Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55.931
Filter
1.
Sci Signal ; 17(843): eabq7038, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38954638

ABSTRACT

Mini-G proteins are engineered, thermostable variants of Gα subunits designed to stabilize G protein-coupled receptors (GPCRs) in their active conformations. Because of their small size and ease of use, they are popular tools for assessing GPCR behaviors in cells, both as reporters of receptor coupling to Gα subtypes and for cellular assays to quantify compartmentalized signaling at various subcellular locations. Here, we report that overexpression of mini-G proteins with their cognate GPCRs disrupted GPCR endocytic trafficking and associated intracellular signaling. In cells expressing the Gαs-coupled GPCR glucagon-like peptide 1 receptor (GLP-1R), coexpression of mini-Gs, a mini-G protein derived from Gαs, blocked ß-arrestin 2 recruitment and receptor internalization and disrupted endosomal GLP-1R signaling. These effects did not involve changes in receptor phosphorylation or lipid nanodomain segregation. Moreover, we found that mini-G proteins derived from Gαi and Gαq also inhibited the internalization of GPCRs that couple to them. Finally, we developed an alternative intracellular signaling assay for GLP-1R using a nanobody specific for active Gαs:GPCR complexes (Nb37) that did not affect GLP-1R internalization. Our results have important implications for designing methods to assess intracellular GPCR signaling.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Protein Engineering , Receptors, G-Protein-Coupled , Signal Transduction , Humans , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , HEK293 Cells , Protein Engineering/methods , Endocytosis/physiology , Protein Transport , Animals
2.
Methods Enzymol ; 700: 385-411, 2024.
Article in English | MEDLINE | ID: mdl-38971608

ABSTRACT

Plasma membranes are flexible and can exhibit numerous shapes below the optical diffraction limit. The shape of cell periphery can either induce or be a product of local protein density changes, encoding numerous cellular functions. However, quantifying membrane curvature and the ensuing sorting of proteins in live cells remains technically demanding. Here, we demonstrate the use of simple widefield fluorescence microscopy to study the geometrical properties (i.e., radius, length, and number) of thin membrane protrusions. Importantly, the quantification of protrusion radius establishes a platform for studying the curvature preferences of membrane proteins.


Subject(s)
Membrane Proteins , Microscopy, Fluorescence , Protein Transport , Microscopy, Fluorescence/methods , Humans , Membrane Proteins/metabolism , Membrane Proteins/analysis , Cell Membrane/metabolism , Cell Membrane/chemistry , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Animals
3.
J Cell Biol ; 223(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38958655

ABSTRACT

Export of secretory cargoes from the endoplasmic reticulum (ER) requires COPII proteins, which were first identified for their ability to coat small vesicles that bud from the ER. Recent data indicate that COPII proteins can also organize into a collar at the necks of tubules, as well as phase-separate into liquid-like condensates. Thus, COPII assemblies seem to be tailored to accommodate variations in the size and quantities of cargo secreted.


Subject(s)
COP-Coated Vesicles , Endoplasmic Reticulum , Protein Transport , Vesicular Transport Proteins , COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Humans , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Animals
4.
Cells ; 13(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38995007

ABSTRACT

Primary cilia are finger-like sensory organelles that extend from the bodies of most cell types and have a distinct lipid and protein composition from the plasma membrane. This partitioning is maintained by a diffusion barrier that restricts the entry of non-ciliary proteins, and allows the selective entry of proteins harboring a ciliary targeting sequence (CTS). However, CTSs are not stereotyped and previously reported sequences are insufficient to drive efficient ciliary localisation across diverse cell types. Here, we describe a short peptide sequence that efficiently targets transmembrane proteins to primary cilia in all tested cell types, including human neurons. We generate human-induced pluripotent stem cell (hiPSC) lines stably expressing a transmembrane construct bearing an extracellular HaloTag and intracellular fluorescent protein, which enables the bright, specific labeling of primary cilia in neurons and other cell types to facilitate studies of cilia in health and disease. We demonstrate the utility of this resource by developing an image analysis pipeline for the automated measurement of primary cilia to detect changes in their length associated with altered signaling or disease state.


