Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(5)2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35269915

ABSTRACT

Glioblastoma is the most aggressive brain tumour with short survival, partly due to resistance to conventional therapy. Glioma stem cells (GSC) are likely to be involved in treatment resistance, by releasing extracellular vesicles (EVs) containing specific molecular cargoes. Here, we studied the EVs secreted by glioma stem cells (GSC-EVs) and their effects on radiation resistance and glioma progression. EVs were isolated from 3 GSCs by serial centrifugation. NanoSight measurement, cryo-electron microscopy and live imaging were used to study the EVs size, morphology and uptake, respectively. The non-GSC glioma cell lines LN229 and U118 were utilised as a recipient cell model. Wound healing assays were performed to detect cell migration. Colony formation, cell viability and invadopodium assays were conducted to detect cell survival of irradiated recipient cells and cell invasion post GSC-EV treatment. NanoString miRNA global profiling was used to select for the GSC-EVs' specific miRNAs. All three GSC cell lines secreted different amounts of EVs, and all expressed consistent levels of CD9 but different level of Alix, TSG101 and CD81. EVs were taken up by both LN229 and U118 recipient cells. In the presence of GSC-EVs, these recipient cells survived radiation exposure and initiated colony formation. After GSC-EVs exposure, LN229 and U118 cells exhibited an invasive phenotype, as indicated by an increase in cell migration. We also identified 25 highly expressed miRNAs in the GSC-EVs examined, and 8 of these miRNAs can target PTEN. It is likely that GSC-EVs and their specific miRNAs induced the phenotypic changes in the recipient cells due to the activation of the PTEN/Akt pathway. This study demonstrated that GSC-EVs have the potential to induce radiation resistance and modulate the tumour microenvironment to promote glioma progression. Future therapeutic studies should be designed to interfere with these GSC-EVs and their specific miRNAs.


Subject(s)
Extracellular Vesicles , Glioma , MicroRNAs , Cryoelectron Microscopy , Extracellular Vesicles/metabolism , Glioma/genetics , Glioma/metabolism , Glioma/radiotherapy , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Tumor Microenvironment
2.
Mol Ther ; 25(6): 1269-1278, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28412169

ABSTRACT

Exosomes represent an attractive vehicle for the delivery of biomolecules. However, mechanisms for loading functional molecules into exosomes are relatively unexplored. Here we report the use of the evolutionarily conserved late-domain (L-domain) pathway as a mechanism for loading exogenous proteins into exosomes. We demonstrate that labeling of a target protein, Cre recombinase, with a WW tag leads to recognition by the L-domain-containing protein Ndfip1, resulting in ubiquitination and loading into exosomes. Our results show that Ndfip1 expression acts as a molecular switch for exosomal packaging of WW-Cre that can be suppressed using the exosome inhibitor GW4869. When taken up by floxed reporter cells, exosomes containing WW-Cre were capable of inducing DNA recombination, indicating functional delivery of the protein to recipient cells. Engineered exosomes were administered to the brain of transgenic reporter mice using the nasal route to test for intracellular protein delivery in vivo. This resulted in the transport of engineered exosomes predominantly to recipient neurons in a number of brain regions, including the olfactory bulb, cortex, striatum, hippocampus, and cerebellum. The ability to engineer exosomes to deliver biologically active proteins across the blood-brain barrier represents an important step for the development of therapeutics to treat brain diseases.


Subject(s)
Drug Delivery Systems , Exosomes/metabolism , Genetic Engineering , Protein Transport , Animals , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Extracellular Vesicles/metabolism , Gene Expression , Genetic Engineering/methods , Integrases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nasal Absorption , Permeability , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
3.
J Mol Cell Biol ; 7(2): 119-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25801959

