Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Immunol ; 195(3): 953-64, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26116508

ABSTRACT

NF-κB-inducing kinase (NIK) is a primary regulator of the noncanonical NF-κB signaling pathway, which plays a vital role downstream of BAFF, CD40L, lymphotoxin, and other inflammatory mediators. Germline deletion or inactivation of NIK in mice results in the defective development of B cells and secondary lymphoid organs, but the role of NIK in adult animals has not been studied. To address this, we generated mice containing a conditional allele of NIK. Deletion of NIK in adult mice results in decreases in B cell populations in lymph nodes and spleen, similar to what is observed upon blockade of BAFF. Consistent with this, B cells from mice in which NIK is acutely deleted fail to respond to BAFF stimulation in vitro and in vivo. In addition, mice with induced NIK deletion exhibit a significant decrease in germinal center B cells and serum IgA, which is indicative of roles for NIK in additional pathways beyond BAFF signaling. Our conditional NIK-knockout mice may be broadly useful for assessing the postdevelopmental and cell-specific roles of NIK and the noncanonical NF-κB pathway in mice.


Subject(s)
B-Cell Activating Factor/genetics , B-Lymphocytes/immunology , Lymphocyte Activation/genetics , NF-kappa B p52 Subunit/biosynthesis , Protein Serine-Threonine Kinases/genetics , Animals , B-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Germ-Line Mutation , I-kappa B Kinase/metabolism , Immunoglobulin A/blood , Lymph Nodes/cytology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , NF-kappa B p52 Subunit/genetics , Sequence Deletion , Signal Transduction/genetics , Signal Transduction/immunology , Spleen/cytology , Tamoxifen/pharmacology , NF-kappaB-Inducing Kinase
2.
Oncologist ; 19(4): 336-43, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24664487

ABSTRACT

PURPOSE: The success of precision oncology relies on accurate and sensitive molecular profiling. The Ion AmpliSeq Cancer Panel, a targeted enrichment method for next-generation sequencing (NGS) using the Ion Torrent platform, provides a fast, easy, and cost-effective sequencing workflow for detecting genomic "hotspot" regions that are frequently mutated in human cancer genes. Most recently, the U.K. has launched the AmpliSeq sequencing test in its National Health Service. This study aimed to evaluate the clinical application of the AmpliSeq methodology. METHODS: We used 10 ng of genomic DNA from formalin-fixed, paraffin-embedded human colorectal cancer (CRC) tumor specimens to sequence 46 cancer genes using the AmpliSeq platform. In a validation study, we developed an orthogonal NGS-based resequencing approach (SimpliSeq) to assess the AmpliSeq variant calls. RESULTS: Validated mutational analyses revealed that AmpliSeq was effective in profiling gene mutations, and that the method correctly pinpointed "true-positive" gene mutations with variant frequency >5% and demonstrated high-level molecular heterogeneity in CRC. However, AmpliSeq enrichment and NGS also produced several recurrent "false-positive" calls in clinically druggable oncogenes such as PIK3CA. CONCLUSION: AmpliSeq provided highly sensitive and quantitative mutation detection for most of the genes on its cancer panel using limited DNA quantities from formalin-fixed, paraffin-embedded samples. For those genes with recurrent "false-positive" variant calls, caution should be used in data interpretation, and orthogonal verification of mutations is recommended for clinical decision making.


Subject(s)
Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , DNA, Neoplasm/analysis , Genes, Neoplasm/genetics , Base Sequence , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , DNA Mutational Analysis/methods , Formaldehyde , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Mutation/genetics , Paraffin , Phosphatidylinositol 3-Kinases/genetics , Sequence Analysis, DNA , Tissue Embedding , Tissue Fixation
3.
Pigment Cell Melanoma Res ; 32(2): 269-279, 2019 03.
Article in English | MEDLINE | ID: mdl-30156010

ABSTRACT

The deubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with a high risk of mesothelioma and melanocytic tumors. Here, we show that Bap1 deletion in melanocytes cooperates with the constitutively active, oncogenic form of BRAF (BRAFV600E ) and UV to cause melanoma in mice, albeit at very low frequency. In addition, Bap1-null melanoma cells derived from mouse tumors are more aggressive and colonize and grow at distant sites more than their wild-type counterparts. Molecularly, Bap1-null melanoma cell lines have increased DNA damage measured by γH2aX and hyperubiquitination of histone H2a. Therapeutically, these Bap1-null tumors are completely responsive to BRAF- and MEK-targeted therapies. Therefore, BAP1 functions as a tumor suppressor and limits tumor progression in melanoma.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Melanoma/genetics , Melanoma/pathology , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , DNA Damage , Epithelial-Mesenchymal Transition/genetics , Gene Deletion , Gene Expression Regulation, Neoplastic , Histones/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Melanocytes/metabolism , Melanocytes/pathology , Mice, Inbred C57BL , Mice, Knockout , Transcription, Genetic , Ubiquitination , Melanoma, Cutaneous Malignant
4.
Mol Cell Biol ; 25(16): 7054-68, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16055717

