Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38617277

ABSTRACT

Optineurin (OPTN) mutations are linked to amyotrophic lateral sclerosis (ALS) and normal tension glaucoma (NTG), but a relevant animal model is lacking, and the molecular mechanisms underlying neurodegeneration are unknown. We found that OPTN C-terminus truncation (OPTN∆C) causes late-onset neurodegeneration of retinal ganglion cells (RGCs), optic nerve (ON), and spinal cord motor neurons, preceded by a striking decrease of axonal mitochondria. Surprisingly, we discover that OPTN directly interacts with both microtubules and the mitochondrial transport complex TRAK1/KIF5B, stabilizing them for proper anterograde axonal mitochondrial transport, in a C-terminus dependent manner. Encouragingly, overexpressing OPTN/TRAK1/KIF5B reverses not only OPTN truncation-induced, but also ocular hypertension-induced neurodegeneration, and promotes striking ON regeneration. Therefore, in addition to generating new animal models for NTG and ALS, our results establish OPTN as a novel facilitator of the microtubule-dependent mitochondrial transport necessary for adequate axonal mitochondria delivery, and its loss as the likely molecular mechanism of neurodegeneration.

2.
Sci Rep ; 11(1): 9052, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33907301

ABSTRACT

Recently, we established silicone oil-induced ocular hypertension (SOHU) mouse model with significant glaucomatous neurodegeneration. Here we characterize two additional variations of this model that simulate two distinct glaucoma types. The first is a chronic model produced by high frequency (HF) pupillary dilation after SO-induced pupillary block, which shows sustained moderate IOP elevation and corresponding slow, mild glaucomatous neurodegeneration. We also demonstrate that although SO removal quickly returns IOP to normal, the glaucomatous neurodegeneration continues to advance to a similar degree as in the HF group without SO removal. The second, an acute model created by no pupillary dilation (ND), shows a greatly elevated IOP and severe inner retina degeneration at an early time point. Therefore, by a straightforward dilation scheme, we extend our original SOHU model to recapitulate phenotypes of two major glaucoma forms, which will be invaluable for selecting neuroprotectants and elucidating their molecular mechanisms.


Subject(s)
Disease Models, Animal , Glaucoma/pathology , Ocular Hypertension/physiopathology , Retinal Degeneration/pathology , Retinal Ganglion Cells/pathology , Silicone Oils/toxicity , Acute Disease , Animals , Female , Glaucoma/chemically induced , Intraocular Pressure , Male , Mice , Mice, Inbred C57BL , Ocular Hypertension/chemically induced , Retinal Degeneration/chemically induced , Retinal Ganglion Cells/drug effects , Silicone Oils/administration & dosage
3.
Brain Struct Funct ; 226(4): 951-961, 2021 May.
Article in English | MEDLINE | ID: mdl-33710409

ABSTRACT

The impressive functions of the brain rely on an extensive connectivity matrix between specific neurons, the architecture of which is frequently characterized by one brain nucleus/region connecting to multiple targets, either via collaterals of the same projection neuron or several, differentially specified neurons. Delineating the fine architecture of projection neuron subsets in a specific brain region could greatly facilitate its circuit, computational, and functional resolution. Here, we developed multiple fluorescent rabies viruses (RV) to delineate the fine organization of corticothalamic projection neuron subsets in the primary visual cortex (V1). By simultaneously retrograde labeling multiple distinct subsets of corticothalamic projection neurons in V1 from their target nuclei in thalamus (dLGN, LP, LD), we observed that V1-dLGN corticothalamic projection neurons were densely concentrated in layer VI, except for several sparsely scattered neurons in layer V, while V1-LP and V1-LD corticothalamic projection neurons were localized to both layers V and VI. Meanwhile, we observed a fraction of V1 corticothalamic projection neurons targeting two thalamic nuclei, which was further confirmed by fMOST whole-brain imaging. The multiple fluorescent RV tracing tools can be extensively applied to resolve the architecture of projection neuron subsets in certain brain regions, with a strong potential to delineate the computational and functional organization of these brain regions.


Subject(s)
Rabies virus , Visual Cortex , Interneurons , Rabies , Thalamus/diagnostic imaging
4.
Am J Pathol ; 191(6): 1020-1035, 2021 06.
Article in English | MEDLINE | ID: mdl-33705750

