Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
bioRxiv ; 2023 Oct 16.
Article in English | MEDLINE | ID: mdl-37873486

ABSTRACT

STING activation by cyclic dinucleotides in mammals induces interferon- and NFκB -related gene expression, and the lipidation of LC3B at Golgi membranes. While mechanisms of the interferon response are well understood, the mechanisms of NFκB activation mediated by STING remain unclear. We report that STING activation induces K63- and M1-linked/linear ubiquitin chain formation at LC3B-associated Golgi membranes. Loss of the LUBAC E3 ubiquitin ligase prevents formation of linear, but not K63-linked ubiquitin chains or STING activation and inhibits STING-induced NFκB and IRF3-mediated signaling in monocytic THP1 cells. The proton channel activity of STING is also important for both K63 and linear ubiquitin chain formation, and NFκB- and interferon-related gene expression. Thus, LUBAC synthesis of linear ubiquitin chains regulates STING-mediated innate immune signaling.

2.
EMBO J ; 42(22): e113491, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37621214

ABSTRACT

Nix is a membrane-anchored outer mitochondrial protein that induces mitophagy. While Nix has an LC3-interacting (LIR) motif that binds to ATG8 proteins, it also contains a minimal essential region (MER) that induces mitophagy through an unknown mechanism. We used chemically induced dimerization (CID) to probe the mechanism of Nix-mediated mitophagy and found that both the LIR and MER are required for robust mitophagy. We find that the Nix MER interacts with the autophagy effector WIPI2 and recruits WIPI2 to mitochondria. The Nix LIR motif is also required for robust mitophagy and converts a homogeneous WIPI2 distribution on the surface of the mitochondria into puncta, even in the absence of ATG8s. Together, this work reveals unanticipated mechanisms in Nix-induced mitophagy and the elusive role of the MER, while also describing an interesting example of autophagy induction that acts downstream of the canonical initiation complexes.


Subject(s)
Autophagy , Mitophagy , Mitochondria/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Autophagy-Related Protein 8 Family/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
3.
Mol Cell ; 83(6): 927-941.e8, 2023 03 16.
Article in English | MEDLINE | ID: mdl-36898370

ABSTRACT

Mitophagy is a form of selective autophagy that disposes of superfluous and potentially damage-inducing organelles in a tightly controlled manner. While the machinery involved in mitophagy induction is well known, the regulation of the components is less clear. Here, we demonstrate that TNIP1 knockout in HeLa cells accelerates mitophagy rates and that ectopic TNIP1 negatively regulates the rate of mitophagy. These functions of TNIP1 depend on an evolutionarily conserved LIR motif as well as an AHD3 domain, which are required for binding to the LC3/GABARAP family of proteins and the autophagy receptor TAX1BP1, respectively. We further show that phosphorylation appears to regulate its association with the ULK1 complex member FIP200, allowing TNIP1 to compete with autophagy receptors, which provides a molecular rationale for its inhibitory function during mitophagy. Taken together, our findings describe TNIP1 as a negative regulator of mitophagy that acts at the early steps of autophagosome biogenesis.


Subject(s)
Autophagy-Related Proteins , Autophagy , Mitophagy , Humans , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Autophagy-Related Protein 8 Family/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mitophagy/genetics , Neoplasm Proteins/metabolism
4.
Neurobiol Dis ; 172: 105832, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35907632

ABSTRACT

Synaptojanin 2 binding protein (SYNJ2BP) is an outer mitochondrial membrane protein with a cytosolic PDZ domain that functions as a cellular signaling hub. Few studies have evaluated its role in disease. Here we use induced pluripotent stem cell (iPSC)-derived motor neurons and post-mortem tissue from patients with two hereditary motor neuron diseases, spinal and bulbar muscular atrophy (SBMA) and amyotrophic lateral sclerosis type 4 (ALS4), and show that SYNJ2BP expression is increased in diseased motor neurons. Similarly, we show that SYNJ2BP expression increases in iPSC-derived motor neurons undergoing stress. Using proteomic analysis, we found that elevated SYNJ2BP alters the cellular distribution of mitochondria and increases mitochondrial-ER membrane contact sites. Furthermore, decreasing SYNJ2BP levels improves mitochondrial oxidative function in the diseased motor neurons. Together, our observations offer new insight into the molecular pathology of motor neuron disease and the role of SYNJ2BP in mitochondrial dysfunction.