Subject(s)
Cilia , Induced Pluripotent Stem Cells , Membrane Proteins , Cilia/metabolism , Humans , Membrane Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Animals , Neurons/metabolism , Amino Acid Sequence , Cell Line , Protein Transport
5.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000460

ABSTRACT

Aberrant aggregation of misfolded alpha-synuclein (α-syn), a major pathological hallmark of related neurodegenerative diseases such as Parkinson's disease (PD), can translocate between cells. Ubiquitin-like 3 (UBL3) is a membrane-anchored ubiquitin-fold protein and post-translational modifier. UBL3 promotes protein sorting into small extracellular vesicles (sEVs) and thereby mediates intercellular communication. Our recent studies have shown that α-syn interacts with UBL3 and that this interaction is downregulated after silencing microsomal glutathione S-transferase 3 (MGST3). However, how MGST3 regulates the interaction of α-syn and UBL3 remains unclear. In the present study, we further explored this by overexpressing MGST3. In the split Gaussia luciferase complementation assay, we found that the interaction between α-syn and UBL3 was upregulated by MGST3. While Western blot and RT-qPCR analyses showed that silencing or overexpression of MGST3 did not significantly alter the expression of α-syn and UBL3, the immunocytochemical staining analysis indicated that MGST3 increased the co-localization of α-syn and UBL3. We suggested roles for the anti-oxidative stress function of MGST3 and found that the effect of MGST3 overexpression on the interaction between α-syn with UBL3 was significantly rescued under excess oxidative stress and promoted intracellular α-syn to extracellular transport. In conclusion, our results demonstrate that MGST3 upregulates the interaction between α-syn with UBL3 and promotes the interaction to translocate intracellular α-syn to the extracellular. Overall, our findings provide new insights and ideas for promoting the modulation of UBL3 as a therapeutic agent for the treatment of synucleinopathy-associated neurodegenerative diseases.


Subject(s)
Glutathione Transferase , Oxidative Stress , Ubiquitins , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Humans , Glutathione Transferase/metabolism , Glutathione Transferase/genetics , Ubiquitins/metabolism , Ubiquitins/genetics , Up-Regulation , Protein Transport , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Protein Binding
6.
J Cell Biol ; 223(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-38967608

ABSTRACT

Peroxisomes are membrane-bound organelles harboring metabolic enzymes. In humans, peroxisomes are required for normal development, yet the genes regulating peroxisome function remain unclear. We performed a genome-wide CRISPRi screen to identify novel factors involved in peroxisomal homeostasis. We found that inhibition of RNF146, an E3 ligase activated by poly(ADP-ribose), reduced the import of proteins into peroxisomes. RNF146-mediated loss of peroxisome import depended on the stabilization and activity of the poly(ADP-ribose) polymerases TNKS and TNKS2, which bind the peroxisomal membrane protein PEX14. We propose that RNF146 and TNKS/2 regulate peroxisome import efficiency by PARsylation of proteins at the peroxisome membrane. Interestingly, we found that the loss of peroxisomes increased TNKS/2 and RNF146-dependent degradation of non-peroxisomal substrates, including the ß-catenin destruction complex component AXIN1, which was sufficient to alter the amplitude of ß-catenin transcription. Together, these observations not only suggest previously undescribed roles for RNF146 in peroxisomal regulation but also a novel role in bridging peroxisome function with Wnt/ß-catenin signaling during development.