ABSTRACT

Glucocorticoid-induced glaucoma is a secondary open-angle glaucoma. About 40% of the general population may develop elevated intraocular pressure on prolonged glucocorticoid treatment secondary to damages in the trabecular meshwork (TM), a tissue that regulates intraocular pressure. Therefore, identifying the key molecules responsible for glucocorticoid-induced ocular hypertension is crucial. In this study, Dickkopf-related protein 1 (Dkk1), a canonical Wnt signaling inhibitor, was found to be elevated in the aqueous humor and TM of glaucoma patients. At the signaling level, Dkk1 enhanced glucocorticoid receptor (GR) signaling, whereas Dkk1 knockdown or Wnt signaling activators decreased GR signaling in human TM cells as indicated by luciferase assays. Similarly, activation of the GR signaling inhibited Wnt signaling. At the protein level, glucocorticoid-induced extracellular matrix was inhibited by Wnt activation using Wnt activators or Dkk1 knockdown in primary human TM cells. In contrast, inhibition of canonical Wnt signaling by ß-catenin knockdown increased glucocorticoid-induced extracellular matrix proteins. At the physiological level, adenovirus-mediated Wnt3a expression decreased glucocorticoid-induced ocular hypertension in mouse eyes. In summary, Wnt and GR signaling inhibit each other in the TM, and canonical Wnt signaling activators may prevent the adverse effect of glucocorticoids in the eye.


Subject(s)
Glaucoma/metabolism , Receptors, Glucocorticoid/metabolism , Trabecular Meshwork/metabolism , Wnt Signaling Pathway/physiology , Animals , Female , Glaucoma/chemically induced , Glucocorticoids/adverse effects , Humans , Immunosuppressive Agents/adverse effects , Mice , Mice, Inbred C57BL
5.
J Clin Invest ; 130(11): 5875-5892, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33016930

ABSTRACT

The undruggable nature of oncogenic Myc transcription factors poses a therapeutic challenge in neuroblastoma, a pediatric cancer in which MYCN amplification is strongly associated with unfavorable outcome. Here, we show that CYC065 (fadraciclib), a clinical inhibitor of CDK9 and CDK2, selectively targeted MYCN-amplified neuroblastoma via multiple mechanisms. CDK9 - a component of the transcription elongation complex P-TEFb - bound to the MYCN-amplicon superenhancer, and its inhibition resulted in selective loss of nascent MYCN transcription. MYCN loss led to growth arrest, sensitizing cells for apoptosis following CDK2 inhibition. In MYCN-amplified neuroblastoma, MYCN invaded active enhancers, driving a transcriptionally encoded adrenergic gene expression program that was selectively reversed by CYC065. MYCN overexpression in mesenchymal neuroblastoma was sufficient to induce adrenergic identity and sensitize cells to CYC065. CYC065, used together with temozolomide, a reference therapy for relapsed neuroblastoma, caused long-term suppression of neuroblastoma growth in vivo, highlighting the clinical potential of CDK9/2 inhibition in the treatment of MYCN-amplified neuroblastoma.


Subject(s)
Adenosine/analogs & derivatives , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 9/antagonists & inhibitors , N-Myc Proto-Oncogene Protein/biosynthesis , Neuroblastoma/drug therapy , Temozolomide/pharmacology , Adenosine/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 9/metabolism , Enhancer Elements, Genetic , Humans , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , Transcription, Genetic/drug effects
6.
Diabetes ; 69(11): 2481-2489, 2020 11.
Article in English | MEDLINE | ID: mdl-32816962

ABSTRACT

Loss-of-function mutations in both alleles of the human insulin receptor gene (INSR) cause extreme insulin resistance (IR) and usually death in childhood, with few effective therapeutic options. Bivalent antireceptor antibodies can elicit insulin-like signaling by mutant INSR in cultured cells, but whether this translates into meaningful metabolic benefits in vivo, wherein the dynamics of insulin signaling and receptor recycling are more complex, is unknown. To address this, we adopted a strategy to model human insulin receptoropathy in mice, using Cre recombinase delivered by adeno-associated virus to knockout endogenous hepatic Insr acutely in floxed Insr mice (liver insulin receptor knockout [L-IRKO] + GFP), before adenovirus-mediated add back of wild-type (WT) or mutant human INSR Two murine anti-INSR monoclonal antibodies, previously shown to be surrogate agonists for mutant INSR, were then tested by intraperitoneal injections. As expected, L-IRKO + GFP mice showed glucose intolerance and severe hyperinsulinemia. This was fully corrected by add back of WT but not with either D734A or S350L mutant INSR. Antibody injection improved glucose tolerance in D734A INSR-expressing mice and reduced hyperinsulinemia in both S350L and D734A INSR-expressing animals. It did not cause hypoglycemia in WT INSR-expressing mice. Antibody treatment also downregulated both WT and mutant INSR protein, attenuating its beneficial metabolic effects. Anti-INSR antibodies thus improve IR in an acute model of insulin receptoropathy, but these findings imply a narrow therapeutic window determined by competing effects of antibodies to stimulate receptors and induce their downregulation.