Subject(s)
Amyotrophic Lateral Sclerosis , Motor Neuron Disease , Muscular Atrophy, Spinal , Amyotrophic Lateral Sclerosis/metabolism , Humans , Membrane Proteins/metabolism , Mitochondria/metabolism , Motor Neuron Disease/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/pathology , Proteomics
5.
Mol Cell Proteomics ; 21(10): 100262, 2022 10.
Article in English | MEDLINE | ID: mdl-35753663

ABSTRACT

The nonpsychoactive cannabinoid, cannabidiol (CBD), is Food and Dug Administration approved for treatment of two drug-resistant epileptic disorders and is seeing increased use among the general public, yet the mechanisms that underlie its therapeutic effects and side-effect profiles remain unclear. Here, we report a systems-level analysis of CBD action in human cell lines using temporal multiomic profiling. FRET-based biosensor screening revealed that CBD elicits a sharp rise in cytosolic calcium, and activation of AMP-activated protein kinase in human keratinocyte and neuroblastoma cell lines. CBD treatment leads to alterations in the abundance of metabolites, mRNA transcripts, and proteins associated with activation of cholesterol biosynthesis, transport, and storage. We found that CBD rapidly incorporates into cellular membranes, alters cholesterol accessibility, and disrupts cholesterol-dependent membrane properties. Sustained treatment with high concentrations of CBD induces apoptosis in a dose-dependent manner. CBD-induced apoptosis is rescued by inhibition of cholesterol synthesis and potentiated by compounds that disrupt cholesterol trafficking and storage. Our data point to a pharmacological interaction of CBD with cholesterol homeostasis pathways, with potential implications in its therapeutic use.


Subject(s)
Cannabidiol , Cannabinoids , Humans , Cannabidiol/pharmacology , Calcium/metabolism , AMP-Activated Protein Kinases , Cell Line , Cannabinoids/pharmacology , Homeostasis , RNA, Messenger/metabolism , Cholesterol
6.
J Vis Exp ; (178)2021 12 18.
Article in English | MEDLINE | ID: mdl-34978296

ABSTRACT

Collective cellular migration plays a key role in many fundamental biological processes including development, wound healing, and cancer metastasis. To understand the regulation of cell motility, we must be able to measure it easily and consistently under different conditions. Here we describe a method for measuring and quantifying single-cell and bulk motility of HaCaT keratinocytes using a nuclear stain. This method includes a MATLAB script for analyzing TrackMate output files to calculate displacements, motility rates, and trajectory angles in single cells and in bulk for an imaging site. This motility analysis script allows for quick, straightforward, and scalable analysis of cell motility rates from TrackMate data and could be broadly used to identify and study the regulation of motility in epithelial cells. We also provide a MATLAB script for reorganizing microscopy videos collected on a microscope and converting them to TIF stacks, which can be analyzed using the ImageJ TrackMate plugin in bulk. Using this methodology to explore the roles of adherens junctions and actin cytoskeletal dynamics in regulating cell motility in HaCaT keratinocytes, we demonstrate evidence that Arp2/3 activity is required for the elevated motility seen after α-catenin depletion in HaCaT keratinocytes.


Subject(s)
Adherens Junctions , Keratinocytes , Cell Movement , Cell Nucleus , Wound Healing
7.
Mol Biol Cell ; 32(4): 348-361, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33378218

ABSTRACT

Sustained cell migration is essential for wound healing and cancer metastasis. The epidermal growth factor receptor (EGFR) signaling cascade is known to drive cell migration and proliferation. While the signal transduction downstream of EGFR has been extensively investigated, our knowledge of the initiation and maintenance of EGFR signaling during cell migration remains limited. The metalloprotease TACE (tumor necrosis factor alpha converting enzyme) is responsible for producing active EGFR family ligands in the via ligand shedding. Sustained TACE activity may perpetuate EGFR signaling and reduce a cell's reliance on exogenous growth factors. Using a cultured keratinocyte model system, we show that depletion of α-catenin perturbs adherens junctions, enhances cell proliferation and motility, and decreases dependence on exogenous growth factors. We show that the underlying mechanism for these observed phenotypical changes depends on enhanced autocrine/paracrine release of the EGFR ligand transforming growth factor alpha in a TACE-dependent manner. We demonstrate that proliferating keratinocyte epithelial cell clusters display waves of oscillatory extracellular signal-regulated kinase (ERK) activity, which can be eliminated by TACE knockout, suggesting that these waves of oscillatory ERK activity depend on autocrine/paracrine signals produced by TACE. These results provide new insights into the regulatory role of adherens junctions in initiating and maintaining autocrine/paracrine signaling with relevance to wound healing and cellular transformation.


Subject(s)
ADAM17 Protein/metabolism , Adherens Junctions/metabolism , alpha Catenin/metabolism , ADAM17 Protein/physiology , Adherens Junctions/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , ErbB Receptors/metabolism , HaCaT Cells , Humans , Metalloproteases/metabolism , Paracrine Communication/physiology , Phosphorylation , Signal Transduction , Transforming Growth Factor alpha/metabolism , alpha Catenin/physiology
8.
JCI Insight ; 5(13)2020 07 09.
Article in English | MEDLINE | ID: mdl-32641584

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is a neuromuscular disorder caused by a polyglutamine expansion in the androgen receptor (AR). Using gene expression analysis and ChIP sequencing, we mapped transcriptional changes in genetically engineered patient stem cell-derived motor neurons. We found that transcriptional dysregulation in SBMA can occur through AR-mediated histone modification. We detected reduced histone acetylation, along with decreased expression of genes encoding compensatory metabolic proteins and reduced substrate availability for mitochondrial function. Furthermore, we found that pyruvate supplementation corrected this deficiency and improved mitochondrial function and SBMA motor neuron viability. We propose that epigenetic dysregulation of metabolic genes contributes to reduced mitochondrial ATP production. Our results show a molecular link between altered epigenetic regulation and mitochondrial metabolism that contributes to neurodegeneration.


Subject(s)
Epigenesis, Genetic/physiology , Mitochondria/metabolism , Motor Neurons/metabolism , Muscular Atrophy, Spinal/physiopathology , Humans , Muscular Atrophy, Spinal/metabolism , Peptides/metabolism , Receptors, Androgen/metabolism
9.
Methods Mol Biol ; 1978: 343-353, 2019.
Article in English | MEDLINE | ID: mdl-31119673

ABSTRACT

Living cells employ complex and highly dynamic signaling networks and transcriptional circuits to maintain homeostasis and respond appropriately to constantly changing environments. These networks enable cells to maintain tight control on intracellular concentrations of ions, metabolites, proteins, and other biomolecules and ensure a careful balance between a cell's energetic needs and catabolic processes required for growth. Establishing molecular mechanisms of genetic and pharmacological perturbations remains challenging, due to the interconnected nature of these networks and the extreme sensitivity of cellular systems to their external environment. Live cell imaging with genetically encoded fluorescent biosensors provides a powerful new modality for nondestructive spatiotemporal tracking of ions, small molecules, enzymatic activities, and molecular interactions in living systems, from cells, tissues, and even living organisms. By deploying large panels of cell lines, each with distinct biosensors, many critical biochemical pathways can be monitored in a highly parallel and high-throughput fashion to identify pharmacological vulnerabilities and combination therapies unique to a given cell type or genetic background. Here we describe the experimental and analytical methods required to conduct multiplexed parallel fluorescence microscopy experiments on live cells expressing stable transgenic synthetic protein biosensors.


Subject(s)
Biosensing Techniques/methods , Enzymes/chemistry , Microscopy, Fluorescence/methods , Proteins/chemistry , Enzymes/genetics , Fluorescence Resonance Energy Transfer , Ions/chemistry , Proteins/genetics , Signal Transduction/genetics
10.
Cell Death Differ ; 26(12): 2682-2694, 2019 12.
Article in English | MEDLINE | ID: mdl-30976095

ABSTRACT

Bok (Bcl-2-related ovarian killer) is a member of the Bcl-2 protein family that governs the intrinsic apoptosis pathway, but the cellular role that Bok plays is controversial. Remarkably, endogenous Bok is constitutively bound to inositol 1,4,5-trisphosphate receptors (IP3Rs) and is stabilized by this interaction. Here we report that despite the strong association with IP3Rs, deletion of Bok expression by CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein-9 nuclease)-mediated gene editing does not alter calcium mobilization via IP3Rs or calcium influx into the mitochondria. Rather, Bok deletion significantly reduces mitochondrial fusion rate, resulting in mitochondrial fragmentation. This phenotype is reversed by exogenous wild-type Bok and by an IP3R binding-deficient Bok mutant, and may result from a decrease in mitochondrial motility. Bok deletion also enhances mitochondrial spare respiratory capacity and membrane potential. Finally, Bok does not play a major role in apoptotic signaling, since Bok deletion does not alter responsiveness to various apoptotic stimuli. Overall, despite binding to IP3Rs, Bok does not alter IP3R-mediated Ca2+ signaling, but is required to maintain normal mitochondrial fusion, morphology, and bioenergetics.


Subject(s)
Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Calcium Signaling , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Knockout , Oxygen Consumption , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics
11.
Mol Cell ; 74(2): 347-362.e6, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30853401

ABSTRACT

Selective autophagy recycles damaged organelles and clears intracellular pathogens to prevent their aberrant accumulation. How ULK1 kinase is targeted and activated during selective autophagic events remains to be elucidated. In this study, we used chemically inducible dimerization (CID) assays in tandem with CRISPR KO lines to systematically analyze the molecular basis of selective autophagosome biogenesis. We demonstrate that ectopic placement of NDP52 on mitochondria or peroxisomes is sufficient to initiate selective autophagy by focally localizing and activating the ULK1 complex. The capability of NDP52 to induce mitophagy is dependent on its interaction with the FIP200/ULK1 complex, which is facilitated by TBK1. Ectopically tethering ULK1 to cargo bypasses the requirement for autophagy receptors and TBK1. Focal activation of ULK1 occurs independently of AMPK and mTOR. Our findings provide a parsimonious model of selective autophagy, which highlights the coordination of ULK1 complex localization by autophagy receptors and TBK1 as principal drivers of targeted autophagosome biogenesis.


Subject(s)
Autophagy-Related Protein-1 Homolog/genetics , Autophagy/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Autophagy-Related Proteins , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , HeLa Cells , Humans , Mitochondria/chemistry , Mitochondria/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Peroxisomes/chemistry , Peroxisomes/genetics , Phosphorylation , Protein Kinases/genetics , Protein Multimerization , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
12.
J Biol Chem ; 294(18): 7472-7487, 2019 05 03.
Article in English | MEDLINE | ID: mdl-30885942

ABSTRACT

A critical function of the PTEN-induced kinase 1 (PINK1)-Parkin pathway is to mediate the clearing of unhealthy or damaged mitochondria via mitophagy. Loss of either PINK1 or Parkin protein expression is associated with Parkinson's disease. Here, using a high-throughput screening approach along with recombinant protein expression and kinase, immunoblotting, and immunofluorescence live-cell imaging assays, we report that celastrol, a pentacyclic triterpenoid isolated from extracts of the medicinal plant Tripterygium wilfordii, blocks recruitment pof Parkin to mitochondria, preventing mitophagy in response to mitochondrial depolarization induced by carbonyl cyanide m-chlorophenylhydrazone or to gamitrinib-induced inhibition of mitochondrial heat shock protein 90 (HSP90). Celastrol's effect on mitophagy was independent of its known role in microtubule disruption. Instead, we show that celastrol suppresses Parkin recruitment by inactivating PINK1 and preventing it from phosphorylating Parkin and also ubiquitin. We also observed that PINK1 directly and strongly associates with TOM20, a component of the translocase of outer mitochondrial membrane (TOM) machinery and relatively weak binding to another TOM subunit, TOM70. Moreover, celastrol disrupted binding between PINK1 and TOM20 both in vitro and in vivo but did not affect binding between TOM20 and TOM70. Using native gel analysis, we also show that celastrol disrupts PINK1 complex formation upon mitochondrial depolarization and sequesters PINK1 to high-molecular-weight protein aggregates. These results reveal that celastrol regulates the mitochondrial quality control pathway by interfering with PINK1-TOM20 binding.


Subject(s)
Membrane Transport Proteins/metabolism , Mitophagy/drug effects , Protein Kinases/metabolism , Receptors, Cell Surface/metabolism , Tripterygium/chemistry , Triterpenes/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HeLa Cells , Humans , Hydrazones/antagonists & inhibitors , Hydrazones/pharmacology , Membrane Potential, Mitochondrial/drug effects , Microtubules/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Pentacyclic Triterpenes , Polymerization , Protein Binding , Ubiquitin-Protein Ligases/metabolism
13.
Mol Cell ; 73(5): 1028-1043.e5, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30733118

ABSTRACT

Mutations in PTEN-induced kinase 1 (PINK1) can cause recessive early-onset Parkinson's disease (PD). Import arrest results in PINK1 kinase activation specifically on damaged mitochondria, triggering Parkin-mediated mitophagy. Here, we show that PINK1 import is less dependent on Tim23 than on mitochondrial membrane potential (ΔΨm). We identified a negatively charged amino acid cluster motif that is evolutionarily conserved just C-terminal to the PINK1 transmembrane. PINK1 that fails to accumulate at the outer mitochondrial membrane, either by mutagenesis of this negatively charged motif or by deletion of Tom7, is imported into depolarized mitochondria and cleaved by the OMA1 protease. Some PD patient mutations also are defective in import arrest and are rescued by the suppression of OMA1, providing a new potential druggable target for PD. These results suggest that ΔΨm loss-dependent PINK1 import arrest does not result solely from Tim23 inactivation but also through an actively regulated "tug of war" between Tom7 and OMA1.


Subject(s)
Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Mitochondria/enzymology , Mitochondrial Membranes/enzymology , Mitochondrial Proteins/metabolism , Parkinson Disease/enzymology , Protein Kinases/metabolism , Amino Acid Motifs , Antiparkinson Agents/pharmacology , Biological Transport , Drug Design , Enzyme Activation , HeLa Cells , Humans , Membrane Potential, Mitochondrial , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/genetics , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Protein Interaction Domains and Motifs , Protein Kinases/genetics , Proteolysis , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
Proc Natl Acad Sci U S A ; 115(2): 319-324, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29279382

ABSTRACT

Ubiquitin-dependent proteolysis of cyclin D1 is associated with normal and tumor cell proliferation and survival. The SCFFBXO31 (Skp1-Cul1-Rbx1-FBXO31) ubiquitin ligase complex mediates genotoxic stress-induced cyclin D1 degradation. Previous studies have suggested that cyclin D1 levels are maintained at steady state by phosphorylation-dependent nuclear export and subsequent proteolysis in the cytoplasm. Here we present the crystal structures of the Skp1-FBXO31 complex alone and bound to a phosphorylated cyclin D1 C-terminal peptide. FBXO31 possesses a unique substrate-binding domain consisting of two ß-barrel motifs, whereas cyclin D1 binds to FBXO31 by tucking its free C-terminal carboxylate tail into an open cavity of the C-terminal FBXO31 ß-barrel. Biophysical and functional studies demonstrate that SCFFBXO31 is capable of recruiting and ubiquitinating cyclin D1 in a phosphorylation-independent manner. Our findings provide a conceptual framework for understanding the substrate specificity of the F-box protein FBXO31 and the mechanism of FBXO31-regulated cyclin D1 protein turnover.


Subject(s)
Cyclin D1/chemistry , F-Box Proteins/chemistry , Multiprotein Complexes/chemistry , Protein Domains , Tumor Suppressor Proteins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Cyclin D1/genetics , Cyclin D1/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , HeLa Cells , Humans , Models, Molecular , Multiprotein Complexes/metabolism , Phosphorylation , Protein Binding , Proteolysis , Sequence Homology, Amino Acid , Substrate Specificity , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitination
15.
Nucleic Acids Res ; 46(4): 1756-1776, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29240919

ABSTRACT

Histone deacetylase inhibitors (HDACIs) are known to alter gene expression by both up- and down-regulation of protein-coding genes in normal and cancer cells. However, the exact regulatory mechanisms of action remain uncharacterized. Here we investigated genome wide dose-dependent epigenetic and transcriptome changes in response to HDACI largazole in a transformed and a non-transformed cell line. Exposure to low nanomolar largazole concentrations (

Subject(s)
Depsipeptides/pharmacology , Enhancer Elements, Genetic , Histone Code/drug effects , Histone Deacetylase Inhibitors/pharmacology , Thiazoles/pharmacology , Acetylation , Cell Line , Cell Line, Transformed , Cytostatic Agents/pharmacology , Dose-Response Relationship, Drug , Enhancer Elements, Genetic/drug effects , Genome , Histone Deacetylases/physiology , Histones/metabolism , Oncogenes , Promoter Regions, Genetic , RNA Polymerase II/metabolism , RNA, Messenger/metabolism
16.
J Biol Chem ; 292(36): 15105-15120, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28673964

ABSTRACT

Sorafenib (Nexavar) is a broad-spectrum multikinase inhibitor that proves effective in treating advanced renal-cell carcinoma and liver cancer. Despite its well-characterized mechanism of action on several established cancer-related protein kinases, sorafenib causes variable responses among human tumors, although the cause for this variation is unknown. In an unbiased screening of an oncology drug library, we found that sorafenib activates recruitment of the ubiquitin E3 ligase Parkin to damaged mitochondria. We show that sorafenib inhibits the activity of both complex II/III of the electron transport chain and ATP synthase. Dual inhibition of these complexes, but not inhibition of each individual complex, stabilizes the serine-threonine protein kinase PINK1 on the mitochondrial outer membrane and activates Parkin. Unlike the protonophore carbonyl cyanide m-chlorophenylhydrazone, which activates the mitophagy response, sorafenib treatment triggers PINK1/Parkin-dependent cellular apoptosis, which is attenuated upon Bcl-2 overexpression. In summary, our results reveal a new mechanism of action for sorafenib as a mitocan and suggest that high Parkin activity levels could make tumor cells more sensitive to sorafenib's actions, providing one possible explanation why Parkin may be a tumor suppressor gene. These insights could be useful in developing new rationally designed combination therapies with sorafenib.


Subject(s)
Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex II/antagonists & inhibitors , Mitochondria/drug effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Cells, Cultured , Electron Transport/drug effects , Electron Transport Complex II/metabolism , Electron Transport Complex III/metabolism , HEK293 Cells , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Niacinamide/pharmacology , Sorafenib
17.
Cell Microbiol ; 19(1)2017 01.
Article in English | MEDLINE | ID: mdl-27376507

ABSTRACT

Salmonella Typhimurium is an intracellular bacterial pathogen that infects both epithelial cells and macrophages. Salmonella effector proteins, which are translocated into the host cell and manipulate host cell components, control the ability to replicate and/or survive in host cells. Due to the complexity and heterogeneity of Salmonella infections, there is growing recognition of the need for single-cell and live-cell imaging approaches to identify and characterize the diversity of cellular phenotypes and how they evolve over time. Here, we establish a pipeline for long-term (17 h) live-cell imaging of infected cells and subsequent image analysis methods. We apply this pipeline to track bacterial replication within the Salmonella-containing vacuole in epithelial cells, quantify vacuolar replication versus survival in macrophages and investigate the role of individual effector proteins in mediating these parameters. This approach revealed that dispersed bacteria can coalesce at later stages of infection, that the effector protein SseG influences the propensity for cytosolic hyper-replication in epithelial cells, and that while SteA only has a subtle effect on vacuolar replication in epithelial cells, it has a profound impact on infection parameters in immunocompetent macrophages, suggesting differential roles for effector proteins in different infection models.


Subject(s)
Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Macrophages/microbiology , Salmonella typhimurium/pathogenicity , Vacuoles/microbiology , Virulence Factors/metabolism , Image Processing, Computer-Assisted , Microbial Viability , Optical Imaging , Salmonella typhimurium/growth & development , Single-Cell Analysis
18.
Nucleic Acids Res ; 44(8): 3788-800, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27034466

ABSTRACT

The Musashi-2 (Msi2) RNA-binding protein maintains stem cell self-renewal and promotes oncogenesis by enhancing cell proliferation in hematopoietic and gastrointestinal tissues. However, it is unclear how Msi2 recognizes and regulates mRNA targets in vivo and whether Msi2 primarily controls cell growth in all cell types. Here we identified Msi2 targets with HITS-CLIP and revealed that Msi2 primarily recognizes mRNA 3'UTRs at sites enriched in multiple copies of UAG motifs in epithelial progenitor cells. RNA-seq and ribosome profiling demonstrated that Msi2 promotes targeted mRNA decay without affecting translation efficiency. Unexpectedly, the most prominent Msi2 targets identified are key regulators that govern cell motility with a high enrichment in focal adhesion and extracellular matrix-receptor interaction, in addition to regulators of cell growth and survival. Loss of Msi2 stimulates epithelial cell migration, increases the number of focal adhesions and also compromises cell growth. These findings provide new insights into the molecular mechanisms of Msi2's recognition and repression of targets and uncover a key function of Msi2 in restricting epithelial cell migration.


Subject(s)
Cell Movement/genetics , Gene Expression Regulation , Keratinocytes/physiology , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Animals , Binding Sites , Cell Proliferation/genetics , Cell Survival , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Immunoprecipitation , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Sequence Analysis, RNA , Transcriptome
19.
Sci Signal ; 8(365): rs1, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25714465

ABSTRACT

Diverse environmental conditions stimulate protein "shedding" from the cell surface through proteolytic cleavage. The protease TACE [tumor necrosis factor-α (TNFα)--converting enzyme, encoded by ADAM17] mediates protein shedding, thereby regulating the maturation and release of various extracellular substrates, such as growth factors and cytokines, that induce diverse cellular responses. We developed a FRET (fluorescence resonance energy transfer)-based biosensor called TSen that quantitatively reports the kinetics of TACE activity in live cells. In combination with chemical biology approaches, we used TSen to probe the dependence of TACE activation on the induction of the kinases p38 and ERK (extracellular signal-regulated kinase) in various epithelial cell lines. Using TSen, we found that disruption of the actin cytoskeleton in keratinocytes induced rapid and robust TSen cleavage and the accumulation of TACE at the plasma membrane. Cytoskeletal disruption also increased the cleavage of endogenous TACE substrates, including transforming growth factor-α. Thus, TSen is a useful tool for unraveling the mechanisms underlying the spatiotemporal activation of TACE in live cells.


Subject(s)
ADAM Proteins/metabolism , Biosensing Techniques/methods , Epithelial Cells/enzymology , Fluorescence Resonance Energy Transfer/methods , Keratinocytes/enzymology , ADAM Proteins/genetics , ADAM17 Protein , Animals , Cytoskeleton/genetics , Cytoskeleton/metabolism , Enzyme Activation/genetics , Epithelial Cells/cytology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Keratinocytes/cytology , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL