Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.867
Filter
1.
Nat Commun ; 15(1): 3978, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38729926

ABSTRACT

A key mechanism employed by plants to adapt to salinity stress involves maintaining ion homeostasis via the actions of ion transporters. While the function of cation transporters in maintaining ion homeostasis in plants has been extensively studied, little is known about the roles of their anion counterparts in this process. Here, we describe a mechanism of salt adaptation in plants. We characterized the chloride channel (CLC) gene AtCLCf, whose expression is regulated by WRKY transcription factor under salt stress in Arabidopsis thaliana. Loss-of-function atclcf seedlings show increased sensitivity to salt, whereas AtCLCf overexpression confers enhanced resistance to salt stress. Salt stress induces the translocation of GFP-AtCLCf fusion protein to the plasma membrane (PM). Blocking AtCLCf translocation using the exocytosis inhibitor brefeldin-A or mutating the small GTPase gene AtRABA1b/BEX5 (RAS GENES FROM RAT BRAINA1b homolog) increases salt sensitivity in plants. Electrophysiology and liposome-based assays confirm the Cl-/H+ antiport function of AtCLCf. Therefore, we have uncovered a mechanism of plant adaptation to salt stress involving the NaCl-induced translocation of AtCLCf to the PM, thus facilitating Cl- removal at the roots, and increasing the plant's salinity tolerance.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cell Membrane , Chloride Channels , Golgi Apparatus , Salt Stress , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/physiology , Arabidopsis/drug effects , Cell Membrane/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Golgi Apparatus/metabolism , Chloride Channels/metabolism , Chloride Channels/genetics , Gene Expression Regulation, Plant , Protein Transport/drug effects , Salt Tolerance/genetics , Sodium Chloride/pharmacology , Plants, Genetically Modified
2.
Nature ; 626(8000): 874-880, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38297121

ABSTRACT

Stress response pathways detect and alleviate adverse conditions to safeguard cell and tissue homeostasis, yet their prolonged activation induces apoptosis and disrupts organismal health1-3. How stress responses are turned off at the right time and place remains poorly understood. Here we report a ubiquitin-dependent mechanism that silences the cellular response to mitochondrial protein import stress. Crucial to this process is the silencing factor of the integrated stress response (SIFI), a large E3 ligase complex mutated in ataxia and in early-onset dementia that degrades both unimported mitochondrial precursors and stress response components. By recognizing bifunctional substrate motifs that equally encode protein localization and stability, the SIFI complex turns off a general stress response after a specific stress event has been resolved. Pharmacological stress response silencing sustains cell survival even if stress resolution failed, which underscores the importance of signal termination and provides a roadmap for treating neurodegenerative diseases caused by mitochondrial import defects.


Subject(s)
Mitochondria , Mitochondrial Proteins , Mutation , Neurodegenerative Diseases , Stress, Physiological , Ubiquitin-Protein Ligases , Apoptosis/drug effects , Ataxia/genetics , Cell Survival/drug effects , Dementia/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Stability/drug effects , Protein Transport/drug effects , Proteolysis/drug effects , Stress, Physiological/drug effects , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
3.
J Biol Chem ; 299(11): 105269, 2023 11.
Article in English | MEDLINE | ID: mdl-37739036

ABSTRACT

Prenylation is an irreversible post-translational modification that supports membrane interactions of proteins involved in various cellular processes, including migration, proliferation, and survival. Dysregulation of prenylation contributes to multiple disorders, including cancers and vascular and neurodegenerative diseases. Prenyltransferases tether isoprenoid lipids to proteins via a thioether linkage during prenylation. Pharmacological inhibition of the lipid synthesis pathway by statins is a therapeutic approach to control hyperlipidemia. Building on our previous finding that statins inhibit membrane association of G protein γ (Gγ) in a subtype-dependent manner, we investigated the molecular reasoning for this differential inhibition. We examined the prenylation of carboxy-terminus (Ct) mutated Gγ in cells exposed to Fluvastatin and prenyl transferase inhibitors and monitored the subcellular localization of fluorescently tagged Gγ subunits and their mutants using live-cell confocal imaging. Reversible optogenetic unmasking-masking of Ct residues was used to probe their contribution to prenylation and membrane interactions of the prenylated proteins. Our findings suggest that specific Ct residues regulate membrane interactions of the Gγ polypeptide, statin sensitivity, and extent of prenylation. Our results also show a few hydrophobic and charged residues at the Ct are crucial determinants of a protein's prenylation ability, especially under suboptimal conditions. Given the cell and tissue-specific expression of different Gγ subtypes, our findings indicate a plausible mechanism allowing for statins to differentially perturb heterotrimeric G protein signaling in cells depending on their Gγ-subtype composition. Our results may also provide molecular reasoning for repurposing statins as Ras oncogene inhibitors and the failure of using prenyltransferase inhibitors in cancer treatment.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Protein Prenylation , Humans , Amino Acid Motifs , Drug Resistance/genetics , HeLa Cells , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Models, Molecular , Mutation , Protein Prenylation/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Signal Transduction/drug effects
4.
Int J Mol Sci ; 24(5)2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36902257

