Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Res Sq ; 2024 Jan 17.
Article En | MEDLINE | ID: mdl-38313284

Tyrosine kinase (TK) fusions are frequently found in cancers, either as initiating events or as a mechanism of resistance to targeted therapy. Partner genes and exons in most TK fusions are typical and recurrent, but the underlying mechanisms and clinical implications of these patterns are poorly understood. Here, we investigated structures of > 8,000 kinase fusions and explore their generative mechanisms by applying newly developed experimental framework integrating high-throughput genome-wide gene fusion sequencing and clonal selection called Functionally Active Chromosomal Translocation Sequencing (FACTS). We discovered that typical oncogenic TK fusions recurrently seen in patients are selected from large pools of chromosomal rearrangements spontaneously occurring in cells based on two major determinants: active transcription of the fusion partner genes and protein stability. In contrast, atypical TK fusions that are rarely seen in patients showed reduced protein stability, decreased downstream oncogenic signaling, and were less responsive to inhibition. Consistently, patients with atypical TK fusions were associated with a reduced response to TKI therapies, as well as a shorter progression-free survival (PFS) and overall survival (OS) compared to patients with typical TK fusions. These findings highlight the principles of oncogenic TK fusion formation and their selection in cancers, with clinical implications for guiding targeted therapy.

2.
Nature ; 627(8004): 594-603, 2024 Mar.
Article En | MEDLINE | ID: mdl-38383780

Although KDM5C is one of the most frequently mutated genes in X-linked intellectual disability1, the exact mechanisms that lead to cognitive impairment remain unknown. Here we use human patient-derived induced pluripotent stem cells and Kdm5c knockout mice to conduct cellular, transcriptomic, chromatin and behavioural studies. KDM5C is identified as a safeguard to ensure that neurodevelopment occurs at an appropriate timescale, the disruption of which leads to intellectual disability. Specifically, there is a developmental window during which KDM5C directly controls WNT output to regulate the timely transition of primary to intermediate progenitor cells and consequently neurogenesis. Treatment with WNT signalling modulators at specific times reveal that only a transient alteration of the canonical WNT signalling pathway is sufficient to rescue the transcriptomic and chromatin landscapes in patient-derived cells and to induce these changes in wild-type cells. Notably, WNT inhibition during this developmental period also rescues behavioural changes of Kdm5c knockout mice. Conversely, a single injection of WNT3A into the brains of wild-type embryonic mice cause anxiety and memory alterations. Our work identifies KDM5C as a crucial sentinel for neurodevelopment and sheds new light on KDM5C mutation-associated intellectual disability. The results also increase our general understanding of memory and anxiety formation, with the identification of WNT functioning in a transient nature to affect long-lasting cognitive function.


Cognition , Embryo, Mammalian , Embryonic Development , Histone Demethylases , Wnt Signaling Pathway , Animals , Humans , Mice , Anxiety , Chromatin/drug effects , Chromatin/genetics , Chromatin/metabolism , Embryo, Mammalian/metabolism , Gene Expression Profiling , Histone Demethylases/genetics , Histone Demethylases/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Intellectual Disability/genetics , Memory , Mice, Knockout , Mutation , Neurogenesis/genetics , Wnt Signaling Pathway/drug effects
3.
STAR Protoc ; 4(1): 101975, 2023 03 17.
Article En | MEDLINE | ID: mdl-36580401

Experimentally targeting mouse choroid plexus (ChP) provides a valuable approach for investigating mechanisms of ChP-cerebrospinal fluid (CSF) biology. Here, we provide a protocol to deliver adeno-associated viral vectors (AAVs) by in utero intracerebroventricular (ICV) injection to ChP epithelial cells. We begin by describing steps for induction anesthesia of the pregnant dam, laparotomy, and in utero ICV injection. We also detail post-surgical care and immunoblot validation. For complete details on the use and execution of this protocol, please refer to Jang et al. (2022).1.


