Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Blood Adv ; 8(13): 3453-3463, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38608257

ABSTRACT

ABSTRACT: Clonal hematopoiesis (CH) is an age-associated phenomenon leading to an increased risk of both hematologic malignancy and nonmalignant organ dysfunction. Increasingly available genetic testing has made the incidental discovery of CH clinically common yet evidence-based guidelines and effective management strategies to prevent adverse CH health outcomes are lacking. To address this gap, the prospective CHIVE (clonal hematopoiesis and inflammation in the vasculature) registry and biorepository was created to identify and monitor individuals at risk, support multidisciplinary CH clinics, and refine taxonomy and standards of practice for CH risk mitigation. Data from the first 181 patients enrolled in this prospective registry recapitulate the molecular epidemiology of CH from biobank-scale retrospective studies, with DNMT3A, TET2, ASXL1, and TP53 as the most commonly mutated genes. Blood counts across all hematopoietic lineages trended lower in patients with CH. In addition, patients with CH had higher rates of end organ dysfunction, in particular chronic kidney disease. Among patients with CH, variant allele frequency was independently associated with the presence of cytopenias and progression to hematologic malignancy, whereas other common high-risk CH clone features were not clear. Notably, accumulation of multiple distinct high-risk clone features was also associated with cytopenias and hematologic malignancy progression, supporting a recently published CH risk score. Surprisingly, ∼30% of patients enrolled in CHIVE from CH clinics were adjudicated as not having clonal hematopoiesis of indeterminate potential, highlighting the need for molecular standards and purpose-built assays in this field. Maintenance of this well-annotated cohort and continued expansion of CHIVE to multiple institutions are underway and will be critical to understanding how to thoughtfully care for this patient population.


Subject(s)
Clonal Hematopoiesis , Inflammation , Humans , Prospective Studies , Female , Male , Middle Aged , Aged , Registries , Hematologic Neoplasms/genetics , Mutation , Adult
2.
Clin Transl Sci ; 16(9): 1628-1638, 2023 09.
Article in English | MEDLINE | ID: mdl-37353859

ABSTRACT

Despite complex pathways of drug disposition, clinical pharmacogenetic predictors currently rely on only a few high effect variants. Quantification of the polygenic contribution to variability in drug disposition is necessary to prioritize target drugs for pharmacogenomic approaches and guide analytic methods. Dexmedetomidine and fentanyl, often used in postoperative care of pediatric patients, have high rates of inter-individual variability in dosing requirements. Analyzing previously generated population pharmacokinetic parameters, we used Bayesian hierarchical mixed modeling to measure narrow-sense (additive) heritability ( h SNP 2 ) of dexmedetomidine and fentanyl clearance in children and identify relative contributions of small, moderate, and large effect-size variants to h SNP 2 . We used genome-wide association studies (GWAS) to identify variants contributing to variation in dexmedetomidine and fentanyl clearance, followed by functional analyses to identify associated pathways. For dexmedetomidine, median clearance was 33.0 L/h (interquartile range [IQR] 23.8-47.9 L/h) and h SNP 2 was estimated to be 0.35 (90% credible interval 0.00-0.90), with 45% of h SNP 2 attributed to large-, 32% to moderate-, and 23% to small-effect variants. The fentanyl cohort had median clearance of 8.2 L/h (IQR 4.7-16.7 L/h), with estimated h SNP 2 of 0.30 (90% credible interval 0.00-0.84). Large-effect variants accounted for 30% of h SNP 2 , whereas moderate- and small-effect variants accounted for 37% and 33%, respectively. As expected, given small sample sizes, no individual variants or pathways were significantly associated with dexmedetomidine or fentanyl clearance by GWAS. We conclude that clearance of both drugs is highly polygenic, motivating the future use of polygenic risk scores to guide appropriate dosing of dexmedetomidine and fentanyl.


Subject(s)
Dexmedetomidine , Humans , Child , Fentanyl , Genome-Wide Association Study , Bayes Theorem
3.
Nat Commun ; 14(1): 3720, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37349305

ABSTRACT

Transmission and secretion of signals via the choroid plexus (ChP) brain barrier can modulate brain states via regulation of cerebrospinal fluid (CSF) composition. Here, we developed a platform to analyze diurnal variations in male mouse ChP and CSF. Ribosome profiling of ChP epithelial cells revealed diurnal translatome differences in metabolic machinery, secreted proteins, and barrier components. Using ChP and CSF metabolomics and blood-CSF barrier analyses, we observed diurnal changes in metabolites and cellular junctions. We then focused on transthyretin (TTR), a diurnally regulated thyroid hormone chaperone secreted by the ChP. Diurnal variation in ChP TTR depended on Bmal1 clock gene expression. We achieved real-time tracking of CSF-TTR in awake TtrmNeonGreen mice via multi-day intracerebroventricular fiber photometry. Diurnal changes in ChP and CSF TTR levels correlated with CSF thyroid hormone levels. These datasets highlight an integrated platform for investigating diurnal control of brain states by the ChP and CSF.


