Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
2.
Am J Hum Genet ; 111(2): 338-349, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38228144

ABSTRACT

Clinical exome and genome sequencing have revolutionized the understanding of human disease genetics. Yet many genes remain functionally uncharacterized, complicating the establishment of causal disease links for genetic variants. While several scoring methods have been devised to prioritize these candidate genes, these methods fall short of capturing the expression heterogeneity across cell subpopulations within tissues. Here, we introduce single-cell tissue-specific gene prioritization using machine learning (STIGMA), an approach that leverages single-cell RNA-seq (scRNA-seq) data to prioritize candidate genes associated with rare congenital diseases. STIGMA prioritizes genes by learning the temporal dynamics of gene expression across cell types during healthy organogenesis. To assess the efficacy of our framework, we applied STIGMA to mouse limb and human fetal heart scRNA-seq datasets. In a cohort of individuals with congenital limb malformation, STIGMA prioritized 469 variants in 345 genes, with UBA2 as a notable example. For congenital heart defects, we detected 34 genes harboring nonsynonymous de novo variants (nsDNVs) in two or more individuals from a set of 7,958 individuals, including the ortholog of Prdm1, which is associated with hypoplastic left ventricle and hypoplastic aortic arch. Overall, our findings demonstrate that STIGMA effectively prioritizes tissue-specific candidate genes by utilizing single-cell transcriptome data. The ability to capture the heterogeneity of gene expression across cell populations makes STIGMA a powerful tool for the discovery of disease-associated genes and facilitates the identification of causal variants underlying human genetic disorders.


Subject(s)
Heart Defects, Congenital , Transcriptome , Humans , Animals , Mice , Exome/genetics , Heart Defects, Congenital/genetics , Exome Sequencing , Machine Learning , Single-Cell Analysis/methods , Ubiquitin-Activating Enzymes/genetics
3.
Nature ; 623(7988): 772-781, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37968388

ABSTRACT

Mouse models are a critical tool for studying human diseases, particularly developmental disorders1. However, conventional approaches for phenotyping may fail to detect subtle defects throughout the developing mouse2. Here we set out to establish single-cell RNA sequencing of the whole embryo as a scalable platform for the systematic phenotyping of mouse genetic models. We applied combinatorial indexing-based single-cell RNA sequencing3 to profile 101 embryos of 22 mutant and 4 wild-type genotypes at embryonic day 13.5, altogether profiling more than 1.6 million nuclei. The 22 mutants represent a range of anticipated phenotypic severities, from established multisystem disorders to deletions of individual regulatory regions4,5. We developed and applied several analytical frameworks for detecting differences in composition and/or gene expression across 52 cell types or trajectories. Some mutants exhibit changes in dozens of trajectories whereas others exhibit changes in only a few cell types. We also identify differences between widely used wild-type strains, compare phenotyping of gain- versus loss-of-function mutants and characterize deletions of topological associating domain boundaries. Notably, some changes are shared among mutants, suggesting that developmental pleiotropy might be 'decomposable' through further scaling of this approach. Overall, our findings show how single-cell profiling of whole embryos can enable the systematic molecular and cellular phenotypic characterization of mouse mutants with unprecedented breadth and resolution.


Subject(s)
Developmental Disabilities , Embryo, Mammalian , Mutation , Phenotype , Single-Cell Gene Expression Analysis , Animals , Mice , Cell Nucleus/genetics , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Gain of Function Mutation , Genotype , Loss of Function Mutation , Models, Genetic , Disease Models, Animal
4.
Development ; 150(3)2023 02 01.
Article in English | MEDLINE | ID: mdl-36715020

ABSTRACT

Thyroid hormone and its receptor TRα1 play an important role in brain development. Several animal models have been used to investigate this function, including mice heterozygous for the TRα1R384C mutation, which confers receptor-mediated hypothyroidism. These mice display abnormalities in several autonomic functions, which was partially attributed to a developmental defect in hypothalamic parvalbumin neurons. However, whether other cell types in the hypothalamus are similarly affected remains unknown. Here, we used single-nucleus RNA sequencing to obtain an unbiased view on the importance of TRα1 for hypothalamic development and cellular diversity. Our data show that defective TRα1 signaling has surprisingly little effect on the development of hypothalamic neuronal populations, but it heavily affects hypothalamic oligodendrocytes. Using selective reactivation of the mutant TRα1 during specific developmental periods, we find that early postnatal thyroid hormone action seems to be crucial for proper hypothalamic oligodendrocyte maturation. Taken together, our findings underline the well-known importance of postnatal thyroid health for brain development and provide an unbiased roadmap for the identification of cellular targets of TRα1 action in mouse hypothalamic development.