Subject(s)
Axin Protein , Peroxisomes , Ubiquitin-Protein Ligases , Wnt Signaling Pathway , Peroxisomes/metabolism , Peroxisomes/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Humans , Axin Protein/metabolism , Axin Protein/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , beta Catenin/metabolism , beta Catenin/genetics , HEK293 Cells , Protein Transport , CRISPR-Cas Systems
7.
Commun Biol ; 7(1): 826, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38972875

ABSTRACT

Classically, G protein-coupled receptors (GPCRs) promote signaling at the plasma membrane through activation of heterotrimeric Gαßγ proteins, followed by the recruitment of GPCR kinases and ßarrestin (ßarr) to initiate receptor desensitization and internalization. However, studies demonstrated that some GPCRs continue to signal from internalized compartments, with distinct cellular responses. Both ßarr and Gßγ contribute to such non-canonical endosomal G protein signaling, but their specific roles and contributions remain poorly understood. Here, we demonstrate that the vasopressin V2 receptor (V2R)-ßarr complex scaffolds Gßγ at the plasma membrane through a direct interaction with ßarr, enabling its transport to endosomes. Gßγ subsequently potentiates Gαs endosomal translocation, presumably to regenerate an endosomal pool of heterotrimeric Gs. This work shines light on the mechanism underlying G protein subunits translocation from the plasma membrane to the endosomes and provides a basis for understanding the role of ßarr in mediating sustained G protein signaling.


Subject(s)
Endosomes , GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Protein Transport , Receptors, Vasopressin , beta-Arrestins , Humans , beta-Arrestins/metabolism , Cell Membrane/metabolism , Endosomes/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , GTP-Binding Protein gamma Subunits/genetics , HEK293 Cells , Receptors, Vasopressin/metabolism , Receptors, Vasopressin/genetics , Signal Transduction
8.
Acta Biochim Pol ; 71: 13004, 2024.
Article in English | MEDLINE | ID: mdl-39041003

ABSTRACT

CD36 is a type 2 cell surface scavenger receptor expressed in various tissues. In macrophages, CD36 recognizes oxidized low-density lipoprotein (ox-LDL), which promotes the formation of foam cells, the first step toward an atherosclerotic arterial lesion. CD36 possesses a variety of posttranslational modifications, among them N-glycosylation and O-GlcNAc modification. Some of the roles of these modifications on CD36 are known, such as N-linked glycosylation, which provides proper folding and trafficking to the plasma membrane in the human embryonic kidney. This study aimed to determine whether variations in the availability of UDP-GlcNAc could impact Rab-5-mediated endocytic trafficking and, therefore, the cellular localization of CD36. These preliminary results suggest that the availability of the substrate UDP-GlcNAc, modulated in response to treatment with Thiamet G (TMG), OSMI-1 (O-GlcNAcylation enzymes modulators) or Azaserine (HBP modulator), influences the localization of CD36 in J774 macrophages, and the endocytic trafficking as evidenced by the regulatory protein Rab-5, between the plasma membrane and the cytoplasm.


Subject(s)
CD36 Antigens , Macrophages , CD36 Antigens/metabolism , Macrophages/metabolism , Animals , Mice , Cell Line , Glycosylation , Cell Membrane/metabolism , Humans , Lipoproteins, LDL/metabolism , Hexosamines/metabolism , Hexosamines/biosynthesis , rab5 GTP-Binding Proteins/metabolism , Protein Transport , Biosynthetic Pathways , Protein Processing, Post-Translational
9.
J Agric Food Chem ; 72(29): 16298-16311, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38982710

ABSTRACT

From the fruits of Cordia dichotoma, 11 new phenolic compounds, dichotomins A-K, were isolated, together with 19 known compounds. Through the analysis of detailed NMR data and HRESIMS data, the planar structures of all compounds were confirmed. Using NMR calculations, the absolute configuration of dichotomins A-K was elucidated by comparing their observed and computed electronic circular dichroism (ECD) spectra. Dichotomin H (8) and dichotomin I (9) were determined as two pairs of enantiomers. The enantiomers of compounds 8 and 9 were separated using chiral-phase high-performance liquid chromatography (HPLC), and the stereostructure of each enantiomer was determined by similarly calculating the ECD. Compounds 3, 5, 7, 17, 18, 23-25, and 27-30 increased glucose uptake by 1.04- to 2.85-folds at concentrations of 30 µg/mL. Further studies revealed that compounds 3 and 5 had a moderate effect on glucose transporter 4 (GLUT4) translocation activity in L6 cells. At 30 µg/mL, compound 3 significantly enhanced AMPK phosphorylation and GLUT4 expression. As a whole, compound 3 has the potential to be a drug candidate for the treatment of type 2 diabetes mellitus (T2DM).


