Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Nature ; 624(7991): 355-365, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38092919

ABSTRACT

Single-cell analyses parse the brain's billions of neurons into thousands of 'cell-type' clusters residing in different brain structures1. Many cell types mediate their functions through targeted long-distance projections allowing interactions between specific cell types. Here we used epi-retro-seq2 to link single-cell epigenomes and cell types to long-distance projections for 33,034 neurons dissected from 32 different regions projecting to 24 different targets (225 source-to-target combinations) across the whole mouse brain. We highlight uses of these data for interrogating principles relating projection types to transcriptomics and epigenomics, and for addressing hypotheses about cell types and connections related to genetics. We provide an overall synthesis with 926 statistical comparisons of discriminability of neurons projecting to each target for every source. We integrate this dataset into the larger BRAIN Initiative Cell Census Network atlas, composed of millions of neurons, to link projection cell types to consensus clusters. Integration with spatial transcriptomics further assigns projection-enriched clusters to smaller source regions than the original dissections. We exemplify this by presenting in-depth analyses of projection neurons from the hypothalamus, thalamus, hindbrain, amygdala and midbrain to provide insights into properties of those cell types, including differentially expressed genes, their associated cis-regulatory elements and transcription-factor-binding motifs, and neurotransmitter use.


Subject(s)
Brain , Epigenomics , Neural Pathways , Neurons , Animals , Mice , Amygdala , Brain/cytology , Brain/metabolism , Consensus Sequence , Datasets as Topic , Gene Expression Profiling , Hypothalamus/cytology , Mesencephalon/cytology , Neural Pathways/cytology , Neurons/metabolism , Neurotransmitter Agents/metabolism , Regulatory Sequences, Nucleic Acid , Rhombencephalon/cytology , Single-Cell Analysis , Thalamus/cytology , Transcription Factors/metabolism
2.
Cell Rep ; 40(7): 111222, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977501

ABSTRACT

Perception of threats is essential for survival. Previous findings suggest that parallel pathways independently relay innate threat signals from different sensory modalities to multiple brain areas, such as the midbrain and hypothalamus, for immediate avoidance. Yet little is known about whether and how multi-sensory innate threat cues are integrated and conveyed from each sensory modality to the amygdala, a critical brain area for threat perception and learning. Here, we report that neurons expressing calcitonin gene-related peptide (CGRP) in the parvocellular subparafascicular nucleus in the thalamus and external lateral parabrachial nucleus in the brainstem respond to multi-sensory threat cues from various sensory modalities and relay negative valence to the lateral and central amygdala, respectively. Both CGRP populations and their amygdala projections are required for multi-sensory threat perception and aversive memory formation. The identification of unified innate threat pathways may provide insights into developing therapeutic candidates for innate fear-related disorders.


Subject(s)
Central Amygdaloid Nucleus , Parabrachial Nucleus , Calcitonin Gene-Related Peptide/metabolism , Central Amygdaloid Nucleus/metabolism , Cues , Parabrachial Nucleus/metabolism , Thalamus/metabolism
3.
Neuron ; 110(8): 1318-1326.e4, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35108498

ABSTRACT

We tested whether social signal processing in more traditional, head-restrained contexts is representative of the putative natural analog-social communication-by comparing responses to vocalizations within individual neurons in marmoset prefrontal cortex (PFC) across a series of behavioral contexts ranging from traditional to naturalistic. Although vocalization-responsive neurons were evident in all contexts, cross-context consistency was notably limited. A response to these social signals when subjects were head-restrained was not predictive of a comparable neural response to the identical vocalizations during natural communication. This pattern was evident both within individual neurons and at a population level, as PFC activity could be reliably decoded for the behavioral context in which vocalizations were heard. These results suggest that neural representations of social signals in primate PFC are not static but highly flexible and likely reflect how nuances of the dynamic behavioral contexts affect the perception of these signals and what they communicate.


