIK kinase domain fragment that amplifies pathogenic NF-κB signaling. Lentiviral-mediated transduction of this NIK fragment into normal SCs promotes proliferation, survival, and adhesion while inducing schwannoma formation in a novel in vivo orthotopic transplant model. Furthermore, we describe an NF-κB-potentiated hepatocyte growth factor (HGF) to MET proto-oncogene receptor tyrosine kinase (c-Met) autocrine feed-forward loop promoting SC proliferation. These innovative studies identify a novel signaling axis underlying schwannoma formation, revealing new and potentially druggable schwannoma vulnerabilities with future therapeutic potential.


Subject(s)
Neurilemmoma/genetics , Neurofibromatosis 2/genetics , Neurofibromin 2/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Autocrine Communication/genetics , Carcinogenesis/genetics , Caspase 1/genetics , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Hepatocyte Growth Factor/genetics , Humans , Mice , Molecular Targeted Therapy , NF-kappa B/genetics , Neurilemmoma/complications , Neurilemmoma/drug therapy , Neurilemmoma/pathology , Neurofibromatosis 2/complications , Neurofibromatosis 2/drug therapy , Neurofibromatosis 2/pathology , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/genetics , Schwann Cells , Signal Transduction/genetics , NF-kappaB-Inducing Kinase
18.
Oncotarget ; 9(1): 718-725, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29416648

ABSTRACT

Mutations in the tumor suppressor gene NF2 lead to Neurofibromatosis type 2 (NF2), a tumor predisposition syndrome characterized by the development of schwannomas, including bilateral vestibular schwannomas with complete penetrance. Recent work has implicated the importance of COX-2 in schwannoma growth. Using a genetically engineered murine model of NF2, we demonstrate that selective inhibition of COX-2 with celecoxib fails to prevent the spontaneous development of schwannomas or sensorineural hearing loss in vivo, despite elevated expression levels of COX-2 in Nf2-deficient tumor tissue. These results suggest that COX-2 is nonessential to schwannomagenesis and that the proposed tumor suppressive effects of NSAIDs on schwannomas may occur through COX-2 independent mechanisms.

19.
Mol Ecol ; 20(2): 299-310, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21091561

ABSTRACT

The expression of cone opsin genes is a primary determinant of the characteristics of colour vision. Interspecific variation in opsin expression is common in African cichlids. It is correlated with foraging among cichlids from Lake Malawi, and with ambient light environment among cichlids from Lake Victoria. In this study, we tested whether gene expression varied within species such that it might be important in contributing to divergence. We hypothesized that light attenuation with depth would be correlated with predictable changes in gene expression in Lake Malawi, and that this variation would tune visual sensitivities to match the ambient light environment. We observed significant differences in cone opsin expression in three different comparisons among populations of the same species. Higher LWS expression was found in shallow versus deep Copadichromis eucinostomus. In Metriaclima zebra, individuals from Zimbawe Rock expressed significantly more SWS2B than those from Thumbi West Island, although these locales have similar ambient light environments. Finally, Tropheops gracilior from deeper water had significantly more variation in expression than their shallow counterparts. These results support that gene expression varies significantly between populations of the same species. Surprisingly, these results could not be explained by predicted visual performance as models predicted that differential expression patterns did not confer sensitivity advantages at different depths. This suggested that expression variation did not confer a local sensitivity advantage. Therefore, our findings were contrary to a primary requirement of the sensory bias hypothesis. As such, other explanations for intraspecific gene expression variation need to be tested.


Subject(s)
Cichlids/genetics , Cone Opsins/genetics , Gene Expression , Retinal Cone Photoreceptor Cells/metabolism , Visual Acuity/genetics , Africa , Animals , Cichlids/physiology , Color Vision , Evolution, Molecular , Fresh Water , Genetic Variation , Light , Phylogeny , Polymerase Chain Reaction , Sequence Analysis, DNA , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...