Subject(s)
RNA , Thyroid Hormone Receptors alpha , Mice , Animals , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormones , Thyroid Gland , Hypothalamus/metabolism
5.
Mamm Genome ; 34(2): 276-284, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36401619

ABSTRACT

The structure and function of the circulatory system, including the heart, have undergone substantial changes with the vertebrate evolution. Although the basic function of the heart is to pump blood through the body, its size, shape, speed, regeneration capacity, etc. vary considerably across species. Here, we address the differences among vertebrate hearts using a single-cell transcriptomics approach. Published datasets of macaque (Macaca fascicularis), mouse, and zebrafish hearts were integrated and compared to the human heart as a reference. While the three mammalian hearts integrated well, the zebrafish heart showed very little overlap with the other species. Our analysis revealed a mouse-specific cell subpopulation of ventricular cardiomyocytes (CM), represented by strikingly different expression patterns of specific genes related to high-energy metabolism. Interestingly, the observed differences between mouse and human CM coincided with actual biological differences between the two species. Smooth muscle and endothelial cells (EC) exhibited species-specific differences in clustering and gene expression, respectively, which we attribute to the tissues selected for sequencing, given different focuses of the original studies. Finally, we compared human and zebrafish heart-specific fibroblasts (FB) and identified a distinctively high expression of genes associated with heart regeneration following injury in zebrafish. Together, our results show that integration of numerous datasets of different species and different sequencing technologies is feasible and that this approach can identify species-specific differences and similarities in the heart.


Subject(s)
Endothelial Cells , Zebrafish , Adult , Animals , Mice , Humans , Zebrafish/genetics , Regeneration/genetics , Myocytes, Cardiac/metabolism , Gene Expression Profiling , Mammals/genetics
6.
J Med Genet ; 59(9): 827-839, 2022 09.
Article in English | MEDLINE | ID: mdl-35790352

ABSTRACT

Single-cell sequencing is a powerful approach that can detect genetic alterations and their phenotypic consequences in the context of human development, with cellular resolution. Humans start out as single-cell zygotes and undergo fission and differentiation to develop into multicellular organisms. Before fertilisation and during development, the cellular genome acquires hundreds of mutations that propagate down the cell lineage. Whether germline or somatic in nature, some of these mutations may have significant genotypic impact and lead to diseased cellular phenotypes, either systemically or confined to a tissue. Single-cell sequencing enables the detection and monitoring of the genotype and the consequent molecular phenotypes at a cellular resolution. It offers powerful tools to compare the cellular lineage between 'normal' and 'diseased' conditions and to establish genotype-phenotype relationships. By preserving cellular heterogeneity, single-cell sequencing, unlike bulk-sequencing, allows the detection of even small, diseased subpopulations of cells within an otherwise normal tissue. Indeed, the characterisation of biopsies with cellular resolution can provide a mechanistic view of the disease. While single-cell approaches are currently used mainly in basic research, it can be expected that applications of these technologies in the clinic may aid the detection, diagnosis and eventually the treatment of rare genetic diseases as well as cancer. This review article provides an overview of the single-cell sequencing technologies in the context of human genetics, with an aim to empower clinicians to understand and interpret the single-cell sequencing data and analyses. We discuss the state-of-the-art experimental and analytical workflows and highlight current challenges/limitations. Notably, we focus on two prospective applications of the technology in human genetics, namely the annotation of the non-coding genome using single-cell functional genomics and the use of single-cell sequencing data for in silico variant prioritisation.