Subject(s)
Prefrontal Cortex , Vocalization, Animal , Animals , Callithrix , Humans , Neurons/physiology , Prefrontal Cortex/physiology , Vocalization, Animal/physiology
4.
Nature ; 598(7879): 167-173, 2021 10.
Article in English | MEDLINE | ID: mdl-34616065

ABSTRACT

Neuronal cell types are classically defined by their molecular properties, anatomy and functions. Although recent advances in single-cell genomics have led to high-resolution molecular characterization of cell type diversity in the brain1, neuronal cell types are often studied out of the context of their anatomical properties. To improve our understanding of the relationship between molecular and anatomical features that define cortical neurons, here we combined retrograde labelling with single-nucleus DNA methylation sequencing to link neural epigenomic properties to projections. We examined 11,827 single neocortical neurons from 63 cortico-cortical and cortico-subcortical long-distance projections. Our results showed unique epigenetic signatures of projection neurons that correspond to their laminar and regional location and projection patterns. On the basis of their epigenomes, intra-telencephalic cells that project to different cortical targets could be further distinguished, and some layer 5 neurons that project to extra-telencephalic targets (L5 ET) formed separate clusters that aligned with their axonal projections. Such separation varied between cortical areas, which suggests that there are area-specific differences in L5 ET subtypes, which were further validated by anatomical studies. Notably, a population of cortico-cortical projection neurons clustered with L5 ET rather than intra-telencephalic neurons, which suggests that a population of L5 ET cortical neurons projects to both targets. We verified the existence of these neurons by dual retrograde labelling and anterograde tracing of cortico-cortical projection neurons, which revealed axon terminals in extra-telencephalic targets including the thalamus, superior colliculus and pons. These findings highlight the power of single-cell epigenomic approaches to connect the molecular properties of neurons with their anatomical and projection properties.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Epigenome , Epigenomics , Neural Pathways , Neurons/classification , Neurons/metabolism , Animals , Brain Mapping , Female , Male , Mice , Neurons/cytology
5.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34074761

ABSTRACT

Opioid-induced respiratory depression (OIRD) causes death following an opioid overdose, yet the neurobiological mechanisms of this process are not well understood. Here, we show that neurons within the lateral parabrachial nucleus that express the µ-opioid receptor (PBL Oprm1 neurons) are involved in OIRD pathogenesis. PBL Oprm1 neuronal activity is tightly correlated with respiratory rate, and this correlation is abolished following morphine injection. Chemogenetic inactivation of PBL Oprm1 neurons mimics OIRD in mice, whereas their chemogenetic activation following morphine injection rescues respiratory rhythms to baseline levels. We identified several excitatory G protein-coupled receptors expressed by PBL Oprm1 neurons and show that agonists for these receptors restore breathing rates in mice experiencing OIRD. Thus, PBL Oprm1 neurons are critical for OIRD pathogenesis, providing a promising therapeutic target for treating OIRD in patients.


Subject(s)
Analgesics, Opioid/adverse effects , Morphine/adverse effects , Neurons/metabolism , Receptors, Opioid, mu/metabolism , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/metabolism , Analgesics, Opioid/pharmacology , Animals , Mice , Mice, Transgenic , Morphine/administration & dosage , Morphine/pharmacology , Neurons/pathology , Receptors, Opioid, mu/genetics , Respiratory Insufficiency/genetics , Respiratory Insufficiency/pathology
6.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33372152

ABSTRACT

Defense of the central nervous system (CNS) against infection must be accomplished without generation of potentially injurious immune cell-mediated or off-target inflammation which could impair key functions. As the CNS is an immune-privileged compartment, inducible innate defense mechanisms endogenous to the CNS likely play an essential role in this regard. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide known to regulate neurodevelopment, emotion, and certain stress responses. While PACAP is known to interact with the immune system, its significance in direct defense of brain or other tissues is not established. Here, we show that our machine-learning classifier can screen for immune activity in neuropeptides, and correctly identified PACAP as an antimicrobial neuropeptide in agreement with previous experimental work. Furthermore, synchrotron X-ray scattering, antimicrobial assays, and mechanistic fingerprinting provided precise insights into how PACAP exerts antimicrobial activities vs. pathogens via multiple and synergistic mechanisms, including dysregulation of membrane integrity and energetics and activation of cell death pathways. Importantly, resident PACAP is selectively induced up to 50-fold in the brain in mouse models of Staphylococcus aureus or Candida albicans infection in vivo, without inducing immune cell infiltration. We show differential PACAP induction even in various tissues outside the CNS, and how these observed patterns of induction are consistent with the antimicrobial efficacy of PACAP measured in conditions simulating specific physiologic contexts of those tissues. Phylogenetic analysis of PACAP revealed close conservation of predicted antimicrobial properties spanning primitive invertebrates to modern mammals. Together, these findings substantiate our hypothesis that PACAP is an ancient neuro-endocrine-immune effector that defends the CNS against infection while minimizing potentially injurious neuroinflammation.


Subject(s)
Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Amino Acid Sequence/genetics , Animals , Anti-Infective Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Brain/immunology , Brain/metabolism , Cell Death/drug effects , Computer Simulation , Databases, Genetic , Inflammation/metabolism , Mice , Mice, Inbred BALB C , Neuropeptides/metabolism , Phylogeny , Signal Transduction/physiology
7.
PLoS Genet ; 15(3): e1007948, 2019 03.
Article in English | MEDLINE | ID: mdl-30870413

ABSTRACT

Glial cells regulate multiple aspects of synaptogenesis. In the absence of Schwann cells, a peripheral glial cell, motor neurons initially innervate muscle but then degenerate. Here, using a genetic approach, we show that neural activity-regulated negative factors produced by muscle drive neurodegeneration in Schwann cell-deficient mice. We find that thrombin, the hepatic serine protease central to the hemostatic coagulation cascade, is one such negative factor. Trancriptomic analysis shows that expression of the antithrombins serpin C1 and D1 is significantly reduced in Schwann cell-deficient mice. In the absence of peripheral neuromuscular activity, neurodegeneration is completely blocked, and expression of prothrombin in muscle is markedly reduced. In the absence of muscle-derived prothrombin, neurodegeneration is also markedly reduced. Together, these results suggest that Schwann cells regulate NMJs by opposing the effects of activity-regulated, muscle-derived negative factors and provide the first genetic evidence that thrombin plays a central role outside of the coagulation system.


Subject(s)
Antithrombin III/genetics , Heparin Cofactor II/genetics , Neuromuscular Junction/genetics , Prothrombin/genetics , Synapses/genetics , Animals , Gene Expression Profiling , Mice , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Nerve Degeneration/genetics , Neuroglia , Neuromuscular Junction/growth & development , Schwann Cells/metabolism , Thrombin/genetics
8.
PLoS Genet ; 15(3): e1007857, 2019 03.
Article in English | MEDLINE | ID: mdl-30870432

ABSTRACT

Schwann cells are integral components of vertebrate neuromuscular synapses; in their absence, pre-synaptic nerve terminals withdraw from post-synaptic muscles, leading to muscle denervation and synapse loss at the developing neuromuscular junction (NMJ). Here, we report a rescue of muscle denervation and neuromuscular synapses loss in type III Neuregulin 1 mutant mice (CRD-Nrg1-/-), which lack Schwann cells. We found that muscle denervation and neuromuscular synapse loss were prevented in CRD-Nrg1-/-mice when presynaptic activity was blocked by ablating a specific gene, such as Snap25 (synaptosomal-associated 25 kDa protein) or Chat (choline acetyltransferase). Further, these effects were mediated by a pathway that requires postsynaptic acetylcholine receptors (AChRs), because ablating Chrna1 (acetylcholine receptor α1 subunit), which encodes muscle-specific AChRs in CRD-Nrg1-/-mice also rescued muscle denervation. Moreover, genetically ablating muscle dihydropyridine receptor (DHPR) ß1 subunit (Cacnb1) or ryanodine receptor 1 (Ryr1) also rescued muscle denervation and neuromuscular synapse loss in CRD-Nrg1-/-mice. Thus, these genetic manipulations follow a pathway-from presynaptic to postsynaptic, and, ultimately to muscle activity mediated by DHPRs and Ryr1. Importantly, electrophysiological analyses reveal robust synaptic activity in the rescued, Schwann-cell deficient NMJs in CRD-Nrg1-/-Cacnb1-/-or CRD-Nrg1-/-Ryr1-/-mutant mice. Thus, a blockade of synaptic activity, although sufficient, is not necessary to preserve NMJs that lack Schwann cells. Instead, a blockade of muscle activity mediated by DHRPs and Ryr1 is both necessary and sufficient for preserving NMJs that lack Schwann cells. These findings suggest that muscle activity mediated by DHPRs/Ryr1 may destabilize developing NMJs and that Schwann cells play crucial roles in counteracting such a destabilizing activity to preserve neuromuscular synapses during development.


Subject(s)
Calcium Channels, L-Type/genetics , Neuregulin-1/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Synapses/genetics , Animals , Axons/metabolism , Electrophysiology , Humans , Mice , Motor Neurons/metabolism , Muscle Denervation , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Nerve Regeneration/genetics , Neuromuscular Junction/genetics , Presynaptic Terminals/metabolism , Receptors, Nicotinic/genetics , Schwann Cells/metabolism , Synapses/physiology , Synaptosomal-Associated Protein 25/genetics
9.
Proc Natl Acad Sci U S A ; 115(23): E5289-E5297, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29784793

ABSTRACT

In response to cold exposure, placental mammals maintain body temperature by increasing sympathetic nerve activity in brown adipose tissue (BAT). Triggering of ß-adrenergic receptors on brown adipocytes stimulates thermogenesis via induction of the cAMP/PKA pathway. Although cAMP response element-binding protein (CREB) and its coactivators-the cAMP-regulated transcriptional coactivators (CRTCs)-mediate transcriptional effects of cAMP in most tissues, other transcription factors such as ATF2 appear critical for induction of thermogenic genes by cAMP in BAT. Brown adipocytes arise from Myf5-positive mesenchymal cells under the control of PRDM16, a coactivator that concurrently represses differentiation along the skeletal muscle lineage. Here, we show that the CREB coactivator CRTC3 is part of an inhibitory feedback pathway that antagonizes PRDM16-dependent differentiation. Mice with a knockout of CRTC3 in BAT (BKO) have increased cold tolerance and reduced adiposity, whereas mice overexpressing constitutively active CRTC3 in adipose tissue are more cold sensitive and have greater fat mass. CRTC3 reduced sympathetic nerve activity in BAT by up-regulating the expression of miR-206, a microRNA that promotes differentiation along the myogenic lineage and that we show here decreases the expression of VEGFA and neurotrophins critical for BAT innervation and vascularization. Sympathetic nerve activity to BAT was enhanced in BKO mice, leading to increases in catecholamine signaling that stimulated energy expenditure. As reexpression of miR-206 in BAT from BKO mice reversed the salutary effects of CRTC3 depletion on cold tolerance, our studies suggest that small-molecule inhibitors against this coactivator may provide therapeutic benefit to overweight individuals.


Subject(s)
Adipose Tissue, Brown/metabolism , Thermogenesis/physiology , Transcription Factors/metabolism , Adipocytes, Brown/metabolism , Adiposity/genetics , Adiposity/physiology , Animals , Cell Differentiation/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Energy Metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , Signal Transduction , Sympathetic Nervous System/metabolism , Transcription Factors/genetics
10.
Cell Rep ; 21(3): 707-720, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29045838

ABSTRACT

Producing the neuronal diversity required to adequately discriminate all elements of somatosensation is a complex task during organogenesis. The mechanisms guiding this process during dorsal root ganglion (DRG) sensory neuron specification remain poorly understood. Here, we show that the p75 neurotrophin receptor interacts with Ret and its GFRα co-receptor upon stimulation with glial cell line-derived neurotrophic factor (GDNF). Furthermore, we demonstrate that p75 is required for GDNF-mediated Ret activation, survival, and cell surface localization of Ret in DRG neurons. In mice in which p75 is deleted specifically within sensory neurons beginning at E12.5, we observe that approximately 20% of neurons are lost between P14 and adulthood, and these losses selectively occur within a subpopulation of Ret+ nonpeptidergic nociceptors, with neurons expressing low levels of Ret impacted most heavily. These results suggest that p75 is required for the development of the nonpeptidergic nociceptor lineage by fine-tuning Ret-mediated trophic support.