ABSTRACT

Glycosylphosphatidylinositol-anchored proteins (GPI-APs) are anchored at the outer leaflet of eukaryotic plasma membranes (PMs) only by carboxy-terminal covalently coupled GPI. GPI-APs are known to be released from the surface of donor cells in response to insulin and antidiabetic sulfonylureas (SUs) by lipolytic cleavage of the GPI or upon metabolic derangement as full-length GPI-APs with the complete GPI attached. Full-length GPI-APs become removed from extracellular compartments by binding to serum proteins, such as GPI-specific phospholipase D (GPLD1), or insertion into the PMs of acceptor cells. Here, the interplay between the lipolytic release and intercellular transfer of GPI-APs and its potential functional impact was studied using transwell co-culture with human adipocytes as insulin-/SU-responsive donor cells and GPI-deficient erythroleukemia as acceptor cells (ELCs). Measurement of the transfer as the expression of full-length GPI-APs at the ELC PMs by their microfluidic chip-based sensing with GPI-binding α-toxin and GPI-APs antibodies and of the ELC anabolic state as glycogen synthesis upon incubation with insulin, SUs and serum yielded the following results: (i) Loss of GPI-APs from the PM upon termination of their transfer and decline of glycogen synthesis in ELCs, as well as prolongation of the PM expression of transferred GPI-APs upon inhibition of their endocytosis and upregulated glycogen synthesis follow similar time courses. (ii) Insulin and SUs inhibit both GPI-AP transfer and glycogen synthesis upregulation in a concentration-dependent fashion, with the efficacies of the SUs increasing with their blood glucose-lowering activity. (iii) Serum from rats eliminates insulin- and SU-inhibition of both GPI-APs' transfer and glycogen synthesis in a volume-dependent fashion, with the potency increasing with their metabolic derangement. (iv) In rat serum, full-length GPI-APs bind to proteins, among them (inhibited) GPLD1, with the efficacy increasing with the metabolic derangement. (v) GPI-APs are displaced from serum proteins by synthetic phosphoinositolglycans and then transferred to ELCs with accompanying stimulation of glycogen synthesis, each with efficacies increasing with their structural similarity to the GPI glycan core. Thus, both insulin and SUs either block or foster transfer when serum proteins are depleted of or loaded with full-length GPI-APs, respectively, i.e., in the normal or metabolically deranged state. The transfer of the anabolic state from somatic to blood cells over long distance and its "indirect" complex control by insulin, SUs and serum proteins support the (patho)physiological relevance of the intercellular transfer of GPI-APs.


Subject(s)
Adipocytes , Adipose Tissue , Blood Cells , Glycosylphosphatidylinositols , Hypoglycemic Agents , Insulin , Sulfonylurea Compounds , Animals , Humans , Rats , Blood Cells/metabolism , Glycogen/metabolism , Glycosylphosphatidylinositols/metabolism , Insulin/pharmacology , Sulfonylurea Compounds/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Protein Transport/drug effects , Hypoglycemic Agents/pharmacology , Adipocytes/drug effects , Coculture Techniques
5.
EBioMedicine ; 89: 104451, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36738481

ABSTRACT

BACKGROUND: Vacuolar protein sorting-associated protein 35 (VPS35) is a core component of the retromer complex which mediates intracellular protein transport. It is well known that dysfunctional VPS35 functions in the accumulation of pathogenic proteins. In our previous study, VPS35 was found to be a potential gene related to poor prognosis in gastric cancer. However, the biological functions of VPS35 in gastric cancer remain unclear. METHODS: Cell viability assays were performed to examine whether VPS35 affected cell proliferation. Immunoprecipitation and biotin assays showed that VPS35 bound to epidermal growth factor receptor (EGFR) in the cytoplasm and recycled it to the cell surface. Patient-derived xenografts and organoids were used to evaluate the effect of VPS35 on the response of gastric cancer to EGFR inhibitors. FINDINGS: VPS35 expression levels were upregulated in tumour tissues and correlated with local tumour invasion and poor survival in patients with gastric cancer. VPS35 promoted cell proliferation and increased tumour growth. Mechanistically, VPS35 selectively bound to endocytosed EGFR in early endosomes and recycled it back to the cell surface, leading to the downstream activation of the ERK1/2 pathway. We also found that high VPS35 expression levels increased the sensitivity of the xenograft and organoid models to EGFR inhibitors. INTERPRETATION: VPS35 promotes cell proliferation by recycling EGFR to the cell surface, amplifying the network of receptor trafficking. VPS35 expression levels are positively correlated with gastric cancer sensitivity to EGFR inhibitors, which offers a potential method to stratify patients for EGFR inhibitor utilisation. FUNDING: National Natural Science Foundation of China.