Choroid Plexus , Epithelial Cells , Mice , Animals , Female , Pregnancy
4.
Neuron ; 110(20): 3288-3301.e8, 2022 10 19.
Article En | MEDLINE | ID: mdl-36070751

For many cancer patients, chemotherapy produces untreatable life-long neurologic effects termed chemotherapy-related cognitive impairment (CRCI). We discovered that the chemotherapy methotrexate (MTX) adversely affects oxidative metabolism of non-cancerous choroid plexus (ChP) cells and the cerebrospinal fluid (CSF). We used a ChP-targeted adeno-associated viral (AAV) vector approach in mice to augment CSF levels of the secreted antioxidant SOD3. AAV-SOD3 gene therapy increased oxidative defense capacity of the CSF and prevented MTX-induced lipid peroxidation in the hippocampus. Furthermore, this gene therapy prevented anxiety and deficits in short-term learning and memory caused by MTX. MTX-induced oxidative damage to cultured human cortical neurons and analyses of CSF samples from MTX-treated lymphoma patients demonstrated that MTX diminishes antioxidant capacity of patient CSF. Collectively, our findings motivate the advancement of ChP- and CSF-targeted anti-oxidative prophylactic measures to relieve CRCI.


Antioxidants , Neoplasms , Humans , Animals , Mice , Antioxidants/pharmacology , Antioxidants/therapeutic use , Choroid Plexus , Methotrexate/toxicity , Oxidative Stress , Hippocampus , Neoplasms/chemically induced
5.
Fluids Barriers CNS ; 19(1): 36, 2022 May 26.
Article En | MEDLINE | ID: mdl-35619113

Choroid plexus (ChP) epithelial cells are crucial for the function of the blood-cerebrospinal fluid barrier (BCSFB) in the developing and mature brain. The ChP is considered the primary source and regulator of CSF, secreting many important factors that nourish the brain. It also performs CSF clearance functions including removing Amyloid beta and potassium. As such, the ChP is a promising target for gene and drug therapy for neurodevelopmental and neurological disorders in the central nervous system (CNS). This review describes the current successful and emerging experimental approaches for targeting ChP epithelial cells. We highlight methodological strategies to specifically target these cells for gain or loss of function in vivo. We cover both genetic models and viral gene delivery systems. Additionally, several lines of reporters to access the ChP epithelia are reviewed. Finally, we discuss exciting new approaches, such as chemical activation and transplantation of engineered ChP epithelial cells. We elaborate on fundamental functions of the ChP in secretion and clearance and outline experimental approaches paving the way to clinical applications.


Amyloid beta-Peptides , Choroid Plexus , Cell Count , Epithelial Cells , Epithelium
7.
Development ; 148(10)2021 05 15.
Article En | MEDLINE | ID: mdl-34032267

The choroid plexus (ChP) produces cerebrospinal fluid and forms an essential brain barrier. ChP tissues form in each brain ventricle, each one adopting a distinct shape, but remarkably little is known about the mechanisms underlying ChP development. Here, we show that epithelial WNT5A is crucial for determining fourth ventricle (4V) ChP morphogenesis and size in mouse. Systemic Wnt5a knockout, or forced Wnt5a overexpression beginning at embryonic day 10.5, profoundly reduced ChP size and development. However, Wnt5a expression was enriched in Foxj1-positive epithelial cells of 4V ChP plexus, and its conditional deletion in these cells affected the branched, villous morphology of the 4V ChP. We found that WNT5A was enriched in epithelial cells localized to the distal tips of 4V ChP villi, where WNT5A acted locally to activate non-canonical WNT signaling via ROR1 and ROR2 receptors. During 4V ChP development, MEIS1 bound to the proximal Wnt5a promoter, and gain- and loss-of-function approaches demonstrated that MEIS1 regulated Wnt5a expression. Collectively, our findings demonstrate a dual function of WNT5A in ChP development and identify MEIS transcription factors as upstream regulators of Wnt5a in the 4V ChP epithelium.