Subject(s)
Proto-Oncogene Proteins c-ret/metabolism , Receptor, Nerve Growth Factor/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction , Animals , Animals, Newborn , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Core Binding Factor Alpha 2 Subunit/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Integrases/metabolism , Ligands , Mice , Nociception/drug effects , Nociceptors/drug effects , Nociceptors/metabolism , Peptides/metabolism
11.
J Clin Invest ; 127(2): 569-582, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-28067668

ABSTRACT

Failure of trabecular myocytes to undergo appropriate cell cycle withdrawal leads to ventricular noncompaction and heart failure. Signaling of growth factor receptor ERBB2 is critical for myocyte proliferation and trabeculation. However, the mechanisms underlying appropriate downregulation of trabecular ERBB2 signaling are little understood. Here, we have found that the endocytic adaptor proteins NUMB and NUMBL were required for downregulation of ERBB2 signaling in maturing trabeculae. Loss of NUMB and NUMBL resulted in a partial block of late endosome formation, resulting in sustained ERBB2 signaling and STAT5 activation. Unexpectedly, activated STAT5 overrode Hippo-mediated inhibition and drove YAP1 to the nucleus. Consequent aberrant cardiomyocyte proliferation resulted in ventricular noncompaction that was markedly rescued by heterozygous loss of function of either ERBB2 or YAP1. Further investigations revealed that NUMB and NUMBL interacted with small GTPase Rab7 to transition ERBB2 from early to late endosome for degradation. Our studies provide insight into mechanisms by which NUMB and NUMBL promote cardiomyocyte cell cycle withdrawal and highlight previously unsuspected connections between pathways that are important for cardiomyocyte cell cycle reentry, with relevance to ventricular noncompaction cardiomyopathy and regenerative medicine.


Subject(s)
Cell Cycle , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proteolysis , Receptor, ErbB-2/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Cycle Proteins , Endosomes/genetics , Endosomes/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nerve Tissue Proteins/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptor, ErbB-2/genetics , Signal Transduction/genetics , Transcription Factors , YAP-Signaling Proteins , rab GTP-Binding Proteins , rab7 GTP-Binding Proteins
12.
Sci Rep ; 6: 31692, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27558862

ABSTRACT

Several lines of evidence suggest that neuregulin 1 (NRG1) signaling may influence cognitive function and neuropathology in Alzheimer's disease (AD). To test this possibility, full-length type I or type III NRG1 was overexpressed via lentiviral vectors in the hippocampus of line 41 AD mouse. Both type I and type III NRG1 improves deficits in the Morris water-maze behavioral task. Neuropathology was also significantly ameliorated. Decreased expression of the neuronal marker MAP2 and synaptic markers PSD95 and synaptophysin in AD mice was significantly reversed. Levels of Aß peptides and plaques were markedly reduced. Furthermore, we showed that soluble ectodomains of both type I and type III NRG1 significantly increased expression of Aß-degrading enzyme neprilysin (NEP) in primary neuronal cultures. Consistent with this finding, immunoreactivity of NEP was increased in the hippocampus of AD mice. These results suggest that NRG1 provides beneficial effects in candidate neuropathologic substrates of AD and, therefore, is a potential target for the treatment of AD.