Subject(s)
Antibodies , Disease Models, Animal , Hyperglycemia/therapy , Insulin/immunology , Receptor, Insulin/immunology , Receptor, Insulin/metabolism , Animals , Blood Glucose , Gene Expression Regulation , Liver/metabolism , Male , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Receptor, Insulin/genetics
7.
J Neurosci ; 40(20): 3896-3914, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32300046

ABSTRACT

Optic neuropathies are a group of optic nerve (ON) diseases caused by various insults including glaucoma, inflammation, ischemia, trauma, and genetic deficits, which are characterized by retinal ganglion cell (RGC) death and ON degeneration. An increasing number of genes involved in RGC intrinsic signaling have been found to be promising neural repair targets that can potentially be modulated directly by gene therapy, if we can achieve RGC specific gene targeting. To address this challenge, we first used adeno-associated virus (AAV)-mediated gene transfer to perform a low-throughput in vivo screening in both male and female mouse eyes and identified the mouse γ-synuclein (mSncg) promoter, which specifically and potently sustained transgene expression in mouse RGCs and also works in human RGCs. We further demonstrated that gene therapy that combines AAV-mSncg promoter with clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing can knock down pro-degenerative genes in RGCs and provide effective neuroprotection in optic neuropathies.SIGNIFICANCE STATEMENT Here, we present an RGC-specific promoter, mouse γ-synuclein (mSncg) promoter, and perform extensive characterization and proof-of-concept studies of mSncg promoter-mediated gene expression and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing in RGCs in vivo To our knowledge, this is the first report demonstrating in vivo neuroprotection of injured RGCs and optic nerve (ON) by AAV-mediated CRISPR/Cas9 inhibition of genes that are critical for neurodegeneration. It represents a powerful tool to achieve RGC-specific gene modulation, and also opens up a promising gene therapy strategy for optic neuropathies, the most common form of eye diseases that cause irreversible blindness.


Subject(s)
Gene Expression Regulation/genetics , RNA Editing/genetics , Retinal Ganglion Cells/metabolism , gamma-Synuclein/genetics , Animals , CRISPR-Cas Systems , Dependovirus/genetics , Female , Gene Deletion , Genetic Therapy , Humans , Induced Pluripotent Stem Cells , Male , Mice , Mice, Inbred C57BL , Optic Nerve/pathology , Optic Nerve Diseases/genetics , Optic Nerve Diseases/therapy , Retinal Ganglion Cells/pathology , Transgenes/genetics
8.
Biochem Pharmacol ; 172: 113770, 2020 02.
Article in English | MEDLINE | ID: mdl-31862449

ABSTRACT

The antimetabolite 6-mercaptopurine (6-MP) is an important component in the treatment of specific cancer subtypes, however, the development of drug resistance and dose-limiting toxicities can limit its effectiveness. The therapeutic activity of 6-MP requires cellular uptake, enzymatic conversion to thio-GMP and incorporation of thio-GTP into RNA and DNA, as well as inhibition of de novo purine synthesis by methyl-thio-IMP. Mechanisms that prevent 6-MP entry into the cell, prevent 6-MP metabolism or deplete thiopurine intermediates, can all lead to 6-MP resistance. We previously conducted a high-throughput screen for inhibitors of the multidrug transporter MRP4 using 6-MP sensitivity as the readout. In addition to MRP4-specific inhibitors, we identified a compound, CCI52, that sensitized cell lines to 6-MP independent of this transporter. CCI52 and its more stable analogue CCI52-14 also function as effective chemosensitizers in vivo, substantially extending survival in a transgenic mouse cancer model treated with 6-MP. Chemosensitization was associated with an increase in thio-IMP, suggesting that CCI52 functions directly on 6-MP uptake or metabolism. In addition to its chemosensitizing effects, CCI52 and CCI52-14 inhibited the growth of MYCN-amplified high-risk neuroblastoma cell lines and delayed tumor progression in a MYCN-driven, transgenic mouse model of neuroblastoma. These multifunctional inhibitors may be useful for the further development of anticancer agents and as tools to better understand 6-MP metabolism.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Mercaptopurine/administration & dosage , Mercaptopurine/pharmacology , Neuroblastoma/drug therapy , Thiazoles/pharmacology , Animals , Antimetabolites, Antineoplastic/administration & dosage , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Transgenic , Molecular Structure , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neoplasms, Experimental/drug therapy , Neuroblastoma/pathology , Thiazoles/adverse effects , Thiazoles/chemistry
9.
J Vis Exp ; (153)2019 11 15.
Article in English | MEDLINE | ID: mdl-31789319