Choroid Plexus/embryology , Epithelium/metabolism , Fourth Ventricle/embryology , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Wnt-5a Protein/metabolism , Animals , Brain/embryology , CRISPR-Cas Systems/genetics , Cell Line , Epithelial Cells/metabolism , Female , HEK293 Cells , Humans , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Signal Transduction/physiology , Wnt-5a Protein/genetics
8.
Cell ; 184(11): 3056-3074.e21, 2021 05 27.
Article En | MEDLINE | ID: mdl-33932339

The choroid plexus (ChP) in each brain ventricle produces cerebrospinal fluid (CSF) and forms the blood-CSF barrier. Here, we construct a single-cell and spatial atlas of each ChP in the developing, adult, and aged mouse brain. We delineate diverse cell types, subtypes, cell states, and expression programs in epithelial and mesenchymal cells across ages and ventricles. In the developing ChP, we predict a common progenitor pool for epithelial and neuronal cells, validated by lineage tracing. Epithelial and fibroblast cells show regionalized expression by ventricle, starting at embryonic stages and persisting with age, with a dramatic transcriptional shift with maturation, and a smaller shift in each aged cell type. With aging, epithelial cells upregulate host-defense programs, and resident macrophages upregulate interleukin-1ß (IL-1ß) signaling genes. Our atlas reveals cellular diversity, architecture and signaling across ventricles during development, maturation, and aging of the ChP-brain barrier.


Choroid Plexus/embryology , Choroid Plexus/metabolism , Age Factors , Aging/physiology , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/physiology , Brain Diseases/genetics , Brain Diseases/physiopathology , Cell Differentiation/genetics , Cell Lineage/genetics , Choroid Plexus/physiology , Epithelial Cells/metabolism , Female , Male , Mice/embryology , Mice, Inbred C57BL , Signal Transduction , Single-Cell Analysis
9.
Neuron ; 99(5): 905-913.e7, 2018 09 05.
Article En | MEDLINE | ID: mdl-30146301

Channelopathies are disorders caused by abnormal ion channel function in differentiated excitable tissues. We discovered a unique neurodevelopmental channelopathy resulting from pathogenic variants in SCN3A, a gene encoding the voltage-gated sodium channel NaV1.3. Pathogenic NaV1.3 channels showed altered biophysical properties including increased persistent current. Remarkably, affected individuals showed disrupted folding (polymicrogyria) of the perisylvian cortex of the brain but did not typically exhibit epilepsy; they presented with prominent speech and oral motor dysfunction, implicating SCN3A in prenatal development of human cortical language areas. The development of this disorder parallels SCN3A expression, which we observed to be highest early in fetal cortical development in progenitor cells of the outer subventricular zone and cortical plate neurons and decreased postnatally, when SCN1A (NaV1.1) expression increased. Disrupted cerebral cortical folding and neuronal migration were recapitulated in ferrets expressing the mutant channel, underscoring the unexpected role of SCN3A in progenitor cells and migrating neurons.


Cerebral Cortex/diagnostic imaging , Cerebral Cortex/growth & development , Language Development , NAV1.3 Voltage-Gated Sodium Channel/genetics , Sodium Channels/genetics , Adolescent , Adult , Animals , Cell Movement/physiology , Cells, Cultured , Cerebral Cortex/pathology , Child , Child, Preschool , Female , Ferrets , HEK293 Cells , Humans , Infant , Male , Megalencephaly/diagnostic imaging , Megalencephaly/genetics , Megalencephaly/pathology , Middle Aged , Pedigree , Polymicrogyria/diagnostic imaging , Polymicrogyria/genetics , Polymicrogyria/pathology
10.
Lab Anim Res ; 32(4): 194-199, 2016 Dec.
Article En | MEDLINE | ID: mdl-28053612

Dynamin-like protein I (DLP-1) is an important mitochondrial fission and fusion protein that is associated with apoptotic cell death in neurodegenerative diseases. In this study, we investigated DLP-1 expression in a focal cerebral ischemia animal model and glutamate-exposed hippocampal-derived cell line. Middle cerebral artery occlusion (MCAO) was surgically induced in adult male rats to induce focal cerebral ischemic injury. Brain tissues were collected 24 hours after the onset of MCAO. MCAO induces an increase in infarct volume and histopathological changes in the cerebral cortex. We identified a decrease in DLP-1 in the cerebral cortices of MCAO-injured animals using a proteomic approach and Western blot analysis. Moreover, glutamate treatment significantly decreased DLP-1 expression in a hippocampal-derived cell line. The decrease in DLP-1 indicates mitochondrial dysfunction. Thus, these results suggest that neuronal cell injury induces a decrease in DLP-1 levels and consequently leads to neuronal cell death.