Subject(s)
Stomach Neoplasms , Vesicular Transport Proteins , Humans , Carrier Proteins/metabolism , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Protein Transport/drug effects , Protein Transport/genetics , Stomach Neoplasms/genetics , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
6.
Proc Natl Acad Sci U S A ; 119(25): e2122477119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35700362

ABSTRACT

Alcohol intoxication at early ages is a risk factor for the development of addictive behavior. To uncover neuronal molecular correlates of acute ethanol intoxication, we used stable-isotope-labeled mice combined with quantitative mass spectrometry to screen more than 2,000 hippocampal proteins, of which 72 changed synaptic abundance up to twofold after ethanol exposure. Among those were mitochondrial proteins and proteins important for neuronal morphology, including MAP6 and ankyrin-G. Based on these candidate proteins, we found acute and lasting molecular, cellular, and behavioral changes following a single intoxication in alcohol-naïve mice. Immunofluorescence analysis revealed a shortening of axon initial segments. Longitudinal two-photon in vivo imaging showed increased synaptic dynamics and mitochondrial trafficking in axons. Knockdown of mitochondrial trafficking in dopaminergic neurons abolished conditioned alcohol preference in Drosophila flies. This study introduces mitochondrial trafficking as a process implicated in reward learning and highlights the potential of high-resolution proteomics to identify cellular mechanisms relevant for addictive behavior.


Subject(s)
Alcoholic Intoxication , Dopaminergic Neurons , Ethanol , Hippocampus , Nerve Tissue Proteins , Alcoholic Intoxication/metabolism , Alcoholic Intoxication/pathology , Animals , Behavior, Addictive/chemically induced , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dose-Response Relationship, Drug , Drosophila melanogaster , Ethanol/administration & dosage , Ethanol/toxicity , Gene Knockdown Techniques , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mitochondria/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Transport/drug effects
7.
Cell Mol Life Sci ; 79(4): 193, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35298717

ABSTRACT

Aberrant insulin-like growth factor 1 (IGF-1) signaling has been proposed as a contributing factor to the development of neurodegenerative disorders including diabetic neuropathy, and delivery of exogenous IGF-1 has been explored as a treatment for Alzheimer's disease and amyotrophic lateral sclerosis. However, the role of autocrine/paracrine IGF-1 in neuroprotection has not been well established. We therefore used in vitro cell culture systems and animal models of diabetic neuropathy to characterize endogenous IGF-1 in sensory neurons and determine the factors regulating IGF-1 expression and/or affecting neuronal health. Single-cell RNA sequencing (scRNA-Seq) and in situ hybridization analyses revealed high expression of endogenous IGF-1 in non-peptidergic neurons and satellite glial cells (SGCs) of dorsal root ganglia (DRG). Brain cortex and DRG had higher IGF-1 gene expression than sciatic nerve. Bidirectional transport of IGF-1 along sensory nerves was observed. Despite no difference in IGF-1 receptor levels, IGF-1 gene expression was significantly (P < 0.05) reduced in liver and DRG from streptozotocin (STZ)-induced type 1 diabetic rats, Zucker diabetic fatty (ZDF) rats, mice on a high-fat/ high-sugar diet and db/db type 2 diabetic mice. Hyperglycemia suppressed IGF-1 gene expression in cultured DRG neurons and this was reversed by exogenous IGF-1 or the aldose reductase inhibitor sorbinil. Transcription factors, such as NFAT1 and CEBPß, were also less enriched at the IGF-1 promoter in DRG from diabetic rats vs control rats. CEBPß overexpression promoted neurite outgrowth and mitochondrial respiration, both of which were blunted by knocking down or blocking IGF-1. Suppression of endogenous IGF-1 in diabetes may contribute to neuropathy and its upregulation at the transcriptional level by CEBPß can be a promising therapeutic approach.