ABSTRACT

Elevated intraocular pressure (IOP) is a well-documented risk factor for glaucoma. Here we describe a novel, effective method for consistently inducing stable IOP elevation in mice that mimics the post-operative complication of using silicone oil (SO) as a tamponade agent in human vitreoretinal surgery. In this protocol, SO is injected into the anterior chamber of the mouse eye to block the pupil and prevent inflow of aqueous humor. The posterior chamber accumulates aqueous humor and this in turn increases the IOP of the posterior segment. A single SO injection produces reliable, sufficient, and stable IOP elevation, which induces significant glaucomatous neurodegeneration. This model is a true replicate of secondary glaucoma in the eye clinic. To further mimic the clinical setting, SO can be removed from the anterior chamber to reopen the drainage pathway and allow inflow of aqueous humor, which is drained through the trabecular meshwork (TM) at the angle of the anterior chamber. Because IOP quickly returns to normal, the model can be used to test the effect of lowering IOP on glaucomatous retinal ganglion cells. This method is straightforward, does not require special equipment or repeat procedures, closely simulates clinical situations, and may be applicable to diverse animal species. However, minor modifications may be required.


Subject(s)
Disease Models, Animal , Ocular Hypertension/chemically induced , Ocular Hypertension/pathology , Silicon/toxicity , Animals , Aqueous Humor/drug effects , Aqueous Humor/physiology , Injections, Intraocular , Intraocular Pressure/drug effects , Intraocular Pressure/physiology , Male , Mice , Mice, Inbred C57BL , Oils/administration & dosage , Oils/toxicity , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/physiology , Silicon/administration & dosage
10.
Cancer Res ; 79(20): 5382-5393, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31405846

ABSTRACT

Neuroblastoma is a pediatric cancer that is frequently metastatic and resistant to conventional treatment. In part, a lack of natively metastatic, chemoresistant in vivo models has limited our insight into the development of aggressive disease. The Th-MYCN genetically engineered mouse model develops rapidly progressive chemosensitive neuroblastoma and lacks clinically relevant metastases. To study tumor progression in a context more reflective of clinical therapy, we delivered multicycle treatment with cyclophosphamide to Th-MYCN mice, individualizing therapy using MRI, to generate the Th-MYCN CPM32 model. These mice developed chemoresistance and spontaneous bone marrow metastases. Tumors exhibited an altered immune microenvironment with increased stroma and tumor-associated fibroblasts. Analysis of copy number aberrations revealed genomic changes characteristic of human MYCN-amplified neuroblastoma, specifically copy number gains at mouse chromosome 11, syntenic with gains on human chromosome 17q. RNA sequencing revealed enriched expression of genes associated with 17q gain and upregulation of genes associated with high-risk neuroblastoma, such as the cell-cycle regulator cyclin B1-interacting protein 1 (Ccnb1ip1) and thymidine kinase (TK1). The antiapoptotic, prometastatic JAK-STAT3 pathway was activated in chemoresistant tumors, and treatment with the JAK1/JAK2 inhibitor CYT387 reduced progression of chemoresistant tumors and increased survival. Our results highlight that under treatment conditions that mimic chemotherapy in human patients, Th-MYCN mice develop genomic, microenvironmental, and clinical features reminiscent of human chemorefractory disease. The Th-MYCN CPM32 model therefore is a useful tool to dissect in detail mechanisms that drive metastasis and chemoresistance, and highlights dysregulation of signaling pathways such as JAK-STAT3 that could be targeted to improve treatment of aggressive disease. SIGNIFICANCE: An in vivo mouse model of high-risk treatment-resistant neuroblastoma exhibits changes in the tumor microenvironment, widespread metastases, and sensitivity to JAK1/2 inhibition.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Genes, myc , Neoplasm Metastasis/drug therapy , Neuroblastoma/drug therapy , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Benzamides/therapeutic use , Child , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Disease Models, Animal , Disease Progression , Gene Dosage , Gene Expression Regulation, Neoplastic , Humans , Janus Kinases/antagonists & inhibitors , Magnetic Resonance Imaging , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein/genetics , Neoplasm Metastasis/diagnostic imaging , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neuroblastoma/diagnostic imaging , Neuroblastoma/genetics , Neuroblastoma/pathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Signal Transduction , Synteny , Tumor Burden , Tumor Microenvironment
11.
Elife ; 82019 05 15.
Article in English | MEDLINE | ID: mdl-31090540

ABSTRACT

Understanding the molecular mechanism of glaucoma and development of neuroprotectants is significantly hindered by the lack of a reliable animal model that accurately recapitulates human glaucoma. Here, we sought to develop a mouse model for the secondary glaucoma that is often observed in humans after silicone oil (SO) blocks the pupil or migrates into the anterior chamber following vitreoretinal surgery. We observed significant intraocular pressure (IOP) elevation after intracameral injection of SO, and that SO removal allows IOP to return quickly to normal. This simple, inducible and reversible mouse ocular hypertension model shows dynamic changes of visual function that correlate with progressive retinal ganglion cell (RGC) loss and axon degeneration. It may be applicable with only minor modifications to a range of animal species in which it will generate stable, robust IOP elevation and significant neurodegeneration that will facilitate selection of neuroprotectants and investigating the pathogenesis of ocular hypertension-induced glaucoma.