11.
FASEB J ; 29(10): 4133-44, 2015 Oct.
Article En | MEDLINE | ID: mdl-26103986

14-3-3 proteins act as adapters that exert their function by interacting with their various protein partners. 14-3-3 proteins have been implicated in a variety of human diseases including neurodegenerative diseases. 14-3-3 proteins have recently been reported to be abundant in the neurofibrillary tangles (NFTs) observed inside the neurons of brains affected by Alzheimer's disease (AD). These NFTs are mainly constituted of phosphorylated Tau protein, a microtubule-associated protein known to bind 14-3-3. Despite this indication of 14-3-3 protein involvement in the AD pathogenesis, the role of 14-3-3 in the Tauopathy remains to be clarified. In the present study, we shed light on the role of 14-3-3 proteins in the molecular pathways leading to Tauopathies. Overexpression of the 14-3-3σ isoform resulted in a disruption of the tubulin cytoskeleton and prevented neuritic outgrowth in neurons. NMR studies validated the phosphorylated residues pSer214 and pSer324 in Tau as the 2 primary sites for 14-3-3 binding, with the crystal structure of 14-3-3σ in complex with Tau-pSer214 and Tau-pSer324 revealing the molecular details of the interaction. These data suggest a rationale for a possible pharmacologic intervention of the Tau/14-3-3 interaction.


14-3-3 Proteins/metabolism , Axons/metabolism , Biomarkers, Tumor/metabolism , Exoribonucleases/metabolism , Tubulin/metabolism , tau Proteins/metabolism , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , Axons/physiology , Binding Sites/genetics , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Crystallography, X-Ray , Cytoskeleton/metabolism , Exoribonucleases/chemistry , Exoribonucleases/genetics , Humans , Magnetic Resonance Spectroscopy , Microscopy, Confocal , Models, Molecular , Mutation , Neurites/metabolism , Neurites/physiology , Neurons/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Serine/chemistry , Serine/genetics , Serine/metabolism , Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/chemistry , tau Proteins/genetics
12.
Curr Alzheimer Res ; 10(8): 829-43, 2013 Oct.
Article En | MEDLINE | ID: mdl-23905994

The activities of CDK5 and p35 are thought to be important in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease (AD). We studied the effect of p35 deletion in Tg2576 mice, which is an AD animal model. To obtain the desired mice, we crossed p35(-/-) with Tg2576 mice. The resulting p35(-/-)/Tg2576 (KO/Tg) mice displayed higher mortality rates and exhibited impaired spatial learning and memory at 6 months of age. Using immunohistochemical and biochemical approaches, we observed a reduction in the expression of pre- and post-synaptic markers such as NMDAR1, synaptophysin and GluR1. In addition, the intensity of MAP-2-positive dendrites extending from neuronal cell bodies was significantly decreased in KO/Tg mice compared with KO/WT and WT/Tg mice. We also detected increased neuronal cell death in the hippocampus, along with thinned and collapsed morphological changes in the alveus region and a dramatic increase in the number of microglial cells. Microglial infiltration in the hippocampus could result in the increased secretion of the soluble high mobility group box-1 protein (HMGB-1). The secretion of HMGB-1 is increased by Aß, and secretion of HMGB-1 promotes neuronal cell death. Moreover, we found that HMGB-1 secretion induced by Aß in KO/Tg mice gave rise to ER-mediated cell death. In summary, during the stages of KO/Tg mice model, the microglial infiltration and secretion of soluble HMGB-1 were significantly increased in the hippocampus. These conditions promote neuronal death, synaptic destruction and behavioral deficits.