Subject(s)
Aging/metabolism , Axons/pathology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Energy Metabolism , Insulin-Like Growth Factor I/metabolism , Sensory Receptor Cells/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Axons/drug effects , Axons/metabolism , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Respiration/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Energy Metabolism/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Glycolysis/drug effects , HEK293 Cells , Humans , Insulin-Like Growth Factor I/genetics , Liver/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , NFATC Transcription Factors/metabolism , Neuronal Outgrowth/drug effects , Polymers/metabolism , Promoter Regions, Genetic/genetics , Protein Transport/drug effects , Rats, Sprague-Dawley , Sensory Receptor Cells/pathology , Signal Transduction/drug effects
8.
Cell Rep ; 38(7): 110396, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172151

ABSTRACT

Fasciculation and elongation protein zeta-1 (FEZ1) is a multifunctional kinesin adaptor involved in processes ranging from neurodegeneration to retrovirus and polyomavirus infection. Here, we show that, although modulating FEZ1 expression also impacts infection by large DNA viruses in human microglia, macrophages, and fibroblasts, this broad antiviral phenotype is associated with the pre-induction of interferon-stimulated genes (ISGs) in a STING-independent manner. We further reveal that S58, a key phosphorylation site in FEZ1's kinesin regulatory domain, controls both binding to, and the nuclear-cytoplasmic localization of, heat shock protein 8 (HSPA8), as well as ISG expression. FEZ1- and HSPA8-induced changes in ISG expression further involved changes in DNA-dependent protein kinase (DNA-PK) accumulation in the nucleus. Moreover, phosphorylation of endogenous FEZ1 at S58 was reduced and HSPA8 and DNA-PK translocated to the nucleus in cells stimulated with DNA, suggesting that FEZ1 is a regulatory component of the recently identified HSPA8/DNA-PK innate immune pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation , HSC70 Heat-Shock Proteins/metabolism , Interferons/pharmacology , Nerve Tissue Proteins/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chlorocebus aethiops , DNA Viruses/physiology , DNA-Activated Protein Kinase/metabolism , Female , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Immunity, Innate/drug effects , Interferon Regulatory Factors/metabolism , Membrane Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Vero Cells
9.
Cell Mol Life Sci ; 79(2): 120, 2022 Feb 04.
Article in English | MEDLINE | ID: mdl-35119496

ABSTRACT

Loss of neuronal polarity and missorting of the axonal microtubule-associated-protein TAU are hallmarks of Alzheimer's disease (AD) and related tauopathies. Impairment of mitochondrial function is causative for various mitochondriopathies, but the role of mitochondria in tauopathies and in axonal TAU-sorting is unclear. The axon-initial-segment (AIS) is vital for maintaining neuronal polarity, action potential generation, and-here important-TAU-sorting. Here, we investigate the role of mitochondria in the AIS for maintenance of TAU cellular polarity. Using not only global and local mitochondria impairment via inhibitors of the respiratory chain and a locally activatable protonophore/uncoupler, but also live-cell-imaging and photoconversion methods, we specifically tracked and selectively impaired mitochondria in the AIS in primary mouse and human iPSC-derived forebrain/cortical neurons, and assessed somatic presence of TAU. Global application of mitochondrial toxins efficiently induced tauopathy-like TAU-missorting, indicating involvement of mitochondria in TAU-polarity. Mitochondria show a biased distribution within the AIS, with a proximal cluster and relative absence in the central AIS. The mitochondria of this cluster are largely immobile and only sparsely participate in axonal mitochondria-trafficking. Locally constricted impairment of the AIS-mitochondria-cluster leads to detectable increases of somatic TAU, reminiscent of AD-like TAU-missorting. Mechanistically, mitochondrial impairment sufficient to induce TAU-missorting results in decreases of calcium oscillation but increases in baseline calcium, yet chelating intracellular calcium did not prevent mitochondrial impairment-induced TAU-missorting. Stabilizing microtubules via taxol prevented TAU-missorting, hinting towards a role for impaired microtubule dynamics in mitochondrial-dysfunction-induced TAU-missorting. We provide evidence that the mitochondrial distribution within the proximal axon is biased towards the proximal AIS and that proper function of this newly described mitochondrial cluster may be essential for the maintenance of TAU polarity. Mitochondrial impairment may be an upstream event in and therapeutic target for AD/tauopathy.


Subject(s)
Axons/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Calcium/metabolism , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Microtubules/metabolism , Mitochondria/pathology , Neurons/cytology , Protein Transport/drug effects , Rotenone/pharmacology
10.
Biochem J ; 479(3): 445-462, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35147164

ABSTRACT

Insulin rapidly stimulates GLUT4 translocation and glucose transport in fat and muscle cells. Signals from the occupied insulin receptor are translated into downstream signalling changes in serine/threonine kinases within timescales of seconds, and this is followed by delivery and accumulation of the glucose transporter GLUT4 at the plasma membrane. Kinetic studies have led to realisation that there are distinct phases of this stimulation by insulin. There is a rapid initial burst of GLUT4 delivered to the cell surface from a subcellular reservoir compartment and this is followed by a steady-state level of continuing stimulation in which GLUT4 recycles through a large itinerary of subcellular locations. Here, we provide an overview of the phases of insulin stimulation of GLUT4 translocation and the molecules that are currently considered to activate these trafficking steps. Furthermore, we suggest how use of new experimental approaches together with phospho-proteomic data may help to further identify mechanisms for activation of these trafficking processes.