Subject(s)
Disease Models, Animal , Glaucoma/pathology , Neurodegenerative Diseases/pathology , Ocular Hypertension/chemically induced , Ocular Hypertension/complications , Silicone Oils/adverse effects , Animals , Cell Survival/drug effects , Injections, Intraocular , Mice , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Silicone Oils/administration & dosage
13.
Invest Ophthalmol Vis Sci ; 59(3): 1454-1466, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29625468

ABSTRACT

Purpose: Wnt/ß-catenin signaling in the trabecular meshwork (TM) is required for maintaining normal intraocular pressure (IOP), although the mechanism(s) behind this are unknown. We hypothesize that Wnt/ß-catenin signaling regulates IOP via ß-catenin's effects on cadherin junctions. Methods: Nonglaucomatous primary human TM (NTM) cells were treated with or without 100 ng/ml Wnt3a, 1 µg/ml sFRP1, or both for 4 to 48 hours. Cells were immunostained for ß-catenin, total cadherins, or cadherin isoforms. Membrane proteins or whole-cell lysates were isolated for Western immunoblotting and probed for cadherin isoforms. RNA was extracted for cDNA synthesis and qPCR analysis of cadherin expression. Some NTM cells were cultured on electric plates for cell impedance assays. Ad5.CMV recombinant adenoviruses encoding K-cadherin, and/or sFRP1 were injected into eyes of 4- to 6-month-old female BALB/cJ mice (n = 8-10). Conscious IOPs were assessed for 35 days. Results: Upon Wnt3a treatment, total cadherin expression increased and ß-catenin accumulated at the TM cell membrane and on processes formed between TM cells. qPCR showed that Wnt3a significantly increased K-cadherin expression in NTM cells (P < 0.01, n = 3), and Western immunoblotting showed that Wnt3a increased K-cadherin in NTM cells, which was inhibited by the addition of sFRP1. Cell impedance assays showed that Wnt3a treatment increased transcellular resistance and anti-K-cadherin siRNA decreased transcellular resistance (P < 0.001, n = 4-6). Our in vivo study showed that K-cadherin significantly decreased sFRP1-induced ocular hypertension (P < 0.05, n = 6). Western immunoblotting also showed that K-cadherin alleviated sFRP1-induced ß-catenin decrease in mouse anterior segments. Conclusions: Our results suggest that cadherins play important roles in the regulation of TM homeostasis and IOP via the Wnt/ß-catenin pathway.


Subject(s)
Cadherins/physiology , Intraocular Pressure/physiology , Trabecular Meshwork/drug effects , Trabecular Meshwork/metabolism , Wnt Signaling Pathway/physiology , Wnt3A Protein/pharmacology , beta Catenin/physiology , Animals , Blotting, Western , Cadherins/metabolism , Cell Membrane/metabolism , Cells, Cultured , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Protein Isoforms/metabolism , Signal Transduction/physiology , beta Catenin/metabolism
14.
Br J Cancer ; 117(6): 791-800, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28787429

ABSTRACT

BACKGROUND: The use of clinical MRI scanners to conduct pre-clinical research facilitates comparisons with clinical studies. Here the utility and sensitivity of anatomical and functional MRI data/biomarkers acquired from transgenic mouse models of neuroblastoma using a dedicated radiofrequency (RF) coil on a clinical 3T scanner was evaluated. METHODS: Multiparametric MRI of transgenic mice bearing abdominal neuroblastomas was performed at 3T, and data cross-referenced to that acquired from the same mice on a pre-clinical 7T MRI system. T2-weighted imaging, quantitation of the native longitudinal relaxation time (T1) and the transverse relaxation rate (R2*), and dynamic contrast-enhanced (DCE)-MRI, was used to assess tumour volume, phenotype and response to cyclophosphamide or cabozantinib. RESULTS: Excellent T2-weighted image contrast enabled clear tumour delineation at 3T. Significant correlations of tumour volume (R=0.98, P<0.0001) and R2* (R=0.87, P<0.002) measured at 3 and 7T were established. Mice with neuroblastomas harbouring the anaplastic lymphoma kinase mutation exhibited a significantly slower R2* (P<0.001), consistent with impaired tumour perfusion. DCE-MRI was performed simultaneously on three transgenic mice, yielding estimates of Ktrans for each tumour (median Ktrans values of 0.202, 0.168 and 0.114 min-1). Cyclophosphamide elicited a significant reduction in both tumour burden (P<0.002) and native T1 (P<0.01), whereas cabozantinib induced significant (P<0.01) tumour growth delay. CONCLUSIONS: Simultaneous multiparametric MRI of multiple tumour-bearing animals using this coil arrangement at 3T can provide high efficiency/throughput for both phenotypic characterisation and evaluation of novel therapeutics, and facilitate the introduction of functional MRI biomarkers into aligned imaging-embedded clinical trials.