ABSTRACT

The Drosophila Fused (Fu) kinase is an integral component of the Hedgehog (Hh) pathway that helps promote Hh-dependent gene transcription. Vertebrate homologues of Fu function in the Hh pathway in vitro, suggesting that Fu is evolutionarily conserved. We have generated fused (stk36) knockout mice to address the in vivo function of the mouse Fu (mFu) homologue. fused knockouts develop normally, being born in Mendelian ratios, but fail to thrive within 2 weeks, displaying profound growth retardation with communicating hydrocephalus and early mortality. The fused gene is expressed highly in ependymal cells and the choroid plexus, tissues involved in the production and circulation of cerebral spinal fluid (CSF), suggesting that loss of mFu disrupts CSF homeostasis. Similarly, fused is highly expressed in the nasal epithelium, where fused knockouts display bilateral suppurative rhinitis. No obvious defects were observed in the development of organs where Hh signaling is required (limbs, face, bones, etc.). Specification of neuronal cell fates by Hh in the neural tube was normal in fused knockouts, and induction of Hh target genes in numerous tissues is not affected by the loss of mFu. Furthermore, stimulation of fused knockout cerebellar granule cells to proliferate with Sonic Hh revealed no defect in Hh signal transmission. These results show that the mFu homologue is not required for Hh signaling during embryonic development but is required for proper postnatal development, possibly by regulating the CSF homeostasis or ciliary function.


Subject(s)
Cerebrospinal Fluid/metabolism , Gene Expression Regulation, Developmental , Hydrocephalus/etiology , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Axin Protein , Cell Lineage , Cell Proliferation , Dose-Response Relationship, Drug , Genes, Reporter , Genotype , Heterozygote , Hydrocephalus/genetics , Hydrocephalus/metabolism , In Situ Hybridization , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Models, Genetic , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rhinitis/genetics , Signal Transduction , Time Factors , Tissue Distribution , Transcription, Genetic , beta-Galactosidase/metabolism
5.
Toxicol Sci ; 152(1): 72-84, 2016 07.
Article in English | MEDLINE | ID: mdl-27103662

ABSTRACT

CRTh2 is expressed on immune cells that drive asthma pathophysiology. Current treatment options for severe asthma are inadequate and therapeutic antibody-mediated depletion of CRTh2-expressing cells represents a promising new therapeutic strategy. Here we report for the first time that CRTh2 is not only expressed on immune cells, but also on microvasculature in the central nervous system (CNS) and gastric mucosa in humans. Microvascular expression of CRTh2 raises a safety concern because a therapeutic antiCRTh2 antibody with enhanced depletion capacity could lead to vascular damage. To evaluate this safety risk, we characterized microvascular expression in human and in transgenic mice expressing human CRTh2 protein (hCRTh2.BAC.Tg) and found that CRTh2 is not localized to microvascular endothelium that is directly exposed to circulating therapeutic antibody, but rather, to pericytes that in the CNS are shielded from direct circulatory exposure by the blood-brain barrier. Immunohistochemical visualization of an intravenously administered antiCRTh2 antibody in transgenic mice revealed localization to microvascular pericytes in the gastric mucosa but not in the CNS, suggesting the blood-brain barrier effectively limits pericyte exposure to circulating therapeutic antibody in the CNS. Repeated dosing with a depleting antiCRTh2 antibody in hCRTh2.BAC.Tg mice revealed linear pharmacokinetics and no drug-related adverse findings in any tissues, including the CNS and gastric mucosa, despite complete depletion of CRTh2 expressing circulating eosinophils and basophils. Collectively, these studies demonstrate that the likelihood of drug-related CNS or gastrointestinal toxicity in humans treated with a therapeutic depleting antiCRTh2 antibody is low despite pericyte expression of CRTh2 in these tissues.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Asthma/drug therapy , Central Nervous System/drug effects , Gastric Mucosa/drug effects , Pericytes/drug effects , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Animals , Anti-Asthmatic Agents/administration & dosage , Anti-Asthmatic Agents/pharmacokinetics , Anti-Asthmatic Agents/toxicity , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/toxicity , Asthma/immunology , Asthma/metabolism , Blood-Brain Barrier/metabolism , Capillary Permeability , Central Nervous System/immunology , Central Nervous System/metabolism , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Humans , Injections, Intravenous , Mice, Inbred C57BL , Mice, Transgenic , Pericytes/immunology , Pericytes/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/immunology , Receptors, Prostaglandin/metabolism , Risk Assessment , Tissue Distribution
6.
Clin Cancer Res ; 21(14): 3252-62, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25862760