Subject(s)
Alzheimer Disease/therapy , Neuregulin-1/physiology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured , Cognition , Female , Hippocampus/pathology , Humans , Maze Learning , Mice, Transgenic , Neprilysin/metabolism , Rats
13.
J Am Chem Soc ; 138(30): 9473-8, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27379525

ABSTRACT

Apolipoprotein E (ApoE) belongs to a large class of proteins that solubilize lipids for physiological transport. Humans have three different APOE alleles, APOE ε2, APOE ε3, and APOE ε4, and genetic studies identified ApoE4 as the strongest genetic risk factor for Alzheimer's disease (AD). People who are homozygous for ApoE4 (i.e., ApoE4/E4) are an order of magnitude more likely to develop late-onset AD (LOAD) than ApoE3/E3 carriers. Several differences between ApoE3 and ApoE4 may contribute to AD including the observation that ApoE4 is degraded to a greater extent than ApoE3 in the human brain. Experiments with high-temperature requirement serine peptidase A1 (HtrA1), which is found in the nervous system, demonstrate that HtrA1 is an allele-selective ApoE-degrading enzyme that degrades ApoE4 more quickly than ApoE3. This activity is specific to HtrA1, as similar assays with HtrA2 showed minimal ApoE4 proteolysis and trypsin had no preference between ApoE4 and ApoE3. HtrA1 has also been reported to cleave the tau protein (Tau) and the amyloid protein precursor (APP) to hinder the formation of toxic amyloid deposits associated with AD. Competition assays with ApoE4, ApoE3, and Tau revealed that ApoE4 inhibits Tau degradation. Thus, the identification of ApoE4 as an in vitro HtrA1 substrate suggests a potential biochemical mechanism that links ApoE4 regulation of AD proteins such as Tau.


Subject(s)
Alleles , Apolipoproteins E/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Proteolysis , HEK293 Cells , High-Temperature Requirement A Serine Peptidase 1/chemistry , Humans , Models, Molecular , Protein Conformation
14.
Nanoscale ; 8(23): 11840-50, 2016 Jun 09.
Article in English | MEDLINE | ID: mdl-27228391

ABSTRACT

Composite colloidal structures with multi-functional properties have wide applications in targeted delivery of therapeutics and imaging contrast molecules and high-throughput molecular bio-sensing. We have constructed a multifunctional composite magnetic nanobowl using the bottom-up approach on an asymmetric silica/polystyrene Janus template consisting of a silica shell around a partially exposed polystyrene core. The nanobowl consists of a silica bowl and a gold exterior shell with iron oxide magnetic nanoparticles sandwiched between the silica and gold shells. The nanobowls were characterized by electron microscopy, atomic force microscopy, magnetometry, vis-NIR and FTIR spectroscopy. Magnetically vectored transport of these nanobowls was ascertained by time-lapsed imaging of their flow in fluid through a porous hydrogel under a defined magnetic field. These magnetically-responsive nanobowls show distinct surface enhanced Raman spectroscopy (SERS) imaging capability. The PEGylated magnetically-responsive nanobowls show size-dependent cellular uptake in vitro.


Subject(s)
Gold/chemistry , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Spectrum Analysis, Raman , Cell Line, Tumor , Humans , Polystyrenes
15.
Science ; 350(6267): 1525-9, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26680198

ABSTRACT

Dysfunction of microRNA (miRNA) metabolism is thought to underlie diseases affecting motoneurons. One miRNA, miR-218, is abundantly and selectively expressed by developing and mature motoneurons. Here we show that mutant mice lacking miR-218 die neonatally and exhibit neuromuscular junction defects, motoneuron hyperexcitability, and progressive motoneuron cell loss, all of which are hallmarks of motoneuron diseases such as amyotrophic lateral sclerosis and spinal muscular atrophy. Gene profiling reveals that miR-218 modestly represses a cohort of hundreds of genes that are neuronally enriched but are not specific to a single neuron subpopulation. Thus, the set of messenger RNAs targeted by miR-218, designated TARGET(218), defines a neuronal gene network that is selectively tuned down in motoneurons to prevent neuromuscular failure and neurodegeneration.