Subject(s)
Magnetic Resonance Imaging/methods , Magnets , Neuroblastoma/diagnostic imaging , Stomach Neoplasms/diagnostic imaging , Anaplastic Lymphoma Kinase , Anilides/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Contrast Media , Cyclophosphamide/therapeutic use , Disease Models, Animal , Female , Magnetic Resonance Imaging/instrumentation , Male , Mice , Mice, Transgenic , Mutation , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , Phantoms, Imaging , Phenotype , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/genetics , Signal-To-Noise Ratio , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Burden/drug effects
15.
Invest Ophthalmol Vis Sci ; 58(2): 1288-1295, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28241317

ABSTRACT

Purpose: Increased intraocular pressure results from increased aqueous humor (AH) outflow resistance at the trabecular meshwork (TM) due to pathologic changes including the formation of cross-linked actin networks (CLANs). Transforming growth factor ß2 (TGFß2) is elevated in the AH and TM of primary open angle glaucoma (POAG) patients and induces POAG-associated TM changes, including CLANs. We determined the role of individual TGFß2 signaling pathways in CLAN formation. Methods: Cultured nonglaucomatous human TM (NTM) cells were treated with control or TGFß2, with or without the inhibitors of TGFß receptor, Smad3, c-Jun N-terminal kinases (JNK), extracellular signal regulated kinase (ERK), P38, or Rho-associated protein kinase (ROCK). NTM cells were cotreated with TGFß2 plus inhibitors for 10 days or pretreated with TGFß2 for 10 days followed by 1-hour inhibitor treatment. NTM cells were immunostained with phalloidin-Alexa-488 and 4',6-diamidino-2-phenylindole (DAPI). Data were analyzed using 1-way ANOVA and Dunnett's post hoc test. Results: TGFß2 significantly induced CLAN formation (n = 6 to 12, P < 0.05), which was completely inhibited by TGFß receptor, Smad3, and ERK inhibitors, as well as completely or partially inhibited by JNK, P38, and ROCK inhibitors, depending on cell strains. One-hour exposure to ROCK inhibitor completely resolved formed CLANs (P < 0.05), whereas TGFß receptor, Smad3 inhibitor, and ERK inhibitors resulted in partial or complete resolution. The JNK and P38 inhibitors showed partial or no resolution. Among these inhibitors, the ROCK inhibitor was the most disruptive to the actin stress fibers, whereas ERK inhibition showed the least disruption. Conclusions: TGFß2-induced CLANs in NTM cells were prevented and resolved using various pathway inhibitors. Apart from CLAN inhibition, some of these inhibitors also had different effects on actin stress fibers.


Subject(s)
Actins/metabolism , Smad3 Protein/metabolism , Trabecular Meshwork/drug effects , Transforming Growth Factor beta2/pharmacology , Analysis of Variance , Aqueous Humor/metabolism , Blotting, Western , Cells, Cultured , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta , Signal Transduction/physiology , Trabecular Meshwork/metabolism , Transforming Growth Factor beta2/antagonists & inhibitors , Transforming Growth Factor beta2/physiology
16.
PLoS One ; 12(1): e0169671, 2017.
Article in English | MEDLINE | ID: mdl-28068412

ABSTRACT

The most common ocular side effect of glucocorticoid (GC) therapy is GC-induced ocular hypertension (OHT) and GC-induced glaucoma (GIG). GC-induced OHT occurs in about 40% of the general population, while the other 60% are resistant. This study aims to determine the genes and pathways involved in differential GC responsiveness in the trabecular meshwork (TM). Using paired bovine eyes, one eye was perfusion-cultured with 100nM dexamethasone (DEX), while the fellow eye was used to establish a bovine TM (BTM) cell strain. Based on maximum IOP change in the perfused eye, the BTM cell strain was identified as a DEX-responder or non-responder strain. Three responder and three non-responder BTM cell strains were cultured, treated with 0.1% ethanol or 100nM DEX for 7 days. RNA and proteins were extracted for RNA sequencing (RNAseq), qPCR, and Western immunoblotting (WB), respectively. Data were analyzed using the human and bovine genome databases as well as Tophat2 software. Genes were grouped and compared using Student's t-test. We found that DEX induced fibronectin expression in responder BTM cells but not in non-responder cells using WB. RNAseq showed between 93 and 606 differentially expressed genes in different expression groups between responder and non-responder BTM cells. The data generated by RNAseq were validated using qPCR. Pathway analyses showed 35 pathways associated with differentially expressed genes. These genes and pathways may play important roles in GC-induced OHT and will help us to better understand differential ocular responsiveness to GCs.