Subject(s)
Fruit , Glucose Transporter Type 4 , Glucose , Phenols , Plant Extracts , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 4/genetics , Fruit/chemistry , Glucose/metabolism , Phenols/chemistry , Phenols/pharmacology , Phenols/metabolism , Animals , Rats , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/metabolism , Biological Transport/drug effects , Molecular Structure , Cell Line , Protein Transport , Humans , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/chemistry
10.
J Physiol Pharmacol ; 75(3)2024 Jun.
Article in English | MEDLINE | ID: mdl-39042386

ABSTRACT

Suicide is a significant public health challenge worldwide. Statistical data confirm a strong relationship between suicidal behavior and depressive disorders (DDs), but the molecular mechanisms of these diseases are still poorly understood. A growing body of research suggests that the Klotho-mediated pathway may be a novel intracellular target for the development of suicide-related disorders (including DDs). To verify this hypothesis, the link between α-Klotho levels, Nrf2-related inflammatory status (IL-1α, IL-1ß, Keap1, NFκB p65), AMPA (GluA1, GluA2, p-S831-GluA1, p-S845-GluA1) receptor subunit trafficking and AMPK (AMPKα1/2; pT172-AMPKα1) signalling pathways in the brain of suicide victims as compared to controls were investigated. Commercially available enzyme-linked immunoassay (ELISA) and Western blot analysis were performed in the hippocampus (HP) and frontal cortex (FCx) of suicide victims and matched controls. Group differences were assessed using an unpaired Student's t-test. A statistically significant decrease in the level of α-Klotho (HP: p=0.001; FCx: p=0.012) with an increase in IL-1ß (HP: p=0.0108) and IL-1α (FCx: p=0.009) concentrations were shown. These alterations were associated with increased Keap1 (FCx: p=0.023) and NF-κB-p65 (HP: p=0.039; FCx: p=0.013 nuclear fraction) protein levels. Furthermore, a significant reduction in p-S831-GluA1 (HP: p=0.029; FCx=0.002) and p-S845-GluA1 (HP: p=0.0012) proteins was observed. Similarly, the level of GluA2 (HP: p=0.011; FCx: p=0.002) and in p-T172-AMPKα1 (HP: p=0.0288; FCx: p=0.0338) protein were statistically decreased. Our findings demonstrate that a reduction in α-Klotho levels in brain structures related to mood disorders (HP, FCx) correlates with suicidal behavior. Moreover, our study provides novel insights into the molecular mechanisms underlying suicide-related disorders, highlighting the role of α-Klotho, Nrf2-related inflammatory status, AMPA receptor trafficking, and AMPK signaling pathways in the pathophysiology of suicidal behavior. These results may have implications for the development of targeted interventions for individuals at risk of suicide.


Subject(s)
Glucuronidase , Kelch-Like ECH-Associated Protein 1 , Klotho Proteins , NF-E2-Related Factor 2 , Receptors, AMPA , Signal Transduction , Suicide , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Male , Suicide/psychology , Receptors, AMPA/metabolism , Female , Adult , Glucuronidase/metabolism , Middle Aged , Protein Transport , Brain/metabolism , Interleukin-1beta/metabolism , Hippocampus/metabolism , Frontal Lobe/metabolism , Young Adult
11.
Med Microbiol Immunol ; 213(1): 15, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39008129