ABSTRACT

Pten controls a signaling axis that is implicated to regulate cell proliferation, growth, survival, migration, and metabolism. The molecular mechanisms underlying the specificity of Pten responses to such diverse cellular functions are currently poorly understood. Here we report the control of Pten activity and signaling specificity during the cell cycle by Ndfip1 regulation of Pten spatial distribution. Genetic deletion of Ndfip1 resulted in a loss of Pten nuclear compartmentalization and increased cell proliferation, despite cytoplasmic Pten remaining active in regulating PI3K/Akt signaling. Cells lacking nuclear Pten were found to have dysregulated levels of Plk1 and cyclin D1 that could drive cell proliferation. In vivo, transgene expression of Ndfip1 in the developing brain increased nuclear Pten and lengthened the cell cycle of neuronal progenitors, resulting in microencephaly. Our results show that local partitioning of Pten from the cytoplasm to the nucleus represents a key mechanism contributing to the specificity of Pten signaling during cell proliferation.


Subject(s)
Carrier Proteins/physiology , Cell Proliferation , Membrane Proteins/physiology , PTEN Phosphohydrolase/metabolism , Active Transport, Cell Nucleus , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cyclin D1/metabolism , Female , Indazoles/pharmacology , Intercellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Microcephaly/metabolism , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Rats , Signal Transduction , Sirolimus/pharmacology , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Polo-Like Kinase 1
4.
J Biol Chem ; 290(11): 7141-50, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25631046

ABSTRACT

During injury, cells are vulnerable to apoptosis from a variety of stress conditions including DNA damage causing double-stranded breaks. Without repair, these breaks lead to aberrations in DNA replication and transcription, leading to apoptosis. A major response to DNA damage is provided by the protein kinase ATM (ataxia telangiectasia mutated) that is capable of commanding a plethora of signaling networks for DNA repair, cell cycle arrest, and even apoptosis. A key element in the DNA damage response is the mobilization of activating proteins into the cell nucleus to repair damaged DNA. BRAT1 is one of these proteins, and it functions as an activator of ATM by maintaining its phosphorylated status while also keeping other phosphatases at bay. However, it is unknown how BRAT1 is trafficked into the cell nucleus to maintain ATM phosphorylation. Here we demonstrate that Ndfip1-mediated ubiquitination of BRAT1 leads to BRAT1 trafficking into the cell nucleus. Without Ndfip1, BRAT1 failed to translocate to the nucleus. Under genotoxic stress, cells showed increased expression of both Ndfip1 and phosphorylated ATM. Following brain injury, neurons show increased expression of Ndfip1 and nuclear translocation of BRAT1. These results point to Ndfip1 as a sensor protein during cell injury and Ndfip1 up-regulation as a cue for BRAT1 ubiquitination by Nedd4 E3 ligases, followed by nuclear translocation of BRAT1.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Carrier Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Active Transport, Cell Nucleus , Animals , Brain Injuries/metabolism , Cell Line , DNA Damage , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins , Mice, Inbred C57BL , Nedd4 Ubiquitin Protein Ligases , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps , Proteolysis , Signal Transduction , Ubiquitination
5.
Methods ; 77-78: 157-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25542098

ABSTRACT

PTEN was discovered as a membrane-associated tumor suppressor protein nearly two decades ago, but the concept that it can be secreted and taken up by recipient cells is revolutionary. Since then, various laboratories have reported that PTEN is indeed secreted and available for uptake by other cells in at least two different guises. First, PTEN may be packaged and exported within extracellular vesicles (EV) called exosomes. Second, PTEN may also be secreted as a naked protein in a longer isoform called PTEN-long. While the conditions favouring the secretion of PTEN-long remain unknown, PTEN secretion in exosomes is enhanced by the Ndfip1/Nedd4 ubiquitination system. In this report, we describe conditions for packaging PTEN in exosomes and their potential use for mediating non cell-autonomous functions in recipient cells. We suggest that this mode of PTEN transfer may potentially provide beneficial PTEN for tumor suppression, however it may also propagate deleterious versions of mutated PTEN causing tumorigenesis.