Alzheimer Disease/metabolism , Cell Death/physiology , HMGB1 Protein/metabolism , Hippocampus/metabolism , Neurons/metabolism , Phosphotransferases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Hippocampus/pathology , Maze Learning/physiology , Memory/physiology , Mice , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Neurons/pathology , Phosphotransferases/genetics
13.
J Appl Toxicol ; 33(10): 1089-96, 2013 Oct.
Article En | MEDLINE | ID: mdl-23408656

Zinc oxide (ZnO) nanoparticles (NPs) are used in diverse applications ranging from paints and cosmetics to biomedicine and food. Although micron-sized ZnO is a traditional food supplement, ZnO NPs are an unknown public health risk because of their unique physicochemical properties. Herein, we studied the 13-week subchronic toxicity of ZnO NPs administered via the oral route according to Organization for Economic Cooperation and Development (OECD) test guideline 408. Well-dispersed ZnO NPs were administered to Sprague-Dawley (SD) rats (11/sex/group) at doses of 67.1, 134.2, 268.4 or 536.8 mg kg(-1) per body weight over a 13-week period. The mean body weight gain in males given 536.8 mg kg(-1) ZnO NPs was significantly lower than that of control male rats, whereas no significant differences were observed between the other treatment groups and the controls. Male and female rats dosed at 536.8 mg kg(-1) ZnO NPs had significant changes in anemia-related hematologic parameters. Mild to moderate pancreatitis also developed in both sexes dosed at 536.8 mg kg(-1) , whereas no histological changes were observed in the other treatment groups. To evaluate the mechanism of toxicity, we performed a bio-persistence study and evaluated the effects of the ZnO NPs on cell proliferation. The treatment of a human gastric adenocarcinoma cell line with ZnO NPs resulted in a significant inhibition of cellular proliferation. The anti-proliferative effect of ZnO NPs or Zn(2+) was effectively blocked by treatment with chelators. These results indicate that the bio-persistence of ZnO NPs after ingestion is key to their toxicity; the no-observed-adverse effect level (NOAEL) of ZnO NPs was found to be 268.4 mg kg(-1) per day for both sexes.


Nanoparticles/toxicity , Pancreatitis/chemically induced , Pancreatitis/physiopathology , Zinc Oxide/toxicity , Administration, Oral , Animals , Carcinogens/administration & dosage , Carcinogens/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Edetic Acid/pharmacology , Ethylenediamines/pharmacology , Female , Humans , Male , Nanoparticles/chemistry , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Toxicity Tests, Subchronic , Zinc Oxide/chemistry
14.
Biochem Biophys Res Commun ; 424(3): 597-603, 2012 Aug 03.
Article En | MEDLINE | ID: mdl-22776201

α-Synuclein can be degraded by both the ubiquitin-proteasomal system and the chaperone-lysosomal system. However, the switching mechanism between the two pathways is not clearly understood. In our study, we investigated the mutual association between the binding of α-synuclein to heat shock cognate 70 and the lysosomal translocation of α-synuclein. Tyrosine phosphorylation of Y136 on α-synuclein increased when it bound to heat shock protein 70. We also found that tyrosine phosphorylation of α-synuclein can be regulated by focal adhesion kinase pp125 and protein tyrosine phosphatase 1B. Furthermore, protein tyrosine phosphatase 1B inhibitor protected dopaminergic neurons against cell death and rescued rotarod performance in a Parkinson's disease animal model. This study provides evidence that the regulation of Y136 phosphorylation of α-synuclein can improve behavioral performance and protect against neuronal death by promoting the turnover of lysosomal degradation of α-synuclein. As a result, protein tyrosine phosphatase 1B inhibitor may be used as a potential therapeutic agent against Parkinson's disease.


Parkinson Disease/enzymology , Proteasome Endopeptidase Complex/metabolism , alpha-Synuclein/metabolism , Animals , Cell Line , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HSC70 Heat-Shock Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Tyrosine/metabolism
...