ABSTRACT

Chlamydiae are a large group of obligate endosymbionts of eukaryotes that includes the Chlamydiaceae family, comprising several animal pathogens. Among Chlamydiaceae, Chlamydia trachomatis causes widespread ocular and urogenital infections in humans. Like many bacterial pathogens, all Chlamydiae manipulate host cells by injecting them with type III secretion effector proteins. We previously characterized the C. trachomatis effector CteG, which localizes at the host cell Golgi and plasma membrane during distinct phases of the chlamydial infectious cycle. Here, we show that CteG is a Chlamydiaceae-specific effector with over 60 homologs phylogenetically categorized into two distinct clades (CteG I and CteG II) and exhibiting several inparalogs and outparalogs. Notably, cteG I homologs are syntenic to C. trachomatis cteG, whereas cteG II homologs are syntenic among themselves but not with C. trachomatis cteG. This indicates a complex evolution of cteG homologs, which is unique among C. trachomatis effectors, marked by numerous events of gene duplication and loss. Despite relatively modest sequence conservation, nearly all tested CteG I and CteG II proteins were identified as type III secretion substrates using Yersinia as a heterologous bacterial host. Moreover, most of the type III secreted CteG I and CteG II homologs were delivered by C. trachomatis into host cells, where they localized at the Golgi region and cell periphery. Overall, this provided insights into the evolution of bacterial effectors and revealed a Chlamydiaceae family of type III secreted proteins that underwent substantial divergence during evolution while conserving the capacity to localize at specific host cell compartments.


Subject(s)
Bacterial Proteins , Chlamydia trachomatis , Phylogeny , Type III Secretion Systems , Humans , Chlamydia trachomatis/genetics , Chlamydia trachomatis/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Type III Secretion Systems/metabolism , Type III Secretion Systems/genetics , Virulence Factors/metabolism , Virulence Factors/genetics , HeLa Cells , Yersinia/genetics , Yersinia/metabolism , Protein Transport , Host-Pathogen Interactions , Evolution, Molecular , Chlamydiaceae/genetics , Chlamydiaceae/metabolism , Chlamydiaceae/classification
12.
Proc Natl Acad Sci U S A ; 121(28): e2317833121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968112

ABSTRACT

Parkinson's disease (PD) is a multifactorial disease that affects multiple brain systems and circuits. While defined by motor symptoms caused by degeneration of brainstem dopamine neurons, debilitating non-motor abnormalities in fronto-striatal-based cognitive function are common, appear early, and are initially independent of dopamine. Young adult mice expressing the PD-associated G2019S missense mutation in Lrrk2 also exhibit deficits in fronto-striatal-based cognitive tasks. In mice and humans, cognitive functions require dynamic adjustments in glutamatergic synapse strength through cell-surface trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptors (AMPARs), but it is unknown how LRRK2 mutation impacts dynamic features of AMPAR trafficking in striatal projection neurons (SPNs). Here, we used Lrrk2G2019S knockin mice to show that surface AMPAR subunit stoichiometry is altered biochemically and functionally in mutant SPNs in dorsomedial striatum to favor the incorporation of GluA1 over GluA2. GluA1-containing AMPARs were resistant to internalization from the cell surface, leaving an excessive accumulation of GluA1 on the surface within and outside synapses. This negatively impacted trafficking dynamics that normally support synapse strengthening, as GluA1-containing AMPARs failed to increase at synapses in response to a potentiating stimulus and showed significantly reduced surface mobility. Surface GluA2-containing AMPARs were expressed at normal levels in synapses, indicating subunit-selective impairment. Abnormal surface accumulation of GluA1 was independent of PKA activity and was limited to D1R SPNs. Since LRRK2 mutation is thought to be part of a common PD pathogenic pathway, our data suggest that sustained, striatal cell-type specific changes in AMPAR composition and trafficking contribute to cognitive or other impairments associated with PD.


Subject(s)
Corpus Striatum , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease , Protein Transport , Receptors, AMPA , Animals , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Receptors, AMPA/metabolism , Receptors, AMPA/genetics , Mice , Corpus Striatum/metabolism , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Mutation, Missense , Humans , Synapses/metabolism
13.
Methods Mol Biol ; 2814: 133-147, 2024.
Article in English | MEDLINE | ID: mdl-38954203