Subject(s)
Exosomes/metabolism , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Mice
6.
Traffic ; 15(7): 749-61, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24798731

ABSTRACT

The spatial regulation of Pten is critical for its role as a tumour suppressor with both nuclear and cytoplasmic locations being implicated with distinct functions. In the cytoplasm, Pten plays a central role in opposing PI3K/Akt cell signalling, whereas in the nucleus, Pten is important for maintaining genome stability and enhancing the tumour suppressor activity of APC-CDH1. Despite this diversity in protein function at different subcellular locations, there is limited knowledge on how Pten is able to find different cellular niches. Here, we report that Rab5 GTPase is required for efficient trafficking and ubiquitination of Pten on endosomes inside the cytosol. Using bimolecular fluorescence complementation (BiFC) for imaging protein interactions, we observed that ubiquitinated Pten is localized to peri-nuclear and nuclear regions of the cell. Nuclear trafficking of Pten required both Rab5 as well as the E3 ligase adaptor protein Ndfip1. Rab5 colocalization with Pten was observed on endosomes and expression of a dominant negative form of Rab5 significantly reduced Pten ubiquitination and nuclear trafficking. Genomic deletion of Ndfip1 abrogated nuclear trafficking of ubiquitinated Pten, even in the presence of Rab5. Our findings show that endosomal trafficking and ubiquitination are important mechanisms for the subcellular distribution of Pten.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleus/metabolism , Membrane Proteins/metabolism , PTEN Phosphohydrolase/metabolism , rab5 GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Endosomes/metabolism , Intercellular Signaling Peptides and Proteins , Mice , Protein Transport , Ubiquitination
7.
PLoS One ; 9(1): e87119, 2014.
Article in English | MEDLINE | ID: mdl-24475238

ABSTRACT

Iron misregulation is a central component in the neuropathology of Parkinson's disease. The iron transport protein DMT1 is known to be increased in Parkinson's brains linking functional transport mechanisms with iron accumulation. The regulation of DMT1 is therefore critical to the management of iron uptake in the disease setting. We previously identified post-translational control of DMT1 levels through a ubiquitin-mediated pathway led by Ndfip1, an adaptor for Nedd4 family of E3 ligases. Here we show that loss of Ndfip1 from mouse dopaminergic neurons resulted in misregulation of DMT1 levels and increased susceptibility to iron induced death. We report that in human Parkinson's brains increased iron concentrations in the substantia nigra are associated with upregulated levels of Ndfip1 in dopaminergic neurons containing α-synuclein deposits. Additionally, Ndfip1 was also found to be misexpressed in astrocytes, a cell type normally devoid of this protein. We suggest that in Parkinson's disease, increased iron levels are associated with increased Ndfip1 expression for the regulation of DMT1, including abnormal Ndfip1 activation in non-neuronal cell types such as astrocytes.


Subject(s)
Astrocytes/metabolism , Carrier Proteins/metabolism , Dopaminergic Neurons/metabolism , Iron/metabolism , Membrane Proteins/metabolism , Parkinson Disease/genetics , Substantia Nigra/metabolism , Transcription Factors/metabolism , Aged , Aged, 80 and over , Animals , Astrocytes/drug effects , Astrocytes/pathology , Carrier Proteins/genetics , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Embryo, Mammalian , Female , Gene Expression Regulation , Humans , Ion Transport , Iron/pharmacology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Parkinson Disease/metabolism , Parkinson Disease/pathology , Primary Cell Culture , Signal Transduction , Substantia Nigra/pathology , Transcription Factors/genetics , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
8.
Exp Neurol ; 252: 37-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275527

ABSTRACT

There is controversy whether accumulation of the tumor suppressor PTEN protein in the cell nucleus under stress conditions such as trauma and stroke causes cell death. A number of in vitro studies have reported enhanced apoptosis in neurons possessing nuclear PTEN, with the interpretation that its nuclear phosphatase activity leads to reduction of the survival protein phospho-Akt. However, there have been no in vivo studies to show that nuclear PTEN in neurons under stress is detrimental. Using a mouse model of injury, we demonstrate here that brain trauma altered the nucleo-cytoplasmic distribution of Pten, resulting in increased nuclear Pten but only in surviving neurons near the lesion. This event was driven by Ndfip1, an adaptor and activator of protein ubiquitination by Nedd4 E3 ligases. Neurons next to the lesion with nuclear PTEN were invariably negative for TUNEL, a marker for cell death. These neurons also showed increased Ndfip1 which we previously showed to be associated with neuron survival. Biochemical assays revealed that overall levels of Pten in the affected cortex were unchanged after trauma, suggesting that Pten abundance globally had not increased but rather Pten subcellular location in affected neurons had changed. Following experimental injury, the number of neurons with nuclear Pten was reduced in heterozygous mice (Ndfip1(+/-)) although lesion volumes were increased. We conclude that nuclear trafficking of Pten following injury leads to neuron survival not death.


Subject(s)
Brain Injuries/pathology , Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Neurons , PTEN Phosphohydrolase/metabolism , Analysis of Variance , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Carrier Proteins/genetics , Cell Survival/genetics , Cytoplasm , Disease Models, Animal , Functional Laterality , Immunoprecipitation , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Oncogene Protein v-akt , PTEN Phosphohydrolase/genetics , Protein Transport/genetics
9.
Cereb Cortex ; 24(12): 3289-300, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23897647

ABSTRACT

Ubiquitin ligases of the Nedd4 family are important for axon and dendrite development, but little is known about their adaptor, Nedd4 family-interacting protein 1 (Ndfip1), that is responsible for their enzymatic activation. To study the function of Ndfip1 in cortical development, we generated a conditional knock-out (conditional KO) in neurons. The Ndfip1 conditional KO mice were viable; however, cortical neurons in the adult brain exhibited atrophic characteristics, including stunted dendritic arbors, blebbing of dendrites, and fewer dendritic spines. In electron micrographs, these neurons appeared shrunken with compacted somata and involutions of the nuclear membrane. In culture, Ndfip1 KO neurons exhibited exuberant sprouting suggesting loss of developmental control. Biochemical analysis of postsynaptic density (PSD) fractions from Ndfip1 KO cortical and hippocampal neurons showed that the postsynaptic proteins (Arc and PSD-95) were reduced compared with wild-type controls. In addition, the PI3 kinase/Akt signaling pathway was altered. These results indicate that Ndfip1, through its Nedd4 effectors, is important for the development of dendrites and dendritic spines in the cortex.


Subject(s)
Carrier Proteins/genetics , Dendritic Spines/metabolism , Gene Expression Regulation, Developmental/physiology , Membrane Proteins/genetics , Neocortex , Pyramidal Cells/diagnostic imaging , Animals , Animals, Newborn , Cell Fractionation , Cells, Cultured , Disks Large Homolog 4 Protein , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Guanylate Kinases/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/cytology , Neocortex/embryology , Neocortex/growth & development , Nestin/genetics , Nestin/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Ultrasonography
10.
Neurosci Lett ; 555: 225-30, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24036464

ABSTRACT

During development, protein substrates need to be removed and degraded when they are no longer required. The E3 ubiquitin ligases, including Nedd4 family proteins, are a major group of enzymes responsible for adding ubiquitin chains to protein substrates prior to their degradation. Ndfip1 (Nedd4 family-interacting protein 1) is an adaptor and activator for Nedd4-family ubiquitin ligases for increasing substrate specificity. To study Nedd4-mediated ubiquitination during cortical development, we have mapped the spatio-temporal dynamics of Ndfip1 protein expression by immunocytochemistry. Ndfip1 expression was observed from embryonic day 11 (E11.5) until adult stages. Its presence increased during the postnatal stages and peaked at postnatal day 7 (P7). Spatially, Ndfip1 was found in the ventricular and marginal zones during corticogenesis but also in the cortical plate and subplate during midstage cortical development (E15.5). Postnatally, Ndfip1 was expressed in all cortical neurons (but not in glial cells) and this expression was both ubiquitous and uniform across cortical layers involving both pyramidal and non-pyramidal neurons. This consistent but dynamic pattern of Ndfip1 expression in temporal and spatial domains of the cortical landscape is indicative of complex programs of protein ubiquitination during corticogenesis.


Subject(s)
Carrier Proteins/metabolism , Cerebral Cortex/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins , Interneurons/metabolism , Mice , Mice, Inbred C57BL , Morphogenesis , Nedd4 Ubiquitin Protein Ligases , Pyramidal Cells/metabolism , Ubiquitination
11.
Sci Signal ; 5(243): ra70, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-23012657

ABSTRACT

Exosomes are microvesicles of endosomal origin that are secreted, and their contents (proteins, lipids, DNA, or microRNAs) can alter the physiological states of recipient cells. We demonstrated that phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a tumor suppressor protein normally localized in the cytoplasm and nucleus, was secreted in exosomes. Secreted PTEN was internalized by recipient cells with resultant functional activity, which resulted in reduced phosphorylation of the serine and threonine kinase Akt and reduced cellular proliferation. PTEN secretion in exosomes required Ndfip1, an adaptor protein for members of the Nedd4 family of E3 ubiquitin ligases. Without Ndfip1, neither Nedd4-1 nor Nedd4-2 promoted the recruitment of PTEN into exosomes. In addition, lysine 13 within PTEN, which is required for its ubiquitination by Nedd4-1, was required for exosomal transport of PTEN. These results implicate Ndfip1 as a molecular regulator of the exosomal export of PTEN, with consequences for non-cell-autonomous PTEN activity. Thus, we suggest that the ability of PTEN to exert phosphatase activity beyond the cell in which it is produced has implications for PTEN function during development, health, and disease.


Subject(s)
Exosomes/metabolism , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/metabolism , Animals , Blotting, Western , Carrier Proteins/metabolism , Cell Proliferation , Electrophoresis, Polyacrylamide Gel , Endosomal Sorting Complexes Required for Transport/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Intercellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microscopy, Electron , Nedd4 Ubiquitin Protein Ligases , Phosphorylation , Tetrazolium Salts , Thiazoles , Trypan Blue , Ubiquitin-Protein Ligases/metabolism
12.
J Cell Biol ; 196(1): 29-36, 2012 Jan 09.
Article in English | MEDLINE | ID: mdl-22213801

ABSTRACT

PTEN (phosphatase and tensin homologue deleted on chromosome TEN) is the major negative regulator of phosphatidylinositol 3-kinase signaling and has cell-specific functions including tumor suppression. Nuclear localization of PTEN is vital for tumor suppression; however, outside of cancer, the molecular and physiological events driving PTEN nuclear entry are unknown. In this paper, we demonstrate that cytoplasmic Pten was translocated into the nuclei of neurons after cerebral ischemia in mice. Critically, this transport event was dependent on a surge in the Nedd4 family-interacting protein 1 (Ndfip1), as neurons in Ndfip1-deficient mice failed to import Pten. Ndfip1 binds to Pten, resulting in enhanced ubiquitination by Nedd4 E3 ubiquitin ligases. In vitro, Ndfip1 overexpression increased the rate of Pten nuclear import detected by photobleaching experiments, whereas Ndfip1(-/-) fibroblasts showed negligible transport rates. In vivo, Ndfip1 mutant mice suffered larger infarct sizes associated with suppressed phosphorylated Akt activation. Our findings provide the first physiological example of when and why transient shuttling of nuclear Pten occurs and how this process is critical for neuron survival.


Subject(s)
Brain Ischemia/metabolism , Carrier Proteins/physiology , Membrane Proteins/physiology , Neurons/physiology , PTEN Phosphohydrolase/metabolism , Animals , Brain Ischemia/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Survival , Endosomal Sorting Complexes Required for Transport/physiology , Intercellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nedd4 Ubiquitin Protein Ligases , Photobleaching , Protein Transport , Ubiquitin-Protein Ligases/physiology , Ubiquitination
13.
J Biol Chem ; 286(10): 8555-8564, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21187286

ABSTRACT

The delivery of metal ions using cell membrane-permeable metal complexes represents a method for activating cellular pathways. Here, we report the synthesis and characterization of new [Co(III)(salen)(acac)] complexes capable of up-regulating the ubiquitin ligase adaptor protein Ndfip1. Ndfip1 is a neuroprotective protein that is up-regulated in the brain after injury and functions in combination with Nedd4 ligases to ubiquitinate harmful proteins for removal. We previously showed that Ndfip1 can be increased in human neurons using CoCl(2) that is toxic at high concentration. Here we demonstrate a similar effect can be achieved by low concentrations of synthetic Co(III) complexes that are non-toxic and designed to be activated following cellular entry. Activation is achieved by intracellular reduction of Co(III) to Co(II) leading to release of Co(II) ions for Ndfip1 up-regulation. The cellular benefit of Ndfip1 up-regulation by Co(III) complexes includes demonstrable protection against cell death in SH-SY5Y cells during stress. In vivo, focal delivery of Co(III) complexes into the adult mouse brain was observed to up-regulate Ndfip1 in neurons. These results demonstrate that a cellular response pathway can be advantageously manipulated by chemical modification of metal complexes, and represents a significant step of harnessing low concentration metal complexes for therapeutic benefit.


Subject(s)
Brain/metabolism , Carrier Proteins/biosynthesis , Cobalt/pharmacology , Membrane Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Up-Regulation/drug effects , Animals , Brain/cytology , Carrier Proteins/genetics , Cell Death/drug effects , Cell Death/physiology , Cell Line , Humans , Intercellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mice , Neurons/cytology , Stress, Physiological/drug effects , Stress, Physiological/physiology , Up-Regulation/physiology
14.
Proc Natl Acad Sci U S A ; 106(36): 15489-94, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19706893

ABSTRACT

The regulation of metal ion transport within neurons is critical for normal brain function. Of particular importance is the regulation of redox metals such as iron (Fe), where excess levels can contribute to oxidative stress and protein aggregation, leading to neuronal death. The divalent metal transporter 1 (DMT1) plays a central role in the regulation of Fe as well as other metals; hence, failure of DMT1 regulation is linked to human brain pathology. However, it remains unclear how DMT1 is regulated in the brain. Here, we show that DMT1 is regulated by Ndfip1 (Nedd4 family-interacting protein 1), an adaptor protein that recruits E3 ligases to ubiquitinate target proteins. Using human neurons we show the Ndfip1 is upregulated and binds to DMT1 in response to Fe and cobalt (Co) exposure. This interaction results in the ubiquitination and degradation of DMT1, resulting in reduced metal entry. Induction of Ndfip1 expression protects neurons from metal toxicity, and removal of Ndfip1 by shRNAi results in hypersensitivity to metals. We identify Nedd4-2 as an E3 ligase recruited by Ndfip1 for the ubiquitination of DMT1 within human neurons. Comparison of brains from Ndfip1(-/-) with Ndfip1(+/+) mice exposed to Fe reveals that Ndfip1(-/-) brains accumulate Fe within neurons. Together, this evidence suggests a critical role for Ndfip1 in regulating metal transport in human neurons.


Subject(s)
Carrier Proteins/metabolism , Cation Transport Proteins/metabolism , Cobalt/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Gene Expression Regulation/physiology , Iron/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Blotting, Western , Cobalt/toxicity , Gene Expression Regulation/drug effects , Humans , Immunohistochemistry , Immunoprecipitation , Ion Transport , Iron/toxicity , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases , Neurons/drug effects , RNA Interference , Ubiquitination
15.
J Neurosci ; 29(13): 4016-22, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19339597

ABSTRACT

Although brain-derived neurotrophic factor (BDNF) has been shown to promote peripheral myelination during development and remyelination after injury, the precise mechanisms mediating this effect remain unknown. Here, we determine that BDNF promotes myelination of nerve growth factor-dependent neurons, an effect dependent on neuronal expression of the p75 neurotrophin receptor, whereas BDNF inhibits myelination of BDNF-dependent neurons via the full-length TrkB receptor. Thus, BDNF exerts contrasting effects on Schwann cell myelination, depending on the complement of BDNF receptors that are expressed by different subpopulations of dorsal root ganglion neurons. These results demonstrate that BDNF exerts contrasting modulatory roles in peripheral nervous system myelination, and that its mechanism of action is acutely regulated and specifically targeted to neurons.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Ganglia, Spinal/cytology , Myelin Proteins/metabolism , Nerve Growth Factor/physiology , Neurons/physiology , Animals , Animals, Genetically Modified , Animals, Newborn , Brain-Derived Neurotrophic Factor/pharmacology , Carbazoles/pharmacology , Cells, Cultured , Coculture Techniques/methods , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , Indole Alkaloids/pharmacology , Mice , Myelin Basic Protein/metabolism , Myelin P0 Protein/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Growth Factor/genetics , Receptors, Nerve Growth Factor/genetics , Schwann Cells/drug effects , Tissue Culture Techniques , Transfection
16.
J Biol Chem ; 283(47): 32621-7, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18819914

ABSTRACT

The ability to remove unwanted proteins is an important cellular feature. Classically, this involves the enzymatic addition of ubiquitin moieties followed by degradation in the proteasome. Nedd4 proteins are ubiquitin ligases important not only for protein degradation, but also for protein trafficking. Nedd4 proteins can bind to target proteins either by themselves or through adaptor protein Ndfip1 (Nedd4 family-interacting protein 1). An alternative mechanism for protein removal and trafficking is provided by exosomes, which are small vesicles (50-90-nm diameter) originating from late endosomes and multivesicular bodies (MVBs). Exosomes provide a rapid means of shedding obsolete proteins and also for cell to cell communication. In the present work, we show that Ndfip1 is detectable in exosomes secreted from transfected cells and also from primary neurons. Compared with control, Ndfip1 increases exosome secretion from transfected cells. Furthermore, while Nedd4, Nedd4-2, and Itch are normally absent from exosomes, expression of Ndfip1 results in recruitment of all three Nedd4 proteins into exosomes. Together, these results suggest that Ndfip1 is important for protein trafficking via exosomes, and provides a mechanism for cargoing passenger proteins such as Nedd4 family proteins. Given the positive roles of Ndfip1/Nedd4 in improving neuronal survival during brain injury, it is possible that exosome secretion provides a novel route for rapid sequestration and removal of proteins during stress.


Subject(s)
Carrier Proteins/metabolism , Carrier Proteins/physiology , Membrane Proteins/metabolism , Membrane Proteins/physiology , Ubiquitin-Protein Ligases/metabolism , Brain/pathology , Brefeldin A/pharmacology , Cell Communication , Cell Line , Cell Survival , Endosomal Sorting Complexes Required for Transport , Exosomes , Humans , Microscopy, Electron, Transmission , Models, Biological , Nedd4 Ubiquitin Protein Ligases , Neurons/metabolism , Repressor Proteins/metabolism , Transfection
17.
Nat Neurosci ; 8(3): 322-31, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15711540

ABSTRACT

The balance between proliferation and apoptosis is critical for proper development of the nervous system. Yet, little is known about molecules that regulate apoptosis of proliferative neurons. Here we identify a soluble, secreted form of CPG15 expressed in embryonic rat brain regions undergoing rapid proliferation and apoptosis, and show that it protects cultured cortical neurons from apoptosis by preventing activation of caspase 3. Using a lentivirus-delivered small hairpin RNA, we demonstrate that endogenous CPG15 is essential for the survival of undifferentiated cortical progenitors in vitro and in vivo. We further show that CPG15 overexpression in vivo expands the progenitor pool by preventing apoptosis, resulting in an enlarged, indented cortical plate and cellular heterotopias within the ventricular zone, similar to the phenotypes of mutant mice with supernumerary forebrain progenitors. CPG15 expressed during mammalian forebrain morphogenesis may help balance neuronal number by countering apoptosis in specific neuroblasts subpopulations, thus influencing final brain size and shape.


Subject(s)
Apoptosis/physiology , Cerebral Cortex/cytology , Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Stem Cells/physiology , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/embryology , Embryo, Mammalian , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...