Subject(s)
Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Trabecular Meshwork/cytology , Transcriptome , Animals , Cattle , Computational Biology/methods , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Protein Interaction Mapping , Sequence Analysis, RNA , Signal Transduction , Trabecular Meshwork/metabolism
17.
Invest Ophthalmol Vis Sci ; 57(8): 3698-707, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27403998

ABSTRACT

PURPOSE: Elevated intraocular pressure (IOP) in primary open-angle glaucoma (POAG) results from glaucomatous damage to the trabecular meshwork (TM). The glaucoma-associated factor TGFß2 is increased in aqueous humor and TM of POAG patients. We hypothesize that histone acetylation has a role in dysregulated TGFß2 expression. METHODS: Protein acetylation was compared between nonglaucomatous TM (NTM) and glaucomatous TM (GTM) cells using Western immunoblotting (WB). Nonglaucomatous TM cells were treated with 10 nM thailandepsin-A (TDP-A), a potent histone deacetylase inhibitor for 4 days. Total and nuclear proteins, RNA, and nuclear protein-DNA complexes were harvested for WB, quantitative PCR (qPCR), and chromatin immunoprecipitation (ChIP) assays, respectively. Paired bovine eyes were perfused with TDP-A versus DMSO, or TDP-A versus TDP-A plus the TGFß pathway inhibitor LY364947 for 5 to 9 days. Intraocular pressure, TM, and perfusate proteins were compared. RESULTS: We found increased acetylated histone 3 and total protein acetylation in the GTM cells and TDP-A treated NTM cells. Chromatin immunoprecipitation assays showed that TDP-A induced histone hyperacetylation associated with the TGFß2 promoter. This change of acetylation significantly increased TGFß2 mRNA and protein expression in NTM cells. In perfusion-cultured bovine eyes, TDP-A increased TGFß2 in the perfusate as well as elevated IOP. Histologic and immunofluorescent analyses showed increased extracellular matrix and cytoskeletal proteins in the TM of TDP-A treated bovine eyes. Cotreatment with the TGFß pathway inhibitor LY364947 blocked TDP-A-induced ocular hypertension. CONCLUSIONS: Our results suggest that histone acetylation has an important role in increased expression of the glaucoma-associated factor TGFß2. Histone hyperacetylation may be the initiator of glaucomatous damage to the TM.


Subject(s)
Epigenesis, Genetic/genetics , Glaucoma, Open-Angle/genetics , Histone Deacetylase Inhibitors/pharmacology , Trabecular Meshwork/physiology , Transforming Growth Factor beta2/genetics , Acetylation , Animals , Cattle , Cells, Cultured , Pyrazoles/pharmacology , Pyrroles/pharmacology , RNA, Messenger/metabolism
18.
Cancer Res ; 76(10): 3025-35, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27197232

ABSTRACT

Neuroblastoma is the most common childhood extracranial solid tumor. In high-risk cases, many of which are characterized by amplification of MYCN, outcome remains poor. Mutations in the p53 (TP53) tumor suppressor are rare at diagnosis, but evidence suggests that p53 function is often impaired in relapsed, treatment-resistant disease. To address the role of p53 loss of function in the development and pathogenesis of high-risk neuroblastoma, we generated a MYCN-driven genetically engineered mouse model in which the tamoxifen-inducible p53ER(TAM) fusion protein was expressed from a knock-in allele (Th-MYCN/Trp53(KI)). We observed no significant differences in tumor-free survival between Th-MYCN mice heterozygous for Trp53(KI) (n = 188) and Th-MYCN mice with wild-type p53 (n = 101). Conversely, the survival of Th-MYCN/Trp53(KI/KI) mice lacking functional p53 (n = 60) was greatly reduced. We found that Th-MYCN/Trp53(KI/KI) tumors were resistant to ionizing radiation (IR), as expected. However, restoration of functional p53ER(TAM) reinstated sensitivity to IR in only 50% of Th-MYCN/Trp53(KI/KI) tumors, indicating the acquisition of additional resistance mechanisms. Gene expression and metabolic analyses indicated that the principal acquired mechanism of resistance to IR in the absence of functional p53 was metabolic adaptation in response to chronic oxidative stress. Tumors exhibited increased antioxidant metabolites and upregulation of glutathione S-transferase pathway genes, including Gstp1 and Gstz1, which are associated with poor outcome in human neuroblastoma. Accordingly, glutathione depletion by buthionine sulfoximine together with restoration of p53 activity resensitized tumors to IR. Our findings highlight the complex pathways operating in relapsed neuroblastomas and the need for combination therapies that target the diverse resistance mechanisms at play. Cancer Res; 76(10); 3025-35. ©2016 AACR.