Subject(s)
Blood-Brain Barrier , Choroid Plexus , Mice , Male , Animals , Choroid Plexus/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Thyroid Hormones/metabolism , Prealbumin/genetics , Prealbumin/metabolism , Biological Transport
4.
Dev Cell ; 55(5): 617-628.e6, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33038331

ABSTRACT

The choroid plexus (ChP) regulates brain development by secreting instructive cues and providing a protective brain barrier. Here, we show that polyI:C-mediated maternal immune activation leads to an inflammatory response in the developing embryonic mouse brain that manifests as pro-inflammatory cerebrospinal fluid (CSF) and accumulation of ChP macrophages. Elevation of CSF-CCL2 was sufficient to drive ChP immune cell recruitment, activation, and proliferation. In addition, ChP macrophages abandoned their regular tiling pattern and relocated to the ChP-free margin where they breached the weakened epithelial barrier. We further found that these immune cells entered from the ChP into the brain via anatomically specialized "hotspots" at the distal tips of ChP villi. In vivo two-photon imaging demonstrated that surveillance behaviors in ChP macrophages had already emerged at this early stage of embryogenesis. Thus, the embryonic ChP forms a functional brain barrier that can mount an inflammatory response to external insults.


Subject(s)
Choroid Plexus/embryology , Choroid Plexus/immunology , Inflammation/pathology , Animals , Calcium-Binding Proteins/metabolism , Cell Proliferation , Cerebrospinal Fluid/metabolism , Chemokine CCL2/metabolism , Imaging, Three-Dimensional , Inflammation Mediators/metabolism , Macrophage Activation , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Receptors, CCR2/metabolism , Signal Transduction , Tight Junctions/metabolism , Up-Regulation
5.
Neuron ; 108(4): 623-639.e10, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32961128

ABSTRACT

The choroid plexus (ChP) epithelium is a source of secreted signaling factors in cerebrospinal fluid (CSF) and a key barrier between blood and brain. Here, we develop imaging tools to interrogate these functions in adult lateral ventricle ChP in whole-mount explants and in awake mice. By imaging epithelial cells in intact ChP explants, we observed calcium activity and secretory events that increased in frequency following delivery of serotonergic agonists. Using chronic two-photon imaging in awake mice, we observed spontaneous subcellular calcium events as well as strong agonist-evoked calcium activation and cytoplasmic secretion into CSF. Three-dimensional imaging of motility and mobility of multiple types of ChP immune cells at baseline and following immune challenge or focal injury revealed a range of surveillance and defensive behaviors. Together, these tools should help illuminate the diverse functions of this understudied body-brain interface.


Subject(s)
Calcium/metabolism , Cerebrospinal Fluid/immunology , Cerebrospinal Fluid/metabolism , Choroid Plexus/immunology , Choroid Plexus/metabolism , Optical Imaging/methods , Animals , Choroid Plexus/drug effects , Epithelium/metabolism , Mice , Serotonin Receptor Agonists/pharmacology
6.
Development ; 146(20)2019 10 24.
Article in English | MEDLINE | ID: mdl-31575649

ABSTRACT

Massive, coordinated cellular changes accompany the transition of central nervous system (CNS) progenitors from forebrain neurectodermal cells to specified neuroepithelial cells. We have previously found that MYC regulates the changing ribosomal and proteostatic landscapes in mouse forebrain precursors at embryonic days E8.5 and E10.5 (before and after neural tube closure; NTC) (Chau et al., 2018). Here, we demonstrate parallel coordinated transcriptional changes in metabolic machinery during this same stage of forebrain specification. Progenitors showed striking mitochondrial structural changes transitioning from glycolytic cristae at E8.5, to more traditional mitochondria at E10.5. Accordingly, glucose use shifted in progenitors such that E8.5 progenitors relied on glycolysis, and after NTC increasingly used oxidative phosphorylation. This metabolic shift was matched by changes in surrounding amniotic and cerebrospinal fluid proteomes. Importantly, these mitochondrial morphological shifts depend on MYC downregulation. Together, our findings demonstrate that metabolic shifting accompanies dynamic organelle and proteostatic remodeling of progenitor cells during the earliest stages of forebrain development.