ABSTRACT

PURPOSE: Chemotherapies are limited by a narrow therapeutic index resulting in suboptimal exposure of the tumor to the drug and acquired tumor resistance. One approach to overcome this is through antibody-drug conjugates (ADC) that facilitate greater potency via target-specific delivery of highly potent cytotoxic agents. EXPERIMENTAL DESIGN: In this study, we used a bioinformatics approach to identify the lymphocyte antigen 6 complex locus E (LY6E), an IFN-inducible glycosylphosphatidylinositol (GPI)-linked cell membrane protein as a promising ADC target. We developed a monoclonal anti-LY6E antibody and characterized in situ LY6E expression in over 750 cancer specimens and normal tissues. Target-dependent anti-LY6E ADC killing was investigated both in vitro and in vivo using patient-derived xenograft models. RESULTS: Using in silico approaches, we found that LY6E was significantly overexpressed and amplified in a wide array of different human solid tumors. IHC analysis revealed high LY6E protein expression in a number of tumor types, such as breast, lung, gastric, ovarian, pancreatic, kidney and head/neck carcinomas. Characterization of the endocytic pathways for LY6E revealed that the LY6E-specific antibody is internalized into cells leading to lysosomal accumulation. Consistent with this, a LY6E-specific ADC inhibited in vitro cell proliferation and produced durable tumor regression in vivo in clinically relevant LY6E-expressing xenograft models. CONCLUSIONS: Our results identify LY6E as a highly promising molecular ADC target for a variety of solid tumor types with current unmet medical need.


Subject(s)
Antigens, Neoplasm/pharmacology , Antigens, Surface/immunology , Antineoplastic Agents/pharmacology , Immunoconjugates/pharmacology , Neoplasms/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Cell Line, Tumor , Flow Cytometry , GPI-Linked Proteins/immunology , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, SCID , Polymerase Chain Reaction , RNA, Small Interfering , Transfection , Xenograft Model Antitumor Assays
7.
PLoS One ; 2(6): e575, 2007 Jun 27.
Article in English | MEDLINE | ID: mdl-17593974

ABSTRACT

BACKGROUND: Heparan sulfate proteoglycans (HSPGs) use highly sulfated polysaccharide side-chains to interact with several key growth factors and morphogens, thereby regulating their accessibility and biological activity. Various sulfotransferases and sulfatases with differing specificities control the pattern of HSPG sulfation, which is functionally critical. Among these enzymes in the mouse are two secreted 6-O-endosulfatases, Sulf1 and Sulf2, which modify HSPGs in the extracellular matrix and on the cell surface. The roles of Sulf1 and Sulf2 during normal development are not well understood. METHODS/RESULTS: To investigate the importance of Sulf1 and Sulf2 for embryonic development, we generated mice genetically deficient in these genes and assessed the phenotypes of the resulting secreted sulfatase-deficient mice. Surprisingly, despite the established crucial role of HSPG interactions during development, neither Sulf1- nor Sulf2-deficient mice showed significant developmental flaws. In contrast, mice deficient in both Sulf1and Sulf2 exhibited highly penetrant neonatal lethality. Loss of viability was associated with multiple, although subtle, developmental defects, including skeletal and renal abnormalities. CONCLUSIONS: These results show that Sulf1 and Sulf2 play overlapping yet critical roles in mouse development and are redundant and essential for neonatal survival.


Subject(s)
Animals, Newborn/growth & development , Genes, Lethal/physiology , Heparan Sulfate Proteoglycans/metabolism , Sulfatases/physiology , Sulfotransferases/physiology , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Development , Female , Fibroblasts/cytology , Fibroblasts/metabolism , In Situ Hybridization , Kidney/abnormalities , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/abnormalities , Pregnancy , Signal Transduction , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL