Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 40(3): 557-568, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31776210

ABSTRACT

Mitochondria are important sources of energy, but they are also the target of cellular stress, toxin exposure, and aging-related injury. Persistent accumulation of damaged mitochondria has been implicated in many neurodegenerative diseases. One highly conserved mechanism to clear damaged mitochondria involves the E3 ubiquitin ligase Parkin and PTEN-induced kinase 1 (PINK1), which cooperatively initiate the process called mitophagy that identifies and eliminates damaged mitochondria through the autophagosome and lysosome pathways. Parkin is a mostly cytosolic protein, but is rapidly recruited to damaged mitochondria and target them for mitophagy. Moreover, Parkin interactomes also involve signaling pathways and transcriptional machinery critical for survival and cell death. However, the mechanism that regulates Parkin protein level remains poorly understood. Here, we show that the loss of homeodomain interacting protein kinase 2 (HIPK2) in neurons and mouse embryonic fibroblasts (MEFs) has a broad protective effect from cell death induced by mitochondrial toxins. The mechanism by which Hipk2-/- neurons and MEFs are more resistant to mitochondrial toxins is in part due to the role of HIPK2 and its kinase activity in promoting Parkin degradation via the proteasome-mediated mechanism. The loss of HIPK2 leads to higher cytosolic Parkin protein levels at basal conditions and upon exposure to mitochondrial toxins, which protects mitochondria from toxin-induced damage. In addition, Hipk2-/- neurons and MEFs show increased expression of PGC-1α (peroxisome proliferator-activated receptor-γ coactivator 1), a Parkin downstream target that can provide additional benefits via transcriptional activation of mitochondrial genes. Together, these results reveal a previously unrecognized avenue to target HIPK2 in neuroprotection via the Parkin-mediated pathway.SIGNIFICANCE STATEMENT In this study, we provide evidence that homeodomain interacting protein kinase 2 (HIPK2) and its kinase activity promote Parkin degradation via the proteasome-mediated pathway. The loss of HIPK2 increases cytosolic and mitochondrial Parkin protein levels under basal conditions and upon exposure to mitochondrial toxins, which protect mitochondria from toxin-induced damage. In addition, Hipk2-/- neurons and mouse embryonic fibroblasts also show increased expression of PGC-1α (peroxisome proliferator-activated receptor-γ coactivator 1), a Parkin downstream target that can provide additional benefits via transcriptional activation of mitochondrial genes. These results indicate that targeting HIPK2 and its kinase activity can have neuroprotective effects by elevating Parkin protein levels.


Subject(s)
Mitochondria/drug effects , Neurons , Neuroprotective Agents , Neurotoxins/toxicity , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Dopaminergic Neurons/ultrastructure , Female , Fibroblasts/metabolism , Gene Expression Regulation/genetics , Male , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/ultrastructure , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Protein Kinases/genetics
2.
Methods Enzymol ; 535: 403-18, 2014.
Article in English | MEDLINE | ID: mdl-24377936

ABSTRACT

Ligand-dependent regulation of adenylyl cyclase by the large family of seven-transmembrane G protein-coupled receptors (GPCRs) represents a deeply conserved and widely deployed cellular signaling mechanism. Studies of adenylyl cyclase regulation by catecholamine receptors have led to a remarkably detailed understanding of the basic biochemistry of G protein-linked signal transduction and have elaborated numerous mechanisms of regulation. Endocytosis of GPCRs plays a significant role in controlling longer-term cellular responses, such as under conditions of prolonged or repeated receptor activation occurring over a course of hours or more. It has been more challenging to investigate regulatory effects occurring over shorter time intervals, within the minutes to tens of minutes spanning the time course of many acute cyclic AMP (cAMP)-mediated signaling processes. A main reason for this is that biochemical methods used traditionally to assay changes in cytoplasmic cAMP concentration are limited in spatiotemporal resolution and typically require perturbing cellular structure and/or function for implementation. Recent developments in engineering genetically encoded cAMP biosensors linked to optical readouts, which can be expressed in cells or tissues and detected without cellular disruption or major functional perturbation, represent a significant step toward overcoming these limitations. Here, we describe the application of two such cAMP biosensors, one based on enzyme complementation and luminescence detection and another using Förster resonance energy transfer and fluorescence detection. We focus on applying these approaches to investigate cAMP signaling by catecholamine receptors and then on combining these analytical approaches with manipulations of receptor endocytic trafficking.


Subject(s)
Endocytosis , Receptors, G-Protein-Coupled/metabolism , Second Messenger Systems , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Microscopy, Fluorescence , Protein Transport
3.
Mol Pharmacol ; 83(3): 633-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23239825

ABSTRACT

Differences in the ability of opioid drugs to promote regulated endocytosis of µ-opioid receptors are related to their tendency to produce drug tolerance and dependence. Here we show that drug-specific differences in receptor internalization are determined by a conserved, 10-residue sequence in the receptor's carboxyl-terminal cytoplasmic tail. Diverse opioids induce receptor phosphorylation at serine (S)375, present in the middle of this sequence, but opioids differ markedly in their ability to drive higher-order phosphorylation on flanking residues [threonine (T)370, T376, and T379]. Multi-phosphorylation is required for the endocytosis-promoting activity of this sequence and occurs both sequentially and hierarchically, with S375 representing the initiating site. Higher-order phosphorylation involving T370, T376, and T379 specifically requires GRK2/3 isoforms, and the same sequence controls opioid receptor internalization in neurons. These results reveal a biochemical mechanism differentiating the endocytic activity of opioid drugs.


Subject(s)
Analgesics, Opioid/pharmacology , Receptors, Opioid/metabolism , Animals , Endocytosis/drug effects , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 3/metabolism , HEK293 Cells , Humans , Mice , Neurons/drug effects , Neurons/metabolism , Phosphorylation , Protein Isoforms/metabolism , Serine/metabolism , Threonine/metabolism
4.
Sci Signal ; 4(185): ra52, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21868358

ABSTRACT

In comparison to endogenous ligands of seven-transmembrane receptors, which typically act as full agonists, many drugs act as partial agonists. Partial agonism is best described as a "macroscopic" property that is manifest at the level of physiological systems or cell populations; however, whether partial agonists also encode discrete regulatory information at the "microscopic" level of individual receptors is not known. Here, we addressed this question by focusing on morphine, a partial agonist drug for µ-type opioid peptide receptors (MORs), and by combining quantitative mass spectrometry with cell biological analysis to investigate the reduced efficacy of morphine, compared to that of a peptide full agonist, in promoting receptor endocytosis. We showed that these chemically distinct ligands produced a complex and qualitatively similar mixture of phosphorylated opioid receptor forms in intact cells. Quantitatively, however, the different agonists promoted disproportionate multisite phosphorylation of a specific serine and threonine motif, and we found that modification at more than one residue was essential for the efficient recruitment of the adaptor protein ß-arrestin that mediated subsequent endocytosis of MORs. Thus, quantitative encoding of agonist-selective endocytosis at the level of individual opioid receptors was based on the conserved biochemical principles of multisite phosphorylation and threshold detection.


Subject(s)
Endocytosis/drug effects , Morphine/pharmacology , Narcotics/pharmacology , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Amino Acid Motifs , Animals , Humans , Mice , Phosphorylation/drug effects , Receptors, Opioid, mu/genetics
5.
Neuron ; 71(2): 278-90, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21791287

ABSTRACT

D(1) dopamine receptors are primary mediators of dopaminergic signaling in the CNS. These receptors internalize rapidly following agonist-induced activation, but the functional significance of this process is unknown. We investigated D(1) receptor endocytosis and signaling in HEK293 cells and cultured striatal neurons using real-time fluorescence imaging and cAMP biosensor technology. Agonist-induced activation of D(1) receptors promoted endocytosis of receptors with a time course overlapping that of acute cAMP accumulation. Inhibiting receptor endocytosis blunted acute D(1) receptor-mediated signaling in both dissociated cells and striatal slice preparations. Although endocytic inhibition markedly attenuated acute cAMP accumulation, inhibiting the subsequent recycling of receptors had no effect. Further, D(1) receptors localized in close proximity to endomembrane-associated trimeric G protein and adenylyl cyclase immediately after endocytosis. Together, these results suggest a previously unanticipated role of endocytosis, and the early endocytic pathway, in supporting rapid dopaminergic neurotransmission.


Subject(s)
Dopamine/metabolism , Endocytosis/physiology , Neurons/physiology , Signal Transduction/physiology , Action Potentials/drug effects , Action Potentials/genetics , Adenylyl Cyclases/pharmacology , Animals , Benzazepines/pharmacology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cells, Cultured , Corpus Striatum/cytology , Cyclic AMP/pharmacology , Dopamine/pharmacology , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , Embryo, Mammalian , Endocytosis/drug effects , Flow Cytometry/methods , Guanine Nucleotide Exchange Factors/metabolism , Humans , Hydrazones/pharmacology , Microscopy, Fluorescence/methods , Neurons/drug effects , Protein Transport/drug effects , RNA, Small Interfering/pharmacology , Rats , Receptors, Dopamine D1/genetics , Time Factors , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...