Subject(s)
Gene Expression Regulation , MicroRNAs/physiology , Motor Neuron Disease/genetics , Motor Neurons/physiology , Neurodegenerative Diseases/genetics , Animals , Gene Regulatory Networks , Mice , Mice, Knockout , MicroRNAs/genetics , Motor Neuron Disease/physiopathology , Motor Neurons/metabolism , Motor Neurons/pathology , Neurodegenerative Diseases/pathology , Spinal Cord/metabolism , Spinal Cord/physiopathology
16.
Neuron ; 86(3): 617-31, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25950631

ABSTRACT

One of the great strengths of the mouse model is the wide array of genetic tools that have been developed. Striking examples include methods for directed modification of the genome, and for regulated expression or inactivation of genes. Within neuroscience, it is now routine to express reporter genes, neuronal activity indicators, and opsins in specific neuronal types in the mouse. However, there are considerable anatomical, physiological, cognitive, and behavioral differences between the mouse and the human that, in some areas of inquiry, limit the degree to which insights derived from the mouse can be applied to understanding human neurobiology. Several recent advances have now brought into reach the goal of applying these tools to understanding the primate brain. Here we describe these advances, consider their potential to advance our understanding of the human brain and brain disorders, discuss bioethical considerations, and describe what will be needed to move forward.


Subject(s)
Brain/physiology , Genes , Primates/genetics , Animals , Biological Evolution , Humans , Mice , Models, Biological
17.
PLoS Biol ; 12(8): e1001918, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25093680

ABSTRACT

The p75 neurotrophin receptor, a member of the tumor necrosis factor receptor superfamily, is required as a co-receptor for the Nogo receptor (NgR) to mediate the activity of myelin-associated inhibitors such as Nogo, MAG, and OMgp. p45/NRH2/PLAIDD is a p75 homologue and contains a death domain (DD). Here we report that p45 markedly interferes with the function of p75 as a co-receptor for NgR. P45 forms heterodimers with p75 and thereby blocks RhoA activation and inhibition of neurite outgrowth induced by myelin-associated inhibitors. p45 binds p75 through both its transmembrane (TM) domain and DD. To understand the underlying mechanisms, we have determined the three-dimensional NMR solution structure of the intracellular domain of p45 and characterized its interaction with p75. We have identified the residues involved in such interaction by NMR and co-immunoprecipitation. The DD of p45 binds the DD of p75 by electrostatic interactions. In addition, previous reports suggested that Cys257 in the p75 TM domain is required for signaling. We found that the interaction of the cysteine 58 of p45 with the cysteine 257 of p75 within the TM domain is necessary for p45-p75 heterodimerization. These results suggest a mechanism involving both the TM domain and the DD of p45 to regulate p75-mediated signaling.


Subject(s)
Protein Multimerization , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/metabolism , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Cysteine/metabolism , HEK293 Cells , Humans , Magnetic Resonance Spectroscopy , Mice , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Interaction Mapping , Protein Stability , Receptors, Cell Surface/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Solutions , Structure-Activity Relationship , Up-Regulation
18.
PLoS Genet ; 10(6): e1004451, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24945252

ABSTRACT

Infertility and adverse gynecological outcomes such as preeclampsia and miscarriage represent significant female reproductive health concerns. The spatiotemporal expression of growth factors indicates that they play an important role in pregnancy. The goal of this study is to define the role of the ERBB family of growth factor receptors in endometrial function. Using conditional ablation in mice and siRNA in primary human endometrial stromal cells, we identified the epidermal growth factor receptor (Egfr) to be critical for endometrial function during early pregnancy. While ablation of Her2 or Erbb3 led to only a modest reduction in litter size, mice lacking Egfr expression are severely subfertile. Pregnancy demise occurred shortly after blastocyst implantation due to defects in decidualization including decreased proliferation, cell survival, differentiation and target gene expression. To place Egfr in a genetic regulatory hierarchy, transcriptome analyses was used to compare the gene signatures from mice with conditional ablation of Egfr, wingless-related MMTV integration site 4 (Wnt4) or boneless morphogenic protein 2 (Bmp2); revealing that not only are Bmp2 and Wnt4 key downstream effectors of Egfr, but they also regulate distinct physiological functions. In primary human endometrial stromal cells, marker gene expression, a novel high content image-based approach and phosphokinase array analysis were used to demonstrate that EGFR is a critical regulator of human decidualization. Furthermore, inhibition of EGFR signaling intermediaries WNK1 and AKT1S1, members identified in the kinase array and previously unreported to play a role in the endometrium, also attenuate decidualization. These results demonstrate that EGFR plays an integral role in establishing the cellular context necessary for successful pregnancy via the activation of intricate signaling and transcriptional networks, thereby providing valuable insight into potential therapeutic targets.