Subject(s)
Adaptation, Physiological/radiation effects , Apoptosis/radiation effects , N-Myc Proto-Oncogene Protein/physiology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Radiation Tolerance , Tumor Suppressor Protein p53/physiology , Animals , Blotting, Western , Cell Proliferation/radiation effects , Female , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Transgenic , Neuroblastoma/radiotherapy , RNA, Messenger/genetics , Radiation, Ionizing , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
Exp Eye Res ; 148: 97-102, 2016 07.
Article in English | MEDLINE | ID: mdl-27091054

ABSTRACT

Primary Open Angle Glaucoma (POAG) is an irreversible, vision-threatening disease that affects millions worldwide. The principal risk factor of POAG is increased intraocular pressure (IOP) due to pathological changes in the trabecular meshwork (TM). The TGFß signaling pathway activator TGFß2 and the Wnt signaling pathway inhibitor secreted frizzled-related protein 1 (sFRP1) are elevated in the POAG TM. In this study, we determined whether there is a crosstalk between the TGFß/Smad pathway and the canonical Wnt pathway using luciferase reporter assays. Lentiviral luciferase reporter vectors for studying the TGFß/Smad pathway or the canonical Wnt pathway were transduced into primary human non-glaucomatous TM (NTM) cells. Cells were treated with or without a combination of 5 µg/ml TGFß2 and/or 100 ng/ml Wnt3a recombinant proteins, and luciferase levels were measured using a plate reader. We found that TGFß2 inhibited Wnt3a-induced canonical Wnt pathway activation, while Wnt3a inhibited TGFß2-induced TGFß/Smad pathway activation (n = 6, p < 0.05) in 3 NTM cell strains. We also found that knocking down of Smad4 or ß-catenin using siRNA in HTM5 cells transfected with similar luciferase reporter plasmids abolished the inhibitory effect of TGFß2 and/or Wnt3a on the other pathway (n = 6). Our results suggest the existence of a cross-inhibition between the TGFß/Smad and canonical Wnt pathways in the TM, and this cross-inhibition may be mediated by Smad4 and ß-catenin.


Subject(s)
Glaucoma, Open-Angle/metabolism , Trabecular Meshwork/metabolism , Transforming Growth Factor beta2/physiology , Wnt Signaling Pathway/physiology , Wnt3A Protein/physiology , Aged , Aged, 80 and over , Blotting, Western , Female , Humans , Luciferases/metabolism , Male , Signal Transduction/physiology , Smad4 Protein/metabolism , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/pharmacology , Wnt3A Protein/metabolism , Wnt3A Protein/pharmacology , beta Catenin/metabolism
20.
Clin Cancer Res ; 22(17): 4391-404, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27012811

ABSTRACT

PURPOSE: Deregulated MYC drives oncogenesis in many tissues yet direct pharmacologic inhibition has proven difficult. MYC coordinately regulates polyamine homeostasis as these essential cations support MYC functions, and drugs that antagonize polyamine sufficiency have synthetic-lethal interactions with MYC Neuroblastoma is a lethal tumor in which the MYC homologue MYCN, and ODC1, the rate-limiting enzyme in polyamine synthesis, are frequently deregulated so we tested optimized polyamine depletion regimens for activity against neuroblastoma. EXPERIMENTAL DESIGN: We used complementary transgenic and xenograft-bearing neuroblastoma models to assess polyamine antagonists. We investigated difluoromethylornithine (DFMO; an inhibitor of Odc, the rate-limiting enzyme in polyamine synthesis), SAM486 (an inhibitor of Amd1, the second rate-limiting enzyme), and celecoxib (an inducer of Sat1 and polyamine catabolism) in both the preemptive setting and in the treatment of established tumors. In vitro assays were performed to identify mechanisms of activity. RESULTS: An optimized polyamine antagonist regimen using DFMO and SAM486 to inhibit both rate-limiting enzymes in polyamine synthesis potently blocked neuroblastoma initiation in transgenic mice, underscoring the requirement for polyamines in MYC-driven oncogenesis. Furthermore, the combination of DFMO with celecoxib was found to be highly active, alone, and combined with numerous chemotherapy regimens, in regressing established tumors in both models, including tumors harboring highest risk genetic lesions such as MYCN amplification, ALK mutation, and TP53 mutation with multidrug resistance. CONCLUSIONS: Given the broad preclinical activity demonstrated by polyamine antagonist regimens across diverse in vivo models, clinical investigation of such approaches in neuroblastoma and potentially other MYC-driven tumors is warranted. Clin Cancer Res; 22(17); 4391-404. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Transformation, Neoplastic/drug effects , Neuroblastoma/etiology , Neuroblastoma/pathology , Polyamines/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Celecoxib/pharmacology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Drug Synergism , Eflornithine/pharmacology , Genes, myc , Homeostasis/drug effects , Humans , Mice , Mice, Transgenic , Neuroblastoma/drug therapy , Neuroblastoma/mortality , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Polyamines/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...