Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38701782

ABSTRACT

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Subject(s)
Epigenesis, Genetic , Myelin Sheath , Oligodendroglia , Remyelination , Animals , Myelin Sheath/metabolism , Humans , Mice , Remyelination/drug effects , Oligodendroglia/metabolism , Central Nervous System/metabolism , Mice, Inbred C57BL , Rejuvenation , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Organoids/metabolism , Organoids/drug effects , Demyelinating Diseases/metabolism , Demyelinating Diseases/genetics , Cell Differentiation/drug effects , Small Molecule Libraries/pharmacology , Male , Regeneration/drug effects , Multiple Sclerosis/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology
2.
J Neurosci ; 44(20)2024 May 15.
Article in English | MEDLINE | ID: mdl-38565291

ABSTRACT

Microglia undergo two-stage activation in neurodegenerative diseases, known as disease-associated microglia (DAM). TREM2 mediates the DAM2 stage transition, but what regulates the first DAM1 stage transition is unknown. We report that glucose dyshomeostasis inhibits DAM1 activation and PKM2 plays a role. As in tumors, PKM2 was aberrantly elevated in both male and female human AD brains, but unlike in tumors, it is expressed as active tetramers, as well as among TREM2+ microglia surrounding plaques in 5XFAD male and female mice. snRNAseq analyses of microglia without Pkm2 in 5XFAD mice revealed significant increases in DAM1 markers in a distinct metabolic cluster, which is enriched in genes for glucose metabolism, DAM1, and AD risk. 5XFAD mice incidentally exhibited a significant reduction in amyloid pathology without microglial Pkm2 Surprisingly, microglia in 5XFAD without Pkm2 exhibited increases in glycolysis and spare respiratory capacity, which correlated with restoration of mitochondrial cristae alterations. In addition, in situ spatial metabolomics of plaque-bearing microglia revealed an increase in respiratory activity. These results together suggest that it is not only glycolytic but also respiratory inputs that are critical to the development of DAM signatures in 5XFAD mice.


Subject(s)
Glucose , Homeostasis , Mice, Transgenic , Microglia , Animals , Microglia/metabolism , Microglia/pathology , Mice , Homeostasis/physiology , Glucose/metabolism , Male , Female , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Glycolysis/physiology , Thyroid Hormone-Binding Proteins
3.
ACS Med Chem Lett ; 12(10): 1546-1552, 2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34676036

ABSTRACT

An indazole/aza-indazole scaffold was developed as a novel chemotype for JNK3 inhibition. Extensive structure activity relationship (SAR) studies utilizing various in vitro and in vivo assays led to potent and highly selective JNK3 inhibitors with good oral bioavailability and high brain penetration. One lead compound, 29, was a potent and selective JNK3 inhibitor (IC50 = 0.005 µM) that had significant inhibition (>80% at 1 µM) to only JNK3 and JNK2 in a panel profiling of 374 wild-type kinases, had high potency in functional cell-based assays, had high stability in the human liver microsome (t 1/2 = 92 min), and was orally bioavailable and brain penetrant (brain/plasma ratio: 56%). The cocrystal structure of 29 in human JNK3 at a 2.1 Å resolution showed that indazole or aza-indazole-based JNK3 inhibitors demonstrated a type I kinase inhibition/binding.