Subject(s)
Glucose Transporter Type 4/physiology , Adipocytes/metabolism , Animals , Cell Membrane/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Insulin/pharmacology , Models, Biological , Muscle Cells/metabolism , Phosphorylation , Protein Processing, Post-Translational , Protein Transport/drug effects , Signal Transduction , Subcellular Fractions/metabolism
11.
Sci Rep ; 12(1): 1054, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058496

ABSTRACT

Neonatal microcephaly and adult Guillain-Barré syndrome are severe complications of Zika virus (ZIKV) infection. The robustly induced inflammatory cytokine expressions in ZIKV-infected patients may constitute a hallmark for severe disease. In the present study, the potential role of high mobility group box 1 protein (HMGB1) in ZIKV infection was investigated. HMGB1 protein expression was determined by the enzyme-linked immunosorbent assay (ELISA) and immunoblot assay. HMGB1's role in ZIKV infection was also explored using treatment with dexamethasone, an immunomodulatory drug, and HMGB1-knockdown (shHMGB1) Huh7 cells. Results showed that the Huh7 cells were highly susceptible to ZIKV infection. The infection was found to induce HMGB1 nuclear-to-cytoplasmic translocation, resulting in a > 99% increase in the cytosolic HMGB1 expression at 72-h post-infection (h.p.i). The extracellular HMGB1 level was elevated in a time- and multiplicity of infection (MOI)-dependent manner. Treatment of the ZIKV-infected cells with dexamethasone (150 µM) reduced HMGB1 extracellular release in a dose-dependent manner, with a maximum reduction of 71 ± 5.84% (P < 0.01). The treatment also reduced virus titers by over 83 ± 0.50% (P < 0.01). The antiviral effects, however, were not observed in the dexamethasone-treated shHMGB1 cells. These results suggest that translocation of HMGB1 occurred during ZIKV infection and inhibition of the translocation by dexamethasone coincided with a reduction in ZIKV replication. These findings highlight the potential of targeting the localization of HMGB1 in affecting ZIKV infection.


Subject(s)
Dexamethasone/pharmacokinetics , HMGB1 Protein/metabolism , Zika Virus Infection/drug therapy , Zika Virus/drug effects , Cell Line, Tumor , Dexamethasone/metabolism , Gene Knockdown Techniques , HMGB1 Protein/genetics , Humans , Protein Transport/drug effects , Virus Replication/drug effects , Zika Virus/physiology
12.
Int J Mol Sci ; 23(2)2022 Jan 11.
Article in English | MEDLINE | ID: mdl-35054947

ABSTRACT

The cAMP-dependent aquaporin-2 (AQP2) redistribution from intracellular vesicles into the plasma membrane of renal collecting duct principal cells induces water reabsorption and fine-tunes body water homeostasis. However, the mechanisms controlling the localization of AQP2 are not understood in detail. Using immortalized mouse medullary collecting duct (MCD4) and primary rat inner medullary collecting duct (IMCD) cells as model systems, we here discovered a key regulatory role of Aurora kinase A (AURKA) in the control of AQP2. The AURKA-selective inhibitor Aurora-A inhibitor I and novel derivatives as well as a structurally different inhibitor, Alisertib, prevented the cAMP-induced redistribution of AQP2. Aurora-A inhibitor I led to a depolymerization of actin stress fibers, which serve as tracks for the translocation of AQP2-bearing vesicles to the plasma membrane. The phosphorylation of cofilin-1 (CFL1) inactivates the actin-depolymerizing function of CFL1. Aurora-A inhibitor I decreased the CFL1 phosphorylation, accounting for the removal of the actin stress fibers and the inhibition of the redistribution of AQP2. Surprisingly, Alisertib caused an increase in actin stress fibers and did not affect CFL1 phosphorylation, indicating that AURKA exerts its control over AQP2 through different mechanisms. An involvement of AURKA and CFL1 in the control of the localization of AQP2 was hitherto unknown.


Subject(s)
Aquaporin 2/metabolism , Aurora Kinase A/metabolism , Kidney Tubules, Collecting/metabolism , Actins/metabolism , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Cell Proliferation , Cell Survival/drug effects , Cyclic AMP/metabolism , Gene Silencing , Immunohistochemistry , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Male , Molecular Structure , Phosphorylation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Rats
13.
J Med Chem ; 65(3): 2507-2521, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35077161

ABSTRACT

Androgen receptor (AR) antagonists have been widely used for the treatment of prostate cancer (PCa). As a link between the AR and its transcriptional function, the activation function 2 (AF2) region has recently been revealed as a novel targeting site for developing AR antagonists. Here, we reported a series of N-(4-(benzyloxy)-phenyl)-sulfonamide derivatives as new-scaffold AR antagonists targeting the AR AF2. Therein, compound T1-12 showed excellent AR antagonistic activity (IC50 = 0.47 µM) and peptide displacement activity (IC50 = 18.05 µM). Furthermore, the in vivo LNCaP xenograft study confirmed that T1-12 offered effective inhibition on tumor growth when administered intratumorally. The study represents the first successful attempt to identify a small molecule targeting the AR AF2 with submicromolar AR antagonistic activity by structure-based virtual screening and provides important clues for the development of novel therapeutics for PCa treatment.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Receptors, Androgen/metabolism , Sulfonamides/therapeutic use , Androgen Receptor Antagonists/chemical synthesis , Androgen Receptor Antagonists/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Binding Sites , Cell Proliferation/drug effects , Gene Expression/drug effects , Humans , Male , Mice, SCID , Molecular Docking Simulation , Molecular Structure , Protein Transport/drug effects , Receptors, Androgen/chemistry , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/metabolism , Xenograft Model Antitumor Assays
14.
PLoS One ; 17(1): e0261098, 2022.
Article in English | MEDLINE | ID: mdl-35089929

ABSTRACT

JAK2 is cytokine-activated non-receptor tyrosine kinase. Although JAK2 is mainly localized at the plasma membrane, it is also present on the centrosome. In this study, we demonstrated that JAK2 localization to the centrosome depends on the SH2 domain and intact kinase activity. We created JAK2 mutants deficient in centrosomal localization ΔSH2, K882E and (ΔSH2, K882E). We showed that JAK2 WT clone strongly enhances cell proliferation as compared to control cells while JAK2 clones ΔSH2, K882E and (ΔSH2, K882E) proliferate slower than JAK2 WT cells. These mutant clones also progress much slower through the cell cycle as compared to JAK2 WT clone and the enhanced proliferation of JAK2 WT cells is accompanied by increased S -> G2 progression. Both the SH2 domain and the kinase activity of JAK2 play a role in prolactin-dependent activation of JAK2 substrate STAT5. We showed that JAK2 is an important regulator of centrosome function as the SH2 domain of JAK2 regulates centrosome amplification. The cells overexpressing ΔSH2 and (ΔSH2, K-E) JAK2 have almost three-fold the amplified centrosomes of WT cells. In contrast, the kinase activity of JAK2 is dispensable for centrosome amplification. Our observations provide novel insight into the role of SH2 domain and kinase activity of JAK2 in centrosome localization of JAK2 and in the regulation of cell growth and centrosome biogenesis.


Subject(s)
Cell Proliferation , Centrosome/metabolism , Janus Kinase 2/metabolism , src Homology Domains/genetics , Animals , COS Cells , Cell Cycle Checkpoints , Cell Line , Chlorocebus aethiops , Humans , Interferon-gamma/pharmacology , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Mutagenesis, Site-Directed , Protein Transport/drug effects , STAT5 Transcription Factor/metabolism
15.
J Cell Physiol ; 237(2): 1521-1531, 2022 02.
Article in English | MEDLINE | ID: mdl-34724217

ABSTRACT

Mechanical forces can modulate the immune response, mostly described as promoting the activation of immune cells, but the role and mechanism of pathological levels of mechanical stress in lymphocyte activation have not been focused on before. By an ex vivo experimental approach, we observed that mechanical stressing of murine spleen lymphocytes with 50 mmHg for 3 h induced the nuclear localization of NFAT1, increased C-Jun, and increased the expression of early activation marker CD69 in resting CD8+ cells. Interestingly, 50 mmHg mechanical stressing induced the nuclear localization of NFAT1; but conversely decreased C-Jun and inhibited the expression of CD69 in lymphocytes under lipopolysaccharide or phorbol 12-myristate 13-acetate/ionomycin stimulation. Additionally, we observed similar changes trends when comparing RNA-seq data of hypertensive and normotensive COVID-19 patients. Our results indicate a biphasic effect of mechanical stress on lymphocyte activation, which provides insight into the variety of immune responses in pathologies involving elevated mechanical stress.


Subject(s)
Lymphocyte Activation/immunology , Stress, Mechanical , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Biomarkers/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , COVID-19/complications , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Comorbidity , Gene Expression Regulation/drug effects , Humans , Hypertension/complications , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ion Channels/metabolism , Lectins, C-Type/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Male , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology
16.
Biotechnol Bioeng ; 119(1): 199-210, 2022 01.
Article in English | MEDLINE | ID: mdl-34698368

ABSTRACT

Ligand inducible proteins that enable precise and reversible control of nuclear translocation of passenger proteins have broad applications ranging from genetic studies in mammals to therapeutics that target diseases such as cancer and diabetes. One of the drawbacks of the current translocation systems is that the ligands used to control nuclear localization are either toxic or prone to crosstalk with endogenous protein cascades within live animals. We sought to take advantage of salicylic acid (SA), a small molecule that has been extensively used in humans. In plants, SA functions as a hormone that can mediate immunity and is sensed by the nonexpressor of pathogenesis-related (NPR) proteins. Although it is well recognized that nuclear translocation of NPR1 is essential to promoting immunity in plants, the exact subdomain of Arabidopsis thaliana NPR1 (AtNPR1) essential for SA-mediated nuclear translocation is controversial. Here, we utilized the fluorescent protein mCherry as the reporter to investigate the ability of SA to induce nuclear translocation of the full-length NPR1 protein or its C-terminal transactivation (TAD) domain using HEK293 cells as a heterologous system. HEK293 cells lack accessory plant proteins including NPR3/NPR4 and are thus ideally suited for studying the impact of SA-induced changes in NPR1. Our results obtained using a stable expression system show that the TAD of AtNPR1 is sufficient to enable the reversible SA-mediated nuclear translocation of mCherry. Our studies advance a basic understanding of nuclear translocation mediated by the TAD of AtNPR1 and uncover a biotechnological tool for SA-mediated nuclear localization.


Subject(s)
Arabidopsis Proteins , Cell Nucleus/metabolism , Recombinant Fusion Proteins , Salicylic Acid/pharmacology , Synthetic Biology/methods , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cytoplasm/metabolism , Gene Expression/drug effects , HEK293 Cells , Humans , Protein Transport/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Salicylic Acid/chemistry
17.
Am J Physiol Lung Cell Mol Physiol ; 322(1): L1-L12, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34704843

ABSTRACT

Respiratory syncytial virus (RSV) is an important human pathogen that causes severe lower respiratory tract infections in young children, the elderly, and the immunocompromised, yet no effective treatments or vaccines are available. The precise mechanism underlying RSV-induced acute airway disease and associated sequelae are not fully understood; however, early lung inflammatory and immune events are thought to play a major role in the outcome of the disease. Moreover, oxidative stress responses in the airways play a key role in the pathogenesis of RSV. Oxidative stress has been shown to elevate cytosolic calcium (Ca2+) levels, which in turn activate Ca2+-dependent enzymes, including transglutaminase 2 (TG2). Transglutaminase 2 is a multifunctional cross-linking enzyme implicated in various physiological and pathological conditions; however, its involvement in respiratory virus-induced airway inflammation is largely unknown. In this study, we demonstrated that RSV-induced oxidative stress promotes enhanced activation and release of TG2 from human lung epithelial cells as a result of its translocation from the cytoplasm and subsequent release into the extracellular space, which was mediated by Toll-like receptor (TLR)-4 and NF-κB pathways. Antioxidant treatment significantly inhibited RSV-induced TG2 extracellular release and activation via blocking viral replication. Also, treatment of RSV-infected lung epithelial cells with TG2 inhibitor significantly reduced RSV-induced matrix metalloprotease activities. These results suggested that RSV-induced oxidative stress activates innate immune receptors in the airways, such as TLRs, that can activate TG2 via the NF-κB pathway to promote cross-linking of extracellular matrix proteins, resulting in enhanced inflammation.


Subject(s)
Epithelial Cells/enzymology , Epithelial Cells/virology , Lung/pathology , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Respiratory Syncytial Virus Infections/enzymology , Respiratory Syncytial Virus, Human/physiology , Antioxidants/pharmacology , Cell Line , Epithelial Cells/drug effects , Fibronectins/metabolism , Gene Expression Regulation/drug effects , Humans , Matrix Metalloproteinases/metabolism , Models, Biological , NF-kappa B/metabolism , Protein Transport/drug effects , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus, Human/drug effects , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Virus Replication/drug effects , Virus Replication/physiology
18.
J Cell Biol ; 221(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34747980

ABSTRACT

Mitochondria and peroxisomes are independent but functionally closely related organelles. A few proteins have been characterized as dual-organelle locating proteins with distinct or similar roles on mitochondria and peroxisomes. MARCH5 is a mitochondria-associated ubiquitin ligase best known for its regulatory role in mitochondria quality control, fission, and fusion. Here, we used a proximity tagging system, PUP-IT, and identified new interacting proteins of MARCH5. Our data uncover that MARCH5 is a dual-organelle locating protein that interacts with several peroxisomal proteins. PEX19 binds the transmembrane region on MARCH5 and targets it to peroxisomes. On peroxisomes, MARCH5 binds and mediates the ubiquitination of PMP70. Furthermore, we find PMP70 ubiquitination and pexophagy induced by mTOR inhibition are blocked in the absence of MARCH5. Our study suggests novel roles of MARCH5 on peroxisomes.