ABSTRACT

Activation processes at the plasma membrane have been studied with life-cell imaging using GFP fused to a protein that binds to a component of the activation process. In this way, PIP3 formation has been monitored with CRAC-GFP, Ras-GTP with RBD-Raf-GFP, and Rap-GTP with Ral-GDS-GFP. The fluorescent sensors translocate from the cytoplasm to the plasma membrane upon activation of the process. Although this translocation assay can provide very impressive images and movies, the method is not very sensitive, and amount of GFP-sensor at the plasma membrane is not linear with the amount of activator. The fluorescence in pixels at the cell boundary is partly coming from the GFP-sensor that is bound to the activated membrane and partly from unbound GFP-sensor in the cytosolic volume of that boundary pixel. The variable and unknown amount of cytosol in boundary pixels causes the low sensitivity and nonlinearity of the GFP-translocation assay. Here we describe a method in which the GFP-sensor is co-expressed with cytosolic-RFP. For each boundary pixels, the RFP fluorescence is used to determine the amount of cytosol of that pixel and is subtracted from the GFP fluorescence of that pixel yielding the amount of GFP-sensor that is specifically associated with the plasma membrane in that pixel. This GRminusRD method using GFP-sensor/RFP is at least tenfold more sensitive, more reproducible, and linear with activator compared to GFP-sensor alone.


Subject(s)
Cell Membrane , Green Fluorescent Proteins , Cell Membrane/metabolism , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Humans , Luminescent Proteins/metabolism , Luminescent Proteins/genetics , Protein Transport , Microscopy, Fluorescence/methods , Cytosol/metabolism , Animals
14.
Mol Biol Cell ; 35(8): ar113, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38985515

ABSTRACT

Coat protein complex II (COPII) governs the initial steps of biosynthetic secretory protein transport from the endoplasmic reticulum (ER), facilitating the movement of a wide variety of cargoes. Here, we demonstrate that Trk-fused gene (TFG) regulates the rate at which inner COPII coat proteins are concentrated at ER subdomains. Specifically, in cells lacking TFG, the GTPase-activating protein (GAP) Sec23 accumulates more rapidly at budding sites on the ER as compared with control cells, potentially altering the normal timing of GTP hydrolysis on Sar1. Under these conditions, anterograde trafficking of several secretory cargoes is delayed, irrespective of their predicted size. We propose that TFG controls the local, freely available pool of Sec23 during COPII coat formation and limits its capacity to prematurely destabilize COPII complexes on the ER. This function of TFG enables it to act akin to a rheostat, promoting the ordered recruitment of Sec23, which is critical for efficient secretory cargo export.


Subject(s)
COP-Coated Vesicles , Endoplasmic Reticulum , Monomeric GTP-Binding Proteins , Protein Transport , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Vesicular Transport Proteins , COP-Coated Vesicles/metabolism , Endoplasmic Reticulum/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Monomeric GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Golgi Apparatus/metabolism
15.
Proc Natl Acad Sci U S A ; 121(29): e2320709121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38985760

ABSTRACT

The Type-I interferon (IFN-I) response is the major outcome of stimulator of interferon genes (STING) activation in innate cells. STING is more abundantly expressed in adaptive T cells; nevertheless, its intrinsic function in T cells remains unclear. Intriguingly, we previously demonstrated that STING activation in T cells activates widespread IFN-independent activities, which stands in contrast to the well-known STING-mediated IFN response. Here, we have identified that STING activation induces regulatory T cells (Tregs) differentiation independently of IRF3 and IFN. Specifically, the translocation of STING from the endoplasmic reticulum to the Golgi activates mitogen-activated protein kinase (MAPK) activity, which subsequently triggers transcription factor cAMP response element-binding protein (CREB) activation. The activation of the STING-MAPK-CREB signaling pathway induces the expression of many cytokine genes, including interleukin-2 (IL-2) and transforming growth factor-beta 2 (TGF-ß2), to promote the Treg differentiation. Genetic knockdown of MAPK p38 or pharmacological inhibition of MAPK p38 or CREB markedly inhibits STING-mediated Treg differentiation. Administration of the STING agonist also promotes Treg differentiation in mice. In the Trex1-/- autoimmune disease mouse model, we demonstrate that intrinsic STING activation in CD4+ T cells can drive Treg differentiation, potentially counterbalancing the autoimmunity associated with Trex1 deficiency. Thus, STING-MAPK-CREB represents an IFN-independent signaling axis of STING that may have profound effects on T cell effector function and adaptive immunity.