Subject(s)
Abortion, Spontaneous/genetics , ErbB Receptors/genetics , Fertility/genetics , Pregnancy Complications/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Morphogenetic Protein 2/genetics , Cell Differentiation/genetics , Decidua/metabolism , Endometriosis/genetics , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Minor Histocompatibility Antigens , Pregnancy , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Signal Transduction/genetics , WNK Lysine-Deficient Protein Kinase 1 , Wnt4 Protein/genetics
19.
PLoS One ; 8(7): e69286, 2013.
Article in English | MEDLINE | ID: mdl-23935974

ABSTRACT

Fas-associated death domain (DD) adaptor (FADD), a member of the DD superfamily, contains both a DD and a death effector domain (DED) that are important in mediating FAS ligand-induced apoptotic signaling. P45 is a unique member of the DD superfamily in that it has a domain with sequence and structural characteristics of both DD and DED. We show that p45 forms a complex with FADD and diminishes Fas-FADD mediated death signaling. The DED of FADD is required for the complex formation with p45. Following spinal cord injury, transgenic mice over-expressing p45 exhibit increased neuronal survival, decreased retraction of corticospinal tract fibers and improved functional recovery. Understanding p45-mediated cellular and molecular mechanisms may provide insights into facilitating nerve regeneration in humans.


Subject(s)
Fas-Associated Death Domain Protein/metabolism , Membrane Glycoproteins/metabolism , Neurons/metabolism , Neurons/pathology , Receptors, Death Domain/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Animals , Caspase 3/metabolism , Caspase 8/metabolism , Cell Death , Cell Survival , Enzyme Activation , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/chemistry , Female , HEK293 Cells , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Protein Structure, Tertiary , Signal Transduction , Thy-1 Antigens/metabolism
20.
Virol J ; 10: 130, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23617998

ABSTRACT

BACKGROUND: ZASC1 is a zinc finger-containing transcription factor that was previously shown to bind to specific DNA binding sites in the Moloney murine leukemia virus (Mo-MuLV) promoter and is required for efficient viral mRNA transcription (J. Virol. 84:7473-7483, 2010). METHODS: To determine whether this cellular factor influences Mo-MuLV replication and viral disease pathogenesis in vivo, we generated a ZASC1 knockout mouse model and completed both early infection and long term disease pathogenesis studies. RESULTS: Mice lacking ZASC1 were born at the expected Mendelian ratio and showed no obvious physical or behavioral defects. Analysis of bone marrow samples revealed a specific increase in a common myeloid progenitor cell population in ZASC1-deficient mice, a result that is of considerable interest because osteoclasts derived from the myeloid lineage are among the first bone marrow cells infected by Mo-MuLV (J. Virol. 73: 1617-1623, 1999). Indeed, Mo-MuLV infection of neonatal mice revealed that ZASC1 is required for efficient early virus replication in the bone marrow, but not in the thymus or spleen. However, the absence of ZASC1 did not influence the timing of subsequent tumor progression or the types of tumors resulting from virus infection. CONCLUSIONS: These studies have revealed that ZASC1 is important for myeloid cell differentiation in the bone marrow compartment and that this cellular factor is required for efficient Mo-MuLV replication in this tissue at an early time point post-infection.


Subject(s)
Bone Marrow/virology , DNA-Binding Proteins/metabolism , Leukemia, Experimental/virology , Moloney murine leukemia virus/physiology , Nuclear Proteins/metabolism , Retroviridae Infections/virology , Tumor Virus Infections/virology , Virus Replication , Animals , DNA-Binding Proteins/deficiency , Host-Pathogen Interactions , Mice , Mice, Knockout , Nuclear Proteins/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...