Subject(s)
Mitochondria/metabolism , Proteome/metabolism , Animals , Central Nervous System/metabolism , Epithelium/metabolism , Female , Glycolysis , Immunoblotting , Male , Mice , Mice, Mutant Strains , Microscopy, Electron, Transmission , Neuroepithelial Cells/cytology , Neuroepithelial Cells/metabolism , Prosencephalon/cytology , Prosencephalon/metabolism , RNA-Seq , Reverse Transcriptase Polymerase Chain Reaction
7.
Am J Pathol ; 189(12): 2440-2449, 2019 12.
Article in English | MEDLINE | ID: mdl-31541646

ABSTRACT

Cells of the developing central nervous system are particularly susceptible to formation of double-stranded DNA breaks (DSBs) arising from physiological and/or environmental insults. Therefore, efficient repair of DSBs is especially vital for maintaining cellular health and proper functioning in the developing brain. Here, increased expression of DSB initiating and nonhomologous end joining repair machinery in newborn neurons in the developing brains of both mouse and human are demonstrated. In parallel, the first characterization is provided of the brain phenotype in the Lig4R278H/R278H (Lig4R/R) mouse model of DNA Ligase 4 (LIG4) syndrome, in which a hypomorphic Lig4 mutation, originally identified in patients, impedes nonhomologous end joining. It is shown that Lig4R/R mice develop nonprogressive microcephaly, resulting primarily from apoptotic death of newborn neurons that is both spatially and temporally specific during peak cortical neurogenesis. This apoptosis leads to a reduction in neurons throughout the postnatal cerebral cortex, but with a more prominent impact on those of the lower cortical layers. Together, these findings begin to uncover the pathogenesis of microcephaly in LIG4 syndrome and open avenues to more focused investigations on the critical roles of DSB formation and repair in vulnerable neuronal populations of the brain.


Subject(s)
Apoptosis , Cerebral Cortex/pathology , Craniofacial Abnormalities/complications , DNA Ligase ATP/metabolism , Disease Models, Animal , Growth Disorders/complications , Immunologic Deficiency Syndromes/complications , Microcephaly/etiology , Neurons/pathology , Animals , Cerebral Cortex/metabolism , DNA Breaks, Double-Stranded , DNA Ligase ATP/genetics , Female , Gene Knock-In Techniques , Male , Mice , Microcephaly/pathology , Mutation , Neurons/metabolism , Spatio-Temporal Analysis
8.
Elife ; 72018 05 10.
Article in English | MEDLINE | ID: mdl-29745900

ABSTRACT

Forebrain precursor cells are dynamic during early brain development, yet the underlying molecular changes remain elusive. We observed major differences in transcriptional signatures of precursor cells from mouse forebrain at embryonic days E8.5 vs. E10.5 (before vs. after neural tube closure). Genes encoding protein biosynthetic machinery were strongly downregulated at E10.5. This was matched by decreases in ribosome biogenesis and protein synthesis, together with age-related changes in proteomic content of the adjacent fluids. Notably, c-MYC expression and mTOR pathway signaling were also decreased at E10.5, providing potential drivers for the effects on ribosome biogenesis and protein synthesis. Interference with c-MYC at E8.5 prematurely decreased ribosome biogenesis, while persistent c-MYC expression in cortical progenitors increased transcription of protein biosynthetic machinery and enhanced ribosome biogenesis, as well as enhanced progenitor proliferation leading to subsequent macrocephaly. These findings indicate large, coordinated changes in molecular machinery of forebrain precursors during early brain development.


Subject(s)
Down-Regulation , Gene Expression Regulation, Developmental , Organelle Biogenesis , Prosencephalon/embryology , Ribosomes/metabolism , Animals , Mice , Time Factors
9.
Am J Pathol ; 188(6): 1334-1344, 2018 06.
Article in English | MEDLINE | ID: mdl-29545198

ABSTRACT

Choroid plexus tumors and ciliary body medulloepithelioma are predominantly pediatric neoplasms. Progress in understanding the pathogenesis of these tumors has been hindered by their rarity and lack of models that faithfully recapitulate the disease. Here, we find that endogenous Myc proto-oncogene protein is down-regulated in the forebrain neuroepithelium, whose neural plate border domains give rise to the anterior choroid plexus and ciliary body. To uncover the consequences of persistent Myc expression, MYC expression was forced in multipotent neural precursors (nestin-Cre:Myc), which produced fully penetrant models of choroid plexus carcinoma and ciliary body medulloepithelioma. Nestin-mediated MYC expression in the epithelial cells of choroid plexus leads to the regionalized formation of choroid plexus carcinoma in the posterior domain of the lateral ventricle choroid plexus and the fourth ventricle choroid plexus that is accompanied by loss of multiple cilia, up-regulation of protein biosynthetic machinery, and hydrocephalus. Parallel MYC expression in the ciliary body leads also to up-regulation of protein biosynthetic machinery. Additionally, Myc expression in human choroid plexus tumors increases with aggressiveness of disease. Collectively, our findings expose a select vulnerability of the neuroepithelial lineage to postnatal tumorigenesis and provide a new mouse model for investigating the pathogenesis of these rare pediatric neoplasms.


Subject(s)
Carcinogenesis/pathology , Choroid Plexus Neoplasms/pathology , Ciliary Body/pathology , Disease Models, Animal , Neurons/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Adolescent , Adult , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Child , Child, Preschool , Choroid Plexus Neoplasms/genetics , Choroid Plexus Neoplasms/metabolism , Ciliary Body/metabolism , Female , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...