Subject(s)
Genetic Variation , Genomics , Genotype , Human Genetics , Humans , Phenotype
7.
Cell Rep ; 40(1): 111037, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35793629

ABSTRACT

One key factor underlying the functional balance of cortical networks is the ratio of excitatory and inhibitory neurons. The mechanisms controlling the ultimate number of interneurons are beginning to be elucidated, but to what extent similar principles govern the survival of the large diversity of cortical inhibitory cells remains to be investigated. Here, we investigate the mechanisms regulating developmental cell death in neurogliaform cells, bipolar cells, and basket cells, the three main populations of interneurons originating from the caudal ganglionic eminence and the preoptic region. We found that all three subclasses of interneurons undergo activity-dependent programmed cell death. However, while neurogliaform cells and basket cells require glutamatergic transmission to survive, the final number of bipolar cells is instead modulated by serotonergic signaling. Together, our results demonstrate that input-specific modulation of neuronal activity controls the survival of cortical interneurons during the critical period of programmed cell death.


Subject(s)
Cerebral Cortex , Interneurons , Apoptosis , Cell Survival , Neurons
8.
Proc Natl Acad Sci U S A ; 119(26): e2121868119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35727980

ABSTRACT

Proper orientation of the mitotic spindle plays a crucial role in embryos, during tissue development, and in adults, where it functions to dissipate mechanical stress to maintain tissue integrity and homeostasis. While mitotic spindles have been shown to reorient in response to external mechanical stresses, the subcellular cues that mediate spindle reorientation remain unclear. Here, we used a combination of optogenetics and computational modeling to investigate how mitotic spindles respond to inhomogeneous tension within the actomyosin cortex. Strikingly, we found that the optogenetic activation of RhoA only influences spindle orientation when it is induced at both poles of the cell. Under these conditions, the sudden local increase in cortical tension induced by RhoA activation reduces pulling forces exerted by cortical regulators on astral microtubules. This leads to a perturbation of the balance of torques exerted on the spindle, which causes it to rotate. Thus, spindle rotation in response to mechanical stress is an emergent phenomenon arising from the interaction between the spindle positioning machinery and the cell cortex.


Subject(s)
Microtubules , Spindle Apparatus , Stress, Mechanical , Actomyosin/metabolism , Computer Simulation , Cytoplasm , Microtubules/metabolism , Optogenetics , Spindle Apparatus/physiology , rhoA GTP-Binding Protein/metabolism
9.
Proc Natl Acad Sci U S A ; 119(20): e2118430119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35533272

ABSTRACT

The assembly of functional neuronal circuits requires appropriate numbers of distinct classes of neurons, but the mechanisms through which their relative proportions are established remain poorly defined. Investigating the mouse striatum, we found that the two most prominent subtypes of striatal interneurons, parvalbumin-expressing (PV+) GABAergic and cholinergic (ChAT+) interneurons, undergo extensive programmed cell death between the first and second postnatal weeks. Remarkably, the survival of PV+ and ChAT+ interneurons is regulated by distinct mechanisms mediated by their specific afferent connectivity. While long-range cortical inputs control PV+ interneuron survival, ChAT+ interneuron survival is regulated by local input from the medium spiny neurons. Our results identify input-specific circuit mechanisms that operate during the period of programmed cell death to establish the final number of interneurons in nascent striatal networks.


Subject(s)
Corpus Striatum , Interneurons , Cerebral Cortex/physiology , Corpus Striatum/physiology , GABAergic Neurons/physiology , Interneurons/physiology , Parvalbumins
10.
Int J Mol Sci ; 23(4)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35216353

ABSTRACT

X-linked dystonia-parkinsonism (XDP) is a severe neurodegenerative disorder that manifests as adult-onset dystonia combined with parkinsonism. A SINE-VNTR-Alu (SVA) retrotransposon inserted in an intron of the TAF1 gene reduces its expression and alters splicing in XDP patient-derived cells. As a consequence, increased levels of the TAF1 intron retention transcript TAF1-32i can be found in XDP cells as compared to healthy controls. Here, we investigate the sequence of the deep intronic region included in this transcript and show that it is also present in cells from healthy individuals, albeit in lower amounts than in XDP cells, and that it undergoes degradation by nonsense-mediated mRNA decay. Furthermore, we investigate epigenetic marks (e.g., DNA methylation and histone modifications) present in this intronic region and the spanning sequence. Finally, we show that the SVA evinces regulatory potential, as demonstrated by its ability to repress the TAF1 promoter in vitro. Our results enable a better understanding of the disease mechanisms underlying XDP and transcriptional alterations caused by SVA retrotransposons.