4.
J Neurosci ; 41(42): 8710-8724, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34507952

ABSTRACT

We report that the neurotrophin receptor p75 contributes to sensory neuron survival through the regulation of cholesterol metabolism in Schwann cells. Selective deletion of p75 in mouse Schwann cells of either sex resulted in a 30% loss of dorsal root ganglia (DRG) neurons and diminished thermal sensitivity. P75 regulates Schwann cell cholesterol biosynthesis in response to BDNF, forming a co-receptor complex with ErbB2 and activating ErbB2-mediated stimulation of sterol regulatory element binding protein 2 (SREBP2), a master regulator of cholesterol synthesis. Schwann cells lacking p75 exhibited decreased activation of SREBP2 and a reduction in 7-dehydrocholesterol (7-DHC) reductase (DHCR7) expression, resulting in accumulation of the neurotoxic intermediate, 7-dehyrocholesterol in the sciatic nerve. Restoration of DHCR7 in p75 null Schwann cells in mice significantly attenuated DRG neuron loss. Together, these results reveal a mechanism by which the disruption of lipid metabolism in glial cells negatively influences sensory neuron survival, which has implications for a wide range of peripheral neuropathies.SIGNIFICANCE STATEMENT Although expressed in Schwann cells, the role of p75 in myelination has remained unresolved in part because of its dual expression in sensory neurons that Schwann cells myelinate. When p75 was deleted selectively among Schwann cells, myelination was minimally affected, while sensory neuron survival was reduced by 30%. The phenotype is mainly due to dysregulation of cholesterol biosynthesis in p75-deficient Schwann cells, leading to an accumulation of neurotoxic cholesterol precursor, 7-dehydrocholesterol (7-DHC). Mechanism-wise, we discovered that in response to BDNF, p75 recruits and activates ErbB2 independently of ErbB3, thereby stimulating the master regulator, sterol regulatory element binding protein 2 (SREBP2). These results together highlight a novel role of p75 in Schwann cells in regulating DRG neuron survival by orchestrating proper cholesterol metabolism.


Subject(s)
Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Schwann Cells/metabolism , Sensory Receptor Cells/metabolism , Animals , Cell Survival/physiology , Cells, Cultured , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Schwann Cells/ultrastructure , Sensory Receptor Cells/ultrastructure
5.
ACS Med Chem Lett ; 12(1): 24-29, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33488960

ABSTRACT

Potent JNK3 isoform selective inhibitors were developed from a thiophenyl-pyrazolourea scaffold. Through structure activity relationship (SAR) studies utilizing enzymatic and cell-based assays, and in vitro and in vivo drug metabolism and pharmacokinetic (DMPK) studies, potent and highly selective JNK3 inhibitors with oral bioavailability and brain penetrant capability were developed. Inhibitor 17 was a potent and isoform selective JNK3 inhibitor (IC50 = 35 nM), had significant inhibition to only JNK3 in a panel profiling of 374 wild-type kinases, had high potency in functional cell-based assays, had high stability in human liver microsome (t 1/2 = 66 min) and a clean CYP-450 inhibition profile, and was orally bioavailable and brain penetrant. Moreover, cocrystal structures of compounds 17 and 27 in human JNK3 were solved at 1.84 Å, which showed that these JNK3 isoform selective inhibitors bound to the ATP pocket, had interactions in both hydrophobic pocket-I and hydrophobic pocket-II.

6.
Dev Biol ; 462(1): 36-49, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32113830

ABSTRACT

Embryonic morphogenesis relies on the intrinsic ability of cells, often through remodeling the cytoskeleton, to shape epithelial tissues during development. Epithelial invagination is an example of morphogenesis that depends on this remodeling but the cellular mechanisms driving arrangement of cytoskeletal elements needed for tissue deformation remain incompletely characterized. To elucidate these mechanisms, live fluorescent microscopy and immunohistochemistry on fixed specimens were performed on chick and mouse lens placodes. This analysis revealed the formation of peripherally localized, circumferentially orientated and aligned junctions enriched in F-actin and MyoIIB. Once formed, the aligned junctions contract in a Rho-kinase and non-muscle myosin dependent manner. Further molecular characterization of these junctions revealed a Rho-kinase dependent accumulation of Arhgef11, a RhoA-specific guanine exchange factor known to regulate the formation of actomyosin cables and junctional contraction. In contrast, the localization of the Par-complex protein Par3, was reduced in these circumferentially orientated junctions. In an effort to determine if Par3 plays a negative role in MyoIIB accumulation, Par3-deficient mouse embryos were analyzed which not only revealed an increase in bicellular junctional accumulation of MyoIIB, but also a reduction of Arhgef11. Together, these results highlight the importance of the formation of the multicellular actomyosin cables that appear essential to the initiation of epithelial invagination and implicate the potential role of Arhgef11 and Par3 in their contraction and formation.


Subject(s)
Actomyosin/metabolism , Lens, Crystalline/embryology , Actin Cytoskeleton/metabolism , Actins/metabolism , Actomyosin/physiology , Adaptor Proteins, Signal Transducing/metabolism , Adherens Junctions/metabolism , Animals , Cell Cycle Proteins/metabolism , Chick Embryo , Cytoskeleton/metabolism , Embryonic Development , Epithelial Cells/metabolism , Female , Guanine Nucleotide Exchange Factors/metabolism , Mice , Mice, Knockout , Morphogenesis , Rho Guanine Nucleotide Exchange Factors/metabolism , rho-Associated Kinases/metabolism
7.
Elife ; 92020 01 29.
Article in English | MEDLINE | ID: mdl-31995032

ABSTRACT

Networks of neurons control feeding and activity patterns by integrating internal metabolic signals of energy balance with external environmental cues such as time-of-day. Proper circadian alignment of feeding behavior is necessary to prevent metabolic disease, and thus it is imperative that molecular players that maintain neuronal coordination of energy homeostasis are identified. Here, we demonstrate that mice lacking the p75 neurotrophin receptor, p75NTR, decrease their feeding and food anticipatory behavior (FAA) in response to daytime, but not nighttime, restricted feeding. These effects lead to increased weight loss, but do not require p75NTR during development. Instead, p75NTR is required for fasting-induced activation of neurons within the arcuate hypothalamus. Indeed, p75NTR specifically in AgRP neurons is required for FAA in response to daytime restricted feeding. These findings establish p75NTR as a novel regulator gating behavioral response to food scarcity and time-of-day dependence of circadian food anticipation.


In many animals, specific types of neurons, such as the hypothalamic hunger neurons, are tasked with regulating food consumption, integrating internal signals of hunger. In general, individuals eat if food becomes available when they are hungry; if food is absent, they will start moving to find new resources. Finally, if food always comes at the same time, animals will increase their activity just before it is delivered. Neurotrophins are a family of proteins that have many essential roles in the brain. In recent years, they have been shown to interact with the circadian clock, the built-in mechanism that helps animals stay synchronized with the cycle of day and night. A protein known as p75NTR is present in nerve cells, including hypothalamic hunger neurons: there, it helps to relay messages from a neurotrophin which, amongst other roles, controls food intake. However, it was unclear whether p75NTR played a role in regulating feeding behaviors, especially in a circadian manner. To investigate this question, Podyma et al. genetically engineered a group of mice lacking p75NTR, and a group missing the protein only in their hypothalamic hunger neurons. Both types of mutants had abnormal control of their feeding behavior: compared to normal mice, they fed less (and lost more weight) after they had been deprived of food overnight, or when they faced food shortage over multiple days. In addition, the mutants failed to move more before being fed. However, these feeding patterns were only affected during daytime, while they were preserved at night. These results reveal a new role for p75NTR in hypothalamic hunger neurons. Dissecting the biological processes that control food intake is key since obesity levels are increasing around the world. In particular, the relationship between food intake and the circadian clock is an important avenue of research as time-restricted diets (where food intake is only allowed during specific periods of the day) are growing in popularity.


Subject(s)
Agouti-Related Protein/metabolism , Feeding Behavior , Homeostasis , Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Blood Chemical Analysis , Circadian Rhythm , Germ-Line Mutation , Mice , Mice, Knockout , Receptors, Nerve Growth Factor/genetics
8.
Anal Chem ; 91(3): 2304-2311, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30608652

ABSTRACT

Sensitivity and resolution are key considerations for NMR applications in general and for metabolomics in particular, where complex mixtures containing hundreds of metabolites over a large range of concentrations are commonly encountered. There is a strong demand for advanced methods that can provide maximal information in the shortest possible time frame. Here, we present the optimization and application of the recently introduced 2D real-time BIRD 1H-13C HSQC experiment for NMR-based metabolomics of aqueous samples at 13C natural abundance. For mouse urine samples, it is demonstrated how this real-time pure shift sensitivity-improved heteronuclear single quantum correlation method provides broadband homonuclear decoupling along the proton detection dimension and thereby significantly improves spectral resolution in regions that are affected by spectral overlap. Moreover, the collapse of the scalar multiplet structure of cross-peaks leads to a sensitivity gain of about 40-50% over a traditional 2D HSQC-SI experiment. The experiment works well over a range of magnetic field strengths and is particularly useful when resonance overlap in crowded regions of the HSQC spectra hampers accurate metabolite identification and quantitation.


Subject(s)
Metabolome , Metabolomics/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Urine/chemistry , Animals , Mice
9.
J Clin Invest ; 128(5): 1772-1786, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29584618

ABSTRACT

Loss of bladder control is a challenging outcome facing patients with spinal cord injury (SCI). We report that systemic blocking of pro-nerve growth factor (proNGF) signaling through p75 with a CNS-penetrating small-molecule p75 inhibitor resulted in significant improvement in bladder function after SCI in rodents. The usual hyperreflexia was attenuated with normal bladder pressure, and automatic micturition was acquired weeks earlier than in the controls. The improvement was associated with increased excitatory input to the spinal cord, in particular onto the tyrosine hydroxylase-positive fibers in the dorsal commissure. The drug also had an effect on the bladder itself, as the urothelial hyperplasia and detrusor hypertrophy that accompany SCI were largely prevented. Urothelial cell loss that precedes hyperplasia was dependent on p75 in response to urinary proNGF that is detected after SCI in rodents and humans. Surprisingly, death of urothelial cells and the ensuing hyperplastic response were beneficial to functional recovery. Deleting p75 from the urothelium prevented urothelial death, but resulted in reduction in overall voiding efficiency after SCI. These results unveil a dual role of proNGF/p75 signaling in bladder function under pathological conditions with a CNS effect overriding the peripheral one.


Subject(s)
Nerve Growth Factor/metabolism , Nerve Tissue Proteins/metabolism , Protein Precursors/metabolism , Receptors, Nerve Growth Factor/metabolism , Signal Transduction , Spinal Cord Injuries/metabolism , Urinary Bladder Diseases/metabolism , Urinary Bladder/metabolism , Animals , Female , Gene Deletion , Humans , Male , Mice , Mice, Knockout , Nerve Growth Factor/genetics , Nerve Tissue Proteins/genetics , Protein Precursors/genetics , Receptors, Nerve Growth Factor/genetics , Spinal Cord Injuries/complications , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Urinary Bladder/pathology , Urinary Bladder Diseases/etiology , Urinary Bladder Diseases/genetics , Urinary Bladder Diseases/pathology , Urothelium/metabolism , Urothelium/pathology
10.
Nat Med ; 24(3): 338-351, 2018 03.
Article in English | MEDLINE | ID: mdl-29431744

ABSTRACT

Deficits in Schwann cell-mediated remyelination impair functional restoration after nerve damage, contributing to peripheral neuropathies. The mechanisms mediating block of remyelination remain elusive. Here, through small-molecule screening focusing on epigenetic modulators, we identified histone deacetylase 3 (HDAC3; a histone-modifying enzyme) as a potent inhibitor of peripheral myelinogenesis. Inhibition of HDAC3 enhanced myelin growth and regeneration and improved functional recovery after peripheral nerve injury in mice. HDAC3 antagonizes the myelinogenic neuregulin-PI3K-AKT signaling axis. Moreover, genome-wide profiling analyses revealed that HDAC3 represses promyelinating programs through epigenetic silencing while coordinating with p300 histone acetyltransferase to activate myelination-inhibitory programs that include the HIPPO signaling effector TEAD4 to inhibit myelin growth. Schwann cell-specific deletion of either Hdac3 or Tead4 in mice resulted in an elevation of myelin thickness in sciatic nerves. Thus, our findings identify the HDAC3-TEAD4 network as a dual-function switch of cell-intrinsic inhibitory machinery that counters myelinogenic signals and maintains peripheral myelin homeostasis, highlighting the therapeutic potential of transient HDAC3 inhibition for improving peripheral myelin repair.


Subject(s)
DNA-Binding Proteins/genetics , E1A-Associated p300 Protein/genetics , Muscle Proteins/genetics , Nerve Regeneration/genetics , Peripheral Nerve Injuries/genetics , Remyelination/genetics , Transcription Factors/genetics , Animals , Genome , Histone Deacetylases , Humans , Mice, Transgenic , Myelin Sheath/genetics , Myelin Sheath/metabolism , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Peripheral Nerve Injuries/physiopathology , Peripheral Nerve Injuries/rehabilitation , Recovery of Function/genetics , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/growth & development , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Signal Transduction , TEA Domain Transcription Factors
11.
Cell Death Differ ; 25(4): 663-678, 2018 03.
Article in English | MEDLINE | ID: mdl-29238071

ABSTRACT

Axonal injury is a common feature of central nervous system insults. Upregulation of amyloid precursor protein (APP) is observed following central nervous system neurotrauma and is regarded as a marker of central nervous system axonal injury. However, the underlying mechanism by which APP mediates neuronal death remains to be elucidated. Here, we used mouse optic nerve axotomy (ONA) to model central nervous system axonal injury replicating aspects of retinal ganglion cell (RGC) death in optic neuropathies. APP and APP intracellular domain (AICD) were upregulated in retina after ONA and APP knockout reduced Tuj1+ RGC loss. Pathway analysis of microarray data combined with chromatin immunoprecipitation and a luciferase reporter assay demonstrated that AICD interacts with the JNK3 gene locus and regulates JNK3 expression. Moreover, JNK3 was found to be upregulated after ONA and to contribute to Tuj1+ RGC death. APP knockout reduced the ONA-induced enhanced expression of JNK3 and phosphorylated JNK (pJNK). Gamma-secretase inhibitors prevented production of AICD, reduced JNK3 and pJNK expression similarly, and protected Tuj1+ RGCs from ONA-induced cell death. Together these data indicate that ONA induces APP expression and that gamma-secretase cleavage of APP releases AICD, which upregulates JNK3 leading to RGC death. This pathway may be a novel target for neuronal protection in optic neuropathies and other forms of neurotrauma.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Gene Expression Regulation, Enzymologic , Mitogen-Activated Protein Kinase 10/biosynthesis , Optic Nerve Diseases/metabolism , Optic Nerve/metabolism , Retinal Ganglion Cells/metabolism , Up-Regulation , Amyloid beta-Protein Precursor/genetics , Animals , Axotomy , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 10/genetics , Optic Nerve/pathology , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Retinal Ganglion Cells/pathology
12.
Dev Cell ; 40(6): 566-582.e5, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28350989

ABSTRACT

A lack of sufficient oligodendrocyte myelination contributes to remyelination failure in demyelinating disorders. miRNAs have been implicated in oligodendrogenesis; however, their functions in myelin regeneration remained elusive. Through developmentally regulated targeted mutagenesis, we demonstrate that miR-219 alleles are critical for CNS myelination and remyelination after injury. Further deletion of miR-338 exacerbates the miR-219 mutant hypomyelination phenotype. Conversely, miR-219 overexpression promotes precocious oligodendrocyte maturation and regeneration processes in transgenic mice. Integrated transcriptome profiling and biotin-affinity miRNA pull-down approaches reveal stage-specific miR-219 targets in oligodendrocytes and further uncover a novel network for miR-219 targeting of differentiation inhibitors including Lingo1 and Etv5. Inhibition of Lingo1 and Etv5 partially rescues differentiation defects of miR-219-deficient oligodendrocyte precursors. Furthermore, miR-219 mimics enhance myelin restoration following lysolecithin-induced demyelination as well as experimental autoimmune encephalomyelitis, principal animal models of multiple sclerosis. Together, our findings identify context-specific miRNA-regulated checkpoints that control myelinogenesis and a therapeutic role for miR-219 in CNS myelin repair.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/pathology , MicroRNAs/metabolism , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Regeneration , Wound Healing , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Central Nervous System/drug effects , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Deletion , Lecithins/pharmacology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Myelin Sheath/drug effects , Nerve Regeneration/drug effects , Nerve Regeneration/genetics , Nerve Tissue Proteins/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Optic Nerve/pathology , Optic Nerve/ultrastructure , Phenotype , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , Wound Healing/drug effects , Wound Healing/genetics
13.
Nat Commun ; 6: 6576, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25807892

ABSTRACT

Angiogenesis produces primitive vascular networks that need pruning to yield hierarchically organized and functional vessels. Despite the critical importance of vessel pruning to vessel patterning and function, the mechanisms regulating this process are not clear. Here we show that EphrinB2, a well-known player in angiogenesis, is an essential regulator of endothelial cell death and vessel pruning. This regulation depends upon phosphotyrosine-EphrinB2 signalling repressing c-jun N-terminal kinase 3 activity via STAT1. JNK3 activation causes endothelial cell death. In the absence of JNK3, hyaloid vessel physiological pruning is impaired, associated with abnormal persistence of hyaloid vessels, defective retinal vasculature and microphthalmia. This syndrome closely resembles human persistent hyperplastic primary vitreus (PHPV), attributed to failed involution of hyaloid vessels. Our results provide evidence that EphrinB2/STAT1/JNK3 signalling is essential for vessel pruning, and that defects in this pathway may contribute to PHPV.


Subject(s)
Endothelial Cells/metabolism , Ephrin-B2/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Neovascularization, Physiologic/genetics , Retinal Vessels/metabolism , STAT1 Transcription Factor/metabolism , Animals , Cell Proliferation , Cell Survival , Chromatin Immunoprecipitation , Flow Cytometry , Fluorescent Antibody Technique , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Immunoblotting , Immunoprecipitation , In Vitro Techniques , Mice , Mice, Knockout , Neovascularization, Pathologic/genetics , Persistent Hyperplastic Primary Vitreous/genetics , Signal Transduction
14.
J Biol Chem ; 289(45): 31458-72, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25253694

ABSTRACT

p75 is expressed among Purkinje cells in the adult cerebellum, but its function has remained obscure. Here we report that p75 is involved in maintaining the frequency and regularity of spontaneous firing of Purkinje cells. The overall spontaneous firing activity of Purkinje cells was increased in p75(-/-) mice during the phasic firing period due to a longer firing period and accompanying reduction in silence period than in the wild type. We attribute these effects to a reduction in small conductance Ca(2+)-activated potassium (SK) channel activity in Purkinje cells from p75(-/-) mice compared with the wild type littermates. The mechanism by which p75 regulates SK channel activity appears to involve its ability to activate Rac1. In organotypic cultures of cerebellar slices, brain-derived neurotrophic factor increased RacGTP levels by activating p75 but not TrkB. These results correlate with a reduction in RacGTP levels in synaptosome fractions from the p75(-/-) cerebellum, but not in that from the cortex of the same animals, compared with wild type littermates. More importantly, we demonstrate that Rac1 modulates SK channel activity and firing patterns of Purkinje cells. Along with the finding that spine density was reduced in p75(-/-) cerebellum, these data suggest that p75 plays a role in maintaining normalcy of Purkinje cell firing in the cerebellum in part by activating Rac1 in synaptic compartments and modulating SK channels.


Subject(s)
Cerebellum/metabolism , Neuropeptides/metabolism , Purkinje Cells/metabolism , Receptors, Nerve Growth Factor/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Electrophysiology , Golgi Apparatus/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Patch-Clamp Techniques , Potassium Channels/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Synaptosomes/metabolism , Tetraethylammonium/chemistry , rac GTP-Binding Proteins/metabolism
15.
Neuro Oncol ; 16(10): 1354-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24879047

ABSTRACT

BACKGROUNDS: Piperlongumine, a natural plant product, kills multiple cancer types with little effect on normal cells. Piperlongumine raises intracellular levels of reactive oxygen species (ROS), a phenomenon that may underlie the cancer-cell killing. Although these findings suggest that piperlongumine could be useful for treating cancers, the mechanism by which the drug selectively kills cancer cells remains unknown. METHODS: We treated multiple high-grade glioma (HGG) sphere cultures with piperlongumine and assessed its effects on ROS and cell-growth levels as well as changes in downstream signaling. We also examined the levels of putative piperlongumine targets and their roles in HGG cell growth. RESULTS: Piperlongumine treatment increased ROS levels and preferentially killed HGG cells with little effect in normal brain cells. Piperlongumine reportedly increases ROS levels after interactions with several redox regulators. We found that HGG cells expressed higher levels of the putative piperlongumine targets than did normal neural stem cells (NSCs). Furthermore, piperlongumine treatment in HGG cells, but not in normal NSCs, increased oxidative inactivation of peroxiredoxin 4 (PRDX4), an ROS-reducing enzyme that is overexpressed in HGGs and facilitates proper protein folding in the endoplasmic reticulum (ER). Moreover, piperlongumine exacerbated intracellular ER stress, an effect that was mimicked by suppressing PRDX4 expression. CONCLUSIONS: Our results reveal that the mechanism by which piperlongumine preferentially kills HGG cells involves PRDX4 inactivation, thereby inducing ER stress. Therefore, piperlongumine treatment could be considered as a novel therapeutic option for HGG treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Dioxolanes/administration & dosage , Endoplasmic Reticulum Stress/drug effects , Glioma/drug therapy , Peroxiredoxins/metabolism , Animals , Apoptosis/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Databases, Factual , Glioma/metabolism , Glioma/mortality , Humans , Mice , Reactive Oxygen Species/metabolism , Survival Analysis , Tumor Cells, Cultured
16.
J Neurosci ; 33(6): 2388-97, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23392668

ABSTRACT

In the developing peripheral nervous system, axon-derived signals stimulate Schwann cells to undergo a global genetic reprogramming involving the cessation of cellular division and the upregulation of myelin genes. How such a comprehensive change in gene transcription is regulated is poorly understood. Here we report that BRG1/SMARCA4, the central helicase of the mammalian SWI/SNF-related chromatin remodeling complex, is required for Schwann cells to differentiate and form myelin, both in vitro and in vivo, in the mouse. BRG1 was highly activated in Schwann cells at early stages of myelination, and loss of the enzyme inhibited their differentiation and completely prevented myelin formation. Furthermore, we identify NF-κB as a key transcription factor that associates with the BRG1 complex in response to neuregulin 1 type III. During myelination, BRG1 was activated through the formation of a complex with NF-κB, and both proteins bound to the promoter region of Sox10, an inducer of myelination. These findings delineate a novel mechanism whereby axonal signals promote myelination through the remodeling of chromatin structure.


Subject(s)
Cell Differentiation/physiology , Chromatin/metabolism , DNA Helicases/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Schwann Cells/physiology , Transcription Factors/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Chromatin/physiology , Coculture Techniques , DNA Helicases/physiology , Female , HEK293 Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , NF-kappa B/physiology , Nuclear Proteins/physiology , Rats , Schwann Cells/cytology , Transcription Factors/physiology
17.
J Neurosci ; 33(2): 397-410, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23303920

ABSTRACT

The lack of effective therapies for spinal cord injury points to the need for identifying novel targets for therapeutic intervention. Here we report that a small molecule, LM11A-31, developed to block proNGF-p75 interaction and p75-mediated cell death crosses the blood-brain barrier efficiently when delivered orally. Administered starting 4 h postinjury, LM11A-31 promotes functional recovery without causing any toxicity or increased pain in a mouse model of spinal contusion injury. In both weight-bearing open-field tests and nonweight-bearing swim tests, LM11A-31 was effective in improving motor function and coordination. Such functional improvement correlated with a >50% increase in the number of surviving oligodendrocytes and myelinated axons. We also demonstrate that LM11A-31 indeed inhibits proNGF-p75 interaction in vivo, thereby curtailing the JNK3-mediated apoptotic cascade. These results thus highlight p75 as a novel therapeutic target for an orally delivered treatment for spinal cord injury.


Subject(s)
Isoleucine/analogs & derivatives , Morpholines/therapeutic use , Myelin Sheath/metabolism , Nerve Growth Factor/metabolism , Protein Precursors/metabolism , Receptor, Nerve Growth Factor/drug effects , Receptor, Nerve Growth Factor/metabolism , Spinal Cord Injuries/drug therapy , Animals , Blotting, Western , DNA/genetics , Dose-Response Relationship, Drug , Forelimb/physiology , Hindlimb/physiology , Hyperalgesia/drug therapy , Immunohistochemistry , Isoleucine/therapeutic use , Locomotion/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 10/metabolism , Polymerase Chain Reaction , Spinal Cord Injuries/pathology , Swimming/physiology
18.
Cerebellum ; 12(3): 300-3, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23307658

ABSTRACT

The p75 neurotrophin receptor is highly expressed in the developing nervous system and is required for neuronal survival, growth, and synaptic transmission. In young mice, p75 is present in both granular cells and Purkinje cells of the cerebellum. Although p75 has been implicated in modulation of neuronal excitability in several neuronal types, whether and how it affects the excitability of cerebellar Purkinje neurons remained unclear. Using extracellular recordings of spontaneous firing of Purkinje neurons in cerebellar slices prepared from wild type and p75 knockout mice, we measured intrinsic firing properties in the presence of fast synaptic blockers of more than 200 Purkinje cells, each for a period of 5 min, for each genotype. We detected a significant increase in the mean firing frequency in p75(-/-) neurons comparing to the wild type littermates. Upon separating tonically firing from phasically firing cells, i.e., cells with firing pauses of longer than 300 ms, we observed that the change mainly arose from phasic firing cells and can be explained by an increase in the firing/silence ratio and a decrease in the number of long pauses during the 5-min recording period. We conclude that p75 plays an important role in regulating the firing-to-silence transition during the phasic firing period of the spontaneous firing of Purkinje cells. Thus, p75 exerts a modulatory function on Purkinje cell firing patterns, through which it may act as a key player in motor coordination and other cerebellum-regulated activities since Purkinje cells represent the sole neuronal output of the cerebellar cortex.


Subject(s)
Action Potentials/genetics , Cerebellum/cytology , Purkinje Cells/physiology , Receptors, Nerve Growth Factor/deficiency , Animals , Animals, Newborn , In Vitro Techniques , Mice , Mice, Knockout
19.
Neuron ; 75(5): 824-37, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22958823

ABSTRACT

Although Aß peptides are causative agents in Alzheimer's disease (AD), the underlying mechanisms are still elusive. We report that Aß42 induces a translational block by activating AMPK, thereby inhibiting the mTOR pathway. This translational block leads to widespread ER stress, which activates JNK3. JNK3 in turn phosphorylates APP at T668, thereby facilitating its endocytosis and subsequent processing. In support, pharmacologically blocking translation results in a significant increase in Aß42 in a JNK3-dependent manner. Thus, JNK3 activation, which is increased in human AD cases and a familial AD (FAD) mouse model, is integral to perpetuating Aß42 production. Concomitantly, deletion of JNK3 from FAD mice results in a dramatic reduction in Aß42 levels and overall plaque loads and increased neuronal number and improved cognition. This reveals AD as a metabolic disease that is under tight control by JNK3.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Mitogen-Activated Protein Kinase 10/metabolism , Peptide Fragments/metabolism , Stress, Physiological/physiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/toxicity , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred Strains , Mice, Knockout , Mitogen-Activated Protein Kinase 10/deficiency , Mitogen-Activated Protein Kinase 10/genetics , Organ Culture Techniques , Peptide Fragments/biosynthesis , Peptide Fragments/toxicity , Primary Cell Culture , Rats
20.
PLoS One ; 7(8): e42818, 2012.
Article in English | MEDLINE | ID: mdl-22916164

ABSTRACT

Glioblastoma multiforme (GBM), the most common and aggressive primary brain malignancy, is incurable despite the best combination of current cancer therapies. For the development of more effective therapies, discovery of novel candidate tumor drivers is urgently needed. Here, we report that peroxiredoxin 4 (PRDX4) is a putative tumor driver. PRDX4 levels were highly increased in a majority of human GBMs as well as in a mouse model of GBM. Reducing PRDX4 expression significantly decreased GBM cell growth and radiation resistance in vitro with increased levels of ROS, DNA damage, and apoptosis. In a syngenic orthotopic transplantation model, Prdx4 knockdown limited GBM infiltration and significantly prolonged mouse survival. These data suggest that PRDX4 can be a novel target for GBM therapies in the future.


Subject(s)
Apoptosis , Cell Division , Glioblastoma/pathology , Peroxiredoxins/metabolism , Animals , DNA Damage , Disease Models, Animal , Glioblastoma/metabolism , Humans , Mice , Reactive Oxygen Species/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...