Subject(s)
Cell Differentiation , Cyclic AMP Response Element-Binding Protein , Membrane Proteins , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Mice , Signal Transduction , MAP Kinase Signaling System , Mice, Inbred C57BL , Protein Transport , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Mice, Knockout , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Nat Commun ; 15(1): 6068, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025931

ABSTRACT

Neurexins are key adhesion proteins that coordinate extracellular and intracellular synaptic components. Nonetheless, the low abundance of these multidomain proteins has complicated any localization and structure-function studies. Here we combine an ALFA tag (AT)/nanobody (NbALFA) tool with classic genetics, cell biology and electrophysiology to examine the distribution and function of the Drosophila Nrx-1 in vivo. We generate full-length and ΔPDZ ALFA-tagged Nrx-1 variants and find that the PDZ binding motif is key to Nrx-1 surface expression. A PDZ binding motif provided in trans, via genetically encoded cytosolic NbALFA-PDZ chimera, fully restores the synaptic localization and function of NrxΔPDZ-AT. Using cytosolic NbALFA-mScarlet intrabody, we achieve compartment-specific detection of endogenous Nrx-1, track live Nrx-1 transport along the motor neuron axons, and demonstrate that Nrx-1 co-migrates with Rab2-positive vesicles. Our findings illustrate the versatility of the ALFA system and pave the way towards dissecting functional domains of complex proteins in vivo.


Subject(s)
Drosophila Proteins , Single-Domain Antibodies , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Single-Domain Antibodies/metabolism , Drosophila melanogaster/metabolism , Motor Neurons/metabolism , PDZ Domains , Axons/metabolism , Neural Cell Adhesion Molecules/metabolism , Neural Cell Adhesion Molecules/genetics , Protein Transport , Cell Adhesion Molecules, Neuronal
17.
Cell Mol Life Sci ; 81(1): 302, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39008111

ABSTRACT

DNAX-activating protein of 12 kDa (DAP12) is a transmembrane adapter protein expressed in lymphoid and myeloid lineage cells. It interacts with several immunoreceptors forming functional complexes that trigger intracellular signaling pathways. One of the DAP12 associated receptors is the triggering receptor expressed on myeloid cells 2 (TREM2). Mutations in both DAP12 and TREM2 have been linked to neurodegenerative diseases. However, mechanisms involved in the regulation of subcellular trafficking and turnover of these proteins are not well understood. Here, we demonstrate that proteasomal degradation of DAP12 is increased in the absence of TREM2. Interestingly, unassembled DAP12 is also retained in early secretory compartments, including the endoplasmic reticulum (ER) and the ER-Golgi intermediate compartment (ERGIC), thereby preventing its transport to the plasma membrane. We also show that unassembled DAP12 interacts with the retention in ER sorting receptor 1 (RER1). The deletion of endogenous RER1 decreases expression of functional TREM2-DAP12 complexes and membrane proximal signaling, and resulted in almost complete inhibition of phagocytic activity in THP-1 differentiated macrophage-like cells. These results indicate that RER1 acts as an important regulator of DAP12 containing immunoreceptor complexes and immune cell function.


Subject(s)
Adaptor Proteins, Signal Transducing , Endoplasmic Reticulum , Membrane Glycoproteins , Receptors, Immunologic , Secretory Pathway , Humans , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Endoplasmic Reticulum/metabolism , Secretory Pathway/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , HEK293 Cells , Signal Transduction , Phagocytosis/genetics , Macrophages/metabolism , Protein Transport , Protein Binding , Animals , Golgi Apparatus/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Cell Membrane/metabolism
18.
Mol Biol Rep ; 51(1): 829, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39037581

ABSTRACT

BACKGROUND: The roles of Caveolin-1 (Cav-1) and the Wnt/ß-catenin signaling pathways in cerebral ischemia-reperfusion (I/R) injury are well established. The translocation of ß-catenin into the nucleus is critical for regulating neuronal apoptosis, repair, and neurogenesis within the ischemic brain. It has been reported that the scaffold domain of Caveolin-1 (Cav-1) (residues 95-98) interacts with ß-catenin (residues 330-337). However, the specific contribution of the Cav-1/ß-catenin complex to I/R injury remains unknown. METHODS AND RESULTS: To investigate the mechanism underlying the involvement of the Cav-1/ß-catenin complex in the subcellular translocation of ß-catenin and its subsequent effects on cerebral I/R injury, we treated ischemic brains with ASON (Cav-1 antisense oligodeoxynucleotides) or FTVT (a competitive peptide antagonist of the Cav-1 and ß-catenin interaction). Our study demonstrated that the binding of Cav-1 to ß-catenin following I/R injury prevented the nuclear accumulation of ß-catenin. Treatment with ASON or FTVT after I/R injury significantly increased the levels of nuclear ß-catenin. Furthermore, ASON reduced the phosphorylation of ß-catenin at Ser33, Ser37, and Thr41, which contributes to its proteasomal degradation, while FTVT increased phosphorylation at Tyr333, which is associated with its nuclear translocation. CONCLUSIONS: The above results indicate that the formation of the Cav-1/ß-catenin complex anchors ß-catenin in the cytoplasm following I/R injury. Additionally, both ASON and FTVT treatments attenuated neuronal death in ischemic brains. Our study suggests that targeting the interaction between Cav-1 and ß-catenin serve as a novel therapeutic strategy to protect against neuronal damage during cerebral injury.


Subject(s)
Caveolin 1 , Cell Nucleus , Neurons , Reperfusion Injury , beta Catenin , beta Catenin/metabolism , Animals , Reperfusion Injury/metabolism , Caveolin 1/metabolism , Caveolin 1/genetics , Neurons/metabolism , Neurons/pathology , Cell Nucleus/metabolism , Male , Rats , Brain Ischemia/metabolism , Brain Ischemia/pathology , Apoptosis , Wnt Signaling Pathway , Rats, Sprague-Dawley , Protein Binding , Protein Transport , Cell Death
19.
Methods Mol Biol ; 2814: 107-118, 2024.
Article in English | MEDLINE | ID: mdl-38954201

ABSTRACT

A given protein can perform numerous roles in a cell with its participation in protein complexes and distinct localization within the cell playing a critical role in its diverse functions. Thus, the ability to artificially dimerize proteins and recruit proteins to specific locations in a cell has become a powerful tool for the investigation of protein function and the understanding of cell biology. Here, we discuss two systems that have been used to activate signal transduction pathways, a chemically inducible dimerization (CID) and a light-inducible (LI) system to control signaling and cytoskeletal regulation in a spatial and temporal manner.


Subject(s)
Optogenetics , Protein Multimerization , Signal Transduction , Optogenetics/methods , Humans , Animals , Light , Protein Transport
20.
Nat Commun ; 15(1): 5618, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965227

ABSTRACT

Naturally generated lipid nanoparticles termed extracellular vesicles (EVs) hold significant promise as engineerable therapeutic delivery vehicles. However, active loading of protein cargo into EVs in a manner that is useful for delivery remains a challenge. Here, we demonstrate that by rationally designing proteins to traffic to the plasma membrane and associate with lipid rafts, we can enhance loading of protein cargo into EVs for a set of structurally diverse transmembrane and peripheral membrane proteins. We then demonstrate the capacity of select lipid tags to mediate increased EV loading and functional delivery of an engineered transcription factor to modulate gene expression in target cells. We envision that this technology could be leveraged to develop new EV-based therapeutics that deliver a wide array of macromolecular cargo.


Subject(s)
Extracellular Vesicles , Nanoparticles , Extracellular Vesicles/metabolism , Humans , Nanoparticles/chemistry , Protein Engineering/methods , Membrane Microdomains/metabolism , Lipids/chemistry , Cell Membrane/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Animals , Drug Delivery Systems , Protein Transport , HEK293 Cells , Liposomes
SELECTION OF CITATIONS
SEARCH DETAIL
...