Subject(s)
Dystonic Disorders/genetics , Genetic Diseases, X-Linked/genetics , Parkinsonian Disorders/genetics , Retroelements/genetics , Transcription, Genetic/genetics , Adolescent , Adult , DNA Methylation/genetics , Female , Histone Acetyltransferases/genetics , Humans , Introns/genetics , Male , Middle Aged , Promoter Regions, Genetic/genetics , Short Interspersed Nucleotide Elements/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Young Adult
11.
Am J Hum Genet ; 108(9): 1725-1734, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34433009

ABSTRACT

Copy-number variations (CNVs) are a common cause of congenital limb malformations and are interpreted primarily on the basis of their effect on gene dosage. However, recent studies show that CNVs also influence the 3D genome chromatin organization. The functional interpretation of whether a phenotype is the result of gene dosage or a regulatory position effect remains challenging. Here, we report on two unrelated families with individuals affected by bilateral hypoplasia of the femoral bones, both harboring de novo duplications on chromosome 10q24.32. The ∼0.5 Mb duplications include FGF8, a key regulator of limb development and several limb enhancer elements. To functionally characterize these variants, we analyzed the local chromatin architecture in the affected individuals' cells and re-engineered the duplications in mice by using CRISPR-Cas9 genome editing. We found that the duplications were associated with ectopic chromatin contacts and increased FGF8 expression. Transgenic mice carrying the heterozygous tandem duplication including Fgf8 exhibited proximal shortening of the limbs, resembling the human phenotype. To evaluate whether the phenotype was a result of gene dosage, we generated another transgenic mice line, carrying the duplication on one allele and a concurrent Fgf8 deletion on the other allele, as a control. Surprisingly, the same malformations were observed. Capture Hi-C experiments revealed ectopic interaction with the duplicated region and Fgf8, indicating a position effect. In summary, we show that duplications at the FGF8 locus are associated with femoral hypoplasia and that the phenotype is most likely the result of position effects altering FGF8 expression rather than gene dosage effects.


Subject(s)
Chromosome Duplication , Chromosomes, Human, Pair 10/chemistry , DNA Copy Number Variations , Fibroblast Growth Factor 8/genetics , Lower Extremity Deformities, Congenital/genetics , Adolescent , Alleles , Animals , CRISPR-Cas Systems , Child, Preschool , Chromatin/chemistry , Chromatin/metabolism , Chromosomes, Human, Pair 10/metabolism , Enhancer Elements, Genetic , Family , Female , Femur/abnormalities , Femur/diagnostic imaging , Femur/metabolism , Fibroblast Growth Factor 8/metabolism , Gene Editing , Heterozygote , Humans , Infant , Lower Extremity Deformities, Congenital/diagnostic imaging , Lower Extremity Deformities, Congenital/metabolism , Lower Extremity Deformities, Congenital/pathology , Male , Mice , Mice, Transgenic , Pedigree , Phenotype
12.
ACS Sens ; 6(3): 1375-1383, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33660984

ABSTRACT

To address the growing demand for simultaneous imaging of multiple biomarkers in highly scattering media such as organotypic cell cultures, we introduce a new type of photoluminescent nanomaterial termed "tau-ruby" composed of ruby nanocrystals (Al2O3:Cr3+) with tunable emission lifetime. The lifetime tuning range from 2.4 to 3.2 ms was achieved by varying the Cr3+ dopant concentration from 0.8% to 0.2%, affording facile implementation of background-free detection. We developed inexpensive scalable production of tau-ruby characterized by bright emission, narrow spectrum (693 ± 2 nm), and virtually unlimited photostability upon excitation with affordable excitation/detection sources, noncytotoxic and insensitive to microenvironmental fluctuations. By functionalizing the surface of tau-rubies with targeting antibodies, we obtained different biomarkers suitable for multiplexed lifetime imaging. As a proof of principle, three tau-ruby bioprobes, characterized by three mean lifetimes, were deployed to label three µ-opioid receptor species expressed on transfected cancer cells, each fused to a unique epitope, so that three types of cells were lifetime-encoded. Robust decoding of photoluminescent signals that report on each cell type was achieved by using a home-built lifetime imaging system and resulted in high-contrast multiplexed lifetime imaging of the cells.


Subject(s)
Biosensing Techniques , Nanoparticles , Nanostructures , Diagnostic Imaging
13.
Methods Mol Biol ; 2201: 59-70, 2021.
Article in English | MEDLINE | ID: mdl-32975789

ABSTRACT

Sensitive and long-term fluorescence imaging of G-protein-coupled receptors enables exploration of molecular level details of these therapeutically relevant proteins, including their expression, localization, signaling, and intracellular trafficking. In this context, labeling these receptors with bright and photostable fluorescent probes is necessary to overcome current imaging problems such as optical background and photobleaching. Here, we describe the procedures to functionalize nanoruby (and other similar nanoparticles) with NeutrAvidin (a streptavidin analog) and to apply this bioconjugate for ultrasensitive, long-term imaging of µ-opioid receptors heterologously expressed in AtT-20 cells. The receptor targeting is mediated via a biotinylated primary antibody, rendering this methodology extendable to other G-protein-coupled or, more generally, cell-surface receptors. Nanoruby-based time-gated imaging enables indefinitely long visualization of single particles even in high-autofluorescence media, such as serum, by completely suppressing autofluorescence and any laser backscatter.


Subject(s)
Avidin/chemistry , Diagnostic Imaging/methods , Receptors, Opioid, mu/metabolism , Biotinylation/methods , Fluorescence , Fluorescent Dyes , Humans , Nanoparticles , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid/metabolism , Streptavidin
14.
Biomed Opt Express ; 11(11): 6137-6153, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-33282480

ABSTRACT

Protein-protein interactions at the plasma membrane mediate transmembrane signaling. Dual-channel fluorescence cross-correlation spectroscopy (dc-FCCS) is a method with which these interactions can be quantified in a cellular context. However, factors such as incomplete maturation of fluorescent proteins, spectral crosstalk, and fluorescence resonance energy transfer (FRET) affect quantification. Some of these can be corrected or accounted for during data analysis and/or interpretation. Here, we experimentally and analytically demonstrate that it is difficult to correct the error caused due to FRET when applying dc-FCCS to measure binding affinity or bound molecular concentrations. Additionally, the presence of dark fluorescent proteins due to incomplete maturation introduces further errors, which too cannot be corrected in the presence of FRET. Based on simulations, we find that modalities such as pulse-interleaved excitation FCCS do not eliminate FRET-induced errors. Finally, we demonstrate that the detrimental effect of FRET can be eliminated with careful experimental design when applying dc-FCCS to quantify protein-protein interactions at the plasma membrane of living cells.

15.
Mol Pharmacol ; 98(4): 410-424, 2020 10.
Article in English | MEDLINE | ID: mdl-32665252

ABSTRACT

Evidence from several novel opioid agonists and knockout animals suggests that improved opioid therapeutic window, notably for analgesia versus respiratory depression, is a result of ligand bias downstream of activation of the µ-opioid receptor (MOR) toward G protein signaling and away from other pathways, such as arrestin recruitment. Here, we argue that published claims of opioid bias based on application of the operational model of agonism are frequently confounded by failure to consider the assumptions of the model. These include failure to account for intrinsic efficacy and ceiling effects in different pathways, distortions introduced by analysis of amplified (G protein) versus linear (arrestin) signaling mechanisms, and nonequilibrium effects in a dynamic signaling cascade. We show on both theoretical and experimental grounds that reduced intrinsic efficacy that is unbiased across different downstream pathways, when analyzed without due considerations, does produce apparent but erroneous MOR ligand bias toward G protein signaling, and the weaker the G protein partial agonism is the greater the apparent bias. Experimentally, such apparently G protein-biased opioids have been shown to exhibit low intrinsic efficacy for G protein signaling when ceiling effects are properly accounted for. Nevertheless, such agonists do display an improved therapeutic window for analgesia versus respiratory depression. Reduced intrinsic efficacy for G proteins rather than any supposed G protein bias provides a more plausible, sufficient explanation for the improved safety. Moreover, genetic models of G protein-biased opioid receptors and replication of previous knockout experiments suggest that reduced or abolished arrestin recruitment does not improve therapeutic window for MOR-induced analgesia versus respiratory depression. SIGNIFICANCE STATEMENT: Efforts to improve safety of µ-opioid analgesics have focused on agonists that show signaling bias for the G protein pathway versus other signaling pathways. This review provides theoretical and experimental evidence showing that failure to consider the assumptions of the operational model can lead to large distortions and overestimation of actual bias. We show that low intrinsic efficacy is a major determinant of these distortions, and pursuit of appropriately reduced intrinsic efficacy should guide development of safer opioids.


Subject(s)
Analgesics, Opioid/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Opioid, mu/metabolism , Animals , Humans , Ligands , Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects
16.
Nature ; 557(7707): 668-673, 2018 05.
Article in English | MEDLINE | ID: mdl-29849154

ABSTRACT

Complex neuronal circuitries such as those found in the mammalian cerebral cortex have evolved as balanced networks of excitatory and inhibitory neurons. Although the establishment of appropriate numbers of these cells is essential for brain function and behaviour, our understanding of this fundamental process is limited. Here we show that the survival of interneurons in mice depends on the activity of pyramidal cells in a critical window of postnatal development, during which excitatory synaptic input to individual interneurons predicts their survival or death. Pyramidal cells regulate interneuron survival through the negative modulation of PTEN signalling, which effectively drives interneuron cell death during this period. Our findings indicate that activity-dependent mechanisms dynamically adjust the number of inhibitory cells in nascent local cortical circuits, ultimately establishing the appropriate proportions of excitatory and inhibitory neurons in the cerebral cortex.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/physiology , Interneurons/cytology , Neural Pathways , Pyramidal Cells/physiology , Animals , Cell Count , Cell Death , Cell Survival , Female , Interneurons/physiology , Male , Mice , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism
17.
Biophys J ; 114(9): 2231-2242, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29742416

ABSTRACT

Cell volume regulation is fundamentally important in phenomena such as cell growth, proliferation, tissue homeostasis, and embryogenesis. How the cell size is set, maintained, and changed over a cell's lifetime is not well understood. In this work we focus on how the volume of nonexcitable tissue cells is coupled to the cell membrane electrical potential and the concentrations of membrane-permeable ions in the cell environment. Specifically, we demonstrate that a sudden cell depolarization using the whole-cell patch clamp results in a 50% increase in cell volume, whereas hyperpolarization results in a slight volume decrease. We find that cell volume can be partially controlled by changing the chloride or the sodium/potassium concentrations in the extracellular environment while maintaining a constant external osmotic pressure. Depletion of external chloride leads to a volume decrease in suspended HN31 cells. Introducing cells to a high-potassium solution causes volume increase up to 50%. Cell volume is also influenced by cortical tension: actin depolymerization leads to cell volume increase. We present an electrophysiology model of water dynamics driven by changes in membrane potential and the concentrations of permeable ions in the cells surrounding. The model quantitatively predicts that the cell volume is directly proportional to the intracellular protein content.


Subject(s)
Cell Size , Electrophysiological Phenomena , Actins/chemistry , Cell Line, Tumor , Chlorides/metabolism , Extracellular Space/metabolism , Humans , Intracellular Space/metabolism , Potassium/metabolism , Protein Multimerization , Protein Structure, Quaternary , Sodium/metabolism
18.
Ann Palliat Med ; 7(3): 304-319, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29156918

ABSTRACT

In the recent years, there has been an increase in awareness with regards to the role of palliative care (PC) in management of neurologic diseases. In 1996, the need to incorporate PC in the care for patients with neurologic conditions was recognized by the American Academy of Neurology (AAN) Ethics and Humanities Subcommittee. The gaps in research, education and the ability to deliver adequate PC were then acknowledged by the National Academy of Sciences with their publication of "Approaching death: improving care at the end of life" and most recently, continued goals in improving PC was highlighted by another recent publication "Dying in America: improving quality and honoring individual preferences near the end of life". The complexity of managing neurologic patients brings about challenges and ethical issues in this setting. The aim of this review is to discuss and summarize the challenges and ethical issues in the context of PC management of patients with advanced acute, rapidly progressive, slowly-progressive or degenerative neurological conditions that are commonly encountered in practice.


Subject(s)
Nervous System Diseases/therapy , Palliative Care/ethics , Brain Injuries/therapy , Decision Making , Ethics , Humans , Palliative Care/methods , Time Factors , Tissue and Organ Procurement , Withholding Treatment
19.
ACS Appl Mater Interfaces ; 9(45): 39197-39208, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29022702

ABSTRACT

At the forefront of developing fluorescent probes for biological imaging applications are enhancements aimed at increasing their brightness, contrast, and photostability, especially toward demanding applications of single-molecule detection. In comparison with existing probes, nanorubies exhibit unlimited photostability and a long emission lifetime (∼4 ms), which enable continuous imaging at single-particle sensitivity in highly scattering and fluorescent biological specimens. However, their wide application as fluorescence probes has so far been hindered by the absence of facile methods for scaled-up high-volume production and molecularly specific targeting. The present work encompasses the large-scale production of colloidally stable nanoruby particles, the demonstration of their biofunctionality and negligible cytotoxicity, as well as the validation of its use for targeted biomolecular imaging. In addition, optical characteristics of nanorubies are found to be comparable or superior to those of state-of-the-art quantum dots. Protocols of reproducible and robust coupling of functional proteins to the nanoruby surface are also presented. As an example, NeutrAvidin-coupled nanoruby show excellent affinity and specificity to µ-opioid receptors in fixed and live cells, allowing wide-field imaging of G-protein coupled receptors with single-particle sensitivity.


Subject(s)
Nanostructures , Biocompatible Materials , Fluorescent Dyes , Quantum Dots , Receptors, G-Protein-Coupled , Time Factors
20.
PLoS One ; 12(9): e0185393, 2017.
Article in English | MEDLINE | ID: mdl-28949989

ABSTRACT

Focussed radiosurgery may provide a means of inducing molecular changes on the luminal surface of diseased endothelium to allow targeted delivery of novel therapeutic compounds. We investigated the potential of ionizing radiation to induce surface expression of intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) on endothelial cells (EC) in vitro and in vivo, to assess their suitability as vascular targets in irradiated arteriovenous malformations (AVMs). Cultured brain microvascular EC were irradiated by linear accelerator at single doses of 0, 5, 15 or 25 Gy and expression of ICAM-1 and VCAM-1 measured by qRT-PCR, Western, ELISA and immunocytochemistry. In vivo, near-infrared (NIR) fluorescence optical imaging using Xenolight 750-conjugated ICAM-1 or VCAM-1 antibodies examined luminal biodistribution over 84 days in a rat AVM model after Gamma Knife surgery at a single 15 Gy dose. ICAM-1 and VCAM-1 were minimally expressed on untreated EC in vitro. Doses of 15 and 25 Gy stimulated expression equally; 5 Gy was not different from the unirradiated. In vivo, normal vessels did not bind or retain the fluorescent probes, however binding was significant in AVM vessels. No additive increases in probe binding were found in response to radiosurgery at a dose of 15 Gy. In summary, radiation induces adhesion molecule expression in vitro but elevated baseline levels in AVM vessels precludes further induction in vivo. These molecules may be suitable targets in irradiated vessels without hemodynamic derangement, but not AVMs. These findings demonstrate the importance of using flow-modulated, pre-clinical animal models for validating candidate proteins for vascular targeting in irradiated AVMs.


Subject(s)
Disease Models, Animal , Intercellular Adhesion Molecule-1/metabolism , Intracranial Arteriovenous Malformations/metabolism , Intracranial Arteriovenous Malformations/surgery , Radiosurgery/methods , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Male , Mice , Radiotherapy Dosage , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...