Subject(s)
Macroautophagy , Membrane Proteins/metabolism , Peroxisomes/metabolism , Ubiquitin-Protein Ligases/metabolism , ATP-Binding Cassette Transporters/metabolism , Blood Proteins/pharmacology , HeLa Cells , Humans , Jurkat Cells , Lipoproteins/metabolism , Macroautophagy/drug effects , Peroxins/metabolism , Peroxisomes/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Ubiquitination
19.
Biomed Pharmacother ; 145: 112471, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34852990

ABSTRACT

NF-κB contributes to the biosynthesis of various chemokines, cytokines, and enzymes. It plays many crucial roles in the upstream neuroinflammatory pathways. Briefly, the inhibitory IkB subunit is cleaved and phosphorylated by the IKK-α/ß enzyme. It leads to the activation and translocation of the NF-κB (p50/p65) complex into the nucleus. Subsequently, the activated NF-κB interacts with the genomic DNA and contributes to expressing various proinflammatory cytokines. In the present study, we developed a novel NF-κB inhibitor encoded (D5) and investigated the efficacy of our druggable compound through several in silico, in vitro, and in situ analysis. The results demonstrated that D5 not only inhibited the mRNA expression of the IKK-α/ß enzyme (around 86-96% suppression rate for both cell lines at 12 and 24 h time frames) but also by interacting to the active site of the mentioned kinase (dock score -6.14 and binding energy -23.60 kcal/mol) reduced the level of phosphorylated IkB-α in the cytosol around 96-99% and p65 subunit in the nucleus around 73-90% (among all groups in 12 and 24 h time points). Additionally, the results indicated that D5 suppressed the NF-κB target mRNA levels of TNF-α and IL-6 in a total average of around 92%. Overall, The results demonstrated that D5 in a considerably lower concentration than Dis (0.71 µM vs. 52.73 µM) showed significantly higher inhibitory efficacy on NF-κB translocation approx. 200-300%. The results suggested D5 as a potent NF-κB silencer, but further investigations are required to validate our outcomes.


Subject(s)
I-kappa B Kinase , NF-kappa B/metabolism , Neuroinflammatory Diseases , Protein Translocation Systems , Alkaloids/pharmacology , Benzodioxoles/pharmacology , Cell Line , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Drug Development/methods , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Phosphorylation/drug effects , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Protein Translocation Systems/drug effects , Protein Translocation Systems/metabolism , Protein Transport/drug effects , Signal Transduction/drug effects , Transcription Factor RelA/metabolism
20.
Anticancer Res ; 42(1): 471-482, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34969757

ABSTRACT

BACKGROUND/AIM: HDAC6, a cytoplasmic localized deacetylase, is a positive regulator of cancer progression via modification of various substrates. We evaluated how the interaction between HDAC6 and glucose regulatory protein 78 (GRP78) affects the growth of cholangiocarcinoma (CCA). MATERIALS AND METHODS: The anti-tumor effects of ACY-1215, an HDAC6 specific inhibitor, in CCA cell lines were analyzed by cell viability assay, western blotting, flow cytometry, co-immunoprecipitation, and biotinylation assays. In vivo effects of ACY-1215 were evaluated in a xenograft model using CCA cell line TFK-1. RESULTS: ACY-1215 increased the acetyl-form of GRP78 by approximately 50% compared to control, which impaired the translocation of GRP78 to the plasma membrane by 50% through alteration of cellular proliferative signaling via PI3K/AKT. Furthermore, ACY-1215 suppressed tumor growth by 50% compared to vehicle control in a CCA xenograft model. CONCLUSION: Increase in GRP78 acetylation by HDAC6 inhibition suppressed GRP78 translocation to the cell surface, which inhibited proliferation and promoted apoptosis in CCA.


Subject(s)
Cell Proliferation/drug effects , Cholangiocarcinoma/drug therapy , Endoplasmic Reticulum Chaperone BiP/genetics , Histone Deacetylase 6/genetics , Animals , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Survival/drug effects , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Endoplasmic Reticulum Chaperone BiP/antagonists & inhibitors , Flow Cytometry , Humans , Hydroxamic Acids/pharmacology , Mice , Phosphatidylinositol 3-Kinases/genetics